1
|
Inthanachai T, Boonkrai C, Phakham T, Pisitkun T, Thaiwong R, Chuthaphakdikun V, Sakunrangsit N, Limprasutr V, Chinsuwan T, Hirankarn N, Suppipat K, Watanabe N, Tawinwung S. Novel B7-H3 CAR T cells show potent antitumor effects in glioblastoma: a preclinical study. J Immunother Cancer 2025; 13:e010083. [PMID: 39863300 PMCID: PMC11784176 DOI: 10.1136/jitc-2024-010083] [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: 07/11/2024] [Accepted: 01/11/2025] [Indexed: 01/27/2025] Open
Abstract
BACKGROUND B7 homolog 3 (B7-H3), an overexpressed antigen across multiple solid cancers, represents a promising target for CAR T cell therapy. This study investigated the expression of B7-H3 across various solid tumors and developed novel monoclonal antibodies (mAbs) targeting B7-H3 for CAR T cell therapy. METHODS Expression of B7-H3 across various solid tumors was evaluated using RNA-seq data from TCGA, TARGET, and GTEx datasets and by flow cytometry staining. B7-H3-specific mAbs were developed by immunizing mice with human B7-H3, screening with ELISA, and analyzing kinetics with surface plasmon resonance. These mAbs were used to create second-generation CAR constructs, which were evaluated in vitro and in vivo for their antitumor function. RESULTS We identified four mAb clones from immunized mice, with three demonstrating high specificity and affinity. The second-generation B7-H3 CAR T cells derived from these mAbs exhibited robust cytotoxicity against B7-H3-positive targets and successfully infiltrated and eliminated tumor spheroids in vitro. In a xenograft mouse model of glioblastoma, these CAR T cells, particularly those derived from clone A2H4, eradicated the primary tumor, and effectively controlled rechallenge tumor, resulting in prolonged survival of the xenograft mice. In vivo T cell trafficking revealed high accumulation and persistence of A2H4-derived CAR T cells at the tumor site. CONCLUSIONS Our results provide novel B7-H3-targeted CAR T cells with high efficacy, paving the way for clinical translation of solid tumor treatment.
Collapse
Affiliation(s)
- Thananya Inthanachai
- Medical Microbiology, Interdisciplinary and International Program, Graduate School, Chulalongkorn University, Bangkok, Thailand
- Cellular Immunotherapy Research Unit, Chulalongkorn University, Bangkok, Thailand
| | - Chatikorn Boonkrai
- Center of Excellence in Systems Biology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Tanapati Phakham
- Center of Excellence in Systems Biology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Trairak Pisitkun
- Center of Excellence in Systems Biology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Rattapoom Thaiwong
- Cellular Immunotherapy Research Unit, Chulalongkorn University, Bangkok, Thailand
- Chulalongkorn Comprehensive Cancer Center, King Chulalongkorn Memorial Hospital, Bangkok, Thailand
| | - Vichaya Chuthaphakdikun
- Cellular Immunotherapy Research Unit, Chulalongkorn University, Bangkok, Thailand
- Chulalongkorn Comprehensive Cancer Center, King Chulalongkorn Memorial Hospital, Bangkok, Thailand
| | - Nithidol Sakunrangsit
- Cellular Immunotherapy Research Unit, Chulalongkorn University, Bangkok, Thailand
- Department of Pharmacology and Physiology, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok, Thailand
| | - Vudhiporn Limprasutr
- Department of Pharmacology and Physiology, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok, Thailand
- Chulalongkorn University Laboratory Animal Center, Chulalongkorn University, Bangkok, Thailand
| | - Thanyavi Chinsuwan
- Cellular Immunotherapy Research Unit, Chulalongkorn University, Bangkok, Thailand
- Department of Microbiology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Nattiya Hirankarn
- Department of Microbiology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Koramit Suppipat
- Cellular Immunotherapy Research Unit, Chulalongkorn University, Bangkok, Thailand
- Department of Research Affairs, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Norihiro Watanabe
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, Texas, USA
| | - Supannikar Tawinwung
- Cellular Immunotherapy Research Unit, Chulalongkorn University, Bangkok, Thailand
- Department of Pharmacology and Physiology, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok, Thailand
| |
Collapse
|
2
|
Charley KR, Ramstead AG, Matous JG, Kumaki Y, Sircy LM, Hale JS, Williams MA. Effector-Phase IL-2 Signals Drive Th1 Effector and Memory Responses Dependently and Independently of TCF-1. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2024; 212:586-595. [PMID: 38149929 PMCID: PMC10872735 DOI: 10.4049/jimmunol.2300570] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Accepted: 12/03/2023] [Indexed: 12/28/2023]
Abstract
Following viral infection, CD4+ T cell differentiation is tightly regulated by cytokines and TCR signals. Although most activated CD4+ T cells express IL-2Rα after lymphocytic choriomeningtis virus infection, by day 3 postinfection, only half of activated T cells maintain expression. IL-2Rα at this time point distinguishes precursors for terminally differentiated Th1 cells (IL-2Rαhi) from precursors for Tfh cells and memory T cells (IL-2Rαlo) and is linked to strong TCR signals. In this study, we test whether TCR-dependent IL-2 links the TCR to CD4+ T cell differentiation. We employ a mixture of anti-IL-2 Abs to neutralize IL-2 throughout the primary CD4+ T cell response to lymphocytic choriomeningitis virus infection in mice or only after the establishment of lineage-committed effector cells (day 3 postinfection). We report that IL-2 signals drive the formation of Th1 precursor cells in the early stages of the immune response and sustain Th1 responses during its later stages (after day 3). Effector-stage IL-2 also shapes the composition and function of resulting CD4+ memory T cells. Although IL-2 has been shown previously to drive Th1 differentiation by reducing the activity of the transcriptional repressor TCF-1, we found that sustained IL-2 signals were still required to drive optimal Th1 differentiation even in the absence of TCF-1. Therefore, we concluded that IL-2 plays a central role throughout the effector phase in regulating the balance between Th1 and Tfh effector and memory cells via mechanisms that are both dependent and independent of its role in modulating TCF-1 activity.
Collapse
Affiliation(s)
- Krystal R. Charley
- Division of Microbiology and Immunology, Department of Pathology, University of Utah School of Medicine, Salt Lake City, Utah 84112
- Huntsman Cancer Institute, University of Utah Health, Salt Lake City, Utah 84112
| | - Andrew G. Ramstead
- Division of Microbiology and Immunology, Department of Pathology, University of Utah School of Medicine, Salt Lake City, Utah 84112
- Huntsman Cancer Institute, University of Utah Health, Salt Lake City, Utah 84112
| | - Joseph G. Matous
- Division of Microbiology and Immunology, Department of Pathology, University of Utah School of Medicine, Salt Lake City, Utah 84112
- Huntsman Cancer Institute, University of Utah Health, Salt Lake City, Utah 84112
| | - Yohichi Kumaki
- Division of Microbiology and Immunology, Department of Pathology, University of Utah School of Medicine, Salt Lake City, Utah 84112
- Huntsman Cancer Institute, University of Utah Health, Salt Lake City, Utah 84112
| | - Linda M. Sircy
- Division of Microbiology and Immunology, Department of Pathology, University of Utah School of Medicine, Salt Lake City, Utah 84112
| | - J. Scott Hale
- Division of Microbiology and Immunology, Department of Pathology, University of Utah School of Medicine, Salt Lake City, Utah 84112
| | - Matthew A. Williams
- Division of Microbiology and Immunology, Department of Pathology, University of Utah School of Medicine, Salt Lake City, Utah 84112
- Huntsman Cancer Institute, University of Utah Health, Salt Lake City, Utah 84112
| |
Collapse
|
3
|
Chinsuwan T, Hirabayashi K, Mishima S, Hasegawa A, Tanaka M, Mochizuki H, Shimoi A, Murakami T, Yagyu S, Shimizu K, Nakazawa Y. Ligand-based, piggyBac-engineered CAR-T cells targeting EGFR are safe and effective against non-small cell lung cancers. Mol Ther Oncolytics 2023; 31:100728. [PMID: 37822488 PMCID: PMC10562194 DOI: 10.1016/j.omto.2023.100728] [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: 02/16/2023] [Accepted: 09/14/2023] [Indexed: 10/13/2023] Open
Abstract
Epidermal growth factor receptor (EGFR) is overexpressed in various cancers, including non-small cell lung cancer (NSCLC), and in some somatic cells at a limited level, rendering it an attractive antitumor target. In this study, we engineered chimeric antigen receptor (CAR)-T cells using the piggyBac transposon system, autologous artificial antigen-presenting cells, and natural ligands of EGFR. We showed that this approach yielded CAR-T cells with favorable phenotypes and CAR positivity. They exhibited potent antitumor activity against NSCLC both in vitro and in vivo. When administered to tumor-bearing mice and non-tumor-bearing cynomolgus macaques, they did not elicit toxicity despite their cross-reactivity to both murine and simian EGFRs. In total we tested three ligands and found that the CAR candidate with the highest affinity consistently displayed greater potency without adverse events. Taken together, our results demonstrate the feasibility and safety of targeting EGFR-expressing NSCLCs using ligand-based, piggyBac-engineered CAR-T cells. Our data also show that lowering the affinity of CAR molecules is not always beneficial.
Collapse
Affiliation(s)
- Thanyavi Chinsuwan
- Department of Pediatrics, Shinshu University School of Medicine, Matsumoto, Nagano, Japan
- Department of Microbiology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Koichi Hirabayashi
- Department of Pediatrics, Shinshu University School of Medicine, Matsumoto, Nagano, Japan
| | - Shuji Mishima
- Division of General Thoracic Surgery, Department of Surgery, Shinshu University School of Medicine, Matsumoto, Nagano, Japan
| | - Aiko Hasegawa
- Department of Pediatrics, Shinshu University School of Medicine, Matsumoto, Nagano, Japan
| | - Miyuki Tanaka
- Department of Pediatrics, Shinshu University School of Medicine, Matsumoto, Nagano, Japan
- Center for Advanced Research of Gene and Cell Therapy, Shinshu University School of Medicine, Matsumoto, Nagano, Japan
| | - Hidemi Mochizuki
- Center for Advanced Research of Gene and Cell Therapy, Shinshu University School of Medicine, Matsumoto, Nagano, Japan
- Ina Research Inc., Ina, Nagano, Japan
| | - Akihito Shimoi
- Center for Advanced Research of Gene and Cell Therapy, Shinshu University School of Medicine, Matsumoto, Nagano, Japan
- Ina Research Inc., Ina, Nagano, Japan
| | - Takashi Murakami
- Department of Microbiology, Faculty of Medicine, Saitama Medical University, Iruma, Saitama, Japan
| | - Shigeki Yagyu
- Department of Pediatrics, Shinshu University School of Medicine, Matsumoto, Nagano, Japan
- Center for Advanced Research of Gene and Cell Therapy, Shinshu University School of Medicine, Matsumoto, Nagano, Japan
| | - Kimihiro Shimizu
- Division of General Thoracic Surgery, Department of Surgery, Shinshu University School of Medicine, Matsumoto, Nagano, Japan
| | - Yozo Nakazawa
- Department of Pediatrics, Shinshu University School of Medicine, Matsumoto, Nagano, Japan
- Center for Advanced Research of Gene and Cell Therapy, Shinshu University School of Medicine, Matsumoto, Nagano, Japan
- Institute for Biomedical Sciences, Interdisciplinary Cluster for Cutting Edge Research, Shinshu University, Matsumoto, Nagano, Japan
| |
Collapse
|
4
|
Tim-3 blockade enhances the clearance of Chlamydia psittaci in the lung by promoting a cell-mediated immune response. Int Immunopharmacol 2023; 116:109780. [PMID: 36720194 DOI: 10.1016/j.intimp.2023.109780] [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: 09/26/2022] [Revised: 01/10/2023] [Accepted: 01/20/2023] [Indexed: 01/30/2023]
Abstract
Chlamydia psittaci is remarkable at disrupting immunity and thus poses a great risk to the animal industry and public health. Immune inhibitory molecule upregulation and the accumulation of specialized cells play key roles in chlamydial clearance. It is clear that the T-cell immunoglobulin and mucin domain protein 3 receptor (Tim-3) can regulate effector T cells in infectious disease. However, the immunomodulatory effect of Tim-3 in C. psittaci infection remains unknown. Thus, the expression of Tim-3 in effector T cells and its immune regulatory function during C. psittaci infection were investigated. The level of Tim-3 on CD4+ and CD8+ T cells was meaningfully higher in C. psittaci-infected mice. Blockade of Tim-3 signaling by anti-Tim-3 antibody showed accelerated C. psittaci clearance and less pathological changes in the lung than isotype immunoglobulin treatment. Furthermore, treatment with anti-Tim-3 antibody greatly enhanced the levels of IFN-γ and interleukin (IL)-22/IL-17, which were correlated with an improved Th1- and Th17-mediated immune response, and decreased IL-10, which were related with a decreased Treg immune response. In conclusion, Tim-3 expression in effector T cells negatively regulates Th1 and Th17 immune responses against C. psittaci respiratory infection.
Collapse
|
5
|
Banerjee H, Nieves-Rosado H, Kulkarni A, Murter B, McGrath KV, Chandran UR, Chang A, Szymczak-Workman AL, Vujanovic L, Delgoffe GM, Ferris RL, Kane LP. Expression of Tim-3 drives phenotypic and functional changes in Treg cells in secondary lymphoid organs and the tumor microenvironment. Cell Rep 2021; 36:109699. [PMID: 34525351 PMCID: PMC8482289 DOI: 10.1016/j.celrep.2021.109699] [Citation(s) in RCA: 55] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Revised: 04/29/2021] [Accepted: 08/18/2021] [Indexed: 12/17/2022] Open
Abstract
Regulatory T cells (Treg cells) are critical mediators of self-tolerance, but they can also limit effective anti-tumor immunity. Although under homeostasis a small fraction of Treg cells in lymphoid organs express the putative checkpoint molecule Tim-3, this protein is expressed by a much larger proportion of tumor-infiltrating Treg cells. Using a mouse model that drives cell-type-specific inducible Tim-3 expression, we show that expression of Tim-3 by Treg cells is sufficient to drive Treg cells to a more effector-like phenotype, resulting in increases in suppressive activity, effector T cell exhaustion, and tumor growth. We also show that T-reg-cell-specific inducible deletion of Tim-3 enhances anti-tumor immunity. Enhancement of Treg cell function by Tim-3 is strongly correlated with increased expression of interleukin-10 (IL-10) and a shift to a more glycolytic metabolic phenotype. Our data demonstrate that Tim-3+ Treg cells may be a relevant therapeutic target cell type for the treatment of cancer.
Collapse
Affiliation(s)
- Hridesh Banerjee
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA
| | - Hector Nieves-Rosado
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA; Medical Scientist Training Program, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA; Graduate Program in Microbiology and Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA
| | - Aditi Kulkarni
- Hillman Cancer Center, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA
| | - Benjamin Murter
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA; Graduate Program in Microbiology and Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA
| | - Kyle V McGrath
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA
| | - Uma R Chandran
- Department of Biomedical Informatics, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA
| | - Alexander Chang
- Department of Biomedical Informatics, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA
| | | | - Lazar Vujanovic
- Hillman Cancer Center, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA; Department of Otolaryngology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA
| | - Greg M Delgoffe
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA; Hillman Cancer Center, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA
| | - Robert L Ferris
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA; Hillman Cancer Center, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA; Department of Otolaryngology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA
| | - Lawrence P Kane
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA.
| |
Collapse
|
6
|
Kataoka S, Manandhar P, Lee J, Workman CJ, Banerjee H, Szymczak-Workman AL, Kvorjak M, Lohmueller J, Kane LP. The costimulatory activity of Tim-3 requires Akt and MAPK signaling and its recruitment to the immune synapse. Sci Signal 2021; 14:eaba0717. [PMID: 34131021 PMCID: PMC9741863 DOI: 10.1126/scisignal.aba0717] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Expression of the transmembrane protein Tim-3 is increased on dysregulated T cells undergoing chronic activation, including during chronic infection and in solid tumors. Thus, Tim-3 is generally thought of as an inhibitory protein. We and others previously reported that under some circumstances, Tim-3 exerts paradoxical costimulatory activity in T cells (and other cells), including enhancement of the phosphorylation of ribosomal S6 protein. Here, we examined the upstream signaling pathways that control Tim-3-mediated increases in phosphorylated S6 in T cells. We also defined the localization of Tim-3 relative to the T cell immune synapse and its effects on downstream signaling. Recruitment of Tim-3 to the immune synapse was mediated exclusively by the transmembrane domain, replacement of which impaired the ability of Tim-3 to costimulate T cell receptor (TCR)-dependent S6 phosphorylation. Furthermore, enforced localization of the Tim-3 cytoplasmic domain to the immune synapse in a chimeric antigen receptor still enabled T cell activation. Together, our findings are consistent with a model whereby Tim-3 enhances TCR-proximal signaling under acute conditions.
Collapse
Affiliation(s)
- Shunsuke Kataoka
- Department of Immunology, University of Pittsburgh, Pittsburgh, PA 15261, USA
- Asahi Kasei Pharma Corporation, Shizuoka, Japan
- Graduate Program in Microbiology and Immunology, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Priyanka Manandhar
- Department of Immunology, University of Pittsburgh, Pittsburgh, PA 15261, USA
- Graduate Program in Microbiology and Immunology, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Judong Lee
- Department of Immunology, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Creg J Workman
- Department of Immunology, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Hridesh Banerjee
- Department of Immunology, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | | | - Michael Kvorjak
- Department of Surgery, Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Jason Lohmueller
- Department of Surgery, Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Lawrence P Kane
- Department of Immunology, University of Pittsburgh, Pittsburgh, PA 15261, USA.
| |
Collapse
|
7
|
Saleh R, Toor SM, Elkord E. Targeting TIM-3 in solid tumors: innovations in the preclinical and translational realm and therapeutic potential. Expert Opin Ther Targets 2020; 24:1251-1262. [PMID: 33103506 DOI: 10.1080/14728222.2020.1841750] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
INTRODUCTION Immune checkpoint inhibitors (ICIs) have shown a great therapeutic efficacy in cancer patients. However, a significant proportion of cancer patients remain unresponsive or show limited response. T cell immunoglobulin and mucin-domain containing protein-3 (TIM-3) is a co-inhibitory receptor expressed on various cell types and is involved in the attenuation of immune responses. TIM-3 and its ligands are highly expressed in various solid malignancies and some studies have reported its association with worse disease outcomes. Thus, targeting TIM-3 could be a promising therapeutic approach to treat cancer patients. AREAS COVERED This review describes the role of TIM-3 and its ligands in regulating anti-tumor immunity and their contribution to cancer progression. Moreover, this review focuses on the preclinical models and translational data from important studies published in PubMed till October 2020, which demonstrate the therapeutic benefits of targeting TIM-3 signaling. EXPERT OPINION Despite the promising data obtained from targeting TIM-3 in preclinical models, precise mechanisms underlying the anti-tumor effects of TIM-3 inhibition are not fully elucidated. Therefore, mechanistic studies are required to provide better insights into the anti-tumor effects of targeting TIM-3, and clinical data are necessary to determine the safety profiles and therapeutic efficacy of TIM-3 inhibition in cancer patients.
Collapse
Affiliation(s)
- Reem Saleh
- Cancer Research Center, Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation (QF) , Doha, Qatar
| | - Salman M Toor
- Cancer Research Center, Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation (QF) , Doha, Qatar
| | - Eyad Elkord
- Biomedical Research Center, School of Science, Engineering and Environment, University of Salford , Manchester, United Kingdom
| |
Collapse
|
8
|
Significance of TIM-3 expression by CD4 + and CD8 + T lymphocytes in tumor-draining lymph nodes from patients with breast cancer. Mol Immunol 2020; 128:47-54. [PMID: 33068833 DOI: 10.1016/j.molimm.2020.10.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Revised: 08/23/2020] [Accepted: 10/05/2020] [Indexed: 02/06/2023]
Abstract
T cell immunoglobulin and mucin domain-containing protein 3 (TIM-3), which is expressed by immune and nonimmune cells, has been shown to play immunoregulatory roles in the tumor microenvironment. In this study we assessed the expression of TIM-3 by T cells from tumor draining lymph nodes (TDLNs) of patients with breast cancer and its association with disease progression. Lymphocytes were isolated from 41 TDLNs, and flow cytometry was used to determine the expression of TIM-3 on CD4+ and CD8+ T cells, along with the simultaneous expression of CD25, Foxp3 and TIM-3 in CD4+ T cells. The results showed that the frequency of TIM-3+CD8+ T cells was associated with higher tumor grade, and the geometric mean fluorescence intensity (gMFI) of TIM-3 in CD4+ and CD8+ T cells was significantly higher in patients with more than 9 involved lymph nodes than those with fewer involved nodes. The gMFI of TIM3 in CD4+ T cells also showed a direct correlation with the number of metastatic lymph nodes. Phenotypic characterization of TIM-3+CD4+ T cells showed that the majority of CD4+TIM3+ lymphocytes were Foxp3 ̶ CD25 ̶, and the majority of Foxp3+CD25+ regulatory T cells were TIM-3-. Our findings showed that TIM-3 was expressed by CD4+, CD8+ and regulatory T cells in breast TDLNs, and that expression on CD4+ and CD8+ T cells was mostly associated with poor prognosticators such as a higher number of involved lymph nodes or higher tumor grade. More studies are required to confirm TIM-3 as a prognostic marker and a target for immunotherapy in breast cancer.
Collapse
|
9
|
Herrmann M, Schulte S, Wildner NH, Wittner M, Brehm TT, Ramharter M, Woost R, Lohse AW, Jacobs T, Schulze zur Wiesch J. Analysis of Co-inhibitory Receptor Expression in COVID-19 Infection Compared to Acute Plasmodium falciparum Malaria: LAG-3 and TIM-3 Correlate With T Cell Activation and Course of Disease. Front Immunol 2020; 11:1870. [PMID: 32983106 PMCID: PMC7479337 DOI: 10.3389/fimmu.2020.01870] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Accepted: 07/13/2020] [Indexed: 12/12/2022] Open
Abstract
Coronavirus disease 2019 (COVID-19) which is caused by the novel SARS-CoV-2 virus is a severe flu-like illness which is associated with hyperinflammation and immune dysfunction. The virus induces a strong T and B cell response but little is known about the immune pathology of this viral infection. Acute Plasmodium falciparum malaria also causes acute clinical illness and is characterized by hyperinflammation due to the strong production of pro-inflammatory cytokines and a massive activation of T cells. In malaria, T cells express a variety of co-inhibitory receptors which might be a consequence of their activation but also might limit their overwhelming function. Thus, T cells are implicated in protection as well as in pathology. The outcome of malaria is thought to be a consequence of the balance between co-activation and co-inhibition of T cells. Following the hypothesis that T cells in COVID-19 might have a similar, dual function, we comprehensively characterized the differentiation (CCR7, CD45RO) and activation status (HLA-DR, CD38, CD69, CD226), the co-expression of co-inhibitory molecules (PD1, TIM-3, LAG-3, BTLA, TIGIT), as well as the expression pattern of the transcription factors T-bet and eomes of CD8+ and CD4+ T cells of PBMC of n = 20 SARS-CoV-2 patients compared to n = 10 P. falciparum infected patients and n = 13 healthy controls. Overall, acute COVID-19 and malaria infection resulted in a comparably elevated activation and altered differentiation status of the CD8+ and CD4+ T cell populations. T effector cells of COVID-19 and malaria patients showed higher frequencies of the inhibitory receptors T-cell immunoglobulin mucin-3 (TIM-3) and Lymphocyte-activation gene-3 (LAG-3) which was linked to increased activation levels and an upregulation of the transcription factors T-bet and eomes. COVID-19 patients with a more severe disease course showed higher levels of LAG-3 and TIM-3 than patients with a mild disease course. During recovery, a rapid normalization of these inhibitory receptors could be observed. In summary, comparing the expression of different co-inhibitory molecules in CD8+ and CD4+ T cells in COVID-19 vs. malaria, there is a transient increase of the expression of certain inhibitory receptors like LAG-3 and TIM-3 in COVID-19 in the overall context of acute immune activation.
Collapse
Affiliation(s)
- Marissa Herrmann
- Infectious Diseases Unit, I. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- German Center for Infection Research (DZIF), Partner Site Hamburg-Lübeck-Borstel-Riems, Hamburg, Germany
| | - Sophia Schulte
- Infectious Diseases Unit, I. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Nils H. Wildner
- Infectious Diseases Unit, I. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Melanie Wittner
- Infectious Diseases Unit, I. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- German Center for Infection Research (DZIF), Partner Site Hamburg-Lübeck-Borstel-Riems, Hamburg, Germany
| | - Thomas Theo Brehm
- Infectious Diseases Unit, I. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- German Center for Infection Research (DZIF), Partner Site Hamburg-Lübeck-Borstel-Riems, Hamburg, Germany
| | - Michael Ramharter
- Infectious Diseases Unit, I. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- German Center for Infection Research (DZIF), Partner Site Hamburg-Lübeck-Borstel-Riems, Hamburg, Germany
- Department of Tropical Medicine, Bernhard-Nocht-Institute for Tropical Medicine (BNITM), Hamburg, Germany
| | - Robin Woost
- Infectious Diseases Unit, I. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- German Center for Infection Research (DZIF), Partner Site Hamburg-Lübeck-Borstel-Riems, Hamburg, Germany
| | - Ansgar W. Lohse
- Infectious Diseases Unit, I. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- German Center for Infection Research (DZIF), Partner Site Hamburg-Lübeck-Borstel-Riems, Hamburg, Germany
| | - Thomas Jacobs
- Protozoa Immunology, Bernhard-Nocht-Institute for Tropical Medicine, Hamburg, Germany
| | - Julian Schulze zur Wiesch
- Infectious Diseases Unit, I. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- German Center for Infection Research (DZIF), Partner Site Hamburg-Lübeck-Borstel-Riems, Hamburg, Germany
| |
Collapse
|
10
|
Differential Expression of Immune Checkpoint Molecules on CD8 + T Cells Specific for Immunodominant and Subdominant Herpes Simplex Virus 1 Epitopes. J Virol 2020; 94:JVI.01132-19. [PMID: 31645447 DOI: 10.1128/jvi.01132-19] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Accepted: 10/14/2019] [Indexed: 12/14/2022] Open
Abstract
Herpes simplex virus 1 (HSV-1) causes a lifelong infection of neurons that innervate barrier sites like the skin and mucosal surfaces like the eye. After primary infection of the cornea, the virus enters latency within the trigeminal ganglion (TG), from which it can reactivate throughout the life of the host. Viral latency is maintained, in part, by virus-specific CD8+ T cells that nonlethally interact with infected neurons. When CD8+ T cell responses are inhibited, HSV-1 can reactivate, and these recurrent reactivation events can lead to blinding scarring of the cornea. In the C57BL/6 mouse, CD8+ T cells specific for the immunodominant epitope from glycoprotein B maintain functionality throughout latency, while CD8+ T cells specific for subdominant epitopes undergo functional impairment that is associated with the expression of the inhibitory checkpoint molecule programmed death 1 (PD-1). Here, we investigate the checkpoint molecule T cell immunoglobulin and mucin domain-containing 3 (Tim-3), which has traditionally been associated with CD8+ T cell exhaustion. Unexpectedly, we found that Tim-3 was preferentially expressed on highly functional ganglionic CD8+ T cells during acute and latent HSV-1 infection. This, paired with data that show that Tim-3 expression on CD8+ T cells in the latently infected TG is influenced by viral gene expression, suggests that Tim-3 is an indicator of recent T cell stimulation, rather than functional compromise, in this model. We conclude that Tim-3 expression is not sufficient to define functional compromise during latency; however, it may be useful in identifying activated cells within the TG during HSV-1 infection.IMPORTANCE Without an effective means of eliminating HSV-1 from latently infected neurons, efforts to control the virus have centered on preventing viral reactivation from latency. Virus-specific CD8+ T cells within the infected TG have been shown to play a crucial role in inhibiting viral reactivation, and with a portion of these cells exhibiting functional impairment, checkpoint molecule immunotherapies have presented a potential solution to enhancing the antiviral response of these cells. In pursuing this potential treatment strategy, we found that Tim-3 (often associated with CD8+ T cell functional exhaustion) is not upregulated on impaired cells but instead is upregulated on highly functional cells that have recently received antigenic stimulation. These findings support a role for Tim-3 as a marker of activation rather than exhaustion in this model, and we provide additional evidence for the hypothesis that there is persistent viral gene expression in the HSV-1 latently infected TG.
Collapse
|
11
|
Toor SM, Sasidharan Nair V, Pfister G, Elkord E. Effect of pembrolizumab on CD4 + CD25 + , CD4 + LAP + and CD4 + TIM-3 + T cell subsets. Clin Exp Immunol 2019; 196:345-352. [PMID: 30693485 PMCID: PMC6514373 DOI: 10.1111/cei.13264] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/15/2019] [Indexed: 12/29/2022] Open
Abstract
Tumor immune evasion involves the expansion of avidly proliferating immunosuppressive cells and inhibition of effector T cell proliferation. Immune checkpoints (IC) block the activation pathways of tumor-reactive T cells. IC pathways are often exploited by tumor cells to evade immune destruction, and blocking these pathways through IC inhibitors (ICI) has shown promising results in multiple malignancies. In this study, we investigated the effects of an ICI, pembrolizumab, on various T cell subsets in vitro. We compared the suppressive activity of CD4+ CD25+ regulatory T cells (conventional Treg ) with T cells expressing T cell immunoglobulin-3+ (TIM-3+ ) and latency-associated peptide (LAP)+ T cells. We found that LAP-expressing T cells were more suppressive than conventional Treg , but TIM-3-expressing T cells were not suppressive. Our results show that pembrolizumab does not modulate functions of Treg and mediates its immunostimulatory effects via the release of effector T cells from suppression. These findings may assist in the development of agents designed to intervene in IC pathways to overcome Treg resistance to ICI.
Collapse
Affiliation(s)
- S. M. Toor
- Cancer Research CenterQatar Biomedical Research Institute, Hamad Bin Khalifa University, Qatar FoundationDohaQatar
| | - V. Sasidharan Nair
- Cancer Research CenterQatar Biomedical Research Institute, Hamad Bin Khalifa University, Qatar FoundationDohaQatar
| | - G. Pfister
- Flow Cytometry CoreQatar Biomedical Research Institute, Hamad Bin Khalifa University, Qatar FoundationDohaQatar
| | - E. Elkord
- Cancer Research CenterQatar Biomedical Research Institute, Hamad Bin Khalifa University, Qatar FoundationDohaQatar
- Institute of Cancer SciencesUniversity of ManchesterManchesterUK
| |
Collapse
|
12
|
Abstract
T-cell immunoglobulin and mucin domain 3 (Tim-3) is a transmembrane protein that in both mice and humans has been shown to possess various functions in a context-dependent manner. Thus, Tim-3 has been associated with both inhibitory and co-stimulatory function, depending in part on the specific cell type and immune response course. Though originally described on T cells, Tim-3 is now known to be expressed by both lymphoid and non-lymphoid cells within the immune system and even by non-immune cells. In addition, though widely thought of as a negative regulator of immunity, Tim-3 has been shown in more recent studies to have a positive function on both myeloid and lymphoid cells, including T cells. Tim-3 is often expressed at a high level on exhausted T cells in tumors and chronic infection and may engage in crosstalk with other so-called "checkpoint" molecules such as PD-1. Thus, Tim-3 has emerged as a possible therapeutic target, which is being actively explored both pre-clinically and clinically. However, recent research suggests a more complex in vivo role for this protein, compared with other targets in this area.
Collapse
Affiliation(s)
- Hridesh Banerjee
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15261, USA
| | - Lawrence P Kane
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15261, USA
| |
Collapse
|