Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 46
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Gastroenterology ; 2024 Apr 16.
Article in English | MEDLINE | ID: mdl-38636680

ABSTRACT

BACKGROUND & AIMS: High expression of phosphatidylinositol 4-kinase III alpha (PI4KIIIα) correlates with poor survival rates in patients with hepatocellular carcinoma. In addition, hepatitis C virus (HCV) infections activate PI4KIIIα and contribute to hepatocellular carcinoma progression. We aimed at mechanistically understanding the impact of PI4KIIIα on the progression of liver cancer and the potential contribution of HCV in this process. METHODS: Several hepatic cell culture and mouse models were used to study the functional importance of PI4KIIIα on liver pathogenesis. Antibody arrays, gene silencing, and PI4KIIIα-specific inhibitor were applied to identify the involved signaling pathways. The contribution of HCV was examined by using HCV infection or overexpression of its nonstructural protein. RESULTS: High PI4KIIIα expression and/or activity induced cytoskeletal rearrangements via increased phosphorylation of paxillin and cofilin. This led to morphologic alterations and higher migratory and invasive properties of liver cancer cells. We further identified the liver-specific lipid kinase phosphatidylinositol 3-kinase C2 domain-containing subunit gamma (PIK3C2γ) working downstream of PI4KIIIα in regulation of the cytoskeleton. PIK3C2γ generates plasma membrane phosphatidylinositol 3,4-bisphosphate-enriched, invadopodia-like structures that regulate cytoskeletal reorganization by promoting Akt2 phosphorylation. CONCLUSIONS: PI4KIIIα regulates cytoskeleton organization via PIK3C2γ/Akt2/paxillin-cofilin to favor migration and invasion of liver cancer cells. These findings provide mechanistic insight into the contribution of PI4KIIIα and HCV to the progression of liver cancer and identify promising targets for therapeutic intervention.

2.
Mol Ther ; 32(2): 540-555, 2024 Feb 07.
Article in English | MEDLINE | ID: mdl-38213030

ABSTRACT

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2)-specific neutralizing antibodies (NAbs) lack cross-reactivity between SARS-CoV species and variants and fail to mediate long-term protection against infection. The maintained protection against severe disease and death by vaccination suggests a role for cross-reactive T cells. We generated vaccines containing sequences from the spike or receptor binding domain, the membrane and/or nucleoprotein that induced only T cells, or T cells and NAbs, to understand their individual roles. In three models with homologous or heterologous challenge, high levels of vaccine-induced SARS-CoV-2 NAbs protected against neither infection nor mild histological disease but conferred rapid viral control limiting the histological damage. With no or low levels of NAbs, vaccine-primed T cells, in mice mainly CD8+ T cells, partially controlled viral replication and promoted NAb recall responses. T cells failed to protect against histological damage, presumably because of viral spread and subsequent T cell-mediated killing. Neither vaccine- nor infection-induced NAbs seem to provide long-lasting protective immunity against SARS-CoV-2. Thus, a more realistic approach for universal SARS-CoV-2 vaccines should be to aim for broadly cross-reactive NAbs in combination with long-lasting highly cross-reactive T cells. Long-lived cross-reactive T cells are likely key to prevent severe disease and fatalities during current and future pandemics.


Subject(s)
Antibodies, Neutralizing , COVID-19 Vaccines , COVID-19 , Animals , Humans , Mice , Antibodies, Viral , CD8-Positive T-Lymphocytes , COVID-19/prevention & control , COVID-19 Vaccines/adverse effects , SARS-CoV-2 , Viral Vaccines
3.
Gut ; 72(6): 1186-1195, 2023 06.
Article in English | MEDLINE | ID: mdl-35977815

ABSTRACT

OBJECTIVE: Chronic HBV/HDV infections are a major cause of liver cancer. Current treatments can only rarely eliminate HBV and HDV. Our previously developed preS1-HDAg immunotherapy could induce neutralising antibodies to HBV in vivo and raise HBV/HDV-specific T-cells. Here, we further investigate if a heterologous prime-boost strategy can circumvent T-cell tolerance and preclude HDV superinfection in vivo. DESIGN: A DNA prime-protein boost strategy was evaluated for immunogenicity in mice and rabbits. Its ability to circumvent T-cell tolerance was assessed in immunocompetent hepatitis B surface antigen (HBsAg)-transgenic mice. Neutralisation of HBV and HDV was evaluated both in vitro and in immunodeficient human-liver chimeric mice upon adoptive transfer. RESULTS: The prime-boost strategy elicits robust HBV/HDV-specific T-cells and preS1-antibodies that can effectively prevent HBV and HDV (co-)infection in vitro and in vivo. In a mouse model representing the chronic HBsAg carrier state, active immunisation primes high levels of preS1-antibodies and HDAg-specific T-cells. Moreover, transfer of vaccine-induced antibodies completely protects HBV-infected human-liver chimeric mice from HDV superinfection. CONCLUSION: The herein described preS1-HDAg immunotherapy is shown to be immunogenic and vaccine-induced antibodies are highly effective at preventing HBV and HDV (super)infection both in vitro and in vivo. Our vaccine can complement current and future therapies for the control of chronic HBV and HDV infection.


Subject(s)
Hepatitis B, Chronic , Hepatitis B , Superinfection , Humans , Mice , Animals , Rabbits , Hepatitis delta Antigens , Hepatitis B Surface Antigens , Hepatitis B, Chronic/prevention & control , Superinfection/prevention & control , Hepatitis Delta Virus/genetics , Hepatitis B/prevention & control , Hepatitis B virus/genetics , Antibodies, Viral , Mice, Transgenic
4.
J Immunol ; 205(9): 2437-2446, 2020 11 01.
Article in English | MEDLINE | ID: mdl-32878912

ABSTRACT

Coronavirus disease 2019 (COVID-19), caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), emerged in late 2019 and has since become a global pandemic. Pathogen-specific Abs are typically a major predictor of protective immunity, yet human B cell and Ab responses during COVID-19 are not fully understood. In this study, we analyzed Ab-secreting cell and Ab responses in 20 hospitalized COVID-19 patients. The patients exhibited typical symptoms of COVID-19 and presented with reduced lymphocyte numbers and increased T cell and B cell activation. Importantly, we detected an expansion of SARS-CoV-2 nucleocapsid protein-specific Ab-secreting cells in all 20 COVID-19 patients using a multicolor FluoroSpot Assay. Out of the 20 patients, 16 had developed SARS-CoV-2-neutralizing Abs by the time of inclusion in the study. SARS-CoV-2-specific IgA, IgG, and IgM Ab levels positively correlated with SARS-CoV-2-neutralizing Ab titers, suggesting that SARS-CoV-2-specific Ab levels may reflect the titers of neutralizing Abs in COVID-19 patients during the acute phase of infection. Last, we showed that IL-6 and C-reactive protein serum concentrations were higher in patients who were hospitalized for longer, supporting the recent observations that IL-6 and C-reactive protein could be used as markers for COVID-19 severity. Altogether, this study constitutes a detailed description of clinical and immunological parameters in 20 COVID-19 patients, with a focus on B cell and Ab responses, and describes tools to study immune responses to SARS-CoV-2 infection and vaccination.


Subject(s)
Antibodies, Neutralizing/immunology , Antibodies, Viral/immunology , B-Lymphocytes/immunology , Betacoronavirus/immunology , Coronavirus Infections/epidemiology , Coronavirus Infections/immunology , Hospitalization , Pneumonia, Viral/epidemiology , Pneumonia, Viral/immunology , Adult , Aged , Biomarkers/blood , C-Reactive Protein/analysis , COVID-19 , Cohort Studies , Coronavirus Infections/virology , Coronavirus Nucleocapsid Proteins , Female , Humans , Immunoglobulin A/blood , Immunoglobulin G/blood , Immunoglobulin M/blood , Interleukin-6/blood , Lymphocyte Activation , Male , Middle Aged , Nucleocapsid Proteins/immunology , Pandemics , Phosphoproteins , Pneumonia, Viral/virology , SARS-CoV-2 , Sweden/epidemiology
5.
J Infect Dis ; 223(1): 128-138, 2021 01 04.
Article in English | MEDLINE | ID: mdl-31994701

ABSTRACT

BACKGROUND: Chronic hepatitis B and D virus (HBV/HDV) infections can cause cancer. Current HBV therapy using nucleoside analogues (NAs) is life-long and reduces but does not eliminate the risk of cancer. A hallmark of chronic hepatitis B is a dysfunctional HBV-specific T-cell response. We therefore designed an immunotherapy driven by naive healthy T cells specific for the HDV antigen (HDAg) to bypass the need for HBV-specific T cells in order to prime PreS1-specific T cells and PreS1 antibodies blocking HBV entry. METHODS: Ten combinations of PreS1 and/or HDAg sequences were evaluated for induction of PreS1 antibodies and HBV- and HDV-specific T cells in vitro and in vivo. Neutralization of HBV by PreS1-specific murine and rabbit antibodies was evaluated in cell culture, and rabbit anti-PreS1 were tested for neutralization of HBV in mice repopulated with human hepatocytes. RESULTS: The best vaccine candidate induced T cells to PreS1 and HDAg, and PreS1 antibodies blocking HBV entry in vitro. Importantly, adoptive transfer of PreS1 antibodies prevented, or modulated, HBV infection after a subsequent challenge in humanized mice. CONCLUSIONS: We here describe a novel immunotherapy for chronic HBV/HDV that targets viral entry to complement NAs and coming therapies inhibiting viral maturation.


Subject(s)
Hepatitis B virus/immunology , Hepatitis B, Chronic/drug therapy , Hepatitis D, Chronic/drug therapy , Hepatitis Delta Virus/immunology , Virus Internalization/drug effects , Animals , Female , Hepatitis B Vaccines , Hepatocytes/drug effects , Humans , Immunotherapy/methods , Mice , Mice, Inbred C57BL , Mice, SCID , Mice, Transgenic , Rabbits
6.
Gut ; 67(8): 1525-1535, 2018 08.
Article in English | MEDLINE | ID: mdl-28646094

ABSTRACT

OBJECTIVE: HCV is characterised by its ability to establish chronic infection in hepatocytes and to replicate in the presence of an inflammation. We mimicked this situation in vivo in immune-competent mice by syngeneic transplantation of HCV replicon-containing mouse hepatoma cells. DESIGN: A total of 5 million H-2b positive Hep56.1D cells, carrying a subgenomic genotype (gt) 2a replicon (HCV replicon cells) or stably expressing comparable levels of the HCV NS3/4A protease/helicase complex (NS3/4A hepatoma cells), were injected subcutaneously into syngeneic H-2b-restricted mice. Kinetics of tumour growth, HCV RNA replication levels and HCV-specific immune responses were monitored. For immune monitoring, new H-2b-restricted cytotoxic T cell epitopes within the gt2a NS3/4A region were mapped. Immune mice were generated by DNA-based vaccination. RESULTS: HCV replicon and NS3/4A hepatoma cells generated solid tumours in vivo. Similar to what is seen in human HCV infection did HCV RNA replicate in the presence of inflammation. NS3/4A-specific CD8+ T cells seemed to transiently reduce HCV RNA levels. Both CD4+ and CD8+ T cells were required for protection against tumour growth. Vaccine-induced NS3/4A(gt2a)-specific T cells protected against HCV replicon tumours in wild-type, but not in HCV NS3/4A(gt1a)-transgenic mice with dysfunctional HCV-specific T cells. Importantly, as in human HCV infection, HCV replicon cells neither primed nor boosted a strong NS3/4A-specific T cell response. CONCLUSION: Syngeneic transplantation of mouse HCV replicon cells into immune-competent animals mirrors many in vivo events in humans. This system is versatile and can be applied to any genetically modified H-2b-restricted mouse strain.


Subject(s)
Carcinoma, Hepatocellular/pathology , Cell Transplantation , Disease Models, Animal , Hepacivirus , Hepatitis C/etiology , Hepatocytes/transplantation , Animals , Hepatocytes/pathology , Mice , Replicon , Serine Proteases , Viral Nonstructural Proteins
7.
J Virol ; 91(9)2017 05 01.
Article in English | MEDLINE | ID: mdl-28228595

ABSTRACT

Therapy with genetically modified autologous T cells has shown great promise in cancer therapy. For an efficient control of hepatitis C virus (HCV) infection, cytotoxic T cells (CTL) are pivotal, but persistence of activated T cells may lead to liver toxicity. Here, anti-HCV T cell receptors (TCRs) recognizing the HCV nonstructural (NS) NS3 or NS5 viral peptide target were examined by mRNA transfection of human peripheral blood lymphocytes (PBLs) derived from healthy donors as well as chronically infected HCV patients. Immunological analysis shows that while the CTLs expressing the NS5-specific TCR reduced HCV RNA replication by a noncytotoxic mechanism, the NS3-specific TCR-redirected CTLs were polyfunctional and inhibited HCV RNA replication through antigen-specific cytotoxicity. Transcriptome signatures from these two types of CTL responses revealed uniquely expressed gene clusters upon encountering hepatoma target cells presenting endogenously expressed HCV proteins. The NS3 TCR induced a rapid expression of apoptotic signaling pathways and formation of embryonic gene clusters, whereas the NS5A TCR activation induced extended proliferative and metabolic pathways as the HCV target cells survived. Our results provide detailed insights into basic HCV T cell immunology and have clinical relevance for redirecting T cells to target virally infected hepatoma cells.IMPORTANCE Due to the protective ability of HCV-specific T cells and the hepatotoxic potential that they possess, there is a great need for the understanding of the functional aspects of HCV-specific T cells. To circumvent the low level of precursor frequency in patients, we engineered primary CD8+ T cells by mRNA TCR vectors to confer HCV specificity to new T cells. HCV TCRs that differ in antigen specificity and polyfunctionality were examined. mRNA TCR engineering of peripheral blood lymphocytes from healthy donors or chronically infected HCV patients resulted in strikingly high levels of HCV TCR expression and HCV-specific responses. While a cytotoxicity response from a polyfunctional T cell activation caused hepatotoxicity and the rapid induction of apoptotic signaling pathways, the noncytotoxic T cell activation showed extended proliferative, metabolic pathways and persistence of HCV target cells. Our results provide detailed insights into basic HCV T cell immunology and have clinical relevance for immune protection of HCV-associated diseases.


Subject(s)
Hepacivirus/immunology , Lymphocyte Activation/immunology , Receptors, Antigen, T-Cell/genetics , T-Lymphocytes, Cytotoxic/immunology , Viral Nonstructural Proteins/immunology , Apoptosis/genetics , Apoptosis/immunology , Cell Line , Cell Proliferation , Coculture Techniques , Cytotoxicity, Immunologic/immunology , Gene Transfer Techniques , Hepacivirus/genetics , Hepatitis C, Chronic/virology , Humans , RNA, Messenger/genetics , RNA, Viral/biosynthesis , RNA, Viral/genetics , Receptors, Antigen, T-Cell/biosynthesis , Viral Nonstructural Proteins/genetics
8.
J Pediatr Gastroenterol Nutr ; 66(1): 106-111, 2018 01.
Article in English | MEDLINE | ID: mdl-28953534

ABSTRACT

OBJECTIVE: Vertical transmission of hepatitis C virus (HCV) infection is uncommon and occurs in approximately 5% of births from HCV-infected mothers. The reason for the low transmission rate is unclear. We aimed to investigate whether there is evidence of HCV exposure also in the noninfected children born to HCV-infected mothers by the presence of a detectable immune response. METHODS: Serum and peripheral blood mononuclear cells from 9 HCV vertically infected children, 32 uninfected children born to HCV infected mothers, and 15 HCV chronically infected mothers, were analyzed. HCV-RNA-negative adults and children were used as controls. HCV-specific T cell responses were analyzed by interferon gamma using an enzyme-linked immunospot assay and 3H-thymidine incorporation assay. HCV antibodies were also analyzed. RESULTS: An HCV-specific T cell response was detected in 73% (11/15) of the HCV-infected mothers, 67% (6/9) of the vertically infected children, 56% (18/32) of the exposed but uninfected children and in 10% and 20% of the control groups, respectively. The 2 groups of HCV-exposed children both had a significantly higher proportion of HCV-specific T cell responders compared to pediatric controls (P = 0.01 and P = 0.02). CONCLUSIONS: HCV-specific immune responses were more common in children born to HCV-infected mothers, regardless of the presence of HCV RNA. We conclude that noninfected children born to HCV-infected mothers may have been exposed to HCV antigens.


Subject(s)
Hepatitis C Antibodies/blood , Hepatitis C Antigens/immunology , Hepatitis C, Chronic/transmission , Infectious Disease Transmission, Vertical , Pregnancy Complications, Infectious/immunology , Prenatal Exposure Delayed Effects/immunology , Biomarkers/blood , Child, Preschool , Cross-Sectional Studies , Enzyme-Linked Immunosorbent Assay , Female , Follow-Up Studies , Hepatitis C, Chronic/diagnosis , Hepatitis C, Chronic/immunology , Humans , Infant , Infant, Newborn , Male , Pregnancy , Pregnancy Complications, Infectious/virology , Prenatal Exposure Delayed Effects/virology , Prospective Studies , T-Lymphocytes/immunology
9.
J Pediatr Gastroenterol Nutr ; 65(6): 608-612, 2017 12.
Article in English | MEDLINE | ID: mdl-28820758

ABSTRACT

OBJECTIVES: Single genetic nucleotide polymorphism (rs12979860) near the gene for interleukin 28B (IL28B) is known to be of importance for frequency of spontaneous clearance and treatment outcome in interferon-based therapies in patients with hepatitis C virus (HCV) infection. The aim of the present study was to investigate whether IL28B polymorphism in children and/or their mothers plays a role in vertical transmission of HCV (HCV-VT). METHODS: Plasma samples from 59 infected women, 76 uninfected children born to infected mothers, and 47 children with known vertically transmitted HCV infection, were analysed for IL28B polymorphism and classified by the IL28B genotype (C/C, C/T, and T/T) and by viral genotype. RESULTS: The proportion of children with genotype C/C was the same in the vertically infected (36%, 17/47) and the exposed uninfected children (38%, 29/76). No difference was seen when stratifying for viral genotype. There was no association between mothers' IL28B genotype and the risk of vertical transmission. CONCLUSIONS: Regardless of viral genotype we found no association between IL28B genotype and the risk of HCV-VT. The IL28B genotype CC, which has been shown to be favourable in other settings, was not protective of HCV-VT. Thus, other factors possibly associated with the risk of HCV-VT need to be explored.


Subject(s)
Hepacivirus , Hepatitis C/transmission , Infectious Disease Transmission, Vertical , Interleukins/genetics , Polymorphism, Single Nucleotide/genetics , Adolescent , Adult , Case-Control Studies , Child , Child, Preschool , Female , Hepatitis C/epidemiology , Hepatitis C/genetics , Humans , Infant , Infant, Newborn , Interferons , Interleukins/blood , Pregnancy , Pregnancy Complications, Infectious , Prospective Studies , Risk Factors , Young Adult
11.
Mol Ther ; 23(3): 578-90, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25492563

ABSTRACT

Current therapies for the hepatitis B virus (HBV), a major cause of severe liver disease, suppress viral replication but replication rebounds if therapy is withdrawn. It is widely accepted that immune activation is needed to control replication off-therapy. To specifically activate T cells crossreactive between the hepatitis B core and e antigens (HBcAg/HBeAg) in chronically infected patients, we developed a therapeutic vaccine candidate. The vaccine encompass codon-optimized HBcAg and IL-12 expressing plasmids delivered using targeted high-pressure injection combined with in vivo electroporation. One dose of the vaccine primed a B-cell-independent polyfunctional T-cell response, in wild-type, and in HBeAg-transgenic mice with an impaired ability to respond to HBc/eAg. The response peaked at 2 weeks and contracted at week 6 after vaccination. Coadministration of IL-12 improved antibody levels, and T-cell expansion and functionality. The vaccine primed T cells that, 2 weeks after a single dose, cleared hepatocytes transiently expressing HBcAg in vaccinated wild-type and HBeAg-transgenic mice. However, 4 weeks later, these functional responses were lost. Booster doses after 8-12 weeks effectively restored function and expansion of the rapidly contracting T cells. Thus, this vaccine strategy primes functional HBcAg-specific T cells in a host with dysfunctional response to HBV.


Subject(s)
Hepatitis B Antibodies/biosynthesis , Hepatitis B Vaccines/immunology , Hepatitis B virus/immunology , Hepatitis B, Chronic/prevention & control , T-Lymphocytes/immunology , Vaccines, DNA/immunology , Animals , Cell Proliferation , Electroporation , Gene Expression , Hepatitis B Core Antigens/genetics , Hepatitis B Core Antigens/immunology , Hepatitis B Vaccines/administration & dosage , Hepatitis B Vaccines/genetics , Hepatitis B e Antigens/genetics , Hepatitis B e Antigens/immunology , Hepatitis B virus/genetics , Hepatitis B, Chronic/genetics , Hepatitis B, Chronic/immunology , Hepatitis B, Chronic/virology , Hepatocytes/immunology , Hepatocytes/virology , Humans , Interleukin-12/genetics , Interleukin-12/immunology , Liver/immunology , Liver/virology , Mice , Mice, Transgenic , Plasmids/chemistry , Plasmids/metabolism , T-Lymphocytes/virology , Vaccination , Vaccines, DNA/administration & dosage , Vaccines, DNA/genetics
12.
Med Microbiol Immunol ; 204(1): 131-5, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25535102

ABSTRACT

DNA vaccination has historically failed to raise strong immune responses in humans. Recent delivery techniques such as the gene gun and in vivo electroporation (EP)/electrotransfer (ET) have completely changed the efficiency of DNA vaccines in humans. In vivo EP exerts multiple effects that contribute to its efficiency. The two central factors are most likely the increased DNA uptake due to the transient membrane destabilization, and the local tissue damage acting as an adjuvant. To date, several studies in humans have used in vivo EP/ET to deliver DNA. Some of these results have been quite promising with strong T cell responses and/or transient effects on the viral replication. This suggests that improved strategies of in vivo EP/ET can be a future way to deliver DNA in humans.


Subject(s)
Electroporation/methods , Vaccination/methods , Vaccines, DNA/administration & dosage , Humans , T-Lymphocytes/immunology , Vaccines, DNA/immunology
13.
J Immunol ; 190(3): 1113-24, 2013 Feb 01.
Article in English | MEDLINE | ID: mdl-23284053

ABSTRACT

The hepatitis C virus (HCV) nonstructural (NS) 5A protein has been shown to promote viral persistence by interfering with both innate and adaptive immunity. At the same time, the HCV NS5A protein has been suggested as a target for antiviral therapy. In this study, we performed a detailed characterization of HCV NS5A immunogenicity in wild-type (wt) and immune tolerant HCV NS5A-transgenic (Tg) C57BL/6J mice. We evaluated how efficiently HCV NS5A-based genetic vaccines could activate strong T cell responses. Truncated and full-length wt and synthetic codon-optimized NS5A genotype 1b genes were cloned into eukaryotic expression plasmids, and the immunogenicity was determined after i.m. immunization in combination with in vivo electroporation. The NS5A-based genetic vaccines primed high Ab levels, with IgG titers of >10(4) postimmunization. With respect to CD8(+) T cell responses, the coNS5A gene primed more potent IFN-γ-producing and lytic cytotoxic T cells in wt mice compared with NS5A-Tg mice. In addition, high frequencies of NS5A-specific CD8(+) T cells were found in wt mice after a single immunization. To test the functionality of the CTL responses, the ability to inhibit growth of NS5A-expressing tumor cells in vivo was analyzed after immunization. A single dose of coNS5A primed tumor-inhibiting responses in both wt and NS5A-Tg mice. Finally, immunization with the coNS5A gene primed polyfunctional NS5A-specific CD8(+) T cell responses. Thus, the coNS5A gene is a promising therapeutic vaccine candidate for chronic HCV infections.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , DNA, Viral/immunology , Hepacivirus/immunology , Vaccines, DNA/immunology , Viral Hepatitis Vaccines/immunology , Viral Nonstructural Proteins/immunology , Animals , Antibody Specificity , Antigens, Neoplasm/immunology , CD8-Positive T-Lymphocytes/metabolism , Cancer Vaccines , Codon/genetics , Cytotoxicity, Immunologic , DNA, Viral/chemical synthesis , DNA, Viral/genetics , Genes, Synthetic , H-2 Antigens/immunology , Hepacivirus/genetics , Hepatitis C Antibodies/biosynthesis , Hepatitis C Antibodies/genetics , Hepatitis C Antibodies/immunology , Immunization , Immunoglobulin G/biosynthesis , Immunoglobulin G/genetics , Immunoglobulin G/immunology , Lymphocyte Activation , Lymphokines/metabolism , Lymphoma, Non-Hodgkin/immunology , Lymphoma, Non-Hodgkin/therapy , Mice , Mice, Inbred C57BL , Mice, Transgenic , Peptide Fragments/genetics , Peptide Fragments/immunology , T-Cell Antigen Receptor Specificity , T-Lymphocytes, Cytotoxic/immunology , Viral Hepatitis Vaccines/therapeutic use , Viral Nonstructural Proteins/chemistry , Viral Nonstructural Proteins/genetics
14.
J Immunol ; 189(9): 4510-9, 2012 Nov 01.
Article in English | MEDLINE | ID: mdl-23024278

ABSTRACT

Virus-specific CTL with high levels of functional avidity have been associated with viral clearance in hepatitis C virus (HCV) infection and with enhanced protective immunity. In chronic HCV infection, lack of antiviral CTL is frequently observed. In this study, we aim to investigate novel HCV TCRs that differ in Ag specificity. This involved isolating new HCV-specific murine TCRs that recognize a conserved HLA-A2-restricted CTL epitope within the nonstructural protein (NS) 5A viral protein and comparing them with TCRs recognizing another conserved CTL target in the NS3 viral protein. This was done by expressing the TCRs in human T cells and analyzing the function of the resulting TCR-transduced T cells. Our result indicates that these TCRs are efficiently assembled in transduced human T cells. They recognize peptide-loaded targets and demonstrate polyfunctional features such as IL-2, IFN-γ, and TNF-α secretion. However, in contrast to NS3-specific TCRs, the NS5A TCR-transduced T cells consist of a smaller proportion of polyfunctional T cells and require more peptide ligands to trigger the effector functions, including degranulation. Despite the differences, NS5A TCRs show effective inhibition of HCV replication in human hepatoma cells with persistent HCV RNA replication. Moreover, cellular injury demonstrated by aspartate aminotransferase release and cell death is less significant in the hepatoma cells following coincubation with NS5A TCR-transduced T cells, which is a property consistent with noncytotoxic antiviral CTLs. Our results suggest that HCV TCR-transduced T cells may be promising for the treatment of patients with chronic HCV infections.


Subject(s)
Antiviral Agents/pharmacology , Cytotoxicity, Immunologic , Epitopes, T-Lymphocyte/physiology , Hepacivirus/immunology , Hepacivirus/pathogenicity , Receptors, Antigen, T-Cell/physiology , Virus Replication/immunology , Amino Acid Sequence , Animals , Antiviral Agents/toxicity , Cell Line , Cell Line, Tumor , Cytotoxicity, Immunologic/genetics , Epitopes, T-Lymphocyte/toxicity , Female , Gene Transfer Techniques , Humans , Leukocytes, Mononuclear/immunology , Male , Mice , Mice, Transgenic , Molecular Sequence Data , Receptors, Antigen, T-Cell/genetics , Transduction, Genetic/methods , Virus Replication/genetics
15.
Mol Ther ; 21(9): 1796-805, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23752314

ABSTRACT

Clearance of infections caused by the hepatitis C virus (HCV) correlates with HCV-specific T cell function. We therefore evaluated therapeutic vaccination in 12 patients with chronic HCV infection. Eight patients also underwent a subsequent standard-of-care (SOC) therapy with pegylated interferon (IFN) and ribavirin. The phase I/IIa clinical trial was performed in treatment naive HCV genotype 1 patients, receiving four monthly vaccinations in the deltoid muscles with 167, 500, or 1,500 µg codon-optimized HCV nonstructural (NS) 3/4A-expressing DNA vaccine delivered by in vivo electroporation (EP). Enrollment was done with 2 weeks interval between patients for safety reasons. Treatment was safe and well tolerated. The vaccinations significantly improved IFN-γ-producing responses to HCV NS3 during the first 6 weeks of therapy. Five patients experienced 2-10 weeks 0.6-2.4 log10 reduction in serum HCV RNA. Six out of eight patients starting SOC therapy within 1-30 months after the last vaccine dose were cured. This first-in-man therapeutic HCV DNA vaccine study with the vaccine delivered by in vivo EP shows transient effects in patients with chronic HCV genotype 1 infection. The interesting result noted after SOC therapy suggests that therapeutic vaccination can be explored in a combination with SOC treatment.


Subject(s)
Antiviral Agents/therapeutic use , Hepatitis C, Chronic/therapy , Interferon-alpha/therapeutic use , Polyethylene Glycols/therapeutic use , Ribavirin/therapeutic use , Vaccines, DNA/therapeutic use , Viral Hepatitis Vaccines/therapeutic use , Adult , Antiviral Agents/administration & dosage , Antiviral Agents/adverse effects , Combined Modality Therapy , Electroporation , Female , Hepacivirus/genetics , Hepacivirus/immunology , Hepacivirus/physiology , Hepatitis C, Chronic/genetics , Hepatitis C, Chronic/virology , Humans , Interferon-alpha/administration & dosage , Interferon-alpha/adverse effects , Interferons , Interleukins/genetics , Lymphocyte Activation , Male , Middle Aged , Polyethylene Glycols/administration & dosage , Polyethylene Glycols/adverse effects , RNA, Viral/blood , Recombinant Proteins/administration & dosage , Recombinant Proteins/adverse effects , Recombinant Proteins/therapeutic use , Ribavirin/administration & dosage , Ribavirin/adverse effects , Standard of Care , T-Lymphocytes/immunology , Vaccines, DNA/administration & dosage , Vaccines, DNA/adverse effects , Vaccines, DNA/immunology , Viral Hepatitis Vaccines/administration & dosage , Viral Hepatitis Vaccines/adverse effects , Viral Load
16.
Drug Resist Updat ; 16(3-5): 60-7, 2013.
Article in English | MEDLINE | ID: mdl-23911647

ABSTRACT

Presently the development of new therapies for hepatitis C virus (HCV) is rapidly moving forward. Almost every week new data appear on how direct acting antivirals (DAAs) succeed or fail in clinical trials. Despite the potency of many of the DAA combinations, the effect exerted by ribavirin (RBV) is still needed for an effective therapy in many new DAA combinations. Due to the strong antiviral effect of DAAs, it is likely that a major complementary therapeutic effect exerted by RBV is immune modulation resulting in an increased barrier to development of resistance. For HCV genotype 1a infections elimination of pegylated interferon, is not possible in many DAA combinations without jeopardizing the results. The host immune response is thus likely to play a key role even during DAA-based therapies. Hence, T cells may recognize and eliminate viral variants with resistance to the DAAs. We herein show several examples where this may be the case, supporting the rationale of including the host response also in the new therapeutic regimens. This review will describe the potential benefits of combining various DAAs with means to activate the specific immune response against HCV.


Subject(s)
Antiviral Agents/therapeutic use , Hepacivirus/drug effects , Hepatitis C, Chronic/therapy , Immunomodulation/drug effects , Models, Biological , Animals , Antibodies, Neutralizing/therapeutic use , Antibodies, Viral/therapeutic use , Combined Modality Therapy , Drug Resistance, Viral , Hepacivirus/immunology , Hepacivirus/metabolism , Hepatitis C, Chronic/immunology , Hepatitis C, Chronic/metabolism , Hepatitis C, Chronic/virology , Humans , Ligands , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Programmed Cell Death 1 Receptor/metabolism , Ribavirin/therapeutic use , Toll-Like Receptors/agonists , Toll-Like Receptors/metabolism , Viral Hepatitis Vaccines/therapeutic use
17.
J Infect Dis ; 208(6): 1008-19, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23776192

ABSTRACT

BACKGROUND: We explored the concept of heterologous prime/boost vaccination using 2 therapeutic vaccines currently in clinical development aimed at treating chronically infected hepatitis C virus (HCV) patients: prime with a DNA-based vaccine expressing HCV genotype 1a NS3/4A proteins (ChronVac-C) and boost with a modified vaccinia virus Ankara vaccine expressing genotype 1b NS3/4/5B proteins (MVATG16643). METHODS: Two ChronVac-C immunizations 4 weeks apart were delivered intramuscularly in combination with in vivo electroporation and subsequently 5 or 12 weeks later boosted by 3 weekly subcutaneous injections of MVATG16643. Two mouse strains were used, and we evaluated quality, magnitude, and functionality of the T cells induced. RESULTS: DNA prime/MVA boost regimen induced significantly higher levels of interferon γ (IFN-γ) or interleukin 2 (IL-2) ELISpot responses compared with each vaccine alone, independent of the time of analysis and the time interval between vaccinations. Both CD8⁺ and CD4⁺ T-cell responses as well as the spectrum of epitopes recognized was improved. A significant increase in polyfunctional IFN-γ/tumor necrosis factor α (TNF-α)/CD107a⁺ CD8⁺ T cells was detected following ChronVac-C/MVATG16643 vaccination (from 3% to 25%), and prime/boost was the only regimen that activated quadrifunctional T cells (IFN-γ/TNF-α/CD107a/IL-2). In vivo functional protective capacity of DNA prime/MVA boost was demonstrated in a Listeria-NS3-1a challenge model. CONCLUSIONS: We provide a proof-of-concept that immunogenicity of 2 HCV therapeutic vaccines can be improved using their combination, which merits further clinical development.


Subject(s)
Antibody Formation , Hepatitis C/prevention & control , Vaccination/methods , Viral Hepatitis Vaccines/immunology , Animals , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Genotype , Hepacivirus , Hepatitis C/immunology , Immunization, Secondary , Interferon-gamma/blood , Interleukin-2/blood , Mice , Mice, Inbred C57BL , Mice, Transgenic , Tumor Necrosis Factor-alpha/blood , Vaccines, DNA/immunology , Viral Hepatitis Vaccines/genetics
18.
Biochem Biophys Res Commun ; 433(1): 31-5, 2013 Mar 29.
Article in English | MEDLINE | ID: mdl-23454379

ABSTRACT

The hepatitis C virus (HCV) non-structural 3 (NS3) protein plays key roles in both the viral life cycle and in the modulation of intrahepatic signaling and immunity. We recently showed that NS3 cleaves the T cell protein tyrosine phosphatase (TCPTP). To better understand the inactivation of TCPTP in HCV-infected humans, we investigated whether there is an association between TCPTP cleavage, NS3 protein levels and clinical parameters in hepatitis C patients. Liver biopsies were obtained from 69 HCV RNA positive patients with confirmed chronic HCV infection and 16 control patients. Hepatic NS3 and TCPTP protein levels were determined and correlated to viral load or clinical parameters for the severity of liver disease. We found a positive correlation between the viral load and the intrahepatic NS3 protein levels in patients infected with HCV. HCV-infected patients had significantly lower intrahepatic TCPTP levels than non-infected control patients. In HCV-infected patients both intrahepatic NS3 expression and the viral load were inversely correlated with the intrahepatic TCPTP protein levels. Detection of NS3 did not associate with any other clinical parameters such as liver damage, the grade of liver inflammation or fibrosis stage. This is the first study reporting a detailed analysis of HCV NS3 and TCPTP protein levels in the liver. It demonstrates a clear link between HCV viral load, NS3 expression in the liver and intrahepatic TCPTP levels. Thus, the association between TCPTP cleavage and viral replication may have important consequences for the HCV life cycle and HCV-induced liver diseases.


Subject(s)
Hepatitis C, Chronic/metabolism , Hepatitis C, Chronic/virology , Protein Tyrosine Phosphatase, Non-Receptor Type 2/metabolism , RNA, Viral/metabolism , Viral Nonstructural Proteins/metabolism , Adult , Female , Hepacivirus/genetics , Hepacivirus/growth & development , Hepacivirus/physiology , Hepatitis C, Chronic/genetics , Humans , Liver/metabolism , Liver/virology , Male , Middle Aged , RNA, Viral/genetics , Viral Load , Viral Nonstructural Proteins/genetics
19.
J Immunol ; 186(9): 5107-18, 2011 May 01.
Article in English | MEDLINE | ID: mdl-21430225

ABSTRACT

The hepatitis C virus (HCV)-specific T cell response in patients with chronic HCV is dysfunctional. In this study, we aimed at restoring immunological function through therapeutic vaccination in a transgenic mouse model with impaired HCV-specific T cell responses due to a persistent presence of hepatic HCV nonstructural (NS)3/4A Ags. The HCV-specific T cells have an actively maintained dysfunction reflected in reduced frequency, impaired cytokine production, and impaired effector function in vivo, which can be partially restored by blocking regulatory T cells or programmed cell death ligand 1. We hypothesized that the impairment could be corrected by including sequences that created a normal priming environment by recruiting "healthy" heterologous T cells and by activating innate signaling. Endogenously expressed hepatitis B core Ag (HBcAg) can recruit heterologous T cells and activate TLR (TLR7) signaling. Hence, by combining HCV NS3/4A with different forms of HBcAg we found that heterologous sequences somewhat improved activation and expansion of NS3/4A-specific T cells in a wild-type host. Importantly, the signals provided by HBcAg effectively restored the activation of HCV-specific T cells in a tolerant NS3/4A-transgenic mouse model. The adjuvant effect could also be transferred to the priming of dysfunctional HLA-A2-restricted NS3-specific T cells in vivo. Thus, recruiting healthy heterologous T cells to the site of priming may also help restore HCV-specific responses present in a chronically infected host.


Subject(s)
Hepatitis C, Chronic/immunology , T-Lymphocytes/immunology , Vaccination/methods , Viral Hepatitis Vaccines/immunology , Viral Nonstructural Proteins/immunology , Animals , Animals, Genetically Modified , Antigens, Viral/immunology , Antigens, Viral/therapeutic use , Blotting, Western , Cell Line, Tumor , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Hepacivirus/immunology , Hepatitis B Core Antigens/immunology , Hepatitis B Core Antigens/therapeutic use , Hepatitis C, Chronic/therapy , Humans , Immunoprecipitation , Lymphocyte Activation/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Transgenic
20.
Gut ; 61(4): 589-96, 2012 Apr.
Article in English | MEDLINE | ID: mdl-21813471

ABSTRACT

BACKGROUND: The non-structural (NS) 3/4A protease/helicase of the hepatitis C virus is known to modulate signalling pathways in the infected hepatocyte by cleaving CARD adaptor inducing IFNß (Cardif), T-cell protein tyrosine phosphatase (TC-PTP) and TIR domain-containing adaptor inducing IFNß (TRIF), but the effects of NS3/4A in vivo still remain unclear. AIM: To investigate the influence of NS3/4A on intracellular and intercellular signalling in vivo by analysing the intrahepatic inflammatory response of naïve, lipopolysaccharide (LPS)/D-galactosamine (D-galN) or tumour necrosis factor-α (TNFα)/D-galN-treated NS3/4A-transgenic (Tg) mice. METHODS: The intrahepatic immunity of naïve and LPS/D-galN- or TNFα/D-galN-treated NS3/4A-Tg mice was determined using western blot, ELISA, real-time PCR, flow cytometry and survival monitoring. The injection of cytokines or antibodies against signalling components was performed to analyse the relevance of the respective pathways for the investigated issues. A Tg mouse lineage expressing an inactivated NS3/4A protease (NS3/4A(Ile1073Ala)-Tgs) was generated to examine if protective effects were NS3/4A protease dependent. RESULTS: The activation of hepatic signal transducer and activator of transcription 1 and 2 was impaired in NS3/4A-Tg mice after treatment with LPS/D-galN or TNFα/D-galN. This was paralleled by a reduction in hepatic interferon-γ (IFNγ). Reconstitution of IFNγ reverted the resistance to LPS/TNFα in NS3/4A-Tg mice. Subsequently, blocking IFNγ in vivo rendered wild-type mice resistant against treatment with LPS/TNFα. A new Tg mouse expressing an inactivated NS3/4A protease had the same phenotype as wild-type mice with respect to hepatic IFNγ levels and sensitivity to LPS/d-galN. Finally, the chemokine profile was altered in the NS3/4A-Tg mice towards an anti-inflammatory state, which helps to explain the altered immune cell subsets and reduction in hepatic IFNγ production. CONCLUSIONS: Our data demonstrate that the NS3/4A protease reduces the intrahepatic production of IFNγ and alters TNFα-mediated effects, thereby impairing the hepatic inflammatory response. This may contribute to viral persistence.


Subject(s)
Carrier Proteins/physiology , Hepacivirus/metabolism , Interferon-gamma/biosynthesis , Liver/immunology , Viral Nonstructural Proteins/physiology , Animals , Case-Control Studies , Female , Hepatitis C/immunology , Humans , Interferon-alpha/metabolism , Interferon-gamma/pharmacology , Intracellular Signaling Peptides and Proteins , Lipopolysaccharides/pharmacology , Lymphocyte Subsets/immunology , Male , Mice , Mice, Transgenic , Middle Aged , STAT1 Transcription Factor/metabolism , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL