1
|
Zhang X, Zhang C, Lu S, Dong J, Tang N, Wang Y, Han W, Pan X, Zhang X, Liu D, Shyh-Chang N, Wang Y, Feng G, Wang H. Miltefosine reinvigorates exhausted T cells by targeting their bioenergetic state. Cell Rep Med 2024; 5:101869. [PMID: 39657666 PMCID: PMC11722131 DOI: 10.1016/j.xcrm.2024.101869] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2023] [Revised: 09/05/2024] [Accepted: 11/15/2024] [Indexed: 12/12/2024]
Abstract
T cell exhaustion presents a major challenge for the efficacy of both immune checkpoint inhibitors (ICBs) and chimeric antigen receptor T (CAR-T) cell immunotherapies. To address this issue, we generate hypofunctional CAR-T cells that imitate the exhaustion state. By screening a Food and Drug Administration (FDA)-approved small molecule library using this model, we identify miltefosine as a potent molecule that restores the impaired function of CAR-T cells in a PD-1/PD-L1-independent manner. Impressively, in the terminally exhausted state where PD-1 antibody treatment is ineffective, miltefosine still enhances CAR-T cell activity. Single-cell sequencing analysis reveals that miltefosine treatment significantly increases the population of effector cells. Mechanistically, miltefosine improves impaired glycolysis and oxidative phosphorylation in hypofunctional CAR-T cells. In both allogeneic and syngeneic tumor models, miltefosine effectively enhances the solid tumor clearance ability of CAR-T cells and T cells, demonstrating its potential as an effective immunotherapeutic drug.
Collapse
Affiliation(s)
- Xingying Zhang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
| | - Chenze Zhang
- National Key Laboratory of Efficacy and Mechanism on Chinese Medicine for Metabolic Diseases, Beijing Research Institute of Chinese Medicine, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Shan Lu
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Jingxi Dong
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
| | - Na Tang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China
| | - Yao Wang
- Chinese People's Liberation Army General Hospital, Beijing 100176, China
| | - Weidong Han
- Chinese People's Liberation Army General Hospital, Beijing 100176, China
| | - Xi Pan
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Xiang Zhang
- National Key Laboratory of Efficacy and Mechanism on Chinese Medicine for Metabolic Diseases, Beijing Research Institute of Chinese Medicine, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Duan Liu
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
| | - Ng Shyh-Chang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China; Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China; Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, China
| | - Yu Wang
- College of Life Sciences and Oceanography, Shenzhen University, Shenzhen 518060, China
| | - Guihai Feng
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China
| | - Haoyi Wang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China; Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China; Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, China.
| |
Collapse
|
2
|
Jin X, Zhou K, Zhang R, Li J, Guo M, Qiao H, Wu M, Cao X, Dong G, Zhang S. Construction and validation of prognostic signature for transcription factors regulating T cell exhaustion in hepatocellular carcinoma. Medicine (Baltimore) 2024; 103:e38713. [PMID: 38968464 PMCID: PMC11224837 DOI: 10.1097/md.0000000000038713] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2023] [Accepted: 06/06/2024] [Indexed: 07/07/2024] Open
Abstract
In the tumor microenvironment (TME), CD8+ T cells showed stage exhaustion due to the continuous stimulation of tumor antigens. To evaluate the status of CD8+ T cells and reverse the exhaustion is the key to evaluate the prognosis and therapeutic effect of tumor patients. The aim of this study was to establish a prognostic signature that could effectively predict prognosis and response to immunotherapy in patients with hepatocellular carcinoma (HCC). We used univariate Cox analysis to obtain transcription factors associated with CD8+ T cell exhaustion from The Cancer Genome Atlas dataset. Then, the prognostic signature for transcription factors basic leucine zipper ATF-like transcription factor, Eomesodermin, and T-box protein 21 regulating T cell exhaustion was constructed using LASSO Cox regression. The relative expression levels of the mRNA of the 3 transcription factors were detected by reverse transcription-quantitative polymerase chain reaction in 23 pairs of HCC and paracancer tissues, and verified internally in The Cancer Genome Atlas dataset and externally in the International Cancer Genome Consortium dataset. Cox regression analysis showed that risk score was an independent prognostic variable. The overall survival of the high-risk group was significantly lower than that of the low-risk group. The low-risk group had higher immune scores, matrix scores, and ESTIMATE scores, and significantly increased expression levels of most immune checkpoint genes in the low-risk group. Therefore, patients with lower risk scores benefit more from immunotherapy. The combination of the 3 transcription factors can evaluate the exhaustion state of CD8+ T cells in the TME, laying a foundation for evaluating the TME and immunotherapy efficacy in patients with HCC.
Collapse
Affiliation(s)
- Xi Jin
- Scientific Research Center, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Kun Zhou
- Scientific Research Center, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
- Department of Clinical Laboratory, Beidahuang Industry Group General Hospital, Harbin, China
| | - Rongzheng Zhang
- Scientific Research Center, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Jingbo Li
- Department of Anesthesiology Research Institute, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Mengrui Guo
- Scientific Research Center, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Han Qiao
- Scientific Research Center, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Meng Wu
- Scientific Research Center, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Xinyang Cao
- Scientific Research Center, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Guanglu Dong
- Department of Tumor Radiotherapy, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Shuyun Zhang
- Scientific Research Center, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| |
Collapse
|
3
|
Li J, Vranjkovic A, Read D, Delaney SP, Stanford WL, Cooper CL, Crawley AM. Lasting differential gene expression of circulating CD8 T cells in chronic HCV infection with cirrhosis identifies a role for Hedgehog signaling in cellular hyperfunction. Front Immunol 2024; 15:1375485. [PMID: 38887299 PMCID: PMC11180750 DOI: 10.3389/fimmu.2024.1375485] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Accepted: 04/19/2024] [Indexed: 06/20/2024] Open
Abstract
Background The impact of chronic hepatic infection on antigen non-specific immune cells in circulation remains poorly understood. We reported lasting global hyperfunction of peripheral CD8 T cells in HCV-infected individuals with cirrhosis. Whether gene expression patterns in bulk CD8 T cells are associated with the severity of liver fibrosis in HCV infection is not known. Methods RNA sequencing of blood CD8 T cells from treatment naïve, HCV-infected individuals with minimal (Metavir F0-1 ≤ 7.0 kPa) or advanced fibrosis or cirrhosis (F4 ≥ 12.5 kPa), before and after direct-acting antiviral therapy, was performed. CD8 T cell function was assessed by flow cytometry. Results In CD8 T cells from pre-DAA patients with advanced compared to minimal fibrosis, Gene Ontology analysis and Gene Set Enrichment Analysis identified differential gene expression related to cellular function and metabolism, including upregulated Hedgehog (Hh) signaling, IFN-α, -γ, TGF-β response genes, apoptosis, apical surface pathways, phospholipase signaling, phosphatidyl-choline/inositol activity, and second-messenger-mediated signaling. In contrast, genes in pathways associated with nuclear processes, RNA transport, cytoskeletal dynamics, cMyc/E2F regulation, oxidative phosphorylation, and mTOR signaling, were reduced. Hh signaling pathway was the top featured gene set upregulated in cirrhotics, wherein hallmark genes GLI1 and PTCH1 ranked highly. Inhibition of Smo-dependent Hh signaling ablated the expression of IFN-γ and perforin in stimulated CD8 T cells from chronic HCV-infected patients with advanced compared to minimal fibrosis. CD8 T cell gene expression profiles post-DAA remained clustered with pre-DAA profiles and disparately between advanced and minimal fibrosis, suggesting a persistent perturbation of gene expression long after viral clearance. Conclusions This analysis of bulk CD8 T cell gene expression in chronic HCV infection suggests considerable reprogramming of the CD8 T cell pool in the cirrhotic state. Increased Hh signaling in cirrhosis may contribute to generalized CD8 T cell hyperfunction observed in chronic HCV infection. Understanding the lasting nature of immune cell dysfunction may help mitigate remaining clinical challenges after HCV clearance and more generally, improve long term outcomes for individuals with severe liver disease.
Collapse
Affiliation(s)
- Jiafeng Li
- Chronic Disease Program, Ottawa Hospital Research Institute, Ottawa, ON, Canada
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, ON, Canada
- Centre for Infection, Immunity and Inflammation, University of Ottawa, Ottawa, ON, Canada
| | - Agatha Vranjkovic
- Chronic Disease Program, Ottawa Hospital Research Institute, Ottawa, ON, Canada
| | - Daniel Read
- Chronic Disease Program, Ottawa Hospital Research Institute, Ottawa, ON, Canada
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, ON, Canada
| | - Sean P. Delaney
- Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa, ON, Canada
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON, Canada
| | - William L. Stanford
- Centre for Infection, Immunity and Inflammation, University of Ottawa, Ottawa, ON, Canada
- Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa, ON, Canada
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON, Canada
- Ottawa Institute of Systems Biology, University of Ottawa, Ottawa, ON, Canada
| | - Curtis L. Cooper
- Centre for Infection, Immunity and Inflammation, University of Ottawa, Ottawa, ON, Canada
- Division of Infectious Diseases, The Ottawa Hospital, Ottawa, ON, Canada
- School of Epidemiology and Public Health, University of Ottawa, Ottawa, ON, Canada
- Clinical Epidemiology Program, Ottawa Hospital Research Institute, Ottawa, ON, Canada
| | - Angela M. Crawley
- Chronic Disease Program, Ottawa Hospital Research Institute, Ottawa, ON, Canada
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, ON, Canada
- Centre for Infection, Immunity and Inflammation, University of Ottawa, Ottawa, ON, Canada
- Department of Biology and Institute of Biochemistry, Carleton University, Ottawa, ON, Canada
| |
Collapse
|
4
|
Jenkins E, Whitehead T, Fellermeyer M, Davis SJ, Sharma S. The current state and future of T-cell exhaustion research. OXFORD OPEN IMMUNOLOGY 2023; 4:iqad006. [PMID: 37554723 PMCID: PMC10352049 DOI: 10.1093/oxfimm/iqad006] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Revised: 06/16/2023] [Accepted: 06/28/2023] [Indexed: 08/10/2023] Open
Abstract
'Exhaustion' is a term used to describe a state of native and redirected T-cell hypo-responsiveness resulting from persistent antigen exposure during chronic viral infections or cancer. Although a well-established phenotype across mice and humans, exhaustion at the molecular level remains poorly defined and inconsistent across the literature. This is, in part, due to an overreliance on surface receptors to define these cells and explain exhaustive behaviours, an incomplete understanding of how exhaustion arises, and a lack of clarity over whether exhaustion is the same across contexts, e.g. chronic viral infections versus cancer. With the development of systems-based genetic approaches such as single-cell RNA-seq and CRISPR screens applied to in vivo data, we are moving closer to a consensus view of exhaustion, although understanding how it arises remains challenging given the difficulty in manipulating the in vivo setting. Accordingly, producing and studying exhausted T-cells ex vivo are burgeoning, allowing experiments to be conducted at scale up and with high throughput. Here, we first review what is currently known about T-cell exhaustion and how it's being studied. We then discuss how improvements in their method of isolation/production and examining the impact of different microenvironmental signals and cell interactions have now become an active area of research. Finally, we discuss what the future holds for the analysis of this physiological condition and, given the diversity of ways in which exhausted cells are now being generated, propose the adoption of a unified approach to clearly defining exhaustion using a set of metabolic-, epigenetic-, transcriptional-, and activation-based phenotypic markers, that we call 'M.E.T.A'.
Collapse
Affiliation(s)
- Edward Jenkins
- Kennedy Institute of Rheumatology, University of Oxford, Oxford OX3 7FY, UK
| | - Toby Whitehead
- Medical Research Council Human Immunology Unit, John Radcliffe Hospital, University of Oxford, Oxford OX3 9DS, UK
| | - Martin Fellermeyer
- Medical Research Council Human Immunology Unit, John Radcliffe Hospital, University of Oxford, Oxford OX3 9DS, UK
| | - Simon J Davis
- Medical Research Council Human Immunology Unit, John Radcliffe Hospital, University of Oxford, Oxford OX3 9DS, UK
| | - Sumana Sharma
- Medical Research Council Human Immunology Unit, John Radcliffe Hospital, University of Oxford, Oxford OX3 9DS, UK
| |
Collapse
|
5
|
Sun L, Su Y, Jiao A, Wang X, Zhang B. T cells in health and disease. Signal Transduct Target Ther 2023; 8:235. [PMID: 37332039 PMCID: PMC10277291 DOI: 10.1038/s41392-023-01471-y] [Citation(s) in RCA: 266] [Impact Index Per Article: 133.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Revised: 04/21/2023] [Accepted: 04/24/2023] [Indexed: 06/20/2023] Open
Abstract
T cells are crucial for immune functions to maintain health and prevent disease. T cell development occurs in a stepwise process in the thymus and mainly generates CD4+ and CD8+ T cell subsets. Upon antigen stimulation, naïve T cells differentiate into CD4+ helper and CD8+ cytotoxic effector and memory cells, mediating direct killing, diverse immune regulatory function, and long-term protection. In response to acute and chronic infections and tumors, T cells adopt distinct differentiation trajectories and develop into a range of heterogeneous populations with various phenotype, differentiation potential, and functionality under precise and elaborate regulations of transcriptional and epigenetic programs. Abnormal T-cell immunity can initiate and promote the pathogenesis of autoimmune diseases. In this review, we summarize the current understanding of T cell development, CD4+ and CD8+ T cell classification, and differentiation in physiological settings. We further elaborate the heterogeneity, differentiation, functionality, and regulation network of CD4+ and CD8+ T cells in infectious disease, chronic infection and tumor, and autoimmune disease, highlighting the exhausted CD8+ T cell differentiation trajectory, CD4+ T cell helper function, T cell contributions to immunotherapy and autoimmune pathogenesis. We also discuss the development and function of γδ T cells in tissue surveillance, infection, and tumor immunity. Finally, we summarized current T-cell-based immunotherapies in both cancer and autoimmune diseases, with an emphasis on their clinical applications. A better understanding of T cell immunity provides insight into developing novel prophylactic and therapeutic strategies in human diseases.
Collapse
Affiliation(s)
- Lina Sun
- Department of Pathogenic Microbiology and Immunology, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
- Institute of Infection and Immunity, Translational Medicine Institute, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, 710061, China
- Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an, Shaanxi, 710061, China
- Xi'an Key Laboratory of Immune Related Diseases, Xi'an, Shannxi, 710061, China
| | - Yanhong Su
- Department of Pathogenic Microbiology and Immunology, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
- Institute of Infection and Immunity, Translational Medicine Institute, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, 710061, China
- Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an, Shaanxi, 710061, China
- Xi'an Key Laboratory of Immune Related Diseases, Xi'an, Shannxi, 710061, China
| | - Anjun Jiao
- Department of Pathogenic Microbiology and Immunology, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
- Institute of Infection and Immunity, Translational Medicine Institute, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, 710061, China
- Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an, Shaanxi, 710061, China
- Xi'an Key Laboratory of Immune Related Diseases, Xi'an, Shannxi, 710061, China
| | - Xin Wang
- Department of Pathogenic Microbiology and Immunology, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
- Institute of Infection and Immunity, Translational Medicine Institute, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, 710061, China
- Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an, Shaanxi, 710061, China
- Xi'an Key Laboratory of Immune Related Diseases, Xi'an, Shannxi, 710061, China
| | - Baojun Zhang
- Department of Pathogenic Microbiology and Immunology, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China.
- Institute of Infection and Immunity, Translational Medicine Institute, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, 710061, China.
- Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an, Shaanxi, 710061, China.
- Xi'an Key Laboratory of Immune Related Diseases, Xi'an, Shannxi, 710061, China.
| |
Collapse
|
6
|
Casey JL, Dore GJ, Grebely J, Matthews GV, Cherepanov V, Martinello M, Marks P, Janssen HLA, Hansen BE, Kaul R, MacParland SA, Gehring AJ, Feld JJ. Hepatitis C virus-specific immune responses following direct-acting antivirals administered during recent hepatitis C virus infection. J Viral Hepat 2023; 30:64-72. [PMID: 36302162 DOI: 10.1111/jvh.13761] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Accepted: 10/13/2022] [Indexed: 12/09/2022]
Abstract
Individuals who spontaneously clear hepatitis C virus (HCV) infection have demonstrated evidence of partial protective immunity, whereas treatment-induced clearance provides little or no protection against reinfection. We aimed to investigate whether treatment of acute HCV infection with direct-acting antivirals (DAA) prevents establishment of, or reverses, T-cell exhaustion, leading to a virus-specific T-cell immune profile more similar to that seen in spontaneous clearance. The magnitude and breadth of HCV-specific T-cell responses before and after DAA or interferon-based therapy in acute or chronic HCV were compared to those of participants with spontaneous clearance of infection, using Enzyme-linked Immunospot (ELISPOT). PBMCs were available for 55 patients comprising 4 groups: spontaneous clearance (n = 17), acute interferon (n = 14), acute DAA (n = 13) and chronic DAA (n = 11). After controlling for sex, the magnitude of post-treatment HCV-specific responses after acute DAA treatment was greater than after chronic DAA or acute IFN treatment and similar to those found in spontaneous clearers. However, spontaneous clearers responded to more HCV peptide pools indicating greater breadth of response. In conclusion, early treatment with DAAs may prevent or reverse some degree of immune exhaustion and result in stronger HCV-specific responses post-treatment. However, individuals with spontaneous clearance had broader HCV-specific responses.
Collapse
Affiliation(s)
- Julia L Casey
- Toronto Centre for Liver Disease, Toronto General Hospital, University Health Network, Toronto, Ontario, Canada.,Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada
| | - Gregory J Dore
- The Kirby Institute, UNSW Sydney, Sydney, New South Wales, Australia
| | - Jason Grebely
- The Kirby Institute, UNSW Sydney, Sydney, New South Wales, Australia
| | - Gail V Matthews
- The Kirby Institute, UNSW Sydney, Sydney, New South Wales, Australia
| | - Vera Cherepanov
- Toronto Centre for Liver Disease, Toronto General Hospital, University Health Network, Toronto, Ontario, Canada
| | | | - Philippa Marks
- The Kirby Institute, UNSW Sydney, Sydney, New South Wales, Australia
| | - Harry L A Janssen
- Toronto Centre for Liver Disease, Toronto General Hospital, University Health Network, Toronto, Ontario, Canada
| | - Bettina E Hansen
- Toronto Centre for Liver Disease, Toronto General Hospital, University Health Network, Toronto, Ontario, Canada.,Institute of Health Policy Management and Evaluation, University of Toronto, Toronto, Ontario, Canada
| | - Rupert Kaul
- Department of Immunology, University of Toronto, Toronto, Ontario, Canada.,Department of Medicine, University Health Network, Toronto, Ontario, Canada
| | - Sonya A MacParland
- Departments of Laboratory Medicine & Pathobiology, University of Toronto, Toronto, Ontario, Canada.,Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada
| | - Adam J Gehring
- Toronto Centre for Liver Disease, Toronto General Hospital, University Health Network, Toronto, Ontario, Canada.,Department of Immunology, University of Toronto, Toronto, Ontario, Canada
| | - Jordan J Feld
- Toronto Centre for Liver Disease, Toronto General Hospital, University Health Network, Toronto, Ontario, Canada.,Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
7
|
Labrosse R, Boufaied I, Bourdin B, Gona S, Randolph HE, Logan BR, Bourbonnais S, Berthe C, Chan W, Buckley RH, Parrott RE, Cuvelier GDE, Kapoor N, Chandra S, Dávila Saldaña BJ, Eissa H, Goldman FD, Heimall J, O'Reilly R, Chaudhury S, Kolb EA, Shenoy S, Griffith LM, Pulsipher M, Kohn DB, Notarangelo LD, Pai SY, Cowan MJ, Dvorak CC, Haddad É, Puck JM, Barreiro LB, Decaluwe H. Aberrant T-cell exhaustion in severe combined immunodeficiency survivors with poor T-cell reconstitution after transplantation. J Allergy Clin Immunol 2023; 151:260-271. [PMID: 35987350 PMCID: PMC9924130 DOI: 10.1016/j.jaci.2022.08.004] [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: 04/11/2022] [Revised: 08/08/2022] [Accepted: 08/10/2022] [Indexed: 02/04/2023]
Abstract
BACKGROUND Severe combined immunodeficiency (SCID) comprises rare inherited disorders of immunity that require definitive treatment through hematopoietic cell transplantation (HCT) or gene therapy for survival. Despite successes of allogeneic HCT, many SCID patients experience incomplete immune reconstitution, persistent T-cell lymphopenia, and poor long-term outcomes. OBJECTIVE We hypothesized that CD4+ T-cell lymphopenia could be associated with a state of T-cell exhaustion in previously transplanted SCID patients. METHODS We analyzed markers of exhaustion in blood samples from 61 SCID patients at a median of 10.4 years after HCT. RESULTS Compared to post-HCT SCID patients with normal CD4+ T-cell counts, those with poor T-cell reconstitution showed lower frequency of naive CD45RA+/CCR7+ T cells, recent thymic emigrants, and TCR excision circles. They also had a restricted TCR repertoire, increased expression of inhibitory receptors (PD-1, 2B4, CD160, BTLA, CTLA-4), and increased activation markers (HLA-DR, perforin) on their total and naive CD8+ T cells, suggesting T-cell exhaustion and aberrant activation, respectively. The exhaustion score of CD8+ T cells was inversely correlated with CD4+ T-cell count, recent thymic emigrants, TCR excision circles, and TCR diversity. Exhaustion scores were higher among recipients of unconditioned HCT, especially when further in time from HCT. Patients with fewer CD4+ T cells showed a transcriptional signature of exhaustion. CONCLUSIONS Recipients of unconditioned HCT for SCID may develop late post-HCT T-cell exhaustion as a result of diminished production of T-lineage cells. Elevated expression of inhibitory receptors on their T cells may be a biomarker of poor long-term T-cell reconstitution.
Collapse
Affiliation(s)
- Roxane Labrosse
- Pediatric Immunology and Rheumatology Division, Department of Pediatrics, University of Montreal, Montreal, Quebec, Canada
| | - Ines Boufaied
- Cytokines and Adaptive Immunity Laboratory, Sainte-Justine University Hospital Research Center, Montreal, Quebec, Canada
| | - Benoîte Bourdin
- Cytokines and Adaptive Immunity Laboratory, Sainte-Justine University Hospital Research Center, Montreal, Quebec, Canada
| | - Saideep Gona
- Genetics, Genomics, and Systems Biology, Department of Medicine, Section of Genetic Medicine, University of Chicago, Chicago, Ill
| | - Haley E Randolph
- Genetics, Genomics, and Systems Biology, Department of Medicine, Section of Genetic Medicine, University of Chicago, Chicago, Ill
| | - Brent R Logan
- Division of Biostatistics, Medical College of Wisconsin, Milwaukee, Wis
| | - Sara Bourbonnais
- Cytokines and Adaptive Immunity Laboratory, Sainte-Justine University Hospital Research Center, Montreal, Quebec, Canada
| | - Chloé Berthe
- Cytokines and Adaptive Immunity Laboratory, Sainte-Justine University Hospital Research Center, Montreal, Quebec, Canada
| | - Wendy Chan
- Division of Allergy, Immunology, and Blood and Marrow Transplantation, Department of Pediatrics, University of California, San Francisco, and UCSF Benioff Children's Hospital, San Francisco, Calif
| | | | | | - Geoffrey D E Cuvelier
- Manitoba Blood and Marrow Transplant Program, CancerCare Manitoba, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Neena Kapoor
- Blood and Marrow Transplant Program, Division of Hematology, Oncology, and Blood and Marrow Transplantation, Children's Hospital Los Angeles, Keck School of Medicine, University of Southern California, Los Angeles, Calif
| | - Sharat Chandra
- Division of Bone Marrow Transplantation and Immune Deficiency, Cincinnati Children's Hospital Medical Center, and the Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Blachy J Dávila Saldaña
- Division of Blood and Marrow Transplantation, Children's National Hospital, George Washington University School of Medicine and Health Sciences, Washington, DC
| | - Hesham Eissa
- Children's Hospital of Colorado, University of Colorado School of Medicine, Aurora, Colo
| | - Fred D Goldman
- Department of Pediatrics, The University of Alabama at Birmingham, Birmingham, Ala
| | - Jennifer Heimall
- Allergy and Immunology, The Children's Hospital of Philadelphia, Philadelphia, Pa
| | - Richard O'Reilly
- Department of Pediatrics, Bone Marrow Transplant Service, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Sonali Chaudhury
- Division of Hematology, Oncology, and Stem Cell Transplantation, Ann & Robert H. Lurie Children's Hospital of Chicago, Northwestern University Feinberg School of Medicine, Chicago, Ill
| | - Edward A Kolb
- Nemours Children's Health, Center for Cancer and Blood Disorders, Wilmington, Del
| | - Shalini Shenoy
- Division of Pediatric Hematology/Oncology, Department of Pediatrics, Washington University School of Medicine, St Louis, Mo
| | - Linda M Griffith
- Division of Allergy, Immunology, and Transplantation, National Institutes of Health, Bethesda, Md
| | - Michael Pulsipher
- Blood and Marrow Transplant Program, Division of Hematology, Oncology, and Blood and Marrow Transplantation, Children's Hospital Los Angeles, Keck School of Medicine, University of Southern California, Los Angeles, Calif
| | - Donald B Kohn
- Pediatrics, David Geffen School of Medicine at University of California, Los Angeles, Calif
| | - Luigi D Notarangelo
- Laboratory of Clinical Immunology and Microbiology, National Institutes of Health, Bethesda, Md
| | - Sung-Yun Pai
- Immune Deficiency Cellular Therapy Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Md
| | - Morton J Cowan
- Division of Allergy, Immunology, and Blood and Marrow Transplantation, Department of Pediatrics, University of California, San Francisco, and UCSF Benioff Children's Hospital, San Francisco, Calif
| | - Christopher C Dvorak
- Division of Allergy, Immunology, and Blood and Marrow Transplantation, Department of Pediatrics, University of California, San Francisco, and UCSF Benioff Children's Hospital, San Francisco, Calif
| | - Élie Haddad
- Pediatric Immunology and Rheumatology Division, Department of Pediatrics, University of Montreal, Montreal, Quebec, Canada
| | - Jennifer M Puck
- Division of Allergy, Immunology, and Blood and Marrow Transplantation, Department of Pediatrics, University of California, San Francisco, and UCSF Benioff Children's Hospital, San Francisco, Calif
| | - Luis B Barreiro
- Genetics, Genomics, and Systems Biology, Department of Medicine, Section of Genetic Medicine, University of Chicago, Chicago, Ill
| | - Hélène Decaluwe
- Pediatric Immunology and Rheumatology Division, Department of Pediatrics, University of Montreal, Montreal, Quebec, Canada; Cytokines and Adaptive Immunity Laboratory, Sainte-Justine University Hospital Research Center, Montreal, Quebec, Canada.
| |
Collapse
|
8
|
Zhang X, Zhang C, Qiao M, Cheng C, Tang N, Lu S, Sun W, Xu B, Cao Y, Wei X, Wang Y, Han W, Wang H. Depletion of BATF in CAR-T cells enhances antitumor activity by inducing resistance against exhaustion and formation of central memory cells. Cancer Cell 2022; 40:1407-1422.e7. [PMID: 36240777 DOI: 10.1016/j.ccell.2022.09.013] [Citation(s) in RCA: 65] [Impact Index Per Article: 21.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Revised: 01/05/2022] [Accepted: 09/20/2022] [Indexed: 01/09/2023]
Abstract
Chimeric antigen receptor (CAR) T cell therapy has limited efficacy against solid tumors, and one major challenge is T cell exhaustion. To address this challenge, we performed a candidate gene screen using a hypofunction CAR-T cell model and found that depletion of basic leucine zipper ATF-like transcription factor (BATF) improved the antitumor performance of CAR-T cells. In different types of CAR-T cells and mouse OT-1 cells, loss of BATF endows T cells with improved resistance to exhaustion and superior tumor eradication efficacy. Mechanistically, we found that BATF binds to and up-regulates a subset of exhaustion-related genes in human CAR-T cells. BATF regulates the expression of genes involved in development of effector and memory T cells, and knocking out BATF shifts the population toward a more central memory subset. We demonstrate that BATF is a key factor limiting CAR-T cell function and that its depletion enhances the antitumor activity of CAR-T cells against solid tumors.
Collapse
Affiliation(s)
- Xingying Zhang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Chenze Zhang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Miaomiao Qiao
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Chen Cheng
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; School of Life Sciences, University of Science and Technology of China, Hefei, Anhui 230026, China
| | - Na Tang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
| | - Shan Lu
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Wen Sun
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
| | - Beilei Xu
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
| | - Yuanwei Cao
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Xiaofei Wei
- Beijing Cord Blood Bank, Beijing 100176, China
| | - Yao Wang
- Department of Biotherapeutic, the First Medical Center, Chinese PLA General Hospital, Beijing 100853, China
| | - Weidong Han
- Department of Biotherapeutic, the First Medical Center, Chinese PLA General Hospital, Beijing 100853, China
| | - Haoyi Wang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China; Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, China; Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China.
| |
Collapse
|
9
|
Brod SA. The genealogy, methodology, similarities and differences of immune reconstitution therapies for multiple sclerosis and neuromyelitis optica. Autoimmun Rev 2022; 21:103170. [PMID: 35963569 DOI: 10.1016/j.autrev.2022.103170] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Accepted: 08/07/2022] [Indexed: 11/09/2022]
Abstract
Immune reconstitution therapies (IRTs) are a type of short course procedure or pharmaceutical agent within the MS pharmacopeia. They emanate from oncology and induce transient incomplete lympho-ablation with or without myelo-ablation, resulting in potential prolonged immunomodulation. Thus, they provide significant prophylaxis from disease activity without retreatment. Modern IRT for autoimmunity encompasses a heterogeneous group of pulsed lympho- and non-myelo-ablative treatments designed to re-boot the adaptive immune system in a quasi-permanent manner - a re-induction of ontogeny. IRT is the extensive debulking of an auto-aggressive immune system to attempt to reach the Holy Grail of immune tolerance. This incomplete yet significant lympho-ablation induces lymphoproliferation, reduces pathogenic clonal cells, causes thymopoiesis and results in the induction of immune tolerance. Lympho-ablation with immune reconstitution can result in minimal residual autoimmunity. There is a resetting of the immune thermostat - i.e., the immunostat. IRTs have the potential to provide prolonged periods of disease inactivity without retreatment in part through the immunological results of their pulsatile lymphocyte depletion. It is vital to increase our understanding of how IRTs alter a patient's immune response to the antigenic target of the disease so that we can devise newer, more durable and safer forms of such agents. What common features do extant IRTs (i.e., stem cell transplant, alemtuzumab and oral cladribine) have to produce the durable therapeutic response without long term treatment in neuroimmunological diseases such as MS (multiple sclerosis) and NMOSD (neuromyelitis optica spectrum disorders)? Can we learn from these critical features to predict what other maneuvers or agents might effect similar clinical results with equal or greater efficacy and safety?
Collapse
Affiliation(s)
- Staley A Brod
- Division of MS/Neuro-immunology, Department of Neurology, Medical College of Wisconsin, 8701 W Watertown Plank Rd, Milwaukee, WI 53226, USA.
| |
Collapse
|
10
|
The CD226/TIGIT axis is involved in T cell hypo-responsiveness appearance in long-term kidney transplant recipients. Sci Rep 2022; 12:11821. [PMID: 35821240 PMCID: PMC9276733 DOI: 10.1038/s41598-022-15705-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Accepted: 06/28/2022] [Indexed: 11/08/2022] Open
Abstract
T cell exhaustion refers to a dysfunctional state in which effector T cells present a decreased ability to proliferate and to produce cytokines, while the co-expression of inhibitory receptors increases. We investigated global and donor-specific T cell responses in a cohort of stable, living-donor kidney transplant patients that received similar immunosuppression. After transplantation, an increase in the ratio of TIGIT + /CD226 + in mCD4 + T cells (r = 0.47, p = 0.01), and a decrease of CD226 + TIGIT-mCD4 + T cells was observed (r = − 0.55, p = 0.001). This leads to an increase of dysfunctional T cells in patients far from transplantation. In mCD8 + T cells, a decrease of IL-2 production after mitogenic stimulation was observed far from transplantation. Phenotypic analyses revealed an increase of mCD8 + T cells co-expressing PD-1 and TIGIT over time (r = 0.51, p = 0.02). After donor-specific stimulation, the ability of CD4 + T cells to proliferate was decreased compared with third parties. CD4 + T cells expressing CD226 and TIGIT were correlated with allospecific CD4 + proliferation (r = 0.68, p = 0.04). Our study suggests that after kidney transplantation a T cell hyporesponsiveness appears over time, driven by a dysregulation of CD226/TIGIT axis in mCD4 + T cells, associated with an increase of PD1 + TIGIT + in mCD8 + T cells.
Collapse
|
11
|
Yang M, Lin C, Wang Y, Chen K, Han Y, Zhang H, Li W. Cytokine storm promoting T cell exhaustion in severe COVID-19 revealed by single cell sequencing data analysis. PRECISION CLINICAL MEDICINE 2022; 5:pbac014. [PMID: 35694714 PMCID: PMC9172646 DOI: 10.1093/pcmedi/pbac014] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2022] [Revised: 04/28/2022] [Accepted: 05/11/2022] [Indexed: 12/15/2022] Open
Abstract
Background Evidence has suggested that cytokine storms may be associated with T cell exhaustion (TEX) in COVID-19. However, the interaction mechanism between cytokine storms and TEX remains unclear. Methods With the aim of dissecting the molecular relationship of cytokine storms and TEX through single-cell RNA sequencing data analysis, we identified 14 cell types from bronchoalveolar lavage fluid of COVID-19 patients and healthy people. We observed a novel subset of severely exhausted CD8 T cells (Exh T_CD8) that co-expressed multiple inhibitory receptors, and two macrophage subclasses that were the main source of cytokine storms in bronchoalveolar. Results Correlation analysis between cytokine storm level and TEX level suggested that cytokine storms likely promoted TEX in severe COVID-19. Cell–cell communication analysis indicated that cytokines (e.g. CXCL10, CXCL11, CXCL2, CCL2, and CCL3) released by macrophages acted as ligands and significantly interacted with inhibitory receptors (e.g. CXCR3, DPP4, CCR1, CCR2, and CCR5) expressed by Exh T_CD8. These interactions formed the cytokine–receptor axes, which were also verified to be significantly correlated with cytokine storms and TEX in lung squamous cell carcinoma. Conclusions Cytokine storms may promote TEX through cytokine-receptor axes and be associated with poor prognosis in COVID-19. Blocking cytokine-receptor axes may reverse TEX. Our finding provides novel insights into TEX in COVID-19 and new clues for cytokine-targeted immunotherapy development.
Collapse
Affiliation(s)
- Minglei Yang
- Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China
| | - Chenghao Lin
- Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China
| | - Yanni Wang
- Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China
| | - Kang Chen
- Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China
| | - Yutong Han
- Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China
| | - Haiyue Zhang
- Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China
| | - Weizhong Li
- Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China
- Center for Precision Medicine, Sun Yat-sen University, Guangzhou 510080, China
- Key Laboratory of Tropical Disease Control of Ministry of Education, Sun Yat-Sen University, Guangzhou 510080, China
| |
Collapse
|
12
|
Mishra KP, Singh M, Saraswat D, Ganju L, Varshney R. Dysfunctional State of T Cells or Exhaustion During Chronic Viral Infections and COVID-19: A Review. Viral Immunol 2022; 35:284-290. [PMID: 35325564 DOI: 10.1089/vim.2022.0002] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Coronavirus disease 2019 (COVID-19) continuously affecting the lives of millions of people. The virus is spread through the respiratory route to an uninfected person, causing mild-to-moderate respiratory disease-like symptoms that sometimes progress to severe form and can be fatal. When the host is infected with the virus, both innate and adaptive immunity comes into play. The effector T cells act as the master player of adaptive immune response in eradicating the virus from the system. But during cancer and chronic viral infections, the fate of an effector T cell is altered, and the T cell may enters a state of exhaustion, which is marked by loss of effector function, depleted proliferative capacity and cytotoxic effect accomplished by an increased expression of numerous inhibitory receptors such as programmed cell death protein 1 (PD-1), lymphocyte-activation protein 3 (LAG-3), and cytotoxic T lymphocyte-associated antigen 4 (CTLA-4) on their surface. Various other transcriptional and epigenetic changes take place inside the T cell when it enters into an exhausted state. Latest studies point toward the induction of an abnormal immune response such as lymphopenia, cytokine storm, and T cell exhaustion during SARS-CoV-2 (severe acute respiratory syndrome coronavirus 2) infection. This review sheds light on the dysfunctional state of T cells during chronic viral infection and COVID-19. Understanding the cause and the effect of T cell exhaustion observed during COVID-19 may help resolve new therapeutic potentials for treating chronic infections and other diseases.
Collapse
Affiliation(s)
- K P Mishra
- Division of Experimental Biology, Defence Institute of Physiology and Allied Sciences, Delhi, India
| | - Mrinalini Singh
- Division of Experimental Biology, Defence Institute of Physiology and Allied Sciences, Delhi, India
| | - Deepika Saraswat
- Division of Experimental Biology, Defence Institute of Physiology and Allied Sciences, Delhi, India
| | - Lilly Ganju
- Division of Experimental Biology, Defence Institute of Physiology and Allied Sciences, Delhi, India
| | - Rajeev Varshney
- Division of Experimental Biology, Defence Institute of Physiology and Allied Sciences, Delhi, India
| |
Collapse
|
13
|
Titov A, Kaminskiy Y, Ganeeva I, Zmievskaya E, Valiullina A, Rakhmatullina A, Petukhov A, Miftakhova R, Rizvanov A, Bulatov E. Knowns and Unknowns about CAR-T Cell Dysfunction. Cancers (Basel) 2022; 14:1078. [PMID: 35205827 PMCID: PMC8870103 DOI: 10.3390/cancers14041078] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Revised: 01/29/2022] [Accepted: 02/11/2022] [Indexed: 02/01/2023] Open
Abstract
Immunotherapy using chimeric antigen receptor (CAR) T cells is a promising option for cancer treatment. However, T cells and CAR-T cells frequently become dysfunctional in cancer, where numerous evasion mechanisms impair antitumor immunity. Cancer frequently exploits intrinsic T cell dysfunction mechanisms that evolved for the purpose of defending against autoimmunity. T cell exhaustion is the most studied type of T cell dysfunction. It is characterized by impaired proliferation and cytokine secretion and is often misdefined solely by the expression of the inhibitory receptors. Another type of dysfunction is T cell senescence, which occurs when T cells permanently arrest their cell cycle and proliferation while retaining cytotoxic capability. The first section of this review provides a broad overview of T cell dysfunctional states, including exhaustion and senescence; the second section is focused on the impact of T cell dysfunction on the CAR-T therapeutic potential. Finally, we discuss the recent efforts to mitigate CAR-T cell exhaustion, with an emphasis on epigenetic and transcriptional modulation.
Collapse
Affiliation(s)
- Aleksei Titov
- Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, Russia
- Laboratory of Transplantation Immunology, National Research Centre for Hematology, 125167 Moscow, Russia
| | - Yaroslav Kaminskiy
- Laboratory of Transplantation Immunology, National Research Centre for Hematology, 125167 Moscow, Russia
| | - Irina Ganeeva
- Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, Russia
| | - Ekaterina Zmievskaya
- Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, Russia
| | - Aygul Valiullina
- Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, Russia
| | - Aygul Rakhmatullina
- Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, Russia
| | - Alexey Petukhov
- Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, Russia
- Institute of Hematology, Almazov National Medical Research Center, 197341 Saint Petersburg, Russia
| | - Regina Miftakhova
- Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, Russia
| | - Albert Rizvanov
- Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, Russia
| | - Emil Bulatov
- Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, Russia
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 117997 Moscow, Russia
| |
Collapse
|
14
|
Maldonado J, Beach M, Wang Y, Perez P, Yin H, Pelayo E, Fowler S, Alevizos I, Grisius M, Baer A, Walitt B, De Giorgi V, Alter H, Warner B, Chiorini J. HCV Infection Alters Salivary Gland Histology and Saliva Composition. J Dent Res 2022; 101:534-541. [PMID: 35045743 PMCID: PMC9052835 DOI: 10.1177/00220345211049395] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
Hepatitis C virus (HCV) infection is the most common blood-borne chronic infection in the United States. Chronic lymphocytic sialadenitis and sicca syndrome have been reported in chronic HCV infection. Up to 55% of these patients may have xerostomia; the mechanisms of the xerostomia and salivary gland (SG) hypofunction remain controversial. The objectives of this project are to establish if xerostomia associates with SG and HCV infection and to characterize the structural changes in SG and saliva composition. Eighteen HCV-infected patients with xerostomia were evaluated for SG dysfunction; 6 of these patients (patients 1–6) were further evaluated for SG histopathological changes and changes in saliva composition. The techniques used include clinical and laboratory assessment, SG ultrasonography, histological evaluation, sialochemical and proteomics analysis, and RNA in situ hybridization. All the HCV patients had low saliva flow, chronic sialadenitis, and SG fibrosis and lacked Sjögren syndrome (SS) characteristic autoantibodies. Further evaluation of a subgroup of 6 HCV patients (patients 1–6) demonstrated diffuse lymphocytic infiltrates that are predominantly CD8+ T cells with a significant increase in the number of inflammatory cells. Alcian Blue/periodic acid–Schiff staining showed significant changes in the ratio and intensity of the acinar secretory units of the HCV patients’ minor SG. The submandibular glands showed significant ultrasonographic abnormalities in the parenchyma relative to the parotid glands. Significant changes were also observed in the concentration of sodium and mucin 5b. Although no significant correlation was observed between the lymphocytic infiltrates and the years of HCV chronic infection, a positive correlation was observed between HCV RNA–positive epithelial cells and the years of HCV infection. Consistent with the low saliva flow and xerostomia, patients showed changes in several markers of SG acinar and ductal function. Changes in the composition of the saliva suggest that HCV infection can cause xerostomia by mechanisms distinct from SS.
Collapse
Affiliation(s)
- J.O. Maldonado
- AAV Biology Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, USA
- Salivary Disorders Unit, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, USA
| | - M.E. Beach
- Salivary Disorders Unit, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, USA
| | - Y. Wang
- Mass Spectrometry Facility, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, USA
| | - P. Perez
- Salivary Disorders Unit, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, USA
| | - H. Yin
- AAV Biology Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, USA
| | - E. Pelayo
- Salivary Disorders Unit, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, USA
| | - S. Fowler
- Department of Transfusion Medicine, Warren Grant Magnuson Clinical Center, National Institutes of Health, Bethesda, MD, USA
| | - I. Alevizos
- Salivary Disorders Unit, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, USA
| | - M. Grisius
- Salivary Disorders Unit, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, USA
| | - A.N. Baer
- Salivary Disorders Unit, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, USA
- Division of Rheumatology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - B. Walitt
- Salivary Disorders Unit, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, USA
| | - V. De Giorgi
- Department of Transfusion Medicine, Warren Grant Magnuson Clinical Center, National Institutes of Health, Bethesda, MD, USA
| | - H.J. Alter
- Department of Transfusion Medicine, Warren Grant Magnuson Clinical Center, National Institutes of Health, Bethesda, MD, USA
| | - B.M. Warner
- Salivary Disorders Unit, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, USA
| | - J.A. Chiorini
- AAV Biology Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
15
|
Colombo SAP, Hashad R, Denning DW, Kumararatne DS, Ceron-Gutierrez L, Barcenas-Morales G, MacDonald AS, Harris C, Doffinger R, Kosmidis C. Defective interferon-gamma production is common in chronic pulmonary aspergillosis. J Infect Dis 2021; 225:1822-1831. [PMID: 34850023 DOI: 10.1093/infdis/jiab583] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Accepted: 11/25/2021] [Indexed: 01/07/2023] Open
Abstract
BACKGROUND Immune defects in chronic pulmonary aspergillosis (CPA) are poorly characterised. We compared peripheral blood cytokine profiles in patients with CPA vs healthy controls and explored the relationship with disease severity. METHODS Interferon-gamma (IFNγ), IL-17, TNFα, IL-6, IL-12 and IL-10 were measured after in vitro stimulation of whole blood with lipopolysaccharide (LPS), phytohaemagglutinin (PHA), β-glucan, zymosan (ZYM), IL-12 or IL-18, and combinations. Clinical parameters and mortality were correlated with cytokine production. RESULTS Cytokine profiles were evaluated in 133 patients (57.1% male, mean age 61 years). In comparison to controls, patients with CPA had significantly reduced production of IFNγ in response to stimulation with β-glucan+IL-12 (312 vs 988 pg/ml), LPS+IL-12 (252 vs 1033 pg/ml), ZYM+IL-12 (996 vs 2347 pg/ml), and IL-18+IL-12 (7193 vs 12330 pg/ml). Age >60 (p=0.05, HR 1.71, 95%CI 1.00-2.91) and COPD (p=0.039, HR 1.69, 95%CI 1.03-2.78) were associated with worse survival, whereas high IFNγ production in response to beta-glucan+IL-12 stimulation (p=0.026, HR 0.48, 95%CI 0.25-0.92) was associated with reduced mortality. CONCLUSION Patients with CPA show impaired IFNγ production in peripheral blood in response to stimuli. Defective IFNγ production ability correlates with worse outcomes. Immunotherapy with IFNγ could be beneficial for patients showing impaired IFNγ production in CPA.
Collapse
Affiliation(s)
- Stefano A P Colombo
- Lydia Becker Institute of Immunology and Inflammation, Manchester Collaborative Centre for Inflammation Research, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Rola Hashad
- Division of Infection, Immunity and Respiratory Medicine, School of Biological Sciences, The University of Manchester, Manchester Academic Health Science Centre, UK.,Department of Medical Microbiology and Immunology, Faculty of Medicine, Alexandria University, Alexandria, Egypt
| | - David W Denning
- Manchester Fungal Infection Group, Division of Infection, Immunity and Respiratory Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester Academic Health Science Centre, UK
| | - Dinakantha S Kumararatne
- Department of Clinical Biochemistry and Immunology, Addenbrookes Hospital, Cambridge University NHS Foundation Trust, Cambridge, UK
| | - Lourdes Ceron-Gutierrez
- Department of Clinical Biochemistry and Immunology, Addenbrookes Hospital, Cambridge University NHS Foundation Trust, Cambridge, UK
| | | | - Andrew S MacDonald
- Lydia Becker Institute of Immunology and Inflammation, Manchester Collaborative Centre for Inflammation Research, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Chris Harris
- National Aspergillosis Centre, Wythenshawe Hospital, Manchester University NHS Foundation Trust, Manchester, UK
| | - Rainer Doffinger
- Department of Clinical Biochemistry and Immunology, Addenbrookes Hospital, Cambridge University NHS Foundation Trust, Cambridge, UK
| | - Chris Kosmidis
- Manchester Fungal Infection Group, Division of Infection, Immunity and Respiratory Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester Academic Health Science Centre, UK.,National Aspergillosis Centre, Wythenshawe Hospital, Manchester University NHS Foundation Trust, Manchester, UK
| |
Collapse
|
16
|
Echeverría N, Comas V, Aldunate F, Perbolianachis P, Moreno P, Cristina J. In the era of rapid mRNA-based vaccines: Why is there no effective hepatitis C virus vaccine yet? World J Hepatol 2021; 13:1234-1268. [PMID: 34786164 PMCID: PMC8568586 DOI: 10.4254/wjh.v13.i10.1234] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Revised: 05/14/2021] [Accepted: 09/10/2021] [Indexed: 02/06/2023] Open
Abstract
Hepatitis C virus (HCV) is responsible for no less than 71 million people chronically infected and is one of the most frequent indications for liver transplantation worldwide. Despite direct-acting antiviral therapies fuel optimism in controlling HCV infections, there are several obstacles regarding treatment accessibility and reinfection continues to remain a possibility. Indeed, the majority of new HCV infections in developed countries occur in people who inject drugs and are more plausible to get reinfected. To achieve global epidemic control of this virus the development of an effective prophylactic or therapeutic vaccine becomes a must. The coronavirus disease 19 (COVID-19) pandemic led to auspicious vaccine development against severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) virus, which has renewed interest on fighting HCV epidemic with vaccination. The aim of this review is to highlight the current situation of HCV vaccine candidates designed to prevent and/or to reduce HCV infectious cases and their complications. We will emphasize on some of the crossroads encountered during vaccine development against this insidious virus, together with some key aspects of HCV immunology which have, so far, hampered the progress in this area. The main focus will be on nucleic acid-based as well as recombinant viral vector-based vaccine candidates as the most novel vaccine approaches, some of which have been recently and successfully employed for SARS-CoV-2 vaccines. Finally, some ideas will be presented on which methods to explore for the design of live-attenuated vaccines against HCV.
Collapse
Affiliation(s)
- Natalia Echeverría
- Laboratorio de Virología Molecular, Centro de Investigaciones Nucleares, Facultad de Ciencias, Universidad de la República, Montevideo 11400, Uruguay
| | - Victoria Comas
- Departamento de Desarrollo Biotecnológico, Instituto de Higiene, Facultad de Medicina, Universidad de la República, Montevideo 11600, Uruguay
| | - Fabián Aldunate
- Laboratorio de Virología Molecular, Centro de Investigaciones Nucleares, Facultad de Ciencias, Universidad de la República, Montevideo 11400, Uruguay
| | - Paula Perbolianachis
- Laboratorio de Virología Molecular, Centro de Investigaciones Nucleares, Facultad de Ciencias, Universidad de la República, Montevideo 11400, Uruguay
| | - Pilar Moreno
- Laboratorio de Virología Molecular, Centro de Investigaciones Nucleares, Facultad de Ciencias, Universidad de la República, Montevideo 11400, Uruguay
| | - Juan Cristina
- Laboratorio de Virología Molecular, Centro de Investigaciones Nucleares, Facultad de Ciencias, Universidad de la República, Montevideo 11400, Uruguay.
| |
Collapse
|
17
|
Jia X, Chua BY, Loh L, Koutsakos M, Kedzierski L, Olshansky M, Heath WR, Chang SY, Xu J, Wang Z, Kedzierska K. High expression of CD38 and MHC class II on CD8 + T cells during severe influenza disease reflects bystander activation and trogocytosis. Clin Transl Immunology 2021; 10:e1336. [PMID: 34522380 PMCID: PMC8426257 DOI: 10.1002/cti2.1336] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 05/19/2021] [Accepted: 08/10/2021] [Indexed: 11/12/2022] Open
Abstract
Objectives Although co‐expression of CD38 and HLA‐DR reflects T‐cell activation during viral infections, high and prolonged CD38+HLA‐DR+ expression is associated with severe disease. To date, the mechanism underpinning expression of CD38+HLA‐DR+ is poorly understood. Methods We used mouse models of influenza A/H9N2, A/H7N9 and A/H3N2 infection to investigate mechanisms underpinning CD38+MHC‐II+ phenotype on CD8+ T cells. To further understand MHC‐II trogocytosis on murine CD8+ T cells as well as the significance behind the scenario, we used adoptively transferred transgenic OT‐I CD8+ T cells and A/H3N2‐SIINKEKL infection. Results Analysis of influenza‐specific immunodominant DbNP366+CD8+ T‐cell responses showed that CD38+MHC‐II+ co‐expression was detected on both virus‐specific and bystander CD8+ T cells, with increased numbers of both CD38+MHC‐II+CD8+ T‐cell populations observed in immune organs including the site of infection during severe viral challenge. OT‐I cells adoptively transferred into MHC‐II−/− mice had no MHC‐II after infection, suggesting that MHC‐II was acquired via trogocytosis. The detection of CD19 on CD38+MHC‐II+ OT‐I cells supports the proposition that MHC‐II was acquired by trogocytosis sourced from B cells. Co‐expression of CD38+MHC‐II+ on CD8+ T cells was needed for optimal recall following secondary infection. Conclusions Overall, our study demonstrates that both virus‐specific and bystander CD38+MHC‐II+ CD8+ T cells are recruited to the site of infection during severe disease, and that MHC‐II presence occurs via trogocytosis from antigen‐presenting cells. Our findings highlight the importance of the CD38+MHC‐II+ phenotype for CD8+ T‐cell recall.
Collapse
Affiliation(s)
- Xiaoxiao Jia
- Department of Microbiology and Immunology University of Melbourne, at the Peter Doherty Institute for Infection and Immunity Parkville VIC Australia
| | - Brendon Y Chua
- Department of Microbiology and Immunology University of Melbourne, at the Peter Doherty Institute for Infection and Immunity Parkville VIC Australia
| | - Liyen Loh
- Department of Microbiology and Immunology University of Melbourne, at the Peter Doherty Institute for Infection and Immunity Parkville VIC Australia
| | - Marios Koutsakos
- Department of Microbiology and Immunology University of Melbourne, at the Peter Doherty Institute for Infection and Immunity Parkville VIC Australia
| | - Lukasz Kedzierski
- Department of Microbiology and Immunology University of Melbourne, at the Peter Doherty Institute for Infection and Immunity Parkville VIC Australia.,Faculty of Veterinary and Agricultural Sciences University of Melbourne, at the Peter Doherty Institute for Infection and Immunity Parkville VIC Australia
| | - Moshe Olshansky
- Department of Microbiology Monash University Clayton VIC Australia
| | - William R Heath
- Department of Microbiology and Immunology University of Melbourne, at the Peter Doherty Institute for Infection and Immunity Parkville VIC Australia
| | - So Young Chang
- Department of Microbiology and Immunology University of Melbourne, at the Peter Doherty Institute for Infection and Immunity Parkville VIC Australia
| | - Jianqing Xu
- Shanghai Public Health Clinical Center & Institutes of Biomedical Sciences Key Laboratory of Medical Molecular Virology of Ministry of Education/Health Shanghai Medical College Fudan University Shanghai China
| | - Zhongfang Wang
- Department of Microbiology and Immunology University of Melbourne, at the Peter Doherty Institute for Infection and Immunity Parkville VIC Australia.,State Key Laboratory of Respiratory Disease Guangzhou Medical University Guangzhou China
| | - Katherine Kedzierska
- Department of Microbiology and Immunology University of Melbourne, at the Peter Doherty Institute for Infection and Immunity Parkville VIC Australia
| |
Collapse
|
18
|
The Potential of Tissue-Resident Memory T Cells for Adoptive Immunotherapy against Cancer. Cells 2021; 10:cells10092234. [PMID: 34571883 PMCID: PMC8465847 DOI: 10.3390/cells10092234] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Revised: 08/21/2021] [Accepted: 08/26/2021] [Indexed: 12/12/2022] Open
Abstract
Tissue-resident memory T cells (TRM) comprise an important memory T cell subset that mediates local protection upon pathogen re-encounter. TRM populations preferentially localize at entry sites of pathogens, including epithelia of the skin, lungs and intestine, but have also been observed in secondary lymphoid tissue, brain, liver and kidney. More recently, memory T cells characterized as TRM have also been identified in tumors, including but not limited to melanoma, lung carcinoma, cervical carcinoma, gastric carcinoma and ovarian carcinoma. The presence of these memory T cells has been strongly associated with favorable clinical outcomes, which has generated an interest in targeting TRM cells to improve immunotherapy of cancer patients. Nevertheless, intratumoral TRM have also been found to express checkpoint inhibitory receptors, such as PD-1 and LAG-3. Triggering of such inhibitory receptors could induce dysfunction, often referred to as exhaustion, which may limit the effectiveness of TRM in countering tumor growth. A better understanding of the differentiation and function of TRM in tumor settings is crucial to deploy these memory T cells in future treatment options of cancer patients. The purpose of this review is to provide the current status of an important cancer immunotherapy known as TIL therapy, insight into the role of TRM in the context of antitumor immunity, and the challenges and opportunities to exploit these cells for TIL therapy to ultimately improve cancer treatment.
Collapse
|
19
|
Jacob RA, Edgar CR, Prévost J, Trothen SM, Lurie A, Mumby MJ, Galbraith A, Kirchhoff F, Haeryfar SMM, Finzi A, Dikeakos JD. The HIV-1 accessory protein Nef increases surface expression of the checkpoint receptor Tim-3 in infected CD4 + T cells. J Biol Chem 2021; 297:101042. [PMID: 34358561 PMCID: PMC8390549 DOI: 10.1016/j.jbc.2021.101042] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Revised: 07/28/2021] [Accepted: 08/02/2021] [Indexed: 12/17/2022] Open
Abstract
Prolonged immune activation drives the upregulation of multiple checkpoint receptors on the surface of virus-specific T cells, inducing their exhaustion. Reversing HIV-1-induced T cell exhaustion is imperative for efficient virus clearance; however, viral mediators of checkpoint receptor upregulation remain largely unknown. The enrichment of checkpoint receptors on T cells upon HIV-1 infection severely constrains the generation of an efficient immune response. Herein, we examined the role of HIV-1 Nef in mediating the upregulation of checkpoint receptors on peripheral blood mononuclear cells. We demonstrate that the HIV-1 accessory protein Nef upregulates cell surface levels of the checkpoint receptor T-cell immunoglobulin mucin domain-3 (Tim-3) and that this is dependent on Nef's dileucine motif LL164/165. Furthermore, we used a bimolecular fluorescence complementation assay to demonstrate that Nef and Tim-3 form a complex within cells that is abrogated upon mutation of the Nef dileucine motif. We also provide evidence that Nef moderately promotes Tim-3 shedding from the cell surface in a dileucine motif–dependent manner. Treating HIV-1-infected CD4+ T cells with a matrix metalloprotease inhibitor enhanced cell surface Tim-3 levels and reduced Tim-3 shedding. Finally, Tim-3-expressing CD4+ T cells displayed a higher propensity to release the proinflammatory cytokine interferon-gamma. Collectively, our findings uncover a novel mechanism by which HIV-1 directly increases the levels of a checkpoint receptor on the surface of infected CD4+ T cells.
Collapse
Affiliation(s)
- Rajesh Abraham Jacob
- Department of Microbiology and Immunology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada
| | - Cassandra R Edgar
- Department of Microbiology and Immunology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada
| | - Jérémie Prévost
- Centre de Recherche du CHUM, Montreal, Quebec, Canada; Département de Microbiologie, Infectiologie et Immunologie, Université de Montréal, Montreal, Quebec, Canada
| | - Steven M Trothen
- Department of Microbiology and Immunology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada
| | - Antony Lurie
- Department of Microbiology and Immunology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada
| | - Mitchell J Mumby
- Department of Microbiology and Immunology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada
| | - Alexa Galbraith
- Department of Microbiology and Immunology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada
| | - Frank Kirchhoff
- Institute of Molecular Virology, Ulm University Medical Center, UIm, Germany
| | - S M Mansour Haeryfar
- Department of Microbiology and Immunology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada
| | - Andrés Finzi
- Centre de Recherche du CHUM, Montreal, Quebec, Canada; Département de Microbiologie, Infectiologie et Immunologie, Université de Montréal, Montreal, Quebec, Canada; Department of Microbiology and Immunology, McGill University, Montreal, Quebec, Canada
| | - Jimmy D Dikeakos
- Department of Microbiology and Immunology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada.
| |
Collapse
|
20
|
Yates KB, Tonnerre P, Martin GE, Gerdemann U, Al Abosy R, Comstock DE, Weiss SA, Wolski D, Tully DC, Chung RT, Allen TM, Kim AY, Fidler S, Fox J, Frater J, Lauer GM, Haining WN, Sen DR. Epigenetic scars of CD8 + T cell exhaustion persist after cure of chronic infection in humans. Nat Immunol 2021; 22:1020-1029. [PMID: 34312547 DOI: 10.1038/s41590-021-00979-1] [Citation(s) in RCA: 142] [Impact Index Per Article: 35.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Accepted: 06/17/2021] [Indexed: 12/12/2022]
Abstract
T cell exhaustion is an induced state of dysfunction that arises in response to chronic infection and cancer. Exhausted CD8+ T cells acquire a distinct epigenetic state, but it is not known whether that chromatin landscape is fixed or plastic following the resolution of a chronic infection. Here we show that the epigenetic state of exhaustion is largely irreversible, even after curative therapy. Analysis of chromatin accessibility in HCV- and HIV-specific responses identifies a core epigenetic program of exhaustion in CD8+ T cells, which undergoes only limited remodeling before and after resolution of infection. Moreover, canonical features of exhaustion, including super-enhancers near the genes TOX and HIF1A, remain 'epigenetically scarred.' T cell exhaustion is therefore a conserved epigenetic state that becomes fixed and persists independent of chronic antigen stimulation and inflammation. Therapeutic efforts to reverse T cell exhaustion may require new approaches that increase the epigenetic plasticity of exhausted T cells.
Collapse
Affiliation(s)
- Kathleen B Yates
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, MA, USA.,Center for Cancer Research, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA.,Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Pierre Tonnerre
- Division of Gastroenterology, Liver Center, Massachusetts General Hospital, Boston, MA, USA.,Inserm U976, Institut de Recherche Saint-Louis, Paris, France
| | - Genevieve E Martin
- Nuffield Department of Medicine, University of Oxford, Oxford, UK.,Department of Infectious Diseases, Central Clinical School, Monash University, Melbourne, Australia
| | - Ulrike Gerdemann
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Rose Al Abosy
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Dawn E Comstock
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, MA, USA.,Division of Medical Sciences, Harvard Medical School, Boston, MA, USA
| | - Sarah A Weiss
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, MA, USA.,Division of Medical Sciences, Harvard Medical School, Boston, MA, USA
| | - David Wolski
- Division of Gastroenterology, Liver Center, Massachusetts General Hospital, Boston, MA, USA
| | - Damien C Tully
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA.,Department of Infectious Disease Epidemiology, London School of Hygiene and Tropical Medicine, London, UK
| | - Raymond T Chung
- Division of Gastroenterology, Liver Center, Massachusetts General Hospital, Boston, MA, USA
| | - Todd M Allen
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA
| | - Arthur Y Kim
- Division of Infectious Diseases, Massachusetts General Hospital, Boston, MA, USA
| | - Sarah Fidler
- Division of Medicine, Wright Fleming Institute, Imperial College, London, UK.,Imperial College National Institute for Health Research Biomedical Research Centre, London, UK
| | - Julie Fox
- Department of Genitourinary Medicine and Infectious Disease, Guy's and St Thomas' NHS Foundation Trust, London, UK.,King's College National Institute for Health Research Biomedical Research Centre, London, UK
| | - John Frater
- Nuffield Department of Medicine, University of Oxford, Oxford, UK.,Oxford National Institute for Health Research Biomedical Research Centre, Oxford, UK
| | - Georg M Lauer
- Division of Gastroenterology, Liver Center, Massachusetts General Hospital, Boston, MA, USA
| | - W Nicholas Haining
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, MA, USA. .,Merck Research Laboratories, Boston, MA, USA.
| | - Debattama R Sen
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, MA, USA. .,Center for Cancer Research, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA.
| |
Collapse
|
21
|
Tonnerre P, Wolski D, Subudhi S, Aljabban J, Hoogeveen RC, Damasio M, Drescher HK, Bartsch LM, Tully DC, Sen DR, Bean DJ, Brown J, Torres-Cornejo A, Robidoux M, Kvistad D, Alatrakchi N, Cui A, Lieb D, Cheney JA, Gustafson J, Lewis-Ximenez LL, Massenet-Regad L, Eisenhaure T, Aneja J, Haining WN, Chung RT, Hacohen N, Allen TM, Kim AY, Lauer GM. Differentiation of exhausted CD8 + T cells after termination of chronic antigen stimulation stops short of achieving functional T cell memory. Nat Immunol 2021; 22:1030-1041. [PMID: 34312544 PMCID: PMC8323980 DOI: 10.1038/s41590-021-00982-6] [Citation(s) in RCA: 65] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Accepted: 06/22/2021] [Indexed: 12/13/2022]
Abstract
T cell exhaustion is associated with failure to clear chronic infections and malignant cells. Defining the molecular mechanisms of T cell exhaustion and reinvigoration is essential to improving immunotherapeutic modalities. Here we confirmed pervasive phenotypic, functional, and transcriptional differences between memory and exhausted antigen-specific CD8+ T cells in human hepatitis C virus (HCV) infection before and after treatment. After viral cure, phenotypic changes in clonally stable exhausted T cell populations suggested differentiation towards a memory-like profile. However, functionally, the cells showed little improvement and critical transcriptional regulators remained in the exhaustion state. Notably, T cells from chronic HCV infection that were exposed to antigen for less time because of viral escape mutations were functionally and transcriptionally more similar to memory T cells from spontaneously resolved HCV infection. Thus, T cell stimulation duration impacts exhaustion recovery, with antigen removal after long-term exhaustion being insufficient for development of functional T cell memory.
Collapse
Affiliation(s)
- Pierre Tonnerre
- Division of Gastroenterology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA. .,Inserm U976, Université de Paris, Institut de Recherche Saint-Louis, Paris, France.
| | - David Wolski
- Division of Gastroenterology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Sonu Subudhi
- Division of Gastroenterology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Jihad Aljabban
- Division of Gastroenterology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Ruben C Hoogeveen
- Division of Gastroenterology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Marcos Damasio
- Division of Gastroenterology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Hannah K Drescher
- Division of Gastroenterology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Lea M Bartsch
- Division of Gastroenterology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Damien C Tully
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA
| | - Debattama R Sen
- Division of Medical Sciences, Harvard Medical School, Boston, MA, USA.,Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - David J Bean
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA
| | - Joelle Brown
- Division of Gastroenterology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Almudena Torres-Cornejo
- Division of Gastroenterology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Maxwell Robidoux
- Division of Gastroenterology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Daniel Kvistad
- Division of Gastroenterology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Nadia Alatrakchi
- Division of Gastroenterology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Ang Cui
- Broad Institute of MIT and Harvard, Cambridge, MA, USA.,Harvard-MIT Division of Health Sciences and Technology, MIT, Cambridge, MA, USA
| | - David Lieb
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - James A Cheney
- Division of Gastroenterology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Jenna Gustafson
- Division of Gastroenterology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | | | | | | | - Jasneet Aneja
- Division of Gastroenterology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA.,Division of Infectious Diseases, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - W Nicholas Haining
- Division of Medical Sciences, Harvard Medical School, Boston, MA, USA.,Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Raymond T Chung
- Division of Gastroenterology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Nir Hacohen
- Broad Institute of MIT and Harvard, Cambridge, MA, USA.,Center for Cancer Research, Massachusetts General Hospital, Boston, MA, USA
| | - Todd M Allen
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA
| | - Arthur Y Kim
- Division of Infectious Diseases, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Georg M Lauer
- Division of Gastroenterology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
22
|
Where to Next? Research Directions after the First Hepatitis C Vaccine Efficacy Trial. Viruses 2021; 13:v13071351. [PMID: 34372558 PMCID: PMC8310243 DOI: 10.3390/v13071351] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Revised: 07/03/2021] [Accepted: 07/08/2021] [Indexed: 11/17/2022] Open
Abstract
Thirty years after its discovery, the hepatitis C virus (HCV) remains a leading cause of liver disease worldwide. Given that many countries continue to experience high rates of transmission despite the availability of potent antiviral therapies, an effective vaccine is seen as critical for the elimination of HCV. The recent failure of the first vaccine efficacy trial for the prevention of chronic HCV confirmed suspicions that this virus will be a challenging vaccine target. Here, we examine the published data from this first efficacy trial along with the earlier clinical and pre-clinical studies of the vaccine candidate and then discuss three key research directions expected to be important in ongoing and future HCV vaccine development. These include the following: 1. design of novel immunogens that generate immune responses to genetically diverse HCV genotypes and subtypes, 2. strategies to elicit broadly neutralizing antibodies against envelope glycoproteins in addition to cytotoxic and helper T cell responses, and 3. consideration of the unique immunological status of individuals most at risk for HCV infection, including those who inject drugs, in vaccine platform development and early immunogenicity trials.
Collapse
|
23
|
Peña-Asensio J, Calvo H, Torralba M, Miquel J, Sanz-de-Villalobos E, Larrubia JR. Gamma-Chain Receptor Cytokines & PD-1 Manipulation to Restore HCV-Specific CD8 + T Cell Response during Chronic Hepatitis C. Cells 2021; 10:cells10030538. [PMID: 33802622 PMCID: PMC8001543 DOI: 10.3390/cells10030538] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Revised: 02/23/2021] [Accepted: 02/26/2021] [Indexed: 02/05/2023] Open
Abstract
Hepatitis C virus (HCV)-specific CD8+ T cell response is essential in natural HCV infection control, but it becomes exhausted during persistent infection. Nowadays, chronic HCV infection can be resolved by direct acting anti-viral treatment, but there are still some non-responders that could benefit from CD8+ T cell response restoration. To become fully reactive, T cell needs the complete release of T cell receptor (TCR) signalling but, during exhaustion this is blocked by the PD-1 effect on CD28 triggering. The T cell pool sensitive to PD-1 modulation is the progenitor subset but not the terminally differentiated effector population. Nevertheless, the blockade of PD-1/PD-L1 checkpoint cannot be always enough to restore this pool. This is due to the HCV ability to impair other co-stimulatory mechanisms and metabolic pathways and to induce a pro-apoptotic state besides the TCR signalling impairment. In this sense, gamma-chain receptor cytokines involved in memory generation and maintenance, such as low-level IL-2, IL-7, IL-15, and IL-21, might carry out a positive effect on metabolic reprogramming, apoptosis blockade and restoration of co-stimulatory signalling. This review sheds light on the role of combinatory immunotherapeutic strategies to restore a reactive anti-HCV T cell response based on the mixture of PD-1 blocking plus IL-2/IL-7/IL-15/IL-21 treatment.
Collapse
MESH Headings
- Antibodies, Monoclonal/therapeutic use
- B7-H1 Antigen/antagonists & inhibitors
- B7-H1 Antigen/genetics
- B7-H1 Antigen/immunology
- CD8-Positive T-Lymphocytes/drug effects
- CD8-Positive T-Lymphocytes/immunology
- CD8-Positive T-Lymphocytes/virology
- Gene Expression Regulation
- Hepacivirus/immunology
- Hepacivirus/pathogenicity
- Hepatitis C, Chronic/drug therapy
- Hepatitis C, Chronic/genetics
- Hepatitis C, Chronic/immunology
- Hepatitis C, Chronic/virology
- Host-Pathogen Interactions/drug effects
- Host-Pathogen Interactions/genetics
- Host-Pathogen Interactions/immunology
- Humans
- Immune Checkpoint Inhibitors/therapeutic use
- Immunity, Cellular/drug effects
- Immunotherapy/methods
- Interleukins/genetics
- Interleukins/immunology
- Interleukins/therapeutic use
- Lymphocyte Activation/drug effects
- Precursor Cells, T-Lymphoid/drug effects
- Precursor Cells, T-Lymphoid/immunology
- Precursor Cells, T-Lymphoid/virology
- Programmed Cell Death 1 Receptor/antagonists & inhibitors
- Programmed Cell Death 1 Receptor/genetics
- Programmed Cell Death 1 Receptor/immunology
- Receptors, Antigen, T-Cell, gamma-delta/agonists
- Receptors, Antigen, T-Cell, gamma-delta/genetics
- Receptors, Antigen, T-Cell, gamma-delta/immunology
- Signal Transduction
Collapse
Affiliation(s)
- Julia Peña-Asensio
- Translational Hepatology Unit, Guadalajara University Hospital, E-19002 Guadalajara, Spain; (J.P.-A.); (H.C.); (M.T.); (J.M.); (E.S.-d.-V.)
- Department of Biology of Systems, University of Alcalá, E-28805 Alcalá de Henares, Spain
| | - Henar Calvo
- Translational Hepatology Unit, Guadalajara University Hospital, E-19002 Guadalajara, Spain; (J.P.-A.); (H.C.); (M.T.); (J.M.); (E.S.-d.-V.)
- Section of Gastroenterology & Hepatology, Guadalajara University Hospital, E-19002 Guadalajara, Spain
| | - Miguel Torralba
- Translational Hepatology Unit, Guadalajara University Hospital, E-19002 Guadalajara, Spain; (J.P.-A.); (H.C.); (M.T.); (J.M.); (E.S.-d.-V.)
- Service of Internal Medicine, Guadalajara University Hospital, E-19002 Guadalajara, Spain
- Department of Medicine & Medical Specialties, University of Alcalá, E-28805 Alcalá de Henares, Spain
| | - Joaquín Miquel
- Translational Hepatology Unit, Guadalajara University Hospital, E-19002 Guadalajara, Spain; (J.P.-A.); (H.C.); (M.T.); (J.M.); (E.S.-d.-V.)
- Section of Gastroenterology & Hepatology, Guadalajara University Hospital, E-19002 Guadalajara, Spain
| | - Eduardo Sanz-de-Villalobos
- Translational Hepatology Unit, Guadalajara University Hospital, E-19002 Guadalajara, Spain; (J.P.-A.); (H.C.); (M.T.); (J.M.); (E.S.-d.-V.)
- Section of Gastroenterology & Hepatology, Guadalajara University Hospital, E-19002 Guadalajara, Spain
| | - Juan-Ramón Larrubia
- Translational Hepatology Unit, Guadalajara University Hospital, E-19002 Guadalajara, Spain; (J.P.-A.); (H.C.); (M.T.); (J.M.); (E.S.-d.-V.)
- Section of Gastroenterology & Hepatology, Guadalajara University Hospital, E-19002 Guadalajara, Spain
- Department of Medicine & Medical Specialties, University of Alcalá, E-28805 Alcalá de Henares, Spain
- Correspondence: ; Tel.: +34-949-20-9200
| |
Collapse
|
24
|
Hastings KL, Green MD, Gao B, Ganey PE, Roth RA, Burleson GR. Beyond Metabolism: Role of the Immune System in Hepatic Toxicity. Int J Toxicol 2021; 39:151-164. [PMID: 32174281 DOI: 10.1177/1091581819898399] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The liver is primarily thought of as a metabolic organ; however, the liver is also an important mediator of immunological functions. Key perspectives on this emerging topic were presented in a symposium at the 2018 annual meeting of the American College of Toxicology entitled "Beyond metabolism: Role of the immune system in hepatic toxicity." Viral hepatitis is an important disease of the liver for which insufficient preventive vaccines exist. Host immune responses inadequately clear these viruses and often potentiate immunological inflammation that damages the liver. In addition, the liver is a key innate immune organ against bacterial infection. Hepatocytes and immune cells cooperatively control systemic and local bacterial infections. Conversely, bacterial infection can activate multiple types of immune cells and pathways to cause hepatocyte damage and liver injury. Finally, the immune system and specifically cytokines and drugs can interact in idiosyncratic drug-induced liver injury. This rare disease can result in a disease spectrum that ranges from mild to acute liver failure. The immune system plays a role in this disease spectrum.
Collapse
Affiliation(s)
| | | | - Bin Gao
- Laboratory of Liver Diseases, NIH, Bethesda, MD, USA
| | - Patricia E Ganey
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, MI, USA
| | - Robert A Roth
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, MI, USA
| | - Gary R Burleson
- BRT-Burleson Research Technologies, Inc, Morrisville, NC, USA
| |
Collapse
|
25
|
Sepulveda-Crespo D, Resino S, Martinez I. Strategies Targeting the Innate Immune Response for the Treatment of Hepatitis C Virus-Associated Liver Fibrosis. Drugs 2021; 81:419-443. [PMID: 33400242 DOI: 10.1007/s40265-020-01458-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Direct-acting antivirals eliminate hepatitis C virus (HCV) in more than 95% of treated individuals and may abolish liver injury, arrest fibrogenesis, and reverse fibrosis and cirrhosis. However, liver regeneration is usually a slow process that is less effective in the late stages of fibrosis. What is more, fibrogenesis may prevail in patients with advanced cirrhosis, where it can progress to liver failure and hepatocellular carcinoma. Therefore, the development of antifibrotic drugs that halt and reverse fibrosis progression is urgently needed. Fibrosis occurs due to the repair process of damaged hepatic tissue, which eventually leads to scarring. The innate immune response against HCV is essential in the initiation and progression of liver fibrosis. HCV-infected hepatocytes and liver macrophages secrete proinflammatory cytokines and chemokines that promote the activation and differentiation of hepatic stellate cells (HSCs) to myofibroblasts that produce extracellular matrix (ECM) components. Prolonged ECM production by myofibroblasts due to chronic inflammation is essential to the development of fibrosis. While no antifibrotic therapy is approved to date, several drugs are being tested in phase 2 and phase 3 trials with promising results. This review discusses current state-of-the-art knowledge on treatments targeting the innate immune system to revert chronic hepatitis C-associated liver fibrosis. Agents that cause liver damage may vary (alcohol, virus infection, etc.), but fibrosis progression shows common patterns among them, including chronic inflammation and immune dysregulation, hepatocyte injury, HSC activation, and excessive ECM deposition. Therefore, mechanisms underlying these processes are promising targets for general antifibrotic therapies.
Collapse
Affiliation(s)
- Daniel Sepulveda-Crespo
- Unidad de Infección Viral e Inmunidad, Centro Nacional de Microbiología, Instituto de Salud Carlos III (Campus Majadahonda), Carretera Majadahonda-Pozuelo, Km 2.2, 28220, Majadahonda, Madrid, Spain
| | - Salvador Resino
- Unidad de Infección Viral e Inmunidad, Centro Nacional de Microbiología, Instituto de Salud Carlos III (Campus Majadahonda), Carretera Majadahonda-Pozuelo, Km 2.2, 28220, Majadahonda, Madrid, Spain.
| | - Isidoro Martinez
- Unidad de Infección Viral e Inmunidad, Centro Nacional de Microbiología, Instituto de Salud Carlos III (Campus Majadahonda), Carretera Majadahonda-Pozuelo, Km 2.2, 28220, Majadahonda, Madrid, Spain.
| |
Collapse
|
26
|
Thimme R. T cell immunity to hepatitis C virus: Lessons for a prophylactic vaccine. J Hepatol 2021; 74:220-229. [PMID: 33002569 DOI: 10.1016/j.jhep.2020.09.022] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Revised: 09/22/2020] [Accepted: 09/22/2020] [Indexed: 12/12/2022]
Abstract
There is consensus that HCV-specific T cells play a central role in the outcome (clearance vs. persistence) of acute infection and that they contribute to protection against the establishment of persistence after reinfection. However, these T cells often fail and the virus can persist, largely as a result of T cell exhaustion and the emergence of viral escape mutations. Importantly, HCV cure by direct-acting antivirals does not lead to a complete reversion of T cell exhaustion and thus HCV reinfections can occur. The current lack of detailed knowledge about the immunological determinants of viral clearance, persistence and protective immunity is a major roadblock to the development of a prophylactic T cell vaccine. This minireview highlights the basic concepts of successful T cell immunity, major mechanisms of T cell failure and how our understanding of these concepts can be translated into a prophylactic vaccine.
Collapse
Affiliation(s)
- Robert Thimme
- Department of Medicine II, Gastroenterology, Hepatology, Endocrinology and Infectious Diseases, Medical Center - University of Freiburg, Faculty of Medicine, Germany.
| |
Collapse
|
27
|
Colomba GME, Urone N, Marco VD, Ferraro D. Phylodynamic Analysis and Implication of HCV Genotype 4 Variability on Antiviral Drug Response and T-Cell Recognition. Viruses 2020; 12:E1363. [PMID: 33260596 PMCID: PMC7761199 DOI: 10.3390/v12121363] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Revised: 11/22/2020] [Accepted: 11/25/2020] [Indexed: 12/12/2022] Open
Abstract
Therapies for HCV care could change the prevalence and the geographic distribution of genotypes due to differences in Sustained Virologic Response (SVR). In this scenario, uncommon genotypes/subtypes, such as genotype 4, could spread from high-risk groups, replacing genotypes eradicated by antiviral drugs. Genotype eradication is also strongly influenced by the CD8+ T cell response. In this study, the genetic variability in HCV genotype 4 strains obtained from a cohort of 67 patients naïve to DAA therapy was evaluated. We found that the presence of resistance-associated substitutions (RAS) was able to affect drug responses. Next, using a prediction tool, viral mutations were identified by their ability, or lack thereof, to reduce the binding affinity with HLA, which affects T cell recognition. The Bayesian coalescent analysis suggested two different circulation clusters, one in risk groups (IDUs and MSM) and the other due to migration flows, dated to 1940 and 1915, respectively. Most of the RAS overlapped with HLA and a lack of binding mutations was observed in 96% of strains. This study describes the introduction of HCV genotype 4 in a region of the Mediterranean basin and evaluates how HCV genotype 4's genetic variability could affect the response of antiviral drugs and CD8+ T cell recognition.
Collapse
Affiliation(s)
| | | | | | - Donatella Ferraro
- Dipartimento di Scienze per la Promozione della Salute, Materno-Infantile di Medicina Interna e Specialistica di Eccellenza “G. D’Alessandro”, 90133 Palermo, Italy; (G.M.E.C.); (N.U.); (V.d.M.)
| |
Collapse
|
28
|
Batatinha H, Tavares-Silva E, Leite GSF, Resende AS, Albuquerque JAT, Arslanian C, Fock RA, Lancha AH, Lira FS, Krüger K, Thomatieli-Santos R, Rosa-Neto JC. Probiotic supplementation in marathonists and its impact on lymphocyte population and function after a marathon: a randomized placebo-controlled double-blind study. Sci Rep 2020; 10:18777. [PMID: 33139757 PMCID: PMC7608678 DOI: 10.1038/s41598-020-75464-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Accepted: 09/03/2020] [Indexed: 11/16/2022] Open
Abstract
Probiotic supplementation arises as playing an immune-stimulatory role. High-intensity and -volume exercise can inhibit immune cell function, which threatens athletic performance and recovery. We hypothesized that 30 days of probiotic supplementation could stabilize the immune system of athletes preventing immune suppression after a marathon race. Twenty-seven male marathonists were double-blinded randomly into probiotic (Bifidobacterium-animalis-subsp.-Lactis (10 × 109) and Lactobacillus-Acidophilus (10 × 109) + 5 g of maltodextrin) and placebo (5 g of maltodextrin) group. They received 30 sachets and supplemented 1 portion/day during 30 days before the race. Blood were collected 30 days before (rest), 1 day before (pre), 1 h after (post) and 5 days after the race (recovery). Both chronic and acute exercise modulated a different T lymphocyte population (CD3+CD4−CD8− T-cells), increasing pre-race, decreasing post and returning to rest values at the recovery. The total number of CD8 T cell and the memory subsets statistically decreased only in the placebo group post-race. Pro-inflammatory cytokine production by stimulated lymphocytes decreased in the probiotic group after the supplementation period. 30 days of probiotic supplementation maintained CD8 T cell and effector memory cell population and played an immunomodulatory role in stimulated lymphocytes. Both, training and marathon modulated a non-classical lymphocyte population regardless of probiotic supplementation.
Collapse
Affiliation(s)
- Helena Batatinha
- Immunometabolism Research Group, Institute of Biomedical Sciences, University of São Paulo (USP), São Paulo, SP, Brazil. .,Department of Cell and Developmental Biology, University of São Paulo,, 1524, Prof Lineu Prestes Av., Sao Paulo, SP, 05508-000, Brazil.
| | - Edgar Tavares-Silva
- Programa de pós-graduação em psicobiologia, Universidade Federal de São Paulo, Santos, Brazil
| | - Geovana S F Leite
- Laboratory of Applied Nutrition and Metabolism, School of Physical Education and Sports, University of São Paulo, São Paulo, Brazil
| | - Ayane S Resende
- Laboratory of Applied Nutrition and Metabolism, School of Physical Education and Sports, University of São Paulo, São Paulo, Brazil
| | - José A T Albuquerque
- Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Christina Arslanian
- Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Ricardo A Fock
- Department of Clinical and Toxicological Analysis, School of Pharmaceutical Sciences, University of São Paulo, São Paulo, Brazil
| | - Antônio H Lancha
- Laboratory of Applied Nutrition and Metabolism, School of Physical Education and Sports, University of São Paulo, São Paulo, Brazil
| | - Fabio S Lira
- Exercise and Immunometabolism Research Group, Post-Graduation Program in Movement Sciences, Department of Physical Education, Universidade Estadual Paulista (UNESP), School of Technology and Sciences, Presidente Prudente, Brazil
| | - Karsten Krüger
- Department of Exercise Physiology and Sports Therapy, Justus-Liebig-University Giessen, Giessen, Germany
| | - Ronaldo Thomatieli-Santos
- Programa de pós-graduação em psicobiologia, Universidade Federal de São Paulo, Santos, Brazil.,Department of Bioscience, Universidade Federal de São Paulo, Santos, Brazil
| | - José C Rosa-Neto
- Immunometabolism Research Group, Institute of Biomedical Sciences, University of São Paulo (USP), São Paulo, SP, Brazil
| |
Collapse
|
29
|
Hartnell F, Esposito I, Swadling L, Brown A, Phetsouphanh C, de Lara C, Gentile C, Turner B, Dorrell L, Capone S, Folgori A, Barnes E, Klenerman P. Characterizing Hepatitis C Virus-Specific CD4 + T Cells Following Viral-Vectored Vaccination, Directly Acting Antivirals, and Spontaneous Viral Cure. Hepatology 2020; 72:1541-1555. [PMID: 32012325 PMCID: PMC7610807 DOI: 10.1002/hep.31160] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/07/2019] [Accepted: 01/07/2020] [Indexed: 12/21/2022]
Abstract
BACKGROUND AND AIMS Induction of functional helper CD4+ T cells is the hallmark of a protective immune response against hepatitis C virus (HCV), associated with spontaneous viral clearance. Heterologous prime/boost viral vectored vaccination has demonstrated induction of broad and polyfunctional HCV-specific CD8+ T cells in healthy volunteers; however, much less is known about CD4+ T-cell subsets following vaccination. APPROACH AND RESULTS We analyzed HCV-specific CD4+ T-cell populations using major histocompatibility complex class II tetramers in volunteers undergoing HCV vaccination with recombinant HCV adenoviral/modified vaccinia Ankara viral vectors. Peptide-specific T-cell responses were tracked over time, and functional (proliferation and cytokine secretion) and phenotypic (cell surface and intranuclear) markers were assessed using flow cytometry. These were compared to CD4+ responses in 10 human leukocyte antigen-matched persons with HCV spontaneous resolution and 21 chronically infected patients treated with directly acting antiviral (DAA) therapy. Vaccination induced tetramer-positive CD4+ T cells that were highest 1-4 weeks after boosting (mean, 0.06%). Similar frequencies were obtained for those tracked following spontaneous resolution of disease (mean, 0.04%). In addition, the cell-surface phenotype (CD28, CD127) memory subset markers and intranuclear transcription factors, as well as functional capacity of peptide-specific CD4+ T-cell responses characterized after vaccination, are comparable to those following spontaneous viral resolution. In contrast, helper responses in chronic infection were infrequently detected and poorly functional and did not consistently recover following HCV cure. CONCLUSIONS Helper CD4+ T-cell phenotype and function following HCV viral vectored vaccination resembles "protective memory" that is observed following spontaneous clearance of HCV. DAA cure does not promote resurrection of exhausted CD4+ T-cell memory in chronic infection.
Collapse
Affiliation(s)
- Felicity Hartnell
- Peter Medawar Building for Pathogen ResearchUniversity of OxfordOxfordUnited Kingdom
| | - Ilaria Esposito
- Peter Medawar Building for Pathogen ResearchUniversity of OxfordOxfordUnited Kingdom
| | - Leo Swadling
- Peter Medawar Building for Pathogen ResearchUniversity of OxfordOxfordUnited Kingdom
| | - Anthony Brown
- Peter Medawar Building for Pathogen ResearchUniversity of OxfordOxfordUnited Kingdom
| | | | - Catherine de Lara
- Peter Medawar Building for Pathogen ResearchUniversity of OxfordOxfordUnited Kingdom
| | | | - Bethany Turner
- Jenner Vaccine TrialsNuffield Department of MedicineUniversity of OxfordOxfordUnited Kingdom
| | - Lucy Dorrell
- Jenner Vaccine TrialsNuffield Department of MedicineUniversity of OxfordOxfordUnited Kingdom
| | | | | | - Eleanor Barnes
- Peter Medawar Building for Pathogen ResearchUniversity of OxfordOxfordUnited Kingdom,Jenner Vaccine TrialsNuffield Department of MedicineUniversity of OxfordOxfordUnited Kingdom,NIHR Biomedical Research Centre OxfordJohn Radcliffe HospitalOxfordUnited Kingdom,Translational Gastroenterology UnitJohn Radcliffe HospitalOxfordUnited Kingdom
| | - Paul Klenerman
- Peter Medawar Building for Pathogen ResearchUniversity of OxfordOxfordUnited Kingdom,Jenner Vaccine TrialsNuffield Department of MedicineUniversity of OxfordOxfordUnited Kingdom,NIHR Biomedical Research Centre OxfordJohn Radcliffe HospitalOxfordUnited Kingdom,Translational Gastroenterology UnitJohn Radcliffe HospitalOxfordUnited Kingdom
| |
Collapse
|
30
|
Origin and fine-tuning of effector CD8 T cell subpopulations in chronic infection. Curr Opin Virol 2020; 46:27-35. [PMID: 33137688 DOI: 10.1016/j.coviro.2020.10.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Revised: 09/27/2020] [Accepted: 10/11/2020] [Indexed: 12/31/2022]
Abstract
Persisting stimulation can skew CD8 T cells towards a hypofunctional state commonly referred to as T cell exhaustion. This functional attenuation likely constitutes a mechanism which evolved to balance T cell mediated viral control versus overwhelming immunopathology. Here, we highlight the recent progress in defining the genetic mechanisms and factors shaping the differentiation of exhausted CD8 T cells. We review how the transcription factor Tox imposes an exhausted phenotype in the Tcf1+ progenitors and how CD4 help fine-tunes the effector subsets that emerge from this progenitor population. Both processes critically shape the spectrum of effector function performed by CD8 T cells and the level of resulting virus control. Finally, we discuss how these insights can be exploited to boost the immune response in chronic infection and cancer.
Collapse
|
31
|
Verdon DJ, Mulazzani M, Jenkins MR. Cellular and Molecular Mechanisms of CD8 + T Cell Differentiation, Dysfunction and Exhaustion. Int J Mol Sci 2020; 21:ijms21197357. [PMID: 33027962 PMCID: PMC7582856 DOI: 10.3390/ijms21197357] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 10/01/2020] [Accepted: 10/02/2020] [Indexed: 02/07/2023] Open
Abstract
T cells follow a triphasic distinct pathway of activation, proliferation and differentiation before becoming functionally and phenotypically “exhausted” in settings of chronic infection, autoimmunity and in cancer. Exhausted T cells progressively lose canonical effector functions, exhibit altered transcriptional networks and epigenetic signatures and gain constitutive expression of a broad coinhibitory receptor suite. This review outlines recent advances in our understanding of exhausted T cell biology and examines cellular and molecular mechanisms by which a state of dysfunction or exhaustion is established, and mechanisms by which exhausted T cells may still contribute to pathogen or tumour control. Further, this review describes our understanding of exhausted T cell heterogeneity and outlines the mechanisms by which checkpoint blockade differentially engages exhausted T cell subsets to overcome exhaustion and recover T cell function.
Collapse
Affiliation(s)
- Daniel J. Verdon
- Immunology Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia; (D.J.V.); (M.M.)
| | - Matthias Mulazzani
- Immunology Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia; (D.J.V.); (M.M.)
| | - Misty R. Jenkins
- Immunology Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia; (D.J.V.); (M.M.)
- Department of Medical Biology, The University of Melbourne, Parkville, VIC 3052, Australia
- Institute of Molecular Science, La Trobe University, Bundoora, VIC 3086, Australia
- Correspondence:
| |
Collapse
|
32
|
Saeed A, Hildebrand H, Park R, Al-Jumayli M, Abbasi S, Melancon T, Saeed A, Al-Rajabi R, Kasi A, Baranda J, Williamson S, Sun W. Immune Checkpoint Inhibitors versus VEGF Targeted Therapy as Second Line Regimen in Advanced Hepatocellular Carcinoma (HCC): A Retrospective Study. J Clin Med 2020; 9:E2682. [PMID: 32824968 PMCID: PMC7563439 DOI: 10.3390/jcm9092682] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Revised: 08/07/2020] [Accepted: 08/14/2020] [Indexed: 12/26/2022] Open
Abstract
Several targeted agents including multi-tyrosine kinase inhibitors (mTKIs) and immunotherapy (IO) agents have been approved for use beyond the frontline setting in patients with advanced hepatocellular carcinoma (HCC). Due to lack of prospective head-to-head comparative trials, there is no standardized way for alternating those agents beyond frontline. Therefore, we performed a retrospective review of the Kansas University (KU) cancer registry to determine whether IO may be superior to non-IO therapy. Patients with advanced HCC were divided into two groups based on the second-line systemic regimen received (IO vs. non-IO). Progression-free survival (PFS) and overall survival (OS) were calculated under the Kaplan-Meier and Cox proportional hazards models. No statistically significant differences in PFS and OS were found, although a non-significant delayed separation in the survival curve favoring IO was identified (median PFS 3.9 months vs. 3 months; median OS 10 months vs. 10 months respectively for IO vs. non-IO). This retrospective analysis is one of the earliest and largest studies comparing second-line IO and non-IO therapies thus far reported. Future studies should aim to define specific biomarkers for response prediction and treatment optimization based on individual patient and tumor characteristics. Furthermore, combinatorial therapeutic strategies is an evolving approach showing early promising signal.
Collapse
Affiliation(s)
- Anwaar Saeed
- Department of Medicine, Division of Medical Oncology, Gastrointestinal Oncology Program, Kansas University Cancer Center, Kansas City, KS 66205, USA; (H.H.); (S.A.); (T.M.); (R.A.-R.); (A.K.); (J.B.); (S.W.); (W.S.)
| | - Hannah Hildebrand
- Department of Medicine, Division of Medical Oncology, Gastrointestinal Oncology Program, Kansas University Cancer Center, Kansas City, KS 66205, USA; (H.H.); (S.A.); (T.M.); (R.A.-R.); (A.K.); (J.B.); (S.W.); (W.S.)
| | - Robin Park
- MetroWest Medical Center, Tufts University School of Medicine, Framingham, MA 02111, USA;
| | - Mohammed Al-Jumayli
- Department of Medicine, Division of Medical Oncology, University of South Florida, Moffitt Cancer Center, Tampa, FL 33620, USA;
| | - Saqib Abbasi
- Department of Medicine, Division of Medical Oncology, Gastrointestinal Oncology Program, Kansas University Cancer Center, Kansas City, KS 66205, USA; (H.H.); (S.A.); (T.M.); (R.A.-R.); (A.K.); (J.B.); (S.W.); (W.S.)
| | - Tina Melancon
- Department of Medicine, Division of Medical Oncology, Gastrointestinal Oncology Program, Kansas University Cancer Center, Kansas City, KS 66205, USA; (H.H.); (S.A.); (T.M.); (R.A.-R.); (A.K.); (J.B.); (S.W.); (W.S.)
| | - Azhar Saeed
- Department of Pathology and Laboratory Medicine, Kansas University Medical Center, Kansas City, KS 66160, USA;
| | - Raed Al-Rajabi
- Department of Medicine, Division of Medical Oncology, Gastrointestinal Oncology Program, Kansas University Cancer Center, Kansas City, KS 66205, USA; (H.H.); (S.A.); (T.M.); (R.A.-R.); (A.K.); (J.B.); (S.W.); (W.S.)
| | - Anup Kasi
- Department of Medicine, Division of Medical Oncology, Gastrointestinal Oncology Program, Kansas University Cancer Center, Kansas City, KS 66205, USA; (H.H.); (S.A.); (T.M.); (R.A.-R.); (A.K.); (J.B.); (S.W.); (W.S.)
| | - Joaquina Baranda
- Department of Medicine, Division of Medical Oncology, Gastrointestinal Oncology Program, Kansas University Cancer Center, Kansas City, KS 66205, USA; (H.H.); (S.A.); (T.M.); (R.A.-R.); (A.K.); (J.B.); (S.W.); (W.S.)
| | - Stephen Williamson
- Department of Medicine, Division of Medical Oncology, Gastrointestinal Oncology Program, Kansas University Cancer Center, Kansas City, KS 66205, USA; (H.H.); (S.A.); (T.M.); (R.A.-R.); (A.K.); (J.B.); (S.W.); (W.S.)
| | - Weijing Sun
- Department of Medicine, Division of Medical Oncology, Gastrointestinal Oncology Program, Kansas University Cancer Center, Kansas City, KS 66205, USA; (H.H.); (S.A.); (T.M.); (R.A.-R.); (A.K.); (J.B.); (S.W.); (W.S.)
| |
Collapse
|
33
|
Churov AV, Mamashov KY, Novitskaia AV. Homeostasis and the functional roles of CD4 + Treg cells in aging. Immunol Lett 2020; 226:83-89. [PMID: 32717201 DOI: 10.1016/j.imlet.2020.07.004] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Revised: 07/08/2020] [Accepted: 07/15/2020] [Indexed: 02/06/2023]
Abstract
OBJECTIVE An upward trend in life expectancy has been observed in a majority of developed countries and leading to increasing in aging-related diseases. Aging is a risk factor for the development of widespread clinical conditions such as cardiovascular and autoimmune diseases, cancer, infections. Although studies have been very active, the problem of aging still remains one of the most obscure aspects of human biology. Regulatory T (Treg) cells with immunosuppressive properties have a pivotal role in the maintenance of immune homeostasis. Alterations in Treg cell functionality appear to be of great importance in the development of immune senescence and contribute to increased susceptibility to immune-mediated diseases with age. DESIGN This review highlights recent findings regarding the age-related changes in the numbers and functional activity of human Tregs. Some of the mechanisms that maintain the balance of Tregs during human aging are discussed. The possible roles of Tregs in the pathogenesis of diseases associated with advanced age are also considered. RESULTS Age-related systemic changes, such as thymic involution, hormonal status, and epigenetic modifications, may affect the state of the Treg population and trigger various diseases. These changes involve decline or amplification in the functional activity of Tregs, an increase in the memory Treg subset and shifting of a Th17/Treg balance. CONCLUSION Taken together, the reviewed data suggest equal or even increased Treg functionality with age. Thus, age-mediated Treg expansion and higher Treg activity may contribute to elevated immune suppression and increased risk of infections and cancer.
Collapse
Affiliation(s)
- Alexey V Churov
- Institute of Biology, Karelian Research Centre, Russian Academy of Sciences, Petrozavodsk, Russia.
| | | | - Anastasiia V Novitskaia
- Institute of Biology, Karelian Research Centre, Russian Academy of Sciences, Petrozavodsk, Russia
| |
Collapse
|
34
|
Im SJ, Ha SJ. Re-defining T-Cell Exhaustion: Subset, Function, and Regulation. Immune Netw 2020; 20:e2. [PMID: 32158590 PMCID: PMC7049579 DOI: 10.4110/in.2020.20.e2] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Revised: 02/11/2020] [Accepted: 02/13/2020] [Indexed: 12/29/2022] Open
Abstract
Acute viral infection or vaccination generates highly functional memory CD8 T cells following the Ag resolution. In contrast, persistent antigenic stimulation in chronic viral infection and cancer leads to a state of T-cell dysfunction termed T-cell exhaustion. We and other have recently identified a novel subset of exhausted CD8 T cells that act as stem cells for maintaining virus-specific CD8 T cells in a mouse model of chronic lymphocytic choriomeningitis virus infection. This stem cell-like CD8 T-cell subset has been also observed in both mouse and human tumor models. Most importantly, in both chronic viral infection and tumor models, the proliferative burst of Ag-specific CD8 T cells driven by PD-1-directed immunotherapy comes exclusively from this stem cell-like CD8 T-cell subset. Therefore, a better understanding of the mechanisms how CD8 T-cell subsets are regulated during chronic viral infection and cancer is required to improve the current immunotherapies that restore the function of exhausted CD8 T cells. In this review, we discuss the differentiation of virus-specific CD8 T cells during chronic viral infection, the characteristics and function of CD8 T-cell subsets, and the therapeutic intervention of PD-1-directed immunotherapy in cancer.
Collapse
Affiliation(s)
- Se Jin Im
- Emory Vaccine Center and Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, GA 30033, USA.,Department of Immunology, Sungkyunkwan University School of Medicine, Suwon 16419, Korea
| | - Sang-Jun Ha
- Department of Biochemistry, College of Life Science & Biotechnology, Yonsei University, Seoul 03722, Korea
| |
Collapse
|
35
|
Ferrari C, Barili V, Varchetta S, Mondelli MU. Immune Mechanisms of Viral Clearance and Disease Pathogenesis During Viral Hepatitis. THE LIVER 2020:821-850. [DOI: 10.1002/9781119436812.ch63] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
36
|
Chen DY, Wolski D, Aneja J, Matsubara L, Robilotti B, Hauck G, de Sousa PSF, Subudhi S, Fernandes CA, Hoogeveen RC, Kim AY, Lewis-Ximenez L, Lauer GM. Hepatitis C virus-specific CD4+ T cell phenotype and function in different infection outcomes. J Clin Invest 2020; 130:768-773. [PMID: 31904582 PMCID: PMC6994113 DOI: 10.1172/jci126277] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Accepted: 10/24/2019] [Indexed: 01/20/2023] Open
Abstract
CD4+ T cell failure is a hallmark of chronic hepatitis C virus (HCV) infection. However, the mechanisms underlying the impairment and loss of virus-specific CD4+ T cells in persisting HCV infection remain unclear. Here we examined HCV-specific CD4+ T cells longitudinally during acute infection with different infection outcomes. We found that HCV-specific CD4+ T cells are characterized by expression of a narrower range of T cell inhibitory receptors compared with CD8+ T cells, with initially high expression levels of PD-1 and CTLA-4 that were associated with negative regulation of proliferation in all patients, irrespective of outcome. In addition, HCV-specific CD4+ T cells were phenotypically similar during early resolving and persistent infection and secreted similar levels of cytokines. However, upon viral control, CD4+ T cells quickly downregulated inhibitory receptors and differentiated into long-lived memory cells. In contrast, persisting viremia continued to drive T cell activation and PD-1 and CTLA-4 expression, and blocked T cell differentiation, until the cells quickly disappeared from the circulation. Our data support an important and physiological role for inhibitory receptor-mediated regulation of CD4+ T cells in early HCV infection, irrespective of outcome, with persistent HCV viremia leading to sustained upregulation of PD-1 and CTLA-4.
Collapse
Affiliation(s)
| | | | - Jasneet Aneja
- Gastrointestinal Unit and
- Infectious Disease Division, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | | | | | | | | | | | | | | | - Arthur Y. Kim
- Infectious Disease Division, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Lia Lewis-Ximenez
- Viral Hepatitis Laboratory, Oswaldo Cruz Institute, Fundação Oswaldo Cruz, Rio de Janeiro, Brazil
| | | |
Collapse
|
37
|
Lombardi A, Mondelli MU. Review article: immune checkpoint inhibitors and the liver, from therapeutic efficacy to side effects. Aliment Pharmacol Ther 2019; 50:872-884. [PMID: 31378985 DOI: 10.1111/apt.15449] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/22/2019] [Revised: 06/21/2019] [Accepted: 07/13/2019] [Indexed: 12/14/2022]
Abstract
BACKGROUND Immune checkpoint inhibitors have revolutionised the oncological landscape in the last few years. Possible applications include the treatment of hepatocellular carcinoma and cholangiocarcinoma. Unfortunately, new immune-related adverse effects have been associated with the use of these agents and the liver is one of the organs most frequently involved. AIMS To provide a general overview of the potential impact of immune checkpoint inhibitors on the liver METHODS: We reviewed the literature and abstracts/presentations on immune checkpoint inhibitors at most relevant hepatology meetings over the last 5 years. RESULTS The role of immune checkpoint inhibitors has been investigated both for the treatment of viral hepatitis and primary liver cancer. Hepatocellular carcinoma and chronic hepatitis B show the greatest potential for treatment with these drugs in the near future. However, immune-related adverse events involving the liver are a growing concern related to their widespread use. CONCLUSIONS Immune checkpoint inhibitors represent an exciting new class of drugs with currently limited application in malignant and non-malignant liver disease. Caution must be exercised on the emergence of potentially severe immune adverse reactions.
Collapse
Affiliation(s)
- Andrea Lombardi
- Division of Infectious Diseases II and Immunology, Department of Medical Sciences and Infectious Diseases, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy.,Department of Internal Medicine and Therapeutics, University of Pavia, Pavia, Italy
| | - Mario U Mondelli
- Division of Infectious Diseases II and Immunology, Department of Medical Sciences and Infectious Diseases, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy.,Department of Internal Medicine and Therapeutics, University of Pavia, Pavia, Italy
| |
Collapse
|
38
|
Vranjkovic A, Deonarine F, Kaka S, Angel JB, Cooper CL, Crawley AM. Direct-Acting Antiviral Treatment of HCV Infection Does Not Resolve the Dysfunction of Circulating CD8 + T-Cells in Advanced Liver Disease. Front Immunol 2019; 10:1926. [PMID: 31456810 PMCID: PMC6700371 DOI: 10.3389/fimmu.2019.01926] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2018] [Accepted: 07/30/2019] [Indexed: 02/06/2023] Open
Abstract
Chronic hepatitis C virus (HCV) infection disrupts immune functions, including that of cytotoxic CD8+ T-cells which are important mediators of immune response. While HCV cure aims to eliminate long term sequelae of infection, whether direct-acting antiviral (DAA) treatment results in immune reconstitution remains unclear. We and others have reported generalized CD8+ T-cell dysfunction in chronic HCV infection and our research suggests that the degree of liver damage is a factor in this process. Our recent research indicates that liver fibrosis is not readily reversed after DAA-mediated clearance of chronic HCV infection. We therefore examined the function of circulating CD8+ T-cell subsets in chronic HCV infection in the context of liver fibrosis severity, determined by ultrasound elastography and Metavir F-score system. We observed progressive shifts in CD8+ T-cell subset distribution in HCV-infected individuals with advanced liver fibrosis (F4) compared to minimal fibrosis (F0-1) or uninfected controls, and this remained unchanged after viral cure. Impaired CD8+ T-cell function was observed as a reduced proportion of CD107+ and perforin+ late effector memory cells in HCV+(F4) and HCV+(F0-1) individuals, respectively. In HCV+(F4) individuals, nearly all CD8+ T-cell subsets had an elevated proportion of perforin+ cells while naïve cells had increased proportions of IFN-γ+ and CD107+ cells. These exaggerated CD8+ T-cell activities were not resolved when evaluated 24 weeks after completion of DAA therapy and HCV clearance. This was further supported by sustained, high levels of cell proliferation and cytolytic activity. Furthermore, DAA therapy had no effect on elevated concentrations of systemic inflammatory cytokines and decreased levels of inhibitory TGF-β in the plasma of HCV+(F4) individuals, suggesting HCV infection and advanced liver disease result in a long-lasting immune activating microenvironment. These data demonstrate that in chronic HCV infection, liver fibrosis severity is associated with generalized hyperfunctional CD8+ T-cells, particularly with perforin production and cytotoxicity, and this persists after viral clearance. Whether DAA therapy will eliminate other related long-term sequelae in HCV+(F4) individuals remains an important research question.
Collapse
Affiliation(s)
- Agatha Vranjkovic
- Chronic Disease Program, Ottawa Hospital Research Institute, Ottawa, ON, Canada
| | - Felicia Deonarine
- Chronic Disease Program, Ottawa Hospital Research Institute, Ottawa, ON, Canada.,Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, ON, Canada
| | - Shaima Kaka
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, ON, Canada
| | - Jonathan B Angel
- Chronic Disease Program, Ottawa Hospital Research Institute, Ottawa, ON, Canada.,Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, ON, Canada.,Division of Infectious Diseases, The Ottawa Hospital, Ottawa, ON, Canada
| | - Curtis L Cooper
- Division of Infectious Diseases, The Ottawa Hospital, Ottawa, ON, Canada.,School of Epidemiology and Public Health, University of Ottawa, Ottawa, ON, Canada.,Clinical Epidemiology Program, Ottawa Hospital Research Institute, Ottawa, ON, Canada
| | - Angela M Crawley
- Chronic Disease Program, Ottawa Hospital Research Institute, Ottawa, ON, Canada.,Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, ON, Canada.,Division of Infectious Diseases, The Ottawa Hospital, Ottawa, ON, Canada.,Department of Biology, Carleton University, Ottawa, ON, Canada
| |
Collapse
|
39
|
Chronic Hepatitis C: Conspectus of immunological events in the course of fibrosis evolution. PLoS One 2019; 14:e0219508. [PMID: 31318916 PMCID: PMC6638930 DOI: 10.1371/journal.pone.0219508] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2019] [Accepted: 06/25/2019] [Indexed: 01/18/2023] Open
Abstract
In chronically infected HCV patients emergence and evolution of fibrosis, as a consequence of virus persistence, can be considered as an indicator of disease advancement. Therefore the aim of this study was to correlate alterations of immune response in chronic HCV patients with liver histopathology. Sera cytokine levels and frequency of circulating and liver infiltrating cells were evaluated using 13plex Kit Flow Cytomix, flow cytometry and immunohistochemistry. We found that the number of circulating T lymphocytes (including CD4+, CD8+ and Treg) and B lymphocytes, as well as DCs, was higher in patients with no fibrosis than in healthy subjects. In patients with fibrosis frequency of these cells decreased, and contrarily, in the liver, number of T and B lymphocytes gradually increased with fibrosis. Importantly, in patients with advanced fibrosis, liver infiltrating regulatory T cells and DC-SIGN+ mononuclear cells with immunosuppressive and wound-healing effector functions were abundantly present. Cytokine profiling showed predominance of proinflammatory cytokines in patients with no fibrosis and a tendency of decline in level of all cytokines with severity of liver injury. Lower but sustained IL-4 production refers to Th2 predominance in higher stages of fibrosis. Altogether, our results reveal graduall alterations of immunological parameters during fibrosis evolution and illustrate the course of immunological events through disease progression.
Collapse
|
40
|
Abd Ellah NH, Tawfeek HM, John J, Hetta HF. Nanomedicine as a future therapeutic approach for Hepatitis C virus. Nanomedicine (Lond) 2019; 14:1471-1491. [PMID: 31166139 DOI: 10.2217/nnm-2018-0348] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Hepatitis C virus (HCV) is not easily cleared from the human body and in most cases turned into chronic infection. This chronicity is a major cause of liver damage, cirrhosis and hepatocellular carcinoma. Therefore, immediate detection and treatment of HCV guarantees eradication of the virus and prevention of chronicity complications. Since discovery of HCV in 1989, several emerging treatments were developed such as polyethylene glycol(PEG)-ylated interferon/ribavirin, direct acting antivirals and host targeting antivirals. Despite the progress in anti-HCV therapy, there is still a pressing need of new approaches for affordable and effective drug delivery systems using nanomedicine. In this review, the contribution of nanoparticles as a promising delivery system for HCV immunizing, diagnostic and therapeutic agents are discussed.
Collapse
Affiliation(s)
- Noura H Abd Ellah
- Division of Pharmaceutical Sciences, James L. Winkle College of Pharmacy, Medical Sciences Building, University of Cincinnati, Cincinnati, OH 45267, USA.,Department of Pharmaceutics, Faculty of Pharmacy, Assiut University, Assiut, 71526, Egypt
| | - Hesham M Tawfeek
- Department of Industrial Pharmacy, Faculty of Pharmacy, Assiut University, Assiut, 71526, Egypt.,Department of Pharmaceutics & Pharmaceutical Technology, Faculty of Pharmacy, Mutah University, Karak, Jordan
| | - James John
- Central Research Facilities, Sri Ramachandra institute of higher education & research, Sri Ramachandra University, Chennai, India
| | - Helal F Hetta
- Department of Medical Microbiology & Immunology, Faculty of Medicine, Assiut University, Assiut, 71526, Egypt.,Department of Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, OH 45267-0595, USA
| |
Collapse
|
41
|
Ghazal K, Morales O, Barjon C, Dahlqvist G, Aoudjehane L, Ouaguia L, Delhem N, Conti F. Early high levels of regulatory T cells and T helper 1 may predict the progression of recurrent hepatitis C after liver transplantation. Clin Res Hepatol Gastroenterol 2019; 43:273-281. [PMID: 30713032 DOI: 10.1016/j.clinre.2018.10.005] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2018] [Revised: 09/25/2018] [Accepted: 10/02/2018] [Indexed: 02/06/2023]
Abstract
BACKGROUND Immune response failure against hepatitis C virus (HCV) has been associated with an increased regulatory T cell (Treg) activity. After liver transplantation (LT), 80% of patients experience an accelerated progression of hepatitis C recurrence. The aim of this work was to assess the involvement of Tregs, T helper (Th) 1, 2 and 17 cells in recurrent hepatitis C. METHODS Peripheral blood cells obtained before and one month after LT from 22 recipients were analysed. Forty-four key molecules related to Treg, Th1, 2 and 17 responses, were evaluated using qRT-PCR. Liver recipients were classified in two groups according to graft fibrosis evaluated by the METAVIR score on the biopsy performed one year after LT (mild: F ≤ 1, n = 13; severe: F > 1, n = 9). Patients developing a severe recurrence were compared with patients with a mild recurrence. RESULTS mRNA levels of Treg markers obtained one month after LT were significantly increased in patients with a severe disease course when compared to patients with a mild recurrence. Markers of the Th1 response were elevated in the same group. No differences in the markers determined before LT were observed. CONCLUSION These findings suggest that Treg, induced by a multifactorial process, which could include a strong Th1 response itself, may play a role in suppressing the early antiviral response, leading to a severe recurrence of hepatitis C.
Collapse
Affiliation(s)
- K Ghazal
- Sorbonne Universités, UPMC Université Paris 06, INSERM, UMR_S 938, Saint-Antoine Research Center, 75012 Paris, France; AP-HP, Bicêtre Hospital, Biochemistry Laboratory, 94275 Le Kremlin-Bicêtre cedex, France.
| | - O Morales
- CNRS, UMR8161, Institut de Biologie de Lille, Université de Lille, Institut Pasteur de Lille, IFR 142, 59021 Lille cedex, France
| | - C Barjon
- Sorbonne Universités, UPMC Université Paris 06, INSERM, UMR_S 938, Saint-Antoine Research Center, 75012 Paris, France; De Duve Institute, Université catholique de Louvain, 1200 Brussels, Belgium
| | - G Dahlqvist
- Sorbonne Universités, UPMC Université Paris 06, INSERM, UMR_S 938, Saint-Antoine Research Center, 75012 Paris, France; Cliniques Universitaires Saint-Luc, 1200 Woluwe-Saint-Lambert, Belgium
| | - L Aoudjehane
- Sorbonne Universités, UPMC Université Paris 06, INSERM, UMR_S 938, Saint-Antoine Research Center, 75012 Paris, France; Institute of Cardiometabolism and Nutrition (ICAN), 75013 Paris, France
| | - L Ouaguia
- CNRS, UMR8161, Institut de Biologie de Lille, Université de Lille, Institut Pasteur de Lille, IFR 142, 59021 Lille cedex, France
| | - N Delhem
- CNRS, UMR8161, Institut de Biologie de Lille, Université de Lille, Institut Pasteur de Lille, IFR 142, 59021 Lille cedex, France
| | - F Conti
- Sorbonne Universités, UPMC Université Paris 06, INSERM, UMR_S 938, Saint-Antoine Research Center, 75012 Paris, France; AP-HP, Pitié-Salpêtrière hospital, Unité Médicale de Transplantation Hépatique, 75013 Paris, France
| |
Collapse
|
42
|
Chatterjee B, Deng Y, Holler A, Nunez N, Azzi T, Vanoaica LD, Müller A, Zdimerova H, Antsiferova O, Zbinden A, Capaul R, Dreyer JH, Nadal D, Becher B, Robinson MD, Stauss H, Münz C. CD8+ T cells retain protective functions despite sustained inhibitory receptor expression during Epstein-Barr virus infection in vivo. PLoS Pathog 2019; 15:e1007748. [PMID: 31145756 PMCID: PMC6542544 DOI: 10.1371/journal.ppat.1007748] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2018] [Accepted: 04/04/2019] [Indexed: 12/15/2022] Open
Abstract
Epstein Barr virus (EBV) is one of the most ubiquitous human pathogens in the world, persistently infecting more than 90% of the adult human population. It drives some of the strongest human CD8+ T cell responses, which can be observed during symptomatic primary infection known as infectious mononucleosis (IM). Despite high viral loads and prolonged CD8+ T cell stimulation during IM, EBV enters latency and is under lifelong immune control in most individuals that experience this disease. We investigated whether changes in T cell function, as frequently characterized by PD-1 up-regulation, occur during IM due to the prolonged exposure to high antigen levels. We readily detected the expansion of PD-1 positive CD8+ T cells together with high frequencies of Tim-3, 2B4, and KLRG1 expression during IM and in mice with reconstituted human immune system components (huNSG mice) that had been infected with a high dose of EBV. These PD-1 positive CD8+ T cells, however, retained proliferation, cytokine production, and cytotoxic abilities. Multiple subsets of CD8+ T cells expanded during EBV infection, including PD-1+Tim-3+KLRG1+ cells that express CXCR5 and TCF-1 germinal center homing and memory markers, and may also contain BATF3. Moreover, blocking the PD-1 axis compromised EBV specific immune control and resulted in virus-associated lymphomagenesis. Finally, PD-1+, Tim-3+, and KLRG1+ CD8+ T cell expansion coincided with declining viral loads during low dose EBV infection. These findings suggest that EBV infection primes PD-1 positive CD8+ T cell populations that rely on this receptor axis for the efficient immune control of this ubiquitous human tumor virus. Since its discovery as a tumor virus by Epstein and colleagues in 1964, Epstein-Barr virus (EBV) has been implicated in many serious diseases, including infectious mononucleosis, Burkitt’s lymphoma, and post-transplant lymphoproliferative disease. Currently, in vivo studies are lacking to understand the comprehensive immune control of EBV in most healthy virus carriers, and, in particular, the characteristics of the CD8+ T cells involved in this process. We find that even though CD8+ T cells express multiple inhibitory receptors including PD-1 during primary EBV infection, they appear to retain an ability to produce cytokines, to kill infected cells, and to proliferate. Importantly, blocking the PD-1 pathway leads to defects in EBV-specific control and increased virus-induced tumor formation, indicating that this axis is important for viral control. This is in contrast to previous studies where releasing an inhibitory block is important for reinvigorating immune responses against cancer. Because PD-1 function is required to keep EBV in check, this study provides evidence against blocking co-inhibitory pathways in disease settings that require improved immune control of chronic virus infections.
Collapse
Affiliation(s)
- Bithi Chatterjee
- Viral Immunobiology, Institute of Experimental Immunology, University of Zurich, Switzerland
| | - Yun Deng
- Viral Immunobiology, Institute of Experimental Immunology, University of Zurich, Switzerland
| | - Angelika Holler
- Institute of Immunity and Transplantation, Royal Free Campus, University College London, United Kingdom
| | - Nicolas Nunez
- Inflammation Research, Institute of Experimental Immunology, University of Zurich, Switzerland
| | - Tarik Azzi
- Division of Infectious Diseases and Hospital Epidemiology, Children’s Research Center, University Children’s Hospital Zurich, Switzerland
| | | | - Anne Müller
- Viral Immunobiology, Institute of Experimental Immunology, University of Zurich, Switzerland
| | - Hana Zdimerova
- Viral Immunobiology, Institute of Experimental Immunology, University of Zurich, Switzerland
| | - Olga Antsiferova
- Viral Immunobiology, Institute of Experimental Immunology, University of Zurich, Switzerland
| | - Andrea Zbinden
- Institute of Medical Virology, University of Zurich, Switzerland
| | - Riccarda Capaul
- Institute of Medical Virology, University of Zurich, Switzerland
| | | | - David Nadal
- Inflammation Research, Institute of Experimental Immunology, University of Zurich, Switzerland
| | - Burkhard Becher
- Inflammation Research, Institute of Experimental Immunology, University of Zurich, Switzerland
| | - Mark D. Robinson
- Institute of Molecular Life Sciences, University of Zurich, Zurich, Switzerland
- SIB Swiss Institute of Bioinformatics, Zurich, Switzerland
| | - Hans Stauss
- Institute of Immunity and Transplantation, Royal Free Campus, University College London, United Kingdom
| | - Christian Münz
- Viral Immunobiology, Institute of Experimental Immunology, University of Zurich, Switzerland
- * E-mail:
| |
Collapse
|
43
|
Masavuli MG, Wijesundara DK, Underwood A, Christiansen D, Earnest-Silveira L, Bull R, Torresi J, Gowans EJ, Grubor-Bauk B. A Hepatitis C Virus DNA Vaccine Encoding a Secreted, Oligomerized Form of Envelope Proteins Is Highly Immunogenic and Elicits Neutralizing Antibodies in Vaccinated Mice. Front Immunol 2019; 10:1145. [PMID: 31178869 PMCID: PMC6543710 DOI: 10.3389/fimmu.2019.01145] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Accepted: 05/07/2019] [Indexed: 12/24/2022] Open
Abstract
Hepatitis C virus (HCV) persistently infects approximately 71 million people globally. To prevent infection a vaccine which elicits neutralizing antibodies against the virus envelope proteins (E1/E2) which are required for entry into host cells is desirable. DNA vaccines are cost-effective to manufacture globally and despite recent landmark studies highlighting the therapeutic efficacy of DNA vaccines in humans against cervical cancer, DNA vaccines encoding E1/E2 developed thus far are poorly immunogenic. We now report a novel and highly immunogenic DNA vaccination strategy that incorporates secreted E1 and E2 (sE1 and sE2) into oligomers by fusion with the oligomerization domain of the C4b-binding protein, IMX313P. The FDA approved plasmid, pVax, was used to encode sE1, sE2, or sE1E2 with or without IMX313P, and intradermal prime-boost vaccination studies in BALB/c mice showed that vaccines encoding IMX313P were the most effective in eliciting humoral and cell-mediated immunity against the envelope proteins. Further boosting with recombinant E1E2 proteins but not DNA nor virus-like particles (VLPs) expressing E1E2 increased the immunogenicity of the DNA prime-boost regimen. Nevertheless, the antibodies generated by the homologous DNA prime-boost vaccinations more effectively inhibited the binding of VLPs to target cells and neutralized transduction with HCV pseudoparticles (HCVpp) derived from different genotypes including genotypes 1, 2, 3, 4, 5, and 6. This report provides the first evidence that IMX313P can be used as an adjuvant for E1/E2-based DNA vaccines and represents a translatable approach for the development of a HCV DNA vaccine.
Collapse
Affiliation(s)
- Makutiro Ghislain Masavuli
- Virology Laboratory, Basil Hetzel Institute for Translational Medicine, Discipline of Surgery, University of Adelaide, Adelaide, SA, Australia
| | - Danushka K Wijesundara
- Virology Laboratory, Basil Hetzel Institute for Translational Medicine, Discipline of Surgery, University of Adelaide, Adelaide, SA, Australia
| | - Alexander Underwood
- Faculty of Medicine, The Kirby Institute, School of Medical Sciences, University of New South Wales, Sydney, NSW, Australia
| | - Dale Christiansen
- Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, University of Melbourne, Parkville, VIC, Australia
| | - Linda Earnest-Silveira
- Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, University of Melbourne, Parkville, VIC, Australia
| | - Rowena Bull
- Faculty of Medicine, The Kirby Institute, School of Medical Sciences, University of New South Wales, Sydney, NSW, Australia
| | - Joseph Torresi
- Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, University of Melbourne, Parkville, VIC, Australia
| | - Eric J Gowans
- Virology Laboratory, Basil Hetzel Institute for Translational Medicine, Discipline of Surgery, University of Adelaide, Adelaide, SA, Australia
| | - Branka Grubor-Bauk
- Virology Laboratory, Basil Hetzel Institute for Translational Medicine, Discipline of Surgery, University of Adelaide, Adelaide, SA, Australia
| |
Collapse
|
44
|
Chigbu DI, Loonawat R, Sehgal M, Patel D, Jain P. Hepatitis C Virus Infection: Host⁻Virus Interaction and Mechanisms of Viral Persistence. Cells 2019; 8:cells8040376. [PMID: 31027278 PMCID: PMC6523734 DOI: 10.3390/cells8040376] [Citation(s) in RCA: 91] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2018] [Revised: 03/25/2019] [Accepted: 04/17/2019] [Indexed: 12/11/2022] Open
Abstract
Hepatitis C (HCV) is a major cause of liver disease, in which a third of individuals with chronic HCV infections may develop liver cirrhosis. In a chronic HCV infection, host immune factors along with the actions of HCV proteins that promote viral persistence and dysregulation of the immune system have an impact on immunopathogenesis of HCV-induced hepatitis. The genome of HCV encodes a single polyprotein, which is translated and processed into structural and nonstructural proteins. These HCV proteins are the target of the innate and adaptive immune system of the host. Retinoic acid-inducible gene-I (RIG-I)-like receptors and Toll-like receptors are the main pattern recognition receptors that recognize HCV pathogen-associated molecular patterns. This interaction results in a downstream cascade that generates antiviral cytokines including interferons. The cytolysis of HCV-infected hepatocytes is mediated by perforin and granzyme B secreted by cytotoxic T lymphocyte (CTL) and natural killer (NK) cells, whereas noncytolytic HCV clearance is mediated by interferon gamma (IFN-γ) secreted by CTL and NK cells. A host-HCV interaction determines whether the acute phase of an HCV infection will undergo complete resolution or progress to the development of viral persistence with a consequential progression to chronic HCV infection. Furthermore, these host-HCV interactions could pose a challenge to developing an HCV vaccine. This review will focus on the role of the innate and adaptive immunity in HCV infection, the failure of the immune response to clear an HCV infection, and the factors that promote viral persistence.
Collapse
Affiliation(s)
- DeGaulle I Chigbu
- Department of Microbiology and Immunology, and the Institute for Molecular Medicine and Infectious Disease, Drexel University College of Medicine, 2900 West Queen Lane, Philadelphia, PA 19129, USA.
- Pennsylvania College of Optometry at Salus University, Elkins Park, PA 19027, USA.
| | - Ronak Loonawat
- Department of Microbiology and Immunology, and the Institute for Molecular Medicine and Infectious Disease, Drexel University College of Medicine, 2900 West Queen Lane, Philadelphia, PA 19129, USA.
| | - Mohit Sehgal
- Immunology, Microenvironment & Metastasis Program, The Wistar Institute, Philadelphia, PA 19104, USA.
| | - Dip Patel
- Department of Microbiology and Immunology, and the Institute for Molecular Medicine and Infectious Disease, Drexel University College of Medicine, 2900 West Queen Lane, Philadelphia, PA 19129, USA.
| | - Pooja Jain
- Department of Microbiology and Immunology, and the Institute for Molecular Medicine and Infectious Disease, Drexel University College of Medicine, 2900 West Queen Lane, Philadelphia, PA 19129, USA.
| |
Collapse
|
45
|
Casey JL, Feld JJ, MacParland SA. Restoration of HCV-Specific Immune Responses with Antiviral Therapy: A Case for DAA Treatment in Acute HCV Infection. Cells 2019; 8:cells8040317. [PMID: 30959825 PMCID: PMC6523849 DOI: 10.3390/cells8040317] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2019] [Revised: 03/26/2019] [Accepted: 03/30/2019] [Indexed: 12/11/2022] Open
Abstract
Worldwide, 71 million individuals are chronically infected with Hepatitis C Virus (HCV). Chronic HCV infection can lead to potentially fatal outcomes including liver cirrhosis and hepatocellular carcinoma. HCV-specific immune responses play a major role in viral control and may explain why approximately 20% of infections are spontaneously cleared before the establishment of chronicity. Chronic infection, associated with prolonged antigen exposure, leads to immune exhaustion of HCV-specific T cells. These exhausted T cells are unable to control the viral infection. Before the introduction of direct acting antivirals (DAAs), interferon (IFN)-based therapies demonstrated successful clearance of viral infection in approximately 50% of treated patients. New effective and well-tolerated DAAs lead to a sustained virological response (SVR) in more than 95% of patients regardless of viral genotype. Researchers have investigated whether treatment, and the subsequent elimination of HCV antigen, can reverse this HCV-induced exhausted phenotype. Here we review literature exploring the restoration of HCV-specific immune responses following antiviral therapy, both IFN and DAA-based regimens. IFN treatment during acute HCV infection results in greater immune restoration than IFN treatment of chronically infected patients. Immune restoration data following DAA treatment in chronically HCV infected patients shows varied results but suggests that DAA treatment may lead to partial restoration that could be improved with earlier administration. Future research should investigate immune restoration following DAA therapies administered during acute HCV infection.
Collapse
Affiliation(s)
- Julia L Casey
- Institute of Medical Science, University of Toronto, Toronto, ON M5S 1A8, Canada.
| | - Jordan J Feld
- Institute of Medical Science, University of Toronto, Toronto, ON M5S 1A8, Canada.
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON M5G 2C4, Canada.
| | - Sonya A MacParland
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON M5G 2C4, Canada.
- Departments of Laboratory Medicine & Pathobiology and Immunology, University of Toronto, Toronto, ON M5S 1A1, Canada.
| |
Collapse
|
46
|
Hartlage AS, Murthy S, Kumar A, Trivedi S, Dravid P, Sharma H, Walker CM, Kapoor A. Vaccination to prevent T cell subversion can protect against persistent hepacivirus infection. Nat Commun 2019; 10:1113. [PMID: 30846697 PMCID: PMC6405742 DOI: 10.1038/s41467-019-09105-0] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2018] [Accepted: 02/20/2019] [Indexed: 12/23/2022] Open
Abstract
Efforts to develop an effective vaccine against the hepatitis C virus (HCV; human hepacivirus) have been stymied by a lack of small animal models. Here, we describe an experimental rat model of chronic HCV-related hepacivirus infection and its response to T cell immunization. Immune-competent rats challenged with a rodent hepacivirus (RHV) develop chronic viremia characterized by expansion of non-functional CD8+ T cells. Single-dose vaccination with a recombinant adenovirus vector expressing hepacivirus non-structural proteins induces effective immunity in majority of rats. Resolution of infection coincides with a vigorous recall of intrahepatic cellular responses. Host selection of viral CD8 escape variants can subvert vaccine-conferred immunity. Transient depletion of CD8+ cells from vaccinated rats prolongs infection, while CD4+ cell depletion results in chronic viremia. These results provide direct evidence that co-operation between CD4+ and CD8+ T cells is important for hepacivirus immunity, and that subversion of responses can be prevented by prophylactic vaccination. Development of a HCV vaccine is hampered by a lack of appropriate small animal models. Here, Hartlage et al. describe a rat model of hepacivirus persistence and show that persistence can be prevented by vaccination with viral non-structural proteins.
Collapse
Affiliation(s)
- Alex S Hartlage
- Center for Vaccines and Immunity, The Research Institute at Nationwide Children's Hospital, Columbus, OH, 43205, USA.,Medical Scientist Training Program, College of Medicine and Public Health, The Ohio State University, Columbus, OH, 43210, USA
| | - Satyapramod Murthy
- Center for Vaccines and Immunity, The Research Institute at Nationwide Children's Hospital, Columbus, OH, 43205, USA
| | - Arvind Kumar
- Center for Vaccines and Immunity, The Research Institute at Nationwide Children's Hospital, Columbus, OH, 43205, USA
| | - Sheetal Trivedi
- Center for Vaccines and Immunity, The Research Institute at Nationwide Children's Hospital, Columbus, OH, 43205, USA
| | - Piyush Dravid
- Center for Vaccines and Immunity, The Research Institute at Nationwide Children's Hospital, Columbus, OH, 43205, USA
| | - Himanshu Sharma
- Center for Vaccines and Immunity, The Research Institute at Nationwide Children's Hospital, Columbus, OH, 43205, USA
| | - Christopher M Walker
- Center for Vaccines and Immunity, The Research Institute at Nationwide Children's Hospital, Columbus, OH, 43205, USA.,Department of Pediatrics, College of Medicine and Public Health, The Ohio State University, Columbus, OH, 43210, USA
| | - Amit Kapoor
- Center for Vaccines and Immunity, The Research Institute at Nationwide Children's Hospital, Columbus, OH, 43205, USA. .,Department of Pediatrics, College of Medicine and Public Health, The Ohio State University, Columbus, OH, 43210, USA.
| |
Collapse
|
47
|
Ping Y, Song M, Wang M, Li Z, Zhang Y. CDR3 repertoire diversity of CD8+ T lymphocytes in patients with HCV. Cell Immunol 2019; 336:34-39. [PMID: 30591202 DOI: 10.1016/j.cellimm.2018.12.007] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2018] [Revised: 12/10/2018] [Accepted: 12/19/2018] [Indexed: 12/25/2022]
Abstract
T cell receptors (TCR) diversity is known to serve as a defining hallmark of the antigen-reactive T cell repertoire. Complementarity determining region 3 (CDR3) was the most important region for the recognition of peptide-major histocompatibility complex (MHC) complexes and represented the diversity of TCR repertoire. In this study, we detected the CDR3 spectratypes by complexity scoring system to assess TCR repertoire diversity and further analyzed the correlation of CDR3 score with CD8+ T cell function and with the prognosis of chronic hepatitis C virus (HCV)-infected patients. The results demonstrated that CDR3 score was related to CD8+ T cell function and prognosis by analyzing the clinical indicators such as viral load (VL), rapid virologic response (RVR), early virologic response (EVR) and sustained virologic response (SVR). Importantly, we found that Vβ27, a member of CDR3 subfamily, might play an important role in the clearance of HCV. These findings indicate that TCR diversity maybe serve as a biomarker to predict the clinical parameters of HCV-infected patients.
Collapse
Affiliation(s)
- Yu Ping
- Biotherapy Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan, China; Engineering Key Laboratory for Cell Therapy of Henan Province, Zhengzhou, Henan 450052, China
| | - Mengjia Song
- Biotherapy Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan, China; Engineering Key Laboratory for Cell Therapy of Henan Province, Zhengzhou, Henan 450052, China
| | - Meng Wang
- Department of Gastroenterology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan, China
| | - Zhiqin Li
- Department of Infection, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan, China
| | - Yi Zhang
- Biotherapy Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan, China; Engineering Key Laboratory for Cell Therapy of Henan Province, Zhengzhou, Henan 450052, China; Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan, China.
| |
Collapse
|
48
|
Abstract
Exhausted CD8 T (Tex) cells are a distinct cell lineage that arise during chronic infections and cancers in animal models and humans. Tex cells are characterized by progressive loss of effector functions, high and sustained inhibitory receptor expression, metabolic dysregulation, poor memory recall and homeostatic self-renewal, and distinct transcriptional and epigenetic programs. The ability to reinvigorate Tex cells through inhibitory receptor blockade, such as αPD-1, highlights the therapeutic potential of targeting this population. Emerging insights into the mechanisms of exhaustion are informing immunotherapies for cancer and chronic infections. However, like other immune cells, Tex cells are heterogeneous and include progenitor and terminal subsets with unique characteristics and responses to checkpoint blockade. Here, we review our current understanding of Tex cell biology, including the developmental paths, transcriptional and epigenetic features, and cell intrinsic and extrinsic factors contributing to exhaustion and how this knowledge may inform therapeutic targeting of Tex cells in chronic infections, autoimmunity, and cancer.
Collapse
Affiliation(s)
- Laura M McLane
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA; .,Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA.,Parker Institute for Cancer Immunotherapy, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| | - Mohamed S Abdel-Hakeem
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA; .,Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA.,Parker Institute for Cancer Immunotherapy, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA.,Department of Microbiology and Immunology, Faculty of Pharmacy, Cairo University, Kasr El-Aini, Cairo 11562, Egypt
| | - E John Wherry
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA; .,Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA.,Parker Institute for Cancer Immunotherapy, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| |
Collapse
|
49
|
Egelston CA, Avalos C, Tu TY, Simons DL, Jimenez G, Jung JY, Melstrom L, Margolin K, Yim JH, Kruper L, Mortimer J, Lee PP. Human breast tumor-infiltrating CD8 + T cells retain polyfunctionality despite PD-1 expression. Nat Commun 2018; 9:4297. [PMID: 30327458 PMCID: PMC6191461 DOI: 10.1038/s41467-018-06653-9] [Citation(s) in RCA: 90] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2018] [Accepted: 09/18/2018] [Indexed: 12/26/2022] Open
Abstract
Functional CD8+ T cells in human tumors play a clear role in clinical prognosis and response to immunotherapeutic interventions. PD-1 expression in T cells involved in chronic infections and tumors such as melanoma often correlates with a state of T-cell exhaustion. Here we interrogate CD8+ tumor-infiltrating lymphocytes (TILs) from human breast and melanoma tumors to explore their functional state. Despite expression of exhaustion hallmarks, such as PD-1 expression, human breast tumor CD8+ TILs retain robust capacity for production of effector cytokines and degranulation capacity. In contrast, melanoma CD8+ TILs display dramatic reduction of cytokine production and degranulation capacity. We show that CD8+ TILs from human breast tumors can potently kill cancer cells via bi-specific antibodies. Our data demonstrate that CD8+ TILs in human breast tumors retain polyfunctionality, despite PD-1 expression, and suggest that they may be harnessed for effective immunotherapies. Expression of the checkpoint molecule programmed cell death protein 1 (PD-1) is considered a marker of T cells exhaustion. Here the authors show that CD8T cells isolated from breast cancer patients are perfectly functional despite PD-1 expression while those isolated from melanoma patients are not.
Collapse
Affiliation(s)
- Colt A Egelston
- Department of Immuno-Oncology, Beckman Research Institute of City of Hope, Duarte, CA, 91010, USA
| | - Christian Avalos
- Department of Immuno-Oncology, Beckman Research Institute of City of Hope, Duarte, CA, 91010, USA
| | - Travis Y Tu
- Department of Immuno-Oncology, Beckman Research Institute of City of Hope, Duarte, CA, 91010, USA
| | - Diana L Simons
- Department of Immuno-Oncology, Beckman Research Institute of City of Hope, Duarte, CA, 91010, USA
| | - Grecia Jimenez
- Department of Immuno-Oncology, Beckman Research Institute of City of Hope, Duarte, CA, 91010, USA
| | - Jae Y Jung
- Department of Dermatologic Oncology, Norton Cancer Institute, Louisville, KY, 40202, USA
| | - Laleh Melstrom
- Department of Surgery, Beckman Research Institute of City of Hope, Duarte, CA, 91010, USA
| | - Kim Margolin
- Department of Medical Oncology, Beckman Research Institute of City of Hope, Duarte, CA, 91010, USA
| | - John H Yim
- Department of Surgery, Beckman Research Institute of City of Hope, Duarte, CA, 91010, USA
| | - Laura Kruper
- Department of Surgery, Beckman Research Institute of City of Hope, Duarte, CA, 91010, USA
| | - Joanne Mortimer
- Department of Medical Oncology, Beckman Research Institute of City of Hope, Duarte, CA, 91010, USA
| | - Peter P Lee
- Department of Immuno-Oncology, Beckman Research Institute of City of Hope, Duarte, CA, 91010, USA.
| |
Collapse
|
50
|
Shrivastava S, Bhatta M, Ward H, Romani S, Lee R, Rosenthal E, Osinusi A, Kohli A, Masur H, Kottilil S, Wilson E. Multitarget Direct-Acting Antiviral Therapy Is Associated With Superior Immunologic Recovery in Patients Coinfected With Human Immunodeficiency Virus and Hepatitis C Virus. Hepatol Commun 2018; 2:1451-1466. [PMID: 30556035 PMCID: PMC6287478 DOI: 10.1002/hep4.1258] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/23/2018] [Accepted: 08/24/2018] [Indexed: 12/13/2022] Open
Abstract
Patients coinfected with human immunodeficiency virus (HIV) and hepatitis C virus (HCV) have higher levels of immune activation, impaired antigen‐specific responses, and accelerated fibrogenesis compared to patients monoinfected with HCV. Whether different direct‐acting antiviral (DAA) combinations have differential effects on immunophenotypes and functions following successful HCV therapy remain unknown. Therefore, we aimed to assess the peripheral T‐cell immunophenotypes and functions in patients coinfected with HIV/HCV who were successfully treated with combination DAA treatment regimens. We analyzed peripheral blood mononuclear cells (PBMCs) at baseline and at the time of sustained viral response (SVR) from subjects treated with three different combination DAA regimens: daclatasvir (DCV) and asunaprevir (ASV) for 24 weeks (CONQUER 2‐DAA), DCV/ASV/beclabuvir (BCV) for 12 weeks (CONQUER 3‐DAA), and sofosbuvir (SOF) and ledipasvir (LDV) for 12 weeks (ERADICATE study). We used flow cytometry to assess T‐cell phenotypes (activation and exhaustion) and HCV‐specific T‐cell functions (cytokine secretion and cytotoxicity). Statistical analyses were conducted using the Wilcoxon matched‐pairs signed‐rank test with P < 0.05 considered significant. Overall, there was an improvement in T‐cell exhaustion markers, a decrease in T‐cell activation, an increase in the effector memory population, and improved T‐cell function after achieving SVR, with the largest effects noted with CONQUER 3‐DAA treatment. Conclusion: Treatment with DCV/ASV/BCV in patients coinfected with HIV/HCV resulted in greater restoration of the T‐cell impairments and perturbations associated with HIV/HCV coinfection to an extent that was greater than that observed in either two‐drug regimens. We showed that different DAA‐based therapies have different immunologic outcomes after successful HCV treatment in patients coinfected with HIV/HCV. This information will be beneficial for providers when selecting the regimens for patients coinfected with HIV/HCV.
Collapse
Affiliation(s)
- Shikha Shrivastava
- Institute of Human Virology University of Maryland School of Medicine Baltimore MD
| | - Manasa Bhatta
- Institute of Human Virology University of Maryland School of Medicine Baltimore MD.,Critical Care Medicine Department, Clinical Center National Institutes of Health Bethesda MD
| | - Haley Ward
- Institute of Human Virology University of Maryland School of Medicine Baltimore MD.,Critical Care Medicine Department, Clinical Center National Institutes of Health Bethesda MD
| | - Sara Romani
- Institute of Human Virology University of Maryland School of Medicine Baltimore MD
| | - Rebecca Lee
- Institute of Human Virology University of Maryland School of Medicine Baltimore MD
| | - Elana Rosenthal
- Institute of Human Virology University of Maryland School of Medicine Baltimore MD
| | | | - Anita Kohli
- Critical Care Medicine Department, Clinical Center National Institutes of Health Bethesda MD
| | - Henry Masur
- Critical Care Medicine Department, Clinical Center National Institutes of Health Bethesda MD
| | - Shyam Kottilil
- Institute of Human Virology University of Maryland School of Medicine Baltimore MD
| | - Eleanor Wilson
- Institute of Human Virology University of Maryland School of Medicine Baltimore MD
| |
Collapse
|