1
|
Li X, Lou C, Ren H, Cui L, Chen K. Fundamental knowledge and research regarding the role of immunity in triple-negative breast cancer from 2014-2024: A bibliometric analysis. Hum Vaccin Immunother 2025; 21:2483022. [PMID: 40135819 PMCID: PMC11951696 DOI: 10.1080/21645515.2025.2483022] [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: 09/13/2024] [Revised: 02/27/2025] [Accepted: 03/18/2025] [Indexed: 03/27/2025] Open
Abstract
Immunity has vital research value and promising applications in triple-negative breast cancer (TNBC). Nevertheless, few bibliometric analyses have systematically investigated this area. This study aimed to comprehensively review the collaboration and impact of countries, institutions, authors, and journals on the role of immunity in TNBC from a bibliometric perspective, evaluate the keyword co-occurrence of the knowledge structure, and identify hot trends and emerging topics. Articles and reviews related to immunity in TNBC were retrieved from the Web of Science core collection using subject search. A bibliometric study was conducted primarily using CiteSpace and VOSviewer. A total of 3,104 articles and reviews were included from January 1, 2014, through December 31, 2024. The number of articles on immunization in TNBC is rising. These publications are mainly from 415 institutions in 82 countries, led by China and the USA. Among these publications, Lajos Pusztai published the most papers, while Peter Schmid was co-cited the most. The most productive journals focused on molecular biology, biological immunology, and clinical medicine. Furthermore, co-citation analysis revealed that tumor microenvironment, biomarkers, and immune checkpoint inhibitors are current and developing research areas. The keywords "immunotherapy" and "nanoparticles" are also likely to be new trends and focal points for future research. This study adopted bibliometric and visualization methods to provide a comprehensive review of the research on immunization in TNBC. This article will help researchers better understand the dynamic evolution of the role of immunity in TNBC and identify areas for future research.
Collapse
Affiliation(s)
- Xudong Li
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, Harbin, China
| | - Chun Lou
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, Harbin, China
| | - He Ren
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, Harbin, China
| | - Lina Cui
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, Harbin, China
| | - Kexin Chen
- Department of Pathology, Harbin Medical University Cancer Hospital, Harbin, China
| |
Collapse
|
2
|
Pöllinger B, Haiderali A, Huang M, Akyol Ersoy B, Abdelaziz AH, Kassem L, Elsisi GH. The cost-effectiveness of treatment for high-risk, early-stage, triple-negative breast cancer in Egypt: an analysis of neoadjuvant pembrolizumab plus chemotherapy followed by adjuvant single-agent pembrolizumab. J Med Econ 2025; 28:105-113. [PMID: 39665251 DOI: 10.1080/13696998.2024.2441073] [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: 11/02/2024] [Revised: 12/09/2024] [Accepted: 12/09/2024] [Indexed: 12/13/2024]
Abstract
OBJECTIVE The cost-effectiveness of neoadjuvant pembrolizumab + chemotherapy followed by adjuvant pembrolizumab compared to neoadjuvant chemotherapy plus placebo followed by adjuvant placebo was assessed in high-risk, early-stage, triple-negative breast cancer patients from an Egyptian societal perspective over a lifetime horizon. METHODS A 4-state Markov cohort model was developed to compare the cost-effectiveness of pembrolizumab + chemotherapy/pembrolizumab vs chemotherapy alone for the treatment of high-risk, early-stage, triple-negative breast cancer. The model simulated the clinical course of high-risk, early-stage, triple-negative breast cancer across four health states: event-free survival, locoregional recurrence, distant metastasis, and death. Clinical inputs for the simulation were derived from modeling of efficacy and safety data collected in the KEYNOTE-522 trial. Direct medical costs and indirect costs were reported in 2022 Egyptian pounds (EGP) and converted to US dollars ($). Probabilistic and deterministic sensitivity analyses were conducted to assess the robustness of model results. RESULTS Compared with chemotherapy alone, pembrolizumab + chemotherapy/pembrolizumab led to expected gains of 2.92 life years and 2.25 quality-adjusted life years, respectively, while increasing overall treatment costs by EGP 491,695 ($102,436). Incremental costs per year gained were EGP 218,285 ($45,476) per quality-adjusted life year and EGP 168,223 ($35,046) per life year, both of which were lower than the 2022 Egyptian cost-effectiveness threshold of EGP 398,439 ($83,008). The findings of sensitivity analyses indicated that the model was robust across a range of inputs and assumptions. CONCLUSIONS In Egypt, pembrolizumab + chemotherapy/pembrolizumab is a cost-effective treatment for high-risk, early-stage, triple-negative breast cancer when considering health-related quality-of-life and years of life gained.
Collapse
Affiliation(s)
- Bernadette Pöllinger
- Center for Outcomes Research and Health Economy, MSD Sharp & Dohme GmbH, Munich, Germany
| | - Amin Haiderali
- Center for Outcomes Research and Health Economy, Merck & Co., Inc, Rahway, NJ, USA
| | - Min Huang
- Center for Outcomes Research and Health Economy, Merck & Co., Inc, Rahway, NJ, USA
| | | | | | - Loay Kassem
- Faculty of Medicine, Cairo University, Giza, Egypt
| | - Gihan Hamdy Elsisi
- HTA Office, Cairo, Egypt
- Faculty of Economics, American University, Cairo, Egypt
| |
Collapse
|
3
|
Fu Y, Yang Q, Xu N, Zhang X. MiRNA affects the advancement of breast cancer by modulating the immune system's response. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167759. [PMID: 40037267 DOI: 10.1016/j.bbadis.2025.167759] [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: 10/29/2024] [Revised: 02/05/2025] [Accepted: 02/26/2025] [Indexed: 03/06/2025]
Abstract
Breast cancer (BC), which is the most common tumor in women, has greatly endangered women's lives and health. Currently, patients with BC receive comprehensive treatments, including surgery, chemotherapy, radiotherapy, endocrine therapy, and targeted therapy. According to the latest research, the development of BC is closely related to the inflammatory immune response, and the immunogenicity of BC has steadily been recognized. As such, immunotherapy is one of the promising and anticipated forms of treatment for BC. The potential values of miRNA in the diagnosis and prognosis of BC have been established, and aberrant expression of associated miRNA can either facilitate or inhibit progression of BC. In the tumor immune microenvironment (TME), miRNAs are considered to be an essential molecular mechanism by which tumor cells interact with immunocytes and immunologic factors. Aberrant expression of miRNAs results in reprogramming of tumor cells actively, which may suppress the generation and activation of immunocytes and immunologic factors, avoid tumor cells apoptosis, and ultimately result in uncontrolled proliferation and deterioration. Therefore, through activating and regulating the immunocytes related to tumors and associated immunologic factors, miRNA can contribute to the advancement of BC. In this review, we assessed the function of miRNA and associated immune system components in regulating the advancement of BC, as well as the potential and viability of using miRNA in immunotherapy for BC.
Collapse
Affiliation(s)
- Yeqin Fu
- Zhejiang cancer hospital, Hangzhou, Zhejiang 310022, China; Postgraduate Training Base Alliance of Wenzhou Medical University (Zhejiang Cancer Hospital), Hangzhou, Zhejiang 310022, China
| | - Qiuhui Yang
- The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Traditional Chinese Medicine), 310006, China
| | - Ning Xu
- Zhejiang cancer hospital, Hangzhou, Zhejiang 310022, China; School of Medicine, Shaoxing University, Shaoxing, Zhejiang 312000, China
| | - Xiping Zhang
- Zhejiang cancer hospital, Hangzhou, Zhejiang 310022, China.
| |
Collapse
|
4
|
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
|
5
|
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
|
6
|
Zhao Y, Pu C, Liu K, Liu Z. Targeting LAT1 with JPH203 to reduce TNBC proliferation and reshape suppressive immune microenvironment by blocking essential amino acid uptake. Amino Acids 2025; 57:27. [PMID: 40379991 PMCID: PMC12084285 DOI: 10.1007/s00726-025-03456-3] [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: 03/15/2025] [Accepted: 04/28/2025] [Indexed: 05/19/2025]
Abstract
The competitive uptake of essential amino acids (EAAs) by breast cancer cells is associated with poor patient prognosis and the development of an immunosuppressive tumor microenvironment. L-type amino acid transporters, LAT1 (SLC7 A5) and LAT2 (SLC7 A8) are major mediators of EAAs transmembrane uptake and are overexpressed in some tumor tissues. However, the distribution and functional roles of these transporters across breast cancer subtypes have not been fully elucidated. This study aims to investigate the therapeutic potential of targeting EAA transporters, particularly LAT1, in triple-negative breast cancer (TNBC) and its role in remodeling the tumor immune microenvironment. The distribution of EAA transporters across breast cancer subtypes was analyzed using multi-omics data. The effects of LAT1 targeting on TNBC cell proliferation and EAA uptake were evaluated using SLC7 A5 knockout and LAT1 inhibitors in vitro experiments. A 4T1-BALB/c tumor-bearing mouse model with normal immune function was constructed to investigate the effects of LAT1 targeting on tumor growth and immune microenvironment remodeling in vivo. TNBC demonstrated a strong dependence on LAT1-mediated EAAs uptake. Targeting LAT1 limited the exogenous supply of EAAs, leading to amino acid starvation, cell cycle arrest, and increased apoptosis in TNBC cells. The in vivo experiments, using a 4T1-BALB/c tumor-bearing mouse model, showed that LAT1 targeting inhibited tumor growth and remodeled the immunosuppressive tumor microenvironment. Targeting LAT1 improved PD-L1-associated immune suppression and improved the efficacy of PD-1 antibody treatment, producing synergistic anti-tumor effects. This study highlights the therapeutic potential of targeting LAT1 in TNBC, particularly in remodeling the tumor immune microenvironment. The findings provide a promising strategy for immune combination therapy in TNBC.
Collapse
Affiliation(s)
- Yajie Zhao
- Department of Breast Disease, Henan Breast Cancer Centre, The Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, Zhengzhou, China
| | - Chunrui Pu
- Department of Breast Disease, Henan Breast Cancer Centre, The Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, Zhengzhou, China
| | - Kangdong Liu
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China.
| | - Zhenzhen Liu
- Department of Breast Disease, Henan Breast Cancer Centre, The Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, Zhengzhou, China.
| |
Collapse
|
7
|
Wiertsema P, Tan YH, Haanen JBAG, Seijkens TTP, Jedema I. Advances in TIL therapy: Expanding the horizons beyond melanoma. MED 2025:100702. [PMID: 40381620 DOI: 10.1016/j.medj.2025.100702] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2025] [Revised: 04/08/2025] [Accepted: 04/22/2025] [Indexed: 05/20/2025]
Abstract
Tumor-infiltrating lymphocyte (TIL) therapy represents a breakthrough in solid tumor treatment, addressing unmet needs for patients with limited options. While its efficacy is established in advanced melanoma, TIL therapy shows early promise in non-small cell lung cancer, breast cancer, gynecological cancers, and head and neck cancers. However, challenges such as reduced T cell infiltration, lower tumor mutational burden (TMB), immunosuppressive tumor microenvironments (TME), and toxicity associated with the TIL therapy regimen hinder its broader application in these patient groups, compared with melanoma. To address these challenges, new approaches focus on the selection of tumor-reactive TIL, optimization of TIL expansion, combination of immune checkpoint inhibitors with TIL therapy to counteract immunosuppressive microenvironments, and genetic modification of TIL to enhance persistence and functionality. Larger clinical trials are essential to validate these innovations and standardize protocols. With continued advancements, TIL therapy has the potential to redefine the treatment landscape for advanced solid cancers.
Collapse
Affiliation(s)
- Pauline Wiertsema
- Division of Molecular Oncology and Immunology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Ya Hwee Tan
- Division of Medical Oncology, National Cancer Centre Singapore, Singapore, Singapore; Division of Medical Oncology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - John B A G Haanen
- Division of Molecular Oncology and Immunology, The Netherlands Cancer Institute, Amsterdam, the Netherlands; Division of Medical Oncology, The Netherlands Cancer Institute, Amsterdam, the Netherlands; Department of Medical Oncology, Leiden University Medical Center, Leiden, the Netherlands; Melanoma Clinic, Centre Hospitalier Universitaire Vaudois, Lausanne, Switzerland
| | - Tom T P Seijkens
- Division of Medical Oncology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Inge Jedema
- Division of Molecular Oncology and Immunology, The Netherlands Cancer Institute, Amsterdam, the Netherlands.
| |
Collapse
|
8
|
Liu S, Chen H, Gagea M, Federico L, Zhang F, Gomez J, Do KA, Symmans WF, Hortobagyi GN, Mills GB, Gonzalez-Angulo AM, Tripathy D. ADAMs contribute to triple negative breast cancer via mTORC1 pathway: targeting ADAM-mTOR axis improves efficacy. Cancer Lett 2025; 626:217775. [PMID: 40339955 DOI: 10.1016/j.canlet.2025.217775] [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: 12/09/2024] [Revised: 04/28/2025] [Accepted: 05/05/2025] [Indexed: 05/10/2025]
Abstract
Breast cancer is the most frequently diagnosed cancer globally and the second leading cause of cancer-related deaths in American women. Triple-negative breast cancer (TNBC) lacks estrogen receptor, progesterone receptor, and human epidermal growth factor receptor 2. Thus, fewer targeting therapies are available for this most aggressive subtype. The A Disintegrin and Metalloproteinase (ADAM) family plays a vital role in cancer pathophysiology. Previous studies focused on single ADAM members. However, none of these have entered into the clinical arena as diagnostics or therapeutics for breast cancer. In this study, we demonstrate the upregulation of a panel of ADAM members in TNBC, and overexpression of all the individual ADAMs tested are correlated with poor patient survival, making it unlikely that targeting a single ADAM member would be effective. Reverse-phase protein array and multiplexed immunofluorescence revealed that ADAM10/15/17 expression was associated with activated mTOR signaling. Individual knockdown of ADAM10, ADAM15, or ADAM17 modestly reduced mTOR signaling, cellular proliferation and survival. However, the concurrent knockdown of the three ADAMs drastically decreased mTOR signaling and cellular aggressiveness. Consistently, combined targeting of ADAMs and mTOR increased inhibitory efficacy compared to monotherapy in ADAM-mTOR-activated tumor growth and invasion in vitro and in immunodeficient and immunocompetent mice. These results establish a functional link between ADAMs and activation of mTOR signaling, suggesting the ADAM-mTOR axis as a therapeutic target and biomarker for ADAM-enriched TNBC and, potentially, other tumor lineages with high ADAM activity.
Collapse
Affiliation(s)
| | - Huiqin Chen
- Department of Breast Medical Oncology, USA; Department of of Biostatistics, USA
| | - Mihai Gagea
- Department of Veterinary Medicine and Surgery, USA
| | | | | | | | | | - William F Symmans
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | | | | | | | | |
Collapse
|
9
|
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
|
10
|
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
|
11
|
Zhao Q, Pramanik J, Lu Y, Homer NZM, Imianowski CJ, Zhang B, Iqbal M, Shaji SK, Morris AC, Roychoudhuri R, Okkenhaug K, Qiu P, Mahata B. Perturbing local steroidogenesis to improve breast cancer immunity. Nat Commun 2025; 16:3945. [PMID: 40287432 PMCID: PMC12033260 DOI: 10.1038/s41467-025-59356-3] [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: 02/14/2024] [Accepted: 04/16/2025] [Indexed: 04/29/2025] Open
Abstract
Breast cancer, particularly triple-negative breast cancer (TNBC), evades the body's immune defences, in part by cultivating an immunosuppressive tumour microenvironment. Here, we show that suppressing local steroidogenesis can augment anti-tumour immunity against TNBC. Through targeted metabolomics of steroids coupled with immunohistochemistry, we profiled the existence of immunosuppressive steroids in TNBC patient tumours and discerned the steroidogenic activity in immune-infiltrating regions. In mouse, genetic inhibition of immune cell steroidogenesis restricted TNBC tumour progression with a significant reduction in immunosuppressive components such as tumour associated macrophages. Steroidogenesis inhibition appears to bolster anti-tumour immune responses in dendritic and T cells by impeding glucocorticoid signalling. Undertaking metabolic modelling of the single-cell transcriptomics and targeted tumour-steroidomics, we pinpointed the predominant steroidogenic cells. Inhibiting steroidogenesis pharmacologically using a identified drug, posaconazole, curtailed tumour expansion in a humanised TNBC mouse model. This investigation paves the way for targeting steroidogenesis and its signalling pathways in breast cancer affected by immune-steroid maladaptation.
Collapse
Affiliation(s)
- Qiuchen Zhao
- Department of Pathology, University of Cambridge, Cambridge, CB2 1QP, UK
- Cancer Research UK Cambridge Centre and Department of Oncology, University of Cambridge, Cambridge, CB2 0XZ, UK
| | - Jhuma Pramanik
- Department of Pathology, University of Cambridge, Cambridge, CB2 1QP, UK
| | - Yongjin Lu
- Breast Cancer Center, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, 250117, China
| | - Natalie Z M Homer
- Mass Spectrometry Core, Edinburgh Clinical Research Facility, Queens Medical Research Institute, University of Edinburgh, Edinburgh, UK
| | | | - Baojie Zhang
- Department of Pathology, University of Cambridge, Cambridge, CB2 1QP, UK
| | - Muhammad Iqbal
- Department of Pathology, University of Cambridge, Cambridge, CB2 1QP, UK
| | | | | | - Rahul Roychoudhuri
- Department of Pathology, University of Cambridge, Cambridge, CB2 1QP, UK
| | - Klaus Okkenhaug
- Department of Pathology, University of Cambridge, Cambridge, CB2 1QP, UK
| | - Pengfei Qiu
- Breast Cancer Center, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, 250117, China.
- The Precision Breast Cancer Institute, Addenbrookes Hospital, Department of Oncology, University of Cambridge, Cambridge, CB2 0QQ, UK.
| | - Bidesh Mahata
- Department of Pathology, University of Cambridge, Cambridge, CB2 1QP, UK.
| |
Collapse
|
12
|
Imani S, Farghadani R, Roozitalab G, Maghsoudloo M, Emadi M, Moradi A, Abedi B, Jabbarzadeh Kaboli P. Reprogramming the breast tumor immune microenvironment: cold-to-hot transition for enhanced immunotherapy. J Exp Clin Cancer Res 2025; 44:131. [PMID: 40281554 PMCID: PMC12032666 DOI: 10.1186/s13046-025-03394-8] [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: 01/16/2025] [Accepted: 04/14/2025] [Indexed: 04/29/2025] Open
Abstract
This review discusses reprogramming the breast tumor immune microenvironment from an immunosuppressive cold state to an immunologically active hot state. A complex interplay is revealed, in which the accumulation of metabolic byproducts-such as lactate, reactive oxygen species (ROS), and ammonia-is shown to impair T-cell function and promote tumor immune escape. It is demonstrated that the tumor microenvironment (TME) is dominated by immunosuppressive cytokines, including interleukin-10 (IL-10), transforming growth factorβ (TGFβ), and IL-35. Notably, IL-35 is produced by regulatory T cells and breast cancer cells. The conversion of conventional T cells into IL-35-producing induced regulatory T cells, along with the inhibition of pro-inflammatory cytokine secretion, contributes to the suppression of anti-tumor immunity. It is further demonstrated that key immune checkpoint molecules-such as PD-1, PDL1, CTLA-4, TIM-3, LAG-3, and TIGIT-are upregulated within the TME, leading to Tcell exhaustion and diminished immune responses. The blockade of these checkpoints is shown to restore T-cell functionality and is proposed as a strategy to convert cold tumors into hot ones with robust effector cell infiltration. The therapeutic potential of chimeric antigen receptor (CAR)T cell therapy is also explored, and targeting specific tumor-associated antigens, such as glycoproteins and receptor tyrosine kinases, is highlighted. It is suggested that CART cell efficacy can be enhanced by combining these cells with immune checkpoint inhibitors and other immunomodulatory agents, thereby overcoming the barriers imposed by the immunosuppressive TME. Moreover, the role of the microbiome in regulating estrogen metabolism and systemic inflammation is reviewed. Alterations in the gut microbiota are shown to affect the TME, and microbiome-based interventions are proposed as an additional means to facilitate the cold-to-hot transition. It is concluded that by targeting the metabolic and immunological pathways that underpin immune suppression-through combination strategies involving checkpoint blockade, CART cell therapies, and microbiome modulation-the conversion of the breast TME from cold to hot can be achieved. This reprogramming is anticipated to enhance immune cell infiltration and function, thereby improving the overall efficacy of immunotherapies and leading to better clinical outcomes for breast cancer patients.
Collapse
Affiliation(s)
- Saber Imani
- Shulan International Medical College, Zhejiang Shuren University, Hangzhou, Zhejiang, China.
| | - Reyhaneh Farghadani
- Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Jalan Lagoon Selatan, Bandar Sunway, Subang Jaya, 47500, Selangor Darul Ehsan, Malaysia
| | - Ghazaal Roozitalab
- Noncommunicable Diseases Research Center, Fasa University of Medical Sciences, Fasa, Iran
| | - Mazaher Maghsoudloo
- Key Laboratory of Epigenetics and Oncology, The Research Center for Preclinical Medicine, Southwest Medical University, Luzhou, 646000, Sichuan, China
| | - Mahdieh Emadi
- Department of Biology, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Atefeh Moradi
- Department of Life Sciences and System Biology, University of Turin, Turin, Italy
| | - Behnaz Abedi
- Department of Basic Sciences, Faculty of Veterinary Medicine, University of Tabriz, Tabriz, Iran
| | - Parham Jabbarzadeh Kaboli
- Department of Biochemistry, Faculty of Medicine, Medical University of Warsaw, Warsaw, 02-091, Poland.
| |
Collapse
|
13
|
Mariano NC, Marotti JD, Chen Y, Karakyriakou B, Salgado R, Christensen BC, Miller TW, Kettenbach AN. Quantitative proteomics analysis of triple-negative breast cancers. NPJ Precis Oncol 2025; 9:117. [PMID: 40269124 PMCID: PMC12019170 DOI: 10.1038/s41698-025-00907-8] [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: 09/11/2024] [Accepted: 04/05/2025] [Indexed: 04/25/2025] Open
Abstract
Triple-negative breast cancer (TNBC) accounts for approximately 15% of all Breast Cancer (BC) cases with poorer prognosis and clinical outcomes compared to other BC subtypes due to greater tumor heterogeneity and few therapeutically targetable oncogenic drivers. To reveal actionable pathways for anti-cancer treatment, we use a proteomic approach to quantitatively compare the abundances of 6306 proteins across 55 formalin-fixed and paraffin-embedded (FFPE) TNBC tumors. We identified four major TNBC clusters by unsupervised clustering analysis of protein abundances. Analyses of clinicopathological characteristics revealed associations between the proteomic profiles and clinical phenotypes exhibited by each subtype. We validate the findings by inferring immune and stromal cell type composition from genome-wide DNA methylation profiles. Finally, quantitative proteomics on TNBC cell lines was conducted to identify in vitro models for each subtype. Collectively, our data provide subtype-specific insights into molecular drivers, clinicopathological phenotypes, tumor microenvironment (TME) compositions, and potential pharmacologic vulnerabilities for further investigations.
Collapse
Affiliation(s)
| | - Jonathan D Marotti
- Department of Pathology and Laboratory Medicine, Lebanon, NH, USA
- Dartmouth Cancer Center, Lebanon, NH, USA
| | | | | | - Roberto Salgado
- Department of Pathology, GZA-ZNA Hospitals, Antwerp, Belgium
- Division of Research, Peter MacCallum Cancer Centre, Melbourne, Australia
| | - Brock C Christensen
- Department of Pathology and Laboratory Medicine, Lebanon, NH, USA
- Dartmouth Cancer Center, Lebanon, NH, USA
- Department of Molecular and Systems Biology, Lebanon, NH, USA
- Department of Epidemiology, Lebanon, NH, USA
- Department of Community and Family Medicine, Lebanon, NH, USA
| | - Todd W Miller
- Dartmouth Cancer Center, Lebanon, NH, USA
- Department of Molecular and Systems Biology, Lebanon, NH, USA
- Department of Pharmacology & Toxicology, Medical College of Wisconsin, Milwaukee, WI, USA
- Department of Pathology, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Arminja N Kettenbach
- Department of Biochemistry and Cell Biology, Hanover, NH, USA.
- Dartmouth Cancer Center, Lebanon, NH, USA.
| |
Collapse
|
14
|
Emara HM, Allam NK, Youness RA. A comprehensive review on targeted therapies for triple negative breast cancer: an evidence-based treatment guideline. Discov Oncol 2025; 16:547. [PMID: 40244488 PMCID: PMC12006628 DOI: 10.1007/s12672-025-02227-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/07/2024] [Accepted: 03/25/2025] [Indexed: 04/18/2025] Open
Abstract
Triple-negative breast cancer (TNBC) is an aggressive malignancy characterized by limited therapeutic options and poor prognosis. Despite advancements in precision oncology, conventional chemotherapy remains the cornerstone of TNBC treatment, often accompanied by debilitating side effects and suboptimal outcomes. This review presents a comprehensive analysis of clinical trials on targeted therapies, aiming to establish a novel, evidence-based treatment strategy exclusively leveraging molecularly targeted agents. By integrating patient-specific genetic profiles with therapeutic responses observed across various clinical trial phases, this approach seeks to optimize efficacy while minimizing toxicity. The proposed targeted therapy combinations hold significant potential to revolutionize TNBC treatment, offering a paradigm shift toward precision medicine and improved patient outcomes.
Collapse
Affiliation(s)
- Hadir M Emara
- Nanotechnology Program, School of Sciences & Engineering, The American University in Cairo, New Cairo, 11835, Egypt.
| | - Nageh K Allam
- Nanotechnology Program, School of Sciences & Engineering, The American University in Cairo, New Cairo, 11835, Egypt.
- Energy Materials Laboratory, Physics Department, School of Sciences & Engineering, The American University in Cairo, New Cairo, 11835, Egypt.
| | - Rana A Youness
- Department of Molecular Biology and Biochemistry, Faculty of Biotechnology, German International University, New Administrative Capital, Cairo, Egypt.
| |
Collapse
|
15
|
Dos Santos ALS, Da Silva JL, De Albuquerque LZ, Neto ALA, Da Silva CF, Cerva LAM, Small IA, Rodrigues FR, De Macedo FC, Marcelino CP, Batista PDM, Rego MADC, Borba MACSM, De Melo AC. Unveiling the Landscape of PD-L1 Expression and Tumor-Infiltrating Lymphocyte Subtypes in Advanced Triple-Negative Breast Cancer in Brazil. BREAST CANCER (DOVE MEDICAL PRESS) 2025; 17:349-358. [PMID: 40256247 PMCID: PMC12009053 DOI: 10.2147/bctt.s499373] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/05/2024] [Accepted: 03/12/2025] [Indexed: 04/22/2025]
Abstract
Purpose This study aimed to assess the frequency and prognostic significance of programmed cell death ligand 1 (PD-L1) expression and tumor-infiltrating lymphocyte (TIL) subtypes in advanced triple-negative breast cancer (TNBC). Patients and Methods A database search was conducted to identify women with previously untreated locally recurrent inoperable or metastatic TNBC treated between January 2018 and December 2022. The inclusion criteria required formalin-fixed paraffin-embedded samples aged less than four years. PD-L1 expression was evaluated using the PD-L1 IHC 22C3 pharmDx assay, and the combined positive score (CPS) was calculated. TIL subtypes were assessed using immunohistochemical staining. Results The study included 150 patients, with a median age of 51.5 years. The majority of patients were younger than 65 years, postmenopausal, non-white, and had metastatic TNBC. CPS≥10 was observed in 20.9% of cases, mainly in postmenopausal women. No significant differences were found in demographic characteristics and clinicopathological variables across PD-L1 subgroups. Tumors with PD-L1 CPS≥10 had higher expression of CD3+, CD4+, and CD8+ TIL subtypes. Most patients received first-line chemotherapy, with smaller proportions undergoing second, third, and fourth-line treatments. No statistically significant differences were observed in median progression-free survival (PFS) or overall survival (OS) across PD-L1 subgroups in this cohort of chemotherapy-treated patients. Conclusion This study provides insights into the expression profiles of PD-L1 and TIL subtypes in advanced TNBC. The PD-L1 CPS status did not significantly affect survival outcomes, but variations in TIL subtype composition were observed based on PD-L1 CPS status.
Collapse
Affiliation(s)
| | - Jesse Lopes Da Silva
- Division of Clinical Research and Technological Development, Brazilian National Cancer Institute, Rio de Janeiro, Brazil
| | - Lucas Zanetti De Albuquerque
- Division of Clinical Research and Technological Development, Brazilian National Cancer Institute, Rio de Janeiro, Brazil
| | - Antônio Lucas Araújo Neto
- Division of Clinical Research and Technological Development, Brazilian National Cancer Institute, Rio de Janeiro, Brazil
| | - Cecília Ferreira Da Silva
- Division of Clinical Research and Technological Development, Brazilian National Cancer Institute, Rio de Janeiro, Brazil
| | - Luana Aguiar Mesquita Cerva
- Division of Clinical Research and Technological Development, Brazilian National Cancer Institute, Rio de Janeiro, Brazil
| | - Isabele Avila Small
- Division of Clinical Research and Technological Development, Brazilian National Cancer Institute, Rio de Janeiro, Brazil
| | | | | | | | | | | | | | - Andreia Cristina De Melo
- Division of Clinical Research and Technological Development, Brazilian National Cancer Institute, Rio de Janeiro, Brazil
| |
Collapse
|
16
|
Liu Z, Mao Y, Wang S, Zheng H, Yang K, Yang L, Huang P. A bibliometric and visual analysis of the impact of senescence on tumor immunotherapy. Front Immunol 2025; 16:1566227. [PMID: 40292294 PMCID: PMC12021824 DOI: 10.3389/fimmu.2025.1566227] [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: 01/24/2025] [Accepted: 03/24/2025] [Indexed: 04/30/2025] Open
Abstract
Background Recently, many studies have focused on the relationship between senescence and immunotherapy in cancer treatment. However, relatively few studies have examined the intrinsic links between the three. Whether these studies can act synergistically in the fight against cancer and the specific links between them are still unclear. Methods We extracted, quantified, and visualized data from the literature (n = 2396) for the period 2004-2023 after rigorous quality control using citespace, GraphPad Prism, the R software package, and VOSviewer. Results Linear fit analyses were generated to predict the number of annual publications and citations as a function of the top-performing authors, journals, countries, and affiliations academically over the past two decades such as Weiwei, Aging-us, China, and the UT MD Anderson Cancer Center. Vosviewer-based hierarchical clustering further categorized study characteristics into six clusters, including two major clusters of immunotherapy research, immunosenescence-related research factors, and timeline distributions suggesting that cellular senescence and tumor progression is a relatively new research cluster that warrants further exploration and development. Study characterization bursts and linear regression analyses further confirmed these findings and revealed other important results, such as aging (a = 1.964, R² = 0.6803) and immunotherapy (a = 16.38, R² = 0.8812). Furthermore, gene frequency analysis in this study revealed the most abundant gene, APOE, and SIRT1-7 proteins. Conclusion The combination of aging therapies with tumor immunotherapies is currently in its preliminary stages. Although senescence has the greatest impact on ICB therapies, mechanistic investigations, and drug development for APOE and sirt1-7 (Sirtuins family) targets may be the key to combining senescence therapies with immunotherapies in the treatment of tumors.
Collapse
Affiliation(s)
- Zixu Liu
- Center for Evidence-Based Medicine, School of Public Health, Jiangxi Medical College. Nanchang University, Nanchang, China
- Jiangxi Province Key Laboratory of Preventive Medicine, Jiangxi Medical College, Nanchang University, Nanchang, China
- First Clinical Medicine School, Nanchang University, Nanchang, China
| | - Yuchen Mao
- First Clinical Medicine School, Nanchang University, Nanchang, China
| | - Shukai Wang
- First Clinical Medicine School, Nanchang University, Nanchang, China
| | - Haoyu Zheng
- School of Stomatology, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Kangping Yang
- Department of Gastroenterological Surgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Liang Yang
- Department of Gastroenterological Surgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Peng Huang
- Center for Evidence-Based Medicine, School of Public Health, Jiangxi Medical College. Nanchang University, Nanchang, China
- Jiangxi Province Key Laboratory of Preventive Medicine, Jiangxi Medical College, Nanchang University, Nanchang, China
| |
Collapse
|
17
|
Li H, Chang Y, Jin T, Zhang M. Progress of PD-1/PD-L1 immune checkpoint inhibitors in the treatment of triple-negative breast cancer. Cancer Cell Int 2025; 25:139. [PMID: 40211301 PMCID: PMC11987362 DOI: 10.1186/s12935-025-03769-z] [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: 10/05/2024] [Accepted: 03/28/2025] [Indexed: 04/13/2025] Open
Abstract
Triple-negative breast cancer (TNBC) is a highly heterogeneous cancer with substantial recurrence potential. Currently, surgery and chemotherapy are the main treatments for this disease. However, chemotherapy is often limited by several factors, including low bioavailability, significant systemic toxicity, inadequate targeting, and multidrug resistance. Immune checkpoint inhibitors (ICIs), including those targeting programmed death protein-1 (PD-1) and its ligand (PD-L1), have been proven effective in the treatment of various tumours. In particular, in the treatment of TNBC with PD-1/PD-L1 inhibitors, both monotherapy and combination chemotherapy, as well as targeted drugs and other therapeutic strategies, have broad therapeutic prospects. In addition, these inhibitors can participate in the tumour immune microenvironment (TIME) through blocking PD-1/PD-L1 binding, which can improve immune efficacy. This article provides an overview of the use of PD-1/PD-L1 inhibitors in the treatment of TNBC and the progress of multiple therapeutic studies. To increase the survival of TNBC patients, relevant biomarkers for predicting the efficacy of PD-1/PD-L1 inhibitor therapy have been explored to identify new strategies for the treatment of TNBC.
Collapse
Affiliation(s)
- Hongshu Li
- Department of Pathology and Cancer Research Center, Yanbian University Medical College, Gong Yuan Road No. 977, Yanji, 133002, P. R. China
- Key Laboratory of the Science and Technology Department of Jilin Province, Yanji, P. R. China
| | - Ying Chang
- Department of Pathology and Cancer Research Center, Yanbian University Medical College, Gong Yuan Road No. 977, Yanji, 133002, P. R. China
- Key Laboratory of the Science and Technology Department of Jilin Province, Yanji, P. R. China
| | - Tiefeng Jin
- Department of Pathology and Cancer Research Center, Yanbian University Medical College, Gong Yuan Road No. 977, Yanji, 133002, P. R. China.
- Key Laboratory of the Science and Technology Department of Jilin Province, Yanji, P. R. China.
| | - Meihua Zhang
- Department of Ultrasound Medicine, Yanbian University Hospital, Yanji, 133000, P. R. China.
- Department of Pathology and Cancer Research Center, Yanbian University Medical College, Gong Yuan Road No. 977, Yanji, 133002, P. R. China.
- Key Laboratory of the Science and Technology Department of Jilin Province, Yanji, P. R. China.
| |
Collapse
|
18
|
Zhang Y, Li Q, Lan J, Xie G, Zhang G, Cui J, Leng P, Wang Y. Triple-negative breast cancer molecular subtypes and potential detection targets for biological therapy indications. Carcinogenesis 2025; 46:bgaf006. [PMID: 39977309 DOI: 10.1093/carcin/bgaf006] [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: 10/12/2024] [Revised: 02/07/2025] [Accepted: 02/17/2025] [Indexed: 02/22/2025] Open
Abstract
Triple-negative breast cancer (TNBC) is a highly aggressive subtype of breast cancer associated with poor prognosis. While chemotherapy remains the conventional treatment approach, its efficacy is limited and often accompanied by significant toxicity. Advances in precision-targeted therapies have expanded treatment options for TNBC, including immunotherapy, poly (ADP-ribose) polymerase inhibitors, androgen receptor inhibitors, cell cycle-dependent kinase inhibitors, and signaling pathway inhibitors. However, the heterogeneous nature of TNBC contributes to variations in treatment outcomes, underscoring the importance of identifying intrinsic molecular subtypes for personalized therapy. Additionally, due to patient-specific variability, the therapeutic response to targeted treatments is inconsistent. This highlights the need to strategize patients based on potential therapeutic targets for targeted drugs to optimize treatment strategies. This review summarizes the classification strategies and immunohistochemical (IHC) biomarkers for TNBC subtypes, along with potential targets for identifying indications for targeted drug therapy. These insights aim to support the development of personalized treatment approaches for TNBC patients.
Collapse
Affiliation(s)
- Yanchuan Zhang
- Chongqing Key Laboratory of Sichuan-Chongqing Co-construction for Diagnosis and Treatment of Infectious Diseases Integrated Traditional Chinese and Western Medicine, Chengdu, China
- Sichuan Key Laboratory of Medical Molecular Testing, College of Medical Technology, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Qinghua Li
- Chongqing Key Laboratory of Sichuan-Chongqing Co-construction for Diagnosis and Treatment of Infectious Diseases Integrated Traditional Chinese and Western Medicine, Chengdu, China
- Sichuan Key Laboratory of Medical Molecular Testing, College of Medical Technology, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Jie Lan
- Division of Head & Neck Tumor Multimodality Treatment, Cancer Center, Institute of Breast Health Medicine, West China Hospital, Sichuan University, Chengdu, P.R. China
| | - Guojing Xie
- Chongqing Key Laboratory of Sichuan-Chongqing Co-construction for Diagnosis and Treatment of Infectious Diseases Integrated Traditional Chinese and Western Medicine, Chengdu, China
- Sichuan Key Laboratory of Medical Molecular Testing, College of Medical Technology, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Guangjie Zhang
- Department of Clinical Laboratory, Chengdu Fifth People's Hospital, Chengdu, China
| | - Junhao Cui
- Chongqing Key Laboratory of Sichuan-Chongqing Co-construction for Diagnosis and Treatment of Infectious Diseases Integrated Traditional Chinese and Western Medicine, Chengdu, China
| | - Ping Leng
- Chongqing Key Laboratory of Sichuan-Chongqing Co-construction for Diagnosis and Treatment of Infectious Diseases Integrated Traditional Chinese and Western Medicine, Chengdu, China
- Sichuan Key Laboratory of Medical Molecular Testing, College of Medical Technology, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yingshuang Wang
- Chongqing Key Laboratory of Sichuan-Chongqing Co-construction for Diagnosis and Treatment of Infectious Diseases Integrated Traditional Chinese and Western Medicine, Chengdu, China
- Sichuan Key Laboratory of Medical Molecular Testing, College of Medical Technology, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|
19
|
Khairi S, Wang CY, Anuraga G, Prayugo FB, Ansar M, Lesmana MHS, Irham LM, Shen CY, Chung MH. Integrative Analysis of DNA Methylation and microRNA Reveals GNPDA1 and SLC25A16 Related to Biopsychosocial Factors Among Taiwanese Women with a Family History of Breast Cancer. J Pers Med 2025; 15:134. [PMID: 40278313 PMCID: PMC12028518 DOI: 10.3390/jpm15040134] [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: 01/24/2025] [Revised: 03/05/2025] [Accepted: 03/27/2025] [Indexed: 04/26/2025] Open
Abstract
Biopsychosocial factors, including family history, influence the development of breast cancer. Malignancies in women with a family history of breast cancer may be detectable based on DNA methylation and microRNA. Objectives: The present study extended an integrative analysis of DNA methylation and microRNA to identify genes associated with biopsychosocial factors. Methods: We identified 3060 healthy women from the Taiwan Biobank and included 32 blood plasma samples for analysis of biopsychosocial factors and epigenetic changes. GEO databases and bioinformatics approaches were used for the identification and validation of potential genes. Results: Our integrative analysis revealed GNPDA1 and SLC25A16 as potential genes. Age, a family history of cancer, and alcohol consumption were associated with GNPDA1 and SLC25A16 based on the current data set and the GEO data set. GNPDA1 and SLC25A16 exhibited significant expression in breast cancer tissues based on UALCAN analysis, where they were overexpressed and underexpressed, respectively. Through a MethSurv analysis, GNPDA1 hypomethylation and SLC25A16 hypermethylation were associated with poor prognoses in terms of overall survival in breast cancer. Moreover, through a MetaCore functional enrichment analysis, GNPDA1 and SLC25A16 were associated with the BRCA1, BRCA2, and pro-oncogenic actions of the androgen receptor in breast cancer. Further, GNPDA1 and SLC25A16 were enriched in known targets of approved cancer drugs as potential genes associated with breast cancer. Conclusions: These two genes might serve as biomarkers for the early detection of breast cancer, especially for women with a family history of breast cancer.
Collapse
Affiliation(s)
- Sabiah Khairi
- School of Nursing, College of Nursing, Taipei Medical University, Taipei City 11031, Taiwan;
| | - Chih-Yang Wang
- Ph.D. Program for Cancer Molecular Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei City 11031, Taiwan;
- Graduate Institute of Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei City 11031, Taiwan
| | - Gangga Anuraga
- Department of Statistics, Faculty of Science and Technology, Universitas PGRI Adi Buana, Surabaya 60234, Indonesia;
| | - Fidelia Berenice Prayugo
- Chang Gung Medical Education Research Centre (CG-MERC), Chang Gung Memorial Hospital, Taoyuan City 33302, Taiwan;
- School of Medicine, Chang Gung University, Taoyuan City 33302, Taiwan
| | - Muhamad Ansar
- Ph.D. Program in the Clinical Drug Development of Herbal Medicine, Taipei Medical University, Taipei City 110301, Taiwan;
| | - Mohammad Hendra Setia Lesmana
- Department of Mental Health and Community, Faculty of Medicine, Public Health, and Nursing, Universitas Gadjah Mada, Yogyakarta 55281, Indonesia;
| | | | - Chen-Yang Shen
- Institute of Biomedical Sciences, Academia Sinica, Taipei City 11529, Taiwan
- Master Program in Clinical Genomics and Proteomics, School of Pharmacy, Taipei Medical University, Taipei City 11031, Taiwan
- College of Public Health, China Medical University, Taichung City 406040, Taiwan
| | - Min-Huey Chung
- School of Nursing, College of Nursing, Taipei Medical University, Taipei City 11031, Taiwan;
- Department of Nursing, Shuang Ho Hospital, Taipei Medical University, New Taipei City 23561, Taiwan
| |
Collapse
|
20
|
Jiang R, Yang L, Liu X, Xu Y, Han L, Chen Y, Gao G, Wang M, Su T, Li H, Fang L, Sun N, Du H, Zheng J, Wang G. Genetically engineered macrophages reverse the immunosuppressive tumor microenvironment and improve immunotherapeutic efficacy in TNBC. Mol Ther 2025:S1525-0016(25)00198-4. [PMID: 40119517 DOI: 10.1016/j.ymthe.2025.03.024] [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: 09/20/2024] [Revised: 01/21/2025] [Accepted: 03/17/2025] [Indexed: 03/24/2025] Open
Abstract
The main challenges in current immunotherapy for triple-negative breast cancer (TNBC) lie in the immunosuppressive tumor microenvironment (TME). Considering tumor-associated macrophages (TAMs) are the most abundant immune cells in the TME, resetting TAMs is a promising strategy for ameliorating the immunosuppressive TME. Here, we developed genetically engineered macrophages (GEMs) with gene-carrying adenoviruses, to maintain the M1-like phenotype and directly deliver the immune regulators interleukin-12 and CXCL9 into local tumors, thereby reversing the immunosuppressive TME. In tumor-bearing mice, GEMs demonstrated targeted enrichment in tumors and successfully reprogramed TAMs to M1-like macrophages. Moreover, GEMs significantly enhanced the accumulation, proliferation, and activation of CD8+ T cells, mature dendritic cells, and natural killer cells within tumors, while diminishing M2-like macrophages, immunosuppressive myeloid-derived suppressor cells, and regulatory T cells. This treatment efficiently suppressed tumor growth. In addition, combination therapy with GEMs and anti-programmed cell death protein 1 further improved interferon-γ+CD8+ T cell percentages and tumor inhibition efficacy in an orthotopic murine TNBC model. Therefore, this study provides a novel strategy for reversing the immunosuppressive TME and improving immunotherapeutic efficacy through live macrophage-mediated gene delivery.
Collapse
Affiliation(s)
- Ranran Jiang
- Department of Oncology, Nanjing Medical University, 101 Longmian Avenue, Nanjing, Jiangsu 211166, China; Cancer Institute, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu 221004, China; Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, 99 Huaihai Road, Xuzhou, Jiangsu 221002, China; Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu 221004, China; Department of Oncology, The Affiliated Hospital of Xuzhou Medical University, 99 Huaihai Road, Xuzhou, Jiangsu 221002, China
| | - Liechi Yang
- Department of Neurosurgery, The Affiliated Hospital of Xuzhou Medical University, 99 Huaihai Road, Xuzhou, Jiangsu 221002, China
| | - Xin Liu
- Cancer Institute, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu 221004, China; Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, 99 Huaihai Road, Xuzhou, Jiangsu 221002, China; Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu 221004, China; Department of Urology, The Affiliated Hospital of Xuzhou Medical University, 99 Huaihai Road, Xuzhou, Jiangsu 221002, China
| | - Yujun Xu
- Cancer Institute, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu 221004, China; Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, 99 Huaihai Road, Xuzhou, Jiangsu 221002, China; Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu 221004, China
| | - Lulu Han
- Cancer Institute, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu 221004, China; Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, 99 Huaihai Road, Xuzhou, Jiangsu 221002, China; Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu 221004, China
| | - Yuxin Chen
- Cancer Institute, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu 221004, China; Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, 99 Huaihai Road, Xuzhou, Jiangsu 221002, China; Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu 221004, China
| | - Ge Gao
- Cancer Institute, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu 221004, China; Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, 99 Huaihai Road, Xuzhou, Jiangsu 221002, China; Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu 221004, China
| | - Meng Wang
- Cancer Institute, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu 221004, China; Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, 99 Huaihai Road, Xuzhou, Jiangsu 221002, China; Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu 221004, China
| | - Tong Su
- Cancer Institute, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu 221004, China; Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, 99 Huaihai Road, Xuzhou, Jiangsu 221002, China; Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu 221004, China
| | - Huizhong Li
- Cancer Institute, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu 221004, China; Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, 99 Huaihai Road, Xuzhou, Jiangsu 221002, China; Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu 221004, China
| | - Lin Fang
- Cancer Institute, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu 221004, China; Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, 99 Huaihai Road, Xuzhou, Jiangsu 221002, China; Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu 221004, China
| | - Nan Sun
- Cancer Institute, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu 221004, China; Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, 99 Huaihai Road, Xuzhou, Jiangsu 221002, China; Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu 221004, China
| | - Hongwei Du
- Cancer Institute, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu 221004, China; Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, 99 Huaihai Road, Xuzhou, Jiangsu 221002, China; Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu 221004, China
| | - Junnian Zheng
- Department of Oncology, Nanjing Medical University, 101 Longmian Avenue, Nanjing, Jiangsu 211166, China; Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, 99 Huaihai Road, Xuzhou, Jiangsu 221002, China; Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu 221004, China.
| | - Gang Wang
- Cancer Institute, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu 221004, China; Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, 99 Huaihai Road, Xuzhou, Jiangsu 221002, China; Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu 221004, China.
| |
Collapse
|
21
|
Al-Sharabass EA, El-Houseini ME, Effat H, Ibrahim SA, Abdellateif MS. The clinical potential of PDL-1 pathway and some related micro-RNAs as promising diagnostic markers for breast cancer. Mol Med 2025; 31:106. [PMID: 40108523 PMCID: PMC11921724 DOI: 10.1186/s10020-025-01137-1] [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: 07/31/2024] [Accepted: 02/19/2025] [Indexed: 03/22/2025] Open
Abstract
BACKGROUND Immune checkpoint pathways play important roles in breast cancer (BC) pathogenesis and therapy. METHODS Expression levels of programmed cell death protein 1 (PD-1), cytotoxic T-lymphocyte-associated antigen 4 (CTLA-4), programmed death-ligand 1 (PD-L1), Forkhead box P3 (FOXP3), miR-155, and miR-195 were assessed in the peripheral blood of 90 BC patients compared to 30 healthy controls using quantitative real-time PCR (qRt-PCR). The plasma level of soluble MHC class I chain related-protein B (MIC-B) protein was assessed using the enzyme linked immunosorbent assay (ELISA) technique. The data were correlated to the clinico-pathological characteristics of the patients. RESULTS There was a significant increase in the expression levels of PDL-1 [17.59 (3.24-123), p < 0.001], CTLA-4 [23.34 (1.3-1267), p = 0.006], PD-1 [10.25 (1-280), p < 0.001], FOXP3 [11.5 (1-234.8), p = 0.001], miR-155 [87.3 (1.5-910), p < 0.001] in BC patients compared to normal controls. The miR-195 was significantly downregulated in BC patients [0.23 (0-0.98, p < 0.001]. The plasma level of MIC-B was significantly increased in the BC patients [0.941 (0.204-6.38) ng/ml], compared to the control group [0.351 (0.211-0.884) ng/mL, p < 0.00]. PDL-1, CTLA-4, PD-1, and FOXP3 achieved a specificity of 100% for distinguishing BC patients, at a sensitivity of 93.3%, 82.2%, 62.2%, and 71.1% respectively. The combined expression of PDL-1 and CTLA-4 scored a 100% sensitivity and 100% specificity for diagnosing BC (p < 0.001). The sensitivity, specificity, and AUC of miR-155 were 88.9%, 96.7%, and 0.934; respectively (p < 0.001). While those of miR-195 were 73.3%, 60%, and 0.716; respectively (p = 0.001). MIC-B expression showed a 77.8% sensitivity, 80% specificity, and 0.811 AUC at a cutoff of 1.17 ng/ml (p < 0.001). Combined expression of miR-155 and miR-195 achieved a sensitivity of 91.1%, a specificity of 96.7%, and AUC of 0.926 (p < 0.001). Multivariate analysis showed that PDL-1 (OR:13.825, p = 0.004), CTLA-4 (OR: 20.958, p = 0.010), PD-1(OR:10.550, p = 0.044), MIC-B (OR: 17.89, p = 0.003), miR-155 (OR: 211.356, P < 0.001), and miR-195(OR:0.006, P < 0.001) were considered as independent risk factors for BC. CONCLUSIONS The PB levels of PDL-1, CTLA-4, PD-1, FOXP3, MIC-B, miR-155, and miR-195 could be used as promising diagnostic markers for BC patients.
Collapse
Affiliation(s)
| | - Motawa E El-Houseini
- Medical Biochemistry and Molecular Biology, Cancer Biology Department, National Cancer Institute, Cairo University, Cairo, Egypt
| | - Heba Effat
- Medical Biochemistry and Molecular Biology, Cancer Biology Department, National Cancer Institute, Cairo University, Cairo, Egypt
| | | | - Mona S Abdellateif
- Medical Biochemistry and Molecular Biology, Cancer Biology Department, National Cancer Institute, Cairo University, Cairo, Egypt.
| |
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
|
Liu K, Zhu C, Liang Y, Min D, Jin Z, Sun X. Discovery of a Novel 1,4-Benzodiazepine Derivative as a Highly Selective ANXA3 Degrader for the Treatment of Triple-Negative Breast Cancer. J Med Chem 2025; 68:5358-5381. [PMID: 40013713 DOI: 10.1021/acs.jmedchem.4c02403] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/28/2025]
Abstract
Annexin A3 has been demonstrated to be a key pathogenic protein in the occurrence and development of triple-negative breast cancer (TNBC); its overexpression in TNBC cells can promote the proliferation, migration, and drug resistance of TNBC. Previously, we reported the first ANXA3 degrader, (R)-SL18, with potent anti-TNBC effects, albeit with moderate ANXA3 binding affinity leading to off-target effects and relatively poor degradation selectivity of family proteins. To obtain molecules with stronger binding with ANXA3 and lower toxicity, we performed further structural optimization of (R)-SL18 to explore structure-activity relationships for a series of 1,4-benzodiazepines. Among them, compound 18a5 exhibited a 14-fold increase in ANXA3 binding activity, along with better cancer cell inhibition and functional activity. In particular, 18a5 showed more desirable ANXA3 selective degradation than (R)-SL18 and displayed excellent inhibitory effect in a TNBC tumor xenograft model (TGI = 96%). Collectively, 18a5 proved to be a promising lead compound to treat TNBC through the degradation of ANXA3.
Collapse
Affiliation(s)
- Kunlin Liu
- School of Pharmacy, Fudan University, 826 Zhangheng Road, Shanghai 201203, China
| | - Chenhao Zhu
- School of Pharmacy, Fudan University, 826 Zhangheng Road, Shanghai 201203, China
| | - Yongxi Liang
- School of Pharmacy, Fudan University, 826 Zhangheng Road, Shanghai 201203, China
| | - Delin Min
- School of Pharmacy, Fudan University, 826 Zhangheng Road, Shanghai 201203, China
| | - Zijun Jin
- School of Pharmacy, Fudan University, 826 Zhangheng Road, Shanghai 201203, China
| | - Xun Sun
- School of Pharmacy, Fudan University, 826 Zhangheng Road, Shanghai 201203, China
- The Institutes of Integrative Medicine of Fudan University, 12 Wulumuqi Zhong Road, Shanghai 200040, China
| |
Collapse
|
24
|
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
|
25
|
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
|
26
|
Guo X, Nie H, Zhang W, Li J, Ge J, Xie B, Hu W, Zhu Y, Zhong N, Zhang X, Zhao X, Wang X, Sun Q, Wei K, Chen X, Ni L, Zhang T, Lu S, Zhang L, Dong C. Contrasting cytotoxic and regulatory T cell responses underlying distinct clinical outcomes to anti-PD-1 plus lenvatinib therapy in cancer. Cancer Cell 2025; 43:248-268.e9. [PMID: 39889705 DOI: 10.1016/j.ccell.2025.01.001] [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: 11/20/2023] [Revised: 09/04/2024] [Accepted: 01/06/2025] [Indexed: 02/03/2025]
Abstract
Combination of anti-PD-1 with lenvatinib showed clinical efficacy in multiple cancers, yet the underlying immunological mechanisms are unclear. Here, we compared T cells in hepatocellular carcinoma (HCC) patients before and after combination treatment using single-cell transcriptomics and T cell receptor (scTCR) clonotype analyses. We found that tumor-infiltrating GZMK+ CD8+ effector/effector memory T (Teff/Tem) cells, showing a favorable response to combination therapy, comprise progenitor exhausted T (Tpex) cells and also unappreciated circulating Tem (cTem) cells enriched with hepatitis B virus (HBV) specificity. Further integrated analyses revealed that cTem cells are specifically associated with responsiveness to the combination therapy, whereas Tpex cells contribute to responses in both combination therapy and anti-PD-1 monotherapy. Notably, an underexplored KIR+ CD8+ T cell subset in the tumor and FOXP3+ CD4+ regulatory T cells are specifically enriched in non-responders after the combination therapy. Our study thus elucidated T cell subsets associated with clinical benefits and resistance in cancer immunotherapy.
Collapse
Affiliation(s)
- Xinyi Guo
- Shanghai Immune Therapy Institute, New Cornerstone Science Laboratory, Shanghai Jiao Tong University School of Medicine - Affiliated Renji Hospital, Shanghai 200127, China; Institute for Immunology, Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing 100084, China
| | - Hu Nie
- Institute of Cancer Research, Shenzhen Bay Laboratory, Shenzhen, Guangdong 518132, China; State Key Laboratory of Chemical Oncogenomics, Shenzhen Key Laboratory of Chemical Genomics, School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen, Guangdong 518055, China
| | - Wenwen Zhang
- Faculty of Hepato-Pancreato-Biliary Surgery, Chinese PLA General Hospital / Key Laboratory of Digital Hepatobiliary Surgery, PLA / Institute of Hepatobiliary Surgery of Chinese PLA, Beijing 100953, China
| | - Jiesheng Li
- Institute of Cancer Research, Shenzhen Bay Laboratory, Shenzhen, Guangdong 518132, China; State Key Laboratory of Chemical Oncogenomics, Shenzhen Key Laboratory of Chemical Genomics, School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen, Guangdong 518055, China
| | - Jing Ge
- Shanghai Immune Therapy Institute, New Cornerstone Science Laboratory, Shanghai Jiao Tong University School of Medicine - Affiliated Renji Hospital, Shanghai 200127, China
| | - Bowen Xie
- Institute for Immunology, Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing 100084, China
| | - Wenbo Hu
- Institute for Immunology, Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing 100084, China
| | - Yicheng Zhu
- Shanghai Immune Therapy Institute, New Cornerstone Science Laboratory, Shanghai Jiao Tong University School of Medicine - Affiliated Renji Hospital, Shanghai 200127, China
| | - Na Zhong
- Shenzhen Peacock Biotechnology Co., Ltd, Shenzhen, Guangdong 518112, China
| | - Xinmei Zhang
- Shenzhen Peacock Biotechnology Co., Ltd, Shenzhen, Guangdong 518112, China
| | - Xiaohong Zhao
- Institute for Immunology, Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing 100084, China
| | - Xiaoshuang Wang
- Shanghai Immune Therapy Institute, New Cornerstone Science Laboratory, Shanghai Jiao Tong University School of Medicine - Affiliated Renji Hospital, Shanghai 200127, China; Institute for Immunology, Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing 100084, China
| | - Qinli Sun
- Institute for Immunology, Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing 100084, China
| | - Kun Wei
- Institute for Immunology, Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing 100084, China
| | - Xiaoyuan Chen
- Tsinghua Clinical Research Institute, School of Medicine, Tsinghua University, Beijing 100084, China
| | - Ling Ni
- Institute for Immunology, Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing 100084, China
| | - Ting Zhang
- Shanghai Immune Therapy Institute, New Cornerstone Science Laboratory, Shanghai Jiao Tong University School of Medicine - Affiliated Renji Hospital, Shanghai 200127, China
| | - Shichun Lu
- Faculty of Hepato-Pancreato-Biliary Surgery, Chinese PLA General Hospital / Key Laboratory of Digital Hepatobiliary Surgery, PLA / Institute of Hepatobiliary Surgery of Chinese PLA, Beijing 100953, China.
| | - Lei Zhang
- Institute of Cancer Research, Shenzhen Bay Laboratory, Shenzhen, Guangdong 518132, China; State Key Laboratory of Chemical Oncogenomics, Shenzhen Key Laboratory of Chemical Genomics, School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen, Guangdong 518055, China; Shenzhen Medical Academy of Research and Translation (SMART), Shenzhen, Guangdong 518107, China.
| | - Chen Dong
- Shanghai Immune Therapy Institute, New Cornerstone Science Laboratory, Shanghai Jiao Tong University School of Medicine - Affiliated Renji Hospital, Shanghai 200127, China; Research Unit of Immune Regulation and Immune Diseases (2022RU001), Chinese Academy of Medical Sciences, Shanghai Jiao Tong University School of Medicine - Affiliated Renji Hospital, Shanghai 200127, China; Westlake University School of Medicine, Hangzhou, Zhejiang 310030, China.
| |
Collapse
|
27
|
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
|
28
|
Zgura A, Chipuc S, Bacalbasa N, Haineala B, Rodica A, Sebastian V. Evaluating Tumour Mutational Burden as a Key Biomarker in Personalized Cancer Immunotherapy: A Pan-Cancer Systematic Review. Cancers (Basel) 2025; 17:480. [PMID: 39941847 PMCID: PMC11816366 DOI: 10.3390/cancers17030480] [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: 12/29/2024] [Revised: 01/23/2025] [Accepted: 01/27/2025] [Indexed: 02/16/2025] Open
Abstract
BACKGROUND Tumour mutational burden (TMB) is an emerging biomarker for predicting the efficacy of immune checkpoint inhibitors (ICIs) in cancer therapy. While its role is well established in lung cancer and melanoma, its predictive value for breast and prostate cancers remains unclear. OBJECTIVE This systematic review aimed to assess the predictive value of TMB for ICI therapy across four major cancer types-lung, melanoma, breast, and prostate-and to explore factors contributing to the variability in its effectiveness as a biomarker. METHODS A systematic search and a review of the literature were conducted in accordance with PRISMA guidelines. Studies examining the relationship between TMB levels and clinical outcomes following ICI therapy in the specified cancers were analyzed. The data were synthesized to evaluate TMB's predictive value and identify gaps in the current research. RESULTS High TMB consistently correlated with improved outcomes in lung cancer and melanoma, confirming its predictive utility in these cancers. Conversely, the findings for breast and prostate cancers were inconclusive. The variability in TMB's predictive value for these cancers suggests the need for complementary biomarkers or refined criteria to enhance its reliability. Methodological inconsistencies in TMB evaluation were also noted as a significant limitation. CONCLUSIONS TMB serves as a robust biomarker for predicting ICI response in lung cancer and melanoma, but demonstrates limited predictive utility in breast and prostate cancers. Future research should prioritize standardizing TMB assessment protocols and investigating additional biomarkers to improve treatment personalization for these cancer types.
Collapse
Affiliation(s)
- Anca Zgura
- Department of Oncology-Radiotherapy, Prof. Dr. Alexandru Trestioreanu Institute of Oncology, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania; (A.Z.)
- Prof. Dr. Alexandru Trestioreanu Institute of Oncology, 022328 Bucharest, Romania
| | - Stefania Chipuc
- Prof. Dr. Alexandru Trestioreanu Institute of Oncology, 022328 Bucharest, Romania
| | - Nicolae Bacalbasa
- Department of Surgery, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania; (N.B.); (V.S.)
| | - Bogdan Haineala
- Department of Urology, “Fundeni” Clinical Institute, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania;
| | - Anghel Rodica
- Department of Oncology-Radiotherapy, Prof. Dr. Alexandru Trestioreanu Institute of Oncology, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania; (A.Z.)
- Prof. Dr. Alexandru Trestioreanu Institute of Oncology, 022328 Bucharest, Romania
| | - Vâlcea Sebastian
- Department of Surgery, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania; (N.B.); (V.S.)
| |
Collapse
|
29
|
Takahashi H. Neurological symptom management in breast cancer meningeal carcinomatosis. TRANSLATIONAL BREAST CANCER RESEARCH : A JOURNAL FOCUSING ON TRANSLATIONAL RESEARCH IN BREAST CANCER 2025; 6:7. [PMID: 39980811 PMCID: PMC11836744 DOI: 10.21037/tbcr-24-47] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/08/2024] [Accepted: 11/25/2024] [Indexed: 02/22/2025]
Abstract
No treatment has been established for meningeal carcinomatosis (MC) in advanced metastatic breast cancer, and its prognosis is poor. In recent years, systemic therapies such as trastuzumab deruxtecan and tucatinib have been reported effective for human epidermal growth factor receptor 2 (HER2)-positive breast cancer, however, these cannot be used for all MC. The difficulty in diagnosing and treating MC is attributed to its diverse pathology. As a result, in clinical practice, diagnosis is often delayed, and symptoms persist. This review focuses on whether neurological symptoms can be effectively alleviated even with unestablished treatments by classifying the pathology of MC into meningitis, hydrocephalus-related intracranial hypertension symptoms, focal brain damage such as epilepsy, cranial nerve disorders, and spinal cord symptoms and evaluating the diagnosis and condition. Hydrocephalus can be managed with drainage and ventriculoperitoneal shunt surgery, and meningitis symptoms and cranial nerve disorders can be managed with whole brain radiotherapy. Antiepileptic drugs are essential for epilepsy, and supportive care is necessary, as are steroids for cranial nerve disorders. However, MC is not caused by a single condition but can occur in combination thus the therapeutic effectiveness of palliative therapy for neurological symptoms is currently unknown, and research is limited. In the future, if a lineup of highly effective systemic therapies such as tyrosine kinase inhibitors for ALK gene-positive lung cancer is established, treatment strategies for MC may change. However at present, rapid diagnosis and prompt neurological palliative treatment play an important role in the neurological symptoms management of MC.
Collapse
Affiliation(s)
- Hideaki Takahashi
- Department of Neurosurgery, Niigata Cancer Center Hospital, Niigata, Japan
| |
Collapse
|
30
|
Zhu B, Cai Y, Zhou L, Zhao L, Chen J, Shan X, Sun X, You Q, Gong X, Zhang W, Zhu HH, Zhang P, Li Y. Injectable supramolecular hydrogel co-loading abemaciclib/NLG919 for neoadjuvant immunotherapy of triple-negative breast cancer. Nat Commun 2025; 16:687. [PMID: 39814714 PMCID: PMC11735626 DOI: 10.1038/s41467-025-55904-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: 11/15/2023] [Accepted: 01/04/2025] [Indexed: 01/18/2025] Open
Abstract
The efficacy of cancer immunotherapy relies on a sufficient amount of functional immune cells. Triple-negative breast cancer lacks enough immune cell infiltration, and adjuvant therapy is necessary to prime anti-tumor immunity. However, the improvement in efficacy is unsatisfactory with concern about inducing systemic immunotoxicity. Herein, we create an abemaciclib-loaded supramolecular peptide hydrogel formed by peptide-drug amphiphiles for neoadjuvant immunotherapy of triple-negative breast cancer, where the amphiphile is a conjugate of a β-sheet-forming peptide with 1-cyclohexyl-2-(5H-imidazo[5,1-a]isoindol-5-yl)ethanol (NLG919), an inhibitor of indoleamine 2,3-dioxygenase 1. The hydrogel can be injected into the tumor site and retained for at least one week for the sustained release of both abemaciclib and NLG919. The abemaciclib is able to induce immunogenic cell death of cancer cells and increase interleukin-2 secretion by cytotoxic T lymphocytes. Abemaciclib adversely upregulates indoleamine 2,3-dioxygenase 1, whose kynurenine production activity is inhibited by NLG919. The neoadjuvant immunotherapy reduces tumor recurrence and pulmonary metastasis and prolongs the survival of animals. This hydrogel provides a potential platform for neoadjuvant immunotherapy of triple-negative breast cancer with reduced toxicity compared with free abemaciclib.
Collapse
Affiliation(s)
- Binyu Zhu
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, No.19A Yuquan Road, Beijing, China
| | - Ying Cai
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, No.19A Yuquan Road, Beijing, China
- Yantai Key Laboratory of Nanomedicine & Advanced Preparations, Yantai Institute of Pharmaceutical Science, Shandong, China
| | - Lingli Zhou
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, China
| | - Lei Zhao
- School of Chemical Engineering, Zhengzhou University, Zhengzhou, China
| | - Jiameng Chen
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, No.19A Yuquan Road, Beijing, China
| | - Xiaoting Shan
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, No.19A Yuquan Road, Beijing, China
| | - Xujie Sun
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, No.19A Yuquan Road, Beijing, China
| | - Qian You
- State Key Laboratory of Oncogenes and Related Genes, Renji-Med-X Stem Cell Research Center, Department of Urology, Ren Ji Hospital, School of Medicine and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Xiang Gong
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
- National Advanced Medical Engineering Research Center, China State Institute of Pharmaceutical Industry, Shanghai, China
| | - Wen Zhang
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
- National Advanced Medical Engineering Research Center, China State Institute of Pharmaceutical Industry, Shanghai, China
| | - Helen He Zhu
- State Key Laboratory of Oncogenes and Related Genes, Renji-Med-X Stem Cell Research Center, Department of Urology, Ren Ji Hospital, School of Medicine and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Pengcheng Zhang
- School of Biomedical Engineering & State Key Laboratory of Advanced Medical Materials and Devices, ShanghaiTech University, Shanghai, China.
| | - Yaping Li
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China.
- University of Chinese Academy of Sciences, No.19A Yuquan Road, Beijing, China.
- Yantai Key Laboratory of Nanomedicine & Advanced Preparations, Yantai Institute of Pharmaceutical Science, Shandong, China.
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, China.
- Shandong Laboratory of Yantai Drug Discovery, Bohai rim Advanced Research Institute for Drug Discovery, Shandong, China.
| |
Collapse
|
31
|
Xiong H, Song Z, Wang T, Huang K, Yu F, Sun W, Liu X, Liu L, Jiang H, Wang X. Photoswitchable dynamics and RNAi synergist with tailored interface and controlled release reprogramming tumor immunosuppressive niche. Biomaterials 2025; 312:122712. [PMID: 39098305 DOI: 10.1016/j.biomaterials.2024.122712] [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: 02/27/2024] [Revised: 06/29/2024] [Accepted: 07/25/2024] [Indexed: 08/06/2024]
Abstract
Immunosuppressive tumor microenvironment (ITM) severely limited the efficacy of immunotherapy against triple-negative breast cancer (TNBC). Herein, Apt-LPR, a light-activatable photodynamic therapy (PDT)/RNAi immune synergy-enhancer was constructed by co-loading miR-34a and photosensitizers in cationic liposomes (in phase III clinical trial). Interestingly, the introduction of tumor-specific aptamers creates a special "Liposome-Aptamer-Target" interface, where the aptamers are initially in a "lying down" state but transform to "standing up" after target binding. The interfacing mechanism was elaborately revealed by computational and practical experiments. This unique interface endowed Apt-LPR with neutralized surface potential of cationic liposomes to reduce non-specific cytotoxicity, enhanced DNase resistance to protect aptamers, and preserved target-binding ability for selective drug delivery. Upon near-infrared irradiation, the generated reactive oxygen species would oxidize unsaturated phospholipids to destabilize both liposomes and lysosomes, realizing stepwise lysosomal escape of miR-34a for tumor cell apoptosis and downregulation of PD-L1 to suppress immune escape. Together, tumor-associated antigens released from PDT-damaged mitochondria and endoplasmic reticulum could activate the suppressive immune cells to establish an "immune hot" milieu. The collaborative immune-enhancing strategy effectively aroused systemic antitumor immunity and inhibited primary and distal tumor progression as well as lung metastasis in 4T1 xenografted mouse models. The photo-controlled drug release and specific tumor-targeting capabilities of Apt-LPR were also visualized in MDA-MB-231 xenografted zebrafish models. Therefore, this photoswitchable PDT/RNAi immune stimulator offered a powerful approach to reprogramming ITM and reinforcing cancer immunotherapy efficacy.
Collapse
Affiliation(s)
- Hongjie Xiong
- State Key Laboratory of Digital Medical Engineering, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, PR China
| | - Zhongquan Song
- Department of Respiratory Medicine, Zhongda Hospital, Medical School, Southeast University, Nanjing, 210009, PR China
| | - Tingya Wang
- Department of Oncology, Zhongda Hospital, Medical School, Southeast University, Nanjing, 210009, PR China
| | - Ke Huang
- State Key Laboratory of Digital Medical Engineering, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, PR China
| | - Fangfang Yu
- State Key Laboratory of Digital Medical Engineering, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, PR China
| | - Wenyu Sun
- State Key Laboratory of Digital Medical Engineering, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, PR China
| | - Xiaohui Liu
- State Key Laboratory of Digital Medical Engineering, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, PR China
| | - Liu Liu
- State Key Laboratory of Digital Medical Engineering, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, PR China.
| | - Hui Jiang
- State Key Laboratory of Digital Medical Engineering, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, PR China.
| | - Xuemei Wang
- State Key Laboratory of Digital Medical Engineering, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, PR China.
| |
Collapse
|
32
|
Chen C, Pang X, Zhang M, Li F, Zhang W, Chen W. Long-Term and Sustained Remission of Advanced Triple-Negative Breast Cancer with Large Chest Wall Lesions after Transient Chemoimmunotherapy: A Case Report. Case Rep Oncol 2025; 18:198-205. [PMID: 39980521 PMCID: PMC11793911 DOI: 10.1159/000543292] [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: 08/24/2024] [Accepted: 12/18/2024] [Indexed: 02/22/2025] Open
Abstract
Introduction We report a case of advanced triple-negative breast cancer (TNBC) with special clinical manifestations, in which a durable remission was achieved after short-term administration of toripalimab combined with chemotherapy. The progress, advantages, and unique experience of chemoimmunotherapy in TNBC were examined. Case Presentation A patient with TNBC with local recurrence 2 years following surgery, with inoperable large chest wall lesions and positive PD-L1 as the main manifestations, was treated with toripalimab plus paclitaxel (albumin-bound) for 5 months and achieved a partial remission. Twenty-five months after the discontinuation of treatment, the chest wall lesions exhibited a slow but continuous decline, until they achieved a nearly complete remission; however, the patient eventually died from cancer progression. Conclusion Typically, chest wall recurrence in TNBC has a poor prognosis; however, recurrence was rapidly controlled, sustained remission was achieved after immunotherapy combined with chemotherapy, and the curative effect continued after drug withdrawal, which is a rare occurrence. Thus, the tailing effect of immune checkpoint inhibitors was confirmed in the treatment of TNBC.
Collapse
Affiliation(s)
- Chanjuan Chen
- Department of Oncology, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Xiaonan Pang
- Department of Oncology, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Mingjun Zhang
- Department of Oncology, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Fanfan Li
- Department of Oncology, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Wanying Zhang
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Wei Chen
- Department of Oncology, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
| |
Collapse
|
33
|
Varshini MS, Krishnamurthy PT, Reddy RA, Wadhwani A, Chandrashekar VM. Insights into the Emerging Therapeutic Targets of Triple-negative Breast Cancer. Curr Cancer Drug Targets 2025; 25:3-25. [PMID: 38385495 DOI: 10.2174/0115680096280750240123054936] [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: 09/23/2023] [Revised: 12/24/2023] [Accepted: 01/09/2024] [Indexed: 02/23/2024]
Abstract
Triple-negative Breast Cancer (TNBC), the most aggressive breast cancer subtype, is characterized by the non-appearance of estrogen receptor (ER), progesterone receptor (PR), and human epidermal growth factor receptor 2 (HER2). Clinically, TNBC is marked by its low survival rate, poor therapeutic outcomes, high aggressiveness, and lack of targeted therapies. Over the past few decades, many clinical trials have been ongoing for targeted therapies in TNBC. Although some classes, such as Poly (ADP Ribose) Polymerase (PARP) inhibitors and immunotherapies, have shown positive therapeutic outcomes, however, clinical effects are not much satisfiable. Moreover, the development of drug resistance is the major pattern observed in many targeted monotherapies. The heterogeneity of TNBC might be the cause for limited clinical benefits. Hence,, there is a need for the potential identification of new therapeutic targets to address the above limitations. In this context, some novel targets that can address the above-mentioned concerns are emerging in the era of TNBC therapy, which include Hypoxia Inducible Factor (HIF-1α), Matrix Metalloproteinase 9 (MMP-9), Tumour Necrosis Factor-α (TNF-α), β-Adrenergic Receptor (β-AR), Voltage Gated Sodium Channels (VGSCs), and Cell Cycle Regulators. Currently, we summarize the ongoing clinical trials and discuss the novel therapeutic targets in the management of TNBC.
Collapse
Affiliation(s)
- Magham Sai Varshini
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education and Research, Ooty, 643001, TN, India
| | | | - Ramakamma Aishwarya Reddy
- Department of Pharmaceutics, JSS College of Pharmacy, JSS Academy of Higher Education and Research, Ooty, 643001, TN, India
| | - Ashish Wadhwani
- Department of Pharmaceutical Biotechnology, JSS College of Pharmacy, JSS Academy of Higher Education and Research, Ooty, 643001, TN, India
- Faculty of Health Sciences, School of Pharmacy, JSS Academy of Higher Education and Research, Mauritius, Vacoas, 73304, Mauritius
| | - V M Chandrashekar
- Department of Pharmacology, HSK College of Pharmacy, Bagalkot, 587101, Karnataka, India
| |
Collapse
|
34
|
Chu M, Huang J, Wang Q, Fang Y, Cui D, Jin Y. A Circadian Rhythm-related Signature to Predict Prognosis, Immune Infiltration, and Drug Response in Breast Cancer. Curr Med Chem 2025; 32:608-626. [PMID: 39279697 DOI: 10.2174/0109298673320179240803071001] [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: 04/03/2024] [Revised: 07/01/2024] [Accepted: 07/08/2024] [Indexed: 09/18/2024]
Abstract
PURPOSE Circadian rhythm-related genes (CRRGs) play essential roles in cancer occurrence and development. However, the prognostic significance of CRRGs in breast cancer (BC) has not been fully elucidated. Our study aimed to develop a prognostic gene signature based on CRRGs that can accurately and stably predict the prognosis of BC. METHODS The transcriptome data and clinical information for BC patients were obtained from The Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO) databases. A consensus unsupervised clustering analysis was carried out to investigate the roles of CRRGs in BC. A CRRGs-related prognostic risk model was established by using logistic least absolute shrinkage and selection operator (LASSO) Cox regression and univariate Cox regression analyses. Kaplan-Meier (KM) curves analysis, time-dependent receptor operation characteristics (ROC) curves analysis, and nomogram were plotted to evaluate the predictive efficacy of the model. The relevance of risk score to the immune cell infiltration, tumor burden mutation (TMB), and therapeutic response was assessed. RESULTS A risk model comprising six CRRGs (SLC44A4, SLC16A6, TPRG1, FABP7, GLYATL2, and FDCSP) was constructed and validated, demonstrating an effective predictor for the prognosis of BC. The low-risk group displayed a higher expression of immune checkpoint genes and a lower burden of tumor mutation. Additionally, drug sensitivity analysis demonstrated that the prognostic signature may serve as a potential chemosensitivity predictor. CONCLUSION We established a CRRGs-related risk signature, which is of great value in predicting the prognosis of patients with BC and guiding the treatment for BC.
Collapse
Affiliation(s)
- Mingyu Chu
- Department of Medical Genetics, School of Basic Medical Sciences, Nanjing Medical University, 101 Longmian Avenue, Nanjing, 211166, China
| | - Jing Huang
- Department of Medical Genetics, School of Basic Medical Sciences, Nanjing Medical University, 101 Longmian Avenue, Nanjing, 211166, China
| | - Qianyu Wang
- Department of Medical Genetics, School of Basic Medical Sciences, Nanjing Medical University, 101 Longmian Avenue, Nanjing, 211166, China
| | - Yaqun Fang
- Department of Medical Genetics, School of Basic Medical Sciences, Nanjing Medical University, 101 Longmian Avenue, Nanjing, 211166, China
| | - Dina Cui
- Department of Medical Genetics, School of Basic Medical Sciences, Nanjing Medical University, 101 Longmian Avenue, Nanjing, 211166, China
| | - Yucui Jin
- Department of Medical Genetics, School of Basic Medical Sciences, Nanjing Medical University, 101 Longmian Avenue, Nanjing, 211166, China
- Jiangsu Key Laboratory of Xenotransplantation, School of Basic Medical Sciences, Nanjing Medical University, 101 Longmian Avenue, Nanjing, 211166, China
| |
Collapse
|
35
|
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
|
36
|
Qu F, Wang G, Wen P, Liu X, Zeng X. Knowledge mapping of immunotherapy for breast cancer: A bibliometric analysis from 2013 to 2022. Hum Vaccin Immunother 2024; 20:2335728. [PMID: 38563136 PMCID: PMC10989689 DOI: 10.1080/21645515.2024.2335728] [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: 01/17/2024] [Accepted: 03/24/2024] [Indexed: 04/04/2024] Open
Abstract
Breast cancer is the leading cause of cancer-related death among women globally. Immunotherapy has emerged as a major milestone in contemporary oncology. This study aims to conduct a bibliometric analysis in the field of immunotherapy for breast cancer, providing a comprehensive overview of the current research status, identifying trends and hotspots in research topics. We searched and retrieved data from the Web of Science Core Collection, and performed a bibliometric analysis of publications on immunotherapy for breast cancer from 2013 to 2022. Current status and hotspots were evaluated by co-occurrence analysis using VOSviewer. Evolution and bursts of knowledge base were assessed by co-citation analysis using CiteSpace. Thematic evolution by bibliometrix package was used to discover keywords trends. The attribution and collaboration of countries/regions, institutions and authors were also explored. A total of 7,975 publications were included. In co-occurrence analysis of keywords, 6 major clusters were revealed: tumor microenvironment, prognosis biomarker, immune checkpoints, novel drug delivery methods, immune cells and therapeutic approaches. The top three most frequently mentioned keywords were tumor microenvironment, triple-negative breast cancer, and programmed cell death ligand 1. The most productive country, institution and author were the USA (2926 publications), the University of Texas MD Anderson Cancer Center (219 publications), and Sherene Loi (28 publications), respectively. There has been a rapid growth in studies on immunotherapy for breast cancer worldwide. This research area has gained increasing attention from different countries and institutions. With the rising incidence of breast cancer, immunotherapy represents a research field of significant clinical value and potential.
Collapse
Affiliation(s)
- Fanli Qu
- Department of Breast Cancer Center, Chongqing University Cancer Hospital, Chongqing, China
- Chongqing Key Laboratory for Intelligent Oncology in Breast Cancer (iCQBC), Chongqing University Cancer Hospital, Chongqing, China
| | - Guanwen Wang
- Department of Breast Cancer Center, Chongqing University Cancer Hospital, Chongqing, China
- Chongqing Key Laboratory for Intelligent Oncology in Breast Cancer (iCQBC), Chongqing University Cancer Hospital, Chongqing, China
| | - Ping Wen
- School of Medicine, Chongqing University, Chongqing, China
| | - Xiaoyu Liu
- Department of Breast Cancer Center, Chongqing University Cancer Hospital, Chongqing, China
- Chongqing Key Laboratory for Intelligent Oncology in Breast Cancer (iCQBC), Chongqing University Cancer Hospital, Chongqing, China
| | - Xiaohua Zeng
- Department of Breast Cancer Center, Chongqing University Cancer Hospital, Chongqing, China
- Chongqing Key Laboratory for Intelligent Oncology in Breast Cancer (iCQBC), Chongqing University Cancer Hospital, Chongqing, China
| |
Collapse
|
37
|
Xie Z, Cai D, Ye R, Shan Z, Lin Y, Gao F, Shao N, Kuang X. Integrated immune-related gene signature predicts clinical outcome for patients with Luminal B breast cancer. Gland Surg 2024; 13:2253-2263. [PMID: 39822362 PMCID: PMC11733651 DOI: 10.21037/gs-24-377] [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: 08/27/2024] [Accepted: 12/03/2024] [Indexed: 01/19/2025]
Abstract
Background Luminal B breast cancer is routinely treated with chemotherapy and endocrine therapy. However, its sensitivity to treatment remains heterogeneous; therefore, identifying patients who may most benefit remains crucial. Immune-related genes are reportedly related to the prognosis of breast cancer. The purpose of this study was to evaluate the impact of an immune-related gene signature (IRGS) in predicting the prognosis of patients with Luminal B breast cancer. Methods We selected patients with Luminal B breast cancer from two large datasets: 488 from the Metabric dataset (training cohort) and 250 patients from The Cancer Genome Atlas (TCGA) dataset (validation cohort). Prognostic analysis was performed to test the predictive value of IRGS, and enrichment analysis and ESTIMATE were used for deeper function analysis. Results A prognostic IRGS model containing 12 immune-related genes was developed. After which, we separated patients with Luminal B breast cancer into low- and high-risk groups in terms of disease-free survival (DFS) (P<0.001). Multivariate analysis identified IRGS as an independent prognostic factor. Furthermore, functional analysis showed that the 12 genes were mainly enriched in pathways related to chemotherapy response, whose expression levels showed completely opposing trends in low- and high-risk groups. Conclusions The novel IRGS is a satisfactory and reliable biomarker to predict the clinical outcome of patients with Luminal B breast cancer which potentially facilitating individualised management. Further studies are needed to assess the clinical potential in predicting prognosis and the treatment options for Luminal B breast cancer patients.
Collapse
Affiliation(s)
- Zhen Xie
- Department of Breast Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Du Cai
- Department of Colorectal Surgery, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Runyi Ye
- Department of Breast Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Zhen Shan
- Department of Breast Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Ying Lin
- Department of Breast Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Feng Gao
- Department of Colorectal Surgery, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Nan Shao
- Department of Breast Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Xiaying Kuang
- Department of Breast Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
38
|
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
|
39
|
Vishnubalaji R, Alajez NM. Disrupted Lipid Metabolism, Cytokine Signaling, and Dormancy: Hallmarks of Doxorubicin-Resistant Triple-Negative Breast Cancer Models. Cancers (Basel) 2024; 16:4273. [PMID: 39766172 PMCID: PMC11674486 DOI: 10.3390/cancers16244273] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Revised: 11/30/2024] [Accepted: 12/16/2024] [Indexed: 01/11/2025] Open
Abstract
BACKGROUND Chemoresistance in triple-negative breast cancer (TNBC) presents a significant clinical hurdle, limiting the efficacy of treatments like doxorubicin. This study aimed to explore the molecular changes associated with doxorubicin resistance and identify potential therapeutic targets to overcome this resistance, thereby improving treatment outcomes for TNBC patients. METHODS Doxorubicin-resistant (DoxR) TNBC models (MDA-MB-231 and BT-549) were generated by exposing cells to increasing concentrations of doxorubicin. RNA sequencing (RNA-Seq) was performed using the Illumina platform, followed by bioinformatics analysis with CLC Genomics Workbench and iDEP. Functional assays assessed proliferation, sphere formation, migration, and cell cycle changes. Protein expression and phosphorylation were confirmed via Western blotting. Pathway and network analyses were conducted using Ingenuity Pathway Analysis (IPA) and STRING, while survival analysis was performed using Kaplan-Meier Plotter database. RESULTS DoxR cells exhibited reduced proliferation, sphere formation, and migration, but showed enhanced tolerance to doxorubicin. Increased CHK2 and p53 phosphorylation indicated cellular dormancy as a resistance mechanism. RNA-Seq analysis revealed upregulation of cytokine signaling and stress-response pathways, while cholesterol and lipid biosynthesis were suppressed. Activation of the IL1β cytokine network was prominent in DoxR cells, and CRISPR-Cas9 screens data identified dependencies on genes involved in rRNA biogenesis and metabolism. A 27-gene signature associated with doxorubicin resistance was linked to worse clinical outcomes in a large breast cancer cohort (HR = 1.76, FDR p < 2.0 × 10-13). CONCLUSIONS This study uncovers potential therapeutic strategies for overcoming TNBC resistance, including dormancy reversal and targeting onco-ribosomal pathways and cytokine signaling networks, to improve the efficacy of doxorubicin-based treatments.
Collapse
Affiliation(s)
- Radhakrishnan Vishnubalaji
- Translational Oncology Research Center (TORC), Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation (QF), Doha P.O. Box 34110, Qatar;
| | - Nehad M. Alajez
- Translational Oncology Research Center (TORC), Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation (QF), Doha P.O. Box 34110, Qatar;
- College of Health & Life Sciences, Hamad Bin Khalifa University (HBKU), Qatar Foundation (QF), Doha P.O. Box 34110, Qatar
| |
Collapse
|
40
|
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
|
41
|
Fan Y, Sun L, He J, Chen Y, Ma H, Ding H. Siglec15 in blood system diseases: from bench to bedside. Front Immunol 2024; 15:1490505. [PMID: 39697338 PMCID: PMC11652361 DOI: 10.3389/fimmu.2024.1490505] [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: 09/03/2024] [Accepted: 11/13/2024] [Indexed: 12/20/2024] Open
Abstract
Inhibiting the PD-1/PD-L1 pathway using immunomodulators has demonstrated promising outcomes in clinics. Immunomodulators can effectively target immune checkpoints with a strong preference for the tumor microenvironment (TME). Besides, immunomodulators specifically target the recently discovered inhibitory immune checkpoint, sialic acid-binding immunoglobulin-like lectin (Siglec-15). Distinctive in its molecular composition, Siglec-15 has a unique molecular composition and been shown to be highly prevalent in numerous solid tumor tissues and tumor-associated macrophages (TAMs) in human subjects. Notably, Siglec-15 is up-regulated across various cancer types. As a result, Siglec-15 has attracted significant attention due to its exclusive nature concerning PD-L1 expression, suggesting its role in immune evasion in patients lacking PD-L1. Siglec-15 predominantly appears in certain populations and can promote tumor development by repressing T lymphocyte activation and proliferation, thereby facilitating tumor cell immune escape. Furthermore, Siglec-15 is implicated in osteoclast differentiation and bone remodeling, indicating that it is a promising target for next-generation cancer immunotherapies. Additionally, Siglec-15 can modulate immune responses to microbial infections. The current treatment strategies for hematological conditions predominantly include conventional intensive chemotherapy and transplantation methods. However, emerging immunotherapeutic approaches are increasingly recognized for their overall effectiveness, indicating that specific molecular targets should be identified. The expression of Siglec-15 within tumor cells may indicate a novel pathway for treating hematological malignancies. In this study, the biological attributes, expression patterns, and pathogenic mechanisms of Siglec-15 across various diseases were reviewed. The role of Siglec-15 in the pathogenesis and laboratory diagnosis of hematological disorders was also evaluated.
Collapse
Affiliation(s)
- Yujia Fan
- Baotou Medical College of Inner Mongolia University of Science and Technology, Baotou, Inner Mongolia, China
| | - Liangliang Sun
- Clinical Laboratory Medicine Centre, Inner Mongolia Autonomous Region People’s Hospital, Hohhot, Inner Mongolia, China
| | - Juan He
- Clinical Laboratory Medicine Centre, Inner Mongolia Autonomous Region People’s Hospital, Hohhot, Inner Mongolia, China
| | - Yuetong Chen
- Clinical Laboratory Medicine Centre, Inner Mongolia Autonomous Region People’s Hospital, Hohhot, Inner Mongolia, China
| | - Hongli Ma
- Baotou Medical College of Inner Mongolia University of Science and Technology, Baotou, Inner Mongolia, China
| | - Haitao Ding
- Clinical Laboratory Medicine Centre, Inner Mongolia Autonomous Region People’s Hospital, Hohhot, Inner Mongolia, China
| |
Collapse
|
42
|
Ali F, Iqbal A, Azhar I, Qayyum A, Hassan SA, Hasan MSA, Jawi M, Hassan HM, Al-Emam A, Sajid M. Exploring a novel four-gene system as a diagnostic and prognostic biomarker for triple-negative breast cancer, using clinical variables. Comput Biol Chem 2024; 113:108247. [PMID: 39427606 DOI: 10.1016/j.compbiolchem.2024.108247] [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: 08/03/2024] [Revised: 09/25/2024] [Accepted: 10/09/2024] [Indexed: 10/22/2024]
Abstract
Triple-negative breast cancer (TNBC) is a subtype of breast cancer with a poor prognosis. This research aims to find real hub genes for prognostic biomarkers of TNBC therapy. The GEO datasets GSE27447 and GSE233242 were analyzed using R package limma to explore DEGs. The PPI was generated using the STRING database. Cytoscape software plug-ins were used to screen the hub genes. Using the DAVID database, GO functional enrichment and KEGG pathway enrichment analysis were performed. Different online expression databases were employed to investigate the functions of real hub genes in tumor driving, diagnosis, and prognosis in TNBC patients with various clinicopathologic characteristics. A total of one hundred DEGs were identified between both datasets. The seven hub genes were identified after the topological parameter analysis of the PPI network. The KEGG pathway and GO analysis suggest that four genes (PSMB1, PSMC1, PSMF1, and PSMD8) are highly enriched in proteasome and were finally considered as real hub genes. Additionally, the expression analysis demonstrated that hub genes were notably up-regulated in TNBC patients compared to controls. Furthermore, correlational analyses revealed the positive and negative correlations among the expression of the real hub genes and various ancillary data, including tumor purity, promoter methylation status, overall survival (OS), genetic alterations, infiltration of CD8+ T and CD4+ immune cells, and a few more, across TNBC samples. Finally, our analysis identified a couple of significant chemotherapeutic drugs, miRNAs and transcription factors (TFS) with intriguing curative potential. In conclusion, we identified four real hub genes as novel biomarkers to overcome heterogenetic-particular challenges in diagnosis, prognosis, and therapy for TNBC patients.
Collapse
Affiliation(s)
- Faisal Ali
- Department of Biotechnology, Faculty of Life Sciences, University of Okara, Okara, Punjab 56300, Pakistan
| | - Azhar Iqbal
- Department of Biotechnology, Faculty of Life Sciences, University of Okara, Okara, Punjab 56300, Pakistan
| | - Iqra Azhar
- Department of Biotechnology, Faculty of Life Sciences, University of Okara, Okara, Punjab 56300, Pakistan
| | - Adiba Qayyum
- Department of Biotechnology, Faculty of Life Sciences, University of Okara, Okara, Punjab 56300, Pakistan
| | - Syed Ali Hassan
- Department of Biotechnology, Faculty of Life Sciences, University of Okara, Okara, Punjab 56300, Pakistan
| | - Md Sakib Al Hasan
- Department of Pharmacy, Bangabandhu Sheikh Mujibur Rahman Science And Technology University, Gopalgonj, Dhaka 8100, Bangladesh; Bioinformatics and Drug Innovation Laboratory, BioLuster Research Center Ltd., Gopalganj, 8100, Dhaka, Bangladesh.
| | - Motasim Jawi
- Department of Basic Medical Sciences, College of Medicine, University of Jeddah, Jeddah, Saudi Arabia
| | - Hesham M Hassan
- Department of Pathology, College of Medicine, King Khalid University, Asir 61421, Saudi Arabia
| | - Ahmed Al-Emam
- Department of Pathology, College of Medicine, King Khalid University, Asir 61421, Saudi Arabia.
| | - Muhammad Sajid
- Department of Biotechnology, Faculty of Life Sciences, University of Okara, Okara, Punjab 56300, Pakistan.
| |
Collapse
|
43
|
Gupta S, Dev J R A, Prakash Prasad C, Kumar A, Kumar Ghosh S. A potent Bioorganic azapodophyllotoxin derivative Suppresses tumor Progression in Triple negative breast Cancer: An Insight into its Inhibitory effect on tubulin polymerization and Disruptive effect on microtubule assembly. Bioorg Chem 2024; 153:107839. [PMID: 39326339 DOI: 10.1016/j.bioorg.2024.107839] [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: 08/07/2024] [Revised: 09/18/2024] [Accepted: 09/19/2024] [Indexed: 09/28/2024]
Abstract
Triple negative breast cancer (TNBC) has long been a challenging disease owing to its high aggressive behaviour, poor prognosis and its limited treatment options. The growing demand of new therapeutics against TNBC enables us to examine the therapeutic efficiency of an emerging class of anticancer compounds, azapodophyllotoxin derivative (HTDQ), a nitrogen analogue of podophyllotoxin, using different biochemical, spectroscopic and computational approaches. The anticancer activities of HTDQ are studied by performing MTT assay in a dose depended manner on Triple negative breast cancer cells using MDA-MB-468 and MDA-MB-231 cell lines with IC50 value 937 nM and 1.13 µM respectively while demonstrating minimal effect on normal epithelial cells. The efficacy of HTDQ was further tested in 3D tumour spheroids formed by the human TNBC cell line MDA-MB468 and also the murine MMTV positive TNBC cell line 4 T1. The shrinkage that observed in the tumor spheroid clearly indicates that HTDQ remarkably decreases the growth of tumor spheroid thereby affirming its cytotoxicity. The 2D cell viability assay shows significant morphological alteration that possibly caused by the cytoskeleton disturbances. Hence the binding interaction of HTDQ with cytoskeleton protein tubulin, its effect on tubulin polymerisation as well as depolymerisation of preformed microtubules along with the conformational alternation in the protein itself have been investigated in detail. Moreover, the apoptotic effects of HTDQ have been examined using a range of apoptotic markers. HTDQ-treated cancer cells showed increased expression of cleaved PARP-1 and pro-caspase-3, suggesting activation of the apoptosis process. HTDQ also upregulated pro-apoptotic Bax expression while inhibiting anti-apoptotic Bcl2 expression, supporting its ability to induce apoptosis in cancer cells. Hence the consolidated biochemical and spectroscopic research described herein may provide enormous information to use azapodophyllotoxin as promising anticancer therapeutics for TNBC cells.
Collapse
Affiliation(s)
- Smruti Gupta
- Department of Chemistry, Visvesvaraya National Institute of Technology, Nagpur 440010, Maharashtra, India
| | - Arundhathi Dev J R
- Department of Medical Oncology (Laboratory), Dr. BRA IRCH, All India Institute of Medical Sciences (AIIMS), New Delhi 110029, India
| | - Chandra Prakash Prasad
- Department of Medical Oncology (Laboratory), Dr. BRA IRCH, All India Institute of Medical Sciences (AIIMS), New Delhi 110029, India
| | - Ajay Kumar
- School of Science, Technology and Environment, Universidad Ana G. Mendez, Cupey Campus, PO Box 21150, San Juan, PR 00928-1150, United States
| | - Sujit Kumar Ghosh
- Department of Chemistry, Visvesvaraya National Institute of Technology, Nagpur 440010, Maharashtra, India.
| |
Collapse
|
44
|
Zhang N, Li Z, Liu Y, Shi X, Shi D, Li Y, Si X, Xun Z, Shao J, Zhao H, Wang H. Management and treatment of severe immune-related hepatotoxicity based on clinical and pathological characteristics. Hepatol Int 2024; 18:1770-1780. [PMID: 38954360 PMCID: PMC11632075 DOI: 10.1007/s12072-024-10688-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Accepted: 04/21/2024] [Indexed: 07/04/2024]
Abstract
BACKGROUND The management of severe immune-related hepatotoxicity (irH) needs to be further optimized. This study aims to analyze the clinical characteristics of severe irH; improve the therapeutic strategy, especially salvage treatment in steroid-refractory irH; and determine the safety of immune checkpoint inhibitor (ICPi)-rechallenge. METHODS This multicenter retrospective study included patients who developed severe irH and those without irH after immunotherapy between May 2019 and June 2023. Propensity score matching was used to match these two cohorts with similar baseline characteristics. RESULTS Among 5,326 patients receiving ICPis, 51 patients developed severe irH. irH occurred after a median duration of 36 days and a median of two doses after the first ICPi administration. Patients receiving PD-L1 inhibitors faced a lower risk of developing severe irH. A higher dose of glucocorticoids (GCS) was administered to grade 4 irH than grade 3 irH. For steroid-sensitive patients, grade 4 irH individuals received a higher dosage of GCS than those with grade 3 irH, with no difference in time to resolution. Meanwhile, a significantly higher dose of GCS plus immunosuppression was needed in the steroid-refractory group. Liver biopsy of the steroid-refractory patients exhibited heterogeneous histological features. Twelve patients were retreated with ICPi. No irH reoccurred after a median follow-up of 9.3 months. CONCLUSION irH requires multidimensional evaluation. PD-L1 inhibitors correlated with a lower risk of severe irH. Grade 4 irH demands a higher dose of GCS than recommended. Pathology may guide the salvage treatment for steroid-refractory irH. ICPi rechallenge in severe irH is feasible and safe.
Collapse
Affiliation(s)
- Nan Zhang
- Department of Liver Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, No.1 Shuaifuyuan, Beijing, 100730, China
| | - Zhaohui Li
- Division of Pulmonary and Critical Care Medicine, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, No. 1 Shuaifuyuan, Beijing, 100730, China
| | - Yutao Liu
- 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
| | - Xiaohua Shi
- Department of Pathology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Di Shi
- Department of Emergency Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Yue Li
- Department of Digestive Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Xiaoyan Si
- Division of Pulmonary and Critical Care Medicine, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, No. 1 Shuaifuyuan, Beijing, 100730, China
| | - Ziyu Xun
- Department of Liver Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, No.1 Shuaifuyuan, Beijing, 100730, China
| | - Jing Shao
- Department of Liver Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, No.1 Shuaifuyuan, Beijing, 100730, China
| | - Haitao Zhao
- Department of Liver Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, No.1 Shuaifuyuan, Beijing, 100730, China.
| | - Hanping Wang
- Division of Pulmonary and Critical Care Medicine, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, No. 1 Shuaifuyuan, Beijing, 100730, China.
| |
Collapse
|
45
|
Jin M, Fang J, Peng J, Wang X, Xing P, Jia K, Hu J, Wang D, Ding Y, Wang X, Li W, Chen Z. PD-1/PD-L1 immune checkpoint blockade in breast cancer: research insights and sensitization strategies. Mol Cancer 2024; 23:266. [PMID: 39614285 PMCID: PMC11605969 DOI: 10.1186/s12943-024-02176-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Accepted: 11/13/2024] [Indexed: 12/01/2024] Open
Abstract
Immunotherapy targeting programmed cell death-1 (PD-1) and PD-L1 immune checkpoints has reshaped treatment paradigms across several cancers, including breast cancer. Combining PD-1/PD-L1 immune checkpoint blockade (ICB) with chemotherapy has shown promising efficacy in both early and metastatic triple-negative breast cancer, although only a subset of patients experiences durable responses. Identifying responders and optimizing immune drug selection are therefore critical. The effectiveness of PD-1/PD-L1 immunotherapy depends on both tumor-intrinsic factors and the extrinsic cell-cell interactions within the tumor microenvironment (TME). This review systematically summarizes the key findings from clinical trials of ICBs in breast cancer and examines the mechanisms underlying PD-L1 expression regulation. We also highlight recent advances in identifying potential biomarkers for PD-1/PD-L1 therapy and emerging evidence of TME alterations following treatment. Among these, the quantity, immunophenotype, and spatial distribution of tumor-infiltrating lymphocytes stand out as promising biomarkers. Additionally, we explore strategies to enhance the effectiveness of ICBs in breast cancer, aiming to support the development of personalized treatment approaches tailored to the unique characteristics of each patient's tumor.
Collapse
Affiliation(s)
- Menglei Jin
- Department of Breast Surgery (Surgical Oncology), Second Affiliated Hospital, Zhejiang University School of Medicine, 88 Jiefang Road, Hangzhou, Zhejiang, China
- Key Laboratory of Tumor Microenvironment and Immune Therapy of Zhejiang Province, Hangzhou, China
| | - Jun Fang
- Department of Radiation Oncology, Zhejiang Cancer Hospital, Hangzhou, Zhejiang, China
| | - Junwen Peng
- Department of General Surgery, The First People's Hospital of Jiande, Hangzhou, China
| | - Xintian Wang
- Department of General Surgery, The Second People's Hospital of Tongxiang, Jiaxing, Zhejiang, China
| | - Ping Xing
- Department of Breast Surgery (Surgical Oncology), Second Affiliated Hospital, Zhejiang University School of Medicine, 88 Jiefang Road, Hangzhou, Zhejiang, China
- Key Laboratory of Tumor Microenvironment and Immune Therapy of Zhejiang Province, Hangzhou, China
| | - Kunpeng Jia
- Department of Breast Surgery (Surgical Oncology), Second Affiliated Hospital, Zhejiang University School of Medicine, 88 Jiefang Road, Hangzhou, Zhejiang, China
- Key Laboratory of Tumor Microenvironment and Immune Therapy of Zhejiang Province, Hangzhou, China
| | - Jianming Hu
- Department of Breast Surgery (Surgical Oncology), Second Affiliated Hospital, Zhejiang University School of Medicine, 88 Jiefang Road, Hangzhou, Zhejiang, China
- Key Laboratory of Tumor Microenvironment and Immune Therapy of Zhejiang Province, Hangzhou, China
| | - Danting Wang
- Department of Breast Surgery (Surgical Oncology), Second Affiliated Hospital, Zhejiang University School of Medicine, 88 Jiefang Road, Hangzhou, Zhejiang, China
- Key Laboratory of Tumor Microenvironment and Immune Therapy of Zhejiang Province, Hangzhou, China
| | - Yuxin Ding
- Department of Breast Surgery (Surgical Oncology), Second Affiliated Hospital, Zhejiang University School of Medicine, 88 Jiefang Road, Hangzhou, Zhejiang, China
- Key Laboratory of Tumor Microenvironment and Immune Therapy of Zhejiang Province, Hangzhou, China
| | - Xinyu Wang
- Department of Radiation Oncology, Zhejiang Cancer Hospital, Hangzhou, Zhejiang, China
| | - Wenlu Li
- Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, China.
| | - Zhigang Chen
- Department of Breast Surgery (Surgical Oncology), Second Affiliated Hospital, Zhejiang University School of Medicine, 88 Jiefang Road, Hangzhou, Zhejiang, China.
- Key Laboratory of Tumor Microenvironment and Immune Therapy of Zhejiang Province, Hangzhou, China.
| |
Collapse
|
46
|
Arulraj T, Wang H, Deshpande A, Varadhan R, Emens LA, Jaffee EM, Fertig EJ, Santa-Maria CA, Popel AS. Virtual patient analysis identifies strategies to improve the performance of predictive biomarkers for PD-1 blockade. Proc Natl Acad Sci U S A 2024; 121:e2410911121. [PMID: 39467131 PMCID: PMC11551325 DOI: 10.1073/pnas.2410911121] [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: 05/31/2024] [Accepted: 09/24/2024] [Indexed: 10/30/2024] Open
Abstract
Patients with metastatic triple-negative breast cancer (TNBC) show variable responses to PD-1 inhibition. Efficient patient selection by predictive biomarkers would be desirable but is hindered by the limited performance of existing biomarkers. Here, we leveraged in silico patient cohorts generated using a quantitative systems pharmacology model of metastatic TNBC, informed by transcriptomic and clinical data, to explore potential ways to improve patient selection. We evaluated and quantified the performance of 90 biomarker candidates, including various cellular and molecular species, at different cutoffs by a cutoff-based biomarker testing algorithm combined with machine learning-based feature selection. Combinations of pretreatment biomarkers improved the specificity compared to single biomarkers at the cost of reduced sensitivity. On the other hand, early on-treatment biomarkers, such as the relative change in tumor diameter from baseline measured at two weeks after treatment initiation, achieved remarkably higher sensitivity and specificity. Further, blood-based biomarkers had a comparable ability to tumor- or lymph node-based biomarkers in identifying a subset of responders, potentially suggesting a less invasive way for patient selection.
Collapse
Affiliation(s)
- Theinmozhi Arulraj
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD21205
| | - Hanwen Wang
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD21205
| | - Atul Deshpande
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD21205
- Convergence Institute, Johns Hopkins University School of Medicine, Baltimore, MD21205
- Bloomberg Kimmel Immunology Institute, Johns Hopkins University School of Medicine, Baltimore, MD21205
| | - Ravi Varadhan
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD21205
| | | | - Elizabeth M. Jaffee
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD21205
- Convergence Institute, Johns Hopkins University School of Medicine, Baltimore, MD21205
- Bloomberg Kimmel Immunology Institute, Johns Hopkins University School of Medicine, Baltimore, MD21205
| | - Elana J. Fertig
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD21205
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD21205
- Convergence Institute, Johns Hopkins University School of Medicine, Baltimore, MD21205
- Bloomberg Kimmel Immunology Institute, Johns Hopkins University School of Medicine, Baltimore, MD21205
- Department of Applied Mathematics and Statistics, Whiting School of Engineering, Johns Hopkins University, Baltimore, MD21218
| | - Cesar A. Santa-Maria
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD21205
| | - Aleksander S. Popel
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD21205
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD21205
| |
Collapse
|
47
|
Wu S, Ge A, Deng X, Liu L, Wang Y. Evolving immunotherapeutic solutions for triple-negative breast carcinoma. Cancer Treat Rev 2024; 130:102817. [PMID: 39154410 DOI: 10.1016/j.ctrv.2024.102817] [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: 05/16/2024] [Revised: 07/29/2024] [Accepted: 08/15/2024] [Indexed: 08/20/2024]
Abstract
Triple-negative breast carcinoma (TNBC) remains a formidable clinical hurdle owing to its high aggressiveness and scant therapeutic options. Nonetheless, the evolving landscape of immunotherapeutic strategies opens up promising avenues for tackling this hurdle. This review discusses the advancing immunotherapy for TNBC, accentuating personalized interventions due to tumor microenvironment (TME) diversity. Immune checkpoint inhibitors (ICIs) hold pivotal significance, both as single-agent therapies and when administered alongside cytotoxic agents. Moreover, the concurrent inhibition of multiple immune checkpoints represents a potent approach to augment the efficacy of cancer immunotherapy. Synergistic effects have been observed when ICIs are combined with targeted treatments like PARP inhibitors, anti-angiogenics, and ADCs (antibody-drug conjugates). Emerging tactics include tumor vaccines, cellular immunotherapy, and oncolytic viruses, leveraging the immune system's ability for selective malignant cell destruction. This review offers an in-depth examination of the diverse landscape of immunotherapy development for TNBC, furnishing meticulous insights into various advancements within this field. In addition, immunotherapeutic interventions offer hope for TNBC, needing further research for optimization.
Collapse
Affiliation(s)
- Shiting Wu
- Department of Galactophore, the First Hospital of Hunan University of Chinese Medicine, Changsha, Hunan Province 410007, China
| | - Anqi Ge
- Department of Galactophore, the First Hospital of Hunan University of Chinese Medicine, Changsha, Hunan Province 410007, China
| | - Xianguang Deng
- Department of Galactophore, the First Hospital of Hunan University of Chinese Medicine, Changsha, Hunan Province 410007, China
| | - Lifang Liu
- Department of Galactophore, the First Hospital of Hunan University of Chinese Medicine, Changsha, Hunan Province 410007, China
| | - Yue Wang
- Department of Galactophore, the First Hospital of Hunan University of Chinese Medicine, Changsha, Hunan Province 410007, China.
| |
Collapse
|
48
|
Wang Y, Niu B, Tian Y, Lan H, Zhou Z, Li Y, Zhao S, Zhang Y, Yang C, Kong L, Zhang Z. Mitoxantrone Combined with Engineered TRAIL-Nanovesicles for Enhanced Cancer Immunotherapy Via Converting Apoptosis into Pyroptosis. Adv Healthc Mater 2024; 13:e2401723. [PMID: 39049538 DOI: 10.1002/adhm.202401723] [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: 05/09/2024] [Revised: 07/15/2024] [Indexed: 07/27/2024]
Abstract
Pyroptosis, a highly inflammatory form of programmed cell death, has emerged as a promising target for cancer immunotherapy. However, in the context of pyroptosis execution, while both caspase-3 and GSDME are essential, it is noteworthy that GSDME is frequently under-expressed in cold tumors. To overcome this limitation, engineered cellular nanovesicles (NVs) presenting TRAIL on their membranes (NVTRAIL) are developed to trigger the upregulation of cleaved caspase-3. When strategically combined with the chemotherapeutic agent mitoxantrone (MTO), known for its ability to enhance GSDME expression, MTO@NVTRAIL can convert cancer cells from apoptosis into pyroptosis, inhibit the tumor growth and metastasis successfully in primary tumor. The microparticles released by pyroptotic tumor cells also exhibited certain cytotoxicity against other tumor cells. In addition, tumor cells exposed to the combination treatment of MTO@NVTRAIL in vitro have also demonstrated potential utility as a novel form of vaccine for cancer immunotherapy. Flow analysis of the tumor microenvironment and draining lymph nodes reveals an increased proportion of matured dendritic cells and activation of T cells. In summary, the research provided a reference and alternative approach to induce cancer pyroptosis for clinical antitumor therapy based on engineered cellular nanovesicles and chemotherapy.
Collapse
Affiliation(s)
- Yi Wang
- Tongji School of Pharmacy, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Boning Niu
- Tongji School of Pharmacy, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Yinmei Tian
- Tongji School of Pharmacy, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Hongbing Lan
- Tongji School of Pharmacy, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Zhanhao Zhou
- Liyuan Hospital, Huazhong University of Science and Technology, Wuhan, 430077, China
| | - Yang Li
- Tongji School of Pharmacy, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Siyu Zhao
- Tongji School of Pharmacy, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Yu Zhang
- Tongji School of Pharmacy, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Conglian Yang
- Tongji School of Pharmacy, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Li Kong
- Tongji School of Pharmacy, Huazhong University of Science and Technology, Wuhan, 430030, China
- National Engineering Research Center for Nanomedicine, Huazhong University of Science and Technology, Wuhan, 430030, China
- Hubei Engineering Research Centre for Novel Drug Delivery System, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Zhiping Zhang
- Tongji School of Pharmacy, Huazhong University of Science and Technology, Wuhan, 430030, China
- National Engineering Research Center for Nanomedicine, Huazhong University of Science and Technology, Wuhan, 430030, China
- Hubei Engineering Research Centre for Novel Drug Delivery System, Huazhong University of Science and Technology, Wuhan, 430030, China
| |
Collapse
|
49
|
Nandi D, Sharma D. Integrating immunotherapy with conventional treatment regime for breast cancer patients- an amalgamation of armamentarium. Front Immunol 2024; 15:1477980. [PMID: 39555066 PMCID: PMC11563812 DOI: 10.3389/fimmu.2024.1477980] [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: 08/08/2024] [Accepted: 10/08/2024] [Indexed: 11/19/2024] Open
Abstract
Immunotherapy stands as the frontrunner in treatment strategies imparting efficient remission in various types of cancer. In fact, emerging breakthroughs with immune checkpoint inhibitors (ICI) in a spectrum of cancers have evoked interest in research related to the potential effects of immunotherapy in breast cancer patients. A major challenge with breast cancer is the molecular heterogeneity that limits the efficacy of many therapeutic regimes. Clinical trials have shown favorable clinical outcomes with immunotherapeutic options in some subtypes of breast cancer. However, ICI monotherapy may not be sufficient for all breast cancer patients, emphasizing the need for combinatorial approaches. Ongoing research is focused on untangling the interplay of ICI with established as well as novel anticancer therapeutic regimens in preclinical models of breast cancer. Our review will analyze the existing research regarding the mechanisms and clinical impact of immunotherapy for the treatment of breast cancer. We shall evaluate the role of immune cell modulation for improved therapeutic response in breast cancer patients. This review will provide collated evidences about the current clinical trials that are testing out the implications of immunotherapy in conjunction with traditional treatment modalities in breast cancer and summarize the potential future research directions in the field. In addition, we shall underline the recent findings related to microbiota modulation as a key regulator of immune therapy response in cancer patients and its plausible applications in breast cancer.
Collapse
Affiliation(s)
- Deeptashree Nandi
- Department of Oncology, Johns Hopkins University School of Medicine and Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD, United States
| | - Dipali Sharma
- Department of Oncology, Johns Hopkins University School of Medicine and Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD, United States
| |
Collapse
|
50
|
Sawant S, Naik GG, Sahu AN, Jagtap VA. Understanding the chemistry & pharmacology of antibody-drug conjugates in triple-negative breast cancer with special reference to exatecan derivatives. Med Oncol 2024; 41:301. [PMID: 39460856 DOI: 10.1007/s12032-024-02542-y] [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: 09/05/2024] [Accepted: 10/15/2024] [Indexed: 10/28/2024]
Abstract
In the spectrum of breast malignancies, triple-negative breast cancer is the most widely spreading subtype of breast cancer due to a low availability of therapeutic remedies. Recently, antibody-drug conjugates dramatically resolved the landscape for the treatment of triple-negative breast cancer. This review mainly focuses on the chemistry, structure, mechanism of action, and role of antibody-drug conjugates in triple-negative breast cancer. Datopotecan Deruxtecan (Dato-DXd) is a new-generation ADC showing encouraging results for TNBC. In this review, we have also emphasized TROP-2-directed Datopotamab deruxtecan ADCs to treat triple-negative breast cancer, its synthesis, mechanism of action, pharmacokinetics, pharmacodynamics, adverse events, and their ongoing clinical trials.
Collapse
Affiliation(s)
- Sanjana Sawant
- Department of Pharmaceutical Chemistry, Yashwantrao Bhonsale College of Pharmacy, Affiliated to Mumbai University, Sawantwadi, 416510, India
| | - Gaurav Gopal Naik
- Department of Pharmaceutical Chemistry, Yashwantrao Bhonsale College of Pharmacy, Affiliated to Mumbai University, Sawantwadi, 416510, India.
| | | | - Vijay A Jagtap
- Department of Pharmaceutical Chemistry, Yashwantrao Bhonsale College of Pharmacy, Affiliated to Mumbai University, Sawantwadi, 416510, India
| |
Collapse
|