1
|
Craig-Meyer D, Hollenbaugh JA, Morgado S, McGee K, Perkins E, Yarzabek B, Lapinski P, Rowse A, Cooper C, Fortunato M, Cocco M, Cadwallader K, Munday J. Immunophenotypical characterization of immune checkpoint receptor expression in cynomolgus monkeys and human healthy volunteers in resting and in T-cell stimulatory conditions in vitro. J Immunotoxicol 2025; 22:2462106. [PMID: 39945090 DOI: 10.1080/1547691x.2025.2462106] [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: 06/12/2024] [Revised: 01/14/2025] [Accepted: 01/29/2025] [Indexed: 04/12/2025] Open
Abstract
Immunotherapeutics targeting immune checkpoint receptors or their ligands (i.e., immune checkpoint inhibitors), have been groundbreaking in the field of oncology, radically changing the approach to treatment and improving the clinical outcomes of an ever-expanding list of solid tumors and hematological malignancies. However, immune checkpoint inhibitors (ICI) are not devoid of side effects, collectively regarded as immune-related adverse events (irAE); they are not easily uncovered in preclinical immunotoxicological investigations and are often due to the very low expression of their targets in immunologically-unchallenged non-clinical species. We have characterized expression of a broad range of immune checkpoint receptors in peripheral blood mononuclear cell (PBMC) subpopulations from cynomolgus monkeys and healthy human volunteers, under resting and T-cell stimulatory conditions by multicolor flow cytometry to inform appropriate species selection for modeling potential irAE in immunotherapeutic preclinical research. Focusing on the response of the main lymphocyte populations to interleukin (IL)-2 alone, or in combination with anti-CD3 and anti-CD28 antibodies, checkpoints with shared similarities and key differences between the two species were identified. The results of this first study provide a database for the expression and response to stimulation for immune checkpoint receptors and can help guide future model selection in the design of preclinical studies involving immunotherapeutics directed against these targets.
Collapse
Affiliation(s)
| | | | - Sara Morgado
- Labcorp Early Development Laboratories Limited, Huntingdon, UK
| | - Karen McGee
- Labcorp Early Development Laboratories Limited, Harrogate, UK
| | - Ethan Perkins
- Labcorp Early Development Laboratories Limited, Harrogate, UK
- Institute of Cancer Therapeutics, Faculty of Life Sciences, University of Bradford, UK
| | | | | | - Amber Rowse
- Labcorp Early Development Laboratories Inc, Ann Arbor, MI
| | - Chris Cooper
- Labcorp Early Development Laboratories Limited, Harrogate, UK
| | - Mara Fortunato
- Labcorp Early Development Laboratories Limited, Huntingdon, UK
| | - Mario Cocco
- Labcorp Early Development Laboratories Limited, Harrogate, UK
| | | | - James Munday
- Labcorp Early Development Laboratories Limited, Harrogate, UK
| |
Collapse
|
2
|
Morchón-Araujo D, Catani G, Mirallas O, Pretelli G, Sánchez-Pérez V, Vieito M, Braña I, Pujol-Borrell R, Garralda E, Hernando-Calvo A. Emerging Immunotherapy Targets in Early Drug Development. Int J Mol Sci 2025; 26:5394. [PMID: 40508202 PMCID: PMC12155519 DOI: 10.3390/ijms26115394] [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: 04/01/2025] [Revised: 05/24/2025] [Accepted: 05/26/2025] [Indexed: 06/16/2025] Open
Abstract
Immunotherapy has significantly changed the treatment paradigm for solid tumors, with immune checkpoint inhibitors now established in the management of many malignancies. Despite initial success, durable responses remain limited to a subset of patients, often less than 30%, due to both intrinsic and acquired resistance mechanisms. These challenges have prompted the development of next-generation immunotherapies. Recent efforts have expanded the scope of immunotherapy beyond PD-1/PD-L1 and CTLA-4 inhibition, focusing on new immune targets currently under investigation in early phase clinical trials. These include novel immune checkpoint inhibitors, immunomodulators targeting the tumor microenvironment, and bispecific antibodies. This review provides a comprehensive overview of emerging immune targets currently being investigated in early drug development, discussing their mechanisms of action, preliminary clinical outcomes, and potential future directions.
Collapse
Affiliation(s)
- Daniel Morchón-Araujo
- Department of Medical Oncology, University Hospital of Salamanca, IBSAL, 37007 Salamanca, Spain;
- Vall d’Hebron Institute of Oncology, 08035 Barcelona, Spain; (G.C.); (O.M.); (G.P.); (V.S.-P.); (M.V.); (I.B.); (E.G.)
| | - Greta Catani
- Vall d’Hebron Institute of Oncology, 08035 Barcelona, Spain; (G.C.); (O.M.); (G.P.); (V.S.-P.); (M.V.); (I.B.); (E.G.)
- Department of Medical Oncology, Alexander Fleming Institute, Buenos Aires 1426, Argentina
| | - Oriol Mirallas
- Vall d’Hebron Institute of Oncology, 08035 Barcelona, Spain; (G.C.); (O.M.); (G.P.); (V.S.-P.); (M.V.); (I.B.); (E.G.)
- Department of Medical Oncology, Vall d’Hebron Barcelona Hospital Campus, 08035 Barcelona, Spain
| | - Giulia Pretelli
- Vall d’Hebron Institute of Oncology, 08035 Barcelona, Spain; (G.C.); (O.M.); (G.P.); (V.S.-P.); (M.V.); (I.B.); (E.G.)
- Department of Medical Oncology, Vall d’Hebron Barcelona Hospital Campus, 08035 Barcelona, Spain
| | - Vicky Sánchez-Pérez
- Vall d’Hebron Institute of Oncology, 08035 Barcelona, Spain; (G.C.); (O.M.); (G.P.); (V.S.-P.); (M.V.); (I.B.); (E.G.)
- Department of Medical Oncology, Vall d’Hebron Barcelona Hospital Campus, 08035 Barcelona, Spain
| | - María Vieito
- Vall d’Hebron Institute of Oncology, 08035 Barcelona, Spain; (G.C.); (O.M.); (G.P.); (V.S.-P.); (M.V.); (I.B.); (E.G.)
- Department of Medical Oncology, Vall d’Hebron Barcelona Hospital Campus, 08035 Barcelona, Spain
| | - Irene Braña
- Vall d’Hebron Institute of Oncology, 08035 Barcelona, Spain; (G.C.); (O.M.); (G.P.); (V.S.-P.); (M.V.); (I.B.); (E.G.)
- Department of Medical Oncology, Vall d’Hebron Barcelona Hospital Campus, 08035 Barcelona, Spain
| | - Ricardo Pujol-Borrell
- Department of Cell Biology, Physiology and Immunology, Autonomous University of Barcelona, 08193 Bellaterra (Barcelona), Spain;
- Tumor Immunology and Immunotherapy Group, Vall d’Hebron Institute of Oncology, Vall d’Hebron Barcelona Hospital Campus, 08035 Barcelona, Spain
| | - Elena Garralda
- Vall d’Hebron Institute of Oncology, 08035 Barcelona, Spain; (G.C.); (O.M.); (G.P.); (V.S.-P.); (M.V.); (I.B.); (E.G.)
- Department of Medical Oncology, Vall d’Hebron Barcelona Hospital Campus, 08035 Barcelona, Spain
| | - Alberto Hernando-Calvo
- Vall d’Hebron Institute of Oncology, 08035 Barcelona, Spain; (G.C.); (O.M.); (G.P.); (V.S.-P.); (M.V.); (I.B.); (E.G.)
- Department of Medical Oncology, Vall d’Hebron Barcelona Hospital Campus, 08035 Barcelona, Spain
| |
Collapse
|
3
|
Zhang K, Mi Y, Zhang B, Xue X, Ding Y, Ma J, Yuan E, Zhao X, Zheng P. Preclinical application of a CD155 targeting chimeric antigen receptor T cell therapy for digestive system cancers. Oncogene 2025; 44:1463-1474. [PMID: 40025231 DOI: 10.1038/s41388-025-03322-2] [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: 07/09/2024] [Revised: 02/03/2025] [Accepted: 02/19/2025] [Indexed: 03/04/2025]
Abstract
Despite intensive multimodal therapy, the prognosis for patients with digestive system cancers remains poor. Cancer cell heterogeneity and immunosuppressive microenvironments are the main barriers to the effective CAR-T cell therapy with solid malignancies. In parallel, tumor-associated macrophages (TAMs) are essential for tumor immunosuppressive microenvironment formation. The limited efficacy of CAR-T cell therapy with solid malignancies prompted us to test whether new therapeutic target could enhance the antitumor activity of CAR-T cells with several digestive system cancer types. We determined CD155 expression in multiple human digestive system cancers, including gastric cancer, esophagus cancer, pancreatic cancer, and colon cancer, normal tissue samples and patient-derived M2-like tumor-associated macrophages. We developed a CD155-based CAR comprising the extracellular domain of human TIGIT, 4-1BB, and CD3z signaling domains (BBz). Furthermore, we validated the killing efficacy and safety of CD155-BBz CAR-T cells in vitro and in vivo using in-house established preclinical tumor models. CD155 was strongly and homogenously expressed in digestive system cancers but mildly in normal tissues, indicating it could be an ideal target for CAR-T cell therapy, moreover, TAMs that express CD155 possess an immunosuppressive M2-like profile. We found that CD155-BBz CAR-T cells can mediate significant antitumor activity in vivo, which induces complete tumor regression and long-lasting immunologic memory of established solid tumors in xenograft models. Our study indicates that CD155 is a promising target for digestive system cancer therapy, and CD155-targeting CAR-T cells perform a detecting power in digestive system cancer clinical trials.
Collapse
Affiliation(s)
- Kai Zhang
- Department of Laboratory Medicine, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China.
- Henan Key Laboratory for Helicobacter pylori & Microbiota and GI cancer, Marshall Medical Research Center, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China.
- Tianjian Laboratory of Advanced Biomedical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, 450052, Henan, China.
- Zhengzhou Key Laboratory for In Vitro Diagnosis of Hypertensive Disorders of Pregnancy, 450052, Zhengzhou, Henan, China.
| | - Yang Mi
- Henan Key Laboratory for Helicobacter pylori & Microbiota and GI cancer, Marshall Medical Research Center, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Bohao Zhang
- Department of Medicine Research Center, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Xia Xue
- Henan Key Laboratory for Helicobacter pylori & Microbiota and GI cancer, Marshall Medical Research Center, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Yangnan Ding
- Department of Laboratory Medicine, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
- Tianjian Laboratory of Advanced Biomedical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, 450052, Henan, China
- Zhengzhou Key Laboratory for In Vitro Diagnosis of Hypertensive Disorders of Pregnancy, 450052, Zhengzhou, Henan, China
| | - Jun Ma
- Department of Laboratory Medicine, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
- Tianjian Laboratory of Advanced Biomedical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, 450052, Henan, China
- Zhengzhou Key Laboratory for In Vitro Diagnosis of Hypertensive Disorders of Pregnancy, 450052, Zhengzhou, Henan, China
| | - Enwu Yuan
- Department of Laboratory Medicine, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China.
- Tianjian Laboratory of Advanced Biomedical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, 450052, Henan, China.
- Zhengzhou Key Laboratory for In Vitro Diagnosis of Hypertensive Disorders of Pregnancy, 450052, Zhengzhou, Henan, China.
| | - Xin Zhao
- Tianjian Laboratory of Advanced Biomedical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, 450052, Henan, China.
- The Radiology Department, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China.
| | - PengYuan Zheng
- Henan Key Laboratory for Helicobacter pylori & Microbiota and GI cancer, Marshall Medical Research Center, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China.
| |
Collapse
|
4
|
Shang Y, He Y, Zhang X, He W, Hua H, Ye F, Zhou X, Li Y, Zhong W, Wu G, Jiang W. Optimization of Immunotherapy Strategies Based on Spatiotemporal Heterogeneity of Tumour-Associated Tissue-Resident Memory T Cells. Immunology 2025; 175:123-133. [PMID: 40114407 PMCID: PMC12052439 DOI: 10.1111/imm.13924] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Revised: 02/26/2025] [Accepted: 03/11/2025] [Indexed: 03/22/2025] Open
Abstract
Tissue-resident memory T cells (TRMs) reside in peripheral tissues and provide rapid immune defence against local infection and tumours. Tumour-associated TRMs share common tissue-resident features and formation mechanisms, representing some unique subsets of tumour-infiltrating lymphocytes (TILs). However, differences in the tumour microenvironment(TME) and tumour evolution stage result in TRMs exhibiting temporal and spatial heterogeneity of phenotype and function not only at different stages, before and after treatment, but also between tumours originating from different tissues, primary and metastatic cancer, and tumour and adjacent normal tissue. The infiltration of TRMs is often associated with immunotherapy response and favourable prognosis; however, due to different definitions, it has been shown that some subtypes of TRMs can also have a negative impact. Therefore, it is crucial to precisely characterise the TRM subpopulations that can influence the therapeutic efficacy and clinical prognosis of various solid tumours. Here, we review the spatiotemporal heterogeneity of tumour-associated TRMs, as well as the differences in their impact on clinical outcomes. We also explore the relationship between TRMs and immune checkpoint blockade (ICB) and TIL therapy, providing insights into potential new targets and strategies for immunotherapy.
Collapse
Affiliation(s)
- Yile Shang
- Department of Colorectal Surgery, The First Affiliated HospitalZhejiang University School of MedicineHangzhouChina
- College of MedicineZhejiang UniversityHangzhouChina
| | - Yinjun He
- College of MedicineZhejiang UniversityHangzhouChina
| | - Xiang Zhang
- Department of Colorectal Surgery, The First Affiliated HospitalZhejiang University School of MedicineHangzhouChina
| | - Wenguang He
- Department of Radiology, First Affiliated HospitalZhejiang University School of MedicineHangzhouChina
| | - Hanju Hua
- Department of Colorectal Surgery, The First Affiliated HospitalZhejiang University School of MedicineHangzhouChina
| | - Feng Ye
- Department of Colorectal Surgery, The First Affiliated HospitalZhejiang University School of MedicineHangzhouChina
| | - Xile Zhou
- Department of Colorectal Surgery, The First Affiliated HospitalZhejiang University School of MedicineHangzhouChina
| | - Yandong Li
- Department of Colorectal Surgery, The First Affiliated HospitalZhejiang University School of MedicineHangzhouChina
| | - Weixiang Zhong
- Department of Pathology, First Affiliated HospitalZhejiang University School of MedicineHangzhouChina
| | - Guosheng Wu
- Department of Colorectal Surgery, The First Affiliated HospitalZhejiang University School of MedicineHangzhouChina
| | - Weiqin Jiang
- Department of Colorectal Surgery, The First Affiliated HospitalZhejiang University School of MedicineHangzhouChina
| |
Collapse
|
5
|
Yan C, Wang G. Advances in research on flavonoids in tumor immunotherapy (Review). Mol Med Rep 2025; 31:150. [PMID: 40211703 PMCID: PMC11995692 DOI: 10.3892/mmr.2025.13515] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Accepted: 02/25/2025] [Indexed: 04/16/2025] Open
Abstract
Cancer immunotherapy is an approach used in anti‑tumor treatment; however, its efficacy is limited to specific tumor types that are inherently sensitive to immune system modulation. Expanding the scope of indications and enhancing the efficacy of cancer immunotherapy are key goals for continued advancement. Flavonoids modulate the tumor‑immunosuppressive microenvironment. Integrating flavonoids with immunotherapeutic modalities, including cancer vaccines, immune checkpoint inhibitors and adoptive immune‑cell therapy, has potential in terms of augmenting the therapeutic efficacy of immunotherapy. The present review aimed to summarize flavonoids that enhance cancer immunotherapy, focusing on their underlying mechanisms and the application of nanotechnology to overcome inherent limitations such as poor solubility, low bioavailability, rapid metabolism, and instability under physiological conditions, thereby highlighting the potential of flavonoids in advancing cancer immunotherapy.
Collapse
Affiliation(s)
- Chaoguang Yan
- Department of Oncology, Weifang Chinese Medicine Hospital, Weifang, Shandong 261000 P.R. China
| | - Guangchun Wang
- Department of Oncology, Weifang Chinese Medicine Hospital, Weifang, Shandong 261000 P.R. China
| |
Collapse
|
6
|
Mandelboim O, Porgador A. Engineered chemokines, resistant to cancer-mediated post-transcriptional modifications, as drugs to improve cancer immunotherapy. Proc Natl Acad Sci U S A 2025; 122:e2507821122. [PMID: 40388627 DOI: 10.1073/pnas.2507821122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/21/2025] Open
Affiliation(s)
- Ofer Mandelboim
- The Concern Foundation Laboratories at the Lautenberg Center for Immunology and Cancer Research, Institute for Medical Research Israel Canada (IMRIC), Hebrew University-Hadassah Medical School, Jerusalem 91120, Israel
| | - Angel Porgador
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel
| |
Collapse
|
7
|
Yu H, Jin S, Zeng M, Yang Z, Wang X. TIGIT antibody with PVR competitive ability enhances cancer immunotherapy and capable of eliciting anti-tumour immune memory. Br J Cancer 2025:10.1038/s41416-025-03046-w. [PMID: 40394151 DOI: 10.1038/s41416-025-03046-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2024] [Revised: 04/01/2025] [Accepted: 04/25/2025] [Indexed: 05/22/2025] Open
Abstract
BACKGROUND T-cell immunoreceptor with immunoglobulin (Ig) and ITIM domains (TIGIT) is a checkpoint receptor thought to be involved in mediating T-cell exhaustion and dysfunction of natural killer (NK) cells in tumours and is emerging as novel promising targets in immunotherapy, however, the ligand binding and the efficacy of its antibody still need to be further explored. METHODS Four different TIGIT antibodies in characteristics of antigen binding, in vitro effects on activated T cells, Fc region functions and tumour inhibition in animal models were compared. The antibody as monotherapy and combined with anti-PD-L1 antibody, effects on PBMC in ex vivo coculture with autologous human CRC organoids as well as PK profile were evaluated. RESULTS Studies demonstrated that TIGIT antibody with PVR-competitive ability as monotherapy resulted in inhibition of tumour growth, sustained anti-tumour immune memory in tumour re-challenge mice, enhanced anti-tumour therapy in combination with anti-PD-L1. Ex vivo coculture assay suggested that TIGIT antibody treatment activated immune cells and promoted infiltration and tumour killing ability of autologous PBMC in human CRC organoids. CONCLUSIONS Our study broadens the knowledge of TIGIT antibody in cancer immunotherapy and may benefit future development of next-generation checkpoint inhibitors with improved clinical outcomes.
Collapse
Affiliation(s)
- Huijuan Yu
- Guangdong Key Laboratory of Animal Conservation and Resource Utilization, Institute of Zoology, Guangdong Academy of Sciences, Guangzhou, China
| | - Shaowen Jin
- Department of Gastrointestinal Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Min Zeng
- Guangdong Annpobio Co., Ltd, Guangzhou, China
| | | | - Xiaofei Wang
- Guangdong Key Laboratory of Animal Conservation and Resource Utilization, Institute of Zoology, Guangdong Academy of Sciences, Guangzhou, China.
- Guangdong Annpobio Co., Ltd, Guangzhou, China.
| |
Collapse
|
8
|
Keshavarz Sadegh R, Saleki K, Rezaei N. Immune checkpoint inhibitor (ICI) therapy in central nervous system cancers: State-of-the-art and future outlook. Int Immunopharmacol 2025; 159:114837. [PMID: 40394797 DOI: 10.1016/j.intimp.2025.114837] [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: 01/11/2025] [Revised: 04/28/2025] [Accepted: 05/07/2025] [Indexed: 05/22/2025]
Abstract
Invasive central nervous system (CNS) cancers are an area where the development of breakthrough therapies is urgently needed. For instance, conditions such as glioblastoma multiforme (GBM) are associated with poor clinical prognosis, with the majority of trials offering no improvement to marginally enhanced survival. Unleashing the potential of targeting the immune system in CNS cancers has gained attention in recent years. Inhibition of immune checkpoints such as CTLA-4, PD-1/PD-L1, TIM-3, and LAG-3 has been attempted in recent trials. While potentially offering a notable edge over other immunotherapies, multi-organ adverse events have been found with the administration of immune checkpoint inhibitors (ICIs). The present review captures the state-of-the-art evidence on ICI treatments in different CNS cancers. Also, we discuss the value of combinational therapies involving ICIs as well as next-generation therapeutics such as bispecific antibodies targeting PD-1/LAG-3/TIM-3 and CRISPR-Cas9-edited PD-1-knock-out checkpoint-resistant CAR T-cells.
Collapse
Affiliation(s)
- Roghaye Keshavarz Sadegh
- Student Research Committee, Qazvin University of Medical Sciences, Qazvin, Iran; USERN Office, Qazvin University of Medical Sciences, Qazvin, Iran
| | - Kiarash Saleki
- Student Research Committee, Babol University of Medical Sciences, Babol, Iran; Research Center for Immunodeficiencies, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran; Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Tehran, Iran; USERN MUBabol Office, Universal Scientific Education and Research Network (USERN), Babol, Iran
| | - Nima Rezaei
- Research Center for Immunodeficiencies, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran; Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Tehran, Iran; Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
9
|
Appadurai MI, Chaudhary S, Shah A, Natarajan G, Alsafwani ZW, Khan P, Shinde DD, Lele SM, Smith LM, Nasser MW, Batra SK, Ganti AK, Lakshmanan I. ST6GalNAc-I regulates tumor cell sialylation via NECTIN2/MUC5AC-mediated immunosuppression and angiogenesis in non-small cell lung cancer. J Clin Invest 2025; 135:e186863. [PMID: 40371640 PMCID: PMC12077904 DOI: 10.1172/jci186863] [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: 09/10/2024] [Accepted: 03/12/2025] [Indexed: 05/16/2025] Open
Abstract
Glycosylation controls immune evasion, tumor progression, and metastasis. However, how tumor cell sialylation regulates immune evasion remains poorly characterized. ST6GalNAc-I, a sialyltransferase that conjugates sialic acid to the glycans in glycoproteins, was overexpressed in an aggressive-type KPA (KrasG12D/+ Trp53R172H/+ Ad-Cre) lung adenocarcinoma (LUAD) model and patient samples. Proteomic and biochemical analysis indicated that ST6GalNAc-I mediated NECTIN2 sialylation in LUAD cells. ST6GalNAc-I-deficient tumor cells cocultured with T cells were more susceptible to T cell-mediated tumor cell killing, indicating a key role for NECTIN2 in T cell dysfunction. Mice injected with St6galnac-I-knockdown syngeneic cells showed reduced lung tumor incidence and Nectin2/Tigit-associated immunosuppression. ST6GalNAc-I-deficient cells exhibited reduced P-DMEA metabolite levels, while administration of P-DMEA promoted LUAD cell proliferation via MUC5AC. MUC5AC interacted and colocalized with PRRC1 in the Golgi, suggesting a potential role for PRRC1 in MUC5AC glycosylation. Mice injected with ST6GalNAc-I/MUC5AC-deficient cells (human LUAD) exhibited reduced lung tumor incidence, angiogenesis, and liver metastases. Mechanistically, ST6GalNAc-I/MUC5AC regulates VCAN-V1, a key factor in tumor matrix remodeling during angiogenesis and metastasis. These findings demonstrate that ST6GalNAc-I-mediated sialylation of NECTIN2/MUC5AC is critical for immune evasion and tumor angiogenesis. Targeting this pathway may prevent LUAD development and/or metastasis.
Collapse
MESH Headings
- Animals
- Humans
- Lung Neoplasms/pathology
- Lung Neoplasms/immunology
- Lung Neoplasms/genetics
- Lung Neoplasms/metabolism
- Lung Neoplasms/blood supply
- Mice
- Nectins/genetics
- Nectins/immunology
- Nectins/metabolism
- Carcinoma, Non-Small-Cell Lung/pathology
- Carcinoma, Non-Small-Cell Lung/immunology
- Carcinoma, Non-Small-Cell Lung/genetics
- Carcinoma, Non-Small-Cell Lung/metabolism
- Carcinoma, Non-Small-Cell Lung/blood supply
- Sialyltransferases/genetics
- Sialyltransferases/immunology
- Sialyltransferases/metabolism
- Neovascularization, Pathologic/immunology
- Neovascularization, Pathologic/genetics
- Neovascularization, Pathologic/pathology
- Neovascularization, Pathologic/metabolism
- Neoplasm Proteins/genetics
- Neoplasm Proteins/immunology
- Neoplasm Proteins/metabolism
- Cell Line, Tumor
- Angiogenesis
Collapse
Affiliation(s)
| | | | - Ashu Shah
- Department of Biochemistry and Molecular Biology
| | | | | | - Parvez Khan
- Department of Biochemistry and Molecular Biology
| | | | | | | | - Mohd Wasim Nasser
- Department of Biochemistry and Molecular Biology
- Fred & Pamela Buffett Cancer Center; and
| | - Surinder Kumar Batra
- Department of Biochemistry and Molecular Biology
- Fred & Pamela Buffett Cancer Center; and
| | - Apar Kishor Ganti
- Department of Biochemistry and Molecular Biology
- Fred & Pamela Buffett Cancer Center; and
- Department of Internal Medicine, University of Nebraska Medical Center, Omaha, Nebraska, USA
- Division of Oncology-Hematology, Department of Internal Medicine, VA Nebraska-Western Iowa Health Care System, Omaha, Nebraska, USA
| | | |
Collapse
|
10
|
Zhang P, Whipp EC, Skuli SJ, Gharghabi M, Saygin C, Sher SA, Carroll M, Pan X, Eisenmann ED, Lai TH, Harrington BK, Chan WK, Youssef Y, Chen B, Penson A, Lewis AM, Castro CR, Fox N, Cihan A, Le Luduec JB, DeWolf S, Kauffman T, Mims AS, Canfield D, Phillips H, Williams KE, Shaffer J, Lozanski A, Doong TJ, Lozanski G, Mao C, Walker CJ, Blachly JS, Daniyan AF, Alinari L, Baiocchi RA, Yang Y, Grieselhuber NR, Campbell MJ, Baker SD, Blaser BW, Abdel-Wahab O, Lapalombella R. TP53 mutations and TET2 deficiency cooperate to drive leukemogenesis and establish an immunosuppressive environment. J Clin Invest 2025; 135:e184021. [PMID: 40111422 PMCID: PMC12077897 DOI: 10.1172/jci184021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Accepted: 03/06/2025] [Indexed: 03/22/2025] Open
Abstract
Mutations and deletions in TP53 are associated with adverse outcomes in patients with myeloid malignancies, and there is an urgent need for the development of improved therapies for TP53-mutant leukemias. Here, we identified mutations in TET2 as the most common co-occurring mutation in patients with TP53-mutant acute myeloid leukemia (AML). In mice, combined hematopoietic-specific deletion of TET2 and TP53 resulted in enhanced self-renewal compared with deletion of either gene alone. Tp53/Tet2 double-KO mice developed serially transplantable AML. Both mice and patients with AML with combined TET2/TP53 alterations upregulated innate immune signaling in malignant granulocyte-monocyte progenitors, which had leukemia-initiating capacity. A20 governs the leukemic maintenance by triggering aberrant noncanonical NF-κB signaling. Mice with Tp53/Tet2 loss had expansion of monocytic myeloid-derived suppressor cells (MDSCs), which impaired T cell proliferation and activation. Moreover, mice and patients with AML with combined TP53/TET2 alterations displayed increased expression of the TIGIT ligand, CD155, on malignant cells. TIGIT-blocking antibodies augmented NK cell-mediated killing of Tp53/Tet2 double-mutant AML cells, reduced leukemic burden, and prolonged survival in Tp53/Tet2 double-KO mice. These findings describe a leukemia-promoting link between TET2 and TP53 mutations and highlight therapeutic strategies to overcome the immunosuppressive bone marrow environment in this adverse subtype of AML.
Collapse
Affiliation(s)
- Pu Zhang
- Division of Hematology, Department of Internal Medicine, The Ohio State University, Columbus, Ohio, USA
- Molecular Pharmacology Program, Sloan Kettering Institute, New York, New York, USA
- Division of Pharmaceutics and Pharmacology, College of Pharmacy, The Ohio State University, Columbus, Ohio, USA
| | - Ethan C. Whipp
- Division of Hematology, Department of Internal Medicine, The Ohio State University, Columbus, Ohio, USA
| | - Sarah J. Skuli
- University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Mehdi Gharghabi
- Division of Hematology, Department of Internal Medicine, The Ohio State University, Columbus, Ohio, USA
| | - Caner Saygin
- Section of Hematology/Oncology, University of Chicago, Chicago, Illinois, USA
| | - Steven A. Sher
- Division of Hematology, Department of Internal Medicine, The Ohio State University, Columbus, Ohio, USA
| | - Martin Carroll
- University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Xiangyu Pan
- Joan and Sanford I. Weill Department of Medicine, Weill Cornell Medicine, New York, New York, USA
| | - Eric D. Eisenmann
- Division of Pharmaceutics and Pharmacology, College of Pharmacy, The Ohio State University, Columbus, Ohio, USA
| | - Tzung-Huei Lai
- Division of Hematology, Department of Internal Medicine, The Ohio State University, Columbus, Ohio, USA
| | - Bonnie K. Harrington
- College of Veterinary Medicine, Michigan State University, East Lansing, Michigan, USA
| | - Wing Keung Chan
- Division of Hematology, Department of Internal Medicine, The Ohio State University, Columbus, Ohio, USA
| | - Youssef Youssef
- Division of Hematology, Department of Internal Medicine, The Ohio State University, Columbus, Ohio, USA
| | - Bingyi Chen
- Molecular Pharmacology Program, Sloan Kettering Institute, New York, New York, USA
| | - Alex Penson
- Molecular Pharmacology Program, Sloan Kettering Institute, New York, New York, USA
| | - Alexander M. Lewis
- Molecular Pharmacology Program, Sloan Kettering Institute, New York, New York, USA
| | - Cynthia R. Castro
- Molecular Pharmacology Program, Sloan Kettering Institute, New York, New York, USA
| | - Nina Fox
- Molecular Pharmacology Program, Sloan Kettering Institute, New York, New York, USA
| | - Ali Cihan
- Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | | | - Susan DeWolf
- Molecular Pharmacology Program, Sloan Kettering Institute, New York, New York, USA
| | - Tierney Kauffman
- Division of Hematology, Department of Internal Medicine, The Ohio State University, Columbus, Ohio, USA
| | - Alice S. Mims
- Division of Hematology, Department of Internal Medicine, The Ohio State University, Columbus, Ohio, USA
| | - Daniel Canfield
- Division of Hematology, Department of Internal Medicine, The Ohio State University, Columbus, Ohio, USA
| | - Hannah Phillips
- Division of Hematology, Department of Internal Medicine, The Ohio State University, Columbus, Ohio, USA
| | - Katie E. Williams
- Division of Hematology, Department of Internal Medicine, The Ohio State University, Columbus, Ohio, USA
| | - Jami Shaffer
- Division of Hematology, Department of Internal Medicine, The Ohio State University, Columbus, Ohio, USA
| | - Arletta Lozanski
- Division of Hematology, Department of Internal Medicine, The Ohio State University, Columbus, Ohio, USA
| | - Tzyy-Jye Doong
- Division of Hematology, Department of Internal Medicine, The Ohio State University, Columbus, Ohio, USA
| | - Gerard Lozanski
- Division of Hematology, Department of Internal Medicine, The Ohio State University, Columbus, Ohio, USA
| | - Charlene Mao
- Division of Hematology, Department of Internal Medicine, The Ohio State University, Columbus, Ohio, USA
| | - Christopher J. Walker
- Division of Hematology, Department of Internal Medicine, The Ohio State University, Columbus, Ohio, USA
- Leukemia Research Program, The Ohio State University James Comprehensive Cancer Center, Columbus, Ohio, USA
| | - James S. Blachly
- Division of Hematology, Department of Internal Medicine, The Ohio State University, Columbus, Ohio, USA
| | | | - Lapo Alinari
- Division of Hematology, Department of Internal Medicine, The Ohio State University, Columbus, Ohio, USA
| | - Robert A. Baiocchi
- Division of Hematology, Department of Internal Medicine, The Ohio State University, Columbus, Ohio, USA
| | - Yiping Yang
- Division of Hematology, Department of Internal Medicine, The Ohio State University, Columbus, Ohio, USA
| | - Nicole R. Grieselhuber
- Division of Hematology, Department of Internal Medicine, The Ohio State University, Columbus, Ohio, USA
| | - Moray J. Campbell
- Division of Cancer Biology, Cedars Sinai Medical Center, Los Angeles, California, USA
| | - Sharyn D. Baker
- Division of Pharmaceutics and Pharmacology, College of Pharmacy, The Ohio State University, Columbus, Ohio, USA
| | - Bradley W. Blaser
- Division of Hematology, Department of Internal Medicine, The Ohio State University, Columbus, Ohio, USA
| | - Omar Abdel-Wahab
- Molecular Pharmacology Program, Sloan Kettering Institute, New York, New York, USA
| | - Rosa Lapalombella
- Division of Hematology, Department of Internal Medicine, The Ohio State University, Columbus, Ohio, USA
| |
Collapse
|
11
|
Li F, Jin C, Pan Y, Zhang Z, Wang L, Deng J, Zhou Y, Guo B, Zhang S. Construction of a stromal cell-related prognostic signature based on a 101-combination machine learning framework for predicting prognosis and immunotherapy response in triple-negative breast cancer. Front Immunol 2025; 16:1544348. [PMID: 40438115 PMCID: PMC12116347 DOI: 10.3389/fimmu.2025.1544348] [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: 12/12/2024] [Accepted: 04/21/2025] [Indexed: 06/01/2025] Open
Abstract
Background Triple-negative breast cancer (TNBC) is a highly aggressive subtype with limited therapeutic targets and poor immunotherapy outcomes. The tumor microenvironment (TME) plays a key role in cancer progression. Advances in single-cell transcriptomics have highlighted the impact of stromal cells on tumor progression, immune suppression, and immunotherapy. This study aims to identify stromal cell marker genes and develop a prognostic signature for predicting TNBC survival outcomes and immunotherapy response. Methods Single-cell RNA sequencing (scRNA-seq) datasets were retrieved from the Gene Expression Omnibus (GEO) database and annotated using known marker genes. Cell types preferentially distributed in TNBC were identified using odds ratios (OR). Bulk transcriptome data were analyzed using Weighted correlation network analysis (WGCNA) to identify myCAF-, VSMC-, and Pericyte-related genes (MVPRGs). A consensus MVP cell-related signature (MVPRS) was developed using 10 machine learning algorithms and 101 model combinations and validated in training and validation cohorts. Immune infiltration and immunotherapy response were assessed using CIBERSORT, ssGSEA, TIDE, IPS scores, and an independent cohort (GSE91061). FN1, a key gene in the model, was validated through qRT-PCR, immunohistochemistry, RNA interference, CCK-8 assay, apoptosis assay and wound-healing assay. Results In TNBC, three stromal cell subpopulations-myofibroblastic cancer-associated fibroblasts (myCAF), vascular smooth muscle cells (VSMCs), and pericytes-were enriched, exhibiting high interaction frequencies and strong associations with poor prognosis. A nine-gene prognostic model (MVPRS), developed from 23 prognostically significant genes among the 259 MVPRGs, demonstrated excellent predictive performance and was validated as an independent prognostic factor. A nomogram integrating MVPRS, age, stage, and tumor grade offered clinical utility. High-risk group showed reduced immune infiltration and increased activity in tumor-related pathways like ANGIOGENESIS and HYPOXIA, while low-risk groups responded better to immunotherapy based on TIDE and IPS scores. FN1, identified as a key oncogene, was highly expressed in TNBC tissues and cell lines, promoting proliferation and migration while inhibiting apoptosis. Conclusion This study reveals TNBC microenvironment heterogeneity and introduces a prognostic signature based on myCAF, VSMC, and Pericyte marker genes. MVPRS effectively predicts TNBC prognosis and immunotherapy response, providing guidance for personalized treatment. FN1 was validated as a key oncogene impacting TNBC progression and malignant phenotype, with potential as a therapeutic target.
Collapse
Affiliation(s)
- Fanrong Li
- Department of Genetics, School of Basic Medical Sciences, Suzhou Medical College of Soochow University, Suzhou, China
| | - Congnan Jin
- Department of Genetics, School of Basic Medical Sciences, Suzhou Medical College of Soochow University, Suzhou, China
| | - Yacheng Pan
- Department of Genetics, School of Basic Medical Sciences, Suzhou Medical College of Soochow University, Suzhou, China
| | - Zheng Zhang
- Department of Genetics, School of Basic Medical Sciences, Suzhou Medical College of Soochow University, Suzhou, China
| | - Liying Wang
- Jiangsu Clinical Medicine Research Institute, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Jieqiong Deng
- Department of Genetics, School of Basic Medical Sciences, Suzhou Medical College of Soochow University, Suzhou, China
| | - Yifeng Zhou
- Department of Genetics, School of Basic Medical Sciences, Suzhou Medical College of Soochow University, Suzhou, China
- Jiangsu Clinical Medicine Research Institute, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Binbin Guo
- Department of Genetics, School of Basic Medical Sciences, Suzhou Medical College of Soochow University, Suzhou, China
| | - Shenghua Zhang
- Department of Genetics, School of Basic Medical Sciences, Suzhou Medical College of Soochow University, Suzhou, China
- Jiangsu Clinical Medicine Research Institute, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| |
Collapse
|
12
|
Kim S, Jeon SH, Kim Y, Park N, Kim IA. TIGIT blockade increases efficacy of PD-1 blockade combined with radiation therapy in triple-negative breast cancer model. Radiother Oncol 2025; 208:110932. [PMID: 40360046 DOI: 10.1016/j.radonc.2025.110932] [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: 11/14/2024] [Revised: 04/17/2025] [Accepted: 05/03/2025] [Indexed: 05/15/2025]
Abstract
BACKGROUND AND PURPOSE T-cell immunoreceptor with Ig and ITIM domains (TIGIT) suppresses functions of CD8+ T cells, and radiation therapy (RT) induces stimulation of regulatory T cells (Tregs), thereby limiting antitumor efficacy. This study aims to investigate the role of TIGIT in the immunosuppressive tumor environment and evaluate the potential of TIGIT blockade (αTIGIT) to enhance antitumor immune responses. METHODS We analyzed public transcriptomic data to identify the expression patterns of TIGIT on T cells in breast cancer and its prognostic impact. In addition, a murine TNBC model was utilized to evaluate the effects of αPD-1, local RT, and αTIGIT. T cells in tumors, tumor-draining lymph nodes (TdLNs), and the spleen were analyzed to assess the antitumor immune responses upon the treatments. RESULTS The analysis revealed that TIGIT is predominantly expressed on T cells within breast cancer, and the expression of TIGIT was associated with poor outcomes in TNBC patients. In the murine model, the combination of αPD-1 and RT increased TIGIT+CD226+CD8+ TILs, which are crucial for the efficacy of αTIGIT. Adding αTIGIT to αPD-1 and RT (αPD-1/RT) resulted in a synergistic antitumor effect, which was accompanied by increased infiltration of CD8+ TILs in both irradiated and nonirradiated tumors by the triple combination therapy compared to αPD-1/RT. The triple combination therapy also resulted in a less exhausted phenotype among CD8+ TILs and increased the proliferation of splenic CD8+ T cells. Moreover, αTIGIT significantly reduced Tregs in tumors, TdLNs, and the spleen when combined with αPD-1/RT. CONCLUSION αTIGIT exhibits synergistic effects when added to αPD-1/RT by increasing the infiltration and activation of CD8+ TILs while reducing Tregs. The study suggests that αTIGIT could be an effective strategy to enhance the antitumor efficacy of αPD-1 and RT in TNBC.
Collapse
Affiliation(s)
- Seongmin Kim
- Department of Tumor Biology and Cancer Research Institute, Graduate School of Medicine, Seoul National University, Seoul, Republic of Korea; Integrated Major in Innovative Medical Science, Seoul National University Graduate School, Seoul, Republic of Korea; Medical Science Research Institute, Seoul National University Bundang Hospital, Seongnam, Republic of Korea
| | - Seung Hyuck Jeon
- Department of Radiation Oncology, Seoul National University Bundang Hospital, Seongnam, Republic of Korea
| | - Yoomin Kim
- Department of Tumor Biology and Cancer Research Institute, Graduate School of Medicine, Seoul National University, Seoul, Republic of Korea; Medical Science Research Institute, Seoul National University Bundang Hospital, Seongnam, Republic of Korea
| | - Nawon Park
- Department of Tumor Biology and Cancer Research Institute, Graduate School of Medicine, Seoul National University, Seoul, Republic of Korea; Medical Science Research Institute, Seoul National University Bundang Hospital, Seongnam, Republic of Korea
| | - In Ah Kim
- Department of Tumor Biology and Cancer Research Institute, Graduate School of Medicine, Seoul National University, Seoul, Republic of Korea; Integrated Major in Innovative Medical Science, Seoul National University Graduate School, Seoul, Republic of Korea; Medical Science Research Institute, Seoul National University Bundang Hospital, Seongnam, Republic of Korea; Department of Radiation Oncology, Seoul National University Bundang Hospital, Seongnam, Republic of Korea; Department of Radiation Oncology, Seoul National University College of Medicine, Seoul, Republic of Korea.
| |
Collapse
|
13
|
Zhou X, Xiao L, Lai F, Chen W, Zhou C, Deng Y, Wang T, Xing S, Diao H, Tang M, Guo W, Luo E. Comprehensive overview of antibody drug-related clinical studies in gynecology: insights from ClinicalTrials.gov. Front Med (Lausanne) 2025; 12:1521587. [PMID: 40417700 PMCID: PMC12098050 DOI: 10.3389/fmed.2025.1521587] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2024] [Accepted: 04/18/2025] [Indexed: 05/27/2025] Open
Abstract
Antibodies have been widely used globally over the past decade and play an increasingly important role in modern medicine. Notably, significant advancements have been achieved in the realm of gynecology, particularly in gynecological cancers. This study endeavors to present a thorough overview of antibody-related drug clinical studies in gynecology registered on ClinicalTrials.gov, focusing on the basic characteristics of trials, geographical distribution, administration routes, indications, and targets. The analysis indicates a rising prevalence of antibody-drug conjugates (ADCs), bispecific antibodies, and Fc-fusion proteins. This study will help develop new ideas for future research on antibodies in gynecology.
Collapse
Affiliation(s)
- Xiaoling Zhou
- Department of Pharmacy, Chengdu Women's and Children's Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Li Xiao
- Department of Medical Affairs, Chengdu Women's and Children's Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Fan Lai
- Department of Obstetrics, Chengdu Women's and Children's Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Wei Chen
- Department of Traditional Chinese Medicine, Chengdu Women's and Children's Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Congrong Zhou
- Department of Traditional Chinese Medicine, Chengdu Women's and Children's Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Yi Deng
- Department of Pharmacy, Chengdu Women's and Children's Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Tao Wang
- Department of Obstetrics, Chengdu Women's and Children's Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Shasha Xing
- Department of Good Clinical Practice, Chengdu Women's and Children's Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Haoyang Diao
- Department of Good Clinical Practice, Chengdu Women's and Children's Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Mi Tang
- Department of Good Clinical Practice, Chengdu Women's and Children's Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Wenmei Guo
- Department of Good Clinical Practice, Chengdu Women's and Children's Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Erdan Luo
- Department of Good Clinical Practice, Chengdu Women's and Children's Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| |
Collapse
|
14
|
Han W, Lv J, Wang M, Wu X, Sun D, Chen W, Wang Y, Zhou W, Yang Y, Bao J, Han Q, Chen X, Guo F, Feng G, Li M, Chen Q. Potential Roles of Serum Exosomal CD155 and its Impact on NK Cell Immunosuppression in Hepatocellular Carcinoma. Balkan Med J 2025; 42:242-253. [PMID: 40326845 PMCID: PMC12060579 DOI: 10.4274/balkanmedj.galenos.2025.2025-1-129] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2025] [Accepted: 04/17/2025] [Indexed: 05/07/2025] Open
Abstract
Background Targeted therapies directed at tumor immune checkpoint, like programmed death-ligand (PD-L)1/programmed death (PD)-1, have shown remarkable progress. Nevertheless, treatment efficacy in hepatocellular carcinoma (HCC) is notably compromised due to the intricate immune microenvironment. Exploring alternative checkpoints beyond PD-L1/PD-1, including those not located on the cell surface, may improve our understanding of their roles in areas like diagnostic potential and immune tolerance in HCC. Aims To explore the roles of serum exosomal CD155 (exo-CD155) in HCC. Study Design Experimental study. Methods We separated and analyzed serum exosomes from HCC patients. We quantified the concentrations of serum soluble CD155 (sCD155) and serum exo-CD155, and examined their association with disease progression, hepatitis B surface antigen (HBsAg) presence, and the concentrations of α-fetoprotein fraction L3 (AFP-L3) or alpha-fetoprotein (AFP). Additionally, we assessed the diagnostic effect through the receiver operating characteristic (ROC) curve, and the immune suppressive effect on natural killer (NK) cells of exo-CD155. Results This study reveal elevated exo-CD155 levels in all HCC patients, with a significant increase in early-stage patients, exhibiting normal AFP/AFP-L3 or HBsAg-positive status. Exo-CD155 is linked to the progression of HCC and shows significant diagnostic effectiveness for the disease. Furthermore, the incubation of NK-92MI with exosomes derived from HCC patients leads to a substantial reduction in immune function, which can be partially counteracted with an antibody that blocks T cell immune receptor immunoglobulin and ITIM domains, (TIGIT)-blocking antibody. Conclusion These results disclose exo-CD155 shows promise for serving as a biomarker for HCC, especially in early-stage patients or those with normal AFP/AFP-L3 levels. Moreover, serum exosomes from HCC patients suppress NK cell immune functions through the TIGIT/CD155 pathway, contributing to immune tolerance in HCC.
Collapse
Affiliation(s)
- Wenzheng Han
- The First Affiliated Hospital, Wannan Medical College, Anhui, China
| | - Jinrong Lv
- Institute of Biology and Medical Sciences, Soochow University, Jiangsu, China
| | - Mintuo Wang
- The First Affiliated Hospital, Wannan Medical College, Anhui, China
| | - Xiaoxin Wu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Centre for Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang, China
| | - Dongdong Sun
- The First Affiliated Hospital, Wannan Medical College, Anhui, China
| | - Wenwen Chen
- Institute of Biology and Medical Sciences, Soochow University, Jiangsu, China
| | - Yingying Wang
- The First Affiliated Hospital, Wannan Medical College, Anhui, China
| | - Wenjie Zhou
- The First Affiliated Hospital, Wannan Medical College, Anhui, China
| | - Yuxuan Yang
- The First Affiliated Hospital, Wannan Medical College, Anhui, China
| | - Jia Bao
- The First Affiliated Hospital, Wannan Medical College, Anhui, China
| | - Qingzhen Han
- Center of Clinical Laboratory and Translational Medicine, The Fourth Affiliated Hospital of Soochow University, Suzhou Dushu Lake Hospital, Suzhou, Jiangsu, China
| | - Xiaopeng Chen
- The First Affiliated Hospital, Wannan Medical College, Anhui, China
| | - Fei Guo
- Department of Urology, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Gang Feng
- The First Affiliated Hospital, Wannan Medical College, Anhui, China
| | - Min Li
- Institute of Biology and Medical Sciences, Soochow University, Jiangsu, China
| | - Qing Chen
- The First Affiliated Hospital, Wannan Medical College, Anhui, China
| |
Collapse
|
15
|
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
|
16
|
Weng D, Guo R, Gao Y, Xu S, Li Y, Zhou J, An R, Xu H. Immuno-PET Imaging of a 68Ga-Labeled Single-Domain Antibody for Detecting Tumor TIGIT Expression. Mol Pharm 2025. [PMID: 40298304 DOI: 10.1021/acs.molpharmaceut.4c00900] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/30/2025]
Abstract
Preclinical studies have shown that the expression of T cell immunoglobulin and the immunoreceptor tyrosine-based inhibitory motif domain (TIGIT) in the tumor microenvironment is associated with the efficacy of anti-TIGIT-based immunotherapy. This study aimed to develop a TIGIT single-domain antibody (sdAb)-based positron emission tomography (PET) radiotracer, [68Ga]Ga-NOTA-NABT3A1, and evaluate its characteristics. The NABT3A1 was modified with a NOTA derivative and radiolabeled with 68Ga. In vitro stability of [68Ga]Ga-NOTA-NABT3A1 was assessed in phosphate-buffered saline (PBS) and fetal bovine serum (FBS), along with its specificity for TIGIT stably transfected A375 Human melanoma cells (A375-TIGIT) was performed. In vivo imaging was conducted on A375-TIGIT tumor-bearing nude mice at different time points after the injection of [68Ga]Ga-NOTA-NABT3A1. The synthesized [68Ga]Ga-NOTA-NABT3A1 achieved a radiochemical yield of 70.56 ± 1.14% and purity levels of 95.80 ± 0.58% in PBS and 96.79 ± 1.69% in FBS at 2 h. Immuno-PET imaging revealed specific accumulation of [68Ga]Ga-NOTA-NABT3A1 in A375-TIGIT tumor-bearing nude mice, with a maximum uptake of 3.86 ± 0.29% injected dose/g at 0.5 h. Biodistribution and immunohistochemical analyses confirmed the in vivo imaging results. In conclusion, we successfully synthesized an NABT3A1-derived PET radiotracer with the potential to noninvasively assess TIGIT expression in tumors.
Collapse
Affiliation(s)
- Dinghu Weng
- Shanghai Key Laboratory of Molecular Imaging, Shanghai University of Medicine and Health Sciences, 201318 Shanghai, China
- Department of Medical Imaging, Zhongnan Hospital of Wuhan University, 430071 Wuhan, China
- Hubei Provincial Engineering Research Center of Multimodal Medical Imaging Technology and Clinical Application, 430071 Wuhan, China
- Wuhan Clinical Research and Development Center of Brain Resuscitation and Functional Imaging, 430071 Wuhan, China
| | - Rong Guo
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430000, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan 430000, China
| | - Yu Gao
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430000, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan 430000, China
| | - Shasha Xu
- Beijing Novaboody Biotechnological Ltd., Beijing 102600, China
| | - Yingying Li
- Beijing Novaboody Biotechnological Ltd., Beijing 102600, China
| | - Jun Zhou
- Interventional Diagnostic and Therapeutic Center, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Rui An
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430000, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan 430000, China
| | - Haibo Xu
- Department of Medical Imaging, Zhongnan Hospital of Wuhan University, 430071 Wuhan, China
- Hubei Provincial Engineering Research Center of Multimodal Medical Imaging Technology and Clinical Application, 430071 Wuhan, China
- Wuhan Clinical Research and Development Center of Brain Resuscitation and Functional Imaging, 430071 Wuhan, China
| |
Collapse
|
17
|
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
|
18
|
Seo ES, Lee SK, Son YM. Multifaceted functions of tissue-resident memory T cells in tumorigenesis and cancer immunotherapy. Cancer Immunol Immunother 2025; 74:184. [PMID: 40285796 PMCID: PMC12033165 DOI: 10.1007/s00262-025-04035-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2024] [Accepted: 03/24/2025] [Indexed: 04/29/2025]
Abstract
Tissue-resident memory T (TRM) cells are well reported as a strong protective first line of defense against foreign antigens in non-lymphoid tissues. Moreover, TRM cells have demonstrated critical protective roles in antitumor immunity, contributing to enhanced survival and tumor growth inhibition across various cancer types. However, surprisingly, recent studies suggest that TRM cells can exhibit paradoxical effects, potentially promoting tumor progression under certain conditions and leading to adverse outcomes during antitumor immune responses. Understanding the complexities of TRM cell functions will enable us to harness their potential in advancing cancer immunotherapy more effectively. Therefore, this review comprehensively investigates the dual roles of TRM cells in different tumor contexts, highlighting their protective functions in combating cancers and their unfavorable potential to exacerbate tumor development. Additionally, we explore the implications of TRM cell behaviors for future cancer treatment strategies, emphasizing the need for further research to optimize the therapeutic exploitation of TRM cells while mitigating their deleterious effects.
Collapse
Affiliation(s)
- Eun Sang Seo
- Department of Systems Biotechnology, Chung-Ang University, Anseong, 17546, Republic of Korea
| | - Sung-Kyu Lee
- Department of Systems Biotechnology, Chung-Ang University, Anseong, 17546, Republic of Korea
| | - Young Min Son
- Department of Systems Biotechnology, Chung-Ang University, Anseong, 17546, Republic of Korea.
| |
Collapse
|
19
|
Sonar PV, Singh AK, Mandadi S, Sharma NK. Expanding horizons of cancer immunotherapy: hopes and hurdles. Front Oncol 2025; 15:1511560. [PMID: 40352591 PMCID: PMC12061710 DOI: 10.3389/fonc.2025.1511560] [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: 10/15/2024] [Accepted: 03/31/2025] [Indexed: 05/14/2025] Open
Abstract
Background Tumor displays various forms of tumor heterogeneity including immune heterogeneity that allow cancer cells to survive during conventional anticancer drug interventions. Thus, there is a strong rationale for overcoming anticancer drug resistance by employing the components of immune cells. Using the immune system to target tumor cells has revolutionized treatment. Recently, significant progress has been achieved at preclinical and clinical levels to benefit cancer patients. Approach A review of literature from the past ten years across PubMed, Scopus, and Web of Science focused on immunotherapy strategies. These include immune checkpoint inhibitors (ICIs), tumor-infiltrating lymphocyte therapy, antibody-drug conjugates (ADCs), cancer vaccines, CAR T-cell therapy, and the role of the gut microbiome. Conclusion While immunotherapy outcomes have improved, particularly for tumor types such as melanoma and non-small cell lung cancer (NSCLC), challenges persist regarding predictive biomarker identification and better management. Ongoing research on modifiers of immune function like gut microbiome-derived metabolites, next-generation ADCs, and new classes of biologics is warranted. Overall, continued investigation toward optimizing synergistic immunotherapeutic combinations through strategic drug delivery systems is imperative for preclinical and clinical success in cancer patients.
Collapse
Affiliation(s)
- Priyanka Vijay Sonar
- Cancer and Translational Research Lab, Dr. D.Y. Patil Biotechnology & Bioinformatics Institute, Dr. D.Y. Patil Vidyapeeth, Pune, Maharashtra, India
| | - Anuj Kumar Singh
- Cancer and Translational Research Lab, Dr. D.Y. Patil Biotechnology & Bioinformatics Institute, Dr. D.Y. Patil Vidyapeeth, Pune, Maharashtra, India
- Ichnos Glenmark Innovation, Glenmark Pharmaceuticals Limited, Navi Mumbai, Maharashtra, India
| | - Sravan Mandadi
- Ichnos Glenmark Innovation, Glenmark Pharmaceuticals Limited, Navi Mumbai, Maharashtra, India
| | - Nilesh Kumar Sharma
- Cancer and Translational Research Lab, Dr. D.Y. Patil Biotechnology & Bioinformatics Institute, Dr. D.Y. Patil Vidyapeeth, Pune, Maharashtra, India
| |
Collapse
|
20
|
Imani S, Farghadani R, Roozitalab G, Maghsoudloo M, Emadi M, Moradi A, Abedi B, Jabbarzadeh Kaboli P. Reprogramming the breast tumor immune microenvironment: cold-to-hot transition for enhanced immunotherapy. J Exp Clin Cancer Res 2025; 44:131. [PMID: 40281554 PMCID: PMC12032666 DOI: 10.1186/s13046-025-03394-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2025] [Accepted: 04/14/2025] [Indexed: 04/29/2025] Open
Abstract
This review discusses reprogramming the breast tumor immune microenvironment from an immunosuppressive cold state to an immunologically active hot state. A complex interplay is revealed, in which the accumulation of metabolic byproducts-such as lactate, reactive oxygen species (ROS), and ammonia-is shown to impair T-cell function and promote tumor immune escape. It is demonstrated that the tumor microenvironment (TME) is dominated by immunosuppressive cytokines, including interleukin-10 (IL-10), transforming growth factorβ (TGFβ), and IL-35. Notably, IL-35 is produced by regulatory T cells and breast cancer cells. The conversion of conventional T cells into IL-35-producing induced regulatory T cells, along with the inhibition of pro-inflammatory cytokine secretion, contributes to the suppression of anti-tumor immunity. It is further demonstrated that key immune checkpoint molecules-such as PD-1, PDL1, CTLA-4, TIM-3, LAG-3, and TIGIT-are upregulated within the TME, leading to Tcell exhaustion and diminished immune responses. The blockade of these checkpoints is shown to restore T-cell functionality and is proposed as a strategy to convert cold tumors into hot ones with robust effector cell infiltration. The therapeutic potential of chimeric antigen receptor (CAR)T cell therapy is also explored, and targeting specific tumor-associated antigens, such as glycoproteins and receptor tyrosine kinases, is highlighted. It is suggested that CART cell efficacy can be enhanced by combining these cells with immune checkpoint inhibitors and other immunomodulatory agents, thereby overcoming the barriers imposed by the immunosuppressive TME. Moreover, the role of the microbiome in regulating estrogen metabolism and systemic inflammation is reviewed. Alterations in the gut microbiota are shown to affect the TME, and microbiome-based interventions are proposed as an additional means to facilitate the cold-to-hot transition. It is concluded that by targeting the metabolic and immunological pathways that underpin immune suppression-through combination strategies involving checkpoint blockade, CART cell therapies, and microbiome modulation-the conversion of the breast TME from cold to hot can be achieved. This reprogramming is anticipated to enhance immune cell infiltration and function, thereby improving the overall efficacy of immunotherapies and leading to better clinical outcomes for breast cancer patients.
Collapse
Affiliation(s)
- Saber Imani
- Shulan International Medical College, Zhejiang Shuren University, Hangzhou, Zhejiang, China.
| | - Reyhaneh Farghadani
- Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Jalan Lagoon Selatan, Bandar Sunway, Subang Jaya, 47500, Selangor Darul Ehsan, Malaysia
| | - Ghazaal Roozitalab
- Noncommunicable Diseases Research Center, Fasa University of Medical Sciences, Fasa, Iran
| | - Mazaher Maghsoudloo
- Key Laboratory of Epigenetics and Oncology, The Research Center for Preclinical Medicine, Southwest Medical University, Luzhou, 646000, Sichuan, China
| | - Mahdieh Emadi
- Department of Biology, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Atefeh Moradi
- Department of Life Sciences and System Biology, University of Turin, Turin, Italy
| | - Behnaz Abedi
- Department of Basic Sciences, Faculty of Veterinary Medicine, University of Tabriz, Tabriz, Iran
| | - Parham Jabbarzadeh Kaboli
- Department of Biochemistry, Faculty of Medicine, Medical University of Warsaw, Warsaw, 02-091, Poland.
| |
Collapse
|
21
|
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
|
22
|
Brauneck E, Leonhardt LG, Assemissen AM, Wahid Y, Kruppa M, Kruppa N, Krüger J, Menzel S, Koch-Nolte F, Kylies J, Weisel K, Bokemeyer C, Wellbrock J, Fiedler W, Viezens L, Brauneck F. Expression of the TIGIT axis and the CD39/CD73 purinergic pathway in bone metastasis-derived immune cells. Cancer Immunol Immunother 2025; 74:182. [PMID: 40274631 PMCID: PMC12022200 DOI: 10.1007/s00262-025-04030-2] [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: 02/02/2025] [Accepted: 03/21/2025] [Indexed: 04/26/2025]
Abstract
BACKGROUND Bone metastases (BM) represent one of the most common sites of metastasis. The study aimed to compare the composition of immune cell infiltration from aspirates of different BM prior to systemic therapy. METHOD Phenotypic and functional analyses were conducted via multiparametric flow cytometry (MFC) on BM-derived aspirates obtained from patients with breast cancer (BC, n = 6), patients with prostate cancer (PC, n = 5), patients with non-small-cell lung cancer (NSCLC) (n = 7), patients with myeloma (MM, n = 10) and bone aspirates from age-matched non-malignant controls (NMC, n = 10). RESULTS Across all tumors aspirates the fraction of CD8+ T cells was reduced. In contrast, infiltration by immunosuppressive CD56+CD16-NK and CD163+CD86+ M2-like macrophages was increased in BM compared to NMC aspirates. BM-derived CD8+ T cells aberrantly co-expressed TIGIT with PVRIG or CD39. Similarly, BM-derived cytotoxic NK cells co-expressed TIGIT and PVRIG. In addition, BM-derived M2-like macrophages exhibited an increased subset of cells co-expressing either TIGIT and PVRL4 or CD112 and CD155. Using a myeloma model, functional in vitro studies showed that blockade of TIGIT and CD39 leads to increased PBMC-mediated lysis of myeloma cells. CONCLUSION The study shows that an altered immune cell composition is present in BM across the different tumor entities. Additionally, molecules of the TIGIT checkpoint as well as of the purinergic pathway are aberrantly expressed by BM-infiltrating CD8+ T cells, NK cells and macrophages and also functionally relevant for tumor cell lysis.
Collapse
Affiliation(s)
- Elias Brauneck
- Division of Spine Surgery, Department of Trauma and Orthopedic Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Leon-Gordian Leonhardt
- Division of Spine Surgery, Department of Trauma and Orthopedic Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Anne Marie Assemissen
- Department of Oncology, Hematology and Bone Marrow Transplantation with Section Pneumology, Hubertus Wald University Cancer Center, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Yagana Wahid
- Department of Oncology, Hematology and Bone Marrow Transplantation with Section Pneumology, Hubertus Wald University Cancer Center, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Moritz Kruppa
- Department of Oncology, Hematology and Bone Marrow Transplantation with Section Pneumology, Hubertus Wald University Cancer Center, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Niklas Kruppa
- Department of Oncology, Hematology and Bone Marrow Transplantation with Section Pneumology, Hubertus Wald University Cancer Center, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Julius Krüger
- Department of Oncology, Hematology and Bone Marrow Transplantation with Section Pneumology, Hubertus Wald University Cancer Center, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Stephan Menzel
- Mildred Scheel Cancer Career Center HaTriCS4, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Core Facility Nanobodies, University of Bonn, Venusberg-Campus 1, 53127, Bonn, Germany
| | - Friedrich Koch-Nolte
- Institute of Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Julian Kylies
- Division of Spine Surgery, Department of Trauma and Orthopedic Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Katja Weisel
- Department of Oncology, Hematology and Bone Marrow Transplantation with Section Pneumology, Hubertus Wald University Cancer Center, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Carsten Bokemeyer
- Department of Oncology, Hematology and Bone Marrow Transplantation with Section Pneumology, Hubertus Wald University Cancer Center, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Jasmin Wellbrock
- Department of Oncology, Hematology and Bone Marrow Transplantation with Section Pneumology, Hubertus Wald University Cancer Center, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Walter Fiedler
- Department of Oncology, Hematology and Bone Marrow Transplantation with Section Pneumology, Hubertus Wald University Cancer Center, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Lennart Viezens
- Division of Spine Surgery, Department of Trauma and Orthopedic Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Franziska Brauneck
- Department of Oncology, Hematology and Bone Marrow Transplantation with Section Pneumology, Hubertus Wald University Cancer Center, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.
- Core Facility Nanobodies, University of Bonn, Venusberg-Campus 1, 53127, Bonn, Germany.
| |
Collapse
|
23
|
Yu Y, Liu H, Liu K, Zhao M, Zhang Y, Jiang R, Wang F. Multi-omics identification of a polyamine metabolism related signature for hepatocellular carcinoma and revealing tumor microenvironment characteristics. Front Immunol 2025; 16:1570378. [PMID: 40330470 PMCID: PMC12052762 DOI: 10.3389/fimmu.2025.1570378] [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: 02/03/2025] [Accepted: 04/01/2025] [Indexed: 05/08/2025] Open
Abstract
Background Accumulating evidence indicates that elevated polyamine levels are closely linked to tumor initiation and progression. However, the precise role of polyamine metabolism in hepatocellular carcinoma (HCC) remains poorly understood. Methods We conducted differential expression analysis on bulk RNA sequencing data from The Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO) to identify 65 polyamine metabolism-related genes. By employing unsupervised consensus clustering, AddModuleScore, single-sample gene set enrichment analysis (ssGSEA), and weighted gene co-expression network analysis (WGCNA), we identified polyamine metabolism-related genes at both the bulk RNA-seq and single-cell RNA-seq (scRNA-seq) levels. Utilizing 101 machine learning algorithms, we constructed a polyamine metabolism-related signature (PMRS) and validated its predictive power across training, testing, and external validation cohorts. Additionally, we developed a prognostic nomogram model by integrating PMRS with clinical variables. To explore immune treatment sensitivity, we assessed tumor mutation burden (TMB), tumor immune dysfunction and exclusion (TIDE) score, mutation frequency, and immune checkpoint genes expression. Immune cell infiltration was analyzed using the CIBERSORT algorithm. Finally, RT-qPCR experiments were conducted to validate the expression of key genes. Results Using 101 machine learning algorithms, we established a polyamine metabolism-related signature comprising 9 genes, which exhibited strong prognostic value for HCC patients. Further analysis revealed significant differences in clinical features, biological functions, mutation profiles, and immune cell infiltration between high-risk and low-risk groups. Notably, TIDE analysis and immune phenotype scoring (IPS) demonstrated distinct immune treatment sensitivities between the two risk groups. RT-qPCR validation confirmed that these 9 genes were highly expressed in normal cells but significantly downregulated in tumor cells. Conclusions Our study developed a polyamine metabolism-based prognostic risk signature for HCC, which may provide valuable insights for personalized treatment strategies in HCC patients.
Collapse
Affiliation(s)
- Yuexi Yu
- Department of gastroenterology &hepatology, Tianjin First Center Hospital, Tianjin Key Laboratory for Organ Transplantation, Tianjin Key Laboratory of Molecular Diagnosis and Treatment of Liver Cancer, Tianjin Medical University, Tianjin, China
| | - Huiru Liu
- Department of gastroenterology &hepatology, Tianjin First Center Hospital, Tianjin Key Laboratory for Organ Transplantation, Tianjin Key Laboratory of Molecular Diagnosis and Treatment of Liver Cancer, Tianjin Medical University, Tianjin, China
| | - Kaipeng Liu
- Department of Hepatobiliary Oncology, Liver Cancer Center, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin’s Clinical Research Center for Cancer, Tianjin Medical University, Tianjin, China
| | - Meiqi Zhao
- Department of gastroenterology &hepatology, Tianjin First Center Hospital, Tianjin Key Laboratory for Organ Transplantation, Tianjin Key Laboratory of Molecular Diagnosis and Treatment of Liver Cancer, Nankai University, Tianjin, China
| | - Yiyan Zhang
- Department of gastroenterology &hepatology, Tianjin First Center Hospital, Tianjin Key Laboratory for Organ Transplantation, Tianjin Key Laboratory of Molecular Diagnosis and Treatment of Liver Cancer, Tianjin Medical University, Tianjin, China
| | - Runci Jiang
- Department of gastroenterology &hepatology, Tianjin First Center Hospital, Tianjin Key Laboratory for Organ Transplantation, Tianjin Key Laboratory of Molecular Diagnosis and Treatment of Liver Cancer, Tianjin Medical University, Tianjin, China
| | - Fengmei Wang
- Department of gastroenterology &hepatology, Tianjin First Center Hospital, Tianjin Key Laboratory for Organ Transplantation, Tianjin Key Laboratory of Molecular Diagnosis and Treatment of Liver Cancer, Tianjin Medical University, Tianjin, China
| |
Collapse
|
24
|
Hung JT, Mynam RS, Patel MA, Ozogbo S, LoConte NK, Kratz JD. Immune-Based Therapies in Pancreatic Cancer: a Systematic Review of Ongoing Clinical Trials (2020-2022). J Gastrointest Cancer 2025; 56:103. [PMID: 40259076 DOI: 10.1007/s12029-025-01194-z] [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: 02/09/2025] [Indexed: 04/23/2025]
Abstract
INTRODUCTION Immune-based treatment strategies have emerged across solid organ malignancies largely with the development of immune checkpoint inhibitors. To date, these strategies have not improved clinical outcomes in pancreatic ductal adenocarcinoma (PDAC). METHODS Here, we perform a systematic review to summarize available evidence for recent immune-based treatment strategies in PDAC. We analyze trends in activated clinical trials queried from clinicaltrials.gov in the years 2020-2022. We review study design, sponsorship, and trends in the phase of development. There is a growing emergence of multiple new classes of immune-based targets and combination strategies in early-phase development. RESULTS Immune-based clinical trials in PDAC are highly collaborative including primarily stakeholders in government, industry, and academic medical centers. In this period, a majority of trials have integrated a non-randomized design (83.2%), including a trend towards an increase in Phase I/II clinical trials. This analysis found a growing list of studies using combinations including inhibitors of vascular endothelial growth factors (VEGF), an expanded set of vaccine-based strategies, and the use of Bispecific T-Cell Engagers (BiTEs). Immune checkpoint inhibitors have been a mainstay of combination strategies including the use of new immune checkpoint inhibitors (CD40, TIGIT). CONCLUSION Immune-based strategies in PDAC have expanded across new targets and the complexities of combinatory approaches. Integrating this work across key stakeholders remains of critical importance to improve clinical outcomes.
Collapse
Affiliation(s)
- Justine T Hung
- Division of Hematology, Medical Oncology and Palliative Care, Department of Medicine, University of Wisconsin School of Medicine and Public Health, University of Wisconsin, Madison, WI, USA
| | - Ritwick S Mynam
- Division of Hematology, Medical Oncology and Palliative Care, Department of Medicine, University of Wisconsin School of Medicine and Public Health, University of Wisconsin, Madison, WI, USA
| | - Monica A Patel
- Division of Hematology, Medical Oncology and Palliative Care, Department of Medicine, University of Wisconsin School of Medicine and Public Health, University of Wisconsin, Madison, WI, USA
- University of Wisconsin Carbone Cancer Center, Madison, WI, USA
| | - Stanley Ozogbo
- Department of Internal Medicine, St Elizabeth Hospital, Youngstown, OH, USA
| | - Noelle K LoConte
- Division of Hematology, Medical Oncology and Palliative Care, Department of Medicine, University of Wisconsin School of Medicine and Public Health, University of Wisconsin, Madison, WI, USA
- University of Wisconsin Carbone Cancer Center, Madison, WI, USA
| | - Jeremy D Kratz
- Division of Hematology, Medical Oncology and Palliative Care, Department of Medicine, University of Wisconsin School of Medicine and Public Health, University of Wisconsin, Madison, WI, USA.
- University of Wisconsin Carbone Cancer Center, Madison, WI, USA.
- William S. Middleton Veterans Administration Health System, Madison, WI, USA.
- Center for Human Genomics and Precision Medicine, University of Wisconsin, 1111 Highland Ave, 2784 West Wedge, MadisonWI, WI, 53705, USA.
| |
Collapse
|
25
|
Cho MM, Song L, Quamine AE, Szewc F, Shi L, Ebben JD, Turicek DP, Kline JM, Burpee DM, Lafeber EO, Phillips MF, Ceas AS, Bates PD, Forsberg MH, Kink JA, Erbe AK, Capitini CM. CD155 blockade enhances allogeneic natural killer cell-mediated antitumor response against osteosarcoma. J Immunother Cancer 2025; 13:e008755. [PMID: 40234092 PMCID: PMC12001373 DOI: 10.1136/jitc-2023-008755] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Accepted: 03/10/2025] [Indexed: 04/17/2025] Open
Abstract
BACKGROUND Allogeneic bone marrow transplant (alloBMT) is curative for hematologic malignancies through the graft-versus-tumor (GVT) effect but has been ineffective for solid tumors like osteosarcoma (OS). OS expresses CD155 which interacts strongly with inhibitory receptors TIGIT and CD96 but also binds to activating receptor DNAM-1 on natural killer (NK) cells. CD155 has never been targeted after alloBMT. Combining adoptively transferred allogeneic NK (alloNK) cells with CD155 blockade after alloBMT may enhance a GVT effect against OS. METHODS Murine NK cells were activated and expanded ex vivo with superagonist interleukin (IL)-15/IL-15Rα. AlloNK and syngeneic NK (synNK) cell phenotype, cytotoxicity, cytokine production, and degranulation against CD155-expressing murine OS cell line K7M2 were assessed in vitro. Mice bearing pulmonary OS metastases underwent alloBMT and alloNK cell infusion with anti-CD155 either before or after tumor induction, with select groups receiving anti-DNAM-1 pretreated alloNK cells. Tumor growth, graft-versus-host disease and survival were monitored, and differential gene expression of lung tissue was assessed by RNA microarray. RESULTS AlloNK cells exhibited superior cytotoxicity against CD155-expressing OS compared with synNK cells, and this activity was enhanced by CD155 blockade. CD155 blockade increased alloNK cell degranulation and interferon gamma production through DNAM-1. In vivo, CD155 blockade with alloNK infusion increased survival when treating OS that relapsed after alloBMT. No benefit was seen for treating established OS before alloBMT. Combining CD155 and anti-DNAM-1 pretreated alloNK did not affect survival and tumor control benefits seen with CD155 blockade alone. RNA microarray showed mice treated with alloNK and CD155 blockade had increased expression of cytotoxicity genes and the NKG2D ligand H60a, whereas mice treated with anti-DNAM-1 pretreated alloNK cells resulted in upregulation of NK cell inhibitory receptor genes. Whereas blocking DNAM-1 on alloNK abrogated cytotoxicity, blocking NKG2D had no effect, implying DNAM-1:CD155 engagement drives alloNK activation against OS. CONCLUSIONS These results demonstrate the safety and efficacy of infusing alloNK cells with CD155 blockade to mount a GVT effect against OS and show benefits are in part through DNAM-1. Defining the hierarchy of receptors that govern alloNK responses is critical to translating alloNK cell infusions and immune checkpoint inhibition for solid tumors treated with alloBMT.
Collapse
Affiliation(s)
- Monica M Cho
- Department of Pediatrics, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, USA
| | - Longzhen Song
- Department of Pediatrics, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, USA
| | - Aicha E Quamine
- Department of Pediatrics, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, USA
| | - Fernanda Szewc
- Department of Pediatrics, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, USA
| | - Lei Shi
- Department of Pediatrics, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, USA
| | - Johnathan D Ebben
- Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, USA
- Carbone Cancer Center, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, USA
| | - David P Turicek
- Department of Pediatrics, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, USA
| | - Jillian M Kline
- Department of Pediatrics, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, USA
| | - Devin M Burpee
- Department of Pediatrics, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, USA
| | - Emily O Lafeber
- Department of Pediatrics, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, USA
| | - Madison F Phillips
- Department of Pediatrics, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, USA
| | - Amanda S Ceas
- Department of Pediatrics, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, USA
| | - Paul D Bates
- Department of Pediatrics, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, USA
| | - Matthew H Forsberg
- Department of Pediatrics, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, USA
| | - John A Kink
- Department of Pediatrics, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, USA
- Carbone Cancer Center, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, USA
| | - Amy K Erbe
- Carbone Cancer Center, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, USA
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, USA
| | - Christian M Capitini
- Department of Pediatrics, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, USA
- Carbone Cancer Center, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, USA
| |
Collapse
|
26
|
Liu M, Gao Y, Li L, Ge L, Yao L, Chu X, Wu F, Tian J. Anti-TIGIT inhibitors plus PD-1 or PD-L1 inhibitors versus PD-1 or PD-L1 inhibitors for first-line treatment of advanced non-small cell lung cancer. Cochrane Database Syst Rev 2025; 4:CD015888. [PMID: 40226904 PMCID: PMC11995688 DOI: 10.1002/14651858.cd015888] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 04/15/2025]
Abstract
OBJECTIVES This is a protocol for a Cochrane Review (intervention). The objectives are as follows: To evaluate the effectiveness and safety of anti-TIGIT inhibitors in combination with PD-1 inhibitors or PD-L1 inhibitors compared to treatment with PD-1 inhibitors or PD-L1 inhibitors in the first-line treatment of advanced NSCLC. To assess the effectiveness and safety of anti-TIGIT inhibitors in combination with chemotherapy and PD-1 inhibitors or PD-L1 inhibitors compared to treatment with chemotherapy plus PD-1 inhibitors or PD-L1 inhibitors in the first-line treatment of advanced NSCLC.
Collapse
Affiliation(s)
- Ming Liu
- Evidence-Based Medicine Center, School of Basic Medical Sciences, Lanzhou University, Lanzhou city, China
| | - Ya Gao
- Department of Medical Data, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, China
- Institute for Medical Dataology, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Lun Li
- Department of Breast Surgery, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Long Ge
- Evidence-Based Social Science Research Centre, School of Public Health, Lanzhou University, Lanzhou city, China
| | - Liang Yao
- Lee Kong Chian School of Medicine, Nanyang Technological University Singapore, Singapore, Singapore
| | - Xiajing Chu
- Department of Health Research Methods, Evidence, and Impact, McMaster University, Hamilton, Canada
| | - Fanqi Wu
- Department of Respirator, Lanzhou University Second Hospital, Lanzhou, China
| | - Jinhui Tian
- Evidence-Based Medicine Center, School of Basic Medical Sciences, Lanzhou University, Lanzhou city, China
| |
Collapse
|
27
|
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
|
28
|
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
|
29
|
Huang S, Kang Y, Liu T, Xiong Y, Yang Z, Zhang Q. The role of immune checkpoints PD-1 and CTLA-4 in cardiovascular complications leading to heart failure. Front Immunol 2025; 16:1561968. [PMID: 40255399 PMCID: PMC12006013 DOI: 10.3389/fimmu.2025.1561968] [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: 01/16/2025] [Accepted: 02/13/2025] [Indexed: 04/22/2025] Open
Abstract
Immune checkpoints, such as PD-1 and CTLA-4, are crucial regulators of immune responses, acting as gatekeepers to balance immunity against foreign antigens and self-tolerance. These checkpoints play a key role in maintaining cardiac homeostasis by preventing immune-mediated damage to critical organs like the heart. In this study, we explored the involvement of PD-1 and CTLA-4 in cardiovascular complications, particularly atherosclerosis and myocarditis, which can lead to heart failure. We conducted a comprehensive analysis using animal models and clinical data to assess the effects of immune checkpoint inhibition on cardiac function. Our findings indicate that disruption of PD-1 and CTLA-4 pathways exacerbates myocardial inflammation, accelerates atherosclerotic plaque formation, and promotes the development of heart failure. Additionally, we observed that immune checkpoint inhibition in these models led to increased infiltration of T lymphocytes, higher levels of pro-inflammatory cytokines, and enhanced tissue damage. These results suggest that PD-1 and CTLA-4 are critical in preserving cardiac health, and their inhibition can result in severe cardiovascular toxicity. Our study emphasizes the need for careful monitoring of cardiovascular health in patients undergoing immune checkpoint inhibitor therapies.
Collapse
Affiliation(s)
- Shoulian Huang
- Department of Cardiology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Department of Cardiology, The Second People’s Hospital of Yibin, Yibin, Sichuan, China
| | - Yu Kang
- Department of Cardiology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Ting Liu
- Department of Cardiology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yan Xiong
- Institute of Cardiovascular Diseases & Department of Cardiology, Sichuan Provincial People’s Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Zixuan Yang
- Department of Cardiology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Qing Zhang
- Department of Cardiology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
30
|
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
|
31
|
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: 1] [Impact Index Per Article: 1.0] [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
|
32
|
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
|
33
|
Fu M, Xue B, Miao X, Gao Z. Overcoming immunotherapy resistance in glioblastoma: challenges and emerging strategies. Front Pharmacol 2025; 16:1584688. [PMID: 40223940 PMCID: PMC11987931 DOI: 10.3389/fphar.2025.1584688] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2025] [Accepted: 03/21/2025] [Indexed: 04/15/2025] Open
Abstract
Glioblastoma (GBM) is the most common and aggressive primary brain tumor in adults, characterized by rapid proliferation, extensive infiltration, and significant intratumoral heterogeneity. Despite advancements in conventional treatments, including surgery, radiotherapy, and chemotherapy, the prognosis for GBM patients remains poor, with a median survival of approximately 15 months. Immunotherapy has emerged as a promising alternative; however, the unique biological and immunological features, including its immunosuppressive tumor microenvironment (TME) and low mutational burden, render it resistant to many immunotherapeutic strategies. This review explores the key challenges in GBM immunotherapy, focusing on immune evasion mechanisms, the blood-brain barrier (BBB), and the TME. Immune checkpoint inhibitors and CAR-T cells have shown promise in preclinical models but have limited clinical success due to antigen heterogeneity, immune cell exhaustion, and impaired trafficking across the BBB. Emerging strategies, including dual-targeting CAR-T cells, engineered immune cells secreting therapeutic molecules, and advanced delivery systems to overcome the BBB, show potential for enhancing treatment efficacy. Addressing these challenges is crucial for improving GBM immunotherapy outcomes.
Collapse
Affiliation(s)
- Maowu Fu
- Department of Neurosurgery, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Bing Xue
- Department of Neurosurgery, Jinan Third People’s Hospital, Jinan, Shandong, China
| | - Xiuming Miao
- Department of Pathology, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Zong Gao
- Department of Neurosurgery, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| |
Collapse
|
34
|
Chan LL, Chan SL. Future perspectives on immunotherapy for hepatocellular carcinoma. Ther Adv Med Oncol 2025; 17:17588359251323199. [PMID: 40144682 PMCID: PMC11938898 DOI: 10.1177/17588359251323199] [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: 10/26/2024] [Accepted: 02/05/2025] [Indexed: 03/28/2025] Open
Abstract
In recent years, several global phase III trials have shown that combinations of immune checkpoint inhibitors (ICIs) offer superior efficacy and survival compared to multi-kinase inhibitors, establishing them as the gold standard for treating patients with advanced hepatocellular carcinoma (HCC). This success has led to investigations into expanding the use of immunotherapy into various other settings and populations, including neoadjuvant and adjuvant therapies, patients with decompensated liver function and those awaiting liver transplantation. Despite its proven efficacy, a significant number of patients still develop resistance to immunotherapy, highlighting the need for innovative strategies to address this challenge. Approaches aimed at enhancing tumour immunogenicity, such as combining immunotherapy with transarterial chemoembolization or radiation therapies, show significant promise. Additionally, novel immunotherapeutics - such as triplet therapy, bispecific antibodies, adoptive T-cell therapy and cancer vaccines - are in early development for HCC. These agents have demonstrated potential for synergistic effects with existing ICIs, with initial studies yielding positive outcomes. In this review, we offer our future perspective on immunotherapy, emphasizing emerging indications, novel combination strategies and the development of new immunotherapeutic agents. Overall, the future of immunotherapy in HCC is brimming with extraordinary potential, set to transform the treatment landscape and redefine the possibilities for managing this challenging disease.
Collapse
Affiliation(s)
- Landon L. Chan
- State Key Laboratory of Translational Oncology, Department of Clinical Oncology, Sir YK Pao Centre for Cancer, SIRT, Hong Kong Cancer Institute, Prince of Wales Hospital, The Chinese University of Hong Kong, Shatin, Hong Kong, China
| | - Stephen L. Chan
- State Key Laboratory of Translational Oncology, Department of Clinical Oncology, Sir YK Pao Centre for Cancer, SIRT, Hong Kong Cancer Institute, Prince of Wales Hospital, The Chinese University of Hong Kong, 30-32 Ngan Shing Street, Shatin, Hong Kong, China
| |
Collapse
|
35
|
Han Z, Chen G, Wang D. Emerging immunotherapies in osteosarcoma: from checkpoint blockade to cellular therapies. Front Immunol 2025; 16:1579822. [PMID: 40170852 PMCID: PMC11958959 DOI: 10.3389/fimmu.2025.1579822] [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: 02/19/2025] [Accepted: 03/04/2025] [Indexed: 04/03/2025] Open
Abstract
Osteosarcoma remains a highly aggressive bone malignancy with limited therapeutic options, necessitating novel treatment strategies. Immunotherapy has emerged as a promising approach, yet its efficacy in osteosarcoma is hindered by an immunosuppressive tumor microenvironment and resistance mechanisms. This review explores recent advancements in checkpoint blockade, cellular therapies, and combination strategies aimed at enhancing immune responses. We highlight key challenges, including tumor heterogeneity, poor immune infiltration, and the need for predictive biomarkers. By integrating immunotherapy with chemotherapy, radiotherapy, and targeted therapy, emerging approaches seek to improve treatment outcomes. This review provides a comprehensive analysis of the evolving landscape of osteosarcoma immunotherapy, offering insights into future directions and potential breakthroughs. Researchers and clinicians will benefit from understanding these developments, as they pave the way for more effective and personalized therapeutic strategies in osteosarcoma.
Collapse
Affiliation(s)
- Zhiwei Han
- Department of Orthopedics, Central Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Guomin Chen
- Laboratory Medicine, Central Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Dongchen Wang
- Department of Orthopedics, Central Hospital Affiliated to Shandong First Medical University, Jinan, China
| |
Collapse
|
36
|
Jaworska J, Tobolski D, Salem SE, Kahler A, Wocławek-Potocka I, de Mestre AM. Single-cell atlas of the pregnant equine endometrium before and after implantation†. Biol Reprod 2025; 112:458-473. [PMID: 39756438 DOI: 10.1093/biolre/ioaf004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Revised: 12/05/2024] [Accepted: 01/03/2025] [Indexed: 01/07/2025] Open
Abstract
Embryo implantation in the mare occurs just over one month after fertilization, coinciding with the production of chorionic gonadotropin. The factors that regulate this late implantation in the mare, and whether they are unique to horses or shared with more invasive embryo implantation in other species, remain poorly understood. This study aimed to determine and compare the transcriptome and subpopulations of endometrial cells before and after embryo implantation in the horse. Single-cell RNA sequencing was used to characterize the transcriptome of nearly 97,000 endometrial cells collected from biopsies of the endometrium at the beginning (day 33 of gestation) and after embryo implantation (day 42 of gestation) in mares. Sixteen immune and 24 non-immune cell clusters were identified, representing known major cell populations as well as novel subpopulations of horse immune cells such as resident innate lymphoid cells and mucosal-associated invariant T cells. Contrary to current knowledge, endometrial natural killer (eNK) cells were the most abundant endometrial leukocyte population during implantation in horses. Moreover, eNK cells not only expressed genes that may interact with fetal MHC I, such as LY49F, but also exert immunoregulatory functions independent of MHC I expression, such as CD96/TIGIT. Analogous to other species studied, upregulation of CXCR4 was found in several subpopulations of immune cells. Our results suggest that despite distinctive and later placentation compared with humans, horses share some key similarities in the mechanisms of embryo implantation.
Collapse
Affiliation(s)
- Joanna Jaworska
- Department of Gamete and Embryo Biology, Institute of Animal Reproduction and Food Research, Polish Academy of Sciences in Olsztyn, Olsztyn, Poland
| | - Dawid Tobolski
- Department of Large Animal Diseases and Clinic, University of Life Sciences, Warsaw, Poland
| | - Shebl E Salem
- Department of Biomedical Sciences, Baker Institute for Animal Health, Cornell University, Ithaca, NY USA
| | - Anne Kahler
- Department of Comparative Biomedical Sciences, Royal Veterinary College, Hatfield, Hertfordshire, UK
| | - Izabela Wocławek-Potocka
- Department of Gamete and Embryo Biology, Institute of Animal Reproduction and Food Research, Polish Academy of Sciences in Olsztyn, Olsztyn, Poland
| | - Amanda M de Mestre
- Department of Biomedical Sciences, Baker Institute for Animal Health, Cornell University, Ithaca, NY USA
- Department of Comparative Biomedical Sciences, Royal Veterinary College, Hatfield, Hertfordshire, UK
| |
Collapse
|
37
|
Carter NM, Hankore WD, Yang YK, Yang C, Hutcherson SM, Fales W, Ghosh A, Mongia P, Mackinnon S, Brennan A, Leone RD, Pomerantz JL. QRICH1 mediates an intracellular checkpoint for CD8 + T cell activation via the CARD11 signalosome. Sci Immunol 2025; 10:eadn8715. [PMID: 40085689 DOI: 10.1126/sciimmunol.adn8715] [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: 01/08/2024] [Accepted: 02/19/2025] [Indexed: 03/16/2025]
Abstract
Antigen receptor signaling pathways that control lymphocyte activation depend on signaling hubs and negative regulatory proteins to fine-tune signaling outputs to ensure host defense and avoid pathogenic responses. Caspase recruitment domain-containing protein 11 (CARD11) is a critical signaling scaffold that translates T cell receptor (TCR) triggering into the activation of nuclear factor κB (NF-κB), c-Jun N-terminal kinase (JNK), mechanistic target of rapamycin (mTOR), and Akt. Here, we identify glutamine-rich protein 1 (QRICH1) as a regulator of CARD11 signaling that mediates an intracellular checkpoint for CD8+ T cell activation. QRICH1 associates with CARD11 after TCR engagement and negatively regulates CARD11 signaling to NF-κB. QRICH1 binding to CARD11 is controlled by an autoregulatory intramolecular interaction between QRICH1 domains of previously uncharacterized function. QRICH1 controls the antigen-induced activation, proliferation, and effector status of CD8+ T cells by regulating numerous genes critical for CD8+ T cell function. Our results define a component of antigen receptor signaling circuitry that fine-tunes effector output in response to antigen recognition.
Collapse
Affiliation(s)
- Nicole M Carter
- Department of Biological Chemistry and Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Wihib D Hankore
- Department of Biological Chemistry and Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Yong-Kang Yang
- Department of Biological Chemistry and Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Chao Yang
- Department of Biological Chemistry and Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Shelby M Hutcherson
- Department of Biological Chemistry and Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Wyatt Fales
- Department of Biological Chemistry and Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Anushka Ghosh
- Department of Biological Chemistry and Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Piyusha Mongia
- Department of Biological Chemistry and Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Sophie Mackinnon
- Department of Biological Chemistry and Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Anna Brennan
- Department of Biological Chemistry and Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Robert D Leone
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Joel L Pomerantz
- Department of Biological Chemistry and Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
38
|
Vigano M, Wang L, As’sadiq A, Samarani S, Ahmad A, Costiniuk CT. Impact of cannabinoids on cancer outcomes in patients receiving immune checkpoint inhibitor immunotherapy. Front Immunol 2025; 16:1497829. [PMID: 40109334 PMCID: PMC11919899 DOI: 10.3389/fimmu.2025.1497829] [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: 09/17/2024] [Accepted: 02/10/2025] [Indexed: 03/22/2025] Open
Abstract
Cannabinoids relieve pain, nausea, anorexia and anxiety, and improve quality of life in several cancer patients. The immunotherapy with checkpoint inhibitors (ICIs), although very successful in a subset of patients, is accompanied by moderate to severe immune-related adverse events (ir-AE) that often necessitate its discontinuation. Because of their role in symptomatic relief, cannabinoids have been used in combination with immune checkpoint inhibitor (ICI) immunotherapy. A few studies strongly suggest that the use of medicinal cannabis in cancer patients attenuates many of the ir-AE associated with the use of ICI immunotherapy and increase its tolerability. However, no significant beneficial effects on overall survival, progression free survival or cancer relapses were observed; rather, some of the studies noted adverse effects of concurrent administration of cannabinoids with ICI immunotherapy on the clinical benefits of the latter. Because of cannabinoids' well documented immunosuppressive effects mediated through the cannabinoid recptor-2 (CB2), we propose considering this receptor as an inhibitory immune checkpoint per se. A simultaneous neutralization of CB2, concurrent with cannabinoid treatment, may lead to better clinical outcomes in cancer patients receiving ICI immunotherapy. In this regard, cannabinoids such as cannabidiol (CBD) and cannabigerol (CBG), with little agonism for CB2, may be better therapeutic choices. Additional strategies e.g., the use of monoacylglycerol lipase (MAGL) inhibitors that degrade some endocannabinoids as well as lipogenesis and formation of lipid bilayers in cancer cells may also be explored. Future studies should take into consideration gut microbiota, CYP450 polymorphism and haplotypes, cannabinoid-drug interactions as well as genetic and somatic variations occurring in the cannabinoid receptors and their signaling pathways in cancer cells for personalized cannabis-based therapies in cancer patients receiving ICIs. This may lead to rational knowledge-based regimens tailored to individual cancer patients.
Collapse
Affiliation(s)
- MariaLuisa Vigano
- Division of Experimental Medicine, Faculty of Medicine and Health Sciences, McGill University, Montreal, QC, Canada
- Infectious Diseases and Immunity in Global Health Program, Research Institute of the McGill University Health Centre, Montreal, QC, Canada
| | - Lixing Wang
- Department of Microbiology and Immunology, McGill University, Montreal, QC, Canada
| | - Alia As’sadiq
- Division of Experimental Medicine, Faculty of Medicine and Health Sciences, McGill University, Montreal, QC, Canada
- Infectious Diseases and Immunity in Global Health Program, Research Institute of the McGill University Health Centre, Montreal, QC, Canada
| | - Suzanne Samarani
- Infectious Diseases and Immunity in Global Health Program, Research Institute of the McGill University Health Centre, Montreal, QC, Canada
| | - Ali Ahmad
- Infectious Diseases and Immunity in Global Health Program, Research Institute of the McGill University Health Centre, Montreal, QC, Canada
| | - Cecilia T. Costiniuk
- Division of Experimental Medicine, Faculty of Medicine and Health Sciences, McGill University, Montreal, QC, Canada
- Infectious Diseases and Immunity in Global Health Program, Research Institute of the McGill University Health Centre, Montreal, QC, Canada
- Division of Infectious Diseases and Chronic Viral Illnesses Service, McGill University Health Centre, Montreal, QC, Canada
| |
Collapse
|
39
|
Liu L, Wu P, Wang B, Dong J, Zhang C, Liu W, Ying J. Exploring the role of TIGIT in patients with Small Cell Lung Cancer as a novel predictor of prognosis and immunotherapy response. Cancer Immunol Immunother 2025; 74:134. [PMID: 40035834 PMCID: PMC11880484 DOI: 10.1007/s00262-025-03985-6] [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: 09/11/2024] [Accepted: 02/17/2025] [Indexed: 03/06/2025]
Abstract
BACKGROUND T-cell immunoreceptor with immunoglobulin and immunoreceptor tyrosine-based inhibitory motif domains (TIGIT) is a novel immune checkpoint playing a crucial role in immunosuppression and immune evasion. This study aims to elucidate the expression patterns, characteristics, and possible mechanisms of TIGIT in small cell lung cancer (SCLC). METHODS TIGIT expression was analyzed across various cancers and normal tissues using The Cancer Genome Atlas (TCGA). Transcriptomic data from SCLC patients, sourced from the Gene Expression Omnibus (GEO) and literature, were analyzed to assess TIGIT-related characteristics. Immunohistochemistry (IHC) was used to verify TIGIT expression in post-surgical and advanced SCLC samples, focusing on expression characteristics, prognostic value, and treatment response. RESULTS TIGIT was significantly overexpressed in various tumors, including SCLC (p < 0.05). Higher expression was associated with better overall survival (OS) (p < 0.05). Notably, a significant positive correlation was observed between TIGIT expression and immune-related metagenes, such as HCK, interferon, and LCK (p < 0.05). Immune infiltration analysis revealed a strong positive correlation between TIGIT expression and immune score in multiple cohorts. Additionally, TIGIT expression correlated positively with immune cells, including CD8 T cells, cytotoxic lymphocytes, and B cells (p < 0.05), and multiple immune checkpoints like BTLA, ICOS, and LAG3 (p < 0.05), while it had a significant negative correlation with the TIDE score (p < 0.05). In the validation section, patients with high TIGIT expression showed significantly prolonged disease-free survival (DFS) and OS (p < 0.05), and demonstrated a better response to adjuvant chemotherapy (ACT) and immunotherapy. CONCLUSION TIGIT serves as a biomarker in SCLC, with its high expression indicating favorable prognosis and treatment response. These effects may be due to TIGIT's unique immune landscape and its association with other immune checkpoints.
Collapse
Affiliation(s)
- Li Liu
- Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Peng Wu
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Bingzhi Wang
- Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Jiyan Dong
- Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Chaoqi Zhang
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Wenchao Liu
- Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Jianming Ying
- Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China.
| |
Collapse
|
40
|
Attrill MH, Shinko D, Viveiros TM, Milighetti M, de Gruijter NM, Jebson B, Kartawinata M, Rosser EC, Wedderburn LR, Pesenacker AM. Treg fitness signatures as a biomarker for disease activity in Juvenile Idiopathic Arthritis. J Autoimmun 2025; 152:103379. [PMID: 39954509 DOI: 10.1016/j.jaut.2025.103379] [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: 10/16/2024] [Revised: 01/06/2025] [Accepted: 02/01/2025] [Indexed: 02/17/2025]
Abstract
Juvenile Idiopathic Arthritis (JIA) is an autoimmune condition characterised by flares of joint inflammation. However, no reliable biomarker exists to predict the erratic disease course. Normally, regulatory T cells (Tregs) maintain tolerance, with altered Tregs associated with autoimmunity. Treg signatures have shown promise in monitoring other conditions, therefore a Treg gene/protein signature could offer novel biomarker potential for predicting disease activity in JIA. Machine learning on our nanoString Treg 48-gene signature on peripheral blood (PB) Tregs generated a model to distinguish active JIA (active joint count, AJC≥1) Tregs from healthy controls (HC, AUC = 0.9875 on test data). Biomarker scores from this model successfully differentiated inactive (AJC = 0) from active JIA PB Tregs. Moreover, scores correlated with clinical activity scores (cJADAS), and discriminated subclinical disease (AJC = 0, cJADAS≥0.5) from remission (cJADAS<0.5). To investigate altered protein expression as a surrogate measure for Treg fitness in JIA, we utilised spectral flow cytometry and unbiased clustering analysis. Three Treg clusters were of interest in active JIA PB, including TIGIThighCD226highCD25low Teff-like Tregs, CD39-TNFR2-Helioshigh, and a 4-1BBlowTIGITlowID2intermediate Treg cluster predominated in inactive JIA PB (AJC = 0). The ratio of these Treg clusters correlated to cJADAS, and higher ratios could potentially predict inactive individuals that flared by 9-month follow-up. Thus, we demonstrate altered Treg signatures and subsets as an important factor, and useful biomarker, for disease progression versus remission in JIA, revealing genes and proteins contributing to Treg fitness. Ultimately, PB Treg fitness measures could serve as routine biomarkers to guide disease and treatment management to sustain remission in JIA.
Collapse
Affiliation(s)
- Meryl H Attrill
- Institute of Immunity and Transplantation, Division of Infection and Immunity, UCL, London, NW3 2PP, UK; Infection, Immunity & Inflammation Research and Teaching Department, UCL Great Ormond Street Institute of Child Health, UCL, London, WC1N 1EH, UK
| | - Diana Shinko
- Institute of Immunity and Transplantation, Division of Infection and Immunity, UCL, London, NW3 2PP, UK
| | - Telma Martins Viveiros
- Institute of Immunity and Transplantation, Division of Infection and Immunity, UCL, London, NW3 2PP, UK
| | - Martina Milighetti
- Institute of Immunity and Transplantation, Division of Infection and Immunity, UCL, London, NW3 2PP, UK; Cancer Institute, UCL, London, WC1E 6DD, UK
| | - Nina M de Gruijter
- Centre for Adolescent Rheumatology Versus Arthritis at UCL UCLH and GOSH, London, WC1E 6JF, UK; Division of Medicine, UCL, London, WC1E 6JF, UK
| | - Bethany Jebson
- Infection, Immunity & Inflammation Research and Teaching Department, UCL Great Ormond Street Institute of Child Health, UCL, London, WC1N 1EH, UK; Centre for Adolescent Rheumatology Versus Arthritis at UCL UCLH and GOSH, London, WC1E 6JF, UK
| | - Melissa Kartawinata
- Infection, Immunity & Inflammation Research and Teaching Department, UCL Great Ormond Street Institute of Child Health, UCL, London, WC1N 1EH, UK; Centre for Adolescent Rheumatology Versus Arthritis at UCL UCLH and GOSH, London, WC1E 6JF, UK
| | - Elizabeth C Rosser
- Centre for Adolescent Rheumatology Versus Arthritis at UCL UCLH and GOSH, London, WC1E 6JF, UK; Division of Medicine, UCL, London, WC1E 6JF, UK
| | - Lucy R Wedderburn
- Infection, Immunity & Inflammation Research and Teaching Department, UCL Great Ormond Street Institute of Child Health, UCL, London, WC1N 1EH, UK; Centre for Adolescent Rheumatology Versus Arthritis at UCL UCLH and GOSH, London, WC1E 6JF, UK; NIHR Biomedical Research Centre at GOSH, London, WC1N 1EH, UK
| | - Anne M Pesenacker
- Institute of Immunity and Transplantation, Division of Infection and Immunity, UCL, London, NW3 2PP, UK.
| |
Collapse
|
41
|
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
|
42
|
Xie Z, Yang T, Zhou C, Xue Z, Wang J, Lu F. Integrative Bioinformatics Analysis and Experimental Study of NLRP12 Reveal Its Prognostic Value and Potential Functions in Ovarian Cancer. Mol Carcinog 2025; 64:383-398. [PMID: 39601513 DOI: 10.1002/mc.23854] [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: 07/26/2024] [Revised: 11/04/2024] [Accepted: 11/08/2024] [Indexed: 11/29/2024]
Abstract
NLRP12 plays a significant role in cellular functional behavior and immune homeostasis, influencing inflammation, tumorigenesis, and prognosis. This study aimed to explore its specific effects on the tumor microenvironment (TME) and its contribution to heterogeneity in ovarian cancer (OV) through bioinformatics analysis and experimental verification. Utilizing various bioinformatics databases and clinical specimens, we investigated NLRP12 expression and its relationship with OV prognosis and immune infiltration. In vitro assays were conducted to assess the impact of NLRP12 on the proliferation and invasion of OV cells. Our findings indicate that NLRP12 is upregulated in OV, with high expression correlating with a negative prognosis. Furthermore, NLRP12 expression demonstrated a positive correlation with the infiltration of various immune cells and the expression of immune checkpoint molecules in OV. Analysis of The Cancer Immunome Atlas (TCIA) database revealed that OV patients with lower NLRP12 expression may exhibit an enhanced response to immunotherapy, particularly CTLA4 blockers, a finding validated in animal experiments. Additionally, the study emphasized the role of NLRP12 in influencing the prognosis of OV patients by promoting epithelial-mesenchymal transition (EMT) in ovarian cancer cells. Finally, we identified a potential therapeutic compound, Schisandrin B (Schi B), which decreases NLRP12 expression in ovarian cancer cells by binding to the transcription factor SPI1 associated with NLRP12. Our findings suggest that NLRP12 serves as a crucial immune-related biomarker predicting poor outcomes in OV, and targeting NLRP12 may represent a promising therapeutic approach for OV patients in the future.
Collapse
Affiliation(s)
- Zhihui Xie
- Department of Medical Oncology, Huaihe Hospital of Henan University, Kaifeng, China
| | - Tiantian Yang
- Department of Immunology, School of Basic Medical Sciences, Henan University, Kaifeng, China
- Joint National Laboratory for Antibody Drug Engineering, Medical School, Henan University, Kaifeng, China
| | - Chuchu Zhou
- Department of Immunology, School of Basic Medical Sciences, Henan University, Kaifeng, China
- Joint National Laboratory for Antibody Drug Engineering, Medical School, Henan University, Kaifeng, China
| | - Zixin Xue
- Department of Immunology, School of Basic Medical Sciences, Henan University, Kaifeng, China
- Joint National Laboratory for Antibody Drug Engineering, Medical School, Henan University, Kaifeng, China
| | - Jianjun Wang
- Department of Medical Oncology, Huaihe Hospital of Henan University, Kaifeng, China
| | - Feng Lu
- Department of Medical Oncology, Huaihe Hospital of Henan University, Kaifeng, China
- Department of Immunology, School of Basic Medical Sciences, Henan University, Kaifeng, China
- Joint National Laboratory for Antibody Drug Engineering, Medical School, Henan University, Kaifeng, China
| |
Collapse
|
43
|
Cao L, Ba Y, Chen F, Li D, Zhang S, Zhang H. The prognostic significance of epoxide hydrolases in colorectal cancer. Biochem Biophys Rep 2025; 41:101912. [PMID: 39850362 PMCID: PMC11754166 DOI: 10.1016/j.bbrep.2024.101912] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2024] [Revised: 12/27/2024] [Accepted: 12/30/2024] [Indexed: 01/25/2025] Open
Abstract
Colorectal cancer (CRC) is a common malignant cancer. Epoxide hydrolases (EHs) are involved in the development of cancer by regulating epoxides, but their relationship with CRC is unclear. We used multiple datasets to confirm the expression of different EPHX family members in CRC tissues, and to explore their association with different clinicopathologic characteristics. The Kaplan-Meier method, correlation analysis and random forest algorithm were used to evaluate the prognostic value of EPHX family members for CRC. Finally, the cell experiment verified function of EPHX4 in CRC. The expressions of EPHX1 and EPHX2 were significantly decreased, while those of EPHX3 and EPHX4 were significantly increased in CRC. The expressions of EPHX family members were correlated with some clinicopathologic features and overall survival. The expressions of the EPHX family were positively associated with CD274, CTLA4, HAVCR2, and TIGIT. EPHX2 and EPHX4 were diagnostic and predictive biomarkers for CRC. EPHX4 promoted the malignant phenotype of CRC cells. Our study firstly elucidated the prognostic significance of EPHX family members in CRC and identified novel diagnostic and prognostic biomarkers for CRC.
Collapse
Affiliation(s)
- Lichao Cao
- Shenzhen Nucleus Gene Technology Co., Ltd., Shenzhen, 518071, China
- Shanghai Nucleus Biotechnology Co., Ltd., Shanghai, 201401, China
- Shenzhen Nucleus Huaxi Medical Laboratory, Shenzhen, 518110, China
| | - Ying Ba
- Shenzhen Nucleus Gene Technology Co., Ltd., Shenzhen, 518071, China
- Shanghai Nucleus Biotechnology Co., Ltd., Shanghai, 201401, China
- Shenzhen Nucleus Huaxi Medical Laboratory, Shenzhen, 518110, China
| | - Fang Chen
- Shenzhen Nucleus Gene Technology Co., Ltd., Shenzhen, 518071, China
- Shanghai Nucleus Biotechnology Co., Ltd., Shanghai, 201401, China
- Shenzhen Nucleus Huaxi Medical Laboratory, Shenzhen, 518110, China
| | - Dandan Li
- Shenzhen Nucleus Gene Technology Co., Ltd., Shenzhen, 518071, China
- Shanghai Nucleus Biotechnology Co., Ltd., Shanghai, 201401, China
- Shenzhen Nucleus Huaxi Medical Laboratory, Shenzhen, 518110, China
| | - Shenrui Zhang
- Shenzhen Nucleus Gene Technology Co., Ltd., Shenzhen, 518071, China
- Shanghai Nucleus Biotechnology Co., Ltd., Shanghai, 201401, China
- Shenzhen Nucleus Huaxi Medical Laboratory, Shenzhen, 518110, China
| | - Hezi Zhang
- Shenzhen Nucleus Gene Technology Co., Ltd., Shenzhen, 518071, China
- Shanghai Nucleus Biotechnology Co., Ltd., Shanghai, 201401, China
- Shenzhen Nucleus Huaxi Medical Laboratory, Shenzhen, 518110, China
| |
Collapse
|
44
|
Son W, Lee Y, Park Y, Park KS, Kim S, Youn H, Seo A, Sung B, Lee SH, Won J. Fc-competent TIGITx4-1BB bispecific antibody exerts potent long-lasting antitumor activity by potentiating CD8 + T cell activity and Fcγ receptor-mediated modulation of the tumor microenvironment. J Immunother Cancer 2025; 13:e010728. [PMID: 40010766 PMCID: PMC12083285 DOI: 10.1136/jitc-2024-010728] [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: 10/03/2024] [Accepted: 02/04/2025] [Indexed: 02/28/2025] Open
Abstract
BACKGROUND TIGIT was identified as a target immune checkpoint for overcoming resistance to PD-(L)1-blocking antibodies. However, the clinical efficacies of TIGIT antibodies were moderate in monotherapy and mixed in combination with PD-(L)1 antibodies. 4-1BB, a strong inducible costimulatory receptor, is another attractive target in antitumor therapeutics. This study investigated whether ABL112, an Fc-competent bispecific antibody targeting TIGIT and 4-1BB (TIGITx4-1BB), would enhance antitumor activity via Fcγ receptor (FcγR)-mediated macrophage activation and antibody-dependent cell-mediated functions. METHODS TIGIT-dependent 4-1BB activation and TIGIT-blocking activity were assessed using reporter Jurkat T cell lines expressing 4-1BB and TIGIT, respectively. In vivo antitumor activity was confirmed in h4-1BB knock-in mice. The main immune cell subsets associated with the antitumor activity of ABL112 were identified using antibodies for depleting specific immune cell subtypes or FcγR-blocking antibodies. The effects of a combined pembrolizumab or atezolizumab treatment with ABL112 were assessed in two mouse models with different genetic backgrounds. Statistical analysis was performed using one-way or two-way analysis of variance (ANOVA) with Dunnett's multiple-comparison test or one-way ANOVA with Fisher's multiple-comparison test. RESULTS ABL112 restored T cell activity by blocking TIGIT-CD155 interactions, based on a TIGIT blockade reporter assay. ABL112, an Fc-competent TIGITx4-1BB bispecific antibody, showed strong FcγRI-dependent 4-1BB activation along with TIGIT-dependent 4-1BB activation. In H22 tumor models expressing high levels of endogenous CD155, both ABL112 and parent TIGIT single-domain Ab showed potent tumor-suppressive activity; however, only ABL112 exerted long-lasting antitumor activity. ABL112 induced a marked decrease in Treg numbers, while augmenting the absolute number of CD8+ T cells and proportion of CD226+ CD8+ T cells. The expressions of CXCL10, CXCL11, IFN-γ, and TNF-α increased, indicating myeloid cell activation and potential modification of the tumor microenvironment to an inflammatory phenotype. ABL112 not only showed outstanding antitumor activity as a monotherapy, but also showed synergistic effects with PD-(L)1 mAb compared with the combined TIGIT-PD-(L)1 mAb treatments. CONCLUSIONS Through multiple mechanisms of action, ABL112 exerted potent tumor-killing activity and immune memory response alone or in combination with anti-PD-(L)1 therapies, representing a promising new cancer treatment strategy.
Collapse
Affiliation(s)
- Wonjun Son
- Oncology Discovery, ABL Bio Inc, Seongnam, Korea (the Republic of)
| | - Yangsoon Lee
- Oncology Discovery, ABL Bio Inc, Seongnam, Korea (the Republic of)
| | - Yelim Park
- Oncology Discovery, ABL Bio Inc, Seongnam, Korea (the Republic of)
| | - Kyeong-Su Park
- Oncology Discovery, ABL Bio Inc, Seongnam, Korea (the Republic of)
| | - Sora Kim
- Oncology Discovery, ABL Bio Inc, Seongnam, Korea (the Republic of)
| | - Hyunseong Youn
- Oncology Discovery, ABL Bio Inc, Seongnam, Korea (the Republic of)
| | - Arim Seo
- Quality Control, ABL Bio Inc, Seongnam, Korea (the Republic of)
| | - Byungje Sung
- Analytics, ABL Bio Inc, Seongnam, Korea (the Republic of)
| | | | - Jonghwa Won
- Oncology Discovery, ABL Bio Inc, Seongnam, Korea (the Republic of)
| |
Collapse
|
45
|
Gao R, Wu P, Yin X, Zhuang L, Meng X. Deep analysis of the trials and major challenges in the first-line treatment for patients with extensive-stage small cell lung cancer. Int Immunopharmacol 2025; 148:114116. [PMID: 39847950 DOI: 10.1016/j.intimp.2025.114116] [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: 05/08/2024] [Revised: 01/15/2025] [Accepted: 01/15/2025] [Indexed: 01/25/2025]
Abstract
The median overall survival (OS) is approximately 10 months when chemotherapy alone is the first-line treatment for extensive-stage small cell lung cancer (ES-SCLC). The approval of the two PD-L1 inhibitors, atezolizumab and durvalumab, marked the beginning of the immunotherapy era for ES-SCLC. Serplulimab, as the first PD-1 inhibitor to achieve success in the first-line treatment of ES-SCLC, has not only demonstrated significant improvements in patient survival outcomes but also ushered in a new era for PD-1 inhibitors in the treatment of ES-SCLC. Recently, antiangiogenic agents with chemo-immunotherapy have achieved breakthroughs in first-line ES-SCLC treatment. Improving the clinical benefits of individualized treatment for patients with ES-SCLC remains challenging. Challenges include identifying biomarkers for targeted therapy, exploring new treatments, developing new medicines, and classifying SCLC molecular subtypes. This review provides an in-depth analysis of research on first-line ES-SCLC treatment. Additionally, it discusses advances in ES-SCLC treatment.
Collapse
Affiliation(s)
- Ran Gao
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Shandong, Jinan, China
| | - Peizhu Wu
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Shandong, Jinan, China
| | - Xiaoyan Yin
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Shandong, Jinan, China
| | - Lulu Zhuang
- Cheeloo College of Cancer Center, Shandong University, Jinan, Shandong, China
| | - Xiangjiao Meng
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Shandong, Jinan, China.
| |
Collapse
|
46
|
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
|
47
|
Arandhara A, Bhuyan P, Das BK. Exploring lung cancer microenvironment: pathways and nanoparticle-based therapies. Discov Oncol 2025; 16:159. [PMID: 39934547 DOI: 10.1007/s12672-025-01902-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Accepted: 02/03/2025] [Indexed: 02/13/2025] Open
Abstract
Lung cancer stands out as a significant global health burden, with staggering incidence and mortality rates primarily linked to smoking and environmental carcinogens. The tumor microenvironment (TME) emerges as a critical determinant of cancer progression and treatment outcomes, comprising a complex interplay of cells, signaling molecules, and extracellular matrix. Through a comprehensive literature review, we elucidate current research trends and therapeutic prospects, aiming to advance our understanding of TME modulation strategies and their clinical implications for lung cancer treatment. Dysregulated immune responses within the TME can facilitate tumor evasion, limiting the efficacy of immune checkpoint inhibitors (ICI). Consequently, TME modulation strategies have become potential avenues to enhance therapeutic responses. However, conventional TME-targeted therapies often face challenges. In contrast, nanoparticle (NP)-based therapies offer promising prospects for improved drug delivery and reduced toxicity, leveraging the enhanced permeability and retention (EPR) effect. Despite NP design and delivery advancements, obstacles like poor tumor cell uptake and off-target effects persist, necessitating further optimization. This review underscores the pivotal role of TME in lung cancer management, emphasizing the synergistic potential of immunotherapy and nano-therapy.
Collapse
Affiliation(s)
- Arunabh Arandhara
- Assam Pharmacy Institute, Titabar, Amgurikhat, Jorhat, Assam, 785632, India
| | - Pallabi Bhuyan
- School of Pharmacy, The Assam Kaziranga University, Koraikhowa, Jorhat, Assam, 785006, India
| | - Bhrigu Kumar Das
- Department of Pharmacology, School of Pharmaceutical Sciences, Girijananda Chowdhury University, Azara, Guwahati, Assam, 781017, India.
| |
Collapse
|
48
|
Nedeljković M, Vuletić A, Mirjačić Martinović K. Divide and Conquer-Targeted Therapy for Triple-Negative Breast Cancer. Int J Mol Sci 2025; 26:1396. [PMID: 40003864 PMCID: PMC11855393 DOI: 10.3390/ijms26041396] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Revised: 01/31/2025] [Accepted: 02/04/2025] [Indexed: 02/27/2025] Open
Abstract
Triple-negative breast cancer (TNBC) is the most aggressive and malignant type of breast cancer with limited treatment options and poor prognosis. One of the most significant impediments in TNBC treatment is the high heterogeneity of this disease, as highlighted by the detection of several molecular subtypes of TNBC. Each subtype is driven by distinct mutations and pathway aberrations, giving rise to specific molecular characteristics closely connected to clinical behavior, outcomes, and drug sensitivity. This review summarizes the knowledge regarding TNBC molecular subtypes and how it can be harnessed to devise tailored treatment strategies instead of blindly using targeted drugs. We provide an overview of novel targeted agents and key insights about new treatment modalities with an emphasis on the androgen receptor signaling pathway, cancer stem cell-associated pathways, phosphatidylinositol 3-kinase (PI3K)/AKT pathway, growth factor signaling, and immunotherapy.
Collapse
Affiliation(s)
- Milica Nedeljković
- Department of Experimental Oncology, Institute for Oncology and Radiology of Serbia, 11000 Belgrade, Serbia; (A.V.); (K.M.M.)
| | | | | |
Collapse
|
49
|
Greene TT, Jo Y, Chiale C, Macal M, Fang Z, Khatri FS, Codrington AL, Kazane KR, Akbulut E, Swaminathan S, Fujita Y, Fitzgerald-Bocarsly P, Cordes T, Metallo C, Scott DA, Zúñiga EI. Metabolic deficiencies underlie reduced plasmacytoid dendritic cell IFN-I production following viral infection. Nat Commun 2025; 16:1460. [PMID: 39920132 PMCID: PMC11805920 DOI: 10.1038/s41467-025-56603-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Accepted: 01/23/2025] [Indexed: 02/09/2025] Open
Abstract
Type I Interferons (IFN-I) are central to host protection against viral infections, with plasmacytoid dendritic cells (pDC) being the most significant source, yet pDCs lose their IFN-I production capacity following an initial burst of IFN-I, resulting in susceptibility to secondary infections. The underlying mechanisms of these dynamics are not well understood. Here we find that viral infection reduces the capacity of pDCs to engage both oxidative and glycolytic metabolism. Mechanistically, we identify lactate dehydrogenase B (LDHB) as a positive regulator of pDC IFN-I production in mice and humans; meanwhile, LDHB deficiency is associated with suppressed IFN-I production, pDC metabolic capacity, and viral control following infection. In addition, preservation of LDHB expression is sufficient to partially retain the function of otherwise exhausted pDCs, both in vitro and in vivo. Furthermore, restoring LDHB in vivo in pDCs from infected mice increases IFNAR-dependent, infection-associated pathology. Our work thus identifies a mechanism for balancing immunity and pathology during viral infections, while also providing insight into the highly preserved infection-driven pDC inhibition.
Collapse
Affiliation(s)
- Trever T Greene
- Department of Biological Sciences, University of California, San Diego, La Jolla, CA, USA
| | - Yeara Jo
- Department of Biological Sciences, University of California, San Diego, La Jolla, CA, USA
| | - Carolina Chiale
- Department of Biological Sciences, University of California, San Diego, La Jolla, CA, USA
| | - Monica Macal
- Department of Biological Sciences, University of California, San Diego, La Jolla, CA, USA
| | - Ziyan Fang
- Department of Biological Sciences, University of California, San Diego, La Jolla, CA, USA
| | - Fawziyah S Khatri
- Department of Biological Sciences, University of California, San Diego, La Jolla, CA, USA
| | - Alicia L Codrington
- Department of Pathology, Immunology and Laboratory Medicine, Rutgers New Jersey Medical School, Newark, NJ, USA
| | - Katelynn R Kazane
- Department of Biological Sciences, University of California, San Diego, La Jolla, CA, USA
| | - Elizabeth Akbulut
- Department of Pathology, Immunology and Laboratory Medicine, Rutgers New Jersey Medical School, Newark, NJ, USA
| | - Shobha Swaminathan
- Department of Medicine, Division of Infectious Disease, The State University of New Jersey, Rutgers, New Jersey Medical School, Newark, NJ, USA
| | - Yu Fujita
- Division of Next-Generation Drug Development, Research Center for Medical Sciences, The Jikei University School of Medicine, Tokyo, Japan
| | | | - Thekla Cordes
- Department of Bioinformatics and Biochemistry, Braunschweig Integrated Centre of Systems Biology (BRICS), Technische Universität Braunschweig, Braunschweig, Germany
- Department of Bioengineering, University of California, San Diego, La Jolla, CA, USA
- Molecular and Cell Biology Laboratory, Salk Institute for Biological Sciences, La Jolla, CA, USA
| | - Christian Metallo
- Molecular and Cell Biology Laboratory, Salk Institute for Biological Sciences, La Jolla, CA, USA
| | - David A Scott
- Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
| | - Elina I Zúñiga
- Department of Biological Sciences, University of California, San Diego, La Jolla, CA, USA.
| |
Collapse
|
50
|
Li G, Li S, Jiang Y, Chen T, An Z. Unleashing the Power of immune Checkpoints: A new strategy for enhancing Treg cells depletion to boost antitumor immunity. Int Immunopharmacol 2025; 147:113952. [PMID: 39764997 DOI: 10.1016/j.intimp.2024.113952] [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: 05/26/2024] [Revised: 12/22/2024] [Accepted: 12/24/2024] [Indexed: 01/29/2025]
Abstract
Regulatory T (Treg) cells, immunosuppressive CD4+ T cells, can impede anti-tumor immunity, complicating cancer treatment. Since their discovery, numerous studies have been dedicated to understand Treg cell biology, with a focus on checkpoint pathways' role in their generation and function. Immune checkpoints, such as PD-1/PD-L1, CTLA-4, TIGIT, TIM-3, and OX40, are pivotal in controlling Treg cell expansion and activity in the tumor microenvironment (TME), affecting their ability to suppress immune responses. This review examines the complex relationship between these checkpoints and Tregs in the TME, and how they influence tumor immunity. We also discuss the therapeutic potential of targeting these checkpoints to enhance anti-tumor immunity, including the use of immune checkpoint blockade (ICB) therapies and novel approaches such as CCR8-targeted therapies. Understanding the interaction between immune checkpoints and Treg cells can lead to more effective immunotherapeutic strategies, such as combining CCR8-targeted therapies with immune checkpoint inhibitors, to improve patient outcomes in cancer treatment.
Collapse
Affiliation(s)
- Guoxin Li
- Department of Oral Biology, School and Hospital of Stomatology, Jilin University, Changchun, China; Key Laboratory of Tooth Development and Bone Remodeling of Jilin Province, School and Hospital of Stomatology, Jilin University, Changchun, China
| | - Siqi Li
- Department of Oral Biology, School and Hospital of Stomatology, Jilin University, Changchun, China
| | - Yilin Jiang
- Department of Oral Biology, School and Hospital of Stomatology, Jilin University, Changchun, China
| | - Tao Chen
- Department of Oral Biology, School and Hospital of Stomatology, Jilin University, Changchun, China
| | - Zhengwen An
- Department of Oral Biology, School and Hospital of Stomatology, Jilin University, Changchun, China; Key Laboratory of Tooth Development and Bone Remodeling of Jilin Province, School and Hospital of Stomatology, Jilin University, Changchun, China.
| |
Collapse
|