1
|
Garbuglia AR, Pauciullo S, Zulian V, Del Porto P. Update on Hepatitis C Vaccine: Results and Challenges. Viruses 2024; 16:1337. [PMID: 39205311 PMCID: PMC11359353 DOI: 10.3390/v16081337] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Revised: 08/15/2024] [Accepted: 08/20/2024] [Indexed: 09/04/2024] Open
Abstract
Therapy against the Hepatitis C virus (HCV) has significantly improved with the introduction of direct-acting antiviral drugs (DAAs), achieving over 95% sustained virological response (SVR). Despite this, the development of an effective anti-HCV vaccine remains a critical challenge due to the low number of patients treated with DAAs and the occurrence of HCV reinfections in high-risk groups. Current vaccine strategies aim to stimulate either B-cell or T-cell responses. Vaccines based on E1 and E2 proteins can elicit broad cross-neutralizing antibodies against all major HCV genotypes, though with varying efficiencies and without full protection against infection. In humans, the neutralizing antibodies induced by such vaccines mainly target the AR3 region, but their levels are generally insufficient for broad neutralization. Various HCV proteins expressed through different viral vectors have been utilized to elicit T cell immune responses, showing sustained expansion of HCV-specific effector memory T cells and improved proliferation and polyfunctionality of memory T cells over time. However, despite these advancements, the frequency and effectiveness of T-cell responses remain limited.
Collapse
Affiliation(s)
- Anna Rosa Garbuglia
- Laboratory of Virology, National Institute for Infectious Diseases “Lazzaro Spallanzani” (IRCCS), 00149 Rome, Italy; (S.P.); (V.Z.)
| | - Silvia Pauciullo
- Laboratory of Virology, National Institute for Infectious Diseases “Lazzaro Spallanzani” (IRCCS), 00149 Rome, Italy; (S.P.); (V.Z.)
| | - Verdiana Zulian
- Laboratory of Virology, National Institute for Infectious Diseases “Lazzaro Spallanzani” (IRCCS), 00149 Rome, Italy; (S.P.); (V.Z.)
| | - Paola Del Porto
- Department of Biology and Biotechnology “Charles Darwin”, Sapienza University of Rome, 00100 Rome, Italy;
| |
Collapse
|
2
|
Cornberg M, Mischke J, Kraft AR, Wedemeyer H. Immunological scars after cure of hepatitis C virus infection: Long-HepC? Curr Opin Immunol 2023; 82:102324. [PMID: 37043890 DOI: 10.1016/j.coi.2023.102324] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Revised: 02/21/2023] [Accepted: 03/15/2023] [Indexed: 04/14/2023]
Abstract
Hepatitis C virus (HCV) infection provides a unique opportunity to study the effects of spontaneous or treatment-induced viral elimination on the human immune system. Twenty to 50% of patients with acute HCV infection spontaneously clear the virus, which is related to the quality of the individual's immune response, while the chronic infection is associated with an altered and impaired immune response. Direct-acting antiviral agents are now available that provide sustained viral elimination in more than 95% of patients with chronic HCV infection. Viral elimination leads to a decrease in disease sequelae such as cirrhosis and hepatocellular carcinoma, and extrahepatic manifestations also improve. However, some patients may still experience long-term complications, and viral elimination does not protect against HCV reinfection. This review addresses the question of whether the altered and impaired immune response caused by HCV normalizes after viral elimination and if this may affect the long-term clinical course after HCV cure.
Collapse
Affiliation(s)
- Markus Cornberg
- Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School (MHH), Hannover, Germany; Centre for Individualised Infection Medicine (CiiM), a joint venture between the Helmholtz Centre for Infection Research (HZI) and Hannover Medical School (MHH), Hannover, Germany; TWINCORE, a joint venture between the Helmholtz-Centre for Infection Research (HZI) and the Hannover Medical School (MHH), Hannover, Germany; German Centre for Infection Research (DZIF), partner site Hannover-Braunschweig, Germany; Cluster of Excellence Resolving Infection Susceptibility (RESIST; EXC 2155), Hannover Medical School, Hannover, Germany.
| | - Jasmin Mischke
- Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School (MHH), Hannover, Germany; Centre for Individualised Infection Medicine (CiiM), a joint venture between the Helmholtz Centre for Infection Research (HZI) and Hannover Medical School (MHH), Hannover, Germany; TWINCORE, a joint venture between the Helmholtz-Centre for Infection Research (HZI) and the Hannover Medical School (MHH), Hannover, Germany; German Centre for Infection Research (DZIF), partner site Hannover-Braunschweig, Germany; Cluster of Excellence Resolving Infection Susceptibility (RESIST; EXC 2155), Hannover Medical School, Hannover, Germany
| | - Anke Rm Kraft
- Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School (MHH), Hannover, Germany; Centre for Individualised Infection Medicine (CiiM), a joint venture between the Helmholtz Centre for Infection Research (HZI) and Hannover Medical School (MHH), Hannover, Germany; TWINCORE, a joint venture between the Helmholtz-Centre for Infection Research (HZI) and the Hannover Medical School (MHH), Hannover, Germany; German Centre for Infection Research (DZIF), partner site Hannover-Braunschweig, Germany; Cluster of Excellence Resolving Infection Susceptibility (RESIST; EXC 2155), Hannover Medical School, Hannover, Germany
| | - Heiner Wedemeyer
- Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School (MHH), Hannover, Germany; German Centre for Infection Research (DZIF), partner site Hannover-Braunschweig, Germany; Cluster of Excellence Resolving Infection Susceptibility (RESIST; EXC 2155), Hannover Medical School, Hannover, Germany
| |
Collapse
|
3
|
Akhbariyoon H, Azizpour Y, Esfahani MF, Firoozabad MSM, Rad MR, Esfahani KS, Khoshavi N, Karimi N, Shirinisaz A, Abedi F, Rad MR, Sharifi P. Immune checkpoint inhibition for the treatment of cancers: An update and critical review of ongoing clinical trials. Clin Immunol 2021; 232:108873. [PMID: 34688855 DOI: 10.1016/j.clim.2021.108873] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Revised: 09/29/2021] [Accepted: 10/14/2021] [Indexed: 12/12/2022]
Abstract
Advances in Cancer immunotherapy in the past few years include the development of medications that modulate immune checkpoint proteins. Cytotoxic T-lymphocyte antigen 4 (CTLA4), programmed cell death protein 1 (PD1), and programmed cell death ligand 1 (PD-L1) are three co-inhibitory receptors that are expressed in the tumor microenvironment. Immune checkpoint inhibitors (ICI) that target these biomarkers unleash the properties of effector T cells that are licensed to kill cancer cells. Immune checkpoint blockade has dramatically changed the treatment landscape of many cancers. In this Review, we describe the current data regarding clinical trials of ICIs in six important cancers, including hepatocellular carcinoma (HCC), renal cell cancer (RCC), hodgkin lymphoma (HL), non-hodgkin lymphoma (NHL), non-small cell lung cancer (NSCLC), and head and neck cancer carcinoma (HNSCC).
Collapse
Affiliation(s)
| | - Yasaman Azizpour
- Department of Biochemistry, Tarbiat Modares University, 14115-175 Tehran, Iran
| | | | | | - Mehrdad Rabiee Rad
- School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | | | - Neda Khoshavi
- Department of Biology, Faculty of Science, Yazd University, Yazd, Iran
| | - Negin Karimi
- Department of Biology, Faculty of Science, Yazd University, Yazd, Iran
| | - Asal Shirinisaz
- Department of Biology, Faculty of Science, Yazd University, Yazd, Iran
| | - Fatemeh Abedi
- Department of Biology, Faculty of Science, Yazd University, Yazd, Iran
| | - Maryam Rabiee Rad
- Department of Biology, Faculty of Science, Yazd University, Yazd, Iran
| | - Parisa Sharifi
- Department of Biology, Faculty of Science, Yazd University, Yazd, Iran
| |
Collapse
|
4
|
Espíndola ODM, Siteur-van Rijnstra E, Frankin E, Weijer K, van der Velden YU, Berkhout B, Blom B, Villaudy J. Early Effects of HTLV-1 Infection on the Activation, Exhaustion, and Differentiation of T-Cells in Humanized NSG Mice. Cells 2021; 10:2514. [PMID: 34685494 PMCID: PMC8534134 DOI: 10.3390/cells10102514] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2021] [Revised: 09/11/2021] [Accepted: 09/15/2021] [Indexed: 12/14/2022] Open
Abstract
Adult T-cell leukemia/lymphoma (ATLL) is an aggressive malignancy of CD4+ T-cells associated with HTLV-1 infection. In this study, we used the model of immunodeficient NSG mice reconstituted with a functional human immune system (HIS) to investigate early events in HTLV-1 pathogenesis. Upon infection, human T-cells rapidly increased in the blood and lymphoid tissues, particularly CD4+CD25+ T-cells. Proliferation of CD4+ T-cells in the spleen and mesenteric lymph nodes (MLN) correlated with HTLV-1 proviral load and CD25 expression. In addition, splenomegaly, a common feature of ATLL in humans, was also observed. CD4+ and CD8+ T-cells predominantly displayed an effector memory phenotype (CD45RA-CCR7-) and expressed CXCR3 and CCR5 chemokine receptors, suggesting the polarization into a Th1 phenotype. Activated CD8+ T-cells expressed granzyme B and perforin; however, the interferon-γ response by these cells was limited, possibly due to elevated PD-1 expression and increased frequency of CD4+FoxP3+ regulatory T-cells in MLN. Thus, HTLV-1-infected HIS-NSG mice reproduced several characteristics of infection in humans, and it may be helpful to investigate ATLL-related events and to perform preclinical studies. Moreover, aspects of chronic infection were already present at early stages in this experimental model. Collectively, we suggest that HTLV-1 infection modulates host immune responses to favor viral persistence.
Collapse
Affiliation(s)
- Otávio de Melo Espíndola
- Laboratory for Clinical Research in Neuroinfections, Evandro Chagas National Institute of Infectious Diseases, Oswaldo Cruz Foundation (FIOCRUZ), Rio de Janeiro 21040-900, Brazil
- Department of Experimental Immunology, Amsterdam University Medical Centers, Location AMC, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands; (E.S.-v.R.); (E.F.); (K.W.); (B.B.)
| | - Esther Siteur-van Rijnstra
- Department of Experimental Immunology, Amsterdam University Medical Centers, Location AMC, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands; (E.S.-v.R.); (E.F.); (K.W.); (B.B.)
| | - Esmay Frankin
- Department of Experimental Immunology, Amsterdam University Medical Centers, Location AMC, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands; (E.S.-v.R.); (E.F.); (K.W.); (B.B.)
| | - Kees Weijer
- Department of Experimental Immunology, Amsterdam University Medical Centers, Location AMC, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands; (E.S.-v.R.); (E.F.); (K.W.); (B.B.)
| | - Yme Ubeles van der Velden
- Laboratory of Experimental Virology, Department of Medical Microbiology, Amsterdam University Medical Centers, Location AMC, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands; (Y.U.v.d.V.); (B.B.); (J.V.)
| | - Ben Berkhout
- Laboratory of Experimental Virology, Department of Medical Microbiology, Amsterdam University Medical Centers, Location AMC, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands; (Y.U.v.d.V.); (B.B.); (J.V.)
| | - Bianca Blom
- Department of Experimental Immunology, Amsterdam University Medical Centers, Location AMC, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands; (E.S.-v.R.); (E.F.); (K.W.); (B.B.)
| | - Julien Villaudy
- Laboratory of Experimental Virology, Department of Medical Microbiology, Amsterdam University Medical Centers, Location AMC, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands; (Y.U.v.d.V.); (B.B.); (J.V.)
- J&S Preclinical Solutions, 5345 RR Oss, The Netherlands
| |
Collapse
|
5
|
Woller N, Engelskircher SA, Wirth T, Wedemeyer H. Prospects and Challenges for T Cell-Based Therapies of HCC. Cells 2021; 10:cells10071651. [PMID: 34209393 PMCID: PMC8304292 DOI: 10.3390/cells10071651] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Revised: 06/25/2021] [Accepted: 06/29/2021] [Indexed: 12/11/2022] Open
Abstract
The scope of therapeutic options for the treatment of hepatocellular carcinoma (HCC) has recently been expanded by immunotherapeutic regimens. T cell-based therapies, especially in combination with other treatments have achieved far better outcomes compared to conventional treatments alone. However, there is an emerging body of evidence that eliciting T cell responses in immunotherapeutic approaches is insufficient for favorable outcomes. Immune responses in HCC are frequently attenuated in the tumor microenvironment (TME) or may even support tumor progress. Hence, therapies with immune checkpoint inhibitors or adoptive cell therapies appear to necessitate additional modification of the TME to unlock their full potential. In this review, we focus on immunotherapeutic strategies, underlying molecular mechanisms of CD8 T cell immunity, and causes of treatment failure in HCC of viral and non-viral origin. Furthermore, we provide an overview of TME features in underlying etiologies of HCC patients that mediate therapy resistance to checkpoint inhibition and discuss strategies from the literature concerning current approaches to these challenges.
Collapse
Affiliation(s)
- Norman Woller
- Clinic for Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, 30625 Hannover, Germany
| | - Sophie Anna Engelskircher
- Clinic for Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, 30625 Hannover, Germany
| | - Thomas Wirth
- Clinic for Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, 30625 Hannover, Germany
| | - Heiner Wedemeyer
- Clinic for Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, 30625 Hannover, Germany
| |
Collapse
|
6
|
Qian H, Song S, Tian Y, Qi L, Gao H, Yuan L. Study on the gene signature related to immune microenvironment on viral and nonviral infections of hepatocellular carcinoma. Medicine (Baltimore) 2021; 100:e25374. [PMID: 33847635 PMCID: PMC8052083 DOI: 10.1097/md.0000000000025374] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2020] [Accepted: 03/10/2021] [Indexed: 01/04/2023] Open
Abstract
The pathogenesis of hepatocellular carcinoma (HCC) can be divided into viral infection (VIR) and nonviral (NVIR) infection. Two types of HCC performed different tumor immune microenvironment (TIME) which directly affected prognosis of HCC. This study aimed to identify an effective 2 types of HCC prognostic gene signature that related to immune TIME.The differential expression genes (DEGs) were analyzed by Limma R package from the Cancer Genome Atlas. Immune related genes getting from IMMport database were matched to DEGs for testing prognosis. Prognostic index (PI) consisted of prognostic immune related genes was calculated in different types of HCC by COX regression and the correlation with the abundance of immune infiltrates, including 6 type cells, via gene modules. Tumor immune estimation resource database was applied to analyze TIME. Finally, the correlations between PI of DEGs and TIICs were analyzed by the Spearman method.Results showed that PI consisted of 11 messenger RNAs in VIR and 12 messenger RNAs in NVIR groups. The PI related to HCC prognosis has different correlations with immune infiltrating cells in VIR and NVIR groups. The PI value of DEGs has significant correlations with neutrophils (R = 0.22, P-value = .029) and dendritic (R = 0.21, P-value = .036) infiltration levels in VIR group. However, in NVIR group, the result showed there were no significant correlations between PI and other 5 type cell infiltration levels (P-value > .05).The 11-gene signature in VIR and 12-gene signature in NVIR group selected based on data from the Cancer Genome Atlas database had a different correlation with immune infiltrating cells of HCC patients.
Collapse
Affiliation(s)
- Hong Qian
- The First Hospital of Lanzhou University, Lanzhou
| | | | - Yunling Tian
- The First Hospital of Lanzhou University, Lanzhou
| | - Lei Qi
- Xifeng District People's Hospital of Qingyang City
| | - Haihong Gao
- Qingyang Huanxian People's Hospital, Qingyang
| | - Lingyan Yuan
- The First Hospital of Lanzhou University, Lanzhou
- The First Clinical Medical College of Lanzhou University
- Evidence Based Medicine Center, School of Basic Medical Science, Lanzhou University, Lanzhou, China
| |
Collapse
|
7
|
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
|
8
|
Huang N, Zhou R, Chen H, Zhang S, Li J, Wei W, Sun J, Ren S, Li B, Deng H, Yang J, Ji F, Li Z. Splenic CD4 + and CD8 + T-cells highly expressed PD-1 and Tim-3 in cirrhotic patients with HCV infection and portal hypertension. Int J Immunopathol Pharmacol 2021; 35:20587384211061051. [PMID: 34930041 PMCID: PMC8725229 DOI: 10.1177/20587384211061051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Accepted: 11/01/2021] [Indexed: 11/15/2022] Open
Abstract
Introduction: The spleen plays an important role in regulating the immune response to infectious pathogens. T-cells dysfunction and exhaustion have been reported in patients with hepatitis B/C virus (HBV/HCV) infection, which contributes to persistent virus infection. The aims of this study were to investigate spleen-related evidence of immunosuppression and immune tolerance in HCV cirrhotic patients with portal hypertension (PH). Methods: The expression of programmed cell death 1 (PD-1), T-cell immunoglobulin domain and mucin domain-containing molecule-3 (Tim-3) and its ligand PD-L1/2, and Galectin-9 in the spleens and livers of HCV cirrhotic patients (n = 15) was analyzed using real-time PCR and immunohistochemistry. Flow cytometry was used to evaluate the expression of PD-1 and Tim-3 on splenic T-cells and the peripheral blood T-cells before and after splenectomy (n = 8). Results: Spleens from patients with PH showed significantly increased mRNA levels of PD-L2, Tim-3, Galectin-9, CD80, and CD86, and decreased levels of CD28 compared to control spleens (spleens removed due to traumatic injury) (all p < 0.05). Additionally, protein expression of inhibitory signaling molecules was significantly increased in both the spleens and livers of cirrhotic patients compared with controls (all p < 0.05). Peripheral blood and splenic CD4+ and CD8+ T-cells also expressed higher protein levels of PD-1, Tim-3, and CTLA-4 in cirrhotic patients as compared with healthy controls (all p < 0.05). The proportion of PD-1+CD4+T lymphocytes (26.2% ± 7.12% vs. 21.0% ± 9.14%, p = 0.0293) and Tim-3+CD8+ T lymphocytes (9.4% ± 3.04% vs. 6.0% ± 2.24%, p = 0.0175) in peripheral blood decreased followed splenectomy. Conclusion: The CD4+ and CD8+ T-cells in spleen and peripheral blood highly expressed PD-1 and Tim-3 in HCV-infected and cirrhotic patients with portal hypertension. Highly expressed PD-1 and Tim-3 in peripheral blood T-lymphocytes can be partly reversed following splenectomy.
Collapse
Affiliation(s)
- Na Huang
- National & Local Joint Engineering
Research Center of Biodiagnosis and Biotherapy, the Second Affiliated Hospital of Xi’an Jiaotong
University, Xi’an, China
- Shaanxi Provincial Clinical Research Center
for Hepatic & Splenic Diseases, the Second Affiliated Hospital of Xi’an Jiaotong
University, Xi’an, China
| | - Rui Zhou
- National & Local Joint Engineering
Research Center of Biodiagnosis and Biotherapy, the Second Affiliated Hospital of Xi’an Jiaotong
University, Xi’an, China
- Shaanxi Provincial Clinical Research Center
for Hepatic & Splenic Diseases, the Second Affiliated Hospital of Xi’an Jiaotong
University, Xi’an, China
| | - Haiyan Chen
- National & Local Joint Engineering
Research Center of Biodiagnosis and Biotherapy, the Second Affiliated Hospital of Xi’an Jiaotong
University, Xi’an, China
- Shaanxi Provincial Clinical Research Center
for Hepatic & Splenic Diseases, the Second Affiliated Hospital of Xi’an Jiaotong
University, Xi’an, China
| | - Shu Zhang
- National & Local Joint Engineering
Research Center of Biodiagnosis and Biotherapy, the Second Affiliated Hospital of Xi’an Jiaotong
University, Xi’an, China
- Shaanxi Provincial Clinical Research Center
for Hepatic & Splenic Diseases, the Second Affiliated Hospital of Xi’an Jiaotong
University, Xi’an, China
| | - Jun Li
- National & Local Joint Engineering
Research Center of Biodiagnosis and Biotherapy, the Second Affiliated Hospital of Xi’an Jiaotong
University, Xi’an, China
- Shaanxi Provincial Clinical Research Center
for Hepatic & Splenic Diseases, the Second Affiliated Hospital of Xi’an Jiaotong
University, Xi’an, China
| | - Wei Wei
- National & Local Joint Engineering
Research Center of Biodiagnosis and Biotherapy, the Second Affiliated Hospital of Xi’an Jiaotong
University, Xi’an, China
- Department of Oncology Surgery, the Second Affiliated Hospital of Xi’an Jiaotong
University, Xi’an, China
| | - Jin Sun
- National & Local Joint Engineering
Research Center of Biodiagnosis and Biotherapy, the Second Affiliated Hospital of Xi’an Jiaotong
University, Xi’an, China
| | - Song Ren
- National & Local Joint Engineering
Research Center of Biodiagnosis and Biotherapy, the Second Affiliated Hospital of Xi’an Jiaotong
University, Xi’an, China
- Shaanxi Provincial Clinical Research Center
for Hepatic & Splenic Diseases, the Second Affiliated Hospital of Xi’an Jiaotong
University, Xi’an, China
| | - Baohua Li
- National & Local Joint Engineering
Research Center of Biodiagnosis and Biotherapy, the Second Affiliated Hospital of Xi’an Jiaotong
University, Xi’an, China
- Shaanxi Provincial Clinical Research Center
for Hepatic & Splenic Diseases, the Second Affiliated Hospital of Xi’an Jiaotong
University, Xi’an, China
| | - Hong Deng
- Department of Infectious Diseases, the Second Affiliated Hospital of Xi’an Jiaotong
University, Xi’an, China
| | - Jun Yang
- National & Local Joint Engineering
Research Center of Biodiagnosis and Biotherapy, the Second Affiliated Hospital of Xi’an Jiaotong
University, Xi’an, China
- Department of Pathology, the Second Affiliated Hospital of Xi’an Jiaotong
University, Xi’an, China
| | - Fanpu Ji
- National & Local Joint Engineering
Research Center of Biodiagnosis and Biotherapy, the Second Affiliated Hospital of Xi’an Jiaotong
University, Xi’an, China
- Shaanxi Provincial Clinical Research Center
for Hepatic & Splenic Diseases, the Second Affiliated Hospital of Xi’an Jiaotong
University, Xi’an, China
- Department of Infectious Diseases, the Second Affiliated Hospital of Xi’an Jiaotong
University, Xi’an, China
- Key Laboratory of Environment and Genes
Related to Diseases, Xi’an Jiaotong University, Ministry of Education of China, Xi’an, China
| | - Zongfang Li
- National & Local Joint Engineering
Research Center of Biodiagnosis and Biotherapy, the Second Affiliated Hospital of Xi’an Jiaotong
University, Xi’an, China
- Shaanxi Provincial Clinical Research Center
for Hepatic & Splenic Diseases, the Second Affiliated Hospital of Xi’an Jiaotong
University, Xi’an, China
- Key Laboratory of Environment and Genes
Related to Diseases, Xi’an Jiaotong University, Ministry of Education of China, Xi’an, China
| |
Collapse
|
9
|
Smith S, Honegger JR, Walker C. T-Cell Immunity against the Hepatitis C Virus: A Persistent Research Priority in an Era of Highly Effective Therapy. Cold Spring Harb Perspect Med 2021; 11:cshperspect.a036954. [PMID: 32205413 PMCID: PMC7778213 DOI: 10.1101/cshperspect.a036954] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Approximately 70% of acute hepatitis C virus (HCV) infections become chronic, indicating that the virus is exceptionally well adapted to persist in humans with otherwise normal immune function. Robust, lifelong replication of this small RNA virus does not require a generalized failure of immunity. HCV effectively subverts innate and adaptive host defenses while leaving immunity against other viruses intact. Here, the role of CD4+ and CD8+ T-cell responses in control of HCV infection and their failure to prevent virus persistence in most individuals are reviewed. Two issues of practical importance remain priorities in an era of highly effective antiviral therapy for chronic hepatitis C. First, the characteristics of successful T-cell responses that promote resolution of HCV infection are considered, as they will underpin development of vaccines that prevent HCV persistence. Second, defects in T-cell immunity that facilitate HCV persistence and whether they are reversed after antiviral cure to provide protection from reinfection are also addressed.
Collapse
Affiliation(s)
- Stephanie Smith
- The Center for Vaccines and Immunity, The Abigail Wexner Research Institute at Nationwide Children's, Columbus, Ohio 43205, USA,Department of Pediatrics, College of Medicine, The Ohio State University, Columbus, Ohio 43004, USA
| | - Jonathan R. Honegger
- The Center for Vaccines and Immunity, The Abigail Wexner Research Institute at Nationwide Children's, Columbus, Ohio 43205, USA,Department of Pediatrics, College of Medicine, The Ohio State University, Columbus, Ohio 43004, USA
| | - Christopher Walker
- The Center for Vaccines and Immunity, The Abigail Wexner Research Institute at Nationwide Children's, Columbus, Ohio 43205, USA,Department of Pediatrics, College of Medicine, The Ohio State University, Columbus, Ohio 43004, USA
| |
Collapse
|
10
|
Orr C, Xu W, Masur H, Kottilil S, Meissner EG. Peripheral blood correlates of virologic relapse after Sofosbuvir and Ribavirin treatment of Genotype-1 HCV infection. BMC Infect Dis 2020; 20:929. [PMID: 33276734 PMCID: PMC7718661 DOI: 10.1186/s12879-020-05657-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Accepted: 11/24/2020] [Indexed: 12/11/2022] Open
Abstract
Background Treatment of chronic hepatitis C virus infection with direct acting antiviral therapy results in viral elimination in over 90% of cases. The duration of treatment required to achieve cure differs between individuals and relapse can occur. We asked whether cellular and transcriptional profiling of peripheral blood collected during treatment could identify biomarkers predictive of treatment outcome. Methods We analyzed peripheral blood collected during treatment of genotype 1 HCV with 24 weeks of sofosbuvir and weight-based or low dose ribavirin in a trial in which 29% of patients relapsed. Changes in host immunity during treatment were assessed by flow cytometry and whole blood gene expression profiling. Differences in expression of immune-relevant transcripts based on treatment outcome were analyzed using the Nanostring Human Immunology V2 panel. Results Multiple cellular populations changed during treatment, but pre-treatment neutrophil counts were lower and natural post-treatment killer cell counts were higher in patients who relapsed. Pre-treatment expression of genes associated with interferon-signaling, T-cell dysfunction, and T-cell co-stimulation differed by treatment outcome. We identified a pre- and post-treatment gene expression signature with high predictive capacity for distinguishing treatment outcome, but neither signature was sufficiently robust to suggest viability for clinical use. Conclusions Patients who relapse after hepatitis C virus therapy differ immunologically from non-relapsers based on expression of transcripts related to interferon signaling and T-cell dysfunction, as well as by peripheral neutrophil and NK-cell concentrations. These data provide insight into the host immunologic basis of relapse after DAA therapy for HCV and suggests mechanisms which may be relevant for understanding outcomes with currently approved regimens.
Collapse
Affiliation(s)
- Cody Orr
- Division of Infectious Diseases, Medical University of South Carolina, 135 Rutledge Ave, MSC752, Charleston, SC, 29425, USA
| | - Wenjie Xu
- Nanostring Technologies, Seattle, WA, USA
| | - Henry Masur
- Critical Care Medicine Department, NIH Clinical Center, National Institutes of Health, Bethesda, MD, USA
| | - Shyam Kottilil
- Division of Clinical Care and Research, Institute of Human Virology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Eric G Meissner
- Division of Infectious Diseases, Medical University of South Carolina, 135 Rutledge Ave, MSC752, Charleston, SC, 29425, USA. .,Department of Microbiology and Immunology, Medical University of South Carolina, 135 Rutledge Ave, MSC752, Charleston, SC, 29425, USA.
| |
Collapse
|
11
|
Bonilla CM, McGrath NA, Fu J, Xie C. Immunotherapy of hepatocellular carcinoma with infection of hepatitis B or C virus. HEPATOMA RESEARCH 2020; 6:68. [PMID: 33134550 PMCID: PMC7597818 DOI: 10.20517/2394-5079.2020.58] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Hepatocellular carcinoma (HCC) has one of highest mortalities globally amongst cancers, but has limited therapeutic options once in the advanced stage. Hepatitis B or C virus infection are the most common drivers for HCC carcinogenesis, triggering chronic liver inflammation and adding to the complexity of the immune microecosystem of HCC. The emergence of immunotherapy has afforded a new avenue of therapeutic options for patients with advanced HCC with a history of hepatitis B or C virus infection. This article reviews the change of immunity elicited by hepatitis B or C virus infection, the immune feature of HCC, and the clinical evidence for immunotherapy in advanced HCC and discusses future directions in this field.
Collapse
Affiliation(s)
- Cecilia Monge Bonilla
- Thoracic and Gastrointestinal Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Nicole A McGrath
- Thoracic and Gastrointestinal Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Jianyang Fu
- Thoracic and Gastrointestinal Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Changqing Xie
- Thoracic and Gastrointestinal Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
12
|
Lymphocyte Landscape after Chronic Hepatitis C Virus (HCV) Cure: The New Normal. Int J Mol Sci 2020; 21:ijms21207473. [PMID: 33050486 PMCID: PMC7589490 DOI: 10.3390/ijms21207473] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Revised: 10/05/2020] [Accepted: 10/07/2020] [Indexed: 12/17/2022] Open
Abstract
Chronic HCV (CHC) infection is the only chronic viral infection for which curative treatments have been discovered. These direct acting antiviral (DAA) agents target specific steps in the viral replication cycle with remarkable efficacy and result in sustained virologic response (SVR) or cure in high (>95%) proportions of patients. These treatments became available 6–7 years ago and it is estimated that their real impact on HCV related morbidity, including outcomes such as cirrhosis and hepatocellular carcinoma (HCC), will not be known for the next decade or so. The immune system of a chronically infected patient is severely dysregulated and questions remain regarding the immune system’s capacity in limiting liver pathology in a cured individual. Another important consequence of impaired immunity in patients cleared of HCV with DAA will be the inability to generate protective immunity against possible re-infection, necessitating retreatments or developing a prophylactic vaccine. Thus, the impact of viral clearance on restoring immune homeostasis is being investigated by many groups. Among the important questions that need to be answered are how much the immune system normalizes with cure, how long after viral clearance this recalibration occurs, what are the consequences of persisting immune defects for protection from re-infection in vulnerable populations, and does viral clearance reduce liver pathology and the risk of developing hepatocellular carcinoma in individuals cured with these agents. Here, we review the recent literature that describes the defects present in various lymphocyte populations in a CHC patient and their status after viral clearance using DAA treatments.
Collapse
|
13
|
PD-L1 Checkpoint Inhibition Narrows the Antigen-Specific T Cell Receptor Repertoire in Chronic Lymphocytic Choriomeningitis Virus Infection. J Virol 2020; 94:JVI.00795-20. [PMID: 32641478 DOI: 10.1128/jvi.00795-20] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Accepted: 06/19/2020] [Indexed: 11/20/2022] Open
Abstract
Checkpoint inhibitors are effective in restoring exhausted CD8+ T cell responses in persistent viral infections or tumors. Several compounds are in clinical use for different malignancies, but trials in patients with chronic viral infections have also been conducted. In a mouse model of persistent lymphocytic choriomeningitis virus (LCMV) infection, it was shown that checkpoint inhibitor treatment increased T cell proliferation and functionality, but its influence on the antigen-specific T cell receptor (TCR) repertoire is unknown. NP396-specific CD8+ T cells dominate during acute LCMV infection and are predominantly exhausted during chronic infection. Next-generation sequencing of NP396-specific TCRs showed that exhaustion corresponds with a significantly reduced NP396-specific TCR repertoire diversity: Shannon indices of 4 in immunized mice to 2.6 in persistently infected mice. Anti-PD-L1 treatment during persistent LCMV infection restored NP396-specific T cell responses and reduced viral titers. Nevertheless, anti-PD-L1-treated mice showed an even more narrowed TCR repertoire, with reduced TCR diversity compared to that of persistently infected control mice (Shannon indices of 2.1 and 2.6, respectively). Interestingly, anti-PD-L1 treatment-induced narrowing of the TCR repertoire negatively correlates with functional and physical restoration of the antigen-specific T cell response. Further, we found that private, hyperexpanded TCR clonotypes dominated the T cell response after anti-PD-L1 treatment. Although being private, these top clonotypes from anti-PD-L1-treated mice revealed a more closely related CDR3 motif than those of top clonotypes from persistently infected control mice. In conclusion, although targeting the PD-1/PD-L1 pathway reinvigorates exhausted CD8+ T cells, it fails to restore T cell repertoire diversity.IMPORTANCE Checkpoint inhibitors are effective immunotherapeutics to restore cancer- and virus-induced exhausted CD8+ T cells, by enhancing the quality and survival of immune responses. Although checkpoint inhibitors are already used as therapy against various cancers, not much is known about their multifaceted impact on the exhausted CD8+ T cell receptor (TCR) repertoire. This report describes for the first time the evolvement of an exhausted antigen-specific CD8+ TCR repertoire under checkpoint inhibitor treatment. By using a well-established virus model, we were able to show major shifts toward oligoclonality of the CD8+ TCR repertoire response against a massively exhausted lymphocytic choriomeningitis virus (LCMV) epitope. While supporting viral control in the LCMV model, oligoclonality and more private of TCR repertoires may impact future pathogenic challenges and may promote viral escape. Our results may explain the ongoing problems of viral escapes, unpredictable autoimmunity, and heterogeneous responses appearing as adverse effects of checkpoint inhibitor treatments.
Collapse
|
14
|
Dash S, Aydin Y, Widmer KE, Nayak L. Hepatocellular Carcinoma Mechanisms Associated with Chronic HCV Infection and the Impact of Direct-Acting Antiviral Treatment. J Hepatocell Carcinoma 2020; 7:45-76. [PMID: 32346535 PMCID: PMC7167284 DOI: 10.2147/jhc.s221187] [Citation(s) in RCA: 74] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Accepted: 03/06/2020] [Indexed: 12/12/2022] Open
Abstract
Hepatitis C virus (HCV) infection is the major risk factor for liver cirrhosis and hepatocellular carcinoma (HCC). The mechanisms of HCC initiation, growth, and metastasis appear to be highly complex due to the decade-long interactions between the virus, immune system, and overlapping bystander effects of host metabolic liver disease. The lack of a readily accessible animal model system for HCV is a significant obstacle to understand the mechanisms of viral carcinogenesis. Traditionally, the primary prevention strategy of HCC has been to eliminate infection by antiviral therapy. The success of virus elimination by antiviral treatment is determined by the SVR when the HCV is no longer detectable in serum. Interferon-alpha (IFN-α) and its analogs, pegylated IFN-α (PEG-IFN-α) alone with ribavirin (RBV), have been the primary antiviral treatment of HCV for many years with a low cure rate. The cloning and sequencing of HCV have allowed the development of cell culture models, which accelerated antiviral drug discovery. It resulted in the selection of highly effective direct-acting antiviral (DAA)-based combination therapy that now offers incredible success in curing HCV infection in more than 95% of all patients, including those with cirrhosis. However, several emerging recent publications claim that patients who have liver cirrhosis at the time of DAAs treatment face the risk of HCC occurrence and recurrence after viral cure. This remains a substantial challenge while addressing the long-term benefit of antiviral medicine. The host-related mechanisms that drive the risk of HCC in the absence of the virus are unknown. This review describes the multifaceted mechanisms that create a tumorigenic environment during chronic HCV infection. In addition to the potential oncogenic programming that drives HCC after viral clearance by DAAs, the current status of a biomarker development for early prediction of cirrhosis regression and HCC detection post viral treatment is discussed. Since DAAs treatment does not provide full protection against reinfection or viral transmission to other individuals, the recent studies for a vaccine development are also reviewed.
Collapse
Affiliation(s)
- Srikanta Dash
- Department of Pathology and Laboratory Medicine, Tulane University Health Sciences Center, New Orleans, LA70112, USA
- Southeast Louisiana Veterans Health Care System, New Orleans, LA70119, USA
- Department of Medicine, Division of Gastroenterology, Tulane University Health Sciences Center, New Orleans, LA70112, USA
| | - Yucel Aydin
- Department of Pathology and Laboratory Medicine, Tulane University Health Sciences Center, New Orleans, LA70112, USA
| | - Kyle E Widmer
- Southeast Louisiana Veterans Health Care System, New Orleans, LA70119, USA
| | - Leela Nayak
- Southeast Louisiana Veterans Health Care System, New Orleans, LA70119, USA
| |
Collapse
|
15
|
Hakim MS, Rahmadika N, Jariah ROA. Expressions of inhibitory checkpoint molecules in acute and chronic HBV and HCV infections: Implications for therapeutic monitoring and personalized therapy. Rev Med Virol 2019; 30:e2094. [DOI: 10.1002/rmv.2094] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2019] [Revised: 11/06/2019] [Accepted: 11/12/2019] [Indexed: 12/14/2022]
Affiliation(s)
- Mohamad S. Hakim
- Department of Microbiology, Faculty of Medicine, Public Health and NursingUniversitas Gadjah Mada Yogyakarta Indonesia
| | - Nofri Rahmadika
- Infectious Disease Research Center, Faculty of MedicineUniversitas Padjadjaran Bandung Indonesia
| | - Rizka O. A. Jariah
- Department of Health Science, Faculty of Vocational StudiesUniversitas Airlangga Surabaya Indonesia
| |
Collapse
|
16
|
Elimination of hepatitis C virus has limited impact on the functional and mitochondrial impairment of HCV-specific CD8+ T cell responses. J Hepatol 2019; 71:889-899. [PMID: 31295532 DOI: 10.1016/j.jhep.2019.06.025] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2019] [Revised: 06/12/2019] [Accepted: 06/19/2019] [Indexed: 12/15/2022]
Abstract
BACKGROUND & AIMS Hepatitis C virus (HCV)-specific CD8+ T cells are functionally impaired in chronic hepatitis C. Even though HCV can now be rapidly and sustainably cleared from chronically infected patients, the repercussions of HCV clearance on virus-specific CD8+ T cells remain elusive. Here, we aimed to investigate if HCV clearance by direct-acting antivirals (DAAs) could restore the functionality of exhausted HCV-specific CD8+ T cell responses. METHODS HCV-specific CD8+ T cells in peripheral blood were obtained from 40 patients with chronic HCV infection, during and 6 months following IFN-free DAA therapy. These cells were analyzed for comprehensive phenotypes, proliferation, cytokine production, mitochondrial fitness and response to immune-checkpoint blockade. RESULTS We show that, unlike activation markers that decreased, surface expression of multiple co-regulatory receptors on exhausted HCV-specific CD8+ T cells remained unaltered after clearance of HCV. Likewise, cytokine production by HCV-specific CD8+ T cells remained impaired following HCV clearance. The proliferative capacity of HCV multimer-specific CD8+ T cells was not restored in the majority of patients. Enhanced in vitro proliferative expansion of HCV-specific CD8+ T cells during HCV clearance was more likely in women, patients with low liver stiffness and low alanine aminotransferase levels in our cohort. Interestingly, HCV-specific CD8+ T cells that did not proliferate following HCV clearance could preferentially re-invigorate their proliferative capacity upon in vitro immune-checkpoint inhibition. Moreover, altered mitochondrial dysfunction exhibited by exhausted HCV-specific CD8+ T cells could not be normalized after HCV clearance. CONCLUSION Taken together, our data implies that exhausted HCV-specific CD8+ T cells remain functionally and metabolically impaired at multiple levels following HCV clearance in most patients with chronic hepatitis C. Our results might have implications in cases of re-infection with HCV and for HCV vaccine development. LAY SUMMARY Direct-acting antiviral therapy results in cure of hepatitis C virus (HCV) in almost all treated patients. However, the impacts of HCV cure on immune responses remain controversial. Whether immune responses to HCV recover is important in cases of re-exposure, or for the resolution of extrahepatic manifestations. The main finding of our study was that HCV-specific T cells remain functionally impaired despite HCV clearance. This finding could explain the fact that HCV cure does not lead to protective immunity and that re-infections have frequently been observed.
Collapse
|
17
|
Is PD-1 blockade a potential therapy for HBV? JHEP Rep 2019; 1:142-144. [PMID: 32040093 PMCID: PMC7001582 DOI: 10.1016/j.jhepr.2019.07.007] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Revised: 07/24/2019] [Accepted: 07/26/2019] [Indexed: 01/05/2023] Open
|
18
|
van den Berg SPH, Pardieck IN, Lanfermeijer J, Sauce D, Klenerman P, van Baarle D, Arens R. The hallmarks of CMV-specific CD8 T-cell differentiation. Med Microbiol Immunol 2019; 208:365-373. [PMID: 30989333 PMCID: PMC6647465 DOI: 10.1007/s00430-019-00608-7] [Citation(s) in RCA: 80] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2019] [Accepted: 04/02/2019] [Indexed: 12/13/2022]
Abstract
Upon cytomegalovirus (CMV) infection, large T-cell responses are elicited that remain high or even increase over time, a phenomenon named memory T-cell inflation. Besides, the maintained robust T-cell response, CMV-specific T cells seem to have a distinctive phenotype, characterized by an advanced differentiation state. Here, we will review this "special" differentiation status by discussing the cellular phenotype based on the expression of CD45 isoforms, costimulatory, inhibitory and natural killer receptors, adhesion and lymphocyte homing molecules, transcription factors, cytokines and cytotoxic molecules. In addition, we focus on whether the differentiation state of CMV-specific CD8 T cells is unique in comparison with other chronic viruses and we will discuss the possible impact of factors such as antigen exposure and aging on the advanced differentiation status of CMV-specific CD8 T cells.
Collapse
Affiliation(s)
- Sara P H van den Berg
- Center for Infectious Disease Control, National Institute for Public Health and the Environment, Bilthoven, The Netherlands
- Laboratory of Translational Immunology, Department of Immunology, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Iris N Pardieck
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, Albinusdreef 2, 2333 ZA, Leiden, The Netherlands
| | - Josien Lanfermeijer
- Center for Infectious Disease Control, National Institute for Public Health and the Environment, Bilthoven, The Netherlands
- Laboratory of Translational Immunology, Department of Immunology, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Delphine Sauce
- Sorbonne Université, INSERM, Centre d'Immunologie et des Maladies Infectieuses (CIMI-Paris), Paris, France
| | - Paul Klenerman
- Nuffield Department of Medicine, Peter Medawar Building for Pathogen Research, University of Oxford, Oxford, UK
- NIHR Biomedical Research Centre, John Radcliffe Hospital, Oxford, UK
| | - Debbie van Baarle
- Center for Infectious Disease Control, National Institute for Public Health and the Environment, Bilthoven, The Netherlands
- Laboratory of Translational Immunology, Department of Immunology, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Ramon Arens
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, Albinusdreef 2, 2333 ZA, Leiden, The Netherlands.
| |
Collapse
|
19
|
Ackermann C, Smits M, Woost R, Eberhard JM, Peine S, Kummer S, Marget M, Kuntzen T, Kwok WW, Lohse AW, Jacobs T, Boettler T, Schulze Zur Wiesch J. HCV-specific CD4+ T cells of patients with acute and chronic HCV infection display high expression of TIGIT and other co-inhibitory molecules. Sci Rep 2019; 9:10624. [PMID: 31337800 PMCID: PMC6650447 DOI: 10.1038/s41598-019-47024-8] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2019] [Accepted: 07/03/2019] [Indexed: 01/08/2023] Open
Abstract
The combined regulation of a network of inhibitory and activating T cell receptors may be a critical step in the development of chronic HCV infection. Ex vivo HCV MHC class I + II tetramer staining and bead-enrichment was performed with baseline and longitudinal PBMC samples of a cohort of patients with acute, chronic and spontaneously resolved HCV infection to assess the expression pattern of the co-inhibitory molecule TIGIT together with PD-1, BTLA, Tim-3, as well as OX40 and CD226 (DNAM-1) of HCV-specific CD4+ T cells, and in a subset of patients of HCV-specific CD8+ T cells. As the main result, we found a higher expression level of TIGIT+ PD-1+ on HCV-specific CD4+ T cells during acute and chronic HCV infection compared to patients with spontaneously resolved HCV infection (p < 0,0001). Conversely, expression of the complementary co-stimulatory receptor of TIGIT, CD226 (DNAM-1) was significantly decreased on HCV-specific CD4+ T cells during chronic infection. The predominant phenotype of HCV-specific CD4+ T cells during acute and chronic infection was TIGIT+, PD-1+, BTLA+, Tim-3−. This comprehensive phenotypic study confirms TIGIT together with PD-1 as a discriminatory marker of dysfunctional HCV-specific CD4+ T cells.
Collapse
Affiliation(s)
- Christin Ackermann
- I. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Maike Smits
- Department of Medicine II, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany.,Faculty of Biology, University of Freiburg, Freiburg, Germany
| | - Robin Woost
- I. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,DZIF partner site (German Center for Infection Research), Hamburg, Germany
| | - Johanna M Eberhard
- I. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,DZIF partner site (German Center for Infection Research), Hamburg, Germany
| | - Sven Peine
- Department of Transfusion Medicine, Germany, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Silke Kummer
- I. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,DZIF partner site (German Center for Infection Research), Hamburg, Germany
| | - Matthias Marget
- Department of Transfusion Medicine, Germany, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Thomas Kuntzen
- Gastroenterologie und Hepatologie; Kantonsspital Aarau, Aarau, Switzerland
| | - William W Kwok
- Benaroya Research Institute at Virginia Mason, Seattle, Washington, United States of America
| | - Ansgar W Lohse
- I. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,DZIF partner site (German Center for Infection Research), Hamburg, Germany
| | - Thomas Jacobs
- Protozoa Immunology, Bernhard Nocht, Institute for Tropical Medicine, Hamburg, Germany
| | - Tobias Boettler
- Department of Medicine II, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Julian Schulze Zur Wiesch
- I. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany. .,DZIF partner site (German Center for Infection Research), Hamburg, Germany.
| |
Collapse
|
20
|
Hansel C, Erschfeld S, Baues M, Lammers T, Weiskirchen R, Trautwein C, Kroy DC, Drescher HK. The Inhibitory T Cell Receptors PD1 and 2B4 Are Differentially Regulated on CD4 and CD8 T Cells in a Mouse Model of Non-alcoholic Steatohepatitis. Front Pharmacol 2019; 10:244. [PMID: 30949049 PMCID: PMC6436071 DOI: 10.3389/fphar.2019.00244] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2018] [Accepted: 02/26/2019] [Indexed: 12/26/2022] Open
Abstract
Infiltrating CD4 and CD8 T cells have been shown to worsen inflammatory liver damage in non-alcoholic steatohepatitis (NASH). Inhibitory T cell receptors such as the programmed cell death protein 1 (PD1) and the natural killer cell receptor 2B4 regulate the activity of CD4 and CD8 T cells and therefore play an important role in immune tolerance required in the liver. In this study, we investigated the expression profile of inhibitory T cell receptors on CD4 and CD8 T cells in a mouse model of NASH. Male B57BL/6J mice were fed a Western diet for 24 weeks. The expression levels of inhibitory receptors on the surface of intrahepatic and peripheral T cells were measured and correlated with markers of activation (CD107a, CD69, and CD44), metabolic disorder (serum triglycerides, serum cholesterol, γ-glutamyl transferase, hepatic triglycerides), inflammation (serum alanine aminotransferase and aspartate aminotransferase) and hepatic fibrosis (collagen 1A1, α-smooth muscle actin, hydroxyproline). Under Western diet, PD1 is exclusively upregulated on intrahepatic and peripheral CD8+ T cells, whereas the expression level on CD4 T cells is unaffected. In contrast, 2B4 is upregulated liver-specifically on both CD4 and CD8 T cells and unchanged on peripheral T cells. Upregulation of PD1 on CD8 T cells is restricted to CD8 effector memory T cells and correlates with lower levels of degranulation. Similarly, the inhibitory function of PD1 on intrahepatic CD4 T cells is shown by a lower CD69 and CD44 expression on PD1-positive CD4 T cells. In murine steatohepatitis, the upregulation of PD1 on CD8 T cells and 2B4 on CD4 and CD8 T cells potentially limits T cell-mediated liver damage. Therefore, these inhibitory T cell receptors could serve as promising targets of immune-modulatory NASH therapy.
Collapse
Affiliation(s)
- Cordula Hansel
- Department of Internal Medicine III, University Hospital Aachen, RWTH Aachen University, Aachen, Germany
| | - Stephanie Erschfeld
- Department of Internal Medicine III, University Hospital Aachen, RWTH Aachen University, Aachen, Germany
| | - Maike Baues
- Institute for Experimental Molecular Imaging, University Hospital Aachen, RWTH Aachen University, Aachen, Germany
| | - Twan Lammers
- Institute for Experimental Molecular Imaging, University Hospital Aachen, RWTH Aachen University, Aachen, Germany
| | - Ralf Weiskirchen
- Institute of Molecular Pathobiochemistry, Experimental Gene Therapy, and Clinical Chemistry (IFMPEGKC), University Hospital Aachen, RWTH Aachen University, Aachen, Germany
| | - Christian Trautwein
- Department of Internal Medicine III, University Hospital Aachen, RWTH Aachen University, Aachen, Germany
| | - Daniela C Kroy
- Department of Internal Medicine III, University Hospital Aachen, RWTH Aachen University, Aachen, Germany
| | - Hannah K Drescher
- Department of Internal Medicine III, University Hospital Aachen, RWTH Aachen University, Aachen, Germany
| |
Collapse
|
21
|
Grant MJ, DeVito N, Salama AKS. Checkpoint inhibitor use in two heart transplant patients with metastatic melanoma and review of high-risk populations. Melanoma Manag 2018; 5:MMT10. [PMID: 30459942 PMCID: PMC6240846 DOI: 10.2217/mmt-2018-0004] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2018] [Accepted: 08/03/2018] [Indexed: 12/12/2022] Open
Abstract
Due to the unique side-effect profile of immune checkpoint inhibitors (ICIs), groups of patients deemed to be at high risk of complications were excluded from trials that proved the efficacy and safety of these agents in patients with various malignancies. Among these excluded patients were those with prior solid organ transplantation, chronic viral infections and pre-existing autoimmune diseases including paraneoplastic syndromes. We present follow-up on a patient from a previously published case report with an orthotopic heart transplantation who was treated with both cytotoxic T-lymphocyte antigen 4 and PD-1 inhibition safely, without organ rejection. Additionally, we describe the case of a patient with a cardiac allograft who also did not experience organ rejection after treatment with pembrolizumab. Through smaller trials, retrospective analyses, case series and individual case reports, we are accumulating initial data on how these agents are tolerated by the aforementioned groups. Our survey of the literature has found more evidence of organ transplant rejection in patients treated with PD-1 inhibitors than those treated with inhibitors of cytotoxic T-lymphocyte antigen 4. Patients with chronic viral infections, especially hepatitis C, seem to have little to no risk of treatment-related increase in serum RNA levels. The literature contains few documented cases of devastating exacerbations of pre-existing autoimmune disease during treatment with ICIs, and flares seem to be easily controlled by immunosuppression in the vast majority of cases. Last, several cases allude to a promising role for disease-specific antibodies and other serum biomarkers in identifying patients at high risk of developing certain immune-related adverse events, detecting subclinical immune-related adverse event onset, and monitoring treatment response to immunosuppressive therapy in patients treated with ICIs. Though these excluded populations have not been well studied in randomized placebo-controlled trials, we may be able to learn and derive hypotheses from the existing observational data in the literature.
Collapse
Affiliation(s)
- Michael J Grant
- Department of Medicine, Duke University Medical Center, Durham, NC 27710, USA.,Department of Medicine, Duke University Medical Center, Durham, NC 27710, USA
| | - Nicholas DeVito
- Duke Cancer Institute, Duke University Medical Center, Durham, NC, USA.,Duke Cancer Institute, Duke University Medical Center, Durham, NC, USA
| | - April K S Salama
- Duke Cancer Institute, Duke University Medical Center, Durham, NC, USA.,Duke Cancer Institute, Duke University Medical Center, Durham, NC, USA
| |
Collapse
|
22
|
Wang W, Tong Z, Zhong J, Zhang L, Zhang H, Su Y, Gao B, Zhang C. Identification of IL-10-secreting CD8 +CD28 -PD-1 + regulatory T cells associated with chronic hepatitis C virus infection. Immunol Lett 2018; 202:16-22. [PMID: 30055200 DOI: 10.1016/j.imlet.2018.07.008] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2018] [Revised: 07/24/2018] [Accepted: 07/24/2018] [Indexed: 12/28/2022]
Abstract
CD8+CD28- regulatory T cells (Tregs) play important roles in chronic viral infections. Programmed death 1 (PD-1) is highly expressed on hepatitis C virus (HCV)-specific CTLs. However, little is known regarding the role of CD8+CD28-PD1+ T cells in hepatitis C. Herein, we found that the frequency of CD8+CD28-PD1+, but not CD8+CD28-PD1- T cells, correlated with markers of chronic hepatitis C virus (HCV) infection and the response to treatment. Our results showed that CD8+CD28-PD1+ T cells were significantly elevated in chronic HCV-infected patients and there was a distinct correlation between the frequency of CD8+CD28-PD1+ T cells and serum levels of HCV RNA. During a 48-week course of treatment with peg-IFN-a2a plus ribavirin, dynamic changes in the frequencies of CD8+CD28-PD1+ T cells were observed, associated with the virologic response. IL-10 secretion may explain the suppressive function of CD8+CD28-PD1+ T cells in chronic HCV-infected patients. Overall, our study demonstrates that PD-1 is an important marker of CD8+CD28- Tregs in chronic HCV infection. Thus, the frequency and regulatory function of CD8+CD28-PD1+ T cells play vital roles in HCV infection and the response to treatment.
Collapse
Affiliation(s)
- Weihong Wang
- Department of Infectious Diseases,Huzhou Central Hospital, Huzhou, Zhejiang 313000, China
| | - Zhaowei Tong
- Department of Infectious Diseases,Huzhou Central Hospital, Huzhou, Zhejiang 313000, China
| | - Jianfeng Zhong
- Department of Infectious Diseases,Huzhou Central Hospital, Huzhou, Zhejiang 313000, China
| | - Longqi Zhang
- Department of Infectious Diseases,Huzhou Central Hospital, Huzhou, Zhejiang 313000, China
| | - Hui Zhang
- School of Medicine, Huzhou University, Huzhou 313000, China
| | - Yanguang Su
- School of Medicine, Huzhou University, Huzhou 313000, China
| | - Bingbing Gao
- School of Medicine, Huzhou University, Huzhou 313000, China
| | - Chun Zhang
- Department of Infectious Diseases,Huzhou Central Hospital, Huzhou, Zhejiang 313000, China.
| |
Collapse
|
23
|
Harding JJ. Immune checkpoint blockade in advanced hepatocellular carcinoma: an update and critical review of ongoing clinical trials. Future Oncol 2018; 14:2293-2302. [PMID: 29663837 DOI: 10.2217/fon-2018-0008] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Systemic treatments for advanced hepatocellular carcinoma (HCC) are evolving rapidly and several multi-targeted tyrosine kinase inhibitors have demonstrated a survival advantage over best supportive care. Despite these treatment advances, the majority of HCC patients will progress on tyrosine kinase inhibitor therapy. Preclinical data indicate that interference with immune checkpoint molecules results in HCC growth suppression. Several clinical trials applying monoclonal antibodies to immune checkpoint molecules have demonstrated durable antitumor activity in advanced HCC patients. As such, pivotal clinical trials are now in progress to assess if these agents will alter the natural history of the disease and further extend the overall survival of advanced HCC patients. This manuscript will review the current status of immune checkpoint blockade in patients with advanced HCC.
Collapse
Affiliation(s)
- James J Harding
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA.,Department of Medicine, Weill Cornell Medical College, New York, NY 10028, USA
| |
Collapse
|
24
|
Al-Ayoubi J, Behrendt P, Bremer B, Suneetha PV, Gisa A, Rinker F, Manns MP, Cornberg M, Wedemeyer H, Kraft ARM. Hepatitis E virus ORF 1 induces proliferative and functional T-cell responses in patients with ongoing and resolved hepatitis E. Liver Int 2018; 38:266-277. [PMID: 28718943 DOI: 10.1111/liv.13521] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/07/2016] [Accepted: 07/08/2017] [Indexed: 12/15/2022]
Abstract
BACKGROUND AND AIMS Hepatitis E virus (HEV) is a major cause of acute viral hepatitis with >3 million symptomatic cases per year accounting for 70 000 HEV-related deaths. HEV-specific T-cell responses have been investigated against structural proteins expressed by open reading frames (ORF) 2 and 3. T-cell responses against non-structural HEV proteins encoded by ORF1 are hardly studied. The aim of this study was to determine HEV ORF1-specific T-cell responses in comparison to ORF2/3 in patients exposed to HEV. METHODS HEV-specific CD4+ and CD8+ T-cell responses against HEV genotype 3 were investigated in patients with acute and chronic hepatitis E as well as in HEV seropositive and seronegative individuals. HEV-specific T-cell responses were determined by proliferation and intracellular cytokine assay upon stimulation of PBMCs with HEV-specific overlapping peptide pools spanning the entire HEV genome. HEV-antigen was measured using an anti-HEV antigen-specific ELISA. RESULTS Broad HEV ORF1-specific T-cell responses were detected in patients with acute, resolved and chronic hepatitis E without distinct dominant regions. The magnitude and frequency in recognition of ORF1-specific T-cell responses were similar compared to responses against HEV ORF2/3. Longitudinal studies of HEV-specific T-cell responses displayed similar behaviour against structural and non-structural proteins. HEV-antigen levels were inversely correlated with HEV-specific T-cell responses. CONCLUSIONS HEV-specific T-cell responses are detectable against the entire HEV genome including the non-structural proteins. HEV-specific T-cell responses are associated with control of HEV infection. These findings have implications for the design of HEV vaccines.
Collapse
Affiliation(s)
- Jana Al-Ayoubi
- Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, Hannover, Germany
| | - Patrick Behrendt
- Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, Hannover, Germany
| | - Birgit Bremer
- Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, Hannover, Germany
| | | | - Anett Gisa
- Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, Hannover, Germany
| | - Franziska Rinker
- Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, Hannover, Germany
| | - Michael P Manns
- Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, Hannover, Germany.,German Center for Infection Research (DZIF), Hannover, Germany
| | - Markus Cornberg
- Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, Hannover, Germany.,German Center for Infection Research (DZIF), Hannover, Germany
| | - Heiner Wedemeyer
- Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, Hannover, Germany.,German Center for Infection Research (DZIF), Hannover, Germany
| | - Anke R M Kraft
- Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, Hannover, Germany.,German Center for Infection Research (DZIF), Hannover, Germany
| |
Collapse
|
25
|
According to Hepatitis C Virus (HCV) Infection Stage, Interleukin-7 Plus 4-1BB Triggering Alone or Combined with PD-1 Blockade Increases TRAF1 low HCV-Specific CD8 + Cell Reactivity. J Virol 2018; 92:JVI.01443-17. [PMID: 29093082 DOI: 10.1128/jvi.01443-17] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2017] [Accepted: 10/23/2017] [Indexed: 12/14/2022] Open
Abstract
Hepatitis C virus (HCV)-specific CD8+ T cells suffer a progressive exhaustion during persistent infection (PI) with HCV. This process could involve the positive immune checkpoint 4-1BB/4-1BBL through the loss of its signal transducer, TRAF1. To address this issue, peripheral HCV-specific CD8+ T cells (pentamer-positive [pentamer+]/CD8+ T cells) from patients with PI and resolved infection (RI) after treatment were studied. The duration of HCV infection and the liver fibrosis progression rate inversely correlated with the likelihood of detection of peripheral pentamer+/CD8+ cells. In PI, pentamer+/CD8+ cells had impaired antigen-specific reactivity that worsened when these cells were not detectable ex vivo Short/midduration PI was characterized by detectable peripheral PD-1+ CD127low TRAF1low cells. After triggering of T cell receptors (TCR), the TRAF1 level positively correlated with the levels of CD127, Mcl-1, and CD107a expression and proliferation intensity but negatively with PD-1 expression, linking TRAF1low to exhaustion. In vitro treatment with interleukin-7 (IL-7) upregulated TRAF1 expression, while treatment with transforming growth factor-β1 (TGF-β1) did the opposite, suggesting that the IL-7/TGF-β1 balance, besides TCR stimulation, could be involved in TRAF1 regulation. In fact, the serum TGF-β1 concentration was higher in patients with PI than in patients with RI, and it negatively correlated with TRAF1 expression. In line with IL-7 increasing the level of TRAF1 expression, IL-7 plus 4-1BBL treatment in vitro enhanced T cell reactivity in patients with short/midduration infection. However, in patients with long-lasting PI, anti-PD-L1, in addition to the combination of IL-7 and 4-1BBL, was necessary to reestablish T cell proliferation in individuals with slowly progressing liver fibrosis (slow fibrosers) but had no effect in rapid fibrosers. In conclusion, a peripheral hyporeactive TRAF1low HCV-specific CD8+ T cell response, restorable by IL-7 plus 4-1BBL treatment, characterizes short/midduration PI. In long-lasting disease, HCV-specific CD8+ T cells are rarely detectable ex vivo, but treatment with IL-7, 4-1BBL, and anti-PD-L1 recovers their reactivity in vitro in slow fibrosers.IMPORTANCE Hepatitis C virus (HCV) infects 71 million people worldwide. Two-thirds develop a chronic disease that can lead to cirrhosis and hepatocellular carcinoma. Direct-acting antivirals clear the infection, but there are still patients who relapse. In these cases, additional immunotherapy could play a vital role. A successful anti-HCV immune response depends on virus-specific CD8+ T cells. During chronic infection, these cells are functionally impaired, which could be due to the failure of costimulation. This study describes exhausted specific T cells, characterized by low levels of expression of the signal transducer TRAF1 of the positive costimulatory pathway 4-1BB/4-1BBL. IL-7 upregulated TRAF1 expression and improved T cell reactivity in patients with short/midduration disease, while in patients with long-lasting infection, it was also necessary to block the negative PD-1/PD-L1 checkpoint. When the results are taken together, this work supports novel ways of restoring the specific CD8+ T cell response, shedding light on the importance of TRAF1 signaling. This could be a promising target for future immunotherapy.
Collapse
|
26
|
Programmed Cell Death 1 (PD-1) and Cytotoxic T Lymphocyte-Associated Antigen 4 (CTLA-4) in Viral Hepatitis. Int J Mol Sci 2017; 18:ijms18071517. [PMID: 28703774 PMCID: PMC5536007 DOI: 10.3390/ijms18071517] [Citation(s) in RCA: 72] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2017] [Revised: 07/03/2017] [Accepted: 07/04/2017] [Indexed: 12/21/2022] Open
Abstract
Virus-specific cluster of differentiation 8 (CD8+) cytotoxic T cells (CTL) recognize viral antigens presented on major histocompatibility complex (MHC) class I chains on infected hepatocytes, with help from CD4+ T cells. However, this CTL response is frequently weak or undetectable in patients with chronic hepatitis B virus (HBV) and hepatitis C virus (HCV) infection. Programmed cell death 1 (PD-1) and cytotoxic T lymphocyte-associated antigen 4 (CTLA-4) are receptors in the CD28 family of costimulatory molecules, providing inhibitory signals to T cells. The overexpressions of PD-1 and CTLA-4 in patients with viral infection have been shown to associate with functional impairment of virus-specific T cells. In acute viral hepatitis, PD-1 and CTLA-4 are up-regulated during the symptomatic phase, and then down-regulated after recovery. These findings suggest that PD-1 and CTLA-4 have protective effects as inhibitory molecules to suppress cytotoxic T cells which induce harmful destruction of viral infected hepatocytes in self-limited viral hepatitis. In chronic viral hepatitis, the extended upregulations of PD-1 and CTLA-4 are associated with T cell exhaustion and persistent viral infection, suggesting positive correlations between expression of immune inhibitory factors and the chronicity of viral disease. In this review, we summarize recent literature relating to PD-1, CTLA-4, and other inhibitory receptors in antigen-specific T cell exhaustion in viral hepatitis, including hepatitis A, B, C, and others.
Collapse
|
27
|
Li BJ, He Y, Zhang Y, Guo YH, Zhou Y, Zhang PX, Wang W, Zhao JR, Li JG, Zuo WZ, Fan C, Jia ZS. Interferon-α-induced CD100 on naïve CD8 + T cells enhances antiviral responses to hepatitis C infection through CD72 signal transduction. J Int Med Res 2017; 45:89-100. [PMID: 28222623 PMCID: PMC5536608 DOI: 10.1177/0300060516676136] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Objectives We investigated the effects of CD100 on naïve CD8+ T cells during hepatitis C virus (HCV) infection after interferon-α (IFN-α) therapy to clarify the mechanism underlying the effect of IFN-α in enhancing the antiviral response. Methods The CD100 molecules on subsets of CD8+ T cells were analysed with flow cytometry. The effects of CD100-overexpressing naïve CD8+ T cells were determined with ELISAs and an MTT cytotoxicity assay. The role of CD100-CD72 signal transduction was analysed with a neutralization and transwell assays. Results HCV infection reduced CD100 expression on CD8+ T cells, whereas IFN-α treatment significantly increased CD100 expression on naïve CD8+ T cells. The increased CD100 interacted with the CD72 receptor and enhanced PBMC cytokine secretion (IFN-γ and tumour necrosis factor-α) and cytotoxicity. Conclusions IFN-α-induced CD100 on naïve CD8+ T cells promotes PBMC cytokine secretion and cytotoxicity through CD100-CD72 signalling during HCV infection.
Collapse
Affiliation(s)
- Bing Jie Li
- 2 First Affiliated Hospital, School of Medicine, Shihezi University, Xinjiang, China
| | - Yu He
- 1 Department of Infectious Diseases and Center of Liver Diseases, Tangdu Hospital, the Fourth Military Medical University, Xi'an, China
| | - Ying Zhang
- 1 Department of Infectious Diseases and Center of Liver Diseases, Tangdu Hospital, the Fourth Military Medical University, Xi'an, China
| | - Yong Hong Guo
- 1 Department of Infectious Diseases and Center of Liver Diseases, Tangdu Hospital, the Fourth Military Medical University, Xi'an, China
| | - Yun Zhou
- 1 Department of Infectious Diseases and Center of Liver Diseases, Tangdu Hospital, the Fourth Military Medical University, Xi'an, China
| | - Pei Xin Zhang
- 1 Department of Infectious Diseases and Center of Liver Diseases, Tangdu Hospital, the Fourth Military Medical University, Xi'an, China
| | - Wei Wang
- 1 Department of Infectious Diseases and Center of Liver Diseases, Tangdu Hospital, the Fourth Military Medical University, Xi'an, China
| | - Jie Ru Zhao
- 1 Department of Infectious Diseases and Center of Liver Diseases, Tangdu Hospital, the Fourth Military Medical University, Xi'an, China
| | - Jin Ge Li
- 1 Department of Infectious Diseases and Center of Liver Diseases, Tangdu Hospital, the Fourth Military Medical University, Xi'an, China
| | - Wei Ze Zuo
- 2 First Affiliated Hospital, School of Medicine, Shihezi University, Xinjiang, China
| | - Chao Fan
- 1 Department of Infectious Diseases and Center of Liver Diseases, Tangdu Hospital, the Fourth Military Medical University, Xi'an, China
| | - Zhan Sheng Jia
- 1 Department of Infectious Diseases and Center of Liver Diseases, Tangdu Hospital, the Fourth Military Medical University, Xi'an, China
| |
Collapse
|
28
|
Schirdewahn T, Grabowski J, Owusu Sekyere S, Bremer B, Wranke A, Lunemann S, Schlaphoff V, Kirschner J, Hardtke S, Manns MP, Cornberg M, Wedemeyer H, Suneetha PV. The Third Signal Cytokine Interleukin 12 Rather Than Immune Checkpoint Inhibitors Contributes to the Functional Restoration of Hepatitis D Virus-Specific T Cells. J Infect Dis 2016; 215:139-149. [PMID: 27803174 DOI: 10.1093/infdis/jiw514] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2016] [Accepted: 10/18/2016] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Hepatitis D virus (HDV) infection affects 15-20 million individuals worldwide and causes severely progressive hepatitis. It is unknown to what extent cellular immune responses contribute to liver disease and control of viral replication in HDV infection. METHODS Immune cell frequencies and phenotypes were determined in 49 HDV-infected patients, 25 individuals with hepatitis B virus monoinfection and 18 healthy controls. T-cell proliferative and cytokine-producing capacities were analyzed by stimulation with overlapping peptides spanning the large HDV antigen. To restore T-cell responses, blocking antibodies (anti-cytotoxic T-lymphocyte antigen 4, anti-programmed death ligand 1) or proinflammatory cytokines (interleukin [IL] 12) were used. RESULTS Immune cell frequencies and phenotypes did not vary between the groups. Exclusively, the senescence marker CD57 was significantly up-regulated in CD8+ T cells from patients with hepatitis delta. HDV-specific T-cell proliferation and cytokine production were weak and could only partly be rescued by blockade of the programmed death 1 pathway. However, a more robust and consistent increase in HDV-specific CD4+ and CD8+ T-cell responses was evident when the third signal cytokine IL-12 was added, which also affected cytomegalovirus- and Epstein-Barr virus-specific T cells. CONCLUSIONS This investigation of virus-specific T-cell immunity in patients with HDV infection, the largest to date, revealed premature aging of immune cells and impaired T-cell functionality. This could be restored by blocking inhibitory pathways and, in particular, by supplementing with IL-12.
Collapse
Affiliation(s)
- Thomas Schirdewahn
- Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School.,German Center for Infection Research, Partner Site Hannover-Braunschweig
| | - Jan Grabowski
- Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School
| | - Solomon Owusu Sekyere
- Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School
| | - Birgit Bremer
- Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School
| | - Anika Wranke
- Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School
| | - Sebastian Lunemann
- Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School
| | - Verena Schlaphoff
- Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School
| | - Janina Kirschner
- Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School
| | - Svenja Hardtke
- Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School.,German Center for Infection Research, Partner Site Hannover-Braunschweig
| | - Michael Peter Manns
- Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School.,German Center for Infection Research, Partner Site Hannover-Braunschweig
| | - Markus Cornberg
- Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School.,German Center for Infection Research, Partner Site Hannover-Braunschweig
| | - Heiner Wedemeyer
- Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School.,German Center for Infection Research, Partner Site Hannover-Braunschweig
| | | |
Collapse
|
29
|
Abstract
Hepatitis A virus (HAV), hepatitis B virus (HBV) and hepatitis C virus (HCV) are responsible for most cases of viral hepatitis. Infection by each type of virus results in a different typical natural disease course and clinical outcome that are determined by virological and immunological factors. HCV tends to establish a chronic persistent infection, whereas HAV does not. HBV is effectively controlled in adults, although it persists for a lifetime after neonatal infection. In this Review, we discuss the similarities and differences in immune responses to and immunopathogenesis of HAV, HBV and HCV infections, which may explain the distinct courses and outcomes of each hepatitis virus infection.
Collapse
|
30
|
Sanchez-Fueyo A, Markmann JF. Immune Exhaustion and Transplantation. Am J Transplant 2016; 16:1953-7. [PMID: 26729653 DOI: 10.1111/ajt.13702] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2015] [Revised: 11/09/2015] [Accepted: 12/13/2015] [Indexed: 01/25/2023]
Abstract
Exhaustion of lymphocyte function through chronic exposure to a high load of foreign antigen is well established for chronic viral infection and antitumor immunity and has been found to be associated with a distinct molecular program and characteristic cell surface phenotype. Although exhaustion has most commonly been studied in the context of CD8 viral responses, recent studies indicate that chronic antigen exposure may affect B cells, NK cells and CD4 T cells in a parallel manner. Limited information is available regarding the extent of lymphocyte exhaustion development in the transplant setting and its impact on anti-graft alloreactivity. By analogy to the persistence of a foreign virus, the large mass of alloantigen presented by an allograft in chronic residence could provide an ideal setting for exhausting donor-reactive T cells. The extent of T cell exhaustion occurring with various allografts, the kinetics of its development, whether exhaustion is influenced positively or negatively by different immunosuppressants, and the impact of exhaustion on graft survival and tolerance development remains a fertile area for investigation. Harnessing or encouraging the natural processes of exhaustion may provide a novel means to promote graft survival and transplantation tolerance.
Collapse
Affiliation(s)
- A Sanchez-Fueyo
- Institute of Liver Studies, Division of Transplantation Immunology and Mucosal Biology, Medical Research Council Centre for Transplantation, Faculty of Life Sciences and Medicine, King's College London University, King's College Hospital, London, UK
| | - J F Markmann
- Division of Transplant Surgery, Center for Transplantation Sciences, Department of Surgery, Massachusetts General Hospital, Boston, MA
| |
Collapse
|
31
|
Sabins NC, Harman BC, Barone LR, Shen S, Santulli-Marotto S. Differential Expression of Immune Checkpoint Modulators on In Vitro Primed CD4(+) and CD8(+) T Cells. Front Immunol 2016; 7:221. [PMID: 27379090 PMCID: PMC4909735 DOI: 10.3389/fimmu.2016.00221] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2016] [Accepted: 05/23/2016] [Indexed: 12/13/2022] Open
Abstract
PD-1, TIM-3, and LAG-3 are molecules shown to have immune modulatory properties, and although initially classified as indicators of T cell hyporesponsiveness, it has become clear that they are also associated with the normal course of T cell activation. Functional studies have focused mainly on CD8+ T cells during chronic inflammation due to interest in co-opting the cellular immune response to eliminate viral or cancerous threats; however, there remains a relative lack of data regarding the expression of these molecules on CD4+ T cells. Here, we report that expression of the immune checkpoint (IC) molecules PD-1, LAG-3, and TIM-3 are differentially expressed on CD4+ and CD8+ T cells in the allogeneic response resulting from a mixed lymphocyte reaction. In these studies, PD-1 expression is higher on CD4+ T cells compared to CD8+ T cells. In contrast, TIM-3 is expressed at higher levels on CD8+ T cells compared to CD4+ T cells with an apparent reciprocity in that PD-1+ CD4+ T cells are frequently TIM-3lo/−, while TIM-3-expressing CD8+ T cells are largely PD-1lo/−. In addition, there is a decrease in the frequency of TIM-3+ CD4+ cells producing IFN-γ and IL-5 compared to TIM-3+ CD8+ cells. Lastly, the memory T cell phenotype within each IC-expressing subset differs between CD4+ and CD8+ T cells. These findings highlight key differences in IC expression patterns between CD4+ and CD8+ T cells and may allow for more effective therapeutic targeting of these molecules in the future.
Collapse
Affiliation(s)
- Nina C Sabins
- Biologics Research, Janssen R&D , Spring House, PA , USA
| | | | - Linda R Barone
- Biologics Research, Janssen R&D , Spring House, PA , USA
| | - Shixue Shen
- Biologics Research, Janssen R&D , Spring House, PA , USA
| | | |
Collapse
|
32
|
Callendret B, Eccleston HB, Satterfield W, Capone S, Folgori A, Cortese R, Nicosia A, Walker CM. Persistent hepatitis C viral replication despite priming of functional CD8+ T cells by combined therapy with a vaccine and a direct-acting antiviral. Hepatology 2016; 63:1442-54. [PMID: 26513111 PMCID: PMC4840073 DOI: 10.1002/hep.28309] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/15/2015] [Revised: 10/22/2015] [Accepted: 10/25/2015] [Indexed: 02/06/2023]
Abstract
UNLABELLED Exhaustion of antiviral CD8(+) T cells contributes to persistence of hepatitis C viral (HCV) infection. This immune response has proved difficult to restore by therapeutic vaccination, even when HCV replication is suppressed using antiviral regimens containing type I interferon. Because immunomodulatory effects of type I interferon may be a factor in poor T-cell priming, we undertook therapeutic vaccination in two chronically infected chimpanzees during treatment with a direct-acting antiviral (DAA) targeting the HCV NS5b polymerase protein. Immunization with genetic vaccines encoding the HCV NS3-NS5b nonstructural proteins during DAA treatment resulted in a multifunctional CD8(+) T-cell response. However, these antiviral CD8(+) T cells did not prevent persistent replication of DAA-resistant HCV variants that emerged during treatment. Most vaccine-induced CD8(+) T cells targeted class I epitopes that were not conserved in the circulating virus. Exhausted intrahepatic CD8(+) T-cell targeting-conserved epitopes did not expand after vaccination, with a notable exception. A sustained, multifunctional CD8(+) T-cell response against at least one intact class I epitope was detected in blood after vaccination. Persistence of HCV was not due to mutational escape of this epitope. Instead, failure to control HCV replication was likely caused by localized exhaustion in the liver, where CD8(+) T-cell expression of the inhibitory receptor programmed cell death 1 increased 25-fold compared with those in circulation. CONCLUSION Treatment with a DAA during therapeutic vaccination provided transient control of HCV replication and a multifunctional T-cell response, primarily against nonconserved class I epitopes; exhaustion of liver-infiltrating CD8(+) T cells that target conserved epitopes may not be averted when DAA therapy fails prematurely due to emergence of resistant HCV variants.
Collapse
Affiliation(s)
- Benoit Callendret
- Center for Vaccines and Immunity, Nationwide Children’s Hospital, Columbus, OH 43205
| | - Heather B. Eccleston
- Center for Vaccines and Immunity, Nationwide Children’s Hospital, Columbus, OH 43205
| | - William Satterfield
- Department of Veterinary Sciences, Michale E. Keeling Center for Comparative Medicine and Research, M. D. Anderson Cancer Center, Bastrop, TX 78602
| | | | | | | | - Alfredo Nicosia
- ReiThera, viale Citta’ d’Europa 679, 00144, Rome, Italy,KEIRES, Bäumleingasse 18, CH 4051, Basel, Switzerland,CEINGE, via Gaetano Salvatore 486, 80145, Naples, Italy
| | - Christopher M. Walker
- Center for Vaccines and Immunity, Nationwide Children’s Hospital, Columbus, OH 43205,Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, via S. Pansini 5, 80131, Naples, Italy
| |
Collapse
|
33
|
Hato T, Zhu AX, Duda DG. Rationally combining anti-VEGF therapy with checkpoint inhibitors in hepatocellular carcinoma. Immunotherapy 2016; 8:299-313. [PMID: 26865127 DOI: 10.2217/imt.15.126] [Citation(s) in RCA: 97] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is a fatal disease with rising incidence in the world. For advanced HCC, sorafenib, a multikinase inhibitor, is the only systemic therapy with proven survival benefits. Sorafenib is a pan-VEGF receptor inhibitor, and thus many studies have focused its antivascular effects. But VEGF also acts as an immunosuppressive molecule. VEGF can inhibit maturation of dendritic cells, promote immune suppressive cell infiltration and enhance immune checkpoint molecules expression. On the other hand, potent VEGF inhibition may increase tumor hypoxia, which could hinder antitumor immunity or immunotherapy. Thus, achieving synergy when combining anti-VEGF therapy with immunotherapy may require proper polarization of the tumor microenvironment by dose titration or combination with other immunomodulating agents.
Collapse
Affiliation(s)
- Tai Hato
- Edwin L. Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA.,Department of Surgery, Keio University School of Medicine, Tokyo 160-8582, Japan
| | - Andrew X Zhu
- Hematology/Oncology, Department of Medicine, Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA 02114, USA
| | - Dan G Duda
- Edwin L. Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| |
Collapse
|
34
|
van Driel BJ, Liao G, Engel P, Terhorst C. Responses to Microbial Challenges by SLAMF Receptors. Front Immunol 2016; 7:4. [PMID: 26834746 PMCID: PMC4718992 DOI: 10.3389/fimmu.2016.00004] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2015] [Accepted: 01/06/2016] [Indexed: 12/24/2022] Open
Abstract
The SLAMF family (SLAMF) of cell surface glycoproteins is comprised of nine glycoproteins and while SLAMF1, 3, 5, 6, 7, 8, and 9 are self-ligand receptors, SLAMF2 and SLAMF4 interact with each other. Their interactions induce signal transduction networks in trans, thereby shaping immune cell-cell communications. Collectively, these receptors modulate a wide range of functions, such as myeloid cell and lymphocyte development, and T and B cell responses to microbes and parasites. In addition, several SLAMF receptors serve as microbial sensors, which either positively or negatively modulate the function of macrophages, dendritic cells, neutrophils, and NK cells in response to microbial challenges. The SLAMF receptor-microbe interactions contribute both to intracellular microbicidal activity as well as to migration of phagocytes to the site of inflammation. In this review, we describe the current knowledge on how the SLAMF receptors and their specific adapters SLAM-associated protein and EAT-2 regulate innate and adaptive immune responses to microbes.
Collapse
Affiliation(s)
- Boaz Job van Driel
- Division of Immunology, Beth Israel Deaconess Medical Center, Harvard Medical School , Boston, MA , USA
| | - Gongxian Liao
- Division of Immunology, Beth Israel Deaconess Medical Center, Harvard Medical School , Boston, MA , USA
| | - Pablo Engel
- Immunology Unit, Department of Cell Biology, Immunology and Neurosciences, Medical School, University of Barcelona , Barcelona , Spain
| | - Cox Terhorst
- Division of Immunology, Beth Israel Deaconess Medical Center, Harvard Medical School , Boston, MA , USA
| |
Collapse
|
35
|
Quarleri JF, Oubiña JR. Hepatitis C virus strategies to evade the specific-T cell response: a possible mission favoring its persistence. Ann Hepatol 2016; 15:17-26. [PMID: 26626636 DOI: 10.5604/16652681.1184193] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Hepatitis C virus (HCV) is a small, enveloped RNA virus. The number of HCV-infected individuals worldwide is estimated to be approximately 200 million. The vast majority of HCV infections persist, with up to 80% of all cases leading to chronic hepatitis associated with liver fibrosis, cirrhosis, and hepatocellular carcinoma. The interaction between HCV and the host have a pivotal role in viral fitness, persistence, pathogenicity, and disease progression. The control of HCV infection requires both effective innate and adaptive immune responses. The HCV clearance during acute infection is associated with an early induction of the innate and a delayed initiation of the adaptive immune responses. However, in the vast majority of acute HCV infections, these responses are overcome and the virus persistence almost inexorably occurs. Recently, several host- and virus-related mechanisms responsible for the failure of both the innate and the adaptive immune responses have been recognized. Among the latter, the wide range of escape mutations to evade the specific-T-and B-cell responses as well as the T cell anergy and the CD8+ T cell exhaustion together with the interference with its function after prolonged virus exposure hold a pivotal role. Other HCV strategies include the modification or manipulation of molecules playing key roles in the induction of the interferon response and its induced effector proteins. In this review, we attempt to gain insights on the main T cell immune evasion strategies used by the virus in order to favor its persistence.
Collapse
Affiliation(s)
- Jorge Fabián Quarleri
- Instituto de Investigaciones Biomédicas en Retrovirus y Sida (INBIRS), Universidad de Buenos Aires-CONICET, Argentina
| | - José Raúl Oubiña
- Instituto de Microbiología y Parasitología Médica (IMPAM), Universidad de Buenos Aires-CONICET, Argentina
| |
Collapse
|
36
|
Chen N, Liu Y, Guo Y, Chen Y, Liu X, Liu M. Lymphocyte activation gene 3 negatively regulates the function of intrahepatic hepatitis C virus-specific CD8+ T cells. J Gastroenterol Hepatol 2015; 30:1788-95. [PMID: 26095288 DOI: 10.1111/jgh.13017] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/03/2015] [Revised: 05/21/2015] [Accepted: 05/26/2015] [Indexed: 12/19/2022]
Abstract
BACKGROUND AND AIM Chronic hepatitis C (CHC) in humans caused by persistent hepatitis C virus (HCV) infection is a global public health problem. The functional exhaustion of HCV-specific CD8(+) T cells regulated by several inhibitory receptors has been shown to contribute to chronic HCV infection. Lymphocyte activation gene 3 (LAG-3), which is an inhibitory receptor, plays an important role in several chronic viral infections. However, its effect on the function of HCV-specific CD8(+) T cells is unclear. METHODS The expression of LAG-3 on the CD8(+) T cells in intrahepatic and peripheral lymphocytes from 17 CHC patients and 15 HCV-negative patients was analyzed by flow cytometry. The LAG-3 expression in CD8(+) T cells was downregulated or upregulated by lentivirus LAG-3 shRNA or lentivirus overexpressing LAG-3. After HCV peptide stimulation, flow cytometry was used to detect cell proliferation and cytokine (γ-interferon [IFN-γ], tumor necrosis factor-α [TNF-α], granzyme B, and perforin) production of CD8(+) T cells. Cytotoxicity functions of HCV-specific CD8(+) T cells were measured using a (51) Cr release assay. RESULTS The frequency of LAG-3-positive intrahepatic and peripheral CD8(+) T cells was higher in CHC patients, compared with HCV-negative patients. The cell proliferation, cytokine (IFN-γ, TNF-α, granzyme B, and perforin) expression and cytotoxicity function of HCV-specific CD8(+) T cells in CHC patients were increased by the knocking down and blockade of LAG-3. In the LAG-3 overexpressed CD8(+) T cells, cell proliferation, cytokine (IFN-γ, TNF-α, granzyme B, and perforin) expression, and cytotoxicity function were inhibited, while the LAG-3 blocking antibody reversed the inhibition. CONCLUSION LAG-3 negatively regulated the function of HCV-specific CD8(+) T cells in CHC patients.
Collapse
Affiliation(s)
- Na Chen
- Department of Infectious Diseases, First Affiliated Hospital, Medical College of Xi'an Jiaotong University, Xi'an, China
| | - Yehong Liu
- Department of Infectious Diseases, First Affiliated Hospital, Medical College of Xi'an Jiaotong University, Xi'an, China
| | - Yonghong Guo
- Department of Infectious Diseases, Second Affiliated Hospital, Medical College of Xi'an Jiaotong University, Xi'an, China
| | - Yunru Chen
- Department of Infectious Diseases, First Affiliated Hospital, Medical College of Xi'an Jiaotong University, Xi'an, China
| | - Xiaojing Liu
- Department of Infectious Diseases, First Affiliated Hospital, Medical College of Xi'an Jiaotong University, Xi'an, China
| | - Min Liu
- Department of Infectious Diseases, First Affiliated Hospital, Medical College of Xi'an Jiaotong University, Xi'an, China
| |
Collapse
|
37
|
Abstract
Despite advances in therapy, hepatitis C virus infection remains a major global health issue with 3 to 4 million incident cases and 170 million prevalent chronic infections. Complex, partially understood, host-virus interactions determine whether an acute infection with hepatitis C resolves, as occurs in approximately 30% of cases, or generates a persistent hepatic infection, as occurs in the remainder. Once chronic infection is established, the velocity of hepatocyte injury and resultant fibrosis is significantly modulated by immunologic as well as environmental factors. Immunomodulation has been the backbone of antiviral therapy despite poor understanding of its mechanism of action.
Collapse
Affiliation(s)
- David E. Kaplan
- Medicine and Research Services, Philadelphia VA Medical Center, Philadelphia PA,Division of Gastroenterology, Department of Medicine, University of Pennsylvania
| |
Collapse
|
38
|
Harding JJ, El Dika I, Abou-Alfa GK. Immunotherapy in hepatocellular carcinoma: Primed to make a difference? Cancer 2015; 122:367-77. [PMID: 26540029 DOI: 10.1002/cncr.29769] [Citation(s) in RCA: 101] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2015] [Revised: 09/04/2015] [Accepted: 09/17/2015] [Indexed: 12/14/2022]
Abstract
Advanced hepatocellular carcinoma (HCC) carries a dismal prognosis and the current treatment is limited to sorafenib, an agent with modest benefit. Preclinical data have indicated that several immunologic mechanisms are at play to promote HCC development and growth while impairing effective antitumor immune surveillance. Several novel approaches geared toward manipulating the immune response to HCC have suggested a therapeutic benefit in early-stage clinical trials, indicating a real potential to augment tumor-specific immunity and improve outcomes in patients with this disease. In the current study, the authors reviewed the barriers to an effective immune response against HCC and contemporary clinical investigations that may be "primed" to alter the natural history of HCC.
Collapse
Affiliation(s)
- James J Harding
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York.,Weill Cornell Medical College, New York, New York
| | - Imane El Dika
- Internal Medicine/Hematology and Oncology, American University of Beirut, Beirut, Lebanon
| | - Ghassan K Abou-Alfa
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York.,Weill Cornell Medical College, New York, New York
| |
Collapse
|
39
|
Wong YC, Tay SS, McCaughan GW, Bowen DG, Bertolino P. Immune outcomes in the liver: Is CD8 T cell fate determined by the environment? J Hepatol 2015; 63:1005-14. [PMID: 26103545 DOI: 10.1016/j.jhep.2015.05.033] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2015] [Revised: 05/22/2015] [Accepted: 05/26/2015] [Indexed: 02/07/2023]
Abstract
The liver is known for its tolerogenic properties. This unique characteristic is associated with persistent infection of the liver by the hepatitis B and C viruses. Improper activation of cellular adaptive immune responses within the liver and immune exhaustion over time both contribute to ineffective cytotoxic T cell responses to liver-expressed antigens in animal models, and likely play a role in incomplete clearance of chronic hepatitis virus infections in humans. However, under some conditions, functional immune responses can be elicited against hepatic antigens, resulting in control of hepatotropic infections. In order to develop improved therapeutics in immune-mediated chronic liver diseases, including viral hepatitis, it is essential to understand how intrahepatic immunity is regulated. This review focuses on CD8 T cell immunity directed towards foreign antigens expressed in the liver, and explores how the liver environment dictates the outcome of intrahepatic CD8 T cell responses. Potential strategies to rescue unresponsive CD8 T cells in the liver are also discussed.
Collapse
Affiliation(s)
- Yik Chun Wong
- Liver Immunology Group, Centenary Institute and AW Morrow Gastroenterology and Liver Centre, University of Sydney and Royal Prince Alfred Hospital, Sydney, NSW, Australia.
| | - Szun Szun Tay
- Liver Immunology Group, Centenary Institute and AW Morrow Gastroenterology and Liver Centre, University of Sydney and Royal Prince Alfred Hospital, Sydney, NSW, Australia
| | - Geoffrey W McCaughan
- Liver Cancer and Injury Group, Centenary Institute and AW Morrow Gastroenterology and Liver Centre, University of Sydney and Royal Prince Alfred Hospital, Sydney, NSW, Australia
| | - David G Bowen
- Liver Immunology Group, Centenary Institute and AW Morrow Gastroenterology and Liver Centre, University of Sydney and Royal Prince Alfred Hospital, Sydney, NSW, Australia
| | - Patrick Bertolino
- Liver Immunology Group, Centenary Institute and AW Morrow Gastroenterology and Liver Centre, University of Sydney and Royal Prince Alfred Hospital, Sydney, NSW, Australia.
| |
Collapse
|
40
|
Xiao W, Zhang Q, Deng XZ, Jiang LF, Zhu DY, Pei JP, Ge CY, Li BJ, Wang CJ, Zhang JH, Zhou ZX, Ding WL, Xu XD, Yue M. HCV F protein amplifies the predictions of IL-28B and CTLA-4 polymorphisms about the susceptibility and outcomes of HCV infection in Southeast China. INFECTION, GENETICS AND EVOLUTION : JOURNAL OF MOLECULAR EPIDEMIOLOGY AND EVOLUTIONARY GENETICS IN INFECTIOUS DISEASES 2015; 34:52-60. [PMID: 26079279 DOI: 10.1016/j.meegid.2015.06.016] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/07/2015] [Revised: 06/01/2015] [Accepted: 06/12/2015] [Indexed: 12/20/2022]
Abstract
Cytotoxic T lymphocyte associated antigen-4(CTLA-4) is an inhibitory receptor with great value in the progression of hepatitis C virus (HCV) infection related diseases. To determine the potential associations of IL-28B rs12979860 and CTLA-4 rs231775, rs3087243 and rs5742909 polymorphisms with the generation of HCV F protein, susceptibility and outcomes of HCV infection, a total of 375 healthy controls, 219 HCV spontaneous recovered patients and 600 chronic HCV patients from Southeast China were recruited and genotyped in this study. And the relative mRNA levels of CTLA-4 in T cells were detected. Logistic regression analysis showed that rs231775 A allele was associated with significantly higher rate of spontaneous viral clearance in anti-HCV F antibody negative patients (adjusted OR=0.512, P=0.008), but allele A was related to higher mRNA level of CTLA-4 with the generation of HCV F protein. And rs5742909 T allele added up to the risk of HCV infection chronicity significantly in patients with the presence of HCV F protein (adjusted OR=2.698, P=0.003). Also, the rs5742909 CC genotype, along with the presence of HCV F protein, indicated a significantly higher CTLA-4 level than that in anti-HCV F antibody negative patients. The AG+AA genotype of rs3087243 significantly increased the susceptibility to HCV infection in subjects over 56 years old (adjusted OR=1.595, P=0.011). Genotype-genotype interaction between IL-28B rs12979860 and CTLA-4 rs3087243 was found to be significantly associated with increased susceptibility to HCV infection (adjusted OR=1.509, P=0.005). Haplotype analysis in CTLA-4 also showed significant association with the generation of HCV F protein. All these results indicated the importance of IL-28B and CTLA-4 polymorphisms and their associations with HCV F protein in the risk and chronicity of HCV infection in Chinese Han population in Southeast China.
Collapse
Affiliation(s)
- Wen Xiao
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, China.
| | - Qi Zhang
- Huadong Research Institute for Medicine and Biotechnics, Nanjing, China
| | - Xiao Zhao Deng
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, China; Huadong Research Institute for Medicine and Biotechnics, Nanjing, China.
| | - Long Feng Jiang
- Department of Infectious Diseases, The First Affiliated Hospital with Nanjing Medical University, Nanjing, China
| | - Dan Yan Zhu
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Nanjing Medical University, Nanjing, China
| | - Jia Ping Pei
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Nanjing Medical University, Nanjing, China
| | - Chi Yu Ge
- Jiangsu Food & Pharmaceutical Science College, Huaian, China
| | - Bing Jun Li
- Huadong Research Institute for Medicine and Biotechnics, Nanjing, China
| | - Chang Jun Wang
- Huadong Research Institute for Medicine and Biotechnics, Nanjing, China.
| | - Jing Hai Zhang
- Huadong Research Institute for Medicine and Biotechnics, Nanjing, China
| | - Zhen Xian Zhou
- Department of Clinical Laboratory, Nanjing Second Hospital, Nanjing, China
| | - Wei Liang Ding
- Department of Clinical Laboratory, Yixing People's Hospital, Yixing, China
| | - Xiao Dong Xu
- Huadong Research Institute for Medicine and Biotechnics, Nanjing, China
| | - Ming Yue
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| |
Collapse
|
41
|
Owusu Sekyere S, Suneetha PV, Hardtke S, Falk CS, Hengst J, Manns MP, Cornberg M, Wedemeyer H, Schlaphoff V. Type I Interferon Elevates Co-Regulatory Receptor Expression on CMV- and EBV-Specific CD8 T Cells in Chronic Hepatitis C. Front Immunol 2015; 6:270. [PMID: 26113847 PMCID: PMC4462106 DOI: 10.3389/fimmu.2015.00270] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2015] [Accepted: 05/15/2015] [Indexed: 01/13/2023] Open
Abstract
Hepatitis C virus (HCV) readily sets up persistence in a large fraction of infected hosts. Mounting epidemiological and immunological evidence suggest that HCV's persistence could influence immune responses toward unrelated pathogens and vaccines. Nonetheless, the fundamental contribution of the inflammatory milieu during persistent HCV infection in impacting immune cells specific for common pathogens such as CMV and EBV has not been fully studied. As the co-regulatory receptors PD-1, Tim-3, and 2B4 have all been shown to be vital in regulating CD8(+) T cell function, we assessed their expression on CMV/EBV-specific CD8(+) T cells from patients with chronic hepatitis C (CHC) and healthy controls ex vivo and upon stimulation with virus-specific peptides in vitro. Total and CMV/EBV-specific CD8(+) T cells expressing PD-1, Tim-3, and 2B4 were highly enriched in patients with CHC compared to healthy individuals ex vivo. In vitro peptide stimulation further potentiated the differential co-regulatory receptor expression of PD-1, Tim-3, and 2B4, which then culminated in an enhanced functionality of CMV/EBV-specific CD8(+) T cells in CHC patients. Comprehensively analyzing plasma cytokines between the two cohorts, we observed that not only was IFNα-2a dominant among 21 other inflammatory mediators elevated in CHC patients but it also correlated with PD-1 and Tim-3 expressions ex vivo. Importantly, IFNα-2a further caused upregulation of these markers upon in vitro peptide stimulation. Finally, we could prospectively study patients receiving novel IFN-free antiviral therapy. Here, we observed that treatment-induced clearance of HCV resulted in a partial reversion of the phenotype of CMV/EBV-specific CD8(+) T cells in patients with CHC. These data reveal an alteration of the plasma concentrations of IFNα-2a together with other inflammatory mediators during CHC, which appeared to pervasively influence co-regulatory receptor expression on CMV/EBV-specific CD8(+) T cells.
Collapse
Affiliation(s)
- Solomon Owusu Sekyere
- Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School , Hannover , Germany
| | | | - Svenja Hardtke
- Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School , Hannover , Germany
| | - Christine Susanne Falk
- TTU-Hepatitis, TTU-IICH, German Center for Infectious Diseases (DZIF) , Hannover-Braunschweig , Germany ; Institute of Transplantation Immunology (IFB-Tx), Hannover Medical School , Hannover , Germany
| | - Julia Hengst
- Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School , Hannover , Germany
| | - Michael Peter Manns
- Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School , Hannover , Germany ; TTU-Hepatitis, TTU-IICH, German Center for Infectious Diseases (DZIF) , Hannover-Braunschweig , Germany
| | - Markus Cornberg
- Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School , Hannover , Germany ; TTU-Hepatitis, TTU-IICH, German Center for Infectious Diseases (DZIF) , Hannover-Braunschweig , Germany
| | - Heiner Wedemeyer
- Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School , Hannover , Germany ; TTU-Hepatitis, TTU-IICH, German Center for Infectious Diseases (DZIF) , Hannover-Braunschweig , Germany
| | - Verena Schlaphoff
- Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School , Hannover , Germany
| |
Collapse
|
42
|
Zhai N, Chi X, Li T, Song H, Li H, Jin X, Crispe IN, Su L, Niu J, Tu Z. Hepatitis C virus core protein triggers expansion and activation of CD4(+)CD25(+) regulatory T cells in chronic hepatitis C patients. Cell Mol Immunol 2014; 12:743-9. [PMID: 25531392 DOI: 10.1038/cmi.2014.119] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2014] [Revised: 11/06/2014] [Accepted: 11/07/2014] [Indexed: 01/08/2023] Open
Abstract
CD4(+)CD25(+)FoxP3(+) regulatory T cells (Tregs) are increased in patients with chronic hepatitis C, which may contribute to the sustained suppression of hepatitis C virus (HCV)-specific T-cell responses and viral persistence in HCV-infected individuals. We postulated that HCV core protein (HCVc) directly contributes to the expansion of Tregs in HCV-infected patients, and we provide evidence to support this hypothesis in the report. Peripheral blood mononuclear cells (PBMCs) and sera were collected from 87 treatment-naïve chronic HCV-infected patients, CD4(+)CD25(+) Tregs were measured by flow cytometry, and HCV RNA and HCVc levels were detected using qPCR and enzyme-linked immunosorbent assay (ELISA), respectively. CD4(+), CD8(+), CD4(+)CD25(+) and CD4(+)CD25(-) T cells were purified from healthy donors and cultured with recombinant HCVc and Toll-like receptor (TLR) ligands. Flow cytometry was used to analyze cell proliferation, and ELISA was performed to measure cytokine production. In the 87 chronic HCV-infected patients, HCVc showed a significant correlation with HCV RNA and CD4(+)CD25(+) Tregs. Mechanistic studies showed that HCVc, together with anti-CD3 antibody, augmented CD4(+)CD25(+) Treg proliferation, but inhibited CD4(+)CD25(-) T-cell proliferation and IFN-γ production, in a dose-dependent and Treg-dependent manner. Moreover, unlike the TLR3 ligand (poly I:C) and the TLR4 ligand (lipopolysaccharide, LPS), the TLR2 ligand (lipoteichoic acid, LTA) and HCVc both inhibited TCR-induced CD4(+) T-cell proliferation and IFN-γ secretion in a Treg-dependent manner. These data indicate that HCVc, like other TLR2 ligands, triggers CD4(+)CD25(+) Treg activation and expansion to inhibit host immune responses, which may play a critical role in viral persistence in HCV-infected patients.
Collapse
Affiliation(s)
- Naicui Zhai
- Institute of Translational Medicine, The First Hospital, Jilin University, Changchun, China
| | - Xiumei Chi
- Department of Hepatology, The First Hospital, Jilin University, Changchun, China
| | - Tianyang Li
- Institute of Translational Medicine, The First Hospital, Jilin University, Changchun, China
| | - Hongxiao Song
- Institute of Translational Medicine, The First Hospital, Jilin University, Changchun, China
| | - Haijun Li
- Institute of Translational Medicine, The First Hospital, Jilin University, Changchun, China
| | - Xia Jin
- Institute Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai, China
| | | | - Lishan Su
- Institute of Translational Medicine, The First Hospital, Jilin University, Changchun, China.,Lineberger Comprehensive Cancer Center, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Junqi Niu
- Department of Hepatology, The First Hospital, Jilin University, Changchun, China
| | - Zhengkun Tu
- Institute of Translational Medicine, The First Hospital, Jilin University, Changchun, China.,Department of Hepatology, The First Hospital, Jilin University, Changchun, China
| |
Collapse
|