Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
1.
J Heart Lung Transplant ; 41(10): 1429-1439, 2022 10.
Article in English | MEDLINE | ID: mdl-35953352

ABSTRACT

BACKGROUND: Previous studies have reported that lung transplant recipients (LTR) develop a poor response to two doses of COVID-19 vaccine, but data regarding the third dose are lacking. We investigated the antibody response after three doses of mRNA vaccine in LTR and its predictive factors. METHODS: A total of 136 LTR, including 10 LTR previously infected and 126 COVID-19-naive LTR, were followed during and after three doses of mRNA vaccine. We retrospectively measured anti-receptor-binding domain (RBD) IgG response and neutralizing antibodies. In a posthoc analysis, we used a multivariate logistic regression model to assess the association between vaccine response and patient characteristics, including viral DNA load (VL) of the ubiquitous Torque teno virus (TTV) (optimal cut-off set by ROC curve analysis), which reflects the overall immunosuppression. RESULTS: After 3 doses, 47/126 (37.3%) COVID-19-naive LTR had positive anti-RBD IgG (responders) and 14/126 (11.1%) had antibody titers above 264 Binding Antibody Units/mL. None neutralized the omicron variant versus 7 of the 10 previously infected LTR. Nonresponse was associated with TTV VL ≥6.2 log10 copies/mL before vaccination (Odds Ratio (OR) = 17.87, 95% confidence interval (CI95) = 3.02-105.72), mycophenolate treatment (OR = 4.73, CI95 = 1.46-15.34) and BNT162b2 (n = 34; vs mRNA-1273, n = 101) vaccine (OR = 6.72, CI95 = 1.75-25.92). In second dose non-responders, TTV VL ≥6.2 or <3.2 log10 copies/mL before the third dose was associated with low (0/19) and high (9/10) rates of seroconversion. CONCLUSION: COVID-19-naive LTR respond poorly to three doses of mRNA vaccine, especially those with high TTV VL. Future studies could further evaluate this biomarker as a guide for vaccine strategies.


Subject(s)
COVID-19 , Torque teno virus , Antibodies, Neutralizing , Antibodies, Viral , Antibody Formation , BNT162 Vaccine , Biomarkers , COVID-19/prevention & control , COVID-19 Vaccines , DNA, Viral , Humans , Immunoglobulin G , Lung , RNA, Messenger , Retrospective Studies , SARS-CoV-2 , Torque teno virus/genetics , Transplant Recipients , Vaccines, Synthetic , mRNA Vaccines
2.
Front Immunol ; 13: 790212, 2022.
Article in English | MEDLINE | ID: mdl-35281046

ABSTRACT

Background: SARS-CoV-2 breakthrough infections after complete vaccination are increasing whereas their determinants remain uncharacterized. Methods: We analyzed two cases of post-vaccination SARS-CoV-2 infections by α and ß variants, respectively. For each participant both humoral (binding and neutralizing antibodies) and cellular (activation markers and cytokine expression) immune responses were characterized longitudinally. Results: The first participant (P1) was infected by an α variant and displayed an extended and short period of viral excretion and symptom. Analysis of cellular and humoral response 72 h post-symptom onset revealed that P1 failed at developing neutralizing antibodies and a potent CD4 memory response (lack of SARS-CoV-2 specific CD4+IL-2+ cells) and CD8 effector response (CD8+IFNγ+ cells). The second participant (P2) developed post-vaccination SARS-CoV-2 infection by a ß variant, associated with a short period of viral excretion and symptoms. Despite displaying initially high levels and polyfunctional T cell responses, P2 lacked initial ß-directed neutralizing antibodies. Both participants developed and/or increased their neutralization activity and cellular responses against all variants, namely, ß and δ variants that lasts up to 3 months after breakthrough infection. Conclusions: An analysis of cellular and humoral response suggests two possible mechanisms of breakthrough infection: a poor immune response to vaccine and viral evasion to neutralizing antibodies.


Subject(s)
BNT162 Vaccine/immunology , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , COVID-19/diagnosis , SARS-CoV-2/physiology , Adult , Antibodies, Neutralizing/blood , Antibodies, Viral/blood , COVID-19 Vaccines , Female , Humans , Immune Evasion , Middle Aged , Vaccination
3.
Sci Transl Med ; 14(636): eabl6141, 2022 03 16.
Article in English | MEDLINE | ID: mdl-35103481

ABSTRACT

Transplant recipients, who receive therapeutic immunosuppression to prevent graft rejection, are characterized by high coronavirus disease 2019 (COVID-19)-related mortality and defective response to vaccines. We observed that previous infection with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), but not the standard two-dose regimen of vaccination, provided protection against symptomatic COVID-19 in kidney transplant recipients. We therefore compared the cellular and humoral immune responses of these two groups of patients. Neutralizing anti-receptor-binding domain (RBD) immunoglobulin G (IgG) antibodies were identified as the primary correlate of protection for transplant recipients. Analysis of virus-specific B and T cell responses suggested that the generation of neutralizing anti-RBD IgG may have depended on cognate T-B cell interactions that took place in germinal center, potentially acting as a limiting checkpoint. High-dose mycophenolate mofetil, an immunosuppressive drug, was associated with fewer antigen-specific B and T follicular helper (TFH) cells after vaccination; this was not observed in patients recently infected with SARS-CoV-2. Last, we observed that, in two independent prospective cohorts, administration of a third dose of SARS-CoV-2 mRNA vaccine restored neutralizing titers of anti-RBD IgG in about 40% of individuals who had not previously responded to two doses of vaccine. Together, these findings suggest that a third dose of SARS-CoV-2 mRNA vaccine improves the RBD-specific responses of transplant patients treated with immunosuppressive drugs.


Subject(s)
COVID-19 , Kidney Transplantation , Antibodies, Neutralizing , Antibodies, Viral , BNT162 Vaccine , COVID-19/prevention & control , COVID-19 Vaccines , Humans , Prospective Studies , SARS-CoV-2 , Transplant Recipients , Vaccines, Synthetic , mRNA Vaccines
4.
Am J Transplant ; 21(1): 329-337, 2021 01.
Article in English | MEDLINE | ID: mdl-32741096

ABSTRACT

BK virus (BKV) replication occurs frequently in kidney transplant recipients (KTR), potentially leading to BKV-associated nephropathy (BKVAN) and graft loss. Patients with high titers of BKV-neutralizing antibodies (NAbs) are protected against BKV replication, and intravenous immunoglobulin (IVIg) infusion can increase NAb titers. We investigated whether early IVIg administration prevents BKV replication in patients with low NAb titers (<4 log10 against the BKV-specific genotype). Based on NAb titers on the day of transplantation, KTR followed in the Strasbourg University Hospital (n = 174) were retrospectively divided into the following 3 risk categories for BKV replication: (1) patients with low NAb titers ("high-risk") who received IVIg for the first 3 posttransplant months (n = 44), (2) patients with low NAb titers ("high-risk") who did not undergo IVIg treatment (n = 41), and (3) patients with high NAb titers ("low-risk") who did not receive IVIg (n = 89). At 12 posttransplant months, the incidence of BKV viremia in the high-risk group treated with IVIg (6.8%) was similar to that observed in the low-risk group (10.1%) and markedly lower than that of the untreated high-risk group (36.6%; P < .001). Similar results were observed with regard to BKVAN. We conclude that IVIg may be a valuable strategy for preventing BKV replication.


Subject(s)
BK Virus , Kidney Transplantation , Polyomavirus Infections , Tumor Virus Infections , Humans , Immunoglobulins, Intravenous , Kidney Transplantation/adverse effects , Polyomavirus Infections/drug therapy , Polyomavirus Infections/prevention & control , Retrospective Studies , Tumor Virus Infections/prevention & control , Viremia/drug therapy , Viremia/etiology , Viremia/prevention & control
5.
Viruses ; 12(12)2020 12 02.
Article in English | MEDLINE | ID: mdl-33276614

ABSTRACT

JC virus (JCV) causes progressive multifocal leukoencephalopathy (PML) in immunosuppressed patients. There is currently no effective specific antiviral treatment and PML management relies on immune restoration. Prognosis markers are crucially needed in this disease because of its high mortality rate. In this work, we investigated the compartmentalization of JCV strains as well as the humoral neutralizing response in various matrices to further understand the pathophysiology of PML and define markers of survival. Four patients were included, of which three died in the few months following PML onset. Cerebrospinal fluid (CSF) viral loads were the highest, with plasma samples having lower viral loads and urine samples being mostly negative. Whether at PML onset or during follow-up, neutralizing antibody (NAb) titers directed against the same autologous strain (genotype or mutant) were the highest in plasma, with CSF titers being on average 430-fold lower and urine titers 500-fold lower at the same timepoint. Plasma NAb titers against autologous genotype or mutant were lower in non-survivor patients, though no neutralization "blind spot" was observed. The surviving patient was followed up until nine months after PML onset and presented, at that time, an increase in neutralizing titers, from 38-fold against the autologous genotype to around 200-fold against PML mutants. Our results suggest that patients' humoral neutralizing response against their autologous strain may play a role in PML outcome, with survivors developing high NAb titers in both plasma and CSF.


Subject(s)
Host-Pathogen Interactions/immunology , Immunity, Humoral , JC Virus/immunology , Leukoencephalopathy, Progressive Multifocal/immunology , Leukoencephalopathy, Progressive Multifocal/pathology , Antibodies, Neutralizing/blood , Antibodies, Neutralizing/immunology , Antibodies, Viral/blood , Antibodies, Viral/immunology , Genotype , Humans , JC Virus/genetics , Leukoencephalopathy, Progressive Multifocal/mortality , Leukoencephalopathy, Progressive Multifocal/virology , Mutation , Neutralization Tests , Viral Load
6.
Crit Rev Microbiol ; 45(4): 472-493, 2019 Aug.
Article in English | MEDLINE | ID: mdl-31267816

ABSTRACT

Tick-borne encephalitis virus (TBEV) is a zoonotic agent causing severe encephalitis. The transmission cycle involves the virus, the Ixodes tick vector, and a vertebrate reservoir, such as small mammals (rodents, or shrews). Humans are accidentally involved in this transmission cycle. Tick-borne encephalitis (TBE) has been a growing public health problem in Europe and Asia over the past 30 years. The mechanisms involved in the development of TBE are very complex and likely multifactorial, involving both host and viral factors. The purpose of this review is to provide an overview of the current literature on TBE neuropathogenesis in the human host and to demonstrate the emergence of common themes in the molecular pathogenesis of TBE in humans. We discuss and review data on experimental study models and on both viral (molecular genetics of TBEV) and host (immune response, and genetic background) factors involved in TBE neuropathogenesis in the context of human infection.


Subject(s)
Encephalitis Viruses, Tick-Borne/genetics , Encephalitis Viruses, Tick-Borne/pathogenicity , Encephalitis, Tick-Borne/pathology , Encephalitis, Tick-Borne/virology , Host-Pathogen Interactions , Virulence Factors/genetics , Virulence Factors/metabolism , Animals , Disease Models, Animal , Humans
7.
Article in English | MEDLINE | ID: mdl-31160292

ABSTRACT

BK polyomavirus (BKPyV)-associated nephropathy (BKPyVAN) is one of the major causes of kidney graft dysfunction, and there are no BKPyV-specific antiviral therapies available. BKPyV neutralizing antibodies (NAbs) play key roles in protecting against BKPyV replication and represent a potential therapeutic or preventive strategy. In this study, we evaluated NAb titers in intravenous immunoglobulin (i.v. Ig) preparations and in kidney transplant recipients (KTR) before and after i.v. Ig administration. NAb titers directed against major BKPyV genotypes were measured using a BKPyV pseudovirion system. Thirty-three KTR receiving high (1 g/kg of body weight/day; n = 17) or low (0.4 g/kg/day; n = 16) i.v. Ig doses were included. Median NAb titers in i.v. Ig preparations ranged from 5.9 log10 50% inhibitory concentration (IC50) for genotype I to 4.1 log10 IC50 for genotype IV. A mean of 90% of patients (range, 88% to 100%) displaying low or negative BKPyV NAb titers against genotype I reached 4 log10 IC50 after the first i.v. Ig administration. This value was reached by a mean of 44% (range, 13% to 83%) and 19% (range, 0% to 38%) of patients against genotype II and genotype IV, respectively. The benefit of i.v. Ig administration persisted until the following course of treatment (day 22 ± 7 days) for genotypes I and II, and no cumulative effect was observed through the three doses. Our findings demonstrate that i.v. Ig administration results in a significant increase in BKPyV NAb titers in KTR. These in vitro and in vivo pharmacokinetic data provide the rationale for a proof-of-concept study investigating the efficacy of i.v. Ig for the prevention of BKPyV infection in KTR.


Subject(s)
Antibodies, Neutralizing/immunology , BK Virus/genetics , Immunoglobulins, Intravenous/pharmacology , Kidney Transplantation , Polyomavirus Infections/prevention & control , Adolescent , Adult , Aged , BK Virus/immunology , Female , Genotype , Humans , Immunoglobulins, Intravenous/adverse effects , Immunologic Deficiency Syndromes/drug therapy , Male , Middle Aged , Polyomavirus Infections/drug therapy , Young Adult
8.
J Am Soc Nephrol ; 29(1): 326-334, 2018 01.
Article in English | MEDLINE | ID: mdl-29042457

ABSTRACT

BK virus-associated nephropathy (BKVAN) causes renal allograft dysfunction. The current management of BKVAN relies on pre-emptive adaptation of immunosuppression according to viral load monitoring. However, this empiric strategy is not always successful. Therefore, pretransplant predictive markers are needed. In a prospective longitudinal study, we enrolled 168 kidney transplant recipients and 69 matched donors. To assess the value of BKV genotype-specific neutralizing antibody (NAb) titers as a predictive marker for BKV replication, we measured BKV DNA load and NAb titers at transplant and followed patients for 24 months. After transplant, 52 (31%) patients displayed BKV replication: 24 (46%) patients were viruric and 28 (54%) patients were viremic, including 13 with biopsy-confirmed BKVAN. At any time, patients with high NAb titers against the replicating strain had a lower risk of developing BKV viremia (hazard ratio [HR], 0.44; 95% confidence interval [95% CI], 0.26 to 0.73; P=0.002). Each log10 increase in NAb titer decreased the risk of developing viremia by 56%. Replicating strains were consistent with donor transmission in 95% of cases of early BKV replication. Genotype mismatch between recipients' neutralization profiles before transplant and their subsequently replicating strain significantly increased the risk of developing viremia (HR, 2.27; 95% CI, 1.06 to 4.88; P=0.04). A NAb titer against the donor's strain <4 log10 before transplant significantly associated with BKV replication after transplant (HR, 1.88; 95% CI, 1.06 to 3.45; P=0.03). BKV genotype-specific NAb titers may be a meaningful predictive marker that allows patient stratification by BKV disease risk before and after transplant.


Subject(s)
Antibodies, Neutralizing/blood , BK Virus/immunology , DNA, Viral/blood , Kidney Diseases/virology , Polyomavirus Infections/complications , Tumor Virus Infections/complications , Adolescent , Adult , Aged , Allografts/physiopathology , Allografts/virology , BK Virus/genetics , BK Virus/physiology , Female , Genotype , Humans , Kidney Diseases/pathology , Kidney Transplantation , Longitudinal Studies , Male , Middle Aged , Prospective Studies , Risk Assessment/methods , Urine/virology , Viral Load , Viremia/virology , Virus Replication , Young Adult
9.
Crit Rev Microbiol ; 43(2): 178-195, 2017 Mar.
Article in English | MEDLINE | ID: mdl-27800693

ABSTRACT

Nearly 45 years after the discovery of the first two human polyomaviruses BK and JC, their life-long persistence and mechanisms of pathogenesis remain poorly understood and efficient antiviral treatments are severely lacking. In this review, we sought to provide an update on recent advances in understanding the life cycle of these two viruses, particularly focusing on their interaction with the host immune system and pathogenesis. We have also discussed novel treatment approaches and highlighted areas of future research.


Subject(s)
BK Virus/isolation & purification , Biomedical Research/trends , JC Virus/isolation & purification , Polyomavirus Infections/pathology , Polyomavirus Infections/therapy , Virology/history , BK Virus/immunology , BK Virus/pathogenicity , History, 20th Century , History, 21st Century , Host-Pathogen Interactions , Humans , JC Virus/immunology , JC Virus/pathogenicity , Virology/trends
10.
Viruses ; 8(10)2016 10 22.
Article in English | MEDLINE | ID: mdl-27782080

ABSTRACT

Developments of genome amplification techniques have rapidly expanded the family of human polyomaviruses (PyV). Following infection early in life, PyV persist in their hosts and are generally of no clinical consequence. High-level replication of PyV can occur in patients under immunosuppressive or immunomodulatory therapy and causes severe clinical entities, such as progressive multifocal leukoencephalopathy, polyomavirus-associated nephropathy or Merkel cell carcinoma. The characterization of known and newly-discovered human PyV, their relationship to human health, and the mechanisms underlying pathogenesis remain to be elucidated. Here, we summarize the most widely-used in vitro and in vivo models to study the PyV-host interaction, pathogenesis and anti-viral drug screening. We discuss the strengths and limitations of the different models and the lessons learned.


Subject(s)
Disease Models, Animal , Host-Pathogen Interactions , Models, Theoretical , Polyomavirus Infections/immunology , Polyomavirus Infections/pathology , Polyomavirus/immunology , Polyomavirus/pathogenicity , Animals , Humans
11.
J Clin Microbiol ; 53(12): 3842-52, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26468499

ABSTRACT

International guidelines define a BK virus (BKV) load of ≥4 log10 copies/ml as presumptive of BKV-associated nephropathy (BKVN) and a cutoff for therapeutic intervention. To investigate whether BKV DNA loads (BKVL) are comparable between laboratories, 2 panels of 15 and 8 clinical specimens (urine, whole blood, and plasma) harboring different BKV genotypes were distributed to 20 and 27 French hospital centers in 2013 and 2014, respectively. Although 68% of the reported results fell within the acceptable range of the expected result ±0.5 log10, the interlaboratory variation ranged from 1.32 to 5.55 log10. Polymorphisms specific to BKV genotypes II and IV, namely, the number and position of mutations in amplification target genes and/or deletion in standards, arose as major sources of interlaboratory disagreements. The diversity of DNA purification methods also contributed to the interlaboratory variability, in particular for urine samples. Our data strongly suggest that (i) commercial external quality controls for BKVL assessment should include all major BKV genotypes to allow a correct evaluation of BKV assays, and (ii) the BKV sequence of commercial standards should be provided to users to verify the absence of mismatches with the primers and probes of their BKV assays. Finally, the optimization of primer and probe design and standardization of DNA extraction methods may substantially decrease interlaboratory variability and allow interinstitutional studies to define a universal cutoff for presumptive BKVN and, ultimately, ensure adequate patient care.


Subject(s)
BK Virus/genetics , BK Virus/isolation & purification , DNA, Viral/genetics , Genetic Variation , Polyomavirus Infections/diagnosis , Viral Load/methods , Viral Load/standards , DNA, Viral/isolation & purification , France , Hospitals , Humans , Laboratory Proficiency Testing , Polyomavirus Infections/virology , Sensitivity and Specificity
12.
J Innate Immun ; 7(5): 530-44, 2015.
Article in English | MEDLINE | ID: mdl-25792183

ABSTRACT

In patients with hepatitis C virus (HCV) infection, enhanced activity of indoleamine-2,3-dioxygenase 1 (IDO) has been reported. IDO - a tryptophan-catabolizing enzyme - has been considered as both an innate defence mechanism and an important regulator of the immune response. The molecular mechanism of IDO induction in HCV infection and its role in the antiviral immune response remain unknown. Using primary human hepatocytes, we show that HCV infection stimulates IDO expression. IDO gene induction was transient and coincided with the expression of types I and III interferons (IFNs) and IFN-stimulated genes in HCV-infected hepatocytes. Overexpression of hepatic IDO prior to HCV infection markedly impaired HCV replication in hepatocytes, suggesting that IDO limits the spread of HCV within the liver. siRNA-mediated IDO knock-down revealed that IDO functions as an IFN-mediated anti-HCV effector. Hepatic IDO was most potently induced by IFN-x03B3;, and ongoing HCV replication could significantly upregulate IDO expression. IRF1 (IFN-regulatory factor 1) and STAT1 (signal transducer and activator of transcription 1) regulated hepatic IDO expression. Hepatic IDO expression also had a significant inhibitory effect on CD4+ T-cell proliferation. Our data suggest that hepatic IDO plays a dual role during HCV infection by slowing down viral replication and also regulating host immune responses.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Hepacivirus/physiology , Hepatitis C/immunology , Hepatocytes/immunology , Indoleamine-Pyrrole 2,3,-Dioxygenase/metabolism , Cell Proliferation/genetics , Cells, Cultured , Hepatocytes/virology , Humans , Immunity, Innate , Immunosuppression Therapy , Interferon Regulatory Factor-1/genetics , Interferon Regulatory Factor-1/metabolism , Interferons/genetics , Interferons/metabolism , RNA, Small Interfering/genetics , STAT1 Transcription Factor/genetics , STAT1 Transcription Factor/metabolism , Virus Replication/genetics
13.
J Clin Microbiol ; 52(12): 4298-304, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25297334

ABSTRACT

Screening of BK virus (BKV) replication is recommended to identify patients at increased risk of BKV-associated diseases. However, the heterogeneity of molecular techniques hinders the establishment of universal guidelines for BKV monitoring. Here we aimed to compare the performance of the CE-marked BK virus R-gene kit (R-gene) to the performance of our in-house assay for quantification of BKV DNA loads (BKVL). A 12-specimen panel from the Quality Control for Molecular Diagnostics (QCMD) organization, 163 urine samples, and 88 paired specimens of plasma and whole blood (WB) from transplant recipients were tested. Both the R-gene and in-house assays showed a good correlation within the QCMD panel (r = 0.995 and r = 0.989, respectively). BKVL were highly correlated between assays, although positive biases were observed with the in-house assay in analysis of urine (0.72 ± 0.83 log10 copies/ml), plasma (1.17 ± 0.63 log10 copies/ml), and WB (1.28 ± 0.37 log10 copies/ml). Recalibration with a common calibrator significantly reduced the bias in comparisons between assays. In contrast, BKVL was underestimated with the in-house PCR in eight samples containing BKV genotype II, presenting point mutations at primer-annealing sites. Using the R-gene assay, plasma and WB specimens were found to be equally suitable for quantification of BKVL, as indicated by the high correlation coefficient (r = 0.965, P < 0.0001). In conclusion, the R-gene assay demonstrated reliable performance and higher accuracy than the in-house assay for quantification of BKVL in urine and blood specimens. Screening of BKV replication by a well-validated commercial kit may enable clinical laboratories to assess viral loads with greater reproducibility and precision.


Subject(s)
BK Virus/isolation & purification , Blood/virology , Polyomavirus Infections/virology , Tumor Virus Infections/virology , Urine/virology , Viral Load/methods , Viral Load/standards , Humans , Reagent Kits, Diagnostic , Reproducibility of Results
14.
PLoS Pathog ; 10(5): e1004128, 2014 May.
Article in English | MEDLINE | ID: mdl-24830295

ABSTRACT

Hepatitis C virus (HCV) is transmitted between hepatocytes via classical cell entry but also uses direct cell-cell transfer to infect neighboring hepatocytes. Viral cell-cell transmission has been shown to play an important role in viral persistence allowing evasion from neutralizing antibodies. In contrast, the role of HCV cell-cell transmission for antiviral resistance is unknown. Aiming to address this question we investigated the phenotype of HCV strains exhibiting resistance to direct-acting antivirals (DAAs) in state-of-the-art model systems for cell-cell transmission and spread. Using HCV genotype 2 as a model virus, we show that cell-cell transmission is the main route of viral spread of DAA-resistant HCV. Cell-cell transmission of DAA-resistant viruses results in viral persistence and thus hampers viral eradication. We also show that blocking cell-cell transmission using host-targeting entry inhibitors (HTEIs) was highly effective in inhibiting viral dissemination of resistant genotype 2 viruses. Combining HTEIs with DAAs prevented antiviral resistance and led to rapid elimination of the virus in cell culture model. In conclusion, our work provides evidence that cell-cell transmission plays an important role in dissemination and maintenance of resistant variants in cell culture models. Blocking virus cell-cell transmission prevents emergence of drug resistance in persistent viral infection including resistance to HCV DAAs.


Subject(s)
Antiviral Agents/pharmacology , Cell Communication , Drug Resistance, Viral , Hepacivirus/drug effects , Hepacivirus/physiology , Hepatitis C/immunology , Hepatitis C/virology , Virus Internalization , Antibodies, Neutralizing/metabolism , Carbamates , Cell Communication/immunology , Cells, Cultured , Drug Resistance, Viral/immunology , Hepacivirus/growth & development , Hepatitis C/pathology , Humans , Imidazoles/pharmacology , Oligopeptides/pharmacology , Proline/analogs & derivatives , Proline/pharmacology , Pyrrolidines , Valine/analogs & derivatives , Viral Load/immunology , Virus Internalization/drug effects
15.
J Hepatol ; 54(1): 48-55, 2011 Jan.
Article in English | MEDLINE | ID: mdl-20932595

ABSTRACT

BACKGROUND AND AIMS: ITX 5061 is a clinical stage small molecule compound that promotes high-density lipoprotein (HDL) levels in animals and patients by targeting the scavenger receptor BI protein pathway. Since SR-BI is a known co-receptor for HCV infection, we evaluated these compounds for their effects on HCV entry. METHODS: We obtained ITX 5061 and related compounds to characterize their interaction with SR-BI and effects on HCV entry and infection. RESULTS: We confirmed that a tritium-labeled compound analog (ITX 7650) binds cells expressing SR-BI, and both ITX 5061 and ITX 7650 compete for HDL-mediated lipid transfer in an SR-BI dependent manner. Both molecules inhibit HCVcc and HCVpp infection of primary human hepatocytes and/or human hepatoma cell lines and have minimal effects on HCV RNA replication. Kinetic studies suggest that the compounds act at an early post-binding step. CONCLUSIONS: These results suggest that the ITX compounds inhibit HCV infection with a mechanism of action distinct from other HCV therapies under development. Since ITX 5061 has already been evaluated in over 280 patients with good pharmacokinetic and safety profiles, it warrants proof-of-concept clinical studies in HCV infected patients.


Subject(s)
Antiviral Agents/pharmacology , Hepacivirus/drug effects , Scavenger Receptors, Class B/antagonists & inhibitors , Virus Internalization/drug effects , Amides/pharmacology , Animals , CHO Cells , Cell Line , Cricetinae , Cricetulus , Hepacivirus/pathogenicity , Hepacivirus/physiology , Hepatitis C, Chronic/drug therapy , Hepatitis C, Chronic/virology , Hepatocytes/drug effects , Hepatocytes/metabolism , Hepatocytes/virology , Humans , Kinetics , Lipoproteins, HDL/metabolism , Receptors, Virus/antagonists & inhibitors
16.
J Exp Med ; 207(9): 2019-31, 2010 Aug 30.
Article in English | MEDLINE | ID: mdl-20713596

ABSTRACT

End-stage liver disease caused by chronic hepatitis C virus (HCV) infection is a leading cause for liver transplantation (LT). Due to viral evasion from host immune responses and the absence of preventive antiviral strategies, reinfection of the graft is universal. The mechanisms by which the virus evades host immunity to reinfect the liver graft are unknown. In a longitudinal analysis of six HCV-infected patients undergoing LT, we demonstrate that HCV variants reinfecting the liver graft were characterized by efficient entry and poor neutralization by antibodies present in pretransplant serum compared with variants not detected after transplantation. Monoclonal antibodies directed against HCV envelope glycoproteins or a cellular entry factor efficiently cross-neutralized infection of human hepatocytes by patient-derived viral isolates that were resistant to autologous host-neutralizing responses. These findings provide significant insights into the molecular mechanisms of viral evasion during HCV reinfection and suggest that viral entry is a viable target for prevention of HCV reinfection of the liver graft.


Subject(s)
Hepacivirus/physiology , Hepatitis C Antibodies/pharmacology , Hepatitis C/transmission , Liver Transplantation/adverse effects , Virus Internalization/drug effects , Adolescent , Adult , Aged , Amino Acid Sequence , Animals , Antigens, CD/genetics , Antigens, CD/immunology , Cell Line , Female , Genetic Variation , Hepacivirus/drug effects , Hepacivirus/genetics , Hepacivirus/isolation & purification , Hepatitis C/immunology , Hepatitis C/virology , Humans , Male , Mice , Mice, SCID , Middle Aged , Molecular Sequence Data , Phylogeny , Sequence Alignment , Tetraspanin 28 , Viral Envelope Proteins/chemistry , Viral Envelope Proteins/genetics , Viral Envelope Proteins/immunology , Young Adult
17.
Gastroenterology ; 139(3): 953-64, 964.e1-4, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20685314

ABSTRACT

BACKGROUND & AIMS: Hepatitis C virus (HCV) infection is a challenge to prevent and treat because of the rapid development of drug resistance and escape. Viral entry is required for initiation, spread, and maintenance of infection, making it an attractive target for antiviral strategies. The tight junction protein claudin-1 (CLDN1) has been shown to be required for entry of HCV into the cell. METHODS: Using genetic immunization, we produced 6 monoclonal antibodies against the host entry factor CLDN1. The effects of antibodies on HCV infection were analyzed in human cell lines and primary human hepatocytes. RESULTS: Competition and binding studies demonstrated that antibodies interacted with conformational epitopes of the first extracellular loop of CLDN1; binding of these antibodies required the motif W(30)-GLW(51)-C(54)-C(64) and residues in the N-terminal third of CLDN1. The monoclonal antibodies against CLDN1 efficiently inhibited infection by HCV of all major genotypes as well as highly variable HCV quasispecies isolated from individual patients. Furthermore, antibodies efficiently blocked cell entry of highly infectious escape variants of HCV that were resistant to neutralizing antibodies. CONCLUSIONS: Monoclonal antibodies against the HCV entry factor CLDN1 might be used to prevent HCV infection, such as after liver transplantation, and might also restrain virus spread in chronically infected patients.


Subject(s)
Antibodies, Monoclonal/pharmacology , Antiviral Agents/pharmacology , Hepacivirus/drug effects , Hepatitis C/prevention & control , Hepatocytes/drug effects , Membrane Proteins/antagonists & inhibitors , Virus Internalization/drug effects , Animals , Antibodies, Monoclonal/metabolism , Antibodies, Monoclonal/toxicity , Antibody Specificity , Antiviral Agents/metabolism , Antiviral Agents/toxicity , Binding Sites, Antibody , Binding, Competitive , CHO Cells , Cell Survival/drug effects , Claudin-1 , Cricetinae , Cricetulus , Dose-Response Relationship, Drug , Epitopes , Genotype , Hep G2 Cells , Hepacivirus/genetics , Hepacivirus/pathogenicity , Hepatitis C/immunology , Hepatocytes/immunology , Hepatocytes/virology , Humans , Membrane Proteins/immunology
18.
Hepatology ; 51(4): 1144-57, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20069648

ABSTRACT

UNLABELLED: The tight junction protein claudin-1 (CLDN1) has been shown to be essential for hepatitis C virus (HCV) entry-the first step of viral infection. Due to the lack of neutralizing anti-CLDN1 antibodies, the role of CLDN1 in the viral entry process is poorly understood. In this study, we produced antibodies directed against the human CLDN1 extracellular loops by genetic immunization and used these antibodies to investigate the mechanistic role of CLDN1 for HCV entry in an infectious HCV cell culture system and human hepatocytes. Antibodies specific for cell surface-expressed CLDN1 specifically inhibit HCV infection in a dose-dependent manner. Antibodies specific for CLDN1, scavenger receptor B1, and CD81 show an additive neutralizing capacity compared with either agent used alone. Kinetic studies with anti-CLDN1 and anti-CD81 antibodies demonstrate that HCV interactions with both entry factors occur at a similar time in the internalization process. Anti-CLDN1 antibodies inhibit the binding of envelope glycoprotein E2 to HCV permissive cell lines in the absence of detectable CLDN1-E2 interaction. Using fluorescent-labeled entry factors and fluorescence resonance energy transfer methodology, we demonstrate that anti-CLDN1 antibodies inhibit CD81-CLDN1 association. In contrast, CLDN1-CLDN1 and CD81-CD81 associations were not modulated. Taken together, our results demonstrate that antibodies targeting CLDN1 neutralize HCV infectivity by reducing E2 association with the cell surface and disrupting CD81-CLDN1 interactions. CONCLUSION: These results further define the function of CLDN1 in the HCV entry process and highlight new antiviral strategies targeting E2-CD81-CLDN1 interactions.


Subject(s)
Antibodies/pharmacology , Antigens, CD/immunology , Cell Adhesion Molecules/immunology , Hepatitis C/therapy , Membrane Proteins/physiology , 12E7 Antigen , Antigens, CD/physiology , Claudin-1 , Humans , Immunization , Membrane Proteins/immunology , Neutralization Tests , Scavenger Receptors, Class B/physiology , Tetraspanin 28 , Tight Junctions/physiology , Virus Internalization
19.
J Infect Dis ; 198(11): 1656-66, 2008 Dec 01.
Article in English | MEDLINE | ID: mdl-18925843

ABSTRACT

BACKGROUND: Nonrandom distribution of hepatitis C virus (HCV) quasispecies (compartmentalization between blood plasma and leukocytes) suggests the presence of HCV leukotropic variants. HCV compartmentalization in the setting of liver transplantation (LT) is poorly understood. To study HCV leukotropic variants, we investigated the evolution of HCV compartmentalization after immunosuppression in liver transplant recipients. METHODS: Plasma and peripheral blood mononuclear cell (PBMC) samples were collected from 5 liver transplant recipients before and after LT. We used clone sequencing to analyze the hypervariable region 1 (HVR1)-E2(384-419) region, which plays a key role in HCV entry and the induction of neutralizing responses, and assessed compartmentalization through phylogenetic analyses and Mantel's test. RESULTS: Compartmentalization was frequent in the LT setting. HCV quasispecies were more homogeneous after LT in both the plasma and PBMC compartments, with a significant decrease in quasispecies complexity (P = .003) and genetic distances (P = .004) after transplantation. Our analysis identified 8 PBMC-related amino acid residues in HVR1. CONCLUSIONS: HCV compartmentalization between plasma and PBMCs and the emergence of leukotropic variants could be potentiated by immunosuppression in liver transplant recipients. The identification of defined leukotropic variants may contribute to the understanding of virus-host interactions after transplantation.


Subject(s)
Amino Acids/blood , Hepacivirus/genetics , Hepacivirus/physiology , Leukocytes, Mononuclear/virology , Liver Transplantation/adverse effects , Amino Acid Sequence , Evolution, Molecular , Genetic Variation , Humans , Molecular Sequence Data , Phylogeny , Selection, Genetic , Viral Proteins/chemistry , Viral Proteins/genetics
20.
Hepatology ; 46(6): 1722-31, 2007 Dec.
Article in English | MEDLINE | ID: mdl-18000990

ABSTRACT

UNLABELLED: Hepatitis C virus (HCV) is a major cause of chronic hepatitis worldwide. Scavenger receptor class B type I (SR-BI) has been shown to bind HCV envelope glycoprotein E2, participate in entry of HCV pseudotype particles, and modulate HCV infection. However, the functional role of SR-BI for productive HCV infection remains unclear. In this study, we investigated the role of SR-BI as an entry factor for infection of human hepatoma cells using cell culture-derived HCV (HCVcc). Anti-SR-BI antibodies directed against epitopes of the human SR-BI extracellular loop specifically inhibited HCVcc infection in a dose-dependent manner. Down-regulation of SR-BI expression by SR-BI-specific short interfering RNAs (siRNAs) markedly reduced the susceptibility of human hepatoma cells to HCVcc infection. Kinetic studies demonstrated that SR-BI acts predominately after binding of HCV at an entry step occurring at a similar time point as CD81-HCV interaction. Although the addition of high-density lipoprotein (HDL) enhanced the efficiency of HCVcc infection, anti-SR-BI antibodies and SR-BI-specific siRNA efficiently inhibited HCV infection independent of lipoprotein. CONCLUSION: Our data suggest that SR-BI (i) represents a key host factor for HCV entry, (ii) is implicated in the same HCV entry pathway as CD81, and (iii) targets an entry step closely linked to HCV-CD81 interaction.


Subject(s)
Antigens, CD/immunology , Hepatitis C/immunology , Scavenger Receptors, Class B/immunology , Carcinoma, Hepatocellular , Cell Line, Tumor , Cells, Cultured , Hepacivirus , Humans , Tetraspanin 28
SELECTION OF CITATIONS
SEARCH DETAIL
...