Saturday, December 18, 2010

Predicting spontaneous clearance of acute hepatitis C virus in a large cohort of HIV-1-infected men


Predicting spontaneous clearance of acute hepatitis C virus in a large cohort of HIV-1-infected men

OPEN ACCESS
Emma C Thomson1,2,3, Vicki M Fleming2, Janice Main1, Paul Klenerman2, Jonathan Weber3, Joseph Eliahoo4, Jennifer Smith2, Myra O McClure3, Peter Karayiannis1
+ Author Affiliations

1Department of Hepatology, Wright-Fleming Institute, Faculty of Medicine, Imperial College London, London, UK
2Nuffield Department of Medicine, Peter Medawar Building for Pathogen Research, University of Oxford, UK
3Department of Infectious Diseases, Jefferiss Research Trust Laboratories, Wright-Fleming Institute, Faculty of Medicine, Imperial College London, London, UK
4Statistical Advisory Service, South Kensington Campus, Imperial College London, UK
Correspondence to
Dr Emma Thomson, Department of Hepatology, 2nd Floor, Wright-Fleming Institute, Faculty of Medicine, Imperial College London, Praed Street, London W2 1PG, UK; e.thomson@imperial.ac.uk
Revised 13 September 2010
Accepted 12 October 2010
Published Online First 7 December 2010
Next SectionAbstract

Objective An epidemic of acute hepatitis C virus (HCV) infection in HIV-positive men-who-have-sex-with-men (MSM) is emerging in Europe, Australia and the USA. The aim of this study was to characterise the natural history of primary HCV in this setting and to assess host and viral factors which predict spontaneous clearance.

Methods This prospective longitudinal cohort study was carried out in 112 HIV-positive patients who were followed in a single centre (the St Mary's Acute HCV Cohort). Plasma and peripheral blood mononuclear cells (PBMCs) were obtained at monthly intervals for 3 months and at 3-monthly intervals thereafter for a median of 45 months (IQR=29–69 months). The primary end point was spontaneous clearance of HCV. Cox regression was used to assess the impact of clinical and virological variables on outcome, including liver function, CD4 count, rate of HCV RNA decline, T cell response and clonal sequence evolution within the HCV E2 envelope gene.

Results 15% of patients cleared HCV spontaneously, while 85% progressed towards chronicity. The latter group included a significant proportion of ‘fluctuating’ progressors (37.5%), in whom a fall followed by a rise (>1 log10) in viraemia was observed. This was associated with superinfection with new HCV strains and partially effective T cell responses. Spontaneous clearance was strongly associated with a 2.2 log10 viral load drop within 100 days of infection (HR=1.78; p<0.0001), hr="5.04;" p="0.006)," hr="2.62;" p="0.048)" hr="2.66;" p="0.045)," n="12)">70 and a >10% genetic distance when compared with other strains derived from the same patient. Superinfection was defined as evidence of unique strains not present at baseline sampling, but evident at a later time point. Mean intrastrain corrected genetic distance and Hamming distances were calculated for all patients.

Non-synonymous substitutions per non-synonymous site divided by synonymous substitutions per synonymous site (dN/dS) ratios were calculated using MEGA 4.0. dN/dS values >1 were considered as evidence of positive selection.

Genetic sequence data
All viral sequences have been submitted to EMBL (accession numbers: HQ616913-HQ619203).

ELISpot assays
PBMCs were separated from whole blood by density gradient centrifugation and stored in liquid nitrogen. Thawed PBMCs were added to 96-well plates coated with mouse antihuman interferon γ (IFNγ) antibody. Peptides were added and the cells were incubated overnight. These were then washed off and mouse antihuman-IFNγ monoclonal antibody and alkaline phosphatase-conjugated antibiotin monoclonal antibody were added followed by incubation with 5-bromo-4-chloro-indolylphosphate-nitroblue-tetrazolium. Spot-forming units (SFUs) were counted on an ELISpot reader. Further details are given in Appendix 1 available online.

Error rates
In order to exclude contamination error as the mechanism for superinfection events, multiple time points were assessed and amplification of NS5B was carried out at different time points. In order to distinguish superinfection from co-infection with varying dominance of virus, PCR amplification was carried out using strain-specific primers. Further methodology details, including measures taken to prevent error, are available in Appendix 2 online.

Statistical analyses
As appropriate, Cox regression survival analysis, Kaplan–Meier survival plots, Mann–Whitney, χ2 and Fisher exact tests were used to analyse the significance of observations using the statistical computer package STATA 10.0. VL observations were right-censored at the time of treatment or end of follow-up. HRs were calculated with 95% CIs. p Values of ≤0.05 were considered to be statistically significant.

Previous SectionNext SectionResults
Cohort characteristics
A total of 112 patients were recruited to the study and followed-up for a median of 45 months (IQR=29–69 months; figure 1). Sequential VL measurements revealed three patterns of infection (figure 2); progression to chronicity with an initial peak viraemia and chronic plateau viraemia (PV) phase (47.5%), fluctuating viraemia (FV) defined as a fall followed by a rise in VL of >1 log10 IU/ml (37.5%); and SC (15%).
Demographic and clinical parameters are shown in table 1.





Figure 1

Patient cohort diagnosis, treatment and follow-up. A total of 112 HIV-positive patients with acute hepatitis C virus (HCV) were recruited to the the St Mary's Acute HCV Cohort study of whom 17 spontaneously cleared HCV. Treatment with pegylated interferon α (IFNα) and ribavirin was given for 48 weeks to 62 patients and withheld or delayed in a further 26. The sustained virological response rate was 66%. Ab, antibody; LFTs, liver function tests; RT-PCR, reverse transcription–PCR.




Figure 2

Clinical measures and outcome in (A) the whole cohort and (B) representative individual patients. Longitudinal hepatitis C virus (HCV) viral load measurements taken prior to treatment revealed three patterns of infection: spontaneous clearance (SC; 15%, red) and two patterns of progression (85%); peak and plateau (PV; 47.5%, black) or fluctuating viraemia (FV; 37.5%, blue) defined as a viral load peak followed by a decline (>1 log10) and subsequent rise in viraemia (>1 log10). Viral loads (VLs) obtained from spontaneous clearers (red), plateau progressors (black) and fluctuating progressors (blue) groups were pooled and plotted using a linear fit model from the time of peak viraemia to 300 days (insert). VL data were right-censored at the time of treatment for HCV. Grey shading indicates 95% CIs.

Table 1

Clinical measures and outcome (spontaneous clearance vs progression)

Sequential bilirubin and alanine aminotransferase (ALT) measurements and outcome
Early infection in progressors (PV and FV groups) and spontaneous clearers (SC) was associated with a rise in ALT and bilirubin. The peak SC bilirubin and ALT levels were significantly higher than in the progressor groups (table 1; figure 2).

Viral dynamics and outcome
SC was associated with an initial increase followed by a rapid fall in VL (2.2 log10 drop within 100 days and 3.46 log10 drop within 200 days; table 2) leading to clearance at a median of 108 days (95% CI 38 to 170 days). Thirteen FV patients became transiently PCR negative, but took on average 154 days to reach negativity (95% CI=113, 323 days; p=0.04). Of the 35 patients who achieved a 2.2 log10 drop in VL, 17 (49%) spontaneously cleared infection and 18 (51%) did not. Those who spontaneously cleared infection took a median of 45 versus 120 days to achieve a 2.2 log10 drop in VL (p=0.01).

Table 2

Median viral load (VL) drop in untreated spontaneous clearers and progressors

Genetic sequence and phylogenetic analyses
Phylogenetic analysis of majority quasispecies compared with reference sequences revealed several monophyletic clusters of infection within genotype 1a (77%), 4d (15%), 3a (7%) and 1b (4%), indicating several introductions of the virus into the MSM population (Appendix 3 online). Six per cent were infected with two genotypes and 2% had three genotypes (1a, 3a and 4d). SC occurred only in patients with genotype 1a infection, and this was significantly associated with outcome (p=0.04).

In order to determine which phylogenetic factors predicted SC, Hamming distance (diversity), number of quasispecies and dN/dS ratios were calculated in 50 patients (40 progressors and 10 clearers) at multiple (median=3) time points (table 3).

View this table:In this window In a new window
Table 3

Viral sequence analyses and predictors of spontaneous clearance

Initially, genetic diversity was noted to vary considerably over time in all groups (Appendix 4 online) but did not correlate with VL. In order to investigate if this variation was the result of superinfection events, Neighbor–Joining phylogenetic analyses of all longitudinally acquired E2 HVR-1 sequences (n=2291) were next carried out, revealing three groups of patients (figure 3); those who spontaneously cleared infection (n=10; 20%), those who progressed to chronicity with increasing quasispecies diversity but no evidence of superinfection (n=20; 40%); and those who progressed to chronicity with evidence of superinfection events (n=20; 40%; figure 5). A number of supplementary analyses were carried out to confirm these results (Appendix 2 online).




Figure 3

Patterns of acute HCV based on phylogenetic analysis in subjects 1–10. (A) Spontaneous clearance and plateau progression, (B) fluctuating viraemia associated with superinfection and (C) reactivation. A circular neighbour-joined phylogenetic tree analysis (Kimura 2-parameter model) of 305 HVR-1 sequences derived from subjects 1–10 at 2–4 time points is shown in order to illustrate patterns observed in different patient groups. Similar trees were created for 50 patients. Clones derived from each patient are given a different colour and those from two representative patients are shown in further detail (filled circles represent time point 1, clear circles time point 2, filled triangles time point 3 and clear triangles time point 4). (A) A gradual increase in genetic diversity can be seen in the progressor patient (black), while clones derived from the spontaneous clearer (red) reveal a narrower repertoire. Bootstrap values greater than 70% are shown. (B) A patient with evidence of superinfection is highlighted. Sequences from time point 4 (clear triangles, red brackets) consist of a dominant superinfecting strain (one clone related to the previous lineage is also present). This patient had a fluctuating viral load, with a negative HCV PCR (limit of detection=12 IU/ml) between time points 3 and 4. (C) A case of HCV reactivation is shown. The patient had a sustained virological response (PCR negative 6 months post-treatment) following 48 weeks of IFNα and ribavirin. One year later, he developed a rapid decrease in CD4 count (nadir 106x106/l) and was hospitalised with Pneumocystis jeroveci pneumonia. During this time, his ALT became elevated and HCV RNA was once again detected in his blood. He denied any risk factors for reacquisition of HCV. Branches derived from patient HVR-1 sequences (n=56) are coloured in filled green (baseline sample) and open green (3 years later). These samples derive from the same lineage.

Mean intrastrain calculations of genetic diversity and dN/dS ratios were used for patients found to have multiple strains of infection. Following this correction, diversity and dN/dS ratios increased gradually with time in progressors, but decreased in clearers (table 3, figure 4).




Figure 4

T cell responses and genetic variation within the E2 hypervariable region-1 (HVR-1) over time. Cumulative genetic variation (corrected Hamming distance and dN/dS ratio (non-synonymous substitutions per non-synonymous site divided by synonymous substitutions per synonymous site)) and T cell responses (interferon γ (IFNγ) spot-forming units (SFU)/106 cells) were assessed in each group of patients at two time points (sample 1, range 28–189 days; and sample 2, range 115–246 days from the last negative PCR test).

T cell responses
IFNγ production in response to pooled peptides spanning the entire viral genome were measured by ELISpot at two time points in 40 patients with genotype 1a infection. Total responses at baseline were significantly higher in SC versus PV (415 versus 115 SFU/million cells; p=0.005) and FV (415 versus 50 SFU/million cells; p=0.04) patients while responses were not significantly different by 3–6 months. In FV patients, transient viral control was associated with strong T cell responses. while viral relapse was often associated with loss of T cell response (Appendix 5 online).

SC was associated with significantly increased early responses to NS2/p7, NS3 protease and NS5A when compared with both PV and FV progressors (figure 5). SC patients also had significantly increased responses to NS3 helicase and NS5B than PV patients but not FV patients.



Figure 5
T cell responses across the hepatitis C virus (HCV) genome at baseline and 3–6 months. (A) Total T cell responses and (B) T cell responses across the HCV genome. T cells responses to pooled peptides spanning the entire HCV genome were assessed by ELISpot in 40 patients with genotype 1a infection at two time points. Significant results (p<0.05) are highlighted with asterisks (red indicates significant difference between SC and both FV and PV patients; green indicates significant difference between SC and PV patients). These responses were measured in total (A) and to 10 overlapping peptide pools (B) from across the entire HCV genome.
Fluctuating viraemia
Patients with a fluctuating VL were more likely to have evidence of superinfecting strains than those with plateau persistence (63% vs 29% p=0.04). However, not all FV patients had evidence of superinfection. Among those with single strain infection, persons with recrudescent viraemia had similar dN/dS ratios at baseline but higher cumulative ratios following a second sample than plateau progressors (1.78 vs 0.76; p=0.02). At baseline, those with single strain recrudescent viraemia had higher viral diversity (Hamming distance 2.84 vs 0.49; p=0.04). Peak bilirubin, ALT and number of quasispecies did not differ between these groups. One patient showed evidence of reactivation of HCV 2 years after successful treatment (figure 3C).
Response to treatment
Patients who did not achieve spontaneous clearance 12 weeks following diagnosis were offered treatment with 48 weeks of IFNα and ribavirin, and 66% achieved an SVR.

Discussion
An emerging epidemic of acute HCV in HIV-positive MSM has for the first time allowed characterisation of the natural history of early HCV infection in a large number of HIV-positive patients. Unravelling the determinants of spontaneous clearance of HCV is crucial in order to increase our understanding of the biological mechanisms which define protective immunity and to improve the management of patients who face a lengthy course of treatment which is toxic, expensive and not always effective. HCV infection in HIV-positive individuals is a growing clinical problem associated with an increased rate of disease progression and reduced response to treatment.21 In this longitudinal study of 112 HIV-positive patients with acute HCV, the key predictors of spontaneous clearance were VL dynamics, liver function, viral diversity and T cell response.
Three courses of infection were observed, based on VL changes over time (figure 2). SC (15%) was associated with a rapid decrease in VL. Patients who progressed towards chronicity exhibited two patterns of viraemia; the majority (47.5%) had an initial rise in VL followed by a PV phase, analogous to that seen in other chronic viral infections when the rate of viral clearance equilibrates with the rate of viral production. Some patients, however, developed a pattern of FV (37.5%) characterised by an initial decline and subsequent rise in VL (>1 log10). This pattern of viraemia has been observed in HIV-negative individuals and experimentally infected chimpanzees, but has not previously been correlated with superinfection and loss of T cell control in a large longitudinal study.
We aimed to employ the viral kinetics of early HCV infection to predict subsequent outcome. SC patients had a 2.2 log10 VL drop versus a 0.03 log10 drop in PV and 0.58 log10 drop in FV within 100 days of infection (HR=1.78, p<0.0001; class="xref-table" id="xref-table-wrap-2-2" href="http://gut.bmj.com/content/early/2010/12/06/gut.2010.217166.full#T2" jquery1292730609327="150">table 2), increasing to a 3.46 versus 0.17 (PV)/0.85 (FV) log10 drop by 200 days (HR=1.68; p<0.0001).
Cox regression analysis indicated that the best diagnostic tests for the prediction of SC were the maximum log10 drop in VL within 100 days, peak bilirubin (≥40 mmol/l), peak ALT(≥1000 IU/l) and baseline CD4 count (≥650×106/l) (tables 1, 2).
A difficulty encountered in predicting the outcome of infection is in distinguishing between those who will develop a FV and those who will go on to spontaneously clear infection, particularly those patients who become transiently RNA negative. Survival analysis was used to compare the time taken to a 2.2 log10 drop or a negative VL in FV and SC patients. The time to a 2.2 log10 drop was significantly shorter in SC versus FV patients (45 vs 120 days; p=0.01) as was the time to a negative VL in the 13 FV patients who did not spontaneously clear infection (108 vs 154 days); thus the rate of decline of viraemia is key in predicting outcome.
A detailed phylogenetic and genetic sequence analysis of 2291 longitudinally acquired clonal E2 HVR-1 sequences allowed us to track the impact of superinfection, and to assess whether measurements of viral diversity and selection had predictive value. Several monophyletic clusters were present (Appendix 3 online), in keeping with multiple transmission events in the MSM population, as previously described.22 Within individuals, the presence of superinfection was suggested by marked non-linear variation in genetic distance over time (Appendix 3 online). Phylogenetic tree analyses (figure 3) revealed evidence of the introduction of new infecting strains in 40%, both of the same genotype and with new genotypes. This high rate of superinfection is in keeping with the likely ongoing high risk behaviour of this group of patients22 and the observation of reinfection events in other cohorts.23 The majority of patients with FV had evidence of reinfection with new strains of HCV (63%). Strain-specific PCR was carried out to reduce overestimation of superinfection due the limitations of clonal analysis.
In order to investigate the influence of positive selection on the diversification of viral strains, mean intrastrain Hamming distance and dN/dS ratios were calculated. Both were significantly higher in progressors by 150 days of infection, consistent with similar observations in HIV-negative individuals.13 In the main, a gradual increase in viral diversity and dN/dS ratio was observed over time in PV patients as a result of generation of positively selected variants associated with a significantly weaker T cell response. In contrast, viral diversity and the dN/dS ratio decreased gradually in SC patients (table 3, figure 4) in association with a strong, multispecific T cell response.
When patients with superinfection were excluded from the analysis, genetic diversity at baseline was greater in the FV group than in the PV group, possibly reflecting a more diverse infecting quasispecies.
FV patients as a group displayed a slow rise in diversity over time and a rise followed by a fall in the dN/dS ratio. However, T cell responses, viral diversity and VL were highly variable in individuals within this group. This variation in FV patients would be in keeping with a model of partial immune control and may reflect within some individuals a delayed partially effective immune response and in others the emergence of escape variants or an unsustained protective immune response due to B or T cell failure, for example as a consequence of PD-1 receptor upregulation (a marker of T cell exhaustion and disease progression both in HIV-positive individuals and in acute HCV infection24 25). Alternatively, it could represent reinfection with the same strain of virus, which has been experimentally demonstrated in the chimpanzee model.26 A third explanation would be the persistence and re-emergence of virus from sites outside the bloodstream, for example the liver, PBMCs or the central nervous system.27 28
Changes observed within the E2 HVR-1 region may reflect direct antibody-driven responses13 29 and indirectly reflect T cell responses to other parts of the genome such as the non-structural proteins.30 Protective immunity in HIV-negative individuals is known to be associated with broad and multispecific CD4 and CD8 T cell responses.14–17 A higher baseline CD4 count (≥650×106/l) was noted in SC patients in this cohort (table 1). We therefore aimed to ascertain if decreased dN/dS ratios and viral diversity within E2 in SC and FV patients were related to a protective T-cell response, by measuring IFNγ responses to overlapping pooled peptides spanning the entire HCV genome. We found that SC patients exhibited significantly stronger multispecific IFNγ responses within 3 months of infection than either progressor group (PV, p=0.006; FV, p=0.01; figure 5). In particular, significant differences in response to NS2/p7, NS3 protease and NS5A were detected (SC vs FV and PV groups). SC patients also had significantly increased responses to NS3 helicase and NS5B than PV progressors. Further studies to characterise specific epitopes within these regions are needed.
One unusual case within the cohort suggested that reactivation of HCV may rarely occur in immunosuppressed individuals. This patient had evidence of reactivation of HCV 2 years after successful treatment for acute disease (figure 5C). This coincided with a period of severe immunosupression (nadir CD4 count 106×106/l and Pneumocystis jerovecii infection) requiring hospitalisation and may have been the result of loss of T cell control of a virus present in the liver but not in the bloodstream, or extremely low level viraemia below the limit of detection. Such cases are rare but have been previously reported in the literature.31
This study is the first longitudinal and detailed analysis of clinical outcome of acute HCV in a large cohort of HIV-positive patients, incorporating viral dynamics, detailed viral sequence evolution and T cell responses. It has a number of limitations. First, it is restricted to an HIV-positive population, although patterns of evolution during early infection may also apply to those with HCV monoinfection. Steps were taken to minimise error rates attributable to PCR contamination and clonal analysis, as detailed in the Supplementary information online. In order to overcome the problem of delayed HCV seroconversion in HIV-positive individuals, as previously reported in this cohort18 (to avoid the inclusion of chronically infected patients), we used retrospective PCR testing on stored HIV VL samples to date the time of infection. More work is needed to further define specific T cell targets and class I/II restriction of these responses, as well as the impact of viral escape.
Acute HCV infection is an emerging global epidemic in HIV-infected men and requires new public health planning and treatment strategies. These data suggest that the outcome of infection can be predicted by the measurement of VL dynamics as well as CD4 count and liver function. Lower genetic diversity and stronger T cell responses are also associated with SC, but were less useful than clinical measures in predicting outcome. Targeting of high risk patients—for example, with no evidence of rapid decline in VL within 100 days—may increase the efficacy of treatment while those more likely to clear infection spontaneously could be monitored for a longer period of time, thus avoiding a rigorous and expensive treatment.
Previous SectionNext Section
Acknowledgments
Hitesh Mistry and David Muir are acknowledged for the location and provision of backdated HIV VL samples.
Previous SectionNext Section
Footnotes
Funding This research was supported by a clinical research training fellowship from the Wellcome Trust (ECT), the Mason Medical Foundation and the NIHR Biomedical Research Centre.
Competing interests None.
Ethics approval This study was conducted with the approval of the Riverside Research Ethics Committee, Charing Cross Hospital, London, UK.
Provenance and peer review Not commissioned; externally peer reviewed.
This is an open-access article distributed under the terms of the Creative Commons Attribution Non-commercial License, which permits use, distribution, and reproduction in any medium, provided the original work is properly cited, the use is non commercial and is otherwise in compliance with the license.

See: http://creativecommons.org/licenses/by-nc/2.0/ and http://creativecommons.org/licenses/by-nc/2.0/legalcode.
Previous Section

References

Choo QL,
Kuo G,
Weiner AJ,
et al. Isolation of a cDNA clone derived from a blood-borne non-A, non-B viral hepatitis genome. Science 1989;244:359–62.
[Abstract/FREE Full text]

Wiese M,
Grungreiff K,
Guthoff W,
et al. Outcome in a hepatitis C (genotype 1b) single source outbreak in Germany—a 25-year multicenter study. J Hepatol 2005;43:590–8.
[CrossRef][Medline][Web of Science]

Martinez-Bauer E,
Forns X,
Armelles M,
et al. Hospital admission is a relevant source of hepatitis C virus acquisition in Spain. J Hepatol 2008;48:20–7.
[CrossRef][Medline][Web of Science]

Santantonio T,
Medda E,
Ferrari C,
et al. Risk factors and outcome among a large patient cohort with community-acquired acute hepatitis C in Italy. Clin Infect Dis 2006;43:1154–9.
[Abstract/FREE Full text]

Page K,
Hahn J,
Evans J,
et al. Acute hepatitis C virus infection in young adult injection drug users: a prospective study of incident infection, resolution, and reinfection. J Infect Dis 2009;200:1216–26.
[Abstract/FREE Full text]

Bica I,
McGovern B,
Dhar R,
et al. Increasing mortality due to end-stage liver disease in patients with human immunodeficiency virus infection. Clin Infect Dis 2001;32:492–7.
[Abstract/FREE Full text]

Alter MJ. Epidemiology of viral hepatitis and HIV co-infection. J Hepatol 2006;44(1 Suppl):S6–9.
[Medline][Web of Science]

Gilleece YC,
Browne RE,
Asboe D,
et al. Transmission of hepatitis C virus among HIV-positive homosexual men and response to a 24-week course of pegylated interferon and ribavirin. J Acquir Immune Defic Syndr 2005;40:41–6.
[CrossRef][Medline][Web of Science]

Dominguez S,
Ghosn J,
Valantin MA,
et al. Efficacy of early treatment of acute hepatitis C infection with pegylated interferon and ribavirin in HIV-infected patients. AIDS 2006;20:1157–61.
[CrossRef][Medline][Web of Science]

Matthews GV,
Hellard M,
Haber P,
et al. Characteristics and treatment outcomes among HIV-infected individuals in the Australian trial in acute hepatitis C. Clin Infect Dis 2009;48:650–8.
[Abstract/FREE Full text]

Fierer DS,
Uriel AJ,
Carriero DC,
et al. Liver fibrosis during an outbreak of acute hepatitis C virus infection in HIV-infected men: a prospective cohort study. J Infect Dis 2008;198:683–6.
[Abstract/FREE Full text]

Neumann AU,
Lam NP,
Dahari H,
et al. Hepatitis C viral dynamics in vivo and the antiviral efficacy of interferon-alpha therapy. Science 1998;282:103–7.
[Abstract/FREE Full text]

Farci P,
Shimoda A,
Coiana A,
et al. The outcome of acute hepatitis C predicted by the evolution of the viral quasispecies. Science 2000;288:339–44.
[Abstract/FREE Full text]

Cooper S,
Erickson AL,
Adams EJ,
et al. Analysis of a successful immune response against hepatitis C virus. Immunity 1999;10:439–49.
[CrossRef][Medline]

Cox AL,
Mosbruger T,
Lauer GM,
et al. Comprehensive analyses of CD8+ T cell responses during longitudinal study of acute human hepatitis C. Hepatology 2005;42:104–12.
[CrossRef][Medline][Web of Science]

Harcourt G,
Hellier S,
Bunce M,
et al. Effect of HLA class II genotype on T helper lymphocyte responses and viral control in hepatitis C virus infection. J Viral Hepat 2001;8:174–9.
[CrossRef][Medline][Web of Science]

Lechner F,
Wong DK,
Dunbar PR,
et al. Analysis of successful immune responses in persons infected with hepatitis C virus. J Exp Med 2000;191:1499–512.
[Abstract/FREE Full text]

Thomson EC,
Nastouli E,
Main J,
et al. Delayed anti-HCV antibody response in HIV-positive men acutely infected with HCV. AIDS 2009;23:89–93.
[CrossRef][Medline][Web of Science]

Lu L,
Nakano T,
He Y,
et al. Hepatitis C virus genotype distribution in china: predominance of closely related subtype 1b isolates and existence of new genotype 6 variants. J Med Virol 2005;75:538–49.
[CrossRef][Medline][Web of Science]

Weiner AJ,
Brauer MJ,
Rosenblatt J,
et al. Variable and hypervariable domains are found in the regions of HCV corresponding to the flavivirus envelope and NS1 proteins and the pestivirus envelope glycoproteins. Virology 1991;180:842–8.
[Medline]

Klenerman P,
Kim A. HCV–HIV coinfection: simple messages from a complex disease. PLoS Med 2007;4:e240.
[CrossRef][Medline]

Danta M,
Brown D,
Bhagani S,
et al. Recent epidemic of acute hepatitis C virus in HIV-positive men who have sex with men linked to high-risk sexual behaviours. AIDS 2007;21:983–91.
[Medline][Web of Science]

Herring BL,
Page-Shafer K,
Tobler LH,
et al. Frequent hepatitis C virus superinfection in injection drug users. J Infect Dis 2004;190:1396–403.
[Abstract/FREE Full text]

Day CL,
Kaufmann DE,
Kiepiela P,
et al. PD-1 expression on HIV-specific T cells is associated with T-cell exhaustion and disease progression. Nature 2006;443:350–4.
[CrossRef][Medline]

Urbani S,
Amadei B,
Tola D,
et al. PD-1 expression in acute hepatitis C virus (HCV) infection is associated with HCV-specific CD8 exhaustion. J Virol 2006;80:11398–403.
[Abstract/FREE Full text]

Bukh J,
Thimme R,
Meunier JC,
et al. Previously infected chimpanzees are not consistently protected against reinfection or persistent infection after reexposure to the identical hepatitis C virus strain. J Virol 2008;82:8183–95.
[Abstract/FREE Full text]

Maggi F,
Fornai C,
Vatteroni ML,
et al. Differences in hepatitis C virus quasispecies composition between liver, peripheral blood mononuclear cells and plasma. J Gen Virol 1997;78:1521–5.
[Abstract]

Forton DM,
Karayiannis P,
Mahmud N,
et al. Identification of unique hepatitis C virus quasispecies in the central nervous system and comparative analysis of internal translational efficiency of brain, liver, and serum variants. J Virol 2004;78:5170–83.
[Abstract/FREE Full text]

Gaud U,
Langer B,
Petropoulou T,
et al. Changes in hypervariable region 1 of the envelope 2 glycoprotein of hepatitis C virus in children and adults with humoral immune defects. J Med Virol 2003;69:350–6.
[CrossRef][Medline][Web of Science]

Klenerman P,
Lechner F,
Kantzanou M,
et al. Viral escape and the failure of cellular immune responses. Science 2000;289:2003.
[Medline]

Kim AY,
Schulze zur Wiesch J,
Kuntzen T,
et al. Impaired hepatitis C virus-specific T cell responses and recurrent hepa

No comments:

Post a Comment