1
|
Srinivasan S, Mishra S, Fan KKH, Wang L, Im J, Segura C, Mukherjee N, Huang G, Rao M, Ma C, Zhang N. Age-Dependent Bi-Phasic Dynamics of Ly49 +CD8 + Regulatory T Cell Population. Aging Cell 2024:e14461. [PMID: 39696807 DOI: 10.1111/acel.14461] [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: 06/07/2024] [Revised: 12/04/2024] [Accepted: 12/07/2024] [Indexed: 12/20/2024] Open
Abstract
Aging is tightly associated with reduced immune protection but increased risk of autoimmunity and inflammatory conditions. Regulatory T cells are one of the key cells to maintaining immune homeostasis. The age-dependent changes in CD4+Foxp3+ regulatory T cells (Tregs) have been well documented. However, the nonredundant Foxp3-CD8+ Tregs were never examined in the context of aging. This study first established clear distinctions between phenotypically overlapping CD8+ Tregs and virtual memory T cells. Then, we elucidated the dynamics of CD8+ Tregs across the lifespan in mice and further extended our investigation to human peripheral blood mononuclear cells (PBMCs). In mice, we discovered a bi-phasic dynamic shift in the frequency of CD8+CD44hiCD122hiLy49+ Tregs, with a steady increase in young adults and a notable peak in middle age followed by a decline in older mice. Transcriptomic analysis revealed that mouse CD8+ Tregs upregulated a selected set of natural killer (NK) cell-associated genes, including NKG2D, with age. Importantly, NKG2D might negatively regulate CD8+ Tregs. Additionally, by analyzing a scRNA-seq dataset of human PBMC, we found a distinct CD8+ Treg-like subset (Cluster 10) with comparable age-dependent frequency changes and gene expression, suggesting a conserved aging pattern in CD8+ Treg across mice and humans. In summary, our findings highlight the importance of CD8+ Tregs in immune regulation and aging.
Collapse
Affiliation(s)
- Saranya Srinivasan
- Department of Microbiology, Immunology and Molecular Genetics, Long School of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA
| | - Shruti Mishra
- Department of Microbiology, Immunology and Molecular Genetics, Long School of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA
- Gilead Sciences Inc, California, USA
| | - Kenneth Ka-Ho Fan
- Department of Microbiology, Immunology and Molecular Genetics, Long School of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA
| | - Liwen Wang
- Department of Microbiology, Immunology and Molecular Genetics, Long School of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA
- Department of Hematology, Third Xiangya Hospital, Central South University, Changsha, Hunan Province, China
| | - John Im
- Department of Microbiology, Immunology and Molecular Genetics, Long School of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA
| | - Courtney Segura
- Department of Microbiology, Immunology and Molecular Genetics, Long School of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA
| | - Neelam Mukherjee
- Department of Urology, University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA
| | - Gang Huang
- Department of Cell Systems and Anatomy, Greehey Children's Cancer Research Institute, San Antonio, Texas, USA
| | - Manjeet Rao
- Department of Cell Systems and Anatomy, Greehey Children's Cancer Research Institute, San Antonio, Texas, USA
| | - Chaoyu Ma
- Department of Microbiology, Immunology and Molecular Genetics, Long School of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA
| | - Nu Zhang
- Department of Microbiology, Immunology and Molecular Genetics, Long School of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA
- South Texas Veterans Health Care System, San Antonio, Texas, USA
| |
Collapse
|
2
|
Im SJ, Lee K, Ha SJ. Harnessing IL-2 for immunotherapy against cancer and chronic infection: a historical perspective and emerging trends. Exp Mol Med 2024; 56:1900-1908. [PMID: 39218982 PMCID: PMC11447265 DOI: 10.1038/s12276-024-01301-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2024] [Revised: 05/19/2024] [Accepted: 06/11/2024] [Indexed: 09/04/2024] Open
Abstract
IL-2 therapy, which enhances the function of CD8 + T cells, was initially employed as the cornerstone of immunotherapy against cancer. However, the impact of this therapy extends beyond CD8 + T cells to cells expressing IL-2R, such as endothelial cells and regulatory T cells (Tregs), resulting in various side effects. Consequently, IL-2 therapy has taken a step back from the forefront of treatment. Immune checkpoint inhibitors (ICIs), such as anti-PD-1/PD-L1 antibodies and CTLA-4 antibodies, are used because of their durable therapeutic responses and the reduced incidence of side effects. Nevertheless, only a small fraction of cancer patients respond to ICIs, and research on IL-2 as a combination treatment to improve the efficacy of these ICIs is ongoing. To mitigate side effects, efforts have focused on developing IL-2 variants that do not strongly bind to cells expressing IL-2Rα and favor signaling through IL-2Rβγ. However, recent studies have suggested that, in the context of persistent antigen stimulation models, effective stimulation of antigen-specific exhausted CD8 + T cells in combination with PD-1 inhibitors requires either 1) binding to IL-2Rα or 2) delivery via a fusion with PD-1. This review explores the historical context of IL-2 as an immunotherapeutic agent and discusses future directions for its use in cancer immunotherapy.
Collapse
Affiliation(s)
- Se Jin Im
- Department of Immunology, Sungkyunkwan University School of Medicine, Suwon, Korea.
| | - Kyungmin Lee
- Department of Immunology, Sungkyunkwan University School of Medicine, Suwon, Korea
| | - Sang-Jun Ha
- Department of Biochemistry, College of Life Science & Biotechnology, Yonsei University, Seoul, Korea.
| |
Collapse
|
3
|
Watson TK, Rosen ABI, Drow T, Medjo JA, MacQuivey MA, Ge Y, Liggitt HD, Grosvenor DA, Dill-McFarland KA, Altman MC, Concannon PJ, Buckner JH, Rawlings DJ, Allenspach EJ. Reduced function of the adaptor SH2B3 promotes T1D via altered gc cytokine-regulated, T cell intrinsic immune tolerance. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.02.606362. [PMID: 39211124 PMCID: PMC11361092 DOI: 10.1101/2024.08.02.606362] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
Genome-wide association studies have identified SH2B3 as an important non-MHC gene for islet autoimmunity and type 1 diabetes (T1D). In this study, we found a single SH2B3 haplotype significantly associated with increased risk for human T1D, and this haplotype carries the single nucleotide variant rs3184504*T in SH2B3. To better characterize the role of SH2B3 in T1D, we used mouse modeling and found a T cell-intrinsic role for SH2B3 regulating peripheral tolerance. SH2B3 deficiency had minimal effect on TCR signaling or proliferation across antigen doses, yet enhanced cell survival and cytokine signaling including common gamma chain-dependent and interferon-gamma receptor signaling. SH2B3 deficient CD8+T cells showed augmented STAT5-MYC and effector-related gene expression partially reversed with blocking autocrine IL-2 in culture. Using the RIP-mOVA model, we found CD8+ T cells lacking SH2B3 promoted early islet destruction and diabetes without requiring CD4+ T cell help. SH2B3-deficient cells demonstrated increased survival post-transfer compared to control cells despite a similar proliferation profile in the same host. Next, we created a spontaneous NOD .Sh2b3 -/- mouse model and found markedly increased incidence and accelerated T1D across sexes. Collectively, these studies identify SH2B3 as a critical mediator of peripheral T cell tolerance limiting the T cell response to self-antigens. Article Highlights The rs3184504 polymorphism, encoding a hypomorphic variant of the negative regulator SH2B3, strongly associates with T1D.SH2B3 deficiency results in hypersensitivity to cytokines, including IL-2, in murine CD4+ and CD8+ T cells.SH2B3 deficient CD8+ T cells exhibit a comparable transcriptome to wild-type CD8+ T cells at baseline, but upon antigen stimulation SH2B3 deficient cells upregulate genes characteristic of enhanced JAK/STAT signaling and effector functions.We found a T-cell intrinsic role of SH2B3 leading to severe islet destruction in an adoptive transfer murine T1D model, while global SH2B3 deficiency accelerated spontaneous NOD diabetes across sexes.
Collapse
|
4
|
Janusova S, Paprckova D, Michalik J, Uleri V, Drobek A, Salyova E, Chorfi L, Neuwirth A, Andreyeva A, Prochazka J, Sedlacek R, Draber P, Stepanek O. ABIN1 is a negative regulator of effector functions in cytotoxic T cells. EMBO Rep 2024; 25:3456-3485. [PMID: 38877170 PMCID: PMC11315980 DOI: 10.1038/s44319-024-00179-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2023] [Revised: 05/31/2024] [Accepted: 06/06/2024] [Indexed: 06/16/2024] Open
Abstract
T cells are pivotal in the adaptive immune defense, necessitating a delicate balance between robust response against infections and self-tolerance. Their activation involves intricate cross-talk among signaling pathways triggered by the T-cell antigen receptors (TCR) and co-stimulatory or inhibitory receptors. The molecular regulation of these complex signaling networks is still incompletely understood. Here, we identify the adaptor protein ABIN1 as a component of the signaling complexes of GITR and OX40 co-stimulation receptors. T cells lacking ABIN1 are hyper-responsive ex vivo, exhibit enhanced responses to cognate infections, and superior ability to induce experimental autoimmune diabetes in mice. ABIN1 negatively regulates p38 kinase activation and late NF-κB target genes. P38 is at least partially responsible for the upregulation of the key effector proteins IFNG and GZMB in ABIN1-deficient T cells after TCR stimulation. Our findings reveal the intricate role of ABIN1 in T-cell regulation.
Collapse
MESH Headings
- Animals
- Humans
- Mice
- Adaptor Proteins, Signal Transducing/metabolism
- Adaptor Proteins, Signal Transducing/genetics
- Diabetes Mellitus, Type 1/immunology
- Diabetes Mellitus, Type 1/genetics
- Diabetes Mellitus, Type 1/metabolism
- Glucocorticoid-Induced TNFR-Related Protein
- Interferon-gamma/metabolism
- Lymphocyte Activation/immunology
- Lymphocyte Activation/genetics
- Mice, Inbred C57BL
- Mice, Knockout
- NF-kappa B/metabolism
- p38 Mitogen-Activated Protein Kinases/metabolism
- Receptors, Antigen, T-Cell/metabolism
- Receptors, OX40/metabolism
- Receptors, OX40/genetics
- Signal Transduction
- T-Lymphocytes, Cytotoxic/immunology
- T-Lymphocytes, Cytotoxic/metabolism
Collapse
Affiliation(s)
- Sarka Janusova
- Laboratory of Adaptive Immunity, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czech Republic
- Faculty of Science, Charles University in Prague, Prague, Czech Republic
| | - Darina Paprckova
- Laboratory of Adaptive Immunity, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czech Republic
| | - Juraj Michalik
- Laboratory of Adaptive Immunity, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czech Republic
| | - Valeria Uleri
- Laboratory of Adaptive Immunity, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czech Republic
- Faculty of Science, Charles University in Prague, Prague, Czech Republic
| | - Ales Drobek
- Laboratory of Adaptive Immunity, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czech Republic
| | - Eva Salyova
- Laboratory of Adaptive Immunity, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czech Republic
| | - Louise Chorfi
- Laboratory of Adaptive Immunity, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czech Republic
| | - Ales Neuwirth
- Laboratory of Adaptive Immunity, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czech Republic
| | - Arina Andreyeva
- Laboratory of Adaptive Immunity, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czech Republic
| | - Jan Prochazka
- Czech Centre for Phenogenomics, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czech Republic
| | - Radislav Sedlacek
- Czech Centre for Phenogenomics, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czech Republic
| | - Peter Draber
- Laboratory of Adaptive Immunity, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czech Republic
- Laboratory of Immunity & Cell Communication, Division BIOCEV, First Faculty of Medicine, Charles University, Vestec, Czech Republic
| | - Ondrej Stepanek
- Laboratory of Adaptive Immunity, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czech Republic.
| |
Collapse
|
5
|
Goodspeed A, Bodlak A, Duffy AB, Nelson-Taylor S, Oike N, Porfilio T, Shirai R, Walker D, Treece A, Black J, Donaldson N, Cost C, Garrington T, Greffe B, Luna-Fineman S, Demedis J, Lake J, Danis E, Verneris M, Adams DL, Hayashi M. Characterization of transcriptional heterogeneity and novel therapeutic targets using single cell RNA-sequencing of primary and circulating Ewing sarcoma cells. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.18.576251. [PMID: 38293103 PMCID: PMC10827204 DOI: 10.1101/2024.01.18.576251] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/01/2024]
Abstract
Ewing sarcoma is the second most common bone cancer in children, accounting for 2% of pediatric cancer diagnoses. Patients who present with metastatic disease at the time of diagnosis have a dismal prognosis, compared to the >70% 5-year survival of those with localized disease. Here, we utilized single cell RNA-sequencing to characterize the transcriptional landscape of primary Ewing sarcoma tumors and surrounding tumor microenvironment (TME). Copy-number analysis identified subclonal evolution within patients prior to treatment. Primary tumor samples demonstrate a heterogenous transcriptional landscape with several conserved gene expression programs, including those composed of genes related to proliferation and EWS targets. Single cell RNA-sequencing and immunofluorescence of circulating tumor cells at the time of diagnosis identified TSPAN8 as a novel therapeutic target.
Collapse
|
6
|
Zhou L, Velegraki M, Wang Y, Mandula JK, Chang Y, Liu W, Song NJ, Kwon H, Xiao T, Bolyard C, Hong F, Xin G, Ma Q, Rubinstein MP, Wen H, Li Z. Spatial and functional targeting of intratumoral Tregs reverses CD8+ T cell exhaustion and promotes cancer immunotherapy. J Clin Invest 2024; 134:e180080. [PMID: 38787791 PMCID: PMC11245154 DOI: 10.1172/jci180080] [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: 02/07/2024] [Accepted: 05/17/2024] [Indexed: 05/26/2024] Open
Abstract
Intratumoral Tregs are key mediators of cancer immunotherapy resistance, including anti-programmed cell death (ligand) 1 [anti-PD-(L)1] immune checkpoint blockade (ICB). The mechanisms driving Treg infiltration into the tumor microenvironment (TME) and the consequence on CD8+ T cell exhaustion remain elusive. Here, we report that heat shock protein gp96 (also known as GRP94) was indispensable for Treg tumor infiltration, primarily through the roles of gp96 in chaperoning integrins. Among various gp96-dependent integrins, we found that only LFA-1 (αL integrin), and not αV, CD103 (αE), or β7 integrin, was required for Treg tumor homing. Loss of Treg infiltration into the TME by genetic deletion of gp96/LFA-1 potently induced rejection of tumors in multiple ICB-resistant murine cancer models in a CD8+ T cell-dependent manner, without loss of self-tolerance. Moreover, gp96 deletion impeded Treg activation primarily by suppressing IL-2/STAT5 signaling, which also contributed to tumor regression. By competing for intratumoral IL-2, Tregs prevented the activation of CD8+ tumor-infiltrating lymphocytes, drove thymocyte selection-associated high mobility group box protein (TOX) induction, and induced bona fide CD8+ T cell exhaustion. By contrast, Treg ablation led to striking CD8+ T cell activation without TOX induction, demonstrating clear uncoupling of the 2 processes. Our study reveals that the gp96/LFA-1 axis plays a fundamental role in Treg biology and suggests that Treg-specific gp96/LFA-1 targeting represents a valuable strategy for cancer immunotherapy without inflicting autoinflammatory conditions.
Collapse
Affiliation(s)
- Lei Zhou
- Pelotonia Institute for Immuno-Oncology (PIIO), The Ohio State University Comprehensive Cancer Center – James Cancer Hospital and Solove Research Institute (OSUCCC), Columbus, Ohio, USA
- Department of Anesthesiology, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Maria Velegraki
- Pelotonia Institute for Immuno-Oncology (PIIO), The Ohio State University Comprehensive Cancer Center – James Cancer Hospital and Solove Research Institute (OSUCCC), Columbus, Ohio, USA
| | - Yi Wang
- Pelotonia Institute for Immuno-Oncology (PIIO), The Ohio State University Comprehensive Cancer Center – James Cancer Hospital and Solove Research Institute (OSUCCC), Columbus, Ohio, USA
- Molecular, Cellular and Developmental Biology Graduate Program, Ohio State University, Columbus, Ohio, USA
| | - J K Mandula
- Pelotonia Institute for Immuno-Oncology (PIIO), The Ohio State University Comprehensive Cancer Center – James Cancer Hospital and Solove Research Institute (OSUCCC), Columbus, Ohio, USA
| | - Yuzhou Chang
- Pelotonia Institute for Immuno-Oncology (PIIO), The Ohio State University Comprehensive Cancer Center – James Cancer Hospital and Solove Research Institute (OSUCCC), Columbus, Ohio, USA
- Department of Biomedical Informatics
| | - Weiwei Liu
- Pelotonia Institute for Immuno-Oncology (PIIO), The Ohio State University Comprehensive Cancer Center – James Cancer Hospital and Solove Research Institute (OSUCCC), Columbus, Ohio, USA
- Molecular, Cellular and Developmental Biology Graduate Program, Ohio State University, Columbus, Ohio, USA
| | - No-Joon Song
- Pelotonia Institute for Immuno-Oncology (PIIO), The Ohio State University Comprehensive Cancer Center – James Cancer Hospital and Solove Research Institute (OSUCCC), Columbus, Ohio, USA
| | - Hyunwoo Kwon
- Pelotonia Institute for Immuno-Oncology (PIIO), The Ohio State University Comprehensive Cancer Center – James Cancer Hospital and Solove Research Institute (OSUCCC), Columbus, Ohio, USA
- Department of Internal Medicine, Ohio State University College of Medicine, Columbus, USA
| | - Tong Xiao
- Pelotonia Institute for Immuno-Oncology (PIIO), The Ohio State University Comprehensive Cancer Center – James Cancer Hospital and Solove Research Institute (OSUCCC), Columbus, Ohio, USA
- Molecular, Cellular and Developmental Biology Graduate Program, Ohio State University, Columbus, Ohio, USA
| | - Chelsea Bolyard
- Pelotonia Institute for Immuno-Oncology (PIIO), The Ohio State University Comprehensive Cancer Center – James Cancer Hospital and Solove Research Institute (OSUCCC), Columbus, Ohio, USA
| | - Feng Hong
- Pelotonia Institute for Immuno-Oncology (PIIO), The Ohio State University Comprehensive Cancer Center – James Cancer Hospital and Solove Research Institute (OSUCCC), Columbus, Ohio, USA
- Division of Medical Oncology, Department of Internal Medicine, Ohio State University Comprehensive Cancer Center, Columbus, USA
| | - Gang Xin
- Pelotonia Institute for Immuno-Oncology (PIIO), The Ohio State University Comprehensive Cancer Center – James Cancer Hospital and Solove Research Institute (OSUCCC), Columbus, Ohio, USA
- Department of Microbial Infection and Immunity, Ohio State University College of Medicine, Columbus, USA
| | - Qin Ma
- Pelotonia Institute for Immuno-Oncology (PIIO), The Ohio State University Comprehensive Cancer Center – James Cancer Hospital and Solove Research Institute (OSUCCC), Columbus, Ohio, USA
- Department of Biomedical Informatics
| | - Mark P. Rubinstein
- Pelotonia Institute for Immuno-Oncology (PIIO), The Ohio State University Comprehensive Cancer Center – James Cancer Hospital and Solove Research Institute (OSUCCC), Columbus, Ohio, USA
- Division of Medical Oncology, Department of Internal Medicine, Ohio State University Comprehensive Cancer Center, Columbus, USA
| | - Haitao Wen
- Pelotonia Institute for Immuno-Oncology (PIIO), The Ohio State University Comprehensive Cancer Center – James Cancer Hospital and Solove Research Institute (OSUCCC), Columbus, Ohio, USA
- Department of Microbial Infection and Immunity, Ohio State University College of Medicine, Columbus, USA
| | - Zihai Li
- Pelotonia Institute for Immuno-Oncology (PIIO), The Ohio State University Comprehensive Cancer Center – James Cancer Hospital and Solove Research Institute (OSUCCC), Columbus, Ohio, USA
- Division of Medical Oncology, Department of Internal Medicine, Ohio State University Comprehensive Cancer Center, Columbus, USA
| |
Collapse
|
7
|
Mehta P, Sanz-Magallón Duque de Estrada B, Denneny EK, Foster K, Turner CT, Mayer A, Milighetti M, Platé M, Worlock KB, Yoshida M, Brown JS, Nikolić MZ, Chain BM, Noursadeghi M, Chambers RC, Porter JC, Tomlinson GS. Single-cell analysis of bronchoalveolar cells in inflammatory and fibrotic post-COVID lung disease. Front Immunol 2024; 15:1372658. [PMID: 38827740 PMCID: PMC11140060 DOI: 10.3389/fimmu.2024.1372658] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Accepted: 05/07/2024] [Indexed: 06/04/2024] Open
Abstract
Background Persistent radiological lung abnormalities are evident in many survivors of acute coronavirus disease 2019 (COVID-19). Consolidation and ground glass opacities are interpreted to indicate subacute inflammation whereas reticulation is thought to reflect fibrosis. We sought to identify differences at molecular and cellular level, in the local immunopathology of post-COVID inflammation and fibrosis. Methods We compared single-cell transcriptomic profiles and T cell receptor (TCR) repertoires of bronchoalveolar cells obtained from convalescent individuals with each radiological pattern, targeting lung segments affected by the predominant abnormality. Results CD4 central memory T cells and CD8 effector memory T cells were significantly more abundant in those with inflammatory radiology. Clustering of similar TCRs from multiple donors was a striking feature of both phenotypes, consistent with tissue localised antigen-specific immune responses. There was no enrichment for known SARS-CoV-2-reactive TCRs, raising the possibility of T cell-mediated immunopathology driven by failure in immune self-tolerance. Conclusions Post-COVID radiological inflammation and fibrosis show evidence of shared antigen-specific T cell responses, suggesting a role for therapies targeting T cells in limiting post-COVID lung damage.
Collapse
Affiliation(s)
- Puja Mehta
- UCL Respiratory, University College London, London, United Kingdom
| | | | - Emma K. Denneny
- UCL Respiratory, University College London, London, United Kingdom
| | - Kane Foster
- UCL Cancer Institute, University College London, London, United Kingdom
| | - Carolin T. Turner
- Division of Infection and Immunity, University College London, London, United Kingdom
| | - Andreas Mayer
- Division of Infection and Immunity, University College London, London, United Kingdom
| | - Martina Milighetti
- Division of Infection and Immunity, University College London, London, United Kingdom
| | - Manuela Platé
- UCL Respiratory, University College London, London, United Kingdom
| | | | - Masahiro Yoshida
- UCL Respiratory, University College London, London, United Kingdom
| | - Jeremy S. Brown
- UCL Respiratory, University College London, London, United Kingdom
| | - Marko Z. Nikolić
- UCL Respiratory, University College London, London, United Kingdom
| | - Benjamin M. Chain
- Division of Infection and Immunity, University College London, London, United Kingdom
| | - Mahdad Noursadeghi
- Division of Infection and Immunity, University College London, London, United Kingdom
| | | | - Joanna C. Porter
- UCL Respiratory, University College London, London, United Kingdom
| | - Gillian S. Tomlinson
- Division of Infection and Immunity, University College London, London, United Kingdom
| |
Collapse
|
8
|
Charan M, Jones TH, Ahirwar DK, Acharya N, Subramaniam VV, Ganju RK, Song JW. Induced electric fields inhibit breast cancer growth and metastasis by modulating the immune tumor microenvironment. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.14.589256. [PMID: 38659909 PMCID: PMC11042207 DOI: 10.1101/2024.04.14.589256] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/26/2024]
Abstract
Despite tremendous advances in oncology, metastatic triple-negative breast cancer remains difficult to treat and manage with established therapies. Here, we show in mice with orthotopic triple-negative breast tumors that alternating (100 kHz), and low intensity (<1 mV/cm) induced electric fields (iEFs) significantly reduced primary tumor growth and distant lung metastases. Non-contact iEF treatment can be delivered safely and non-invasively in vivo via a hollow, rectangular solenoid coil. We discovered that iEF treatment enhances anti-tumor immune responses at both the primary breast and secondary lung sites. In addition, iEF reduces immunosuppressive TME by reducing effector CD8+ T cell exhaustion and the infiltration of immunosuppressive immune cells. Furthermore, iEF treatment reduced lung metastasis by increasing CD8+ T cells and reducing immunosuppressive Gr1+ neutrophils in the lung microenvironment. We also observed that iEFs reduced the metastatic potential of cancer cells by inhibiting epithelial-to-mesenchymal transition. By introducing a non-invasive and non-toxic electrotherapeutic for inhibiting metastatic outgrowth and enhancing anti-tumor immune response in vivo, treatment with iEF technology could add to a paradigm-shifting strategy for cancer therapy.
Collapse
|
9
|
Yu J, Gadwa J, Ross RB, Knitz M, Darragh LB, Abdelazeem KNM, Beynor J, Neupert B, Nguyen A, Nguyen D, Olimpo N, Corbo S, Van Court B, D'Alessandro A, Saviola A, Karam SD. IL7 in combination with radiotherapy stimulates a memory T-cell response to improve outcomes in HNSCC models. Cancer Immunol Immunother 2024; 73:90. [PMID: 38554147 PMCID: PMC10981637 DOI: 10.1007/s00262-024-03664-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Accepted: 02/27/2024] [Indexed: 04/01/2024]
Abstract
Clinically approved head and neck squamous cell carcinoma (HNSCC) immunotherapies manipulate the immune checkpoint blockade (ICB) axis but have had limited success outside of recurrent/metastatic disease. Interleukin-7 (IL7) has been shown to be essential for effector T-cell survival, activation, and proliferation. Here, we show that IL7 in combination with radiotherapy (RT) is effective in activating CD8 + T-cells for reducing tumor growth. Our studies were conducted using both human papillomavirus related and unrelated orthotopic HNSCC murine models. Immune populations from the tumor, draining lymph nodes, and blood were compared between treatment groups and controls using flow cytometry, proteomics, immunofluorescence staining, and RNA sequencing. Treatment with RT and IL7 (RT + IL7) resulted in significant tumor growth reduction, high CD8 T-cell tumor infiltration, and increased proliferation of T-cell progenitors in the bone marrow. IL7 also expanded a memory-like subpopulation of CD8 T-cells. These results indicate that IL7 in combination with RT can serve as an effective immunotherapy strategy outside of the conventional ICB axis to drive the antitumor activity of CD8 T-cells.
Collapse
Affiliation(s)
- Justin Yu
- Department of Otolaryngology - Head and Neck Surgery, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - Jacob Gadwa
- Department of Radiation Oncology, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
- Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - Richard B Ross
- Department of Radiation Oncology, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - Michael Knitz
- Department of Radiation Oncology, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - Laurel B Darragh
- Department of Radiation Oncology, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
- Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - Khalid N M Abdelazeem
- Department of Radiation Oncology, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
- Radiation Biology Research Department, National Center for Radiation Research and Technology, Egyptian Atomic Energy Authority, Cairo, Egypt
| | - Jessica Beynor
- Department of Radiation Oncology, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
- Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - Brooke Neupert
- Department of Radiation Oncology, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - Alexander Nguyen
- Department of Radiation Oncology, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - Diemmy Nguyen
- Department of Radiation Oncology, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - Nicholas Olimpo
- Department of Radiation Oncology, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - Sophia Corbo
- Department of Radiation Oncology, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - Benjamin Van Court
- Department of Radiation Oncology, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - Angelo D'Alessandro
- Department of Biochemistry and Molecular Genetics, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - Anthony Saviola
- Department of Biochemistry and Molecular Genetics, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - Sana D Karam
- Department of Radiation Oncology, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA.
- Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA.
| |
Collapse
|
10
|
Ren JY, Xu M, Niu XD, Ma SX, Jiao YJ, Wang D, Yu M, Cai H. Systemic inflammatory response index is a predictor of prognosis in gastric cancer patients: Retrospective cohort and meta-analysis. World J Gastrointest Surg 2024; 16:382-395. [PMID: 38463377 PMCID: PMC10921201 DOI: 10.4240/wjgs.v16.i2.382] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Revised: 12/25/2023] [Accepted: 02/01/2024] [Indexed: 02/25/2024] Open
Abstract
BACKGROUND The systemic inflammatory response index (SIRI) has been demonstrated to make a significant difference in assessing the prognosis of patients with different solid neoplasms. However, research is needed to ascertain the accuracy and reliability of applying the SIRI to patients who undergo robotic radical gastric cancer surgery. AIM To validate the applicability of the SIRI in assessing the survival of gastric cancer patients and evaluate the clinical contribution of preoperative SIRI levels to predicting long-term tumor outcomes in patients, who received robotic radical gastric cancer surgery. METHODS Initially, an exhaustive retrieval was performed in the PubMed, the Cochrane Library, EMBASE, Web of Science, and Scopus databases to identify relevant studies. Subsequently, a meta-analysis was executed on 6 cohort studies identifying the value of the SIRI in assessing the survival of gastric cancer patients. Additionally, the clinical data of 161 patients undergoing robotic radical gastric cancer surgery were retrospectively analyzed to evaluate their clinicopathological characteristics and relevant laboratory indicators. The association between preoperative SIRI levels and 5-year overall survival (OS) and disease-free survival (DFS) was assessed. RESULTS The findings demonstrated an extensive connection between SIRI values and the outcome of patients with gastric cancer. Preoperative SIRI levels were identified as an independent hazard feature for both OS and DFS among those who received robotic surgery for gastric cancer. SIRI levels in gastric cancer patients were observed to be associated with the presence of comorbidities, T-stage, carcinoembryonic antigen levels, the development of early serious postoperative complications, and the rate of lymph node metastasis. CONCLUSION SIRI values are correlated with adverse in the gastric cancer population and have the potential to be utilized in predicting long-term oncological survival in patients who undergo robotic radical gastric cancer surgery.
Collapse
Affiliation(s)
- Jing-Yao Ren
- School of Clinical Medicine, Ningxia Medical University, Yinchuan 750000, Ningxia Hui autonomous region, China
| | - Meng Xu
- Gansu Provincial Hospital, Gansu Provincial Hospital, Lanzhou 730000, Gansu Province, China
| | - Xiang-Dong Niu
- General Surgery Clinical Medical Center, Gansu Provincial Hospital, Lanzhou 730000, Gansu Province, China
| | - Shi-Xun Ma
- General Surgery Clinical Medical Center, Gansu Provincial Hospital, Lanzhou 730000, Gansu Province, China
| | - Ya-Jun Jiao
- General Surgery Clinical Medical Center, Gansu Provincial Hospital, Lanzhou 730000, Gansu Province, China
| | - Da Wang
- Medical College of Jiangsu University, Jiangsu University, Zhenjiang 212013, Jiangsu Province, China
| | - Miao Yu
- Key Laboratory of Molecular Diagnostics and Precision Medicine for Surgical Oncology in Gansu Province, Gansu Provincial Hospital, Lanzhou 730000, Gansu Province, China
| | - Hui Cai
- General Surgery Clinical Medical Center, Gansu Provincial Hospital, Lanzhou 730000, Gansu Province, China
| |
Collapse
|
11
|
Sprent J, Boyman O. Optimising IL-2 for Cancer Immunotherapy. Immune Netw 2024; 24:e5. [PMID: 38455463 PMCID: PMC10917570 DOI: 10.4110/in.2024.24.e5] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Revised: 01/01/2024] [Accepted: 01/08/2024] [Indexed: 03/09/2024] Open
Abstract
The key role of T cells in cancer immunotherapy is well established and is highlighted by the remarkable capacity of Ab-mediated checkpoint blockade to overcome T-cell exhaustion and amplify anti-tumor responses. However, total or partial tumor remission following checkpoint blockade is still limited to only a few types of tumors. Hence, concerted attempts are being made to devise new methods for improving tumor immunity. Currently, much attention is being focused on therapy with IL-2. This cytokine is a powerful growth factor for T cells and optimises their effector functions. When used at therapeutic doses for cancer treatment, however, IL-2 is highly toxic. Nevertheless, recent work has shown that modifying the structure or presentation of IL-2 can reduce toxicity and lead to effective anti-tumor responses in synergy with checkpoint blockade. Here, we review the complex interaction of IL-2 with T cells: first during normal homeostasis, then during responses to pathogens, and finally in anti-tumor responses.
Collapse
Affiliation(s)
- Jonathan Sprent
- Immunology Division, Garvan Institute of Medical Research, Darlinghurst 2010, Australia
- St. Vincent’s Clinical School, University of New South Wales, Sydney 1466, Australia
- Menzies Institute of Medical Research, Hobart 7000, Australia
| | - Onur Boyman
- Department of Immunology, University Hospital Zurich, Zurich 8091, Switzerland
- Faculty of Medicine and Faculty of Science, University of Zurich, Zurich 8057, Switzerland
| |
Collapse
|
12
|
Vancikova S, Benešová I, Černý V, Lázničková P, Miková E, Šmídová B, Věcek J, Zuani MD, Niederlova V. Czech Young Immunologists: Bound by science, united in purpose. Eur J Immunol 2024; 54:e2350737. [PMID: 37767759 DOI: 10.1002/eji.202350737] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Revised: 09/27/2023] [Accepted: 09/27/2023] [Indexed: 09/29/2023]
Affiliation(s)
- Sabina Vancikova
- Laboratory of Cellular and Molecular Immunology, Institute of Microbiology of the Czech Academy of Sciences, Prague, Czech Republic
| | - Iva Benešová
- Department of Immunology, Second Faculty of Medicine, Charles University and University Hospital Motol, Prague, Czech Republic
| | - Viktor Černý
- Institute of Immunology and Microbiology, First Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Petra Lázničková
- Cellular and Molecular Immunoregulation group, International Clinical Research Center, St. Anne's University Hospital, Brno, Czech Republic
| | - Eliška Miková
- Laboratory of Leukocyte Motility, First Faculty of Medicine, Charles University, Vestec, Czech Republic
| | - Barbora Šmídová
- Department of Parasitology, Faculty of Science, Charles University, Prague, Czech Republic
| | - Jan Věcek
- Institute of Immunology and Microbiology, First Faculty of Medicine, Charles University, Prague, Czech Republic
- Department of Cell Biology, Faculty of Science, Charles University, Prague, Czech Republic
| | - Marco De Zuani
- Cellular and Molecular Immunoregulation group, International Clinical Research Center, St. Anne's University Hospital, Brno, Czech Republic
| | - Veronika Niederlova
- Laboratory of Adaptive Immunity, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czech Republic
| |
Collapse
|
13
|
Niederlova V, Tsyklauri O, Kovar M, Stepanek O. IL-2-driven CD8 + T cell phenotypes: implications for immunotherapy. Trends Immunol 2023; 44:890-901. [PMID: 37827864 PMCID: PMC7615502 DOI: 10.1016/j.it.2023.09.003] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Revised: 09/11/2023] [Accepted: 09/12/2023] [Indexed: 10/14/2023]
Abstract
The therapeutic potential of interleukin (IL)-2 in cancer treatment has been known for decades, yet its widespread adoption in clinical practice remains limited. Recently, chimeric proteins of an anti-PD-1 antibody and suboptimal IL-2 variants were shown to stimulate potent antitumor and antiviral immunity by inducing unique effector CD8+ T cells in mice. A similar subset of cytotoxic T cells is induced by depletion of regulatory T cells (Tregs), suggesting IL-2 sequestration as a major mechanism through which regulatory T cells suppress activated CD8+ T cells. Here, we present our view of how IL-2-based biologicals can boost the antitumor response at a cellular level, and propose that the role of Tregs following such treatments may have been previously overestimated.
Collapse
Affiliation(s)
- Veronika Niederlova
- Laboratory of Adaptive Immunity, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czech Republic
| | - Oksana Tsyklauri
- Laboratory of Adaptive Immunity, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czech Republic
| | - Marek Kovar
- Laboratory of Tumor Immunology, Institute of Microbiology of the Czech Academy of Sciences, Prague, Czech Republic
| | - Ondrej Stepanek
- Laboratory of Adaptive Immunity, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czech Republic.
| |
Collapse
|
14
|
Tsyklauri O, Chadimova T, Niederlova V, Kovarova J, Michalik J, Malatova I, Janusova S, Ivashchenko O, Rossez H, Drobek A, Vecerova H, Galati V, Kovar M, Stepanek O. Regulatory T cells suppress the formation of potent KLRK1 and IL-7R expressing effector CD8 T cells by limiting IL-2. eLife 2023; 12:79342. [PMID: 36705564 PMCID: PMC9977273 DOI: 10.7554/elife.79342] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Accepted: 01/27/2023] [Indexed: 01/28/2023] Open
Abstract
Regulatory T cells (Tregs) are indispensable for maintaining self-tolerance by suppressing conventional T cells. On the other hand, Tregs promote tumor growth by inhibiting anticancer immunity. In this study, we identified that Tregs increase the quorum of self-reactive CD8+ T cells required for the induction of experimental autoimmune diabetes in mice. Their major suppression mechanism is limiting available IL-2, an essential T-cell cytokine. Specifically, Tregs inhibit the formation of a previously uncharacterized subset of antigen-stimulated KLRK1+ IL-7R+ (KILR) CD8+ effector T cells, which are distinct from conventional effector CD8+ T cells. KILR CD8+ T cells show superior cell-killing abilities in vivo. The administration of agonistic IL-2 immunocomplexes phenocopies the absence of Tregs, i.e., it induces KILR CD8+ T cells, promotes autoimmunity, and enhances antitumor responses in mice. Counterparts of KILR CD8+ T cells were found in the human blood, revealing them as a potential target for immunotherapy.
Collapse
Affiliation(s)
- Oksana Tsyklauri
- Laboratory of Adaptive Immunity, Institute of Molecular Genetics of the Czech Academy of SciencesPragueCzech Republic
- Faculty of Science, Charles UniversityPragueCzech Republic
| | - Tereza Chadimova
- Laboratory of Adaptive Immunity, Institute of Molecular Genetics of the Czech Academy of SciencesPragueCzech Republic
- Faculty of Science, Charles UniversityPragueCzech Republic
| | - Veronika Niederlova
- Laboratory of Adaptive Immunity, Institute of Molecular Genetics of the Czech Academy of SciencesPragueCzech Republic
- Faculty of Science, Charles UniversityPragueCzech Republic
| | - Jirina Kovarova
- Institute of Microbiology of the Czech Academy of SciencesPragueCzech Republic
| | - Juraj Michalik
- Laboratory of Adaptive Immunity, Institute of Molecular Genetics of the Czech Academy of SciencesPragueCzech Republic
| | - Iva Malatova
- Institute of Microbiology of the Czech Academy of SciencesPragueCzech Republic
| | - Sarka Janusova
- Laboratory of Adaptive Immunity, Institute of Molecular Genetics of the Czech Academy of SciencesPragueCzech Republic
| | - Olha Ivashchenko
- Laboratory of Adaptive Immunity, Institute of Molecular Genetics of the Czech Academy of SciencesPragueCzech Republic
| | - Helene Rossez
- Department of Biomedicine, University Hospital of BaselBaselSwitzerland
| | - Ales Drobek
- Laboratory of Adaptive Immunity, Institute of Molecular Genetics of the Czech Academy of SciencesPragueCzech Republic
| | - Hana Vecerova
- Laboratory of Adaptive Immunity, Institute of Molecular Genetics of the Czech Academy of SciencesPragueCzech Republic
| | - Virginie Galati
- Department of Biomedicine, University Hospital of BaselBaselSwitzerland
| | - Marek Kovar
- Institute of Microbiology of the Czech Academy of SciencesPragueCzech Republic
| | - Ondrej Stepanek
- Laboratory of Adaptive Immunity, Institute of Molecular Genetics of the Czech Academy of SciencesPragueCzech Republic
- Department of Biomedicine, University Hospital of BaselBaselSwitzerland
| |
Collapse
|