1
|
Misel-Wuchter KM, Thurman AL, Johnson JT, Teghanemt A, Gautam N, Pezzulo AA, Bermick JR, Butler NS, Issuree PD. Developmental epigenetic programming by Tet1/3 determines peripheral CD8 T cell fate. EMBO Rep 2025; 26:2494-2518. [PMID: 40175595 DOI: 10.1038/s44319-025-00439-z] [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: 05/21/2024] [Revised: 03/18/2025] [Accepted: 03/20/2025] [Indexed: 04/04/2025] Open
Abstract
In response to infections, naive CD8 T cells give rise to effector and memory T cells. However, eliciting long-lived memory CD8 T cells remains a challenge for many infections. DNA demethylation of cytosines within CpG dinucleotides by Tet enzymes is a key epigenetic mechanism that regulates short- and long-term transcriptional programs in cells. Currently, their roles in modulating CD8 T-cell effector and memory differentiation are unclear. Here, we report that developing CD8 T cells lacking Tet1/3 preferentially differentiate into short-lived effector and effector memory cells following acute infection. Using genome-wide analyses, mice in which Tet1/3 were ablated during T-cell development and mature CD8 T cells, respectively, we show that Tet1/3 regulates these cell fates by licensing the chromatin landscape of genes downstream of T-cell receptor activation during thymic T-cell maturation. However, in mature CD8 T cells, Tet1/3 are dispensable for effector and memory cell fates. These findings unveil context-specific roles of DNA demethylation, which are essential for defining pathways that contribute to CD8 memory T-cell generation in response to infections.
Collapse
Affiliation(s)
- Kara M Misel-Wuchter
- Inflammation Program, University of Iowa, Iowa City, IA, USA
- Molecular Medicine Graduate Program, University of Iowa, Iowa City, IA, USA
- Department of Internal Medicine, University of Iowa, Iowa City, IA, USA
| | - Andrew L Thurman
- Department of Internal Medicine, University of Iowa, Iowa City, IA, USA
| | - Jordan T Johnson
- Immunology Graduate Program, University of Iowa, Iowa City, IA, USA
| | - Athmane Teghanemt
- Inflammation Program, University of Iowa, Iowa City, IA, USA
- Department of Internal Medicine, University of Iowa, Iowa City, IA, USA
| | - Neelam Gautam
- Inflammation Program, University of Iowa, Iowa City, IA, USA
- Department of Internal Medicine, University of Iowa, Iowa City, IA, USA
| | | | - Jennifer R Bermick
- Inflammation Program, University of Iowa, Iowa City, IA, USA
- Immunology Graduate Program, University of Iowa, Iowa City, IA, USA
- Department of Pediatrics, University of Iowa, Iowa City, IA, USA
| | - Noah S Butler
- Immunology Graduate Program, University of Iowa, Iowa City, IA, USA
- Department of Microbiology and Immunology, University of Iowa, Iowa City, IA, USA
- Graduate Program in Molecular Physiology and Biophysics, University of Iowa, Iowa City, IA, USA
| | - Priya D Issuree
- Inflammation Program, University of Iowa, Iowa City, IA, USA.
- Molecular Medicine Graduate Program, University of Iowa, Iowa City, IA, USA.
- Department of Internal Medicine, University of Iowa, Iowa City, IA, USA.
- Immunology Graduate Program, University of Iowa, Iowa City, IA, USA.
| |
Collapse
|
2
|
Fu Z, Huang Z, Xu H, Liu Q, Li J, Song K, Deng Y, Tao Y, Zhang H, Wang P, Li H, Sheng Y, Zhou A, Han L, Fu Y, Wang C, Choudhary SK, Ye K, Veggiani G, Li Z, August A, Huang W, Shan Q, Peng H. IL-2-inducible T cell kinase deficiency sustains chimeric antigen receptor T cell therapy against tumor cells. J Clin Invest 2024; 135:e178558. [PMID: 39589809 PMCID: PMC11827851 DOI: 10.1172/jci178558] [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: 12/26/2023] [Accepted: 11/19/2024] [Indexed: 11/28/2024] Open
Abstract
Despite the revolutionary achievements of chimeric antigen receptor (CAR) T cell therapy in treating cancers, especially leukemia, several key challenges still limit its therapeutic efficacy. Of particular relevance is the relapse of cancer in large part as a result of exhaustion and short persistence of CAR-T cells in vivo. IL-2-inducible T cell kinase (ITK) is a critical modulator of the strength of T cell receptor signaling, while its role in CAR signaling is unknown. By electroporation of CRISPR-associated protein 9 (Cas9) ribonucleoprotein (RNP) complex into CAR-T cells, we successfully deleted ITK in CD19-CAR-T cells with high efficiency. Bulk and single-cell RNA sequencing analyses revealed downregulation of exhaustion and upregulation of memory gene signatures in ITK-deficient CD19-CAR-T cells. Our results further demonstrated a significant reduction of T cell exhaustion and enhancement of T cell memory, with significant improvement of CAR-T cell expansion and persistence both in vitro and in vivo. Moreover, ITK-deficient CD19-CAR-T cells showed better control of tumor relapse. Our work provides a promising strategy of targeting ITK to develop sustainable CAR-T cell products for clinical use.
Collapse
Affiliation(s)
- Zheng Fu
- Department of Hematology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
- Hubei Jiangxia Laboratory, Wuhan, Hubei, China
- MegaRobo Technologies Co. Ltd., Suzhou, China
- Xinyi Biotech Co. Ltd., Lingang, Shanghai, China
| | - Zineng Huang
- Department of Hematology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
- Institute of Hematology, Central South University, Changsha, Hunan, China
- Hunan Engineering Research Center of Cell Immunotherapy for Hematopoietic Malignancies, Changsha, Hunan, China
| | - Hao Xu
- National Key Laboratory of Immunity and Inflammation, Suzhou Institute of Systems Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Suzhou, Jiangsu, China
| | - Qingbai Liu
- Lianshui People’s Hospital of Kangda College Affiliated to Nanjing Medical University, Huai’an, Jiangsu Province, China
| | - Jing Li
- Key Laboratory of Cluster Science, Ministry of Education of China, School of Chemistry and Chemical Engineering, Beijing Institute of Technology, Beijing, China
| | - Keqing Song
- Tianjin Mogenetics Biotech Co. Ltd., Tianjin, China
| | - Yating Deng
- Department of Hematology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
- Institute of Hematology, Central South University, Changsha, Hunan, China
- Hunan Engineering Research Center of Cell Immunotherapy for Hematopoietic Malignancies, Changsha, Hunan, China
| | - Yujia Tao
- Department of Hematology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
- Institute of Hematology, Central South University, Changsha, Hunan, China
- Hunan Engineering Research Center of Cell Immunotherapy for Hematopoietic Malignancies, Changsha, Hunan, China
| | - Huifang Zhang
- Department of Hematology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
- Institute of Hematology, Central South University, Changsha, Hunan, China
- Hunan Engineering Research Center of Cell Immunotherapy for Hematopoietic Malignancies, Changsha, Hunan, China
| | - Peilong Wang
- Department of Hematology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
- Institute of Hematology, Central South University, Changsha, Hunan, China
- Hunan Engineering Research Center of Cell Immunotherapy for Hematopoietic Malignancies, Changsha, Hunan, China
| | - Heng Li
- Department of Hematology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
- Institute of Hematology, Central South University, Changsha, Hunan, China
- Hunan Engineering Research Center of Cell Immunotherapy for Hematopoietic Malignancies, Changsha, Hunan, China
| | - Yue Sheng
- Department of Hematology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
- Institute of Hematology, Central South University, Changsha, Hunan, China
- Hunan Engineering Research Center of Cell Immunotherapy for Hematopoietic Malignancies, Changsha, Hunan, China
| | - Aijun Zhou
- Lianshui People’s Hospital of Kangda College Affiliated to Nanjing Medical University, Huai’an, Jiangsu Province, China
| | - Lianbin Han
- MegaRobo Technologies Co. Ltd., Suzhou, China
| | - Yan Fu
- MegaRobo Technologies Co. Ltd., Suzhou, China
| | | | | | - Kaixiong Ye
- Institute of Bioinformatics and
- Department of Genetics, Franklin College of Arts and Sciences, University of Georgia, Athens, Georgia, USA
| | - Gianluca Veggiani
- Department of Pathobiological Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, Louisiana, USA
| | - Zhihong Li
- Department of Orthopedics, The Second Xiangya Hospital, Central South University, Changsha, China
- Hunan Key Laboratory of Tumor Models and Individualized Medicine, Changsha, Hunan, China
| | - Avery August
- Department of Microbiology and Immunology, College of Veterinary Medicine, Cornell University, Ithaca, New York, USA
| | - Weishan Huang
- Department of Pathobiological Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, Louisiana, USA
- Department of Microbiology and Immunology, College of Veterinary Medicine, Cornell University, Ithaca, New York, USA
| | - Qiang Shan
- National Key Laboratory of Immunity and Inflammation, Suzhou Institute of Systems Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Suzhou, Jiangsu, China
| | - Hongling Peng
- Department of Hematology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
- Institute of Hematology, Central South University, Changsha, Hunan, China
- Hunan Engineering Research Center of Cell Immunotherapy for Hematopoietic Malignancies, Changsha, Hunan, China
- Hunan Key Laboratory of Tumor Models and Individualized Medicine, Changsha, Hunan, China
| |
Collapse
|
3
|
Gräbnitz F, Oxenius A. CD8 T-cell diversification: Asymmetric cell division and its functional implications. Eur J Immunol 2023; 53:e2250225. [PMID: 36788705 DOI: 10.1002/eji.202250225] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Revised: 02/10/2023] [Accepted: 02/10/2023] [Indexed: 02/16/2023]
Abstract
Establishment of cellular diversity is a basic requirement for the development of multicellular organisms. Cellular diversification can be induced by asymmetric cell division (ACD), during which the emerging two daughter cells unequally inherit lineage specific cargo (including transcription factors, receptors for specific signaling inputs, metabolic platforms, and possibly different epigenetic landscapes), resulting in two daughter cells endowed with different fates. While ACD is strongly involved in lineage choices in mammalian stem cells, its role in fate diversification in lineage committed cell subsets that still exhibit plastic potential, such as T-cells, is currently investigated. In this review, we focus predominantly on the role of ACD in fate diversification of CD8 T-cells. Further, we discuss the impact of differential T-cell receptor stimulation strengths and differentiation history on ACD-mediated fate diversification and highlight a particular importance of ACD in the development of memory CD8 T-cells upon high-affinity stimulation conditions.
Collapse
Affiliation(s)
- Fabienne Gräbnitz
- Institute of Microbiology, ETH Zurich, Vladimir-Prelog-Weg 4, Zurich, 8093, Switzerland
| | - Annette Oxenius
- Institute of Microbiology, ETH Zurich, Vladimir-Prelog-Weg 4, Zurich, 8093, Switzerland
| |
Collapse
|
4
|
Stem cell like memory T cells: A new paradigm in cancer immunotherapy. Clin Immunol 2022; 241:109078. [PMID: 35840054 DOI: 10.1016/j.clim.2022.109078] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Revised: 07/04/2022] [Accepted: 07/09/2022] [Indexed: 11/03/2022]
Abstract
Stem cell like memory T (TSCM) cells have emerged as the apex of memory T cell differentiation for their properties of self-renewal and replenishing progenies. With potent long-term persistence, proliferative capacity and antitumor activity, TSCM cells were thought to be the ideal candidate for cancer immunotherapies. Several strategies have been proposed, such as manipulations of cytokines, metabolic factors, signal pathways, and T cell receptor signal intensity, to induce more TSCM cells in vitro, in the hope that they could reach a clinical order of magnitude to provide more long-lasting and effective anti-tumor effects in vivo. In this review, we summarized the differentiation characteristics of TSCM cells and strategies to generate more TSCM cells. We focused on their roles and application in the cancer immunotherapy especially in adoptive cell transfer therapy and cancer therapeutic vaccines, and hopefully provided clues for future understanding and researches.
Collapse
|
5
|
Huseby ES, Teixeiro E. The perception and response of T cells to a changing environment are based on the law of initial value. Sci Signal 2022; 15:eabj9842. [PMID: 35639856 PMCID: PMC9290192 DOI: 10.1126/scisignal.abj9842] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
αβ T cells are critical components of the adaptive immune system and are capable of inducing sterilizing immunity after pathogen infection and eliminating transformed tumor cells. The development and function of T cells are controlled through the T cell antigen receptor, which recognizes peptides displayed on major histocompatibility complex (MHC) molecules. Here, we review how T cells generate the ability to recognize self-peptide-bound MHC molecules and use signals derived from these interactions to instruct cellular development, activation thresholds, and functional specialization in the steady state and during immune responses. We argue that the basic tenants of T cell development and function follow Weber-Fetcher's law of just noticeable differences and Wilder's law of initial value. Together, these laws argue that the ability of a system to respond and the quality of that response are scalable to the basal state of that system. Manifestation of these laws in T cells generates clone-specific activation thresholds that are based on perceivable differences between homeostasis and pathogen encounter (self versus nonself discrimination), as well as poised states for subsequent differentiation into specific effector cell lineages.
Collapse
Affiliation(s)
- Eric S. Huseby
- Department of Pathology, University of Massachusetts Medical School, Worcester, MA 01655, USA
| | - Emma Teixeiro
- Department of Molecular Microbiology and Immunology, School of Medicine, University of Missouri, Columbia, MO 65212, USA
| |
Collapse
|
6
|
ITK independent development of Th17 responses during hypersensitivity pneumonitis driven lung inflammation. Commun Biol 2022; 5:162. [PMID: 35210549 PMCID: PMC8873479 DOI: 10.1038/s42003-022-03109-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Accepted: 02/03/2022] [Indexed: 12/13/2022] Open
Abstract
T helper 17 (Th17) cells develop in response to T cell receptor signals (TCR) in the presence of specific environments, and produce the inflammatory cytokine IL17A. These cells have been implicated in a number of inflammatory diseases and represent a potential target for ameliorating such diseases. The kinase ITK, a critical regulator of TCR signals, has been shown to be required for the development of Th17 cells. However, we show here that lung inflammation induced by Saccharopolyspora rectivirgula (SR) induced Hypersensitivity pneumonitis (SR-HP) results in a neutrophil independent, and ITK independent Th17 responses, although ITK signals are required for γδ T cell production of IL17A. Transcriptomic analysis of resultant ITK independent Th17 cells suggest that the SR-HP-induced extrinsic inflammatory signals may override intrinsic T cell signals downstream of ITK to rescue Th17 responses in the absence of ITK. These findings suggest that the ability to pharmaceutically target ITK to suppress Th17 responses may be dependent on the type of inflammation.
Collapse
|
7
|
Huang Y, Wang Z, Li H, Hu Z, Hong H, Sun Y, Ke Y, Du X. ITK inhibition promotes long-term survival of cardiac allografts by regulating T cell PLCγ phosphorylation. Am J Transl Res 2020; 12:5762-5771. [PMID: 33042455 PMCID: PMC7540118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2020] [Accepted: 07/18/2020] [Indexed: 06/11/2023]
Abstract
BACKGROUND T cells express interleukin-2 inducible T-cell kinase (ITK), which is an essential modulator of T-cell signaling and function. However, the role of ITK in solid organ transplantation has not been investigated to date. Here, we studied the function of ITK in a murine cardiac transplantation model. METHOD Murine heart transplantation was performed using BALB/C mice as donors and C57BL/6 mice as recipients. Subsequent intraperitoneal injections of an ITK-specific inhibitor (BMS-509744) were performed to assess the effects of the kinase following cardiac transplantation. Additionally, naive T cells were isolated to investigate the inhibitor's potential effects in the alloimmune responses. RESULTS ITK inhibition was found to promote long-term cardiac allograft survival compared with the control group of 36.0 ± 3.8 days vs. 7.0 ± 0.7 days, respectively (P < 0.01). While the Th1/Th17 percentages showed a decrease in prevalence (P < 0.001), the CD4+CD25+Foxp3+ percentages were not markedly affected. In vitro treatment of CD4+ T cells with the ITK inhibitor downregulated the proliferation, possibly by regulating the phosphorylation of PLCγ. CONCLUSION ITK inhibition resulted in lower Th1/Th17 responses after cardiac transplantation and markedly prolonged the mean survival time of the cardiac allografts. Thus, ITK inhibition might be a promising therapeutic target to alleviate alloimmune responses in the cardiac transplantation.
Collapse
Affiliation(s)
- Yajun Huang
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhan, PR China
| | - Zhongyu Wang
- Department of Urology, The First Affiliated Hospital of Yangtze UniversityJingzhou, PR China
| | - Huanhuan Li
- Department of Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhan, PR China
| | - Zhiwei Hu
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhan, PR China
| | - Hao Hong
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhan, PR China
| | - Yongfeng Sun
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhan, PR China
| | - Youli Ke
- Department of General Thoracic Surgery of The No.1 Hospital of Wuhan CityHubei Province, PR China
| | - Xinling Du
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhan, PR China
| |
Collapse
|
8
|
Solouki S, Huang W, Elmore J, Limper C, Huang F, August A. TCR Signal Strength and Antigen Affinity Regulate CD8 + Memory T Cells. THE JOURNAL OF IMMUNOLOGY 2020; 205:1217-1227. [PMID: 32759295 DOI: 10.4049/jimmunol.1901167] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/24/2019] [Accepted: 06/30/2020] [Indexed: 12/14/2022]
Abstract
CD8+ T cells play a critical role in adaptive immunity, differentiating into CD8+ memory T cells that form the basis of protective cellular immunity. Vaccine efficacy is attributed to long-term protective immunity, and understanding the parameters that regulate development of CD8+ T cells is critical to the design of T cell-mediated vaccines. We show in this study using mouse models that two distinct parameters, TCR signal strength (regulated by the tyrosine kinase ITK) and Ag affinity, play important but separate roles in modulating the development of memory CD8+ T cells. Unexpectedly, our data reveal that reducing TCR signal strength along with reducing Ag affinity for the TCR leads to enhanced and accelerated development of CD8+ memory T cells. Additionally, TCR signal strength is able to regulate CD8+ T cell effector cytokine R production independent of TCR Ag affinity. Analysis of RNA-sequencing data reveals that genes for inflammatory cytokines/cytokine receptors are significantly altered upon changes in Ag affinity and TCR signal strength. Furthermore, our findings show that the inflammatory milieu is critical in regulating this TCR signal strength-mediated increase in memory development, as both CpG oligonucleotide treatment or cotransfer of wild-type and Itk-/- T cells eliminates the observed increase in memory cell formation. These findings suggest that TCR signal strength and Ag affinity independently contribute to CD8+ memory T cell development, which is modulated by inflammation, and suggest that manipulating TCR signal strength along with Ag affinity, may be used to tune the development of CD8+ memory T cells during vaccine development.
Collapse
Affiliation(s)
- Sabrina Solouki
- Department of Microbiology and Immunology, Cornell University, Ithaca, NY 14853; and
| | - Weishan Huang
- Department of Microbiology and Immunology, Cornell University, Ithaca, NY 14853; and.,Department of Pathobiological Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA 70803
| | - Jessica Elmore
- Department of Microbiology and Immunology, Cornell University, Ithaca, NY 14853; and
| | - Candice Limper
- Department of Microbiology and Immunology, Cornell University, Ithaca, NY 14853; and
| | - Fei Huang
- Department of Microbiology and Immunology, Cornell University, Ithaca, NY 14853; and
| | - Avery August
- Department of Microbiology and Immunology, Cornell University, Ithaca, NY 14853; and
| |
Collapse
|
9
|
Tuning T helper cell differentiation by ITK. Biochem Soc Trans 2020; 48:179-185. [PMID: 32049330 DOI: 10.1042/bst20190486] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2019] [Revised: 12/28/2019] [Accepted: 01/13/2020] [Indexed: 12/24/2022]
Abstract
CD4+ effector T cells effectuate T cell immune responses, producing cytokines to orchestrate the nature and type of immune responses. The non-receptor tyrosine kinase IL-2 inducible T cell kinase (ITK), a mediator of T cell Receptor signaling, plays a critical role in tuning the development of these effector cells. In this review we discussed the role that signals downstream of ITK, including the Ras/MAPK pathway, play in differentially controlling the differentiation of TH17, Foxp3+ T regulatory (Treg) cells, and Type 1 regulatory T (Tr1) cells, supporting a model of ITK signals controlling a decision point in the effector T cell differentiation process.
Collapse
|
10
|
Parry HM, Mirajkar N, Cutmore N, Zuo J, Long H, Kwok M, Oldrieve C, Hudson C, Stankovic T, Paneesha S, Kelly M, Begum J, McSkeane T, Pratt G, Moss P. Long-Term Ibrutinib Therapy Reverses CD8 + T Cell Exhaustion in B Cell Chronic Lymphocytic Leukaemia. Front Immunol 2019; 10:2832. [PMID: 31921116 PMCID: PMC6921985 DOI: 10.3389/fimmu.2019.02832] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Accepted: 11/18/2019] [Indexed: 12/18/2022] Open
Abstract
Chronic Lymphocytic Leukaemia (CLL) is associated with immune suppression and susceptibility to infection. CD8+ T cell numbers are increased and demonstrate elevated expression of PD-1 and impaired function. The mechanisms driving these features of exhaustion are uncertain but are likely to include chronic immune recognition of tumor and/or infectious agents. We investigated the number, phenotype and function of total and virus-specific CD8+ T cells in 65 patients with CLL and 14 patients undergoing long-term ibrutinib therapy (median 21 months). Ibrutinib substantially reduced the number of both CD3+ T cells and CD8+ T cells. Importantly, this was associated with a reduction in PD-1 expression on CD8+ T cells (median 28 vs. 24%; p = 0.042) and 3.5 fold increase in cytokine production following mitogen stimulation. The influence of ibrutinib on antigen-specific CD8+ T cell function was assessed by HLA-peptide tetramers and revealed increased IFNγ and TNFα cytokine responses following stimulation with CMV or EBV peptides together with a 55% reduction in the frequency of "inflated" virus-specific CD8+ T cells. These findings reveal that long-term ibrutinib therapy is associated with substantial reversal of T cell exhaustion in B-CLL and is likely to contribute to the reduced infection risk seen in association with this agent.
Collapse
MESH Headings
- Adenine/analogs & derivatives
- Aged
- Biomarkers
- CD8-Positive T-Lymphocytes/drug effects
- CD8-Positive T-Lymphocytes/metabolism
- CD8-Positive T-Lymphocytes/pathology
- Cytomegalovirus/immunology
- Duration of Therapy
- Female
- Herpesvirus 4, Human/immunology
- Humans
- Leukemia, Lymphocytic, Chronic, B-Cell/drug therapy
- Leukemia, Lymphocytic, Chronic, B-Cell/etiology
- Leukemia, Lymphocytic, Chronic, B-Cell/metabolism
- Leukemia, Lymphocytic, Chronic, B-Cell/pathology
- Male
- Middle Aged
- Peptides/immunology
- Piperidines
- Protein Kinase Inhibitors/administration & dosage
- Protein Kinase Inhibitors/adverse effects
- Protein Kinase Inhibitors/therapeutic use
- Pyrazoles/administration & dosage
- Pyrazoles/adverse effects
- Pyrazoles/therapeutic use
- Pyrimidines/administration & dosage
- Pyrimidines/adverse effects
- Pyrimidines/therapeutic use
- Treatment Outcome
Collapse
Affiliation(s)
- Helen M. Parry
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, United Kingdom
| | - Nikhil Mirajkar
- Queen Elizabeth Hospital, University Hospitals Birmingham NHS Foundation Trust, Birmingham, United Kingdom
| | - Natasha Cutmore
- St James' University Hospital, Leeds Teaching Hospitals Trust, Leeds, United Kingdom
| | - Jianmin Zuo
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, United Kingdom
| | - Heather Long
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, United Kingdom
| | - Marwan Kwok
- Institute of Cancer and Genomic Sciences University of Birmingham, Birmingham, United Kingdom
| | - Ceri Oldrieve
- Institute of Cancer and Genomic Sciences University of Birmingham, Birmingham, United Kingdom
| | - Chris Hudson
- Faculty of Medicine & Health Sciences, University of Nottingham, Nottingham, United Kingdom
| | - Tatjana Stankovic
- Institute of Cancer and Genomic Sciences University of Birmingham, Birmingham, United Kingdom
| | - Shankara Paneesha
- Heartlands Hospital, University Hospitals Birmingham NHS Foundation Trust, Birmingham, United Kingdom
| | - Melanie Kelly
- Heartlands Hospital, University Hospitals Birmingham NHS Foundation Trust, Birmingham, United Kingdom
| | - Jusnara Begum
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, United Kingdom
| | - Tina McSkeane
- Cancer Research UK Clinical Trials Unit, University of Birmingham, Birmingham, United Kingdom
| | - Guy Pratt
- Queen Elizabeth Hospital, University Hospitals Birmingham NHS Foundation Trust, Birmingham, United Kingdom
| | - Paul Moss
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, United Kingdom
| |
Collapse
|
11
|
Girel S, Arpin C, Marvel J, Gandrillon O, Crauste F. Model-Based Assessment of the Role of Uneven Partitioning of Molecular Content on Heterogeneity and Regulation of Differentiation in CD8 T-Cell Immune Responses. Front Immunol 2019; 10:230. [PMID: 30842771 PMCID: PMC6392104 DOI: 10.3389/fimmu.2019.00230] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2018] [Accepted: 01/28/2019] [Indexed: 12/16/2022] Open
Abstract
Activation of naive CD8 T-cells can lead to the generation of multiple effector and memory subsets. Multiple parameters associated with activation conditions are involved in generating this diversity that is associated with heterogeneous molecular contents of activated cells. Although naive cell polarisation upon antigenic stimulation and the resulting asymmetric division are known to be a major source of heterogeneity and cell fate regulation, the consequences of stochastic uneven partitioning of molecular content upon subsequent divisions remain unclear yet. Here we aim at studying the impact of uneven partitioning on molecular-content heterogeneity and then on the immune response dynamics at the cellular level. To do so, we introduce a multiscale mathematical model of the CD8 T-cell immune response in the lymph node. In the model, cells are described as agents evolving and interacting in a 2D environment while a set of differential equations, embedded in each cell, models the regulation of intra and extracellular proteins involved in cell differentiation. Based on the analysis of in silico data at the single cell level, we show that immune response dynamics can be explained by the molecular-content heterogeneity generated by uneven partitioning at cell division. In particular, uneven partitioning acts as a regulator of cell differentiation and induces the emergence of two coexisting sub-populations of cells exhibiting antagonistic fates. We show that the degree of unevenness of molecular partitioning, along all cell divisions, affects the outcome of the immune response and can promote the generation of memory cells.
Collapse
Affiliation(s)
- Simon Girel
- Univ Lyon, Université Claude Bernard Lyon 1, CNRS UMR 5208, Institut Camille Jordan, Villeurbanne, France
- Inria, Villeurbanne, France
| | - Christophe Arpin
- CIRI, Centre International de Recherche en Infectiologie, Univ Lyon, Inserm, U111, Université Claude Bernard, Lyon 1, CNRS, UMR5308, ENS de Lyon, Lyon, France
| | - Jacqueline Marvel
- CIRI, Centre International de Recherche en Infectiologie, Univ Lyon, Inserm, U111, Université Claude Bernard, Lyon 1, CNRS, UMR5308, ENS de Lyon, Lyon, France
| | - Olivier Gandrillon
- Inria, Villeurbanne, France
- Univ Lyon, ENS de Lyon, Univ Claude Bernard, CNRS UMR 5239, INSERM U1210, Laboratory of Biology and Modelling of the Cell, Lyon, France
| | - Fabien Crauste
- Univ Lyon, Université Claude Bernard Lyon 1, CNRS UMR 5208, Institut Camille Jordan, Villeurbanne, France
- Inria, Villeurbanne, France
| |
Collapse
|
12
|
Purwada A, Tian YF, Huang W, Rohrbach KM, Deol S, August A, Singh A. Self-Assembly Protein Nanogels for Safer Cancer Immunotherapy. Adv Healthc Mater 2016; 5:1413-9. [PMID: 27100566 PMCID: PMC5532174 DOI: 10.1002/adhm.201501062] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2015] [Revised: 03/04/2016] [Indexed: 01/15/2023]
Abstract
Soluble antigen-based cancer vaccines have poor retention in tissues along with suboptimal antigen processing by dendritic cells. Multiple booster doses are often needed, leading to dose-limiting systemic toxicity. A versatile, immunomodulatory, self-assembly protein nanogel vaccine is reported that induces robust immune cell response at lower antigen doses than soluble antigens, an important step towards biomaterials-based safer immunotherapy approaches.
Collapse
Affiliation(s)
- Alberto Purwada
- Meinig School of Biomedical Engineering, College of Engineering, Cornell University, Ithaca, NY 14853, USA
| | - Ye F. Tian
- Sibley School of Mechanical and Aerospace Engineering, College of Engineering, Cornell University, Ithaca, NY 14853, USA
| | - Weishan Huang
- Department of Microbiology and Immunology, College of Veterinary Medicine, Cornell University, Ithaca, NY 14853, USA
| | - Kathleen M. Rohrbach
- Department of Material Science and Engineering, College of Engineering, Cornell University, Ithaca, NY 14853, USA
| | - Simrita Deol
- Department of Microbiology and Immunology, College of Veterinary Medicine, Cornell University, Ithaca, NY 14853, USA
| | - Avery August
- Department of Microbiology and Immunology, College of Veterinary Medicine, Cornell University, Ithaca, NY 14853, USA
| | - Ankur Singh
- Sibley School of Mechanical and Aerospace Engineering, College of Engineering, Cornell University, Ithaca, NY 14853, USA
| |
Collapse
|