1
|
Rajak P. Immune checkpoint inhibitors: From friend to foe. Toxicol Rep 2025; 14:102033. [PMID: 40353246 PMCID: PMC12063143 DOI: 10.1016/j.toxrep.2025.102033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2024] [Revised: 04/18/2025] [Accepted: 04/20/2025] [Indexed: 05/14/2025] Open
Abstract
Immune checkpoints are crucial in regulating the activation of cell-mediated and humoral immune responses. However, cancer cells hijack this mechanism to evade the immune surveillance and anti-cancer response. Typically, receptors like PD-1 and CTLA4, expressed on immune cells, prevent the activation and differentiation of T cells. They also inhibit the development of autoimmune reactions. However, ligands such as PD-L1 for the receptor PD-1 are also expressed on the surface of cancer cells that help prevent the activation of anti-cancer immune responses by blocking the signalling pathways mediated by PD-1 and CTLA4. Immune checkpoint inhibitors (ICIs) have promising therapeutic efficacy for treating several cancers by activating T cells and their differentiation into effector cells against tumours. Nonetheless, hyperactivated immune cells usually contribute to detrimental issues, also known as immune-related adverse effects (IrAE). IrAEs have been observed in multiple organs, leading to neurological issues, colitis, endocrine dysfunction, renal issues, hepatitis, pneumonitis, and dermatitis. The interplay between hyperactivated T cells and Treg cells helps in orchestrating the development of autoimmunity. Moreover, the crosstalk between proinflammatory interleukins and the development of autoantibodies also mediates the multiorgan effects of ICIs in cancer patients. IrAEs are generally managed by terminating the ICI therapy, reducing the ICI dose, and by using corticosteroids to subvert inflammation. Therefore, the present review aims to delineate the impacts of ICIs on the development of autoimmune diseases and inflammatory outcomes in cancer patients. In addition, mechanistic insight involving immune cells, cytokines, and autoantibodies for ICI-mediated IrAEs will also be discussed with updated findings in this field.
Collapse
Affiliation(s)
- Prem Rajak
- Toxicology Research Laboratory, Department of Animal Science, Kazi Nazrul University, Asansol, West Bengal, India
| |
Collapse
|
2
|
Demir T, Moloney C, Mahalingam D. Threading the Needle: Navigating Novel Immunotherapeutics in Pancreatic Ductal Adenocarcinoma. Cancers (Basel) 2025; 17:715. [PMID: 40075563 PMCID: PMC11898821 DOI: 10.3390/cancers17050715] [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: 01/03/2025] [Revised: 02/08/2025] [Accepted: 02/17/2025] [Indexed: 03/14/2025] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a lethal malignancy with a poor prognosis. Currently, chemotherapy is the only option for most patients with advanced-stage PDAC. Further, conventional immunotherapies and targeted therapies improve survival outcomes only in rare PDAC patient subgroups. To date, combinatory immunotherapeutic strategies to overcome the immune-hostile PDAC tumor microenvironment (TME) have resulted in limited efficacy in clinical studies. However, efforts are ongoing to develop new treatment strategies for patients with PDAC with the evolving knowledge of the TME, molecular characterization, and immune resistance mechanisms. Further, the growing arsenal of various immunotherapeutic agents, including novel classes of immune checkpoint inhibitors and oncolytic, chimeric antigen receptor T cell, and vaccine therapies, reinforces these efforts. This review will focus on the place of immunotherapy and future possible strategies in PDAC.
Collapse
Affiliation(s)
| | | | - Devalingam Mahalingam
- Developmental Therapeutics, Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA; (T.D.); (C.M.)
| |
Collapse
|
3
|
Fujimoto D, Shibaki R, Kimura K, Haratani K, Tamiya M, Kijima T, Sato Y, Hata A, Yokoyama T, Taniguchi Y, Uchida J, Tanaka H, Furuya N, Miura S, Onishi MI, Sakata S, Miyauchi E, Yamamoto N, Koh Y, Akamatsu H. Identification of key gene signatures for predicting chemo-immunotherapy efficacy in extensive-stage small-cell lung cancer using machine learning. Lung Cancer 2025; 199:108079. [PMID: 39787635 DOI: 10.1016/j.lungcan.2024.108079] [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: 12/28/2024] [Accepted: 12/31/2024] [Indexed: 01/12/2025]
Abstract
OBJECTIVES The lack of definitive biomarkers presents a significant challenge for chemo-immunotherapy in extensive-stage small-cell lung cancer (ES-SCLC). We aimed to identify key genes associated with chemo-immunotherapy efficacy in ES-SCLC through comprehensive gene expression analysis using machine learning (ML). METHODS A prospective multicenter cohort of patients with ES-SCLC who received first-line chemo-immunotherapy was analyzed. RNA sequencing was performed on tumor samples to assess gene expression levels. ML techniques were applied to identify key gene features associated with treatment efficacy. A panel of genes was then developed and validated using the nCounter system, and the model's performance in predicting 180-day progression-free survival (PFS) was evaluated. RESULTS A total of 93 patients were included in the analysis. ML-based gene selection identified a gene set comprising 83 genes from the comprehensive gene expression data. An nCounter panel was developed using these genes, and an ML model was developed based on their expression levels. In the validation set, the model achieved an accuracy of 0.93, precision of 1.00, a true positive rate of 0.83, and a true negative rate of 1.00. PFS was significantly longer in the high-efficacy group than in the low-efficacy group in the validation set (P < 0.001). CONCLUSIONS These findings provide a foundation for biomarker development in ES-SCLC and highlight the potential of this method as a cost-effective, simple, and rapid tool for assessing treatment efficacy in clinical practice.
Collapse
Affiliation(s)
- Daichi Fujimoto
- Internal Medicine III, Wakayama Medical University, Wakayama, Japan; Department of Respiratory Medicine and Hematology, Hyogo Medical University, Nishinomiya, Japan.
| | - Ryota Shibaki
- Internal Medicine III, Wakayama Medical University, Wakayama, Japan
| | | | - Koji Haratani
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Motohiro Tamiya
- Department of Thoracic Oncology, Osaka International Cancer Institute, Osaka, Japan
| | - Takashi Kijima
- Department of Respiratory Medicine and Hematology, Hyogo Medical University, Nishinomiya, Japan
| | - Yuki Sato
- Department of Respiratory Medicine, Kobe City Medical Center General Hospital, Kobe, Japan
| | - Akito Hata
- Division of Thoracic Oncology, Kobe Minimally Invasive Cancer Center, Kobe, Japan
| | - Toshihide Yokoyama
- Department of Respiratory Medicine, Kurashiki Central Hospital, Kurashiki, Japan
| | - Yoshihiko Taniguchi
- Department of Internal Medicine, NHO Kinki Chuo Chest Medical Center, Sakai, Japan
| | - Junji Uchida
- Department of Respiratory Medicine, Osaka General Medical Center, Osaka, Japan
| | - Hisashi Tanaka
- Department of Respiratory Medicine, Hirosaki University Graduate School of Medicine, Aomori, Japan
| | - Naoki Furuya
- Division of Respiratory Medicine, Department of Internal Medicine, St. Marianna University School of Medicine, Kawasaki, Japan
| | - Satoru Miura
- Department of Internal Medicine, Niigata Cancer Center Hospital, Niigata, Japan
| | | | - Shinya Sakata
- Department of Respiratory Medicine, Kumamoto University Hospital, Kumamoto, Japan
| | - Eisaku Miyauchi
- Department of Respiratory Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Nobuyuki Yamamoto
- Internal Medicine III, Wakayama Medical University, Wakayama, Japan; Center for Biomedical Sciences, Wakayama Medical University, Wakayama, Japan
| | - Yasuhiro Koh
- Internal Medicine III, Wakayama Medical University, Wakayama, Japan; Center for Biomedical Sciences, Wakayama Medical University, Wakayama, Japan
| | - Hiroaki Akamatsu
- Internal Medicine III, Wakayama Medical University, Wakayama, Japan
| |
Collapse
|
4
|
Yin X, Song Y, Deng W, Blake N, Luo X, Meng J. Potential predictive biomarkers in antitumor immunotherapy: navigating the future of antitumor treatment and immune checkpoint inhibitor efficacy. Front Oncol 2024; 14:1483454. [PMID: 39655071 PMCID: PMC11625675 DOI: 10.3389/fonc.2024.1483454] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Accepted: 11/04/2024] [Indexed: 12/12/2024] Open
Abstract
Immune checkpoint inhibitors (ICIs) have revolutionized cancer treatment modality, offering promising outcomes for various malignancies. However, the efficacy of ICIs varies among patients, highlighting the essential need of accurate predictive biomarkers. This review synthesizes the current understanding of biomarkers for ICI therapy, and discusses the clinical utility and limitations of these biomarkers in predicting treatment outcomes. It discusses three US Food and Drug Administration (FDA)-approved biomarkers, programmed cell death ligand 1 (PD-L1) expression, tumor mutational burden (TMB), and microsatellite instability (MSI), and explores other potential biomarkers, including tumor immune microenvironment (TIME)-related signatures, human leukocyte antigen (HLA) diversity, non-invasive biomarkers such as circulating tumor DNA (ctDNA), and combination biomarker strategies. The review also addresses multivariable predictive models integrating multiple features of patients, tumors, and TIME, which could be a promising approach to enhance predictive accuracy. The existing challenges are also pointed out, such as the tumor heterogeneity, the inconstant nature of TIME, nonuniformed thresholds and standardization approaches. The review concludes by emphasizing the importance of biomarker research in realizing the potential of personalized immunotherapy, with the goal of improving patient selection, treatment strategies, and overall outcomes in cancer treatment.
Collapse
Affiliation(s)
- Xiangyu Yin
- Department of Biological Sciences, School of Science, AI University Research Centre, Xi’an Jiaotong-Liverpool University, Suzhou, China
- Institute of Biomedical Research, Regulatory Mechanism and Targeted Therapy for Liver Cancer Shiyan Key Laboratory, Hubei Provincial Clinical Research Center for Precise Diagnosis and Treatment of Liver Cancer, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei, China
- Institute of Infection, Veterinary & Ecological Sciences, University of Liverpool, Liverpool, United Kingdom
- Jiangsu Simcere Diagnostics Co., Ltd., The State Key Laboratory of Neurology and Oncology Drug Development, Nanjing, China
| | - Yunjie Song
- Jiangsu Simcere Diagnostics Co., Ltd., The State Key Laboratory of Neurology and Oncology Drug Development, Nanjing, China
| | - Wanglong Deng
- Jiangsu Simcere Diagnostics Co., Ltd., The State Key Laboratory of Neurology and Oncology Drug Development, Nanjing, China
| | - Neil Blake
- Institute of Infection, Veterinary & Ecological Sciences, University of Liverpool, Liverpool, United Kingdom
| | - Xinghong Luo
- Jiangsu Simcere Diagnostics Co., Ltd., The State Key Laboratory of Neurology and Oncology Drug Development, Nanjing, China
| | - Jia Meng
- Department of Biological Sciences, School of Science, AI University Research Centre, Xi’an Jiaotong-Liverpool University, Suzhou, China
- Institute of Biomedical Research, Regulatory Mechanism and Targeted Therapy for Liver Cancer Shiyan Key Laboratory, Hubei Provincial Clinical Research Center for Precise Diagnosis and Treatment of Liver Cancer, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei, China
- Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, United Kingdom
| |
Collapse
|
5
|
Nunes-Xavier CE. Editorial: Co-inhibitory immune checkpoint proteins as biomarkers and therapeutic targets in cancer. Transl Oncol 2024; 47:102005. [PMID: 39034056 PMCID: PMC11736328 DOI: 10.1016/j.tranon.2024.102005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/23/2024] Open
Affiliation(s)
- Caroline E Nunes-Xavier
- Biobizkaia Health Research Institute, Spain; Institute for Cancer Research, Oslo University Hospital, Norway; CIBERER, ISCIII, Spain.
| |
Collapse
|
6
|
Tang R, Wang H, Tang M. Roles of tissue-resident immune cells in immunotherapy of non-small cell lung cancer. Front Immunol 2023; 14:1332814. [PMID: 38130725 PMCID: PMC10733439 DOI: 10.3389/fimmu.2023.1332814] [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: 11/03/2023] [Accepted: 11/23/2023] [Indexed: 12/23/2023] Open
Abstract
Non-small cell lung cancer (NSCLC) is the most common and lethal type of lung cancer, with limited treatment options and poor prognosis. Immunotherapy offers hope for improving the survival and quality of life of NSCLC patients, but its efficacy depends on the tumor immune microenvironment (TME). Tissue-resident immune cells are a subset of immune cells that reside in various tissues and organs, and play an important role in fighting tumors. In NSCLC, tissue-resident immune cells are heterogeneous in their distribution, phenotype, and function, and can either promote or inhibit tumor progression and response to immunotherapy. In this review, we summarize the current understanding on the characteristics, interactions, and roles of tissue-resident immune cells in NSCLC. We also discuss the potential applications of tissue-resident immune cells in NSCLC immunotherapy, including immune checkpoint inhibitors (ICIs), other immunomodulatory agents, and personalized cell-based therapies. We highlight the challenges and opportunities for developing targeted therapies for tissue-resident immune cells and optimizing existing immunotherapeutic approaches for NSCLC patients. We propose that tissue-resident immune cells are a key determinant of NSCLC outcome and immunotherapy response, and warrant further investigation in future research.
Collapse
Affiliation(s)
- Rui Tang
- School of Basic Medical Sciences, Southwest Medical University, Luzhou, Sichuan, China
- Department of Pathology, Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Haitao Wang
- The School of Clinical Medical Sciences, Southwest Medical University, Sichuan, Luzhou, China
| | - Mingxi Tang
- School of Basic Medical Sciences, Southwest Medical University, Luzhou, Sichuan, China
- Department of Pathology, Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
- Department of Pathology, Yaan People’s Hospital (Yaan Hospital of West China Hospital of Sichuan University), Yaan, Sichuan, China
| |
Collapse
|