1
|
Leon G, Klavina PA, Rehill AM, Cooper SEJ, Dominik A, Basavarajappa SC, O'Donnell JS, Hussey S, Walsh PT, Preston RJS. Tissue factor-dependent colitogenic CD4 + T cell thrombogenicity is regulated by activated protein C signalling. Nat Commun 2025; 16:1677. [PMID: 39956825 PMCID: PMC11830781 DOI: 10.1038/s41467-025-57001-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Accepted: 02/07/2025] [Indexed: 02/18/2025] Open
Abstract
Patients with inflammatory bowel disease (IBD) have an increased risk of venous thromboembolism (VTE), but the underlying mechanistic basis remains poorly defined. Here, we find that colitogenic CD4+ T cells express tissue factor (TF) and promote rapid TF-dependent plasma thrombin generation. TF+CD3+CD4+ T cells are present in both the colons of mice with experimental colitis and blood and colonic tissue from patients with IBD. Expression of genes involved in regulating coagulation, including Protein C (PC; encoded by PROC) and its receptor (PROCR), are dysregulated in IBD patient gut biopsy tissues. Moreover, activated PC signalling reduces the procoagulant activity mediated by TF+CD4+ T cells. Our data thus identify TF-induced, colitogenic T cell-mediated thrombogenicity, and also demonstrate a new function for activated PC signalling in regulating T cell thrombo-inflammatory activity.
Collapse
Affiliation(s)
- Gemma Leon
- Irish Centre for Vascular Biology, School of Pharmacy and Biomolecular Sciences, RCSI University of Medicine and Health Sciences, Dublin, Ireland
| | - Paula A Klavina
- Irish Centre for Vascular Biology, School of Pharmacy and Biomolecular Sciences, RCSI University of Medicine and Health Sciences, Dublin, Ireland
| | - Aisling M Rehill
- Irish Centre for Vascular Biology, School of Pharmacy and Biomolecular Sciences, RCSI University of Medicine and Health Sciences, Dublin, Ireland
| | - Sarah E J Cooper
- National Centre for Paediatric Gastroenterology, CHI-Crumlin, Dublin, Ireland
| | - Anna Dominik
- National Centre for Paediatric Gastroenterology, CHI-Crumlin, Dublin, Ireland
| | | | - James S O'Donnell
- Irish Centre for Vascular Biology, School of Pharmacy and Biomolecular Sciences, RCSI University of Medicine and Health Sciences, Dublin, Ireland
| | - Seamus Hussey
- National Centre for Paediatric Gastroenterology, CHI-Crumlin, Dublin, Ireland
| | - Patrick T Walsh
- Department of Clinical Medicine, Trinity Translational Medicine Institute, Trinity College Dublin, Dublin, Ireland
| | - Roger J S Preston
- Irish Centre for Vascular Biology, School of Pharmacy and Biomolecular Sciences, RCSI University of Medicine and Health Sciences, Dublin, Ireland.
| |
Collapse
|
2
|
Li S, Pan Y, Ye R, Wang Y, Li L. Immune checkpoints in B-cell Lymphoma: Still an Unmet challenge from Basic research to clinical practice. Int Immunopharmacol 2025; 146:113717. [PMID: 39673995 DOI: 10.1016/j.intimp.2024.113717] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2024] [Revised: 11/24/2024] [Accepted: 11/24/2024] [Indexed: 12/16/2024]
Abstract
In the last decade, advancements in immunotherapy knowledge have highlighted CTLA-4, PD-1, LAG-3, TIM-3, and TIGIT, decisive immune checkpoints exhibiting within the tumor microenvironment (TME), as fundamental objects for cancer immunotherapy. The widespread clinical use of immune checkpoint inhibitors (ICls), employing PD-1/PD-L1 or CTLA-4 antibodies to obstruct crucial checkpoint regulators, is noted in treating B-cell lymphoma patients. Nevertheless, the prolonged advantages of the currently employed treatments against CTLA-4, PD-1, and PD-L1 are uncommon among patients. Thus, recent focus has been progressively moved to additional immune checkpoints on T cells, like LAG-3, TIM-3, and TIGIT, which are now seen as reassuring targets for treatment and broadly acknowledged. There are several types of immunecheckpoint molecules expressed by T cells, and inhibitors targeting immune checkpoints can revive and amplify the immune response of T lymphocytes against tumors, a crucial aspect in lymphoma therapy. However, there is little knowledge about their regulation. Herein, we discuss the anti-tumor effects and functions of ICIs in controlling T-cell activity, as well as the progress in combined application with other immunotherapies.
Collapse
Affiliation(s)
- Sijia Li
- Department of Hematology, The Second Hospital of Dalian Medical University, Dalian, PR China
| | - Yuanyuan Pan
- Department of Hematology, The Second Hospital of Dalian Medical University, Dalian, PR China
| | - Ruyu Ye
- Department of Hematology, The Second Hospital of Dalian Medical University, Dalian, PR China
| | - Yu Wang
- Department of Hematology, The Second Hospital of Dalian Medical University, Dalian, PR China
| | - Li Li
- Department of Hematology, The Second Hospital of Dalian Medical University, Dalian, PR China.
| |
Collapse
|
3
|
Zamani MR, Šácha P. TIM3 in COVID-19; A potential hallmark? Heliyon 2024; 10:e40386. [PMID: 39759854 PMCID: PMC11700678 DOI: 10.1016/j.heliyon.2024.e40386] [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: 07/25/2024] [Revised: 11/11/2024] [Accepted: 11/12/2024] [Indexed: 01/07/2025] Open
Abstract
Coronavirus disease 2019 (COVID-19) is a highly contagious viral disease, caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). It can manifest as mild to severe flu-like and non-flu-like symptoms and signs, which are associated with immune dysfunction and increased mortality. The findings from COVID-19 patients imply a link between immune system abnormalities such as impaired T-cell responses or cytokine imbalances and increased risk for worse clinical outcomes, which has not been fully understood. Owing to the regulatory role of inhibitory immune checkpoints during COVID-19 infection, this review summarizes the available studies concerning the TIM3 as a relatively less characterized immune checkpoint in COVID-19 patients.
Collapse
Affiliation(s)
- Mohammad Reza Zamani
- Department of Cell Biology, Faculty of Science, Charles University, Prague, Czech Republic
- Institute of Organic Chemistry and Biochemistry, Czech Academy of Sciences, Prague, Czech Republic
| | - Pavel Šácha
- Institute of Organic Chemistry and Biochemistry, Czech Academy of Sciences, Prague, Czech Republic
| |
Collapse
|
4
|
Nieves-Rosado HM, Banerjee H, Gocher-Demske A, Manandhar P, Mehta I, Ezenwa O, Xie B, Murter B, Das J, Vignali DAA, Delgoffe GM, Kane LP. Tim-3 Is Required for Regulatory T Cell-Mediated Promotion of T Cell Exhaustion and Viral Persistence during Chronic Lymphocytic Choriomeningitis Virus Infection. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2024; 213:1488-1498. [PMID: 39345172 DOI: 10.4049/jimmunol.2400119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Accepted: 09/07/2024] [Indexed: 10/01/2024]
Abstract
Expression of T cell Ig and mucin domain-containing protein 3 (Tim-3) is upregulated on regulatory T cells (Tregs) during chronic viral infections. In several murine and human chronic infections, the expression of Tim-3 is associated with poor control of viral burden and impaired antiviral immune responses. However, the role of Tim-3+ Tregs during persistent viral infections has not been fully defined. We employed an inducible Treg-specific Tim-3 loss-of-function (Tim-3 Treg knockout) murine model to dissect the role of Tim-3 on Tregs during chronic lymphocytic choriomeningitis virus infection. Tim-3 Treg knockout mice exhibited a decrease in morbidity, a more potent virus-specific T cell response, and a significant decrease in viral burden. These mice also had a reduction in the frequency of PD-1+Tim-3+ and PD-1+Tox+ gp33-specific exhausted CD8+ T cells. Our findings demonstrate that modulation of a single surface protein on Tregs can lead to a reduction in viral burden, limit T cell exhaustion, and enhance gp33-specific T cell response. These studies may help to identify Tim-3-directed therapies for the management of persistent infections and cancer.
Collapse
Affiliation(s)
- Hector M Nieves-Rosado
- Department of Immunology, University of Pittsburgh, Pittsburgh, PA
- Medical Scientist Training Program, University of Pittsburgh and Carnegie Mellon University, Pittsburgh, PA
- Graduate Program in Microbiology and Immunology, University of Pittsburgh, Pittsburgh, PA
| | - Hridesh Banerjee
- Department of Immunology, University of Pittsburgh, Pittsburgh, PA
| | | | - Priyanka Manandhar
- Department of Immunology, University of Pittsburgh, Pittsburgh, PA
- Graduate Program in Microbiology and Immunology, University of Pittsburgh, Pittsburgh, PA
| | - Isha Mehta
- Department of Immunology, University of Pittsburgh, Pittsburgh, PA
- Center for Systems Immunology, University of Pittsburgh, Pittsburgh, PA
| | - Ogechukwu Ezenwa
- Department of Immunology, University of Pittsburgh, Pittsburgh, PA
- Center for Systems Immunology, University of Pittsburgh, Pittsburgh, PA
| | - Bingxian Xie
- Department of Immunology, University of Pittsburgh, Pittsburgh, PA
- Tumor Microenvironment Center, UPMC Hillman Cancer Center, Pittsburgh, PA
| | - Ben Murter
- Department of Immunology, University of Pittsburgh, Pittsburgh, PA
- Graduate Program in Microbiology and Immunology, University of Pittsburgh, Pittsburgh, PA
| | - Jishnu Das
- Department of Immunology, University of Pittsburgh, Pittsburgh, PA
- Center for Systems Immunology, University of Pittsburgh, Pittsburgh, PA
| | | | - Greg M Delgoffe
- Department of Immunology, University of Pittsburgh, Pittsburgh, PA
- Tumor Microenvironment Center, UPMC Hillman Cancer Center, Pittsburgh, PA
| | - Lawrence P Kane
- Department of Immunology, University of Pittsburgh, Pittsburgh, PA
| |
Collapse
|
5
|
Mitchell RA, Ubillos I, Requena P, Campo JJ, Ome-Kaius M, Hanieh S, Umbers A, Samol P, Barrios D, Jiménez A, Bardají A, Mueller I, Menéndez C, Rogerson S, Dobaño C, Moncunill G. Chronic malaria exposure is associated with inhibitory markers on T cells that correlate with atypical memory and marginal zone-like B cells. Clin Exp Immunol 2024; 216:172-191. [PMID: 38387476 PMCID: PMC11036110 DOI: 10.1093/cei/uxae015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Revised: 12/05/2023] [Accepted: 02/22/2024] [Indexed: 02/24/2024] Open
Abstract
Chronic immune activation from persistent malaria infections can induce immunophenotypic changes associated with T-cell exhaustion. However, associations between T and B cells during chronic exposure remain undefined. We analyzed peripheral blood mononuclear cells from malaria-exposed pregnant women from Papua New Guinea and Spanish malaria-naïve individuals using flow cytometry to profile T-cell exhaustion markers phenotypically. T-cell lineage (CD3, CD4, and CD8), inhibitory (PD1, TIM3, LAG3, CTLA4, and 2B4), and senescence (CD28-) markers were assessed. Dimensionality reduction methods revealed increased PD1, TIM3, and LAG3 expression in malaria-exposed individuals. Manual gating confirmed significantly higher frequencies of PD1+CD4+ and CD4+, CD8+, and double-negative (DN) T cells expressing TIM3 in malaria-exposed individuals. Increased frequencies of T cells co-expressing multiple markers were also found in malaria-exposed individuals. T-cell data were analyzed with B-cell populations from a previous study where we reported an alteration of B-cell subsets, including increased frequencies of atypical memory B cells (aMBC) and reduction in marginal zone (MZ-like) B cells during malaria exposure. Frequencies of aMBC subsets and MZ-like B cells expressing CD95+ had significant positive correlations with CD28+PD1+TIM3+CD4+ and DN T cells and CD28+TIM3+2B4+CD8+ T cells. Frequencies of aMBC, known to associate with malaria anemia, were inversely correlated with hemoglobin levels in malaria-exposed women. Similarly, inverse correlations with hemoglobin levels were found for TIM3+CD8+ and CD28+PD1+TIM3+CD4+ T cells. Our findings provide further insights into the effects of chronic malaria exposure on circulating B- and T-cell populations, which could impact immunity and responses to vaccination.
Collapse
Affiliation(s)
- Robert A Mitchell
- ISGlobal, Hospital Clínic, Universitat de Barcelona, Barcelona, Spain
| | - Itziar Ubillos
- ISGlobal, Hospital Clínic, Universitat de Barcelona, Barcelona, Spain
| | - Pilar Requena
- ISGlobal, Hospital Clínic, Universitat de Barcelona, Barcelona, Spain
- Facultad de Medicina, Universidad de Granada, Granada, Spain
| | - Joseph J Campo
- ISGlobal, Hospital Clínic, Universitat de Barcelona, Barcelona, Spain
- Antigen Discovery Inc., Irvine, CA, USA
| | - Maria Ome-Kaius
- Papua New Guinea Institute of Medical Research, Madang, Papua New Guinea
| | - Sarah Hanieh
- University of Melbourne, Melbourne, VIC, Australia
| | - Alexandra Umbers
- Papua New Guinea Institute of Medical Research, Madang, Papua New Guinea
| | - Paula Samol
- Papua New Guinea Institute of Medical Research, Madang, Papua New Guinea
| | - Diana Barrios
- ISGlobal, Hospital Clínic, Universitat de Barcelona, Barcelona, Spain
| | - Alfons Jiménez
- ISGlobal, Hospital Clínic, Universitat de Barcelona, Barcelona, Spain
- Consorcio de Investigación Biomédica en Red de Epidemiología y Salud Pública (CIBERESP), Barcelona, Spain
| | - Azucena Bardají
- ISGlobal, Hospital Clínic, Universitat de Barcelona, Barcelona, Spain
- Consorcio de Investigación Biomédica en Red de Epidemiología y Salud Pública (CIBERESP), Barcelona, Spain
- Centro de Investigação em Saúde de Manhiça (CISM), Maputo, Mozambique
| | - Ivo Mueller
- Papua New Guinea Institute of Medical Research, Madang, Papua New Guinea
- Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
| | - Clara Menéndez
- ISGlobal, Hospital Clínic, Universitat de Barcelona, Barcelona, Spain
- Consorcio de Investigación Biomédica en Red de Epidemiología y Salud Pública (CIBERESP), Barcelona, Spain
- Centro de Investigação em Saúde de Manhiça (CISM), Maputo, Mozambique
| | | | - Carlota Dobaño
- ISGlobal, Hospital Clínic, Universitat de Barcelona, Barcelona, Spain
- CIBER de Enfermedades Infecciosas (CIBERINFEC), Barcelona, Spain
| | - Gemma Moncunill
- ISGlobal, Hospital Clínic, Universitat de Barcelona, Barcelona, Spain
- CIBER de Enfermedades Infecciosas (CIBERINFEC), Barcelona, Spain
| |
Collapse
|
6
|
Kristenson L, Badami C, Ljungberg A, Islamagic E, Tian Y, Xie G, Hussein BA, Pesce S, Tang KW, Thorén FB. Deletion of the TMEM30A gene enables leukemic cell evasion of NK cell cytotoxicity. Proc Natl Acad Sci U S A 2024; 121:e2316447121. [PMID: 38557174 PMCID: PMC11009675 DOI: 10.1073/pnas.2316447121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Accepted: 02/15/2024] [Indexed: 04/04/2024] Open
Abstract
Natural killer (NK) cell immunotherapy has gained attention as a promising strategy for treatment of various malignancies. In this study, we used a genome-wide CRISPR screen to identify genes that provide protection or susceptibility to NK cell cytotoxicity. The screen confirmed the role of several genes in NK cell regulation, such as genes involved in interferon-γ signaling and antigen presentation, as well as genes encoding the NK cell receptor ligands B7-H6 and CD58. Notably, the gene TMEM30A, encoding CDC50A-beta-subunit of the flippase shuttling phospholipids in the plasma membrane, emerged as crucial for NK cell killing. Accordingly, a broad range of TMEM30A knock-out (KO) leukemia and lymphoma cells displayed increased surface levels of phosphatidylserine (PtdSer). TMEM30A KO cells triggered less NK cell degranulation, cytokine production and displayed lower susceptibility to NK cell cytotoxicity. Blockade of PtdSer or the inhibitory receptor TIM-3, restored the NK cell ability to eliminate TMEM30A-mutated cells. The key role of the TIM-3 - PtdSer interaction for NK cell regulation was further substantiated by disruption of the receptor gene in primary NK cells, which significantly reduced the impact of elevated PtdSer in TMEM30A KO leukemic cells. Our study underscores the potential significance of agents targeting the interaction between PtdSer and TIM-3 in the realm of cancer immunotherapy.
Collapse
Affiliation(s)
- Linnea Kristenson
- Tumor Immunology (TIMM) Laboratory at Sahlgrenska Center for Cancer Research, University of Gothenburg, Gothenburg413 90, Sweden
- Department of Medical Biochemistry and Cell Biology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg413 90, Sweden
| | - Chiara Badami
- Tumor Immunology (TIMM) Laboratory at Sahlgrenska Center for Cancer Research, University of Gothenburg, Gothenburg413 90, Sweden
- Department of Medical Biochemistry and Cell Biology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg413 90, Sweden
| | - Angelica Ljungberg
- Tumor Immunology (TIMM) Laboratory at Sahlgrenska Center for Cancer Research, University of Gothenburg, Gothenburg413 90, Sweden
| | - Erna Islamagic
- Tumor Immunology (TIMM) Laboratory at Sahlgrenska Center for Cancer Research, University of Gothenburg, Gothenburg413 90, Sweden
- Department of Medical Biochemistry and Cell Biology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg413 90, Sweden
| | - Yarong Tian
- Tumor Immunology (TIMM) Laboratory at Sahlgrenska Center for Cancer Research, University of Gothenburg, Gothenburg413 90, Sweden
- Department of Infectious Diseases, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg413 46, Sweden
| | - Guojiang Xie
- Tumor Immunology (TIMM) Laboratory at Sahlgrenska Center for Cancer Research, University of Gothenburg, Gothenburg413 90, Sweden
- Department of Infectious Diseases, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg413 46, Sweden
| | - Brwa Ali Hussein
- Tumor Immunology (TIMM) Laboratory at Sahlgrenska Center for Cancer Research, University of Gothenburg, Gothenburg413 90, Sweden
- Department of Medical Biochemistry and Cell Biology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg413 90, Sweden
| | - Silvia Pesce
- Tumor Immunology (TIMM) Laboratory at Sahlgrenska Center for Cancer Research, University of Gothenburg, Gothenburg413 90, Sweden
- Dipartimento di Medicina Sperimentale, Università di Genova, Genoa16132, Italy
| | - Ka-Wei Tang
- Tumor Immunology (TIMM) Laboratory at Sahlgrenska Center for Cancer Research, University of Gothenburg, Gothenburg413 90, Sweden
- Department of Infectious Diseases, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg413 46, Sweden
- Department of Clinical Microbiology, Sahlgrenska University Hospital, Gothenburg413 46, Sweden
- Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, Gothenburg413 90, Sweden
| | - Fredrik B. Thorén
- Tumor Immunology (TIMM) Laboratory at Sahlgrenska Center for Cancer Research, University of Gothenburg, Gothenburg413 90, Sweden
- Department of Medical Biochemistry and Cell Biology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg413 90, Sweden
| |
Collapse
|
7
|
Manandhar P, Szymczak-Workman AL, Kane LP. Tim-3 Is Not Required for Establishment of CD8+ T Cell Memory to Lymphocytic Choriomeningitis Virus. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2024; 212:466-474. [PMID: 38108417 PMCID: PMC10906969 DOI: 10.4049/jimmunol.2300401] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Accepted: 11/21/2023] [Indexed: 12/19/2023]
Abstract
Tim-3 is a transmembrane protein that is best known for being highly expressed on terminally exhausted CD8+ T cells associated with chronic infection and tumors, although its expression is not limited to those settings. Tim-3 is also expressed by CD8+ T cells during acute infection and by multiple other immune cell types, including CD4+ Th1 and regulatory T cells, dendritic cells, and mast cells. In this study, we investigated the role of Tim-3 signaling on CD8+ T cell memory using a Tim-3 conditional knockout mouse model and mice lacking the signaling portion of the Tim-3 cytoplasmic domain. Together, our results indicate that Tim-3 has at most a modest effect on the formation and function of CD8+ memory T cells.
Collapse
Affiliation(s)
- Priyanka Manandhar
- Dept. of Immunology, University of Pittsburgh, Pittsburgh, PA 15213
- Graduate Program in Microbiology and Immunology, University of Pittsburgh, Pittsburgh, PA 15213
| | | | - Lawrence P. Kane
- Dept. of Immunology, University of Pittsburgh, Pittsburgh, PA 15213
- Graduate Program in Microbiology and Immunology, University of Pittsburgh, Pittsburgh, PA 15213
| |
Collapse
|
8
|
Chakraborty B, Agarwal S, Kori S, Das R, Kashaw V, Iyer AK, Kashaw SK. Multiple Protein Biomarkers and Different Treatment Strategies for Colorectal Carcinoma: A Comprehensive Prospective. Curr Med Chem 2024; 31:3286-3326. [PMID: 37151060 DOI: 10.2174/0929867330666230505165031] [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: 11/12/2022] [Revised: 02/20/2023] [Accepted: 02/24/2023] [Indexed: 05/09/2023]
Abstract
In this review, we emphasized important biomarkers, pathogenesis, and newly developed therapeutic approaches in the treatment of colorectal cancer (CRC). This includes a complete description of small-molecule inhibitors, phytopharmaceuticals with antiproliferative potential, monoclonal antibodies for targeted therapy, vaccinations as immunotherapeutic agents, and many innovative strategies to intervene in the interaction of oncogenic proteins. Many factors combine to determine the clinical behavior of colorectal cancer and it is still difficult to comprehend the molecular causes of a person's vulnerability to CRC. It is also challenging to identify the causes of the tumor's onset, progression, and responsiveness or resistance to antitumor treatment. Current recommendations for targeted medications are being updated by guidelines throughout the world in light of the growing number of high-quality clinical studies. So, being concerned about the aforementioned aspects, we have tried to present a summarized pathogenic view, including a brief description of biomarkers and an update of compounds with their underlying mechanisms that are currently under various stages of clinical testing. This will help to identify gaps or shortfalls that can be addressed in upcoming colorectal cancer research.
Collapse
Affiliation(s)
- Biswadip Chakraborty
- Integrated Drug Discovery Research Laboratory, Department of Pharmaceutical Sciences, Dr. Harisingh Gour University (A Central University), Sagar (MP), India
| | - Shivangi Agarwal
- Integrated Drug Discovery Research Laboratory, Department of Pharmaceutical Sciences, Dr. Harisingh Gour University (A Central University), Sagar (MP), India
| | - Shivam Kori
- Integrated Drug Discovery Research Laboratory, Department of Pharmaceutical Sciences, Dr. Harisingh Gour University (A Central University), Sagar (MP), India
| | - Ratnesh Das
- Department of Chemistry, ISF College of Pharmacy, Moga-Punjab, India
| | - Varsha Kashaw
- Sagar Institute of Pharmaceutical Sciences, Sagar (M.P.), India
| | - Arun K Iyer
- Use-inspired Biomaterials & Integrated Nano Delivery (U-BiND) Systems Laboratory, Department of Pharmaceutical Sciences, Wayne State University, Detroit, Michigan, USA
- Molecular Imaging Program, Karmanos Cancer Institute, Detroit, Michigan, USA
| | - Sushil Kumar Kashaw
- Integrated Drug Discovery Research Laboratory, Department of Pharmaceutical Sciences, Dr. Harisingh Gour University (A Central University), Sagar (MP), India
| |
Collapse
|
9
|
Lameris R, Shahine A, Veth M, Westerman B, Godfrey DI, Lutje Hulsik D, Brouwer P, Rossjohn J, de Gruijl TD, van der Vliet HJ. Enhanced CD1d phosphatidylserine presentation using a single-domain antibody promotes immunomodulatory CD1d-TIM-3 interactions. J Immunother Cancer 2023; 11:e007631. [PMID: 38040419 PMCID: PMC10693867 DOI: 10.1136/jitc-2023-007631] [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] [Accepted: 11/05/2023] [Indexed: 12/03/2023] Open
Abstract
BACKGROUND CD1d is a monomorphic major histocompatibility complex class I-like molecule that presents lipid antigens to distinct T-cell subsets and can be expressed by various malignancies. Antibody-mediated targeting of CD1d on multiple myeloma cells was reported to induce apoptosis and could therefore constitute a novel therapeutic approach. METHODS To determine how a CD1d-specific single-domain antibody (VHH) enhances binding of the early apoptosis marker annexin V to CD1d+ tumor cells we use in vitro cell-based assays and CRISPR-Cas9-mediated gene editing, and to determine the structure of the VHH1D17-CD1d(endogenous lipid) complex we use X-ray crystallography. RESULTS Anti-CD1d VHH1D17 strongly enhances annexin V binding to CD1d+ tumor cells but this does not reflect induction of apoptosis. Instead, we show that VHH1D17 enhances presentation of phosphatidylserine (PS) in CD1d and that this is saposin dependent. The crystal structure of the VHH1D17-CD1d(endogenous lipid) complex demonstrates that VHH1D17 binds the A'-pocket of CD1d, leaving the lipid headgroup solvent exposed, and has an electro-negatively charged patch which could be involved in the enhanced PS presentation by CD1d. Presentation of PS in CD1d does not trigger phagocytosis but leads to greatly enhanced binding of T-cell immunoglobulin and mucin domain containing molecules (TIM)-1 to TIM-3, TIM-4 and induces TIM-3 signaling. CONCLUSION Our findings reveal the existence of an immune modulatory CD1d(PS)-TIM axis with potentially unexpected implications for immune regulation in both physiological and pathological conditions.
Collapse
Affiliation(s)
- Roeland Lameris
- Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam UMC Location VUmc, Amsterdam, The Netherlands
| | - Adam Shahine
- Infection and Immunity Program and Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia
| | - Myrthe Veth
- Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam UMC Location VUmc, Amsterdam, The Netherlands
| | - Bart Westerman
- Department of Neurosurgery, Cancer Center Amsterdam, Amsterdam UMC Location VUmc, Amsterdam, The Netherlands
| | - Dale I Godfrey
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Melbourne, Victoria, Australia
| | | | | | - Jamie Rossjohn
- Infection and Immunity Program and Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia
- Institute of Infection and Immunity, Cardiff University School of Medicine, Cardiff, UK
| | - Tanja D de Gruijl
- Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam UMC Location VUmc, Amsterdam, The Netherlands
| | - Hans J van der Vliet
- Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam UMC Location VUmc, Amsterdam, The Netherlands
- LAVA Therapeutics, Utrecht, The Netherlands
| |
Collapse
|
10
|
Xue J, Suo L, An Y, Wang X, Zhang S, Liu H, Wu Y, Sun X, Zhao C, Yang P. Phosphatidylserine promotes immunotherapy for airway allergy. Immunol Lett 2023; 264:46-55. [PMID: 38008186 DOI: 10.1016/j.imlet.2023.11.006] [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: 06/02/2023] [Revised: 11/06/2023] [Accepted: 11/23/2023] [Indexed: 11/28/2023]
Abstract
Type 1 regulatory T cells (Tr1 cells) play an important role in the maintenance of the immune homeostasis in the body. The induction of Tr1 cell is to be further investigated. The interaction of phosphatidylserine (PS) with TIM3 has immune regulation functions. The objective of this study is to elucidate the role of PS-TIM3 signals in inducing Tr1 cells. In this study, mice were treated using PS or specific immunotherapy by nasal instillation. A murine model of allergic rhinitis was developed using ovalbumin as a specific antigen. We found that PS-containing nasal instillation induced Tr1 cells in the airway tissues. PS promoted gene activities associated with IL-10 through activation of TIM3 in CD4+ T cells. TIM3 mediated the effects of PS on inducing Tr1 cells, in which the TIM3- PI3K-AKT pathway played a critical role. PS boosted allergen-specific immunotherapy by inducing specific antigen Tr1 cell generation. Concomitant administration of PS and SIT resulted in better therapeutic effects on AR. In conclusion, the data demonstrate that PS can promote the specific immunotherapy for AR through inducing antigen specific Tr1 cells in the airway tissues.
Collapse
Affiliation(s)
- Jinmei Xue
- Department of Otolaryngology, Head & Neck Surgery, Second Hospital, Shanxi Medical University, Taiyuan, China
| | - Limin Suo
- Department of Otolaryngology, Head & Neck Surgery, Second Hospital, Shanxi Medical University, Taiyuan, China
| | - Yunfang An
- Department of Otolaryngology, Head & Neck Surgery, Second Hospital, Shanxi Medical University, Taiyuan, China
| | - Xinxin Wang
- Guangdong Provincial Regional Disease Key Laboratory, Shenzhen, China; Institute of Allergy & Immunology, Shenzhen University School of Medicine, State Key Laboratory of Respiratory Diseases Allergy Division at Shenzhen University, Shenzhen, China
| | - Shuang Zhang
- Guangdong Provincial Regional Disease Key Laboratory, Shenzhen, China; Institute of Allergy & Immunology, Shenzhen University School of Medicine, State Key Laboratory of Respiratory Diseases Allergy Division at Shenzhen University, Shenzhen, China
| | - Huazhen Liu
- Guangdong Provincial Regional Disease Key Laboratory, Shenzhen, China; Institute of Allergy & Immunology, Shenzhen University School of Medicine, State Key Laboratory of Respiratory Diseases Allergy Division at Shenzhen University, Shenzhen, China
| | - Yongjin Wu
- Department of General Practical Medicine, Third Affiliated Hospital, Shenzhen, China
| | - Xizhuo Sun
- Department of General Practical Medicine, Third Affiliated Hospital, Shenzhen, China
| | - Changqing Zhao
- Department of Otolaryngology, Head & Neck Surgery, Second Hospital, Shanxi Medical University, Taiyuan, China.
| | - Pingchang Yang
- Guangdong Provincial Regional Disease Key Laboratory, Shenzhen, China; Institute of Allergy & Immunology, Shenzhen University School of Medicine, State Key Laboratory of Respiratory Diseases Allergy Division at Shenzhen University, Shenzhen, China; Department of General Practical Medicine, Third Affiliated Hospital, Shenzhen, China.
| |
Collapse
|
11
|
Sauer N, Janicka N, Szlasa W, Skinderowicz B, Kołodzińska K, Dwernicka W, Oślizło M, Kulbacka J, Novickij V, Karłowicz-Bodalska K. TIM-3 as a promising target for cancer immunotherapy in a wide range of tumors. Cancer Immunol Immunother 2023; 72:3405-3425. [PMID: 37567938 PMCID: PMC10576709 DOI: 10.1007/s00262-023-03516-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Accepted: 08/02/2023] [Indexed: 08/13/2023]
Abstract
T-cell immunoglobulin and mucin domain-containing protein 3 (TIM-3) expression has been a trending topic in recent years due to its differential expression in a wide range of neoplasms. TIM-3 is one of the key immune checkpoint receptors that interact with GAL-9, PtdSer, HMGB1 and CEACAM1. Initially identified on the surface of T helper 1 (Th1) lymphocytes and later on cytotoxic lymphocytes (CTLs), monocytes, macrophages, natural killer cells (NKs), and dendritic cells (DCs), TIM-3 plays a key role in immunoregulation. Recently, a growing body of evidence has shown that its differential expression in various tumor types indicates a specific prognosis for cancer patients. Here, we discuss which types of cancer TIM-3 can serve as a prognostic factor and the influence of coexpressed immune checkpoint inhibitors, such as LAG-3, PD-1, and CTLA-4 on patients' outcomes. Currently, experimental medicine involving TIM-3 has significantly enhanced the anti-tumor effect and improved patient survival. In this work, we summarized clinical trials incorporating TIM-3 targeting monoclonal and bispecific antibodies in monotherapy and combination therapy and highlighted the emerging role of cell-based therapies.
Collapse
Affiliation(s)
- Natalia Sauer
- Faculty of Pharmacy, Wroclaw Medical University, Wroclaw, Poland
| | - Natalia Janicka
- Faculty of Pharmacy, Wroclaw Medical University, Wroclaw, Poland
| | - Wojciech Szlasa
- Faculty of Medicine, Wroclaw Medical University, Wroclaw, Poland
| | | | | | - Wioletta Dwernicka
- Faculty of Medicine, Poznan University of Medical Sciences, Poznan, Poland
| | | | - Julita Kulbacka
- State Research Institute Centre for Innovative Medicine, Department of Immunology, Vilnius, Lithuania.
- Department of Molecular and Cellular Biology, Faculty of Pharmacy, Wroclaw Medical University, Wroclaw, Poland.
| | - Vitalij Novickij
- State Research Institute Centre for Innovative Medicine, Department of Immunology, Vilnius, Lithuania
- Faculty of Electronics, Vilnius Gediminas Technical University, Vilnius, Lithuania
| | | |
Collapse
|
12
|
Chongsaritsinsuk J, Steigmeyer AD, Mahoney KE, Rosenfeld MA, Lucas TM, Smith CM, Li A, Ince D, Kearns FL, Battison AS, Hollenhorst MA, Judy Shon D, Tiemeyer KH, Attah V, Kwon C, Bertozzi CR, Ferracane MJ, Lemmon MA, Amaro RE, Malaker SA. Glycoproteomic landscape and structural dynamics of TIM family immune checkpoints enabled by mucinase SmE. Nat Commun 2023; 14:6169. [PMID: 37794035 PMCID: PMC10550946 DOI: 10.1038/s41467-023-41756-y] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2023] [Accepted: 09/11/2023] [Indexed: 10/06/2023] Open
Abstract
Mucin-domain glycoproteins are densely O-glycosylated and play critical roles in a host of biological functions. In particular, the T cell immunoglobulin and mucin-domain containing family of proteins (TIM-1, -3, -4) decorate immune cells and act as key regulators in cellular immunity. However, their dense O-glycosylation remains enigmatic, primarily due to the challenges associated with studying mucin domains. Here, we demonstrate that the mucinase SmE has a unique ability to cleave at residues bearing very complex glycans. SmE enables improved mass spectrometric analysis of several mucins, including the entire TIM family. With this information in-hand, we perform molecular dynamics (MD) simulations of TIM-3 and -4 to understand how glycosylation affects structural features of these proteins. Finally, we use these models to investigate the functional relevance of glycosylation for TIM-3 function and ligand binding. Overall, we present a powerful workflow to better understand the detailed molecular structures and functions of the mucinome.
Collapse
Affiliation(s)
| | | | - Keira E Mahoney
- Department of Chemistry, Yale University, New Haven, CT, 06511, USA
| | - Mia A Rosenfeld
- Department of Chemistry and Biochemistry, University of California, San Diego, La Jolla, CA, 92093, USA
| | - Taryn M Lucas
- Department of Chemistry, Yale University, New Haven, CT, 06511, USA
| | - Courtney M Smith
- Yale Cancer Biology Institute and Department of Pharmacology, Yale University School of Medicine, New Haven, CT, 06520, USA
| | - Alice Li
- Yale Cancer Biology Institute and Department of Pharmacology, Yale University School of Medicine, New Haven, CT, 06520, USA
| | - Deniz Ince
- Department of Chemistry, Yale University, New Haven, CT, 06511, USA
| | - Fiona L Kearns
- Department of Chemistry and Biochemistry, University of California, San Diego, La Jolla, CA, 92093, USA
| | | | - Marie A Hollenhorst
- Department of Chemistry and Sarafan ChEM-H, Stanford University, Stanford, CA, 94305, USA
- Department of Pathology, Stanford University, Stanford, CA, 94305, USA
- Department of Medicine, Division of Hematology, Stanford University, Stanford, CA, 94305, USA
| | - D Judy Shon
- Department of Chemistry and Sarafan ChEM-H, Stanford University, Stanford, CA, 94305, USA
| | - Katherine H Tiemeyer
- Department of Chemistry and Sarafan ChEM-H, Stanford University, Stanford, CA, 94305, USA
| | - Victor Attah
- Department of Chemistry, Yale University, New Haven, CT, 06511, USA
| | - Catherine Kwon
- Department of Chemistry, Yale University, New Haven, CT, 06511, USA
| | - Carolyn R Bertozzi
- Department of Chemistry and Sarafan ChEM-H, Stanford University, Stanford, CA, 94305, USA
- Howard Hughes Medical Institute, Stanford University, Stanford, CA, 94305, USA
| | | | - Mark A Lemmon
- Yale Cancer Biology Institute and Department of Pharmacology, Yale University School of Medicine, New Haven, CT, 06520, USA
| | - Rommie E Amaro
- Department of Chemistry and Biochemistry, University of California, San Diego, La Jolla, CA, 92093, USA
- Glycobiology Research and Training Center, University of California, San Diego, La Jolla, CA, 92093, USA
| | - Stacy A Malaker
- Department of Chemistry, Yale University, New Haven, CT, 06511, USA.
| |
Collapse
|
13
|
Chen H, Zha J, Tang R, Chen G. T-cell immunoglobulin and mucin-domain containing-3 (TIM-3): Solving a key puzzle in autoimmune diseases. Int Immunopharmacol 2023; 121:110418. [PMID: 37290326 DOI: 10.1016/j.intimp.2023.110418] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2023] [Revised: 05/25/2023] [Accepted: 05/30/2023] [Indexed: 06/10/2023]
Abstract
Dysfunctional immune cells participate in the pathogenesis of a variety of autoimmune diseases, although the specific mechanisms remain elusive and effective clinical interventions are lacking. Recent research on immune checkpoint molecules has revealed significant expression of T cell immunoglobulin and mucin domain-containing protein 3 (TIM-3) on the surfaces of various immune cells. These include different subsets of T cells, macrophages, dendritic cells, natural killer cells, and mast cells. Further investigation into its protein structure, ligands, and intracellular signaling pathway activation mechanisms has found that TIM-3, by binding with different ligands, is involved in the regulation of crucial biological processes such as proliferation, apoptosis, phenotypic transformation, effector protein synthesis, and cellular interactions of various immune cells. The TIM-3-ligand axis plays a pivotal role in the pathogenesis of numerous conditions, including autoimmune diseases, infections, cancers, transplant rejection, and chronic inflammation. This article primarily focuses on the research findings of TIM-3 in the field of autoimmune diseases, with a special emphasis on the structure and signaling pathways of TIM-3, its types of ligands, and the potential mechanisms implicated in systemic lupus erythematosus, multiple sclerosis, rheumatoid arthritis, as well as other autoimmune diseases and chronic inflammation. The latest research results in the field of immunology suggest that TIM-3 dysfunction affects various immune cells and participates in the pathogenesis of diseases. Monitoring the activity of its receptor-ligand axis can serve as a novel biological marker for disease clinical diagnosis and prognosis evaluation. More importantly, the TIM-3-ligand axis and the downstream signaling pathway molecules may become key targets for targeted intervention treatment of autoimmune-related diseases.
Collapse
Affiliation(s)
- Huihui Chen
- Department of Ophthalmology, The Second Xiangya Hospital of Central South University, Changsha, China; Clinical Immunology Research Center of Central South University, Changsha, China
| | - Jie Zha
- Department of Nephrology, The Second Xiangya Hospital of Central South University, Changsha, China; Hunan Key Laboratory of Kidney Disease and Blood Purification, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Runyan Tang
- Department of Nephrology, The Second Xiangya Hospital of Central South University, Changsha, China; Hunan Key Laboratory of Kidney Disease and Blood Purification, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Guochun Chen
- Clinical Immunology Research Center of Central South University, Changsha, China; Department of Nephrology, The Second Xiangya Hospital of Central South University, Changsha, China; Hunan Key Laboratory of Kidney Disease and Blood Purification, The Second Xiangya Hospital of Central South University, Changsha, China.
| |
Collapse
|
14
|
Goenka A, Khan F, Verma B, Sinha P, Dmello CC, Jogalekar MP, Gangadaran P, Ahn B. Tumor microenvironment signaling and therapeutics in cancer progression. Cancer Commun (Lond) 2023; 43:525-561. [PMID: 37005490 PMCID: PMC10174093 DOI: 10.1002/cac2.12416] [Citation(s) in RCA: 85] [Impact Index Per Article: 42.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Revised: 02/22/2023] [Accepted: 03/20/2023] [Indexed: 04/04/2023] Open
Abstract
Tumor development and metastasis are facilitated by the complex interactions between cancer cells and their microenvironment, which comprises stromal cells and extracellular matrix (ECM) components, among other factors. Stromal cells can adopt new phenotypes to promote tumor cell invasion. A deep understanding of the signaling pathways involved in cell-to-cell and cell-to-ECM interactions is needed to design effective intervention strategies that might interrupt these interactions. In this review, we describe the tumor microenvironment (TME) components and associated therapeutics. We discuss the clinical advances in the prevalent and newly discovered signaling pathways in the TME, the immune checkpoints and immunosuppressive chemokines, and currently used inhibitors targeting these pathways. These include both intrinsic and non-autonomous tumor cell signaling pathways in the TME: protein kinase C (PKC) signaling, Notch, and transforming growth factor (TGF-β) signaling, Endoplasmic Reticulum (ER) stress response, lactate signaling, Metabolic reprogramming, cyclic GMP-AMP synthase (cGAS)-stimulator of interferon genes (STING) and Siglec signaling pathways. We also discuss the recent advances in Programmed Cell Death Protein 1 (PD-1), Cytotoxic T-Lymphocyte Associated Protein 4 (CTLA4), T-cell immunoglobulin mucin-3 (TIM-3) and Lymphocyte Activating Gene 3 (LAG3) immune checkpoint inhibitors along with the C-C chemokine receptor 4 (CCR4)- C-C class chemokines 22 (CCL22)/ and 17 (CCL17), C-C chemokine receptor type 2 (CCR2)- chemokine (C-C motif) ligand 2 (CCL2), C-C chemokine receptor type 5 (CCR5)- chemokine (C-C motif) ligand 3 (CCL3) chemokine signaling axis in the TME. In addition, this review provides a holistic understanding of the TME as we discuss the three-dimensional and microfluidic models of the TME, which are believed to recapitulate the original characteristics of the patient tumor and hence may be used as a platform to study new mechanisms and screen for various anti-cancer therapies. We further discuss the systemic influences of gut microbiota in TME reprogramming and treatment response. Overall, this review provides a comprehensive analysis of the diverse and most critical signaling pathways in the TME, highlighting the associated newest and critical preclinical and clinical studies along with their underlying biology. We highlight the importance of the most recent technologies of microfluidics and lab-on-chip models for TME research and also present an overview of extrinsic factors, such as the inhabitant human microbiome, which have the potential to modulate TME biology and drug responses.
Collapse
Affiliation(s)
- Anshika Goenka
- The Ken & Ruth Davee Department of NeurologyThe Robert H. Lurie Comprehensive Cancer CenterNorthwestern University Feinberg School of MedicineChicago, 60611ILUSA
| | - Fatima Khan
- Department of Neurological SurgeryFeinberg School of MedicineNorthwestern UniversityChicago, 60611ILUSA
| | - Bhupender Verma
- Department of OphthalmologySchepens Eye Research InstituteMassachusetts Eye and Ear InfirmaryHarvard Medical SchoolBoston, 02114MAUSA
| | - Priyanka Sinha
- Department of NeurologyMassGeneral Institute for Neurodegenerative DiseaseMassachusetts General Hospital, Harvard Medical SchoolCharlestown, 02129MAUSA
| | - Crismita C. Dmello
- Department of Neurological SurgeryFeinberg School of MedicineNorthwestern UniversityChicago, 60611ILUSA
| | - Manasi P. Jogalekar
- Helen Diller Family Comprehensive Cancer CenterUniversity of California San FranciscoSan Francisco, 94143CAUSA
| | - Prakash Gangadaran
- BK21 FOUR KNU Convergence Educational Program of Biomedical Sciences for Creative Future TalentsDepartment of Biomedical Science, School of MedicineKyungpook National UniversityDaegu, 41944South Korea
- Department of Nuclear MedicineSchool of Medicine, Kyungpook National University, Kyungpook National University HospitalDaegu, 41944South Korea
| | - Byeong‐Cheol Ahn
- BK21 FOUR KNU Convergence Educational Program of Biomedical Sciences for Creative Future TalentsDepartment of Biomedical Science, School of MedicineKyungpook National UniversityDaegu, 41944South Korea
- Department of Nuclear MedicineSchool of Medicine, Kyungpook National University, Kyungpook National University HospitalDaegu, 41944South Korea
| |
Collapse
|
15
|
Dumolard L, Aspord C, Marche PN, Macek Jilkova Z. Immune checkpoints on T and NK cells in the context of HBV infection: Landscape, pathophysiology and therapeutic exploitation. Front Immunol 2023; 14:1148111. [PMID: 37056774 PMCID: PMC10086248 DOI: 10.3389/fimmu.2023.1148111] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Accepted: 03/10/2023] [Indexed: 03/30/2023] Open
Abstract
In hepatitis B virus (HBV) infection, the interplay between the virus and the host immune system is crucial in determining the pathogenesis of the disease. Patients who fail to mount a sufficient and sustained anti-viral immune response develop chronic hepatitis B (CHB). T cells and natural killer (NK) cells play decisive role in viral clearance, but they are defective in chronic HBV infection. The activation of immune cells is tightly controlled by a combination of activating and inhibitory receptors, called immune checkpoints (ICs), allowing the maintenance of immune homeostasis. Chronic exposure to viral antigens and the subsequent dysregulation of ICs actively contribute to the exhaustion of effector cells and viral persistence. The present review aims to summarize the function of various ICs and their expression in T lymphocytes and NK cells in the course of HBV infection as well as the use of immunotherapeutic strategies targeting ICs in chronic HBV infection.
Collapse
Affiliation(s)
- Lucile Dumolard
- University Grenoble Alpes, Inserm U 1209, CNRS UMR 5309, Team Epigenetics, Immunity, Metabolism, Cell Signaling & Cancer, Institute for Advanced Biosciences, Grenoble, France
| | - Caroline Aspord
- University Grenoble Alpes, Inserm U 1209, CNRS UMR 5309, Team Epigenetics, Immunity, Metabolism, Cell Signaling & Cancer, Institute for Advanced Biosciences, Grenoble, France
- R&D Laboratory, Etablissement Français du Sang Auvergne-Rhone-Alpes, Grenoble, France
| | - Patrice N. Marche
- University Grenoble Alpes, Inserm U 1209, CNRS UMR 5309, Team Epigenetics, Immunity, Metabolism, Cell Signaling & Cancer, Institute for Advanced Biosciences, Grenoble, France
| | - Zuzana Macek Jilkova
- University Grenoble Alpes, Inserm U 1209, CNRS UMR 5309, Team Epigenetics, Immunity, Metabolism, Cell Signaling & Cancer, Institute for Advanced Biosciences, Grenoble, France
- Hepato-Gastroenterology and Digestive Oncology Department, CHU Grenoble Alpes, Grenoble, France
- *Correspondence: Zuzana Macek Jilkova,
| |
Collapse
|
16
|
Gaikwad S, Agrawal MY, Kaushik I, Ramachandran S, Srivastava SK. Immune checkpoint proteins: Signaling mechanisms and molecular interactions in cancer immunotherapy. Semin Cancer Biol 2022; 86:137-150. [PMID: 35341913 DOI: 10.1016/j.semcancer.2022.03.014] [Citation(s) in RCA: 84] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Revised: 03/17/2022] [Accepted: 03/18/2022] [Indexed: 02/06/2023]
Abstract
Immune checkpoint proteins (ICP) are currently one of the most novel and promising areas of immune-oncology research. This novel way of targeting tumor cells has shown favorable success over the past few years with some FDA approvals such as Ipilimumab, Nivolumab, Pembrolizumab etc. Currently, more than 3000 clinical trials of immunotherapeutic agents are ongoing with majority being ICPs. However, as the number of trials increase so do the challenges. Some challenges such as adverse side effects, non-specific binding on healthy tissues and absence of response in some subset populations are critical obstacles. For a safe and effective further therapeutic development of molecules targeting ICPs, understanding their mechanism at molecular level is crucial. Since ICPs are mostly membrane bound receptors, a number of downstream signaling pathways divaricate following ligand-receptor binding. Most ICPs are expressed on more than one type of immune cell populations. Further, the expression varies within a cell type. This naturally varied expression pattern adds to the difficulty of targeting specific effector immune cell types against cancer. Hence, understanding the expression pattern and cellular mechanism helps lay out the possible effect of any immunotherapy. In this review, we discuss the signaling mechanism, expression pattern among various immune cells and molecular interactions derived using interaction database analysis (BioGRID).
Collapse
Affiliation(s)
- Shreyas Gaikwad
- Department of Immunotherapeutics and Biotechnology, and Center for Tumor Immunology and Targeted Cancer Therapy, Texas Tech University Health Sciences Center, Abilene, TX 79601, USA
| | - Manas Yogendra Agrawal
- Department of Immunotherapeutics and Biotechnology, and Center for Tumor Immunology and Targeted Cancer Therapy, Texas Tech University Health Sciences Center, Abilene, TX 79601, USA
| | - Itishree Kaushik
- Department of Immunotherapeutics and Biotechnology, and Center for Tumor Immunology and Targeted Cancer Therapy, Texas Tech University Health Sciences Center, Abilene, TX 79601, USA
| | - Sharavan Ramachandran
- Department of Immunotherapeutics and Biotechnology, and Center for Tumor Immunology and Targeted Cancer Therapy, Texas Tech University Health Sciences Center, Abilene, TX 79601, USA
| | - Sanjay K Srivastava
- Department of Immunotherapeutics and Biotechnology, and Center for Tumor Immunology and Targeted Cancer Therapy, Texas Tech University Health Sciences Center, Abilene, TX 79601, USA.
| |
Collapse
|
17
|
Kuske M, Haist M, Jung T, Grabbe S, Bros M. Immunomodulatory Properties of Immune Checkpoint Inhibitors-More than Boosting T-Cell Responses? Cancers (Basel) 2022; 14:1710. [PMID: 35406483 PMCID: PMC8996886 DOI: 10.3390/cancers14071710] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2022] [Revised: 03/24/2022] [Accepted: 03/24/2022] [Indexed: 12/11/2022] Open
Abstract
The approval of immune checkpoint inhibitors (ICI) that serve to enhance effector T-cell anti-tumor responses has strongly improved success rates in the treatment of metastatic melanoma and other tumor types. The currently approved ICI constitute monoclonal antibodies blocking cytotoxic T-lymphocyte-associated protein (CTLA)-4 and anti-programmed cell death (PD)-1. By this, the T-cell-inhibitory CTLA-4/CD80/86 and PD-1/PD-1L/2L signaling axes are inhibited. This leads to sustained effector T-cell activity and circumvents the immune evasion of tumor cells, which frequently upregulate PD-L1 expression and modulate immune checkpoint molecule expression on leukocytes. As a result, profound clinical responses are observed in 40-60% of metastatic melanoma patients. Despite the pivotal role of T effector cells for triggering anti-tumor immunity, mounting evidence indicates that ICI efficacy may also be attributable to other cell types than T effector cells. In particular, emerging research has shown that ICI also impacts innate immune cells, such as myeloid cells, natural killer cells and innate lymphoid cells, which may amplify tumoricidal functions beyond triggering T effector cells, and thus improves clinical efficacy. Effects of ICI on non-T cells may additionally explain, in part, the character and extent of adverse effects associated with treatment. Deeper knowledge of these effects is required to further develop ICI treatment in terms of responsiveness of patients to treatment, to overcome resistance to ICI and to alleviate adverse effects. In this review we give an overview into the currently known immunomodulatory effects of ICI treatment in immune cell types other than the T cell compartment.
Collapse
Affiliation(s)
| | | | | | | | - Matthias Bros
- Department of Dermatology, University Medical Center Mainz, Langenbeckstraße 1, 55131 Mainz, Germany; (M.K.); (M.H.); (T.J.); (S.G.)
| |
Collapse
|
18
|
Immune Checkpoint Receptors Signaling in T Cells. Int J Mol Sci 2022; 23:ijms23073529. [PMID: 35408889 PMCID: PMC8999077 DOI: 10.3390/ijms23073529] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Revised: 03/16/2022] [Accepted: 03/21/2022] [Indexed: 12/15/2022] Open
Abstract
The characterization of the receptors negatively modulating lymphocyte function is rapidly advancing, driven by success in tumor immunotherapy. As a result, the number of immune checkpoint receptors characterized from a functional perspective and targeted by innovative drugs continues to expand. This review focuses on the less explored area of the signaling mechanisms of these receptors, of those expressed in T cells. Studies conducted mainly on PD-1, CTLA-4, and BTLA have evidenced that the extracellular parts of some of the receptors act as decoy receptors for activating ligands, but in all instances, the tyrosine phosphorylation of their cytoplasmatic tail drives a crucial inhibitory signal. This negative signal is mediated by a few key signal transducers, such as tyrosine phosphatase, inositol phosphatase, and diacylglycerol kinase, which allows them to counteract TCR-mediated activation. The characterization of these signaling pathways is of great interest in the development of therapies for counteracting tumor-infiltrating lymphocyte exhaustion/anergy independently from the receptors involved.
Collapse
|
19
|
Immunotherapy for Colorectal Cancer: Mechanisms and Predictive Biomarkers. Cancers (Basel) 2022; 14:cancers14041028. [PMID: 35205776 PMCID: PMC8869923 DOI: 10.3390/cancers14041028] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 02/07/2022] [Accepted: 02/09/2022] [Indexed: 12/12/2022] Open
Abstract
Simple Summary Late-stage colorectal cancer treatment often involves chemotherapy and radiation that can cause dose-limiting toxicity, and therefore there is great interest in developing targeted therapies for this disease. Immunotherapy is a targeted therapy that uses peptides, cells, antibodies, viruses, or small molecules to engage or train the immune system to kill cancer. Here, we discuss the preclinical and clinical development of immunotherapy for treatment of colorectal cancer and provide an overview of predictive biomarkers for such treatments. We also consider open questions including optimal combination treatments and sensitization of colorectal cancer patients with proficient mismatch repair enzymes. Abstract Though early-stage colorectal cancer has a high 5 year survival rate of 65–92% depending on the specific stage, this probability drops to 13% after the cancer metastasizes. Frontline treatments for colorectal cancer such as chemotherapy and radiation often produce dose-limiting toxicities in patients and acquired resistance in cancer cells. Additional targeted treatments are needed to improve patient outcomes and quality of life. Immunotherapy involves treatment with peptides, cells, antibodies, viruses, or small molecules to engage or train the immune system to kill cancer cells. Preclinical and clinical investigations of immunotherapy for treatment of colorectal cancer including immune checkpoint blockade, adoptive cell therapy, monoclonal antibodies, oncolytic viruses, anti-cancer vaccines, and immune system modulators have been promising, but demonstrate limitations for patients with proficient mismatch repair enzymes. In this review, we discuss preclinical and clinical studies investigating immunotherapy for treatment of colorectal cancer and predictive biomarkers for response to these treatments. We also consider open questions including optimal combination treatments to maximize efficacy, minimize toxicity, and prevent acquired resistance and approaches to sensitize mismatch repair-proficient patients to immunotherapy.
Collapse
|
20
|
Zhao S, Wang C, Lu P, Lou Y, Liu H, Wang T, Yang S, Bao Z, Han L, Liang X, Ma C, Gao L. Switch receptor T3/28 improves long-term persistence and antitumor efficacy of CAR-T cells. J Immunother Cancer 2021; 9:jitc-2021-003176. [PMID: 34853180 PMCID: PMC8638458 DOI: 10.1136/jitc-2021-003176] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/04/2021] [Indexed: 01/14/2023] Open
Abstract
Background Chimeric antigen receptor (CAR) T cells have been successfully used in tumor immunotherapy due to their strong antitumor responses, especially in hematological malignancies such as B cell acute lymphoid leukemia. However, on-target off-tumor toxicity and poor persistence severely limit the clinical application of CAR-T cell therapy. Methods T-cell immunoglobulin mucin domain molecule 3 (TIM-3) was used to develop a second-generation 41BB CD19 CAR linked with a T3/28 chimera, in which truncated extracellular TIM-3 was fused with the CD28 transmembrane and cytoplasmic domains. The efficacy of T3/28 CAR-T cells was evaluated in vitro and in vivo. Results We demonstrated that the switch receptor T3/28 preserved the TCM phenotype, improved proliferative capacity, and reduced exhaustion of CAR-T cells, resulting in superior in vitro and in vivo antitumor activity in B lymphoma. Importantly, the switch receptor T3/28 substantially prolonged the persistence of CAR-T cells, and the interleukin-21/Stat3 axis probably contributed to the enhanced cytotoxicity of T3/28 CAR-T cells. Conclusion Overall, the T3/28 chimera significantly prolonged the persistence of CAR-T cells, and T3/28 CAR-T cells possessed potent antitumor activity in mice, shedding new light on potential improvements in adoptive T cell therapies.
Collapse
Affiliation(s)
- Songbo Zhao
- Key Laboratory for Experimental Teratology of Ministry of Education, Shandong Key Laboratory of Infection and Immunity, and Department of Immunology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Chunhua Wang
- Institute of Marine Science and Technology, Shandong University, Qingdao, Shandong, China
| | - Ping Lu
- Department of Hematology, Zibo Central Hospital, Zibo, Shandong, China
| | - Yalin Lou
- Key Laboratory for Experimental Teratology of Ministry of Education, Shandong Key Laboratory of Infection and Immunity, and Department of Immunology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Huimin Liu
- Key Laboratory for Experimental Teratology of Ministry of Education, Shandong Key Laboratory of Infection and Immunity, and Department of Immunology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Ting Wang
- Biomedical Sciences College & Shandong Medicinal Biotechnology Centre, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Shanshan Yang
- Key Laboratory for Experimental Teratology of Ministry of Education, Shandong Key Laboratory of Infection and Immunity, and Department of Immunology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Ziyou Bao
- Key Laboratory for Experimental Teratology of Ministry of Education, Shandong Key Laboratory of Infection and Immunity, and Department of Immunology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Lin Han
- Institute of Marine Science and Technology, Shandong University, Qingdao, Shandong, China
| | - Xiaohong Liang
- Key Laboratory for Experimental Teratology of Ministry of Education, Shandong Key Laboratory of Infection and Immunity, and Department of Immunology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Chunhong Ma
- Key Laboratory for Experimental Teratology of Ministry of Education, Shandong Key Laboratory of Infection and Immunity, and Department of Immunology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Lifen Gao
- Key Laboratory for Experimental Teratology of Ministry of Education, Shandong Key Laboratory of Infection and Immunity, and Department of Immunology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| |
Collapse
|
21
|
Regulation of Tim-3 function by binding to phosphatidylserine. Biochem J 2021; 478:3999-4004. [PMID: 34813649 DOI: 10.1042/bcj20210652] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 10/27/2021] [Accepted: 10/29/2021] [Indexed: 12/17/2022]
Abstract
Tim-3 is a transmembrane protein that is highly expressed on subsets of chronically stimulated CD4+ helper and CD8+ cytotoxic T cells, with more transient expression during acute activation and infection. Tim-3 is also constitutively expressed by multiple types of myeloid cells. Like other TIM family members, Tim-3 can bind to phosphatidylserine displayed by apoptotic cells, and this interaction has been shown to mediate uptake of such cells by dendritic cells and cross-presentation of antigens to CD8+ T cells. In contrast, how the recognition of PS by Tim-3 might regulate the function of Tim-3+ T cells is not known. In their recent paper, Lemmon and colleagues demonstrate for the first time that recognition of PS by Tim-3 leads to enhanced T cell activation.
Collapse
|