1
|
Zhang YY, He JJ, Liu YL, Shao RN, Bai KH, Li XP, Guo T, Wang PH, Dai YJ. Unlocking the potential of TIGIT in enhancing therapeutic strategies for acute myeloid leukemia through combined azacitidine therapy. NPJ Precis Oncol 2025; 9:142. [PMID: 40374899 PMCID: PMC12081897 DOI: 10.1038/s41698-025-00933-6] [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: 01/17/2025] [Accepted: 05/01/2025] [Indexed: 05/18/2025] Open
Abstract
Immune checkpoint blockade (ICB) therapy has emerged as a pivotal advancement in cancer treatment, yet its efficacy varies among patients and resistance can develop. This study focuses on TIGIT, a newly identified immune checkpoint, to explore its expression, prognostic significance, and therapeutic potential in hematologic malignancies, particularly acute myeloid leukemia (AML). In this study, we found TIGIT highest expression levels in bone marrow and lymphoid tissues, with enrichment in immune cells such as NK-T cells and regulatory T cells (Tregs). A prognostic model incorporating TIGIT expression and other immune-related genes effectively stratified AML patients into high-risk and low-risk groups, with the former displaying significantly shorter overall survival times. Our model outperformed traditional prognostic factors, highlighting TIGIT's potential as a superior predictive biomarker. Additionally, our in vitro and in vivo studies showed that combining tiragolumab with azacitidine (AZA) synergistically enhanced anti-tumor efficacy, reducing tumor burden and extending survival in a murine AML model. Our findings underscore TIGIT's role in hematologic malignancies and its potential as a therapeutic target in AML. The combination of AZA with TIGIT inhibition offers a promising new approach for AML treatment, warranting further clinical evaluation.
Collapse
Affiliation(s)
- Yv-Yin Zhang
- Department of Hematology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Jia-Jun He
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
- Department of Hematologic Oncology, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China
| | - Yi-Lin Liu
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ruo-Nan Shao
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Kun-Hao Bai
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Xue-Ping Li
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
- Department of Hematologic Oncology, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China
| | - Tao Guo
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Pei-Hong Wang
- Department of Hematology, Guangzhou First People's Hospital, Institute of Blood Transfusion and Hematology, Guangzhou Medical University, Guangzhou, Guangdong, China.
| | - Yu-Jun Dai
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China.
- Department of Hematologic Oncology, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China.
| |
Collapse
|
2
|
Tulsian K, Thakker D, Vyas VK. Overcoming chimeric antigen receptor-T (CAR-T) resistance with checkpoint inhibitors: Existing methods, challenges, clinical success, and future prospects : A comprehensive review. Int J Biol Macromol 2025; 306:141364. [PMID: 39988153 DOI: 10.1016/j.ijbiomac.2025.141364] [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: 08/09/2024] [Revised: 11/20/2024] [Accepted: 02/19/2025] [Indexed: 02/25/2025]
Abstract
Immune checkpoint blockade is, as of today, the most successful form of cancer immunotherapy, with more than 43 % of cancer patients in the US eligible to receive it; however, only up to 12.5 % of patients respond to it. Similarly, adoptive cell therapy using bioengineered chimeric antigen receptorT (CAR-T) cells and T-cell receptor (TCR) cells has provided excellent responses against liquid tumours, but both forms of immunotherapy have encountered challenges within a tumour microenvironment that is both lacking in tumour-specific T-cells and is strongly immunosuppressive toward externally administered CAR-T and TCR cells. This review focuses on understanding approved checkpoint blockade and adoptive cell therapy at both biological and clinical levels before delving into how and why their combination holds significant promise in overcoming their individual shortcomings. The advent of next-generation checkpoint inhibitors has further strengthened the immune checkpoint field, and a special section explores how these inhibitors can address existing hurdles in combining checkpoint blockade with adoptive cell therapy and homing in on our cancer target for long-term immunity.
Collapse
Affiliation(s)
- Kartik Tulsian
- Department of Pharmaceutical Chemistry, Institute of Pharmacy, Nirma University, Ahmedabad, 382481, Gujarat, India
| | - Dhinal Thakker
- Department of Pharmaceutical Chemistry, Institute of Pharmacy, Nirma University, Ahmedabad, 382481, Gujarat, India
| | - Vivek K Vyas
- Department of Pharmaceutical Chemistry, Institute of Pharmacy, Nirma University, Ahmedabad, 382481, Gujarat, India.
| |
Collapse
|
3
|
Gupta S, Singh A, Deorah S, Tomar A. Immunotherapy in OSCC: Current trend and challenges. Crit Rev Oncol Hematol 2025; 209:104672. [PMID: 39993651 DOI: 10.1016/j.critrevonc.2025.104672] [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: 01/28/2025] [Revised: 02/14/2025] [Accepted: 02/18/2025] [Indexed: 02/26/2025] Open
Abstract
OBJECTIVES Oral Cancer is one of the most prevalent malignant tumors of the head and neck. The three primary clinical treatments available till now for oral cancer are chemotherapy, radiation, and surgery. The goal of this review was to outline the basic principles of immunotherapy along with various immunotherapeutic agents on Oral Squamous Cell Carcinoma. MATERIALS AND METHODS A comprehensive search in PubMed, Scopus, and Google Scholar was performed using relevant keywords. All the articles, both English as well as non-English were included also with inclusion data from high-incidence countries (South-east Asia) and the compilation was ten done after getting the data reviewed from two pathologists who were blinded to the data. RESULTS All the data has been compiled and the various sections in the manuscript provides an insight into the current trends in immunotherapy. CONCLUSIONS Advanced research studies are needed to counteract the hurdles associated with immunotherapy so that a greater proportion of patients can be treated. CLINICAL RELEVANCE One of the more recent developments that is promising is immunotherapy, which can be quite beneficial when used as a monotherapy or an adjuvant treatment. This more recent treatment approach could serve as the fourth pillar in cancer care, alongside radiation, chemotherapy, and surgery. Because immunotherapy relies on the patient's immunological environment, careful patient selection is essential to its effectiveness.
Collapse
Affiliation(s)
- Shalini Gupta
- Department of Oral Pathology and Microbiology, King George's Medical University, Lucknow 226003, India.
| | - Akanchha Singh
- Department of Oral Pathology and Microbiology, King George's Medical University, Lucknow 226003, India
| | - Sakshi Deorah
- Department of Oral Pathology and Microbiology, King George's Medical University, Lucknow 226003, India
| | - Arushi Tomar
- Department of Oral Pathology and Microbiology, King George's Medical University, Lucknow 226003, India
| |
Collapse
|
4
|
Liu R, Jiang X, Dong R, Zhang Y, Gai C, Wei P. Revealing the mechanisms and therapeutic potential of immune checkpoint proteins across diverse protein families. Front Immunol 2025; 16:1499663. [PMID: 40356928 PMCID: PMC12066663 DOI: 10.3389/fimmu.2025.1499663] [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: 09/21/2024] [Accepted: 03/28/2025] [Indexed: 05/15/2025] Open
Abstract
Host immune responses to antigens are tightly regulated through the activation and inhibition of synergistic signaling networks that maintain homeostasis. Stimulatory checkpoint molecules initiate attacks on infected or tumor cells, while inhibitory molecules halt the immune response to prevent overreaction and self-injury. Multiple immune checkpoint proteins are grouped into families based on common structural domains or origins, yet the variability within and between these families remains largely unexplored. In this review, we discuss the current understanding of the mechanisms underlying the co-suppressive functions of CTLA-4, PD-1, and other prominent immune checkpoint pathways. Additionally, we examine the IgSF, PVR, TIM, SIRP, and TNF families, including key members such as TIGIT, LAG-3, VISTA, TIM-3, SIRPα, and OX40. We also highlight the unique dual role of VISTA and SIRPα in modulating immune responses under specific conditions, and explore potential immunotherapeutic pathways tailored to the distinct characteristics of different immune checkpoint proteins. These insights into the unique advantages of checkpoint proteins provide new directions for drug discovery, emphasizing that emerging immune checkpoint molecules could serve as targets for novel therapies in cancer, autoimmune diseases, infectious diseases, and transplant rejection.
Collapse
Affiliation(s)
| | | | | | | | - Cong Gai
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Peng Wei
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| |
Collapse
|
5
|
Matsudo K, Takada K, Hashinokuchi A, Nagano T, Kinoshita F, Akamine T, Kohno M, Takenaka T, Shimokawa M, Oda Y, Yoshizumi T. CD155 expression and co-expression with PD-L1 are not associated with poor prognosis in patients with stage II and III lung adenocarcinoma undergoing surgical resection. Int J Clin Oncol 2025:10.1007/s10147-025-02771-9. [PMID: 40287900 DOI: 10.1007/s10147-025-02771-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2025] [Accepted: 04/13/2025] [Indexed: 04/29/2025]
Abstract
BACKGROUND CD155 has been identified as a ligand for T-cell immunoreceptor with Ig and ITIM domains. Herein, we investigated the relationship between the expressions of CD155 and programmed cell death-ligand 1 (PD-L1) and clinical outcomes in patients with surgically resected lung adenocarcinoma. METHODS This study included 426 patients diagnosed with pathological stage (pStage) I-III lung adenocarcinoma who underwent surgery at Kyushu University Hospital. The number of tumor cells expressing CD155 and PD-L1 was assessed by immunohistochemistry, and the clinical significance of CD155 expression and CD155/PD-L1 co-expression in prognosis was investigated. RESULTS Among the enrolled cohort, 320 (75.1%), 60 (14.1%), and 46 (10.8%) patients were diagnosed with pStage I, II, and III, respectively. Tissues from 112 patients (26.3%) were classified as having high CD155 expression. Co-expression of CD155 and PD-L1 was observed in 44 patients (10.3%). The High CD155 and CD155/PD-L1 co-expression groups had significantly poorer prognosis in pStage I-III lung adenocarcinoma. However, subgroup analysis revealed that the clinical significance of both CD155 expression and CD155/PD-L1 co-expression differed widely between patients with pStage I and II-III. Multivariate Cox proportional hazards regression analyses showed that high CD155 expression and CD155/PD-L1 co-expression were not independent poor prognostic factors in pStage II-III lung adenocarcinoma. CONCLUSION Our findings suggest that neither CD155 expression or CD155/PD-L1 co-expression are associated with poor prognosis in pStage II-III lung adenocarcinoma.
Collapse
Affiliation(s)
- Kyoto Matsudo
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Kazuki Takada
- Department of Surgery, Saiseikai Fukuoka General Hospital, Fukuoka, Japan
| | - Asato Hashinokuchi
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Taichi Nagano
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Fumihiko Kinoshita
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Takaki Akamine
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Mikihiro Kohno
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Tomoyoshi Takenaka
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Mototsugu Shimokawa
- Department of Biostatistics, Yamaguchi University Graduate School of Medicine, Yamaguchi, Japan
| | - Yoshinao Oda
- Department of Anatomic Pathology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Tomoharu Yoshizumi
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan.
| |
Collapse
|
6
|
Kabut J, Gorzelak-Magiera A, Gisterek-Grocholska I. New Therapeutic Targets TIGIT, LAG-3 and TIM-3 in the Treatment of Advanced, Non-Small-Cell Lung Cancer. Int J Mol Sci 2025; 26:4096. [PMID: 40362333 PMCID: PMC12072094 DOI: 10.3390/ijms26094096] [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: 03/10/2025] [Revised: 04/20/2025] [Accepted: 04/22/2025] [Indexed: 05/15/2025] Open
Abstract
The introduction of immunotherapy and target therapy into clinical practice has become a chance for many patients with cancer to prolong their survival while maintaining optimal quality of life. Treatment of lung cancer is excellent evidence of the progress of medical therapies. An understanding of the mechanisms of tumor development has led to the evolution of new methods of treatment. Immunoreceptors of T cells with the immunoglobulin domain ITIM, TIM-3 (T-cell immunoglobulin- and mucin domain-3-containing molecule 3), and LAG-3 (lymphocyte activation gene-3) represent new interesting therapeutic targets. The combination of anti-PD-1 and anti-CTLA-4 blockade has proven the possibility of strengthening the anti-tumor response by acting via two separate mechanisms. Adding additional checkpoints to the PD-1 blockade offers hope for further improvements in the effects of the treatment of patients and expanding the group responding to immunotherapy. This paper presents new promising molecular targets along with studies demonstrating the treatment results using them.
Collapse
Affiliation(s)
- Jacek Kabut
- Department of Oncology and Radiotherapy, Medical University of Silesia, 40-514 Katowice, Poland;
| | | | | |
Collapse
|
7
|
Ababneh E, Velez S, Zhao J. Immune evasion and resistance in breast cancer. Am J Cancer Res 2025; 15:1517-1539. [PMID: 40371160 PMCID: PMC12070088 DOI: 10.62347/pngt6996] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Accepted: 12/18/2024] [Indexed: 05/16/2025] Open
Abstract
Breast cancer (BC) is the most common malignancy in females with an increasing incidence in the last decade. The previously observed decline in BC mortality rates has also slowed down recently with an increase in the incidence of invasive BC. BC has various molecular subtypes. Among these subtypes, triple-negative breast cancer (TNBC) represents the most aggressive BC, with a poor prognosis. Because lack of the hormonal or human epidermal growth factor receptor 2 (HER2) receptors, TNBC is resistant to hormonal and HER2 targeted therapy effective for other BC subtypes. The good news is that TNBC has recently been considered an immunologically 'hot' tumor. Therefore, immunotherapy, particularly immune checkpoint inhibitor therapy, represents a promising therapeutic approach TNBC. However, a considerable percentage of patients with TNBC do not respond well to immunotherapy, indicating that TNBC seems to adopt several mechanisms to evade immune surveillance. Thus, it is crucial to investigate the mechanisms underlying TNBC immune evasion and resistance to immunotherapy. In this review, we examine and discuss the most recently discovered mechanisms for BC, with a particular focus on TNBC, to evade the immune surveillance via kidnapping the immune checkpoints, suppressing the immune responses in tumor microenvironment and inhibiting the tumor antigen presentation. Evaluation of these mechanisms in BC will hopefully guide future immunotherapeutic research and clinical trials.
Collapse
Affiliation(s)
- Ebaa Ababneh
- Burnett School of Biomedical Science, Medical College, University of Central Florida Orlando, FL, USA
| | - Sarah Velez
- Burnett School of Biomedical Science, Medical College, University of Central Florida Orlando, FL, USA
| | - Jihe Zhao
- Burnett School of Biomedical Science, Medical College, University of Central Florida Orlando, FL, USA
| |
Collapse
|
8
|
Feng M, Ma Q, Zhang B, Chen Y, Yang Y, He X, Zeng Y, Jing M, Ou X, Liu Y, Li Q, Liao W, Li X, Tan S, Qin D, Li D, Li Q, Wang Y. Targeting the poliovirus receptor to activate T cells and induce myeloid-derived suppressor cells to differentiate to pro-inflammatory macrophages via the IFN-γ-p-STAT1-IRF8 axis in cancer therapy. Cell Death Differ 2025:10.1038/s41418-025-01496-6. [PMID: 40229462 DOI: 10.1038/s41418-025-01496-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2024] [Revised: 03/05/2025] [Accepted: 03/21/2025] [Indexed: 04/16/2025] Open
Abstract
T cell immunoglobulin and ITIM domain (TIGIT) is one of the most important immune checkpoints expressed on lymphocytes, and poliovirus receptor (PVR, also CD155) serves as the most crucial ligand for TIGIT, harboring an important function in cancer cells and influencing the tumor microenvironment (TME). While it's well-established that TIGIT blockade could reverse immunosuppression, the question of whether direct inhibition of PVR yields comparable results remains to be fully elucidated. This study investigated the role of PVR within the TME on the LLC, CT26 and MC38 tumor models and found that direct blockade of PVR on tumor cells could trigger T cell activation, enhance the production of immunostimulatory cytokine IFN-γ, and drive the differentiation of intratumoral myeloid-derived suppressor cells (MDSCs) into pro-inflammatory macrophages through the IFN-γ-p-STAT1-IRF8 axis. Furthermore, this study found that the anti-PVR nanobody monotherapy reduced tumor volume in the CT26 and MC38 tumor models. Combination of anti-PVR nanobody and anti-PD-1 antibody was effective in the LLC, CT26 and MC38 tumor models and had acceptable toxicity. These findings collectively suggest that PVR exhibits considerable promise as a therapeutic target in the development of immunotherapies aimed at augmenting the anti-tumor immune response.
Collapse
Affiliation(s)
- Mingyang Feng
- Department of Medical Oncology, Cancer Center, and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Qizhi Ma
- Department of Medical Oncology, Cancer Center, and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Benxia Zhang
- Department of Medical Oncology, Cancer Center, and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
- Clinical Trial Center, West China Hospital, Sichuan University, Chengdu, China
| | - Yue Chen
- Department of Medical Oncology, Cancer Center, and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
- Clinical Trial Center, West China Hospital, Sichuan University, Chengdu, China
| | - Yang Yang
- Department of Medical Oncology, Cancer Center, and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Xia He
- Department of Medical Oncology, Cancer Center, and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
- Clinical Trial Center, West China Hospital, Sichuan University, Chengdu, China
- National Medical Products Administration Key Laboratory for Clinical Research and Evaluation of Innovative Drug, West China Hospital, Sichuan University, Chengdu, China
- Department of Clinical Research Management, West China Hospital, Sichuan University, Chengdu, China
| | - Yao Zeng
- Department of Medical Oncology, Cancer Center, and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Meng Jing
- Department of Pathology, Mianyang Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Mianyang, China
| | - Xuejin Ou
- Department of Medical Oncology, Cancer Center, and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Yixian Liu
- Clinical Trial Center, West China Hospital, Sichuan University, Chengdu, China
| | - Qian Li
- State Key Laboratory of Biotherapy and Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Weiting Liao
- Department of Medical Oncology, Cancer Center, and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Xiaoyu Li
- Department of Medical Oncology, Cancer Center, and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
- Clinical Trial Center, West China Hospital, Sichuan University, Chengdu, China
| | - Sirui Tan
- Department of Medical Oncology, Cancer Center, and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Diyuan Qin
- Department of Medical Oncology, Cancer Center, and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
- Clinical Trial Center, West China Hospital, Sichuan University, Chengdu, China
| | - Dan Li
- Precision Medicine Center, West China Hospital, Sichuan University, Chengdu, China
| | - Qiu Li
- Department of Medical Oncology, Cancer Center, and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China.
| | - Yongsheng Wang
- Division of Thoracic Tumor Multimodality Treatment, Cancer Center, and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China.
| |
Collapse
|
9
|
Wang SL, Chan TA. Navigating established and emerging biomarkers for immune checkpoint inhibitor therapy. Cancer Cell 2025; 43:641-664. [PMID: 40154483 DOI: 10.1016/j.ccell.2025.03.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2025] [Revised: 02/19/2025] [Accepted: 03/04/2025] [Indexed: 04/01/2025]
Abstract
Immune checkpoint inhibitors (ICIs) have improved outcomes of patients with many different cancers. These antibodies target molecules such as programmed cell death 1 (PD-1) or cytotoxic T lymphocyte associated protein 4 (CTLA-4) which normally function to limit immune activity. Treatment with ICIs reactivates T cells to destroy tumor cells in a highly specific manner, which in some patients, results in dramatic remissions and durable disease control. Over the last decade, much effort has been directed at characterizing factors that drive efficacy and resistance to ICI therapy. Food and Drug Administration (FDA)-approved biomarkers for ICI therapy have facilitated more judicious treatment of cancer patients and transformed the field of precision oncology. Yet, adaptive immunity against cancers is complex, and newer data have revealed the potential utility of other biomarkers. In this review, we discuss the utility of currently approved biomarkers and highlight how emerging biomarkers can further improve the identification of patients who benefit from ICIs.
Collapse
Affiliation(s)
- Stephen L Wang
- Center for Immunotherapy and Precision Immuno-Oncology, Cleveland Clinic, Cleveland, OH, USA; Medical Scientist Training Program, Case Western Reserve University School of Medicine, Cleveland, OH, USA; Department of Genetics and Genome Sciences, Case Western Reserve University, Cleveland, OH, USA
| | - Timothy A Chan
- Center for Immunotherapy and Precision Immuno-Oncology, Cleveland Clinic, Cleveland, OH, USA; National Center for Regenerative Medicine, Cleveland, OH, USA.
| |
Collapse
|
10
|
Masmoudi D, Villalba M, Alix-Panabières C. Natural killer cells: the immune frontline against circulating tumor cells. J Exp Clin Cancer Res 2025; 44:118. [PMID: 40211394 PMCID: PMC11983744 DOI: 10.1186/s13046-025-03375-x] [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: 01/10/2025] [Accepted: 03/25/2025] [Indexed: 04/14/2025] Open
Abstract
Natural killer (NK) play a key role in controlling tumor dissemination by mediating cytotoxicity towards cancer cells without the need of education. These cells are pivotal in eliminating circulating tumor cells (CTCs) from the bloodstream, thus limiting cancer spread and metastasis. However, aggressive CTCs can evade NK cell surveillance, facilitating tumor growth at distant sites. In this review, we first discuss the biology of NK cells, focusing on their functions within the tumor microenvironment (TME), the lymphatic system, and circulation. We then examine the immune evasion mechanisms employed by cancer cells to inhibit NK cell activity, including the upregulation of inhibitory receptors. Finally, we explore the clinical implications of monitoring circulating biomarkers, such as NK cells and CTCs, for therapeutic decision-making and emphasize the need to enhance NK cell-based therapies by overcoming immune escape mechanisms.
Collapse
Affiliation(s)
- Doryan Masmoudi
- Laboratory of Rare Circulating Human Cells, University Medical Center of Montpellier, Montpellier, France
| | - Martin Villalba
- IRMB, Univ Montpellier, INSERM, CHU Montpellier, CNRS, Montpellier, France
| | - Catherine Alix-Panabières
- Laboratory of Rare Circulating Human Cells, University Medical Center of Montpellier, Montpellier, France.
- CREEC/CANECEV, MIVEGEC (CREES), University of Montpellier, CNRS, Montpellier, IRD, France.
- European Liquid Biopsy Society (ELBS), Hamburg, Germany.
- LCCRH, Site Unique de Biologie (SUB), 641, Avenue du Doyen Gaston Giraud, Montpellier, 34093, France.
| |
Collapse
|
11
|
Morcillo-Martín-Romo P, Valverde-Pozo J, Ortiz-Bueno M, Arnone M, Espinar-Barranco L, Espinar-Barranco C, García-Rubiño ME. The Role of NK Cells in Cancer Immunotherapy: Mechanisms, Evasion Strategies, and Therapeutic Advances. Biomedicines 2025; 13:857. [PMID: 40299429 PMCID: PMC12024875 DOI: 10.3390/biomedicines13040857] [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: 03/03/2025] [Revised: 03/24/2025] [Accepted: 03/30/2025] [Indexed: 04/30/2025] Open
Abstract
Background/Objectives: Natural killer (NK) cells play a crucial role in tumor surveillance by exerting cytotoxic activity and modulating immune responses. However, tumors employ diverse evasion strategies that limit NK cell effectiveness. This review aims to explore the molecular mechanisms of NK cell activation and inhibition in cancer, the influence of the tumor microenvironment, and the latest advancements in NK cell-based immunotherapies, including adoptive NK cell transfer and Chimeric Antigen Receptor-Natural Killer (CAR-NK) cell therapies. Methods: A comprehensive literature review was conducted, prioritizing peer-reviewed studies from the last decade on NK cell biology, tumor immune evasion, and immunotherapeutic applications. The analysis includes data from preclinical models and clinical trials evaluating NK cell expansion strategies, cytokine-based stimulation, and CAR-NK cell therapy developments. Results: NK cells eliminate tumors through cytotoxic granule release, death receptor pathways, and cytokine secretion. However, tumor cells evade NK-mediated immunity by downregulating activating ligands, secreting immunosuppressive molecules, and altering the tumor microenvironment. Novel NK cell-based therapies, such as CAR-NK cells and combination approaches with immune checkpoint inhibitors, enhance NK cell persistence and therapeutic efficacy against both hematologic and solid malignancies. Clinical trials suggest improved safety profiles compared to CAR-T therapies, with reduced cytokine release syndrome and graft-versus-host disease. Conclusions: While NK cell-based immunotherapies hold great promise, challenges remain, including limited persistence and tumor-induced immunosuppression. Addressing these hurdles will be critical for optimizing NK cell therapies and advancing next-generation, off-the-shelf immunotherapeutics for broader clinical applications.
Collapse
Affiliation(s)
- Paula Morcillo-Martín-Romo
- Centre for Biomedical Research (CIBM), University of Granada, 18100 Granada, Spain; (P.M.-M.-R.); (M.A.)
| | - Javier Valverde-Pozo
- Departamento de Química y Bioquímica, Facultad de Farmacia, Universidad San Pablo-CEU, CEU Universities, Urbanización Montepríncipe, 28668 Boadilla del Monte, Spain;
| | - María Ortiz-Bueno
- Nanoscopy-UGR Laboratory, Department of Physical Chemistry, Faculty of Pharmacy, Unidad de Excelencia en Quimica Aplicada a Biomedicina y Medioambiente (UEQ), University of Granada, C. U. Cartuja, 18071 Granada, Spain; (M.O.-B.); (L.E.-B.)
| | - Maurizio Arnone
- Centre for Biomedical Research (CIBM), University of Granada, 18100 Granada, Spain; (P.M.-M.-R.); (M.A.)
| | - Laura Espinar-Barranco
- Nanoscopy-UGR Laboratory, Department of Physical Chemistry, Faculty of Pharmacy, Unidad de Excelencia en Quimica Aplicada a Biomedicina y Medioambiente (UEQ), University of Granada, C. U. Cartuja, 18071 Granada, Spain; (M.O.-B.); (L.E.-B.)
- Department of Medicine, Translational Transplant Research Center, Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Celia Espinar-Barranco
- Servicio de Análisis Clínicos e Inmunología, Hospital Universitario Virgen de las Nieves, 18014 Granada, Spain;
| | - María Eugenia García-Rubiño
- Centre for Biomedical Research (CIBM), University of Granada, 18100 Granada, Spain; (P.M.-M.-R.); (M.A.)
- Nanoscopy-UGR Laboratory, Department of Physical Chemistry, Faculty of Pharmacy, Unidad de Excelencia en Quimica Aplicada a Biomedicina y Medioambiente (UEQ), University of Granada, C. U. Cartuja, 18071 Granada, Spain; (M.O.-B.); (L.E.-B.)
- Instituto de Investigación Biosanitaria ibs.GRANADA, University Hospitals of Granada-University of Granada, 18100 Granada, Spain
| |
Collapse
|
12
|
Rezazadeh‐Gavgani E, Majidazar R, Lotfinejad P, Kazemi T, Shamekh A. Immune Checkpoint Molecules: A Review on Pathways and Immunotherapy Implications. Immun Inflamm Dis 2025; 13:e70196. [PMID: 40243372 PMCID: PMC12004596 DOI: 10.1002/iid3.70196] [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: 05/07/2024] [Revised: 03/12/2025] [Accepted: 03/27/2025] [Indexed: 04/18/2025] Open
Abstract
BACKGROUND Today, treating cancer patients with monoclonal antibodies (mAbs), by targeting immune checkpoints, is one of the most outstanding immunotherapeutic methods. Immune checkpoints are special molecules having regulatory role in immune system responses. Once these molecules are presented on cancer cells, these cells will be capable of evading the immune system through their own specific pathways. This Evasion can be prevented by counterbalancing immune system responses with immune checkpoints related antibodies. AIMS The current study aimed to highlight immunotherapy and its methods, describe the immune checkpoints pathways, outline the immune checkpoint inhibitors (ICIs), and recent advances in this field, and sketch an outlook on the best treatment options for the most prevalent cancers. MATERIALS & METHODS This research implemented a narrative review method. A comprehensive literature review on the history, molecular and cellular biology, and the clinical aspects of immune checkpoint molecules was performed to illustrate the pathways involved in various cancers. Also, currently-available and future potential immunotherapies targeting these pathways were extracted from the searched studies. RESULTS The immune checkpoint family consists of many molecules, including CTLA-4, PD-1, PD-L1, LAG-3, TIM-3, and TIGIT. Attempts to modify these molecules in cancer treatment led to the development of therapeutic monoclonal antibodies. Most of these antibodies have entered clinical studies and some of them have been approved by the Food and Drug Administration (FDA) to be used in cancer patients' treatment plans. DISCUSSION With these novel treatments and the combination therapies they offer, there is also hope for better treatment outcomes for the previously untreatable metastatic cancers. In spite of the beneficial aspects of immune checkpoint therapy, similar to other treatments, they may cause side effects in some patients. Therefore, more studies are needed to reduce the probable side effects and uncover their underlying mechanism. CONCLUSION Based on the data shown in this review, there is still a lack of knowledge about the complete properties of ICIs and the possible combination therapies that we may be able to implement to achieve a better treatment response in cancer patients.
Collapse
Affiliation(s)
| | - Reza Majidazar
- Student Research CommitteeTabriz University of Medical SciencesTabrizIran
| | - Parisa Lotfinejad
- Immunology Research CenterTabriz University of Medical SciencesTabrizIran
- Department of ImmunologyTabriz University of Medical SciencesTabrizIran
| | - Tohid Kazemi
- Immunology Research CenterTabriz University of Medical SciencesTabrizIran
- Department of ImmunologyTabriz University of Medical SciencesTabrizIran
| | - Ali Shamekh
- Student Research CommitteeTabriz University of Medical SciencesTabrizIran
- Aging Research InstituteTabriz University of Medical SciencesTabrizIran
| |
Collapse
|
13
|
Geng Q, Xu J, Du C, Zhang D, Jin Y, Song J, Qu W, Zhang C, Su G, Jiao P. Small molecules targeting immune checkpoint proteins for cancer immunotherapy: a patent and literature review (2020-2024). Expert Opin Ther Pat 2025; 35:409-440. [PMID: 39907457 DOI: 10.1080/13543776.2025.2462849] [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: 08/06/2024] [Revised: 12/20/2024] [Accepted: 01/29/2025] [Indexed: 02/06/2025]
Abstract
INTRODUCTION Targeting immune checkpoint proteins (ICPs) via small molecules open a new window for cancer immunotherapy. Herein, we summarize recent advances of small molecules with novel chemical structures targeting ICPs, discusses their anti-tumor efficacies, which are important for the development of novel small molecules for cancer immunotherapy. AREAS COVERED In this review, the latest patents and literature were gathered through the comprehensive searches in the databases of European Patent Office (EPO), Cortellis Drug Discovery Intelligence (CDDI), PubMed and Web of Science using ICPs and compounds as key words. EXPERT OPINION To develop novel weapons to fight against cancer, small molecules targeting ICPs including CTLA-4, LAG-3, PD-L1, Siglec-9, TIM-3, TIGIT, and VISTA have been synthesized and evaluated in succession. Chief among them are the small molecules targeting PD-L1, which have been intensively investigated in recent years. Various in vitro assays such as ALPHA, HTRF binding assay, NFAT assay have been successfully developed to screen novel IPCs inhibitors. However, the in vivo assay, for example, using double-humanized PD-1/PD-L1 (hPD-1/hPD-L1) mouse as evaluation model, are seldom reported. Novel pharmacophores with new working mechanisms such as proteolysis targeting chimeras (PROTACs) and peptides are needed to enhance the therapeutic efficacy.
Collapse
Affiliation(s)
- Qiaohong Geng
- School of Chemistry and Chemical Engineering, Qilu Normal University, Jinan, Shandong, China
| | - Juanjuan Xu
- Department of Neurology, Changyi People's Hospital, Weifang, Shandong, China
| | - Chunsheng Du
- School of Chemistry and Chemical Engineering, Qilu Normal University, Jinan, Shandong, China
| | - Deheng Zhang
- School of Chemistry and Chemical Engineering, Qilu Normal University, Jinan, Shandong, China
| | - Yanrui Jin
- School of Chemistry and Chemical Engineering, Qilu Normal University, Jinan, Shandong, China
| | - Jiatong Song
- School of Chemistry and Chemical Engineering, Qilu Normal University, Jinan, Shandong, China
| | - Wenjing Qu
- School of Chemistry and Chemical Engineering, Qilu Normal University, Jinan, Shandong, China
| | - Changnan Zhang
- School of Chemistry and Chemical Engineering, Qilu Normal University, Jinan, Shandong, China
| | - Gaoxing Su
- School of Pharmacy, Nantong University, Nantong, Jiangsu, China
| | - Peifu Jiao
- School of Chemistry and Chemical Engineering, Qilu Normal University, Jinan, Shandong, China
| |
Collapse
|
14
|
Jiang S, Zheng S, Yao C, Ning D, Zou S, Zhan J, Lan T, Yi T, Jin Z, Wu X. Heterogeneity of γδ T-cell subsets and their clinical correlation in patients with AML. Front Immunol 2025; 16:1552235. [PMID: 40236710 PMCID: PMC11996841 DOI: 10.3389/fimmu.2025.1552235] [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: 12/27/2024] [Accepted: 03/10/2025] [Indexed: 04/17/2025] Open
Abstract
Background γδ T cells are integral elements of the immune system and have shown therapeutic potential in the treatment of acute myeloid leukemia (AML). Nevertheless, the influence of distinct functional subsets, including the activating marker NKG2D, the immune exhaustion marker TIGIT, and the regulatory marker Foxp3, on therapeutic outcomes in AML patients remains unknown. Methods First, we analyzed RNA-seq data from 167 patients in The Cancer Genome Atlas (TCGA) database, concentrating on the correlations between NKG2D, TIGIT, and Foxp3 gene expressions and their association with prognosis in AML. We employed flow cytometry to assess the expression of these molecular markers on γδ T cells and the Vδ1/Vδ2 subsets in the peripheral blood of 25 de novo AML (AML-DN) patients, 15 patients in complete remission (CR), and 27 healthy controls (HCs). We also analyzed the relationship between the expression frequencies of NKG2D, TIGIT, and Foxp3 on γδ T cells and their subsets, and their clinical outcomes. Results Based on data from TCGA database, we found that a high expression level of NKG2D in combination with a low expression level of TIGIT was significantly associated with longer overall survival (OS) in AML patients. Clinical data revealed that γδ T cells from AML-DN patients exhibited higher expression levels of TIGIT and Foxp3, whereas NKG2D expression was lower compared to that of HCs. Notably, the expression of the NKG2D+TIGIT- Vδ1 subset was significantly reduced in AML-DN patients compared to CR patients. Univariate logistic regression and Cox regression analyses further indicated that a high expression of the NKG2D+TIGIT- Vδ1 subset was associated with better clinical prognosis. Conclusion This study indicates that NKG2D+TIGIT- Vδ1 T cells are strongly correlated with improved prognosis in AML, and future research should investigate their potential in adoptive immunotherapy to advance more personalized and precise treatment strategies.
Collapse
MESH Headings
- Humans
- Leukemia, Myeloid, Acute/immunology
- Leukemia, Myeloid, Acute/mortality
- Leukemia, Myeloid, Acute/genetics
- Female
- Male
- Middle Aged
- Receptors, Antigen, T-Cell, gamma-delta/metabolism
- Receptors, Antigen, T-Cell, gamma-delta/immunology
- Adult
- Aged
- Receptors, Immunologic/genetics
- Receptors, Immunologic/metabolism
- T-Lymphocyte Subsets/immunology
- T-Lymphocyte Subsets/metabolism
- NK Cell Lectin-Like Receptor Subfamily K/genetics
- NK Cell Lectin-Like Receptor Subfamily K/metabolism
- Prognosis
- Forkhead Transcription Factors/genetics
- Forkhead Transcription Factors/metabolism
- Young Adult
- Intraepithelial Lymphocytes/immunology
Collapse
Affiliation(s)
- Siyuan Jiang
- Institute of Hematology, Medical Laboratory Center, School of Medicine, Jinan University, Guangzhou, China
| | - Shiyu Zheng
- Institute of Hematology, Medical Laboratory Center, School of Medicine, Jinan University, Guangzhou, China
| | - Chao Yao
- Institute of Hematology, Medical Laboratory Center, School of Medicine, Jinan University, Guangzhou, China
| | - Dengchong Ning
- Youjiang Medical University for Nationalities, Baise, China
| | - Shaoyun Zou
- Institute of Hematology, Medical Laboratory Center, School of Medicine, Jinan University, Guangzhou, China
| | - Jiannan Zhan
- Institute of Hematology, Medical Laboratory Center, School of Medicine, Jinan University, Guangzhou, China
| | - Tianbi Lan
- Dongguan Institute of Clinical Cancer Research, Dongguan Key Laboratory of Precision Diagnosis and Treatment for Tumors, The Tenth Affiliated Hospital, Southern Medical University (Dongguan People’s Hospital), Dongguan, China
| | - Tingzhuang Yi
- Department of Oncology, Affiliated Hospital of YouJiang Medical University for Nationalities/Key Laboratory of Molecular Pathology in Tumors of Guangxi Higher Education Institutions, Baise, China
| | - Zhenyi Jin
- Department of Pathology, School of Medicine, Jinan University, Guangzhou, China
- Key Laboratory of Viral Pathogenesis and Infection Prevention and Control (Jinan University), Ministry of Education, Guangzhou, China
- Jinan-Puhua Joint Laboratory, Guangzhou, China
| | - Xiuli Wu
- Institute of Hematology, Medical Laboratory Center, School of Medicine, Jinan University, Guangzhou, China
- Key Laboratory of Viral Pathogenesis and Infection Prevention and Control (Jinan University), Ministry of Education, Guangzhou, China
- Jinan-Puhua Joint Laboratory, Guangzhou, China
| |
Collapse
|
15
|
Zhou J, Li N, Li X, Ye J, Wang M, Sun G. Review on recent advancements in understanding acetylsalicylic acid-induced gastrointestinal injury: mechanisms, medication, and dosage refinement. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025; 398:3297-3320. [PMID: 39545984 DOI: 10.1007/s00210-024-03521-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Accepted: 10/06/2024] [Indexed: 11/17/2024]
Abstract
Acetylsalicylic acid (ASA) is a clinical drug with multiple effects, including prevention of cardiovascular adverse events and anti-cancer effects. However, gastrointestinal side effects, such as gastrointestinal ulcers and bleeding, limit the use of ASA and reduce patient compliance. Various studies have investigated the mechanisms of ASA-induced gastrointestinal injury, and many medicines have been reported to be effective in preventing and treating the adverse gastrointestinal effects of ASA. New formulations of ASA have demonstrated milder gastrointestinal injury than ASA alone. In this article, we summarized the mechanisms of ASA-induced gastrointestinal injury, drugs that resist gastrointestinal side effects of ASA, and progress in research on formulation improvement of ASA to help resolve the clinical dilemma of ASA usage.
Collapse
Affiliation(s)
- Jiahui Zhou
- Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
- Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine, Ministry of Education, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100193, China
- NMPA Key Laboratory for Research and Evaluation of Pharmacovigilance, Beijing, China
- Key Laboratory of New Drug Discovery Based On Classic Chinese Medicine Prescription, Chinese Academy of Medical Sciences, Beijing, China
| | - Na Li
- Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
- Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine, Ministry of Education, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100193, China
- NMPA Key Laboratory for Research and Evaluation of Pharmacovigilance, Beijing, China
- Key Laboratory of New Drug Discovery Based On Classic Chinese Medicine Prescription, Chinese Academy of Medical Sciences, Beijing, China
| | - Xinzhong Li
- Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
- Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine, Ministry of Education, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100193, China
- NMPA Key Laboratory for Research and Evaluation of Pharmacovigilance, Beijing, China
- Key Laboratory of New Drug Discovery Based On Classic Chinese Medicine Prescription, Chinese Academy of Medical Sciences, Beijing, China
| | - Jingxue Ye
- Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China.
- Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine, Ministry of Education, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100193, China.
- NMPA Key Laboratory for Research and Evaluation of Pharmacovigilance, Beijing, China.
- Key Laboratory of New Drug Discovery Based On Classic Chinese Medicine Prescription, Chinese Academy of Medical Sciences, Beijing, China.
| | - Min Wang
- Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China.
- Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine, Ministry of Education, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100193, China.
- NMPA Key Laboratory for Research and Evaluation of Pharmacovigilance, Beijing, China.
- Key Laboratory of New Drug Discovery Based On Classic Chinese Medicine Prescription, Chinese Academy of Medical Sciences, Beijing, China.
| | - Guibo Sun
- Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China.
- Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine, Ministry of Education, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100193, China.
- NMPA Key Laboratory for Research and Evaluation of Pharmacovigilance, Beijing, China.
- Key Laboratory of New Drug Discovery Based On Classic Chinese Medicine Prescription, Chinese Academy of Medical Sciences, Beijing, China.
| |
Collapse
|
16
|
Chakraborty A, Kumar A. Computational screening for natural compounds as potential immune checkpoint inhibitors against TIGIT, a new avenue in cancer immunotherapy. Mol Divers 2025:10.1007/s11030-025-11172-z. [PMID: 40163147 DOI: 10.1007/s11030-025-11172-z] [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: 01/20/2025] [Accepted: 03/16/2025] [Indexed: 04/02/2025]
Abstract
The TIGIT-PVR signalling pathway is a key mechanism of tumour immune evasion, making it an attractive target for cancer immunotherapy. Despite the recent advances in anti-TIGIT antibodies, monoclonal antibody-based therapeutics present significant challenges because of their immunogenicity and immune-related side effects. This study presents a new path involving natural compounds as potential small molecule inhibitors of TIGIT, providing a possible alternative to antibodies in cancer immunotherapy. Through a comprehensive in silico workflow combining structure-based virtual screening, ADMET analysis, Molecular docking and molecular dynamics simulations, six promising candidates, mostly of bacterial origin, were identified: Neomycin K, 4'-Deoxybutirosin A, 5-Glucosyl-neamine, S-11-A, 12-carbamoylstreptothricin E acid, and Zwittermicin A. These candidates demonstrated favourable binding energies, stable interactions, and the capacity to block TIGIT-PVR signalling. The compounds can potentially compete with PVR to bind to TIGIT, limiting the formation of the TIGIT-PVR complex, which typically activates an inhibitory cascade in T cells and NK cells, reducing their anti-tumour activity. By disrupting this interaction, the identified compounds have the potential to stimulate T cell and NK cell responses against cancer cells. Such natural compounds potentially provide better tissue penetration and reduced immunogenicity compared to conventional antibody therapies. The discovery of bacterial-derived compounds as TIGIT inhibitors presents a new direction in the investigation of microbial metabolites for cancer immunotherapy. This strategy not only identifies a new class of TIGIT inhibitors but also provides a robust computational framework for discovering and characterizing small molecule immune checkpoint inhibitors, paving the way for subsequent experimental validation to explore their efficacy in restoring anti-tumour immune responses and improving clinical outcomes for cancer patients.
Collapse
Affiliation(s)
- Aritra Chakraborty
- Department of Biosciences and Biomedical Engineering, Indian Institute of Technology Indore, Simrol, 453552, India
| | - Amit Kumar
- Department of Biosciences and Biomedical Engineering, Indian Institute of Technology Indore, Simrol, 453552, India.
| |
Collapse
|
17
|
Kamrani S, Naseramini R, Khani P, Razavi ZS, Afkhami H, Atashzar MR, Nasri F, Alavimanesh S, Saeidi F, Ronaghi H. Mesenchymal stromal cells in bone marrow niche of patients with multiple myeloma: a double-edged sword. Cancer Cell Int 2025; 25:117. [PMID: 40140850 PMCID: PMC11948648 DOI: 10.1186/s12935-025-03741-x] [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: 10/24/2024] [Accepted: 03/08/2025] [Indexed: 03/28/2025] Open
Abstract
Multiple myeloma (MM) is a hematological malignancy defined by the abnormal proliferation and accumulation of plasma cells (PC) within the bone marrow (BM). While multiple myeloma impacts the bone, it is not classified as a primary bone cancer. The bone marrow microenvironment significantly influences the progression of myeloma and its treatment response. Mesenchymal stromal cells (MSCs) in this environment engage with myeloma cells and other bone marrow components via direct contact and the secretion of soluble factors. This review examines the established roles of MSCs in multiple facets of MM pathology, encompassing their pro-inflammatory functions, contributions to tumor epigenetics, effects on immune checkpoint inhibitors (ICIs), influence on reprogramming, chemotherapy resistance, and senescence. This review investigates the role of MSCs in the development and progression of MM.
Collapse
Affiliation(s)
- Sina Kamrani
- Department of Orthopedic, Faculty of Medicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Reza Naseramini
- Department of Orthopedic, Faculty of Medicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Pouria Khani
- Department of Medical Genetics, School of Medicine, Tehran University of Medical Sciences (TUMS), Tehran, Iran
| | - Zahra Sadat Razavi
- Physiology Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Hamed Afkhami
- Cellular and Molecular Research Center, Qom University of Medical Sciences, Qom, Iran
- Nervous System Stem Cells Research Center, Semnan University of Medical Sciences, Semnan, Iran
- Department of Medical Microbiology, Faculty of Medicine, Shahed University, Tehran, Iran
| | - Mohammad Reza Atashzar
- Department of Immunology, School of Medicine, Fasa University of Medical Sciences, Fasa, Iran
| | - Farzad Nasri
- Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Sajad Alavimanesh
- Student Research Committee, Shahrekord University of Medical Sciences, Shahrekord, Iran
- Cellular and Molecular Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Farzane Saeidi
- Department of Medical Genetics, School of Medical Sciences, Tarbiat Modares University, Tehran, Iran.
| | - Hossein Ronaghi
- Department of Orthopedic, Faculty of Medicine, Guilan University of Medical Sciences, Rasht, Iran.
| |
Collapse
|
18
|
Li F, Gao C, Huang Y, Qiao Y, Xu H, Liu S, Wu H. Unraveling the breast cancer tumor microenvironment: crucial factors influencing natural killer cell function and therapeutic strategies. Int J Biol Sci 2025; 21:2606-2628. [PMID: 40303301 PMCID: PMC12035885 DOI: 10.7150/ijbs.108803] [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: 12/14/2024] [Accepted: 03/04/2025] [Indexed: 05/02/2025] Open
Abstract
Natural killer (NK) cells have emerged as a novel and effective treatment for breast cancer. Nevertheless, the breast cancer tumor microenvironment (TME) manifests multiple immunosuppressive mechanisms, impeding the proper execution of NK cell functions. This review summarizes recent research on the influence of the TME on the functionality of NK cells in breast cancer. It delves into the effects of the internal environment of the TME on NK cells and elucidates the roles of diverse stromal components, immune cells, and signaling molecules in regulating NK cell activity within the TME. It also summarizes therapeutic strategies based on small-molecule inhibitors, antibody therapies, and natural products, as well as the progress of research in preclinical and clinical trials. By enhancing our understanding of the immunosuppressive TME and formulating strategies to counteract its effects, we could fully harness the therapeutic promise of NK cells in breast cancer treatment.
Collapse
Affiliation(s)
- Feifei Li
- Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Shanghai Research Institute of Acupuncture and Meridian, Shanghai, China
- Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Integrated Traditional Chinese and Western Medicine Breast Department, Longhua Hospital Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Chunfang Gao
- Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yan Huang
- Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Shanghai Research Institute of Acupuncture and Meridian, Shanghai, China
| | - Yu Qiao
- Shanghai Research Institute of Acupuncture and Meridian, Shanghai, China
| | - Hongxiao Xu
- Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Sheng Liu
- Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Integrated Traditional Chinese and Western Medicine Breast Department, Longhua Hospital Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Huangan Wu
- Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Shanghai Research Institute of Acupuncture and Meridian, Shanghai, China
| |
Collapse
|
19
|
Narote S, Desai SA, Patel VP, Deshmukh R, Raut N, Dapse S. Identification of new immune target and signaling for cancer immunotherapy. Cancer Genet 2025; 294-295:57-75. [PMID: 40154216 DOI: 10.1016/j.cancergen.2025.03.004] [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: 01/01/2025] [Revised: 03/14/2025] [Accepted: 03/17/2025] [Indexed: 04/01/2025]
Abstract
Immunotherapy has become one of the innovative treatments in malignancy as it activates the immune system to find and eliminate malignant cells. The tumor immunology interface has become increasingly intricate, making the identification of new immune targets and signalling pathways on which to base improved therapeutic strategies an ongoing process. This review, we goal to clarify the contacts between cancer and immune system with a focus on immune surveillance as well as immune evasion mechanisms. Comprehensive immunotherapeutic therapies are overviewed with ICI (CTLA-4, PD-1, PD-L1), CAR-T cell therapy, and cancer vaccines whereas, advanced therapies targeting new immune checkpoints are also elucidated including TIM-3, LAG-3, and TIGIT. The JAK/STAT, MAPK and PI3K-AKT-mTOR pathways are reviewed with regards to cancer progression and immunotherapeutic resistance. The dysregulation of these pathways gives hope for the identification of fresh targets for therapy. Genomics, proteomics, immunopeptidomics, single cell mass spectrometry, CRISPR-based functional genomics and bioinformatics are described as essential for immune target identification and for mapping of cancer relevant signaling pathways. This review also considers some emerging issues in the subject area like the tumor heterogeneity, immune-related adverse events (irAEs), and personalized treatment. These barriers are described to facilitate the understanding of ways to overcome them and increase the efficacy of immunotherapies through combination therapies. This means that by developing new knowledge of immunological targets and pathways, immunoprecision medicine for cancer could greatly enhance outcomes.
Collapse
Affiliation(s)
- Sakshi Narote
- Department of Pharmaceutical Biotechnology, Sanjivani College of Pharmaceutical Education & Research, Savitribai Phule Pune University, Kopargaon, Maharashtra, India
| | - Sharav A Desai
- Department of Pharmaceutical Biotechnology, Sanjivani College of Pharmaceutical Education & Research, Savitribai Phule Pune University, Kopargaon, Maharashtra, India.
| | - Vipul P Patel
- Department of Pharmaceutical Biotechnology, Sanjivani College of Pharmaceutical Education & Research, Savitribai Phule Pune University, Kopargaon, Maharashtra, India
| | - Rutuja Deshmukh
- Department of Pharmaceutical Biotechnology, Sanjivani College of Pharmaceutical Education & Research, Savitribai Phule Pune University, Kopargaon, Maharashtra, India
| | - Nikita Raut
- Department of Pharmaceutical Biotechnology, Sanjivani College of Pharmaceutical Education & Research, Savitribai Phule Pune University, Kopargaon, Maharashtra, India
| | - Sejal Dapse
- Department of Pharmaceutical Biotechnology, Sanjivani College of Pharmaceutical Education & Research, Savitribai Phule Pune University, Kopargaon, Maharashtra, India
| |
Collapse
|
20
|
Maher N, Mouhssine S, Matti BF, Alwan AF, Gaidano G. Molecular Mechanisms in the Transformation from Indolent to Aggressive B Cell Malignancies. Cancers (Basel) 2025; 17:907. [PMID: 40075754 PMCID: PMC11899122 DOI: 10.3390/cancers17050907] [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: 01/16/2025] [Revised: 02/16/2025] [Accepted: 02/28/2025] [Indexed: 03/14/2025] Open
Abstract
Histological transformation (HT) into aggressive lymphoma is a turning point in a significant fraction of patients affected by indolent lymphoproliferative neoplasms, namely, chronic lymphocytic leukemia (CLL), follicular lymphoma (FL), marginal zone lymphomas (MZLs), and lymphoplasmacytic lymphoma (LPL) [...].
Collapse
Affiliation(s)
- Nawar Maher
- Division of Hematology, Department of Translational Medicine, Università del Piemonte Orientale and Azienda Ospedaliero-Universitaria Maggiore della Carità, 28100 Novara, Italy; (N.M.); (S.M.)
| | - Samir Mouhssine
- Division of Hematology, Department of Translational Medicine, Università del Piemonte Orientale and Azienda Ospedaliero-Universitaria Maggiore della Carità, 28100 Novara, Italy; (N.M.); (S.M.)
| | - Bassam Francis Matti
- Department of Hematology and Bone Marrow Transplant, Hematology and Bone Marrow Transplant Center, Medical City, Baghdad 00964, Iraq;
| | - Alaa Fadhil Alwan
- Department of Clinical Hematology, The National Center of Hematology, Mustansiriyah University, Baghdad 10001, Iraq;
| | - Gianluca Gaidano
- Division of Hematology, Department of Translational Medicine, Università del Piemonte Orientale and Azienda Ospedaliero-Universitaria Maggiore della Carità, 28100 Novara, Italy; (N.M.); (S.M.)
| |
Collapse
|
21
|
Roussot N, Kaderbhai C, Ghiringhelli F. Targeting Immune Checkpoint Inhibitors for Non-Small-Cell Lung Cancer: Beyond PD-1/PD-L1 Monoclonal Antibodies. Cancers (Basel) 2025; 17:906. [PMID: 40075753 PMCID: PMC11898530 DOI: 10.3390/cancers17050906] [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: 01/31/2025] [Revised: 02/28/2025] [Accepted: 03/03/2025] [Indexed: 03/14/2025] Open
Abstract
Non-small-cell lung cancer (NSCLC) remains a leading cause of cancer-related mortality worldwide. Immunotherapy targeting the PD-1/PD-L1 axis has revolutionized treatment, providing durable responses in a subset of patients. However, with fewer than 50% of patients achieving significant benefits, there is a critical need to expand therapeutic strategies. This review explores emerging targets in immune checkpoint inhibition beyond PD-1/PD-L1, including CTLA-4, TIGIT, LAG-3, TIM-3, NKG2A, and CD39/CD73. We highlight the biological basis of CD8 T cell exhaustion in shaping the antitumor immune response. Novel therapeutic approaches targeting additional inhibitory receptors (IR) are discussed, with a focus on their distinct mechanisms of action and combinatory potential with existing therapies. Despite significant advancements, challenges remain in overcoming resistance mechanisms and optimizing patient selection. This review underscores the importance of dual checkpoint blockade and innovative bispecific antibody engineering to maximize therapeutic outcomes for NSCLC patients.
Collapse
Affiliation(s)
- Nicolas Roussot
- Department of Medical Oncology, Centre Georges-François Leclerc, 21000 Dijon, France; (C.K.); (F.G.)
- Cancer Biology Transfer Platform, Centre Georges-François Leclerc, 21000 Dijon, France
- Centre de Recherche INSERM LNC-UMR1231, Team TIRECs: Therapies and Immune REsponse in CancerS, 21000 Dijon, France
- UFR Sciences de Santé, Université de Bourgogne, 21000 Dijon, France
| | - Courèche Kaderbhai
- Department of Medical Oncology, Centre Georges-François Leclerc, 21000 Dijon, France; (C.K.); (F.G.)
| | - François Ghiringhelli
- Department of Medical Oncology, Centre Georges-François Leclerc, 21000 Dijon, France; (C.K.); (F.G.)
- Cancer Biology Transfer Platform, Centre Georges-François Leclerc, 21000 Dijon, France
- Centre de Recherche INSERM LNC-UMR1231, Team TIRECs: Therapies and Immune REsponse in CancerS, 21000 Dijon, France
- UFR Sciences de Santé, Université de Bourgogne, 21000 Dijon, France
| |
Collapse
|
22
|
Meng F, Xiang M, Liu Y, Zeng D. TIGIT/PVR axis regulates anti-tumor immunity in hematologic malignancies. Ann Hematol 2025; 104:1415-1426. [PMID: 40074838 PMCID: PMC12031798 DOI: 10.1007/s00277-025-06304-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Accepted: 03/04/2025] [Indexed: 03/14/2025]
Abstract
Hematologic malignancy stands as a grave form of cancer characterized by its arduous treatment and heightened likelihood of recurrence. Over the recent years, immunotherapy has progressively evolved into a pivotal approach for addressing hematologic malignancies. As a novel inhibitory receptor of NK and T cells, TIGIT is similar to PD-1, and blocking TIGIT can play a huge anti-tumor effect. At present, target TIGIT is still in clinical trials. Within this context, the TIGIT/PVR axis, serving as a pivotal element within the immunomodulatory framework, assumes a critical role in tumor immunity orchestration. This composition delves into the advancement of research concerning the TIGIT/PVR axis within hematologic malignancies, elucidating its mechanism for impeding anti-tumor immune responses. Furthermore, potential therapeutic avenues are explored, encompassing immunotherapeutic strategies aimed at targeting the TIGIT/PVR axis, alongside the conceivable integration with alternative immune checkpoint inhibitors. Ultimately, the paper encapsulates forthcoming research trajectories, aspiring to provide a compass for deeper comprehension of the TIGIT/PVR axis's role within hematologic malignancies, consequently fostering the creation of more potent immunotherapeutic tactics. This review details the therapeutic prospects of TIGIT in hematological malignancies, which is expected to advance research targeting TIGIT in hematological malignancies and bring hope for survival to these patients.
Collapse
Affiliation(s)
- Fanqiao Meng
- Department of Hematology, Daping Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Maoyuan Xiang
- Department of Hematology, Daping Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Yu Liu
- Department of Hematology, Daping Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Dongfeng Zeng
- Department of Hematology, Daping Hospital, Third Military Medical University (Army Medical University), Chongqing, China.
- Department of Hematology, Daping Hospital, Third Military Medical University (Army Medical University), No.10, Daping Changjiang Branch Road, Yuzhong District, Chongqing, 400042, China.
| |
Collapse
|
23
|
Bloom M, Podder S, Dang H, Lin D. Advances in Immunotherapy in Hepatocellular Carcinoma. Int J Mol Sci 2025; 26:1936. [PMID: 40076561 PMCID: PMC11900920 DOI: 10.3390/ijms26051936] [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: 12/20/2024] [Revised: 02/10/2025] [Accepted: 02/13/2025] [Indexed: 03/14/2025] Open
Abstract
Over the past several years, the therapeutic landscape for patients with advanced, unresectable, or metastatic hepatocellular carcinoma has been transformed by the incorporation of checkpoint inhibitor immunotherapy into the treatment paradigm. Frontline systemic treatment options have expanded beyond anti-angiogenic tyrosine kinase inhibitors, such as sorafenib, to a combination of immunotherapy approaches, including atezolizumab plus bevacizumab and durvalumab plus tremelimumab, both of which have demonstrated superior response and survival to sorafenib. Additionally, combination treatments with checkpoint inhibitors and tyrosine kinase inhibitors have been investigated with variable success. In this review, we discuss these advances in systemic treatment with immunotherapy, with a focus on understanding both the underlying biology and mechanism of these strategies and their efficacy outcomes in clinical trials. We also review challenges in identifying predictive biomarkers of treatments and discuss future directions with novel immunotherapy targets.
Collapse
Affiliation(s)
- Matthew Bloom
- Department of Medical Oncology, Sidney Kimmel Cancer Center, Thomas Jefferson University Hospital, Philadelphia, PA 19107, USA;
| | - Sourav Podder
- Department of Surgery, Thomas Jefferson University Hospital, Philadelphia, PA 19107, USA; (S.P.); (H.D.)
| | - Hien Dang
- Department of Surgery, Thomas Jefferson University Hospital, Philadelphia, PA 19107, USA; (S.P.); (H.D.)
| | - Daniel Lin
- Department of Medical Oncology, Sidney Kimmel Cancer Center, Thomas Jefferson University Hospital, Philadelphia, PA 19107, USA;
| |
Collapse
|
24
|
Shi H, Yu TK, Johnson B, Selvamani SP, Zhuang L, Lee K, Klebe S, Smith S, Wong K, Chen K, Clark G, Rath EM, Pearson H, Ortega DG, Linton A, Kao S, Silveira P, Cheng YY. A combination of PD-1 and TIGIT immune checkpoint inhibitors elicits a strong anti-tumour response in mesothelioma. J Exp Clin Cancer Res 2025; 44:51. [PMID: 39939955 PMCID: PMC11816573 DOI: 10.1186/s13046-025-03314-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Accepted: 02/02/2025] [Indexed: 02/14/2025] Open
Abstract
BACKGROUND Finding effective and curative treatment for mesothelioma remains challenging. While the introduction of immunotherapy combinations using ipilimumab (anti-CTLA-4) and nivolumab (anti-PD-1) have offered hope for some patients, a large proportion of mesothelioma cases, particularly the epithelial subtype, have minimal benefit from this. METHODS Our study was inspired by the results of the AdvanTG-105 phase I clinical trial, which showed partial response with anti-TIGIT/PD-1 treatment in two epithelioid mesothelioma patients. Here, we conducted a comprehensive in vivo experiment involving eight animal treatment groups administered with either PBS (control group), cisplatin/pemetrexed, anti-PD-1, anti-PD-1 + anti-CTLA-4, anti-TIGIT, anti-PD-1 + anti-TIGIT, anti-PD-1 + anti-CTLA-4 + anti-TIGIT, and cisplatin/pemetrexed + anti-PD-1 + anti-TIGIT. RESULTS Our results indicate that animals receiving anti-PD-1 + TIGIT exhibited a superior anti-tumour response, with 90% of the treatment group exhibiting an objective response, compared to 60%, 20% and 40% for the standard-of-care anti-PD-1 + CTLA-4, single-agent anti-PD-1 and cisplatin/pemetrexed treatment groups, respectively. Animals receiving anti-PD-1 + TIGIT displayed a significantly reduced average tumour size, with improved weight and survival rates, and fewer adverse effects than those receiving anti-PD-1 + CTLA-4 treatment. Anti-PD-1 + TIGIT-treated animals achieved complete tumour regression, with heightened effector CD8 + T cell and NK cell activity, remaining tumour-free for over 300 days without immune-related adverse events. After initial tumour elimination, anti-PD-1 + TIGIT-treated animals showed no tumour regrowth in the rechallenge experiment. CONCLUSION These findings provide rationale for the development of an anti-PD-1 + TIGIT combination immunotherapy trial for mesothelioma patients.
Collapse
Affiliation(s)
- Huaikai Shi
- Asbestos and Dust Disease Research Institute (ADDRI), 3 Hospital Road, Sydney, NSW, Concord, NSW, 2139, Australia.
| | - Ta-Kun Yu
- Asbestos and Dust Disease Research Institute (ADDRI), 3 Hospital Road, Sydney, NSW, Concord, NSW, 2139, Australia
| | - Ben Johnson
- Asbestos and Dust Disease Research Institute (ADDRI), 3 Hospital Road, Sydney, NSW, Concord, NSW, 2139, Australia
| | - Sakthi Priya Selvamani
- Asbestos and Dust Disease Research Institute (ADDRI), 3 Hospital Road, Sydney, NSW, Concord, NSW, 2139, Australia
| | - Ling Zhuang
- Asbestos and Dust Disease Research Institute (ADDRI), 3 Hospital Road, Sydney, NSW, Concord, NSW, 2139, Australia
| | - Kenneth Lee
- Sydney Medical School, University of Sydney, Sydney, NSW, 2050, Australia
- SydPath, St. Vincent Health, Darlinghurst, Sydney, NSW, 2010, Australia
- Institute for Biomedical Materials & Devices (IBMD), Faculty of Science, The University of Technology Sydney, Ultimo, NSW, 15 Broadway, Ultimo, NSW, 2007, Australia
| | - Sonja Klebe
- Asbestos and Dust Disease Research Institute (ADDRI), 3 Hospital Road, Sydney, NSW, Concord, NSW, 2139, Australia
- Pathology, Flinders Health and Medical Research Institute, Flinders University, Bedford Park, South, 5042, Australia
| | - Samuel Smith
- Department of Medical Oncology, Chris O'Brien Lifehouse, Sydney, NSW, 2050, Australia
| | - Kirby Wong
- Department of Radiology, Royal Prince Alfred Hospital, Sydney, 2050, Australia
| | - Kate Chen
- Dendritic Cell Research Group, ANZAC Research Institute, Sydney, NSW, 2139, Australia
| | - Georgina Clark
- Sydney Medical School, University of Sydney, Sydney, NSW, 2050, Australia
- Dendritic Cell Research Group, ANZAC Research Institute, Sydney, NSW, 2139, Australia
| | - Emma M Rath
- Victor Chang Cardiac Research Institute, 405 Liverpool St, Darlinghurst, NSW, 2010, Australia
- School of Clinical Medicine, Faculty of Medicine and Health, UNSW Sydney, Kensington, NSW, Australia
| | - Holly Pearson
- Single Cell Genomics Facility, School of Biomedical Engineering, Faculty of Engineering and IT, The University of Technology Sydney, Ultimo, NSW, 2007, Australia
| | - David Gallego Ortega
- Single Cell Genomics Facility, School of Biomedical Engineering, Faculty of Engineering and IT, The University of Technology Sydney, Ultimo, NSW, 2007, Australia
- School of Clinical Medicine, Faculty of Medicine and Health, University of New South Wales, Kensington, NSW, 2033, Australia
- The Kinghorn Cancer Centre, Garvan Institute of Medical Research, 380 Victoria St. Darlinghurst, Sydney, NSW, 2010, Australia
| | - Anthony Linton
- Asbestos and Dust Disease Research Institute (ADDRI), 3 Hospital Road, Sydney, NSW, Concord, NSW, 2139, Australia
- Sydney Medical School, University of Sydney, Sydney, NSW, 2050, Australia
| | - Steven Kao
- Asbestos and Dust Disease Research Institute (ADDRI), 3 Hospital Road, Sydney, NSW, Concord, NSW, 2139, Australia
- Sydney Medical School, University of Sydney, Sydney, NSW, 2050, Australia
- School of Clinical Medicine, Faculty of Medicine and Health, University of New South Wales, Kensington, NSW, 2033, Australia
| | - Pablo Silveira
- Dendritic Cell Research Group, ANZAC Research Institute, Sydney, NSW, 2139, Australia
| | - Yuen Yee Cheng
- Institute for Biomedical Materials & Devices (IBMD), Faculty of Science, The University of Technology Sydney, Ultimo, NSW, 15 Broadway, Ultimo, NSW, 2007, Australia.
| |
Collapse
|
25
|
Naing A, McKean M, Tolcher A, Victor A, Hu P, Gao W, Nogueira Filho MAF, Kitzing T, Gleicher S, Holland D, Richter E, Tadjalli-Mehr K, Siu LL. TIGIT inhibitor M6223 as monotherapy or in combination with bintrafusp alfa in patients with advanced solid tumors: a first-in-human, phase 1, dose-escalation trial. J Immunother Cancer 2025; 13:e010584. [PMID: 39929671 PMCID: PMC11815413 DOI: 10.1136/jitc-2024-010584] [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: 10/07/2024] [Accepted: 01/20/2025] [Indexed: 02/14/2025] Open
Abstract
BACKGROUND M6223 is an intravenous (IV), Fc-competent, fully human, antagonistic, anti-T cell immunoreceptor with immunoglobulin and immunoreceptor tyrosine-based inhibitory motif domains (TIGIT) antibody. Bintrafusp alfa (BA) is a bifunctional fusion protein that simultaneously blocks nonredundant immunosuppressive TGF-β and PD-(L)1 pathways. METHODS This first-in-human, dose-escalation study in patients with advanced solid tumors (N=58; aged ≥18 years, ECOG PS≤1) evaluated M6223 alone (Part 1A, n=40; M6223 10-2400 mg every 2 weeks, n=32; M6223 2400 mg every 3 weeks, n=8) or with BA (Part 1B, n=18; M6223 300-1600 mg with BA 1200 mg; both every 2 weeks, intravenous). Primary objectives were safety, tolerability, maximum tolerated dose (MTD) and recommended dose for expansion (RDE). Additional objectives included pharmacokinetics, pharmacodynamics and clinical activity (NCT04457778). RESULTS Two dose-limiting toxicities were observed: grade 3 adrenal insufficiency (Part 1A: M6223 900 mg every 2 weeks) and grade 3 anemia (Part 1B: M6223 300 mg, only BA related). MTD was not reached. Overall, median overall survival and progression-free survival were 7.6 (95% CI 4.9, 12.0) and 1.4 (95% CI 1.3, 1.8) months, respectively. Stable disease as best response was observed in 13 (32.5%) and 5 (27.8%) patients in parts 1A and 1B, respectively. M6223±BA displayed a linear pharmacokinetic profile. Anti-TIGIT mode-of-action-related pharmacodynamic effects were observed in peripheral blood and in tumor tissue. RDEs were 1600 mg every 2 weeks or 2400 mg every 3 weeks for M6223 monotherapy and 1600+1200 mg every 2 weeks for M6223+BA. CONCLUSIONS M6223±BA had a manageable safety profile, with RDEs defined for both monotherapy and combination therapy. Further evaluation of M6223 is ongoing in combination with the PD-L1 inhibitor avelumab in patients with advanced urothelial carcinoma (JAVELIN Bladder Medley; NCT05327530). TRIAL REGISTRATION NUMBER NCT04457778.
Collapse
Affiliation(s)
- Aung Naing
- The University of Texas MD Anderson Cancer Centre, Houston, Texas, USA
| | | | | | | | - Ping Hu
- EMD Serono, Billerica, Massachusetts, USA
| | - Wei Gao
- EMD Serono, Billerica, Massachusetts, USA
| | | | | | | | | | | | | | - Lillian L Siu
- Princess Margaret Hospital Cancer Centre, Toronto, Ontario, Canada
| |
Collapse
|
26
|
Ke J, Huang S, He Z, Lei S, Lin S, Duan M. TIGIT Regulates T Cell Inflammation in Airway Inflammatory Diseases. Inflammation 2025; 48:15-24. [PMID: 38780694 DOI: 10.1007/s10753-024-02045-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2024] [Revised: 05/06/2024] [Accepted: 05/06/2024] [Indexed: 05/25/2024]
Abstract
TIGIT, a co-inhibitory receptor found on T cells and NK cells, transmits inhibitory signals upon binding to its ligand. This interaction suppresses the activation of various signaling pathways, leading to functional exhaustion of cells, ultimately dampening excessive inflammatory responses or facilitating immune evasion in tumors. Dysregulated TIGIT expression has been noted in T cells across different inflammatory conditions, exhibiting varying effects based on T cell subsets. TIGIT predominantly restrains the effector function of pro-inflammatory T cells, upholds the suppressive function of regulatory T cells, and influences Tfh maturation. Mechanistically, the IL27-induced transcription factors c-Maf and Blimp-1 are believed to be key regulators of TIGIT expression in T cells. Notably, TIGIT expression in T cells is implicated in lung diseases, particularly airway inflammatory conditions such as lung cancer, obstructive pulmonary disease, interstitial lung disease, sarcoidosis, and COVID-19. This review emphasizes the significance of TIGIT in the context of T cell immunity and airway inflammatory diseases.
Collapse
Affiliation(s)
- Junyi Ke
- Guangxi Medical University, Nanning, China
- The Second Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Shu Huang
- Wuming Hospital of Guangxi Medical University, Nanning, China
| | | | - Siyu Lei
- Wuming Hospital of Guangxi Medical University, Nanning, China
| | - Shiya Lin
- Guangxi Medical University, Nanning, China
| | - Minchao Duan
- Wuming Hospital of Guangxi Medical University, Nanning, China.
- Department of Respiratory Medicine, Wuming Hospital of Guangxi Medical University, No.26 Yongning Road, Wuming District, Nanning, 530100, China.
| |
Collapse
|
27
|
Li S, Pan Y, Ye R, Wang Y, Li L. Immune checkpoints in B-cell Lymphoma: Still an Unmet challenge from Basic research to clinical practice. Int Immunopharmacol 2025; 146:113717. [PMID: 39673995 DOI: 10.1016/j.intimp.2024.113717] [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: 08/11/2024] [Revised: 11/24/2024] [Accepted: 11/24/2024] [Indexed: 12/16/2024]
Abstract
In the last decade, advancements in immunotherapy knowledge have highlighted CTLA-4, PD-1, LAG-3, TIM-3, and TIGIT, decisive immune checkpoints exhibiting within the tumor microenvironment (TME), as fundamental objects for cancer immunotherapy. The widespread clinical use of immune checkpoint inhibitors (ICls), employing PD-1/PD-L1 or CTLA-4 antibodies to obstruct crucial checkpoint regulators, is noted in treating B-cell lymphoma patients. Nevertheless, the prolonged advantages of the currently employed treatments against CTLA-4, PD-1, and PD-L1 are uncommon among patients. Thus, recent focus has been progressively moved to additional immune checkpoints on T cells, like LAG-3, TIM-3, and TIGIT, which are now seen as reassuring targets for treatment and broadly acknowledged. There are several types of immunecheckpoint molecules expressed by T cells, and inhibitors targeting immune checkpoints can revive and amplify the immune response of T lymphocytes against tumors, a crucial aspect in lymphoma therapy. However, there is little knowledge about their regulation. Herein, we discuss the anti-tumor effects and functions of ICIs in controlling T-cell activity, as well as the progress in combined application with other immunotherapies.
Collapse
Affiliation(s)
- Sijia Li
- Department of Hematology, The Second Hospital of Dalian Medical University, Dalian, PR China
| | - Yuanyuan Pan
- Department of Hematology, The Second Hospital of Dalian Medical University, Dalian, PR China
| | - Ruyu Ye
- Department of Hematology, The Second Hospital of Dalian Medical University, Dalian, PR China
| | - Yu Wang
- Department of Hematology, The Second Hospital of Dalian Medical University, Dalian, PR China
| | - Li Li
- Department of Hematology, The Second Hospital of Dalian Medical University, Dalian, PR China.
| |
Collapse
|
28
|
Chennareddy S, Rindler K, Ruggiero JR, Alkon N, Cohenour E, Tran S, Weninger W, Griss J, Jonak C, Brunner PM. Single-cell RNA sequencing comparison of CD4+, CD8+ and T-cell receptor γδ+ cutaneous T-cell lymphomas reveals subset-specific molecular phenotypes. Br J Dermatol 2025; 192:269-282. [PMID: 39133553 PMCID: PMC11758594 DOI: 10.1093/bjd/ljae313] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Revised: 07/17/2024] [Accepted: 07/29/2024] [Indexed: 09/15/2024]
Abstract
BACKGROUND Malignant clones of primary cutaneous T-cell lymphomas (CTCL) can show a CD4+, CD8+ or T-cell receptor (TCR)-γδ+ phenotype, but their individual impact on tumour biology and skin lesion formation remains ill defined. OBJECTIVES To perform a comprehensive molecular characterization of CD4+ vs. CD8+ and TCR-γδ+ CTCL lesions. METHODS We performed single-cell RNA sequencing (scRNAseq) of 18 CTCL skin biopsies to compare classic CD4+ advanced-stage mycosis fungoides (MF) with TCR-γ/δ+ MF and primary cutaneous CD8+ aggressive epidermotropic cytotoxic T-cell lymphoma (Berti lymphoma). RESULTS Malignant clones of TCR-γ/δ+ MF and Bertilymphoma showed similar clustering patterns distinct from CD4+ MF, along with increased expression of cytotoxic markers such as NKG7, CTSW, GZMA and GZMM. Only advanced-stage CD4+ MF clones expressed central memory T-cell markers (SELL, CCR7, LEF1), alongside B1/B2 blood involvement, whereas TCR-γδ+ MF and Berti lymphoma harboured a more tissue-resident phenotype (CD69, CXCR4, NR4A1) without detectable cells in the blood. CD4+ MF and TCR-γδ+ MF skin lesions harboured strong type 2 immune activation across myeloid cells, while Berti lymphoma was more skewed toward type 1 immune responses. Both CD4+ MF and TCR-γδ+ MF lesions showed upregulation of keratinocyte hyperactivation markers such as S100A genes and KRT16. This increase was entirely absent in Berti lymphoma, possibly reflecting an aberrant keratinocyte response to invading tumour cells, which could contribute to the formation of the typical ulceronecrotic lesions within this entity. CONCLUSIONS Our scRNAseq profiling study reveals specific molecular patterns associated with distinct CTCL subtypes.
Collapse
MESH Headings
- Humans
- Skin Neoplasms/genetics
- Skin Neoplasms/pathology
- Skin Neoplasms/immunology
- Lymphoma, T-Cell, Cutaneous/genetics
- Lymphoma, T-Cell, Cutaneous/pathology
- Lymphoma, T-Cell, Cutaneous/immunology
- Receptors, Antigen, T-Cell, gamma-delta/metabolism
- Male
- Single-Cell Analysis
- Female
- CD4-Positive T-Lymphocytes/immunology
- Middle Aged
- Phenotype
- CD8-Positive T-Lymphocytes/immunology
- Aged
- Skin/pathology
- Skin/immunology
- Adult
- Sequence Analysis, RNA
- Mycosis Fungoides/genetics
- Mycosis Fungoides/pathology
- Mycosis Fungoides/immunology
Collapse
Affiliation(s)
- Sumanth Chennareddy
- Department of Dermatology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Katharina Rindler
- Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - John R Ruggiero
- Department of Dermatology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Natalia Alkon
- Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - Emry R Cohenour
- Department of Dermatology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Sophia Tran
- Department of Dermatology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Wolfgang Weninger
- Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - Johannes Griss
- Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - Constanze Jonak
- Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - Patrick M Brunner
- Department of Dermatology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| |
Collapse
|
29
|
Drommi F, Calabrò A, Pezzino G, Vento G, Freni J, Costa G, Cavaliere R, Bonaccorsi I, Allegra A, Ferlazzo G, De Pasquale C, Campana S. Multiple Myeloma Cells Shift the Fate of Cytolytic ILC2s Towards TIGIT-Mediated Cell Death. Cancers (Basel) 2025; 17:263. [PMID: 39858045 PMCID: PMC11763689 DOI: 10.3390/cancers17020263] [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: 11/29/2024] [Revised: 01/05/2025] [Accepted: 01/13/2025] [Indexed: 01/27/2025] Open
Abstract
BACKGROUND Growing evidence attests to the multifaceted roles of group 2 innate lymphoid cells (ILC2s) in cancer immunity. They exhibit either pro- or anticancer activity depending on tumor type but their function in Multiple Myeloma (MM) is still not elucidated. METHODS The bone marrow (BM) and peripheral blood (PB) of patients (pts) with MM or precancerous conditions were collected, and specific properties of ILC2 subsets were assessed by flow cytometry. RESULTS By dissecting ILC2s according to c-Kit marker, we observed that NKp30 and NKG2D were mainly confined to c-Kithi ILC2s, while levels of DNAM-1 was significantly higher in fully mature c-Kitlo cells. Among the total MM-associated ILC2s (MM-ILC2s), we observed a significant increase in c-the Kitlo subset, but the expression of DNAM-1 in these cells was significantly reduced, especially in BM. Interestingly, MM-ILC2s from PB expressed granzyme B (GZMB), but its expression was impaired in BM-ILC2s. Accordingly, MM cells were susceptible to killing by MM-ILC2s derived from PB while eluding ILC2 surveillance in BM. Indeed, in MM-ILC2s derived from BM, the downregulation of DNAM-1 is accompanied by the upregulation of TIGIT, which mediate cell death in ILC2s upon recognition of the cognate ligands expressed by MM cells. These ILC2 changes appeared in clinical precursor conditions and eventually accumulated with disease progression. CONCLUSIONS MM-ILC2s can act as cytolytic immune effectors that are fully competent in PB. However, MM cells shift ILC2 fate towards cell death in BM via the upregulation of TIGIT, thereby representing a potential therapeutic target to restore ILC2 antitumor activity.
Collapse
Affiliation(s)
- Fabiana Drommi
- Laboratory of Immunology and Biotherapy, Department Human Pathology in Adulthood and Childhood “Gaetano Barresi”, University of Messina, 98125 Messina, Italy; (F.D.); (A.C.); (G.P.); (G.C.); (R.C.); (I.B.); (C.D.P.); (S.C.)
| | - Alessia Calabrò
- Laboratory of Immunology and Biotherapy, Department Human Pathology in Adulthood and Childhood “Gaetano Barresi”, University of Messina, 98125 Messina, Italy; (F.D.); (A.C.); (G.P.); (G.C.); (R.C.); (I.B.); (C.D.P.); (S.C.)
| | - Gaetana Pezzino
- Laboratory of Immunology and Biotherapy, Department Human Pathology in Adulthood and Childhood “Gaetano Barresi”, University of Messina, 98125 Messina, Italy; (F.D.); (A.C.); (G.P.); (G.C.); (R.C.); (I.B.); (C.D.P.); (S.C.)
| | - Grazia Vento
- Department of Experimental Medicine (DIMES), University of Genoa, 16132 Genova, Italy;
| | - Josè Freni
- Laboratory of Histology, Department of Biomedical, Dental, Morphological and Functional Imaging Sciences, University of Messina, 98125 Messina, Italy;
| | - Gregorio Costa
- Laboratory of Immunology and Biotherapy, Department Human Pathology in Adulthood and Childhood “Gaetano Barresi”, University of Messina, 98125 Messina, Italy; (F.D.); (A.C.); (G.P.); (G.C.); (R.C.); (I.B.); (C.D.P.); (S.C.)
- Clinical Pathology Unit, University Hospital Policlinico “G. Martino”, 98125 Messina, Italy
| | - Riccardo Cavaliere
- Laboratory of Immunology and Biotherapy, Department Human Pathology in Adulthood and Childhood “Gaetano Barresi”, University of Messina, 98125 Messina, Italy; (F.D.); (A.C.); (G.P.); (G.C.); (R.C.); (I.B.); (C.D.P.); (S.C.)
- Clinical Pathology Unit, University Hospital Policlinico “G. Martino”, 98125 Messina, Italy
| | - Irene Bonaccorsi
- Laboratory of Immunology and Biotherapy, Department Human Pathology in Adulthood and Childhood “Gaetano Barresi”, University of Messina, 98125 Messina, Italy; (F.D.); (A.C.); (G.P.); (G.C.); (R.C.); (I.B.); (C.D.P.); (S.C.)
| | - Alessandro Allegra
- Division of Hematology, Department of Human Pathology in Adulthood and Childhood “Gaetano Barresi”, University of Messina, 98125 Messina, Italy;
| | - Guido Ferlazzo
- Department of Experimental Medicine (DIMES), University of Genoa, 16132 Genova, Italy;
- Unit of Experimental Pathology and Immunology, IRCCS Ospedale Policlinico San Martino, 16132 Genova, Italy
| | - Claudia De Pasquale
- Laboratory of Immunology and Biotherapy, Department Human Pathology in Adulthood and Childhood “Gaetano Barresi”, University of Messina, 98125 Messina, Italy; (F.D.); (A.C.); (G.P.); (G.C.); (R.C.); (I.B.); (C.D.P.); (S.C.)
| | - Stefania Campana
- Laboratory of Immunology and Biotherapy, Department Human Pathology in Adulthood and Childhood “Gaetano Barresi”, University of Messina, 98125 Messina, Italy; (F.D.); (A.C.); (G.P.); (G.C.); (R.C.); (I.B.); (C.D.P.); (S.C.)
| |
Collapse
|
30
|
Glaviano A, Lau HSH, Carter LM, Lee EHC, Lam HY, Okina E, Tan DJJ, Tan W, Ang HL, Carbone D, Yee MYH, Shanmugam MK, Huang XZ, Sethi G, Tan TZ, Lim LHK, Huang RYJ, Ungefroren H, Giovannetti E, Tang DG, Bruno TC, Luo P, Andersen MH, Qian BZ, Ishihara J, Radisky DC, Elias S, Yadav S, Kim M, Robert C, Diana P, Schalper KA, Shi T, Merghoub T, Krebs S, Kusumbe AP, Davids MS, Brown JR, Kumar AP. Harnessing the tumor microenvironment: targeted cancer therapies through modulation of epithelial-mesenchymal transition. J Hematol Oncol 2025; 18:6. [PMID: 39806516 PMCID: PMC11733683 DOI: 10.1186/s13045-024-01634-6] [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: 04/20/2024] [Accepted: 11/11/2024] [Indexed: 01/16/2025] Open
Abstract
The tumor microenvironment (TME) is integral to cancer progression, impacting metastasis and treatment response. It consists of diverse cell types, extracellular matrix components, and signaling molecules that interact to promote tumor growth and therapeutic resistance. Elucidating the intricate interactions between cancer cells and the TME is crucial in understanding cancer progression and therapeutic challenges. A critical process induced by TME signaling is the epithelial-mesenchymal transition (EMT), wherein epithelial cells acquire mesenchymal traits, which enhance their motility and invasiveness and promote metastasis and cancer progression. By targeting various components of the TME, novel investigational strategies aim to disrupt the TME's contribution to the EMT, thereby improving treatment efficacy, addressing therapeutic resistance, and offering a nuanced approach to cancer therapy. This review scrutinizes the key players in the TME and the TME's contribution to the EMT, emphasizing avenues to therapeutically disrupt the interactions between the various TME components. Moreover, the article discusses the TME's implications for resistance mechanisms and highlights the current therapeutic strategies toward TME modulation along with potential caveats.
Collapse
Affiliation(s)
- Antonino Glaviano
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies, University of Palermo, 90123, Palermo, Italy
| | - Hannah Si-Hui Lau
- Division of Cellular and Molecular Research, Humphrey Oei Institute of Cancer Research, National Cancer Centre Singapore, Singapore, 169610, Singapore
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore
| | - Lukas M Carter
- Department of Medical Physics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - E Hui Clarissa Lee
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117600, Singapore
- NUS Center for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119228, Singapore
| | - Hiu Yan Lam
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117600, Singapore
- NUS Center for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119228, Singapore
| | - Elena Okina
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117600, Singapore
- NUS Center for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119228, Singapore
| | - Donavan Jia Jie Tan
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117600, Singapore
- NUS Center for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119228, Singapore
- School of Chemical and Life Sciences, Singapore Polytechnic, Singapore, 139651, Singapore
| | - Wency Tan
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117600, Singapore
- NUS Center for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119228, Singapore
- School of Chemical and Life Sciences, Singapore Polytechnic, Singapore, 139651, Singapore
| | - Hui Li Ang
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117600, Singapore
- NUS Center for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119228, Singapore
| | - Daniela Carbone
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies, University of Palermo, 90123, Palermo, Italy
| | - Michelle Yi-Hui Yee
- Division of Cellular and Molecular Research, Humphrey Oei Institute of Cancer Research, National Cancer Centre Singapore, Singapore, 169610, Singapore
| | - Muthu K Shanmugam
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117600, Singapore
- NUS Center for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119228, Singapore
| | - Xiao Zi Huang
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, 117599, Singapore
| | - Gautam Sethi
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117600, Singapore
- NUS Center for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119228, Singapore
| | - Tuan Zea Tan
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, 117599, Singapore
| | - Lina H K Lim
- Division of Cellular and Molecular Research, Humphrey Oei Institute of Cancer Research, National Cancer Centre Singapore, Singapore, 169610, Singapore
- Immunology Program, Life Sciences Institute, National University of Singapore, Singapore, 117456, Singapore
- Immunology Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119228, Singapore
| | - Ruby Yun-Ju Huang
- School of Medicine and Graduate Institute of Oncology, College of Medicine, National Taiwan University, Taipei, 10051, Taiwan
- Department of Obstetrics & Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117456, Singapore
| | - Hendrik Ungefroren
- First Department of Medicine, University Hospital Schleswig-Holstein (UKSH), Campus Lübeck, 23538, Lübeck, Germany
| | - Elisa Giovannetti
- Department of Medical Oncology, Cancer Center Amsterdam, UMC, Vrije Universiteit, HV Amsterdam, 1081, Amsterdam, The Netherlands
- Cancer Pharmacology Lab, Fondazione Pisana Per La Scienza, 56017, San Giuliano, Italy
| | - Dean G Tang
- Department of Pharmacology and Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, 14263, USA
- Experimental Therapeutics (ET) Graduate Program, University at Buffalo & Roswell Park Comprehensive Cancer Center, Buffalo, NY, 14263, USA
| | - Tullia C Bruno
- Department of Immunology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Peng Luo
- Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Mads Hald Andersen
- National Center for Cancer Immune Therapy, Department of Oncology, Herlev and Gentofte Hospital, Herlev, Denmark
| | - Bin-Zhi Qian
- Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, The Human Phenome Institute, Zhangjiang-Fudan International Innovation Center, Fudan University, Shanghai, China
| | - Jun Ishihara
- Department of Bioengineering, Imperial College London, London, W12 0BZ, UK
| | - Derek C Radisky
- Department of Cancer Biology, Mayo Clinic, Jacksonville, FL, 32224, USA
| | - Salem Elias
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Saurabh Yadav
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Minah Kim
- Herbert Irving Comprehensive Cancer Center, Columbia University, New York, NY, USA
| | - Caroline Robert
- Department of Cancer Medicine, Inserm U981, Gustave Roussy Cancer Center, Université Paris-Saclay, Villejuif, France
- Faculty of Medicine, University Paris-Saclay, Kremlin Bicêtre, Paris, France
| | - Patrizia Diana
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies, University of Palermo, 90123, Palermo, Italy
| | - Kurt A Schalper
- Department of Pathology, Yale School of Medicine, Yale University, New Haven, CT, USA
| | - Tao Shi
- Swim Across America and Ludwig Collaborative Laboratory, Department of Pharmacology, Weill Cornell Medicine, New York, NY, USA
| | - Taha Merghoub
- Swim Across America and Ludwig Collaborative Laboratory, Department of Pharmacology, Weill Cornell Medicine, New York, NY, USA
- Sandra and Edward Meyer Cancer Center, Department of Medicine, Parker Institute for Cancer Immunotherapy, Weill Cornell Medicine, New York, NY, USA
| | - Simone Krebs
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Anjali P Kusumbe
- Tissue and Tumor Microenvironment Group, MRC Human Immunology Unit, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, OX3 9DS, UK
| | - Matthew S Davids
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Jennifer R Brown
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Alan Prem Kumar
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117600, Singapore.
- NUS Center for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119228, Singapore.
| |
Collapse
|
31
|
Song L, Deng Y, Huang J, Zhu X, Zhong Y, Zhong Q, Zhou W, Liu Y, Zhao H, Ge W, Liu D. Effect of curcumin regulated memory Th7 cells in mice with DSS-induced colitis. Int Immunopharmacol 2025; 145:113770. [PMID: 39642569 DOI: 10.1016/j.intimp.2024.113770] [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: 05/11/2024] [Revised: 11/03/2024] [Accepted: 11/30/2024] [Indexed: 12/09/2024]
Abstract
Abnormal activation or dysfunction of memory helper T (mTh) cells is closely associated with the development of ulcerative colitis (UC). Curcumin (Cur), the main component of turmeric, plays a critical role in the treatment of UC due to its favorable anti-inflammatory and immunomodulatory bioactivities. However, whether Cur modulates mTh7 cells to alleviate UC is unknown. In the present study, dextran sulphate sodium (DSS) was administered to establish a colitis model in mice. Our current findings indicated that Cur effectively ameliorated the manifestations of colitis in mice, and had a significant effect in reducing disease activity index (DAI), as well as in the colonic weight and the proportion of colonic weight to colonic length. While Cur reduced the pathological injuries of the colon, restore the length of the colon, inhibited the secretion of IL-7 and IL-21, restored the secretion of IL-2, IL-4, and IL-10. Furthermore, Cur had a regulatory effect on mTh7 cells and their subpopulation status. The results of molecular docking simulations and Surface Plasmon Resonance (SPR) indicated that Cur demonstrates strong interaction capabilities with both IL-7 and IL-7R and reduced the expression levels of IL-7/IL-7R mRNA and protein. It is suggested that the alleviation of DSS-induced colitis by Cur may be achieved by reducing the level of mTh7 cells and inhibiting the activation of IL-7/IL-7R signaling.
Collapse
Affiliation(s)
- Lizhao Song
- Department of Postgraduate, Jiangxi University of Chinese Medicine, Nanchang 330004, Jiangxi Province, China
| | - Yifei Deng
- Clinical Medical School, Jiangxi University of Chinese Medicine, Nanchang 330004, Jiangxi Province, China
| | - Jiaqi Huang
- Department of Postgraduate, Jiangxi University of Chinese Medicine, Nanchang 330004, Jiangxi Province, China
| | - Xiyan Zhu
- Department of Postgraduate, Jiangxi University of Chinese Medicine, Nanchang 330004, Jiangxi Province, China
| | - Youbao Zhong
- Laboratory Animal Research Center for Science and Technology, Jiangxi University of Chinese Medicine, Nanchang 330004, Jiangxi Province, China
| | - Qin Zhong
- School of Nursing, Jiangxi University of Chinese Medicine, Nanchang 330004, Jiangxi Province, China
| | - Wen Zhou
- Nanchang Medical College, Nanchang 330052, Jiangxi Province, China
| | - Yali Liu
- Nanchang Medical College, Nanchang 330052, Jiangxi Province, China
| | - Haimei Zhao
- Formula-Pattern Research Center of Jiangxi University of Chinese Medicine, Nanchang 330004, Jiangxi Province, China.
| | - Wei Ge
- Department of Proctology, Affiliated Hospital of Jiangxi University of Chinese Medicine, 1688 Meiling Road, Nanchang 330004, Jiangxi Province, China.
| | - Duanyong Liu
- School of Nursing, Jiangxi University of Chinese Medicine, Nanchang 330004, Jiangxi Province, China; Formula-Pattern Research Center of Jiangxi University of Chinese Medicine, Nanchang 330004, Jiangxi Province, China.
| |
Collapse
|
32
|
Ghasempour A, Mohseni R, Sharif PM, Hamidieh AA. Natural killer cell-based therapies in neuroblastoma. Cell Immunol 2025; 407:104898. [PMID: 39631142 DOI: 10.1016/j.cellimm.2024.104898] [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: 08/05/2024] [Revised: 11/21/2024] [Accepted: 11/23/2024] [Indexed: 12/07/2024]
Abstract
Neuroblastoma (NB) is the most common extracranial solid tumor of childhood forming around 15 % of all pediatric tumors. Despite advances in the treatment of NB, high-risk patients still face a grave prognosis. Adoptive cell therapies based on NK cells are becoming an assistive treatment for such cases. Moreover, there is also evidence that NKT-based therapies have promising results in the management of NB. Lower complications in comparison with adoptive T cell therapies, various cell sources, and miscellaneous tumor recognition mechanisms are some of the advantages of NK- and NKT-based therapies. This review is dedicated to searching for recent advances in this field.
Collapse
Affiliation(s)
- Abtin Ghasempour
- Pediatric Cell and Gene Therapy Research Center, Gene, Cell & Tissue Research Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Rashin Mohseni
- Pediatric Cell and Gene Therapy Research Center, Gene, Cell & Tissue Research Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Pouya Mahdavi Sharif
- Pediatric Cell and Gene Therapy Research Center, Gene, Cell & Tissue Research Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Amir Ali Hamidieh
- Pediatric Cell and Gene Therapy Research Center, Gene, Cell & Tissue Research Institute, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
33
|
Wistuba‐Hamprecht K, Oberg H, Wesch D. Function and Spatial Organization of Tumor-Invasive Human γδ T Cells-What Do We Know? Eur J Immunol 2025; 55:e202451075. [PMID: 39623788 PMCID: PMC11739682 DOI: 10.1002/eji.202451075] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Revised: 11/15/2024] [Accepted: 11/18/2024] [Indexed: 01/18/2025]
Abstract
Human gammadelta (γδ) T cells not only infiltrate or reside in healthy tissues but also enter solid cancers. A large body of evidence suggests that γδ T cells can exert potent anti-tumor effects, although conflicting or unfavorable effects have been reported in some cancer entities. Infiltration patterns are key to understanding the complexity of the tumor microenvironment (TME) and its interplay with γδ T cells. The limited data available describe different γδ T cell subsets that are located in different areas around and within tumors. Tumor-infiltrating γδ lymphocytes (γδ TIL) exert cytotoxicity, for example, via the CD95- or TRAIL-axis, produce high amounts of granzymes, and after their activation, tumor necrosis factor (TNF)-α or IFN-γ and express immune checkpoint receptors. Under certain conditions, γδ T cell subsets can express low amounts of IL-17 and seem to contribute to immune regulation/suppression. A polarization of γδ T cells can be influenced by the TME. Inflammatory cytokines, growth factors, or tumor promoters can suppress γδ T cell functionality or even push them toward tumor promotion. To avoid this and to exploit the unique features of γδ T cell-mediated anti-cancer and immune-orchestrating capabilities in future immune therapy approaches, a growing body of preclinical but also clinical studies can be observed.
Collapse
Affiliation(s)
- Kilian Wistuba‐Hamprecht
- Skin Cancer UnitGerman Cancer Research Center (DKFZ)HeidelbergGermany
- Department of Dermatology, Venereology and AllergologyUniversity Medical Center Mannheim, Ruprecht‐Karl University of HeidelbergMannheimGermany
- DKFZ Hector Cancer InstituteUniversity Medical Center MannheimMannheimGermany
| | - Hans‐Heinrich Oberg
- Institute of ImmunologyUniversity Medical Center Schleswig‐Holstein and Christian‐Albrechts University of KielKielGermany
| | - Daniela Wesch
- Institute of ImmunologyUniversity Medical Center Schleswig‐Holstein and Christian‐Albrechts University of KielKielGermany
| |
Collapse
|
34
|
Ghaneialvar H, Jahani S, Hashemi E, Khalilzad MA, Falahi S, Rashidi MA, Majidpoor J, Najafi S. Combining anti-checkpoint immunotherapies and cancer vaccines as a novel strategy in oncological therapy: A review. Hum Immunol 2025; 86:111209. [PMID: 39662393 DOI: 10.1016/j.humimm.2024.111209] [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: 07/24/2024] [Revised: 11/17/2024] [Accepted: 11/26/2024] [Indexed: 12/13/2024]
Abstract
The field of cancer immunotherapy has experienced remarkable advancements in the treatment of human cancers over recent decades. Therapeutic cancer vaccines have been employed to elicit antitumor immune responses through the generation of specific reactions against tumor-associated antigens. Although preclinical studies have demonstrated hopeful results and at least one product is approved for clinical use, the overall efficacy of cancer vaccines remains restricted. The co-administration of anti-checkpoint antibodies alongside cancer vaccines is proposed as a potential strategy to enhance the clinical efficacy of immunotherapies. Among the various anti-checkpoint agents, monoclonal antibodies targeting CD127, OX40, and CD40 have been further investigated in combined administration with cancer vaccines, demonstrating a synergistic impact on disease outcomes in both animal models and human subjects. This combinational approach has been shown to suppress tumor regression, improve survival rates, and promote the efficacy of cancer vaccines via multiple mechanisms, including the augmentation of generation, activation, and expansion of CD8+ T cells, as well as the production of tumor-inhibitory cytokines. Importantly, the impact of the concurrent administration of anti-checkpoint agents and cancer vaccines surpass those observed with the sole vaccine, indicating that this strategy may offer significant advantages for clinical application in cancer patients. In this review, we aim to provide a comprehensive overview of the significance and therapeutic potential of the combined administration of checkpoint agonist/antagonist antibodies and cancer vaccines for human tumors.
Collapse
Affiliation(s)
- Hori Ghaneialvar
- Biotechnology and Medicinal Plants Research Center, Ilam University of Medical Sciences, Ilam, Iran
| | - Saleheh Jahani
- Department of Pathology, School of Medicine, University of California, San Diego, USA
| | - Elham Hashemi
- Department of Anatomical Sciences, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | | | - Shahab Falahi
- Zoonotic Diseases Research Center, Ilam University of Medical Sciences, Ilam, Iran
| | - Mohammad Amin Rashidi
- Department of Occupational Health and Safety, School of Public Health and Safety, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Jamal Majidpoor
- Department of Anatomy, Faculty of Medicine, Infectious Disease Research Center, Gonabad University of Medical Sciences, Gonabad, Iran.
| | - Sajad Najafi
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Cellular and Molecular Biology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
35
|
Tomarchio V, Crescenzi A, Tafuri M, Verri M, Di Cecca M, Rigacci L, Annibali O. The past, the present and the future of immune checkpoints inhibitors in multiple myeloma. Expert Rev Hematol 2025; 18:201-214. [PMID: 39987500 DOI: 10.1080/17474086.2025.2469720] [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: 01/20/2025] [Accepted: 02/17/2025] [Indexed: 02/25/2025]
Abstract
INTRODUCTION Myeloma genesis is a very complex mechanism in which the interaction between plasma cells and microenvironments with immune cells, cytokines and chemokines have a central role. In the last years, the improved knowledge of immune checkpoint models led to the development of new drugs (anti-PD1/PD-L1 axis or anti-TIGIT) that now have a crucial role in the treatment of many hematological malignancies. AREAS COVERED In this review, the current significant literature was discussed. In the past, initial trials combining immune checkpoint inhibitors (ICIs) with immunomodulatory drugs or proteasome inhibitors demonstrated suboptimal results in terms of efficacy and safety. On the other hand, recent trials based on the combination of ICIs with immunotherapies, such as CAR-T cells or bispecific antibodies, are a particularly promising area of investigation. EXPERT OPINION Our idea after the evaluation of scientific literature is that despite the past, ICIs may represent a promising therapeutic approach for myeloma, particularly when combined with CAR-T cells or bispecific antibodies. By targeting immune evasion mechanisms, ICIs may enhance the efficacy of these treatments and provide new hope for patients with resistant disease. Future research will be crucial to further elucidate their optimal use in myeloma and to develop personalized treatment strategies.
Collapse
Affiliation(s)
- Valeria Tomarchio
- Operative Research Unit of Hematology, Fondazione Policlinico Universitario Campus Bio-Medico, Roma, Italy
| | - Anna Crescenzi
- Operative Research Unit of Unit of Pathological Anatomy, Fondazione Policlinico Universitario Campus Bio-Medico, Roma, Italy
| | - Mariantonietta Tafuri
- Operative Research Unit of Hematology, Fondazione Policlinico Universitario Campus Bio-Medico, Roma, Italy
| | - Martina Verri
- Operative Research Unit of Unit of Pathological Anatomy, Fondazione Policlinico Universitario Campus Bio-Medico, Roma, Italy
| | - Monica Di Cecca
- Operative Research Unit of Hematology, Fondazione Policlinico Universitario Campus Bio-Medico, Roma, Italy
| | - Luigi Rigacci
- Operative Research Unit of Hematology, Fondazione Policlinico Universitario Campus Bio-Medico, Roma, Italy
| | - Ombretta Annibali
- Operative Research Unit of Hematology, Fondazione Policlinico Universitario Campus Bio-Medico, Roma, Italy
| |
Collapse
|
36
|
Yang X, Li J, Ren M, Pan X, Liu H, Jiang J, Li M, Yang Z, Han B, Ma L, Hao J, Duan Y, Yin Z, Xu Y, Xiang Z, Wu B. Comprehensive analysis of immune signatures in primary biliary cholangitis and autoimmune hepatitis. J Leukoc Biol 2024; 117:qiae085. [PMID: 38652703 DOI: 10.1093/jleuko/qiae085] [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: 01/07/2024] [Revised: 03/14/2024] [Accepted: 03/20/2024] [Indexed: 04/25/2024] Open
Abstract
Primary biliary cholangitis (PBC) and autoimmune hepatitis (AIH) are autoimmune diseases that target hepatocytes and bile duct cells, respectively. Despite their shared autoimmune nature, the differences in immunologic characteristics between them remain largely unexplored. This study seeks to elucidate the unique immunological profiles of PBC and AIH and to identify key differences. We comprehensively analyzed various T cell subsets and their receptor expression in a cohort of 45 patients, including 27 PBC and 18 AIH cases. Both diseases exhibited T cell exhaustion and senescence along with a surge in inflammatory cytokines. Significantly increased CD38+HLA-DR+CD8+ T cell populations were observed in both diseases. AIH was characterized by an upregulation of CD8+ terminally differentiated T, CD4+ effector memory T, and CD4+ terminally differentiated T cells, and a concurrent reduction in regulatory T cells. In contrast, PBC displayed a pronounced presence of T follicular helper (Tfh) cells and a contraction of CD4-CD8- T cell populations. Correlation analysis revealed that NKP46+ natural killer frequency was closely tied to alanine aminotransferase and aspartate aminotransferase levels, and TIGIT expression on T cells was associated with globulin level in AIH. In PBC, there is a significant correlation between Tfh cells and ALP levels. Moreover, the identified immune landscapes in both diseases strongly related to disease severity. Through logistic regression analysis, γδ T, TIGIT+Vδ2 T, and Tfh1 cell frequencies emerged as distinct markers capable of differentiating PBC from AIH. In conclusion, our analyses reveal that PBC and AIH share similarities and differences regarding to immune profiles. γδ T, TIGIT+Vδ2 T, and Tfh1 cell frequencies are potential noninvasive immunological markers that can differentiate PBC from AIH.
Collapse
Affiliation(s)
- Xiaoxue Yang
- Department of Gastroenterology, Third Affiliated Hospital of Sun Yat-Sen University, No. 600 Tianhe Road, Tianhe District, Guangzhou, Guangdong 510630, China
| | - Jiawei Li
- The Biomedical Translational Research Institute, Health Science Center (School of Medicine), Jinan University, No. 601 Huangpu W.Road, Tianhe District, Guangzhou, Guangdong 510632, China
- Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and Treatment, Zhuhai Institute of Translational Medicine, Zhuhai People's Hospital Affiliated with Jinan University, Jinan University, No. 79 Kangning Road, Xiangzhou District, Zhuhai, Guangdong 519000, China
| | - Meiling Ren
- Yuexiu District Center for Disease Control and Prevention, No. 23, Jiaochang West Road, Yuexiu District, Guangzhou, Guangdong 510120, China
| | - Xuemei Pan
- Department of Gastroenterology, Third Affiliated Hospital of Sun Yat-Sen University, No. 600 Tianhe Road, Tianhe District, Guangzhou, Guangdong 510630, China
| | - Huiling Liu
- Department of Gastroenterology, Third Affiliated Hospital of Sun Yat-Sen University, No. 600 Tianhe Road, Tianhe District, Guangzhou, Guangdong 510630, China
| | - Jie Jiang
- Department of Gastroenterology, Third Affiliated Hospital of Sun Yat-Sen University, No. 600 Tianhe Road, Tianhe District, Guangzhou, Guangdong 510630, China
| | - Man Li
- The Biomedical Translational Research Institute, Health Science Center (School of Medicine), Jinan University, No. 601 Huangpu W.Road, Tianhe District, Guangzhou, Guangdong 510632, China
- Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and Treatment, Zhuhai Institute of Translational Medicine, Zhuhai People's Hospital Affiliated with Jinan University, Jinan University, No. 79 Kangning Road, Xiangzhou District, Zhuhai, Guangdong 519000, China
| | - Zhe Yang
- The Biomedical Translational Research Institute, Health Science Center (School of Medicine), Jinan University, No. 601 Huangpu W.Road, Tianhe District, Guangzhou, Guangdong 510632, China
- Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and Treatment, Zhuhai Institute of Translational Medicine, Zhuhai People's Hospital Affiliated with Jinan University, Jinan University, No. 79 Kangning Road, Xiangzhou District, Zhuhai, Guangdong 519000, China
| | - Bingyu Han
- The Biomedical Translational Research Institute, Health Science Center (School of Medicine), Jinan University, No. 601 Huangpu W.Road, Tianhe District, Guangzhou, Guangdong 510632, China
- Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and Treatment, Zhuhai Institute of Translational Medicine, Zhuhai People's Hospital Affiliated with Jinan University, Jinan University, No. 79 Kangning Road, Xiangzhou District, Zhuhai, Guangdong 519000, China
| | - Lina Ma
- The Biomedical Translational Research Institute, Health Science Center (School of Medicine), Jinan University, No. 601 Huangpu W.Road, Tianhe District, Guangzhou, Guangdong 510632, China
- Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and Treatment, Zhuhai Institute of Translational Medicine, Zhuhai People's Hospital Affiliated with Jinan University, Jinan University, No. 79 Kangning Road, Xiangzhou District, Zhuhai, Guangdong 519000, China
| | - Jianlei Hao
- The Biomedical Translational Research Institute, Health Science Center (School of Medicine), Jinan University, No. 601 Huangpu W.Road, Tianhe District, Guangzhou, Guangdong 510632, China
- Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and Treatment, Zhuhai Institute of Translational Medicine, Zhuhai People's Hospital Affiliated with Jinan University, Jinan University, No. 79 Kangning Road, Xiangzhou District, Zhuhai, Guangdong 519000, China
| | - Yuanyuan Duan
- Department of Microbiology and Immunology, Health Science Center, School of Medicine, Jinan University, No. 601 Huangpu W.Road, Tianhe District, Guangzhou, Guangdong 510632, China
- Key Laboratory of Viral Pathogenesis and Infection Prevention and Control, Jinan University, Ministry of Education, No. 601 Huangpu W.Road, Tianhe District, Guangzhou, Guangdong 510632, China
| | - Zhinan Yin
- The Biomedical Translational Research Institute, Health Science Center (School of Medicine), Jinan University, No. 601 Huangpu W.Road, Tianhe District, Guangzhou, Guangdong 510632, China
- Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and Treatment, Zhuhai Institute of Translational Medicine, Zhuhai People's Hospital Affiliated with Jinan University, Jinan University, No. 79 Kangning Road, Xiangzhou District, Zhuhai, Guangdong 519000, China
| | - Yan Xu
- The Biomedical Translational Research Institute, Health Science Center (School of Medicine), Jinan University, No. 601 Huangpu W.Road, Tianhe District, Guangzhou, Guangdong 510632, China
- Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and Treatment, Zhuhai Institute of Translational Medicine, Zhuhai People's Hospital Affiliated with Jinan University, Jinan University, No. 79 Kangning Road, Xiangzhou District, Zhuhai, Guangdong 519000, China
| | - Zheng Xiang
- Department of Microbiology and Immunology, Health Science Center, School of Medicine, Jinan University, No. 601 Huangpu W.Road, Tianhe District, Guangzhou, Guangdong 510632, China
- Key Laboratory of Viral Pathogenesis and Infection Prevention and Control, Jinan University, Ministry of Education, No. 601 Huangpu W.Road, Tianhe District, Guangzhou, Guangdong 510632, China
| | - Bin Wu
- Department of Gastroenterology, Third Affiliated Hospital of Sun Yat-Sen University, No. 600 Tianhe Road, Tianhe District, Guangzhou, Guangdong 510630, China
| |
Collapse
|
37
|
Singer M, Zhang Z, Dayyani F, Zhang Z, Yaghmai V, Choi A, Valerin J, Imagawa D, Abi-Jaoudeh N. Modulation of Tumor-Associated Macrophages to Overcome Immune Suppression in the Hepatocellular Carcinoma Microenvironment. Cancers (Basel) 2024; 17:66. [PMID: 39796695 PMCID: PMC11718901 DOI: 10.3390/cancers17010066] [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: 12/16/2024] [Revised: 12/21/2024] [Accepted: 12/27/2024] [Indexed: 01/13/2025] Open
Abstract
Hepatocellular carcinoma (HCC) is a major global health issue characterized by poor prognosis and complex tumor biology. One of the critical components of the HCC tumor microenvironment (TME) is tumor-associated macrophages (TAMs), which play a pivotal role in modulating tumor growth, immune evasion, and metastasis. Macrophages are divided into two major subtypes: pro-inflammatory M1 and anti-inflammatory M2, both of which may exist in TME with altered function and proportion. The anti-inflammatory M2 macrophages are further subdivided into four distinct immune suppressive subsets. TAMs are generally counted as M2-like macrophages with altered immune suppressive functions that exert a significant influence on both cancer progression and the ability of tumors to escape immune surveillance. Their involvement in modulating immune responses via different mechanisms at the local and systemic levels has made them a key target for therapeutic interventions seeking to enhance treatment outcomes. How TAMs' depletion influences immune responses in cancer is the primary interest in cancer immunotherapies. The purpose of this review is to delve into the recent progress made in TAM-targeting therapies. We will explore the current theories, benefits, and challenges associated with TAMs' depletion or inhibition. The manuscript concludes with future directions and potential implications for clinical practice.
Collapse
Affiliation(s)
- Mahmoud Singer
- Department of Radiological Sciences, School of Medicine, University of California, Irvine, CA 92617, USA; (Z.Z.); (Z.Z.); (V.Y.)
| | - Zhuoli Zhang
- Department of Radiological Sciences, School of Medicine, University of California, Irvine, CA 92617, USA; (Z.Z.); (Z.Z.); (V.Y.)
| | - Farshid Dayyani
- Department of Medicine, Chao Family Comprehensive Cancer Center, University of California, Irvine, CA 92867, USA; (F.D.); (A.C.); (J.V.)
| | - Zigeng Zhang
- Department of Radiological Sciences, School of Medicine, University of California, Irvine, CA 92617, USA; (Z.Z.); (Z.Z.); (V.Y.)
| | - Vahid Yaghmai
- Department of Radiological Sciences, School of Medicine, University of California, Irvine, CA 92617, USA; (Z.Z.); (Z.Z.); (V.Y.)
| | - April Choi
- Department of Medicine, Chao Family Comprehensive Cancer Center, University of California, Irvine, CA 92867, USA; (F.D.); (A.C.); (J.V.)
| | - Jennifer Valerin
- Department of Medicine, Chao Family Comprehensive Cancer Center, University of California, Irvine, CA 92867, USA; (F.D.); (A.C.); (J.V.)
| | - David Imagawa
- Department of Surgery, University of California Irvine, Orange, CA 92697, USA;
| | - Nadine Abi-Jaoudeh
- Department of Radiological Sciences, School of Medicine, University of California, Irvine, CA 92617, USA; (Z.Z.); (Z.Z.); (V.Y.)
| |
Collapse
|
38
|
Wu Y, Zhai Y, Ding Z, Xie T, Zhu W, Zhang C, Lu Y, Chen Y, Ren S, Hu Y, Li X, Zhong F, Liang Y, Wang S. Single-cell transcriptomics reveals tumor microenvironment changes and prognostic gene signatures in hepatocellular carcinoma. Int Immunopharmacol 2024; 143:113317. [PMID: 39447409 DOI: 10.1016/j.intimp.2024.113317] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Revised: 09/23/2024] [Accepted: 10/01/2024] [Indexed: 10/26/2024]
Abstract
BACKGROUND Hepatocellular Carcinoma (HCC) is the most common type of primary liver cancer, accounting for the majority of liver cancer cases. Hepatocellular Carcinoma not only exhibits high heterogeneity but also possesses an immune-suppressive tumor microenvironment that promotes tumor evasion, posing substantial difficulties for efficient therapy. Our aim is to utilize single-cell RNA transcriptome data to investigate the dynamic changes in the tumor microenvironment during the malignant progression of HCC, the communication among immune cells, and the marker genes associated with patient prognosis. METHODS We constructed expression matrices from open single-cell RNA transcriptome data (GSE149614) of HCC patients (representing stages I-IV), establishing single-cell RNA transcriptional atlases for different stages of HCC progression. For each stage, we conducted cell subgroup analysis to identify cell types at each stage. Horizontally, we explored the dynamic changes of the same cell type across different stages, performing trajectory analysis and prognosis analysis. Vertically, we investigated pairwise comparisons of different stages of HCC progression, probing the dynamic alterations in tumor microenvironment immune cell signaling pathways. Finally, potential drugs for the treatment of HCC were predicted based on relevant genes. FINDINGS As the HCC advances towards increased malignancy, there is a shift in the predominant composition of the tumor microenvironment, with a decline in the dominance of hepatic cells. Tumor-infiltrating immune cells migrate and accumulate within the tumor microenvironment, where T cells and myeloid cells display distinct patterns of change. Genes associated with cancer-associated fibroblasts (CAFs) and T cells are correlated with adverse patient outcomes. In the late stages of HCC, the tumor microenvironment is infiltrated by more myeloid-derived suppressor cells (MDSCs), and a prognostic model constructed based on genes related to myeloid cells can predict patient outcomes. Additionally, in the analysis of transcription factors, YY1 and MYC are found to be highly expressed. Cell communication analysis among tumor-infiltrating immune cells reveals significant differences in the main signaling pathways at different stages of HCC progression. Finally, drug sensitivity analysis based on key genes identifies Acetalax, Allopurinol, and Amonafide as potential candidates for HCC treatment.
Collapse
Affiliation(s)
- Yilin Wu
- School of Life Science and Food Engineering, Huaiyin Institute of Technology, Huai'an, China
| | - Yangyang Zhai
- Shanghai Key Laboratory of Biliary Tract Disease Research, Shanghai Research Center of Biliary Tract Disease, Department of General Surgery, Xinhua Hospital, Affiliated with Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhilong Ding
- Department of Hepatobiliary Surgery, The Affiliated Huaian Hospital of Xuzhou Medical University and Huai'an Second People's Hospital, Huai'an, Jiangsu, China
| | - Tong Xie
- School of Life Science and Food Engineering, Huaiyin Institute of Technology, Huai'an, China
| | - WeiJie Zhu
- School of Life Science and Food Engineering, Huaiyin Institute of Technology, Huai'an, China
| | - Cui Zhang
- School of Life Science and Food Engineering, Huaiyin Institute of Technology, Huai'an, China
| | - Ying Lu
- School of Life Science and Food Engineering, Huaiyin Institute of Technology, Huai'an, China
| | - Yunli Chen
- School of Life Science and Food Engineering, Huaiyin Institute of Technology, Huai'an, China
| | - Shiying Ren
- School of Life Science and Food Engineering, Huaiyin Institute of Technology, Huai'an, China
| | - Yihuai Hu
- School of Life Science and Food Engineering, Huaiyin Institute of Technology, Huai'an, China
| | - Xiangqian Li
- School of Life Science and Food Engineering, Huaiyin Institute of Technology, Huai'an, China
| | - Fei Zhong
- Department of Laboratory Medicine, The Affiliated Huaian Hospital of Xuzhou Medical University and Huai'an Second People's Hospital, Huai'an, Jiangsu, China.
| | - Yong Liang
- Department of Laboratory Medicine, The Affiliated Huaian Hospital of Xuzhou Medical University and Huai'an Second People's Hospital, Huai'an, Jiangsu, China.
| | - Shiyan Wang
- School of Life Science and Food Engineering, Huaiyin Institute of Technology, Huai'an, China.
| |
Collapse
|
39
|
Pi JK, Chen XT, Zhang YJ, Chen XM, Wang YC, Xu JY, Zhou JH, Yu SS, Wu SS. Insight of immune checkpoint inhibitor related myocarditis. Int Immunopharmacol 2024; 143:113559. [PMID: 39536487 DOI: 10.1016/j.intimp.2024.113559] [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: 08/20/2024] [Revised: 10/20/2024] [Accepted: 10/30/2024] [Indexed: 11/16/2024]
Abstract
As the understanding of immune-related mechanisms in the development and progression of cancer advances, immunotherapies, notably Immune Checkpoint Inhibitors (ICIs), have become integral in comprehensive cancer treatment strategies. ICIs reactivate T-cell cytotoxicity against tumors by blocking immune suppressive signals on T cells, such as Programmed Death-1 (PD-1) and Cytotoxic T-lymphocyte Antigen-4 (CTLA-4). Despite their beneficial effects, ICIs are associated with immune-related adverse events (irAEs), manifesting as autoimmune side effects across various organ systems. A particularly alarming irAE is life-threatening myocarditis. This rare but severe side effect of ICIs leads to significant long-term cardiac complications, including arrhythmias and heart failure, and has been observed to have a mortality rate of up to 50% in affected patients. This greatly limits the clinical application of ICI-based immunotherapy. In this review, we provide a comprehensive summary of the current knowledge regarding the diagnosis and management of ICI-related myocarditis. We also discuss the utility of preclinical mouse models in understanding and addressing this critical challenge.
Collapse
Affiliation(s)
- Jin-Kui Pi
- Core Facilities, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, PR China
| | - Xiao-Ting Chen
- Animal Experimental Center, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, PR China
| | - Yan-Jing Zhang
- Core Facilities, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, PR China
| | - Xue-Mei Chen
- Core Facilities, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, PR China
| | - Yin-Chan Wang
- Core Facilities, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, PR China
| | - Jia-Yi Xu
- Core Facilities, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, PR China
| | - Jin-Han Zhou
- Core Facilities, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, PR China
| | - Shuai-Shuai Yu
- Core Facilities, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, PR China
| | - Si-Si Wu
- Core Facilities, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, PR China.
| |
Collapse
|
40
|
Arafat Hossain M. A comprehensive review of immune checkpoint inhibitors for cancer treatment. Int Immunopharmacol 2024; 143:113365. [PMID: 39447408 DOI: 10.1016/j.intimp.2024.113365] [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: 08/25/2024] [Revised: 09/28/2024] [Accepted: 10/05/2024] [Indexed: 10/26/2024]
Abstract
Immunology-based therapies are emerging as an effective cancer treatment, using the body's immune system to target tumors. Immune checkpoints, which regulate immune responses to prevent tissue damage and autoimmunity, are often exploited by cancer cells to avoid destruction. The discovery of checkpoint proteins like PD-1/PD-L1 and CTLA-4 was pivotal in developing cancer immunotherapy. Immune checkpoint inhibitors (ICIs) have shown great success, with FDA-approved drugs like PD-1 inhibitors (Nivolumab, Pembrolizumab, Cemiplimab), PD-L1 inhibitors (Atezolizumab, Durvalumab, Avelumab), and CTLA-4 inhibitors (Ipilimumab, Tremelimumab), alongside LAG-3 inhibitor Relatlimab. Research continues on new checkpoints like TIM-3, VISTA, B7-H3, BTLA, and TIGIT. Biomarkers like PDL-1 expression, tumor mutation burden, interferon-γ presence, microbiome composition, and extracellular matrix characteristics play a crucial role in predicting responses to immunotherapy with checkpoint inhibitors. Despite their effectiveness, not all patients experience the same level of benefit, and organ-specific immune-related adverse events (irAEs) such as rash or itching, colitis, diarrhea, hyperthyroidism, and hypothyroidism may occur. Given the rapid advancements in this field and the variability in patient outcomes, there is an urgent need for a comprehensive review that consolidates the latest findings on immune checkpoint inhibitors, covering their clinical status, biomarkers, resistance mechanisms, strategies to overcome resistance, and associated adverse effects. This review aims to fill this gap by providing an analysis of the current clinical status of ICIs, emerging biomarkers, mechanisms of resistance, strategies to enhance therapeutic efficacy, and assessment of adverse effects. This review is crucial to furthering our understanding of ICIs and optimizing their application in cancer therapy.
Collapse
Affiliation(s)
- Md Arafat Hossain
- Department of Pharmacy, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj 8100, Bangladesh.
| |
Collapse
|
41
|
Xue Q, Peng W, Zhang S, Wei X, Ye L, Wang Z, Xiang X, Liu Y, Wang H, Zhou Q. Lactylation-driven TNFR2 expression in regulatory T cells promotes the progression of malignant pleural effusion. J Immunother Cancer 2024; 12:e010040. [PMID: 39721754 PMCID: PMC11683941 DOI: 10.1136/jitc-2024-010040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2024] [Accepted: 11/06/2024] [Indexed: 12/28/2024] Open
Abstract
BACKGROUND Although tumor necrosis factor receptor 2 (TNFR2) has been recognized as an attractive next-generation candidate target for cancer immunotherapy, the factors that regulate the gene expression and their mechanistic effects on tumor-infiltrating regulatory T cells (Treg cells) remain poorly understood. METHODS Single-cell RNA sequencing analysis was employed to analyze the phenotypic and functional differences between TNFR2+ Treg cells and TNFR2- Treg cells. Malignant pleural effusion (MPE) from humans and mouse was used to investigate the potential mechanisms by which lactate regulates TNFR2 expression. RESULTS Treg cells with high TNFR2 expression exhibited elevated levels of immune checkpoint molecules. Additionally, the high expression of TNFR2 on Treg cells was positively correlated with a poor prognosis in MPE patients. Moreover, we revealed that lactate upregulated TNFR2 expression on Treg cells, thereby enhancing their immunosuppressive function in MPE. Mechanistically, lactate modulated the gene transcription of transcription factor nuclear factor-κB p65 (NF-κB p65) through histone H3K18 lactylation (H3K18la), subsequently upregulating the gene expression of TNFR2 and expediting the progression of MPE. Notably, lactate metabolism blockade combined with immune checkpoint blockade (ICB) therapy effectively enhanced the efficacy of ICB therapy, prolonged the survival time of MPE mice, and improved immunosuppression in the microenvironment of MPE. CONCLUSIONS The study explains the mechanism that regulates TNFR2 expression on Treg cells and its function in MPE progression, providing novel insights into the epigenetic regulation of tumor development and metabolic strategies for MPE treatment by targeting lactate metabolism in Treg cells.
Collapse
Affiliation(s)
- Qianqian Xue
- Department of Respiratory and Critical Care Medicine, Huazhong University of Science and Technology, Wuhan, China
| | - Wenbei Peng
- Department of Respiratory and Critical Care Medicine, Huazhong University of Science and Technology, Wuhan, China
| | - Siyu Zhang
- Department of Respiratory and Critical Care Medicine, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaoshan Wei
- Department of Respiratory and Critical Care Medicine, Huazhong University of Science and Technology, Wuhan, China
| | - Linlin Ye
- Department of Respiratory and Critical Care Medicine, Huazhong University of Science and Technology, Wuhan, China
| | - Zihao Wang
- Department of Respiratory and Critical Care Medicine, Huazhong University of Science and Technology, Wuhan, China
| | - Xuan Xiang
- Department of Respiratory and Critical Care Medicine, Huazhong University of Science and Technology, Wuhan, China
| | - Yao Liu
- Department of Respiratory and Critical Care Medicine, Huazhong University of Science and Technology, Wuhan, China
| | - Haolei Wang
- Department of Respiratory and Critical Care Medicine, Huazhong University of Science and Technology, Wuhan, China
| | - Qiong Zhou
- Department of Respiratory and Critical Care Medicine, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
42
|
Hosseininejad-Chafi M, Eftekhari Z, Oghalaie A, Behdani M, Sotoudeh N, Kazemi-Lomedasht F. Nanobodies as innovative immune checkpoint modulators: advancing cancer immunotherapy. Med Oncol 2024; 42:36. [PMID: 39719469 DOI: 10.1007/s12032-024-02588-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: 10/22/2024] [Accepted: 12/14/2024] [Indexed: 12/26/2024]
Abstract
The immune system relies on a delicate balance between attacking harmful pathogens and preserving the body's own tissues, a balance maintained by immune checkpoints. These checkpoints play a critical role in preventing autoimmune diseases by restraining excessive immune responses while allowing the immune system to recognize and destroy abnormal cells, such as tumors. In recent years, immune checkpoint inhibitors (ICIs) have become central to cancer therapy, enabling the immune system to target and eliminate cancer cells that evade detection. Traditional antibodies, such as IgGs, have been widely used in immune therapies but are limited by their size and complexity. Nanobodies (Nbs), derived from camelid heavy-chain-only antibodies, offer a promising alternative. These small, stable antibody fragments retain the antigen-binding specificity of traditional antibodies but have enhanced solubility and the ability to target otherwise inaccessible epitopes. This review explores the use of Nbs as ICIs, emphasizing their potential in cancer immunotherapy and other immune-related treatments. Their unique structural properties and small size make Nbs highly effective tools for modulating immune responses, representing a novel approach in the evolving landscape of checkpoint inhibitor therapies.
Collapse
Affiliation(s)
- Mohammad Hosseininejad-Chafi
- Venom and Biotherapeutics Molecules Laboratory, Biotechnology Department, Biotechnology Research Center, Pasteur Institute of Iran, Tehran, 1316943551, Iran
| | - Zohre Eftekhari
- Venom and Biotherapeutics Molecules Laboratory, Biotechnology Department, Biotechnology Research Center, Pasteur Institute of Iran, Tehran, 1316943551, Iran
| | - Akbar Oghalaie
- Venom and Biotherapeutics Molecules Laboratory, Biotechnology Department, Biotechnology Research Center, Pasteur Institute of Iran, Tehran, 1316943551, Iran
| | - Mahdi Behdani
- Venom and Biotherapeutics Molecules Laboratory, Biotechnology Department, Biotechnology Research Center, Pasteur Institute of Iran, Tehran, 1316943551, Iran
| | - Nazli Sotoudeh
- Venom and Biotherapeutics Molecules Laboratory, Biotechnology Department, Biotechnology Research Center, Pasteur Institute of Iran, Tehran, 1316943551, Iran
| | - Fatemeh Kazemi-Lomedasht
- Venom and Biotherapeutics Molecules Laboratory, Biotechnology Department, Biotechnology Research Center, Pasteur Institute of Iran, Tehran, 1316943551, Iran.
| |
Collapse
|
43
|
Qu F, Wu S, Yu W. Progress of Immune Checkpoint Inhibitors Therapy for pMMR/MSS Metastatic Colorectal Cancer. Onco Targets Ther 2024; 17:1223-1253. [PMID: 39735789 PMCID: PMC11681808 DOI: 10.2147/ott.s500281] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Accepted: 12/12/2024] [Indexed: 12/31/2024] Open
Abstract
Immunotherapy is one of the research hotspots in colorectal cancer field in recent years. The colorectal cancer patients with mismatch repair-deficient (dMMR) or high microsatellite instability (MSI-H) are the primary beneficiaries of immunotherapy. However, the vast majority of colorectal cancers are mismatch repair proficient (pMMR) or microsatellite stability (MSS), and their immune microenvironment is characterized by "cold tumors" that are generally insensitive to single immunotherapy based on immune checkpoint inhibitors (ICIs). Studies have shown that some pMMR/MSS colorectal cancer patients regulate the immune microenvironment by combining other treatments, such as multi-target tyrosine kinase inhibitors, anti-vascular endothelial growth factor (VEGF) monoclonal antibodies, chemotherapy, radiotherapy, anti-epithelial growth factor receptor (EGFR) monoclonal antibodies, and mitogen-activated protein kinase (MAPK) signaling pathway inhibitors and oncolytic viruses, etc. to transform "cold tumor" into "hot tumor", thereby improving the response to immunotherapy. In addition, screening for potential prognostic biomarkers can also enrich the population benefiting from immunotherapy for microsatellite stable colorectal cancer. Therefore, in pMMR or MSS metastatic colorectal cancer (mCRC), the optimization of immunotherapy regimens and the search for effective efficacy prediction biomarkers are currently important research directions. In this paper, we review the progress of efficacy of immunotherapy (mainly ICIs) in pMMR /MSS mCRC, challenges and potential markers, in order to provide research ideas for the development of immunotherapy for mCRC.
Collapse
Affiliation(s)
- Fanjie Qu
- Department of Oncology, Affiliated Dalian Third People’s Hospital of Dalian Medical University, Dalian, Liaoning Province, 116033, People’s Republic of China
| | - Shuang Wu
- Department of Oncology, Affiliated Dalian Third People’s Hospital of Dalian Medical University, Dalian, Liaoning Province, 116033, People’s Republic of China
| | - WeiWei Yu
- Department of Oncology, Affiliated Dalian Third People’s Hospital of Dalian Medical University, Dalian, Liaoning Province, 116033, People’s Republic of China
| |
Collapse
|
44
|
Chantziou A, Brenna C, Ioannidou K, Chen OY, Korkolopoulou P, Antoniadou A, Psichogiou M, Papaioannou M, Tsirigotis P, Foukas PG, de Leval L, Petrovas C. HIV infection is associated with compromised tumor microenvironment adaptive immune reactivity in Hodgkin lymphoma. Blood Adv 2024; 8:6215-6231. [PMID: 39116294 PMCID: PMC11697195 DOI: 10.1182/bloodadvances.2023012116] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Revised: 07/31/2024] [Accepted: 08/01/2024] [Indexed: 08/10/2024] Open
Abstract
ABSTRACT The impact of HIV infection on the tumor microenvironment (TME) of classic Hodgkin lymphoma (cHL), one of the most common comorbidities after HIV infection, is not well understood. Here, we have used multiplexed immunofluorescence and spatial transcriptomic analysis to dissect the impact of viral infections (Epstein-Barr virus [EBV] and HIV/EBV) on cHL TME. HIV-EBV+ cHL TME was characterized by higher cell densities of CD8high T cells coexpressing inhibitory receptors (PD-1 and TIGIT), macrophage subsets, and an in situ inflammatory molecular profile associated with increased expression of T-cell receptor (TCR) and B-cell receptor cell signaling pathways than HIV-EBV- cHL TME. Compared with HIV-EBV+, HIV+EBV+ cHL TME was characterized by significantly less CD8high T cells coexpressing PD-1 and TIGIT, a profile concomitant with significantly increased cell densities of CD155high neoplastic cells. Significant downregulation of in situ TCR signaling and upregulation of extracellular matrix reorganization pathways were found in HIV+EBV+ cHL TME, in line with an altered topological organization of CXCL13 and heparan sulfate, an extracellular matrix glycosaminoglycan. Our data reveal the complexity of the cellular and molecular composition of cHL TME in the presence of viral infections, with possible implications for combinatorial immunotherapies. Furthermore, the data suggest specific molecular targets and pathways for further investigation that could improve our understanding of possible mechanistic links between HIV and lymphomagenesis.
Collapse
Affiliation(s)
- Amanda Chantziou
- Department of Laboratory Medicine and Pathology, Institute of Pathology, Lausanne University Hospital, Lausanne, Switzerland
| | - Cloe Brenna
- Department of Laboratory Medicine and Pathology, Institute of Pathology, Lausanne University Hospital, Lausanne, Switzerland
| | - Kalliopi Ioannidou
- Department of Laboratory Medicine and Pathology, Institute of Pathology, Lausanne University Hospital, Lausanne, Switzerland
| | - Oliver Y. Chen
- Department of Laboratory Medicine and Pathology, Lausanne University Hospital and Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland
| | - Penelope Korkolopoulou
- First Department of Pathology, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Anastasia Antoniadou
- Fourth Department of Internal Medicine, Attikon University Hospital, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Mina Psichogiou
- First Department of Medicine, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Maria Papaioannou
- First Department of Internal Medicine, Hematology Unit, AHEPA University Hospital, School of Medicine, Aristotle University of Thessaloniki, XX, Greece
| | - Panagiotis Tsirigotis
- Division of Hematology, Second Department of Internal Medicine, Attikon University Hospital, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Periklis G. Foukas
- Second Department of Pathology, Attikon University Hospital, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Laurence de Leval
- Department of Laboratory Medicine and Pathology, Institute of Pathology, Lausanne University Hospital, Lausanne, Switzerland
| | - Constantinos Petrovas
- Department of Laboratory Medicine and Pathology, Institute of Pathology, Lausanne University Hospital, Lausanne, Switzerland
| |
Collapse
|
45
|
Xie H, Xi X, Lei T, Liu H, Xia Z. CD8 + T cell exhaustion in the tumor microenvironment of breast cancer. Front Immunol 2024; 15:1507283. [PMID: 39717767 PMCID: PMC11663851 DOI: 10.3389/fimmu.2024.1507283] [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: 10/07/2024] [Accepted: 11/20/2024] [Indexed: 12/25/2024] Open
Abstract
CD8+ T cells are crucial cytotoxic components of the tumor immune system. In chronic inflammation, they become low-responsive, a state known as T cell exhaustion (TEX). The aim of immune checkpoint blockade is to counteract TEX, yet its dynamics in breast cancer remain poorly understood. This review defines CD8+ TEX and outlines its features and underlying mechanisms. It also discusses the primary mechanisms of CD8+ TEX in breast cancer, covering inhibitory receptors, immunosuppressive cells, cytokines, transcriptomic and epigenetic alterations, metabolic reprogramming, and exosome pathways, offering insights into potential immunotherapy strategies for breast cancer.
Collapse
Affiliation(s)
- Hanghang Xie
- Xi’an People’s Hospital (Xi’an Fourth Hospital), Affiliated People’s Hospital of Northwest University, Xi’an, China
| | - Xiaowei Xi
- Technical University of Munich (TUM) School of Medicine and Health, Munich, Germany
| | - Ting Lei
- Xi’an People’s Hospital (Xi’an Fourth Hospital), Affiliated People’s Hospital of Northwest University, Xi’an, China
| | - Hongli Liu
- Xi’an People’s Hospital (Xi’an Fourth Hospital), Affiliated People’s Hospital of Northwest University, Xi’an, China
| | - Zhijia Xia
- Department of General, Visceral, and Transplant Surgery, Ludwig-Maximilians University, Munich, Germany
| |
Collapse
|
46
|
Wei C, Chen G, Chen K, Fang S, He H. Immune alterations and overexpression of CTCF in endometrial carcinoma: insights from molecular subtyping. Cancer Cell Int 2024; 24:392. [PMID: 39623397 PMCID: PMC11613940 DOI: 10.1186/s12935-024-03576-y] [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: 06/24/2024] [Accepted: 11/14/2024] [Indexed: 12/06/2024] Open
Abstract
BACKGROUND Endometrial cancer (EC) is a prevalent epithelial malignancy originating in the female endometrium, and its global incidence has been on the rise over the past decade. Despite significant scientific progress has been achieved recently, the genetic factors underlying EC pathogenesis remain poorly understood, warranting further investigation. METHODS We employed transcriptomic datasets from the Genomic Data Commons database to extract variable and clinical data. Quantile normalization and log2 transformations were applied to obtain a gene expression matrix for the sample cohort. Various assays, such as quantitative real-time polymerase chain reaction (qRT-PCR), western blotting, immunohistochemistry (IHC), wound healing assay, transwell assay, and TUNEL assay, were employed in the study to examine the involvement of CTCF in EC cell biology. Additionally, in vivo experiments were conducted using a subcutaneous transplantation tumor model in athymic nude mice. The potential mechanism of action of CTCF was also illustrated by identifying differentially expressed genes (DEGs) and functions after interfering with CTCF gene expression through the GSPAdb online database. RESULTS After categorizing 543 samples into cohorts with high and low ImmuneScores, we discovered 1025 genes that were differentially expressed, including 745 genes that were up-regulated and 280 genes that were down-regulated in the high scores group compared to the low scores group. Tumor mutation burden (TMB) analysis highlighted 11 genes with the highest mutation frequency. Furthermore, 16 immune checkpoints and 50 immune regulatory factors exhibited differential expression. Among these, CTCF was up-regulated in EC. We found that CTCF knockdown could diminish EC's invasive ability and metastatic potential while enhancing apoptosis. In vivo experiments corroborated that CTCF knockdown could reduce tumor growth. The GSPAdb online database identified differential expression pathways mainly enriched in cellular metabolism as well as some intracellular classical signaling pathways after interfering with CTCF gene expression. In addition, we identified potential downstream regulators of CTCF through protein interaction networks. CONCLUSION This study unveiled comprehensive molecular characteristics and DEGs in EC, emphasizing the up-regulation of CTCF in EC. Our findings collectively suggest that CTCF represents a promising therapeutic target, and our gene molecular typing model offers a novel approach for prognostic evaluation in EC.
Collapse
Affiliation(s)
- Caiping Wei
- Department of Gynecology, Liuzhou Municipal Liutie Central Hospital, Liuzhou, Guangxi, China
| | - Guowei Chen
- Department of Gynecology, Liuzhou Worker's Hospital, Liuzhou, Guangxi, China
| | - Kun Chen
- Department of Gynecology, Liuzhou Municipal Liutie Central Hospital, Liuzhou, Guangxi, China
| | - Shuang Fang
- Department of Gynecology, Liuzhou Worker's Hospital, Liuzhou, Guangxi, China
| | - Hongying He
- Department of Gynecology, Liuzhou Municipal Liutie Central Hospital, Liuzhou, Guangxi, China.
- Key Laboratory of Medical Molecular Diagnostics of Liuzhou, Key Laboratory for Nucleic Acid Molecular Diagnosis and Application of Guangxi Health and Wellness Commission, Liuzhou, Guangxi, China.
- Department of Gynecology, GuangZhou Women and Children's Medical Center Liuzhou Hospital, Liuzhou, Guangxi, China.
| |
Collapse
|
47
|
Liu Y, Liu W, Wu T. TIGIT: Will it be the next star therapeutic target like PD-1 in hematological malignancies? Crit Rev Oncol Hematol 2024; 204:104495. [PMID: 39236904 DOI: 10.1016/j.critrevonc.2024.104495] [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: 07/17/2024] [Revised: 09/01/2024] [Accepted: 09/01/2024] [Indexed: 09/07/2024] Open
Abstract
Research on the mechanism and application of checkpoint inhibitory receptors in hematologic diseases has progressed rapidly. However, in the treatment of relapserefractory (R/R) hematologic malignancies and anti-programmed cell death protein 1 (PD-1), patients who are resistant to anti-cytotoxic T lymphocyte-associated antigen-4 (CTLA-4) are in urgent need of alternative therapeutic targets. T cell immunoreceptor with immunoglobulin and ITIM domains (TIGIT) has a broad prospect as an inhibitory receptor like PD-1, but its more specific mechanism of action and application in hematologic diseases still need to be further studied. In this review, we discuss the mechanism of TIGIT pathway, combined effects with other immune checkpoints, immune-related therapy, the impact of TIGIT on hematopoietic stem cell transplantation (HSCT) and the tumor microenvironment (TME) provides a potential therapeutic target for hematologic malignancies.
Collapse
Affiliation(s)
- Yang Liu
- The 940th Hostipal of Joint Logistics Support force of Chinese People's Liberation Army, China.
| | - Wenhui Liu
- The 940th Hostipal of Joint Logistics Support force of Chinese People's Liberation Army, China.
| | - Tao Wu
- The 940th Hostipal of Joint Logistics Support force of Chinese People's Liberation Army, China.
| |
Collapse
|
48
|
Gayen S, Mukherjee S, Dasgupta S, Roy S. Emerging druggable targets for immune checkpoint modulation in cancer immunotherapy: the iceberg lies beneath the surface. Apoptosis 2024; 29:1879-1913. [PMID: 39354213 DOI: 10.1007/s10495-024-02022-8] [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] [Accepted: 09/18/2024] [Indexed: 10/03/2024]
Abstract
The immune system serves as a fundamental defender against the initiation and progression of cancer. Failure of the immune system augments immunosuppressive action that leading to cancer manifestation. This immunosuppressive effect causes from significant alterations in immune checkpoint expression associated with tumoral progression. The tumor microenvironment promotes immune escape mechanisms that further amplifying immunosuppressive actions. Notably, substantial targeting of immune checkpoints has been pragmatic in the advancement of cancer research. This study highlights a comprehensive review of emerging druggable targets aimed at modulating immune checkpoint co-inhibitory as well as co-stimulatory molecules in response to immune system activation. This modulation has prompted to the development of newer therapeutic insights, eventually inducing immunogenic cell death through immunomodulatory actions. The study emphasizes the role of immune checkpoints in immunogenic regulation of cancer pathogenesis and explores potential therapeutic avenues in cancer immunotherapy.Modulation of Immunosuppressive and Immunostimulatory pathways of immune checkpoints in cancer immunotherapy.
Collapse
Affiliation(s)
- Sakuntala Gayen
- Department of Pharmaceutical Technology, NSHM Knowledge Campus, Kolkata-Group of Institutions, 124, B. L. Saha Road, Tara Park, Behala, Kolkata, West Bengal, 700053, India
| | - Swarupananda Mukherjee
- Department of Pharmaceutical Technology, NSHM Knowledge Campus, Kolkata-Group of Institutions, 124, B. L. Saha Road, Tara Park, Behala, Kolkata, West Bengal, 700053, India
| | - Sandipan Dasgupta
- Department of Pharmaceutical Technology, Maulana Abul Kalam Azad University of Technology, Kolkata, West Bengal, 741249, India
| | - Souvik Roy
- Department of Pharmaceutical Technology, NSHM Knowledge Campus, Kolkata-Group of Institutions, 124, B. L. Saha Road, Tara Park, Behala, Kolkata, West Bengal, 700053, India.
| |
Collapse
|
49
|
Shao Y, Yang WY, Nanayakkara G, Saaoud F, Ben Issa M, Xu K, Lu Y, Jiang X, Mohsin S, Wang H, Yang X. Immune Checkpoints Are New Therapeutic Targets in Regulating Cardio-, and Cerebro-Vascular Diseases and CD4 +Foxp3 + Regulatory T Cell Immunosuppression. INTERNATIONAL JOURNAL OF DRUG DISCOVERY AND PHARMACOLOGY 2024; 3:100022. [PMID: 39926714 PMCID: PMC11804271 DOI: 10.53941/ijddp.2024.100022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/11/2025]
Abstract
Although previous reviews explored the roles of selected immune checkpoints (ICPs) in cardiovascular diseases (CVD) and cerebrovascular diseases from various perspectives, many related aspects have yet to be thoroughly reviewed and analyzed. Our comprehensive review addresses this gap by discussing the cellular functions of ICPs, focusing on the tissue-specific and microenvironment-localized transcriptomic and posttranslational regulation of ICP expressions, as well as their functional interactions with metabolic reprogramming. We also analyze how 14 pairs of ICPs, including CTLA-4/CD86-CD80, PD1-PDL-1, and TIGIT-CD155, regulate CVD pathogenesis. Additionally, the review covers the roles of ICPs in modulating CD4+Foxp3+ regulatory T cells (Tregs), T cells, and innate immune cells in various CVDs and cerebrovascular diseases. Furthermore, we outline seven immunological principles to guide the development of new ICP-based therapies for CVDs. This timely and thorough analysis of recent advancements and challenges provide new insights into the role of ICPs in CVDs, cerebrovascular diseases and Tregs, and will support the development of novel therapeutics strategies for these diseases.
Collapse
Affiliation(s)
- Ying Shao
- Lemole Center for Integrated Lymphatics and Vascular Research, Department of Cardiovascular Sciences, Lewis Katz School of Medicine at Temple University, Philadelphia, PA19140, USA
- Center for Metabolic Disease Research, Department of Cardiovascular Sciences, Lewis Katz School of Medicine at Temple University, Philadelphia, PA19140, USA
| | - William Y. Yang
- Lemole Center for Integrated Lymphatics and Vascular Research, Department of Cardiovascular Sciences, Lewis Katz School of Medicine at Temple University, Philadelphia, PA19140, USA
| | - Gayani Nanayakkara
- Eccles Institute of Human Genetics, University of Utah, Salt Lake City, UT84112, USA
| | - Fatma Saaoud
- Lemole Center for Integrated Lymphatics and Vascular Research, Department of Cardiovascular Sciences, Lewis Katz School of Medicine at Temple University, Philadelphia, PA19140, USA
| | - Mohammed Ben Issa
- Lemole Center for Integrated Lymphatics and Vascular Research, Department of Cardiovascular Sciences, Lewis Katz School of Medicine at Temple University, Philadelphia, PA19140, USA
| | - Keman Xu
- Lemole Center for Integrated Lymphatics and Vascular Research, Department of Cardiovascular Sciences, Lewis Katz School of Medicine at Temple University, Philadelphia, PA19140, USA
| | - Yifan Lu
- Lemole Center for Integrated Lymphatics and Vascular Research, Department of Cardiovascular Sciences, Lewis Katz School of Medicine at Temple University, Philadelphia, PA19140, USA
| | - Xiaohua Jiang
- Lemole Center for Integrated Lymphatics and Vascular Research, Department of Cardiovascular Sciences, Lewis Katz School of Medicine at Temple University, Philadelphia, PA19140, USA
- Center for Metabolic Disease Research, Department of Cardiovascular Sciences, Lewis Katz School of Medicine at Temple University, Philadelphia, PA19140, USA
| | - Sadia Mohsin
- Aging + Cardiovascular Discovery Center (ACDC), Department of Cardiovascular Sciences, Lewis Katz School of Medicine at Temple University, Philadelphia, PA19140, USA
| | - Hong Wang
- Center for Metabolic Disease Research, Department of Cardiovascular Sciences, Lewis Katz School of Medicine at Temple University, Philadelphia, PA19140, USA
| | - Xiaofeng Yang
- Lemole Center for Integrated Lymphatics and Vascular Research, Department of Cardiovascular Sciences, Lewis Katz School of Medicine at Temple University, Philadelphia, PA19140, USA
- Center for Metabolic Disease Research, Department of Cardiovascular Sciences, Lewis Katz School of Medicine at Temple University, Philadelphia, PA19140, USA
| |
Collapse
|
50
|
Yamada Y, Miyoshi H, Takeuchi M, Nakashima K, Yamada K, Kato T, Tanaka K, Kohno K, Imaizumi Y, Miyazaki Y, Ohshima K. TIGIT expression on neoplastic cells is a poor prognostic factor for adult T-cell leukaemia/lymphoma. Pathology 2024; 56:993-999. [PMID: 39266421 DOI: 10.1016/j.pathol.2024.06.003] [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: 12/15/2023] [Revised: 05/02/2024] [Accepted: 06/02/2024] [Indexed: 09/14/2024]
Abstract
Adult T-cell leukaemia/lymphoma (ATLL) is an aggressive peripheral T-cell neoplasm with a poor prognosis. T-cell immunoreceptor with immunoglobulin and immunoreceptor tyrosine-based inhibitory motif domains (TIGIT) is an immune checkpoint receptor expressed on T and natural killer cells. Although increased TIGIT expression in the tumour microenvironment is associated with poor prognosis in various neoplasms, its relevance in ATLL remains unknown. Herein, we investigated the clinicopathological impact of TIGIT expression on ATLL using immunohistochemistry. TIGIT expression was detected in 21 of 84 patients (25%). A partial association between the clinical features and immune checkpoint molecules and the expression of TIGIT was found including sIL-2R, CD86 and GITR. TIGIT-positive patients [median survival time (MST) 8.9 months, 95% confidence interval (CI) 7.7-15.6] had inferior overall survival compared with TIGIT-negative patients (MST 18.7 months, 95% CI 12.0-36.4) (p=0.0124]. TIGIT expression maintained its prognostic value for overall survival in both univariate and multivariate analyses [hazard ratio (HR) 1.909; 95% CI 1.044-3.488; p=0.0356]. Further studies are required to clarify the clinical and biological significance of TIGIT expression in patients with ATLL.
Collapse
Affiliation(s)
- Yuichi Yamada
- Department of Pathology, Kurume University School of Medicine, Kurume, Japan; Department of Hematology, Atomic Bomb Disease and Hibakusha Medicine Unit, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan; Department of Hematology, Nagasaki University Hospital, Nagasaki, Japan
| | - Hiroaki Miyoshi
- Department of Pathology, Kurume University School of Medicine, Kurume, Japan.
| | - Mai Takeuchi
- Department of Pathology, Kurume University School of Medicine, Kurume, Japan
| | - Kazutaka Nakashima
- Department of Pathology, Kurume University School of Medicine, Kurume, Japan
| | - Kyohei Yamada
- Department of Pathology, Kurume University School of Medicine, Kurume, Japan
| | - Takeharu Kato
- Department of Hematology, Nagasaki University Hospital, Nagasaki, Japan
| | - Ken Tanaka
- Department of Hematology/Oncology, Wakayama Medical University, Wakayama, Japan
| | - Kei Kohno
- Department of Pathology, Kurume University School of Medicine, Kurume, Japan
| | - Yoshitaka Imaizumi
- Department of Hematology, National Hospital Organization Nagasaki Medical Center, Omura, Japan
| | - Yasushi Miyazaki
- Department of Hematology, Atomic Bomb Disease and Hibakusha Medicine Unit, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan; Department of Hematology, Nagasaki University Hospital, Nagasaki, Japan; Department of Hematology, Atomic Bomb Disease and Hibakusha Medicine Unit, Atomic Bomb Disease Institute, Nagasaki University, Nagasaki, Japan
| | - Koichi Ohshima
- Department of Pathology, Kurume University School of Medicine, Kurume, Japan
| |
Collapse
|