1
|
Berclaz LM, Altendorf-Hofmann A, Lindner LH, Burkhard-Meier A, Di Gioia D, Dürr HR, Klein A, Albertsmeier M, Schmidt-Hegemann NS, Klauschen F, Knösel T. TIM-3 Qualifies as a Potential Immunotherapeutic Target in Specific Subsets of Patients with High-Risk Soft Tissue Sarcomas (HR-STS). Cancers (Basel) 2023; 15:2735. [PMID: 37345075 DOI: 10.3390/cancers15102735] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Revised: 05/08/2023] [Accepted: 05/10/2023] [Indexed: 06/23/2023] Open
Abstract
(1) Background: The expression of T cell immunoglobulin and mucin domain-containing protein 3 (TIM-3), an immune checkpoint receptor on T cells, has been associated with dismal outcomes and advanced tumor stages in various solid tumors. The blockade of TIM-3 is currently under examination in several clinical trials. This study examines TIM-3 expression in high-risk soft tissue sarcomas (HR-STS). (2) Methods: Tumor cell expression of TIM-3 on protein level was analyzed in pre-treatment biopsies of patients with HR-STS. TIM-3 expression was correlated with clinicopathological parameters including tumor-infiltrating lymphocyte (TIL) counts, programmed cell death 1 (PD-1) and programmed cell death ligand 1 (PDL-1) expression in patients with HR-STS. Survival dependent on the expression of TIM-3 was analyzed. (3) Results: TIM-3 expression was observed in 101 (56%) out of 179 pre-treatment biopsies of patients with HR-STS. TIM-3 expression was significantly more often observed in undifferentiated pleomorphic sarcomas (UPS) compared to other histological subtypes (p < 0.001), high TIL counts (p < 0.001), and high PD-1 (p < 0.001) and PD-L1 expression (p < 0.001). TIM-3 expression did not have a prognostic impact on survival in patients with HR-STS. (4) Conclusions: This is the first study to demonstrate a significant tumor cell expression of TIM-3 in specific subsets of patients with HR-STS. TIM-3 qualifies as a potential immunotherapeutic target in HR-STS.
Collapse
Affiliation(s)
- Luc M Berclaz
- Department of Internal Medicine III, University Hospital, Ludwig-Maximilians-University (LMU) Munich, 81377 Munich, Germany
| | - Annelore Altendorf-Hofmann
- Department of General, Visceral and Vascular Surgery, Friedrich-Schiller University Jena, 07747 Jena, Germany
| | - Lars H Lindner
- Department of Internal Medicine III, University Hospital, Ludwig-Maximilians-University (LMU) Munich, 81377 Munich, Germany
| | - Anton Burkhard-Meier
- Department of Internal Medicine III, University Hospital, Ludwig-Maximilians-University (LMU) Munich, 81377 Munich, Germany
| | - Dorit Di Gioia
- Department of Internal Medicine III, University Hospital, Ludwig-Maximilians-University (LMU) Munich, 81377 Munich, Germany
| | - Hans Roland Dürr
- Orthopaedic Oncology, Department of Orthopaedics and Trauma Surgery, University Hospital, Ludwig-Maximilians-University (LMU) Munich, 81377 Munich, Germany
| | - Alexander Klein
- Orthopaedic Oncology, Department of Orthopaedics and Trauma Surgery, University Hospital, Ludwig-Maximilians-University (LMU) Munich, 81377 Munich, Germany
| | - Markus Albertsmeier
- Department of General, Visceral and Transplantation Surgery, University Hospital, Ludwig-Maximilians-University (LMU) Munich, 81377 Munich, Germany
| | - Nina-Sophie Schmidt-Hegemann
- Department of Radiation Oncology, University Hospital, Ludwig-Maximilians-University (LMU) Munich, Marchioninistr. 15, 81377 Munich, Germany
| | - Frederick Klauschen
- Institute of Pathology, Ludwig-Maximilians-University (LMU) Munich, 81377 Munich, Germany
| | - Thomas Knösel
- Institute of Pathology, Ludwig-Maximilians-University (LMU) Munich, 81377 Munich, Germany
| |
Collapse
|
2
|
Zhang R, Kong D, Chen R, Guo Y, Jian W, Han M, Zhou T. A model‐based meta‐analysis of immune‐related adverse events during immune checkpoint inhibitors treatment for
NSCLC. CPT Pharmacometrics Syst Pharmacol 2022; 11:1135-1146. [PMID: 35763678 PMCID: PMC9381889 DOI: 10.1002/psp4.12834] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Revised: 05/02/2022] [Accepted: 05/24/2022] [Indexed: 11/07/2022] Open
Affiliation(s)
- Renwei Zhang
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery System, Department of Pharmaceutics, School of Pharmaceutical Sciences Peking University Beijing China
| | - Daming Kong
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery System, Department of Pharmaceutics, School of Pharmaceutical Sciences Peking University Beijing China
| | - Rong Chen
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery System, Department of Pharmaceutics, School of Pharmaceutical Sciences Peking University Beijing China
| | - Yuchen Guo
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery System, Department of Pharmaceutics, School of Pharmaceutical Sciences Peking University Beijing China
| | - Weizhe Jian
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery System, Department of Pharmaceutics, School of Pharmaceutical Sciences Peking University Beijing China
| | - Mengyi Han
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery System, Department of Pharmaceutics, School of Pharmaceutical Sciences Peking University Beijing China
| | - Tianyan Zhou
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery System, Department of Pharmaceutics, School of Pharmaceutical Sciences Peking University Beijing China
| |
Collapse
|
3
|
Gomes de Morais AL, Cerdá S, de Miguel M. New Checkpoint Inhibitors on the Road: Targeting TIM-3 in Solid Tumors. Curr Oncol Rep 2022; 24:651-658. [PMID: 35218498 DOI: 10.1007/s11912-022-01218-y] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/02/2021] [Indexed: 12/28/2022]
Abstract
PURPOSE OF REVIEW Even though checkpoint inhibitors have become a recent milestone for the treatment of many different tumor types, eventually, most part of patients will develop resistance mechanisms and their disease will progress. New generations of checkpoint inhibitors, as the ones directed to TIM-3, are on research. RECENT FINDINGS TIM-3 expression has been associated with more advanced stages and shorter survival in several tumor types, due to its association with T-cell dysfunction, and has become an interesting target to explore. Early phase clinical trials with different anti-TIM-3 monoclonal antibodies have shown a safe toxicity profile, as cobolimab, LY3321367, or sabatolimab; however, the general antitumor activity remains to be determined and further investigations are needed. TIM-3 is implicated in resistance to immunotherapy due to its role in T cell exhaustion. However, the TIM-3 pathway is highly complex in terms of non-canonical signaling, broad expression by different immune cells and multiple ligands. Different anti-TIM-3 inhibitors are currently on research, either as monotherapy or in combination with other immunotherapies or chemotherapy, aiming to overcome resistance.
Collapse
Affiliation(s)
- Ana Luiza Gomes de Morais
- START Madrid-Fundación Jiménez Díaz (FJD) Early Phase Program, Fundación Jiménez Díaz Hospital, Madrid, Spain
| | - Sara Cerdá
- START Madrid-HM Centro Integral Oncológico Clara Campal (CIOCC) Early Phase Program, HM Sanchinarro University Hospital, Calle Oña, 10, 28050, Madrid, Spain
| | - Maria de Miguel
- START Madrid-HM Centro Integral Oncológico Clara Campal (CIOCC) Early Phase Program, HM Sanchinarro University Hospital, Calle Oña, 10, 28050, Madrid, Spain.
| |
Collapse
|
4
|
Hu M, Li Y, Lu Y, Wang M, Li Y, Wang C, Li Q, Zhao H. The regulation of immune checkpoints by the hypoxic tumor microenvironment. PeerJ 2021; 9:e11306. [PMID: 34012727 PMCID: PMC8109006 DOI: 10.7717/peerj.11306] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Accepted: 03/29/2021] [Indexed: 12/14/2022] Open
Abstract
The tumor microenvironment (TME) influences the occurrence and progression of tumors, and hypoxia is an important characteristic of the TME. The expression of programmed death 1 (PD1)/programmed death-ligand 1 (PDL1), cytotoxic T-lymphocyte-associated antigen 4 (CTLA4), and other immune checkpoints in hypoxic malignant tumors is often significantly increased, and is associated with poor prognosis. The application of immune checkpoint inhibitors (ICIs) for treating lung cancer, urothelial carcinoma, and gynecological tumors has achieved encouraging efficacy; however, the rate of efficacy of ICI single-drug treatment is only about 20%. In the present review, we discuss the possible mechanisms by which the hypoxic TME regulates immune checkpoints. By activating hypoxia-inducible factor-1α (HIF-1α), regulating the adenosine (Ado)-A2aR pathway, regulating the glycolytic pathway, and driving epithelial-mesenchymal transition (EMT) and other biological pathways, hypoxia regulates the expression levels of CTLA4, PD1, PDL1, CD47, lymphocyte activation gene 3 (LAG3), T-cell immunoglobulin and mucin domain 3 (TIM3), and other immune checkpoints, which interfere with the immune effector cell anti-tumor response and provide convenient conditions for tumors to escape immune surveillance. The combination of HIF-1α inhibitors, Ado-inhibiting tumor immune microenvironment regulatory drugs, and other drugs with ICIs has good efficacy in both preclinical studies and phase I-II clinical studies. Exploring the effects of TME hypoxia on the expression of immune checkpoints and the function of infiltrating immune cells has greatly clarified the relationship between the hypoxic TME and immune escape, which is of great significance for the development of new drugs and the search for predictive markers of the efficacy of immunotherapy for treating malignant tumors. In the future, combination therapy with hypoxia pathway inhibitors and ICIs may be an effective anti-tumor treatment strategy.
Collapse
Affiliation(s)
- Min Hu
- Department of Biochemistry & Molecular Biology, Basic Medical College, Shanxi Medical University, Taiyuan, Shanxi Province, China.,Department of Oncology, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Yongfu Li
- Department of Oncology, Beijing Friendship Hospital, Capital Medical University, Beijing, China.,Department of Oncology, The Second Affiliated Hospital of Hainan Medical University, Haikou, Hainan Province, China
| | - Yuting Lu
- Department of Oncology, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Miao Wang
- Department of Oncology, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Yingrui Li
- Department of Biochemistry & Molecular Biology, Basic Medical College, Shanxi Medical University, Taiyuan, Shanxi Province, China.,Department of Oncology, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Chaoying Wang
- Department of Oncology, The Second Affiliated Hospital of Hainan Medical University, Haikou, Hainan Province, China
| | - Qin Li
- Department of Oncology, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Hong Zhao
- Department of Biochemistry & Molecular Biology, Basic Medical College, Shanxi Medical University, Taiyuan, Shanxi Province, China
| |
Collapse
|
5
|
Ding QQ, Chauvin JM, Zarour HM. Targeting novel inhibitory receptors in cancer immunotherapy. Semin Immunol 2020; 49:101436. [PMID: 33288379 DOI: 10.1016/j.smim.2020.101436] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Revised: 11/18/2020] [Accepted: 11/19/2020] [Indexed: 12/24/2022]
Abstract
T cells play a critical role in promoting tumor regression in both experimental models and humans. Yet, T cells that are chronically exposed to tumor antigen during cancer progression can become dysfunctional/exhausted and fail to induce tumor destruction. Such tumor-induced T cell dysfunction may occur via multiple mechanisms. In particular, immune checkpoint inhibitory receptors that are upregulated by tumor-infiltrating lymphocytes in many cancers limit T cell survival and function. Overcoming this inhibitory receptor-mediated T cell dysfunction has been a central focus of recent developments in cancer immunotherapy. Immunotherapies targeting inhibitory receptor pathways such as programmed cell death 1 (PD-1)/programmed death ligand 1 and cytotoxic T lymphocyte-associated antigen 4 (CTLA-4), alone or in combination, confer significant clinical benefits in multiple tumor types. However, many patients with cancer do not respond to immune checkpoint blockade, and dual PD-1/CTLA-4 blockade may cause serious adverse events, which limits its indications. Targeting novel non-redundant inhibitory receptor pathways contributing to tumor-induced T cell dysfunction in the tumor microenvironment may prove efficacious and non-toxic. This review presents preclinical and clinical findings supporting the roles of two key pathways-T-cell immunoglobulin and mucin-domain containing-3 (TIM-3) and T cell immunoreceptor with Ig and ITIM domain (TIGIT)/CD226/CD96/CD112R-in cancer immunotherapy.
Collapse
Affiliation(s)
- Quan-Quan Ding
- Department of Medicine and Division of Hematology/Oncology, University of Pittsburgh, School of Medicine, Pittsburgh, PA 15213, USA
| | - Joe-Marc Chauvin
- Department of Medicine and Division of Hematology/Oncology, University of Pittsburgh, School of Medicine, Pittsburgh, PA 15213, USA
| | - Hassane M Zarour
- Department of Medicine and Division of Hematology/Oncology, University of Pittsburgh, School of Medicine, Pittsburgh, PA 15213, USA; Department of Immunology, University of Pittsburgh, School of Medicine, Pittsburgh, PA 15213, USA.
| |
Collapse
|
6
|
Abstract
T cell immunoglobulin and mucin domain-containing protein 3 (TIM3), a member of the TIM family, was originally identified as a receptor expressed on interferon-γ-producing CD4+ and CD8+ T cells. Initial data indicated that TIM3 functioned as a 'co-inhibitory' or 'checkpoint' receptor, but due to the lack of a definable inhibitory signalling motif, it was also suggested that TIM3 might act as a co-stimulatory receptor. Recent studies have shown that TIM3 is part of a module that contains multiple co-inhibitory receptors (checkpoint receptors), which are co-expressed and co-regulated on dysfunctional or 'exhausted' T cells in chronic viral infections and cancer. Furthermore, co-blockade of TIM3 and programmed cell death 1 (PD1) can result in tumour regression in preclinical models and can improve anticancer T cell responses in patients with advanced cancers. Here, we highlight the developments in understanding TIM3 biology, including novel ligand identification and the discovery of loss-of-function mutations associated with human disease. In addition, we summarize emerging data from human clinical trials showing that TIM3 indeed acts as a 'checkpoint' receptor and that inhibition of TIM3 enhances the antitumour effect of PD1 blockade.
Collapse
|