Skip to main content
Advertisement
Browse Subject Areas
?

Click through the PLOS taxonomy to find articles in your field.

For more information about PLOS Subject Areas, click here.

  • Loading metrics

Higher rates of HBsAg clearance with tenofovir-containing therapy in HBV/HIV co-infection

  • Pierre Gantner,

    Roles Conceptualization, Formal analysis, Methodology, Writing – original draft

    Affiliation Virology Laboratory, Hôpitaux Universitaires de Strasbourg, Strasbourg University, INSERM, UMR-S U1109, Strasbourg, France

  • Laurent Cotte,

    Roles Conceptualization, Data curation, Formal analysis, Writing – review & editing

    Affiliations Infectious Diseases Department, Croix-Rousse Hospital, Hospices Civils de Lyon, Lyon, France, INSERM U1052, Lyon, France

  • Clotilde Allavena,

    Roles Conceptualization, Data curation, Formal analysis, Methodology, Writing – review & editing

    Affiliation Infectious Diseases Department, CHU de Nantes, Nantes, France

  • Firouzé Bani-Sadr,

    Roles Data curation, Writing – review & editing

    Affiliation Infectious Diseases Department, CHU de Reims, Reims, France

  • Thomas Huleux,

    Roles Data curation, Writing – review & editing

    Affiliation University Department of Infectious Diseases, Tourcoing Hospital, Tourcoing, France

  • Claudine Duvivier,

    Roles Data curation, Writing – review & editing

    Affiliations AP-HP-Necker Hospital, Infectious Diseases Department, Necker-Pasteur Infectiology Center, Medical Center of Pasteur Institut, Necker-Pasteur Infectiology Center; Paris Descartes University, Sorbonne Paris Cité, IHU Imagine, Paris, France

  • Marc-Antoine Valantin,

    Roles Data curation, Writing – review & editing

    Affiliation Infectious Diseases Department, Assistance Publique—Hôpitaux de Paris, Pitié-Salpêtrière Hospital; Sorbonne Universités, UPMC Université Paris 06, INSERM, Institut Pierre Louis d'épidémiologie et de Santé Publique (IPLESP UMRS 1136), Paris, France

  • Christine Jacomet,

    Roles Data curation, Writing – review & editing

    Affiliation Infectious Diseases Department, CHU Clermont-Ferrand, Clermont-Ferrand, France

  • Véronique Joly,

    Roles Data curation, Writing – review & editing

    Affiliation Infectious Diseases Department, APHP Hôpital Bichat, Paris, France

  • Antoine Chéret,

    Roles Data curation, Writing – review & editing

    Affiliation Internal Medicine Department, CHU Bicètre, France; Université Paris Descartes, Sorbonne Paris Cité, Paris, France

  • Pascal Pugliese,

    Roles Data curation, Writing – review & editing

    Affiliation Infectious Diseases Department, Centre Hospitalier Universitaire de Nice, Hôpital l'Archet, Nice, France

  • Pierre Delobel,

    Roles Data curation, Writing – review & editing

    Affiliation Infectious Diseases Department, CHU Toulouse-Purpan, Toulouse, France

  • André Cabié,

    Roles Data curation, Writing – review & editing

    Affiliations Infectious Diseases Department, CHU de Martinique, Fort-de-France, France, Université des Antilles EA4537 and INSERM CIC1424, Fort-de-France, France

  • David Rey ,

    Roles Conceptualization, Data curation, Formal analysis, Methodology, Supervision, Writing – review & editing

    david.rey@chru-strasbourg.fr

    Affiliation Le Trait d’Union, HIV-Infection Care Center, Hôpitaux Universitaires de Strasbourg, Strasbourg, France

  • for the Dat’AIDS Study Group

    Members are listed in the Acknowlegments section.

Abstract

Introduction

Achieving functional cure of chronic HBV infection (Hepatitis B surface antigen [HBsAg] clearance, eventually followed by acquisition of anti-hepatitis B surface antigen [Anti-HBs]) in individuals with HIV and HBV infections is a rare event. In this setting, factors related to HBV cure have not yet been fully characterized.

Methods

HIV-infected individuals with chronic HBV infection enrolled in the French Dat’AIDS cohort (NCT02898987), who started combined antiretroviral (cART)-anti-HBV treatment were retrospectively analyzed for HBsAg loss and Anti-HBs seroconversion.

Results

Overall, 1419 naïve-subjects received three different cART-anti-HBV treatment schedule: (1) 3TC or FTC only (n = 150), (2) TDF with or without 3TC or FTC (n = 489) and (3) 3TC or FTC as first line followed by adding/switching to TDF as second line (n = 780). Individuals were followed-up for a median of 89 months (IQR, 56–118). HBV-DNA was < 15 IU/mL in 91% of individuals at the end of the follow-up. Overall, 97 individuals cleared HBsAg (0.7/100 patient-years), of whom, 67 seroconverted for Anti-HBs (0.5/100 patient-years). A high CD4 nadir, a short delay between HBV diagnosis and treatment, a longer time on HBV therapy, an African origin and TDF-based therapy were independent predictors of HBsAg clearance (Probability of odds ratio [OR]>1, >95%) suggested by Bayesian analysis. Also, TDF-based regimen as first line (OR, 3.03) or second line (OR, 2.95) increased rates of HBsAg clearance compared to 3TC or FTC alone, with a 99% probability.

Conclusions

HBsAg clearance rate was low in HIV-HBV co-infected cART-anti-HBV treated individuals, but was slightly improved on TDF-based regimen.

Introduction

HIV-associated comorbidities remain one major public health challenge worldwide, despite the success of combined antiretroviral therapy (ART) in controlling HIV viral load and in increasing life expectancy of people living with HIV. For instance, individuals with both chronic HBV and HIV infections have an increased risk of developing both cirrhosis and hepatocellular carcinoma, leading to heightened risk of mortality [12]. HIV-infected individuals are at higher risk of both acquiring HBV infection (due to the shared risk of transmission factors between the two viruses) and developing chronic hepatitis B (HBV) after acute infection [3]. In European countries, chronic hepatitis B marked by a positive Hepatitis B surface antigen (HBsAg) for at least 6 months is more prevalent in HIV-infected individuals (1 to 15%) [47] compared to uninfected individuals (< 1%) [8]. Based on HBV infection repartition worldwide, HBV prevalence ranges from 8.5 to 28% in HIV-infected individuals in Asia or Africa [4,910].

Long-term virological suppression of HBV replication represents the main endpoint of current HBV therapy, while HBsAg clearance is considered an optimal endpoint [1112]. Functional cure of chronic HBV infection, defined as HBsAg loss, followed by the acquisition of anti-hepatitis B surface antibody (Anti-HBs), is a rare event: various studies have reported HBsAg seroclearance rates ranging from 1.7 to 2.6/100 patient-years in HBV/HIV co-infected subjects undergoing different therapeutic strategies [1315]. However, the number of participants, a short follow-up duration and the lack of real-life settings limit these data. Of note, even in individuals harboring HBV resolved serological profile, HBV viral reactivation can occur in some clinical conditions, such as immunosuppression [16] due to episomal HBV-DNA persistence in hepatocytes [17].

Nucleoside analog drugs such as lamivudine, 3TC; emtricitabine, FTC; tenofovir disoproxil fumarate, TDF; and tenofovir alafenamide, TAF, suppress HBV replication and can control HIV replication during HBV/HIV co-infection. The use of TDF-based combined antiretroviral therapy (cART) to treat this co-infection has increased over the years [18], and is now the standard for nucleoside analog backbone. However, functional cure achievement rates and related factors to HBV resolution in this setting have not yet been fully characterized [4]. In addition, quantitative HBsAg evolution has not been studied in co-infected individuals. In this study, we aimed at investigating the outcomes of a TDF and 3TC or FTC-based regimen initiation among cART-naïve chronically HBV/HIV co-infected individuals.

Methods

Study population and study design

cART-naïve HIV-infected individuals with chronic HBV infection, enrolled in the French Dat’AIDS cohort, who started undergoing anti-HBV-cART treatment were retrospectively analyzed (NCT02898987) [19]. This study was performed in accordance with the principles of the Declaration of Helsinki and current French legislation relating to biomedical research. As this study relied on already existing clinical data only and as there was no intervention on study participants, there was no need of an ethics committee advice according to French laws available when the study was performed. The DatAIDS cohort is registered on Clinicaltrials.gov under the identifier NCT02898987 and all included individuals provided written informed consent. Among HIV-infected individuals enrolled in the Dat’AIDS cohort, those harboring positive HBsAg for at least 6 months and then initiating HBV/HIV therapy between 2006 and 2016 were included. HCV co-infected individuals and those receiving Entecavir or PEGylated-Interferon were excluded, due to their potential interaction on HBV disease outcomes (Fig 1). Longitudinal data and variables that could influence HBV infection outcomes were recorded (HBV-DNA, HBV serology, laboratory parameters, demographic parameters, HIV-related conditions and HDV co-infection). The main study endpoints were confirmed HBsAg loss or anti-HBs seroconversion. Follow-up was censored if subjects were lost to follow-up or if they stopped anti-HBV treatment. Hepatitis B e antigen (HBeAg) was not taken into account in the analysis [2021]. Individuals were divided into three groups based on the three HBV therapy-cART schedules: (1) those receiving 3TC or FTC only, (2) those receiving TDF with or without 3TC or FTC; and (3) those receiving 3TC or FTC as first line regimen and then switched to TDF with or without 3TC or FTC. Primary outcome measures data (HBsAg and Anti-HBs) were collected from longitudinal medical records with a frequency based on each physician’s practice.

thumbnail
Fig 1. Study flowchart.

Of 59829 HIV-infected individuals enrolled in the Dat’AIDS cohort between 2006 and 2016, 1419 harbored HBsAg for at least 6 months and subsequently initiated cART-anti-HBV treatment. Individuals received one of the following HBV therapy schedule: (1) 3TC or FTC only (n = 150), (2) TDF with or without 3TC or FTC (n = 489); and (3) 3TC or FTC as first line followed by adding/switching to TDF as second line with or without 3TC or FTC (n = 780). TDF, Tenofovir disoproxil fumarate; 3TC, lamivudine; FTC, emtricitabine.

https://doi.org/10.1371/journal.pone.0215464.g001

Individuals with available quantitative HBV-DNA and quantitative HBsAg data at baseline and after more than two years of follow-up were also included in a sub study to assess the dynamics of these two parameters on HBV/HIV therapy.

Primary outcome measures were incidence rates of HBsAg loss and Anti-HBs seroconversion on two consecutive measurements. As secondary outcome measures, predictors of HBsAg loss were also assessed.

Statistical methods

Our data were presented in two different manners. First, we described baseline characteristics of individuals according to the three encountered treatment schedules: (1) 3TC/FTC, (2) TDF, and (3) 3TC/FTC and then switch to TDF. Second, we performed the statistical analysis dividing individuals according to treatment line: (1) 3TC/FTC 1st line, (2) TDF 1st, and (3) TDF 2nd line.

HBsAg loss and anti-HBs seroconversion incidences were expressed for 100 patient-years. Kaplan-Meier curves were used to depict HBsAg loss and anti-HBs seroconversion on follow-up, according to HBV treatment regimen line (and not for the whole therapeutic schedule), with the following cases: first line 3TC or FTC; first line TDF with or without 3TC or FTC; and second line TDF with or without 3TC or FTC (the baseline for the last group corresponds to the time of switching to TDF). Survival analysis results were expressed by cumulative percentages with 95% confidence interval (CoI).

Bayesian methods were used to assess predictors of HBsAg clearance in a multivariable analysis by estimating HBsAg loss posterior probability (mixed logistic model) with non-informative priors (centered at the value of no effect, with very tight precision of 0.001) [22] and using a time-varying approach. Modeling results were expressed as Odd-ratios (OR) with credibility intervals at 95% (CrI) of HBsAg clearance. Bayesian analysis allowed calculation of the probability (Pr) of observing an OR > 1, which was found to be clinically relevant if above 95%. A predictor of HBsAg loss is thus associated with a Pr > 95%. All Bayesian analyses were performed with JAGS using the rjags package in R version 3.1.1 (R Foundation for Statistical Computing, Vienna, Austria). For individuals who received TDF as second line regimen, both time on global anti-HBV and specifically on TDF were included in the model. Of note, the whole follow-up of individuals was taken into account in the multivariable analysis.

Results

Of 59829 HIV-infected individuals enrolled in the Dat’AIDS cohort between 2006 and 2016, 1419 (2.4%) harbored HBsAg for at least 6 months, were cART-naive and subsequently initiated cART-anti-HBV treatment. Individuals received HBV therapy according to one of the following schemes: (1) 3TC or FTC only (n = 150), (2) TDF with or without 3TC or FTC (n = 489); and (3) 3TC or FTC as first line followed by adding/switching to TDF as second line with or without 3TC or FTC (n = 780). When analyzed by treatment regimen line: 930 individuals received 3TC or FTC as first line (individuals who only received 3TC or FTC only plus those who were on 3TC or FTC as first line before switching to TDF as second line), 489 individuals received TDF as first line and 780 received TDF as second line. Subjects were primarily male (76%), had a median age of 36 years at baseline, 90% acquired both HIV and HBV by sexual transmission, and 6% of them were co-infected with hepatitis D virus (Table 1). Median follow-up was of 89 months (IQR, 56–118) but varied among groups of individuals (Fig 1). HIV virological control was achieved in 96% of individuals at the end of follow-up.

thumbnail
Table 1. Baseline characteristics of individuals with chronic HBV/HIV co-infection enrolled in the Dat’AIDS cohort.

https://doi.org/10.1371/journal.pone.0215464.t001

HBV infection related parameters (HBsAg, Anti-HBs and HBV-DNA) were routinely assessed at a median frequency of 24 weeks (IQR, 10–54). Overall, 97 (6.8%) individuals cleared HBsAg (0.7/100 patient-years), of whom, 67 (4.7%) seroconverted for Anti-HBs (0.5/100 patient-years). No occult HBV infection was documented in these individuals, as they all were HBV-DNA negative. HBsAg clearance occurred in 25, 19 and 53 individuals in the different therapeutic schedules 1, 2 and 3 at a median time of 73, 45 and 137 months, respectively. The corresponding median time of HBsAg clearance in group 3 as of 2nd line TDF regimen was of 61 months.

By Kaplan-Meier analysis, HBsAg clearance rate was higher on TDF-containing regimen (versus 3TC or FTC alone) as 1st line regimen (Fig 2A). For instance, after 72 months (half individuals of the global cohort was still on follow-up at month 72) HBsAg loss rate was; 1.7% (95% CoI, 1.2–2.2) with 1st line 3TC or FTC-based regimen, 4.0% (95% CoI, 3.0–5.1) with 1st line TDF with or without 3TC or FTC-based regimen. Of note, HBsAg clearance rate at month 72 was of 5.1% (95% CoI, 4.2–6.0) in individuals switching to TDF as 2nd line (Fig 2B), and thus similar to 1st line TDF. HBsAg clearance incidence was of 0.4/100 patient-years with 1st line 3TC or FTC-based regimen, 0.8/100 patient-years with 1st line TDF-based regimen and 0.9/100 patient-years with 2nd line TDF-based regimen. We also observed a trend toward higher rates of anti-HBs seroconversion on TDF-containing regimen before month 96, either when used as 1st (Fig 2C) or 2nd line (Fig 2D).

thumbnail
Fig 2. Kaplan-Meier analysis of HBsAg clearance and anti-HBs seroconversion.

Kaplan-Meier analysis of HBsAg clearance on 1st line regimen (A): 3TC or FTC-based regimen (n = 930, corresponding to the addition of the follow-up of individuals who only received FTC or 3TC [n = 150], plus the first part of the follow-up of those who then switched to TDF containing regimen [780]), or TDF with or without 3TC or FTC-based regimen (n = 489); and 2nd line regimen (B): TDF with or without 3TC or FTC-based regimen (n = 780, corresponding to the second part of the follow-up of those who first received 3TC or FTC and then TDF). The corresponding Kaplan-Meier analyses of Anti-HBs seroconversion are depicted in (C) for 1st line regimen and (D) for 2nd line regimen. TDF, Tenofovir disoproxil fumarate; 3TC, lamivudine; FTC, emtricitabine.

https://doi.org/10.1371/journal.pone.0215464.g002

A high CD4 nadir, a short delay between HBV diagnosis and treatment, a longer time on HBV therapy, an African origin and TDF-based therapy were independent predictors of HBsAg clearance by multivariable Bayesian analysis in our global cohort (Table 2 and S1 Table). Of note, TDF-based regimen as 1st line or 2nd line increased rates of HBsAg clearance at month 72 when compared to 3TC or FTC alone as 1st line with a > 95% probability, as suggested by Bayesian analysis. Furthermore, when repeating the multivariable analysis on a subset of 259 individuals with available quantitative HBsAg data (S1 Table), lower HBsAg levels at baseline were also associated independently with higher rates of HBsAg clearance (OR, 0.94, 95% CrI [0.89–0.99], Pr = 96%).

thumbnail
Table 2. Bayesian predictors of HBsAg clearance on HIV/HBV therapy.

https://doi.org/10.1371/journal.pone.0215464.t002

In a subset of individuals with available longitudinal follow-up of quantitative HBsAg (n = 259), HBsAg at baseline was of 2511 IU/mL (IQR, 475–5230). A slow decrease in HBsAg serum levels with a mean decrease of -1 IU/mL per year was observed on treatment. Median HBV-DNA was of 3.64 log IU/mL at baseline, and was < 15 IU/mL in 91% of individuals at the end of follow-up. Among Anti-HBs seroconverters (n = 67), median Anti-HBs titer at seroconversion was 62 IU/L (IQR, 22–296). Following seroconversion, anti-HBS titers also increased over time with a mean increase of 31 IU/L per year (Fig 3).

thumbnail
Fig 3. Anti-HBs titers dynamic after seroconversion.

Evolution of Anti-HBs titers was depicted in the subset of 67 individuals who seroconverted for Anti-HBs on HBV therapy.

https://doi.org/10.1371/journal.pone.0215464.g003

Discussion

Chronic HBV co-infection burden among HIV-infected individuals results in higher rates of mortality compared to mono-infected subjects [12] This study in a real-life large cohort of HBV/HIV co-infected individuals showing that HBV functional cure rate remains very low, i.e., <1/100 patient-years under long term cART, including anti-HBV treatment.

Although a TDF-based regimen has not been previously associated with a faster HBsAg level decline [23], we identified this regimen as a major predictor of HBsAg clearance, with an approximately 2-fold higher rate of HBs Ag loss compared to 3TC or FTC only-based-treatments. Of note, in our study TDF was associated with 3TC or FTC in some individuals, and the duration of this association could vary among individuals.

This result is consistent with higher anti-viral efficacy in terms of achieving HBV-DNA blood suppression with TDF compared to 3TC in HBV/HIV co-infected individuals, either as first line regimen [24] or as salvage therapy [25]. Owing to the above stated effects and due to resistance issues [26], 3TC and FTC alone should not be used as part of anti-HBV viral activity, in HBV/HIV co-infected individuals. However, rates of HBsAg seroclearance in our study were lower than previously reported rates with PEGylated-Interferon (3 to 7 /100 patient-years) [11].

When compared to other cohorts of HBV/HIV co-infected individuals, HBsAg clearance and anti-HBs seroconversion rates that we observed are lower [1315]. The large proportion of individuals taking 3TC of FTC only for a long period could partly explain this finding. However, HBsAg loss rates among TDF recipients in our cohort were also lower than the observed loss rates in HBV mono-infected (1.2/100 patient-years) [27] or HBV/HIV co-infected (3.6/100 patient-years) [28] TDF-receiving individuals.

Altogether, these results further reinforce the recommendation of using a TDF-based regimen in HBV/HIV co-infection [11]. Whether the use of the newly commercialized form of tenofovir (i.e. TAF) has a different impact on chronic HBV outcomes or not, is unknown yet and remains to be assessed in long-term follow-up studies [2930].

Moreover, we identified other predictors of HBsAg loss, such as a prompt initiation of HBV therapy, and HBV treatment duration, in accordance with previous reports [3132]. However, the delay between diagnosis and treatment initiation included in the model might not reflect the total duration since HBV infection, as HBV is largely undiagnosed in some key populations [33]. CD4+ T cell count has also been previously identified to negatively correlate with HBsAg clearance [34]. This result further emphasizes our finding that a high CD4+ T cell nadir correlates with higher rates of HBsAg clearance. Finally, we observed that an African origin was linked to a better prognosis of achieving HBV functional cure. This might be related to HBV genotype [35]. However, we could not assess this hypothesis as HBV genotype was poorly documented in our cohort.

We also found a very slow decline in quantitative HBsAg level on treatment. As it has previously been found to be an important predictor of HBsAg seroclearance in HIV/HBV co-infection [3638], it could also be a prognostic factor of HBV functional cure.

Our study has several limitations inherent to its retrospective design. Indeed, some medical records can be partly incomplete, and some variables that could have an impact on HBV-active cART, such as HBV genotype, are scarce. Moreover, we could not accurately estimate the time between HIV or HBV infection and treatment initiation, as medical charts only recorded dates of each infection diagnosis. However, it represents real-life data, on a large cohort of individuals followed for more than seven years. HBeAg was not taken into account in the present analysis, as it was poorly documented in our cohort, and thus could be a limitation of our study. However, HBeAg may have a limited impact on HBsAg clearance as: (1) high rates of precore mutants are observed in France [20], and (2) HBsAg loss has been described to occur at same rates between HBeAg-positive and -negative individuals [21].

In conclusion, HBsAg clearance rates were low while on HBV therapy; higher CD4 nadir, rapid initiation of HBV therapy, mainly with TDF-based regimen, could improve HBsAg clearance and Anti-HBs seroconversion. Quantitative HBsAg decreased slowly but significantly, and therefore could be monitored as a prognosis factor of HBV clearance.

Supporting information

S1 Table. Bayesian Multivariable analysis details.

Pr, Probability; OR, Odd-ratio; MSM, Men who have sex with men; FTC, emtricitabine; 3TC, lamivudine; TDF, tenofovir disoproxil fumarate.

https://doi.org/10.1371/journal.pone.0215464.s001

(DOCX)

Acknowledgments

We thank all the patients and staff of the HIV clinical centers who participated in the study.

Dat’AIDS Study Group: Besançon: C. Drobacheff-Thiébaut, A. Foltzer, K. Bouiller, L. Hustache-Mathieu, C. Chirouze, F. Bozon, O. Babre, P. Muret; Clermont-Ferrand: H. Laurichesse, O. Lesens, M. Vidal, N. Mrozek, C. Aumeran, O. Baud, V. Corbin, P. Letertre, S. Casanova, C. Jacomet; Guadeloupe: B. Hoen, I. Lamaury, I. Fabre, E. Curlier, R. Ouissa, K. Schepers, C. Herrmann-Storck, N. Dournon; La Roche sur Yon: D. Merrien, P. Perré, T. Guimard, O. Bollangier, S. Leautez, M. Morrier; Lyon: F. Ader, F. Biron, A. Boibieux, L. Cotte, T. Ferry, P Miailhes, T. Perpoint, S. Roux, S. Degroodt, C. Brochier, F. Valour, C. Chidiac; Marseille (IHU): C. Dhiver, M. Saadia Mokhtari, A. Ménard, H. Tissot Dupont, C. Toméi, L. Meddeb, AY. Belkhir, I. Ravaux; Marseille (Ste Marguerite): S. Brégigeon, O. Zaegel-Faucher, V. Obry-Roguet, H. Laroche, M. Orticoni, MJ. Soavi, P Geneau de Lamarlière, E. Ressiot, MJ. Ducassou, I. Jaquet, S. Galie, A. Galinier, P. Martinet, M. Landon, AS. Ritleng, A. Ivanova, C. Debreux, C. Lions, I. Poizot-Martin; Martinique: S. Abel, R. Césaire, L. Cuzin, G. Dos Santos, L. Fagour, M. Illiaquer, F. Najioullah, D. Nguyen, M. Ouka, S. Pierre-François, J. Pasquier, M. Pircher, B. Rozé, A. Cabié; Montpellier: N. Atoui, V. Le Moing, A. Makinson, N. Meftah, C. Merle de Boever, B. Montes, A. Montoya Ferrer, J. Reynes; Nancy: M. André, L. Boyer, MP. Bouillon, M. Delestan, T. May; Nantes: C. Allavena, C. Bernaud, E. Billaud, C. Biron, B. Bonnet, S. Bouchez, D. Boutoille, C. Brunet-Cartier, C. Deschanvres, N. Hall, T. Jovelin, P. Morineau, V. Reliquet, H. Hue, S. Sécher, M. Cavellec, A. Soria, V. Ferré, E. André-Garnier, A. Rodallec, M. Lefebvre, O. Grossi, O. Aubry, F. Raffi; Nice: P. Pugliese, S. Breaud, C. Ceppi, J. Courjon, E. Cua, J. Cottalorda, P. Dellamonica, E. Demonchy, A. De Monte, J. Durant, C. Etienne, S. Ferrando, J. G. Fuzibet, R. Garraffo, A. Joulie, K. Risso, V. Mondain, A. Naqvi, N. Oran, I. Perbost, S. Pillet, B. Prouvost-Keller, C. Pradier, S. Wehrlen-Pugliese, V. Rio, E. Rosenthal, S. Sausse, G. Zouzou; Orléans: L. Hocqueloux, T. Prazuck, C. Gubavu, A. Sève, M. Niang, C. Boulard; Paris (Bicêtre): A. Cheret, C. Goujard, Y. Quertainmont, E. Teicher, N. Lerolle, D. Vittecoq, O. Deradji, F. Fourreau, C. Pallier, A. Barrail-Tran; Paris (Bichat): R. Landman, V. Joly, C. Rioux, S. Lariven, A. Gervais, F.X. Lescure, S. Matheron, F. Louni, C. Godard, Z. Julia, M. Chansombat, D. Rahli, C. Mackoumbou-Nkouka, C. Charpentier, D. Descamps, G. Peytavin, Y. Yazdanpanah; Paris (Pasteur-Necker): PH. Consigny, G. Cessot, P. Bossi, J. Goesch, J. Gilquin, G. Benabdelmoumen, F. Lanternier, C. Charlier, K. Amazzough, B. Henry, B. Pilmis, C. Rouzaud, M. Morgand, F Touam, C. Louisin, C. Duvivier, O. Lortholary, R. Guery, F. Danion, J. Lourenco, P. Parize, N. Etienne, M. Launay, C. Rouzioux, V. Avettand Fenoel; Paris (Pitié): M.A. Valantin, F. Caby, R. Tubiana, R. Agher, S. Seang, L. Schneider, R. Palich, C. Blanc, C. Katlama; Reims: JL. Berger, Y. N’Guyen, D. Lambert, D. Lebrun, I. Kmiec, M. Hentzien, V. Brodard, F. Bani-Sadr; Saint-Etienne: E. Botelho-Nevers, A. Gagneux-Brunon, A. Frésard, F. Lucht; Strasbourg: P. Fischer, M. Partisani, C. Cheneau, M. Priester, ML. Batard, C. Bernard-Henry, E. de Mautort, D. Rey; Toulouse: M. Alvarez, N. Biezunski, A. Debard, C. Delpierre, P. Lansalot, L. Lelièvre, G. Martin-Blondel, M. Piffaut, L. Porte, K. Saune, P. Delobel; Tourcoing: F. Ajana, I. Alcaraz, V. Baclet, A. Boucher, P. Choisy, T. Huleux, B. Lafon-Desmurs, H. Melliez, A. Meybeck, A. Pasquet, M. Pradier, O. Robineau, N. Viget, M. Valette.

References

  1. 1. Mak D, Babb de Villiers C, Chasela C, Urban MI, Kramvis A. Analysis of risk factors associated with hepatocellular carcinoma in black South Africans: 2000–2012. PLoS One 2018;13: e0196057. pmid:29718992
  2. 2. Ioannou GN, Bryson CL, Weiss NS, Miller R, Scott JD, Boyko EJ. The prevalence of cirrhosis and hepatocellular carcinoma in patients with human immunodeficiency virus infection. Hepatology 2013;57: 249–57. pmid:22532055
  3. 3. Bodsworth NJ, Cooper DA, Donovan B. The influence of human immunodeficiency virus type 1 infection on the development of the hepatitis B virus carrier state. J Infect Dis 1991; 163: 1138–40. pmid:2019762
  4. 4. Singh KP, Crane M, Audsley J, Avihingsanon A, Sasadeusz J, Lewin SR. HIV-hepatitis B virus coinfection: epidemiology, pathogenesis, and treatment. AIDS 2017;31: 2035–2052. pmid:28692539
  5. 5. Morlat P, Roussillon C, Henard S, Salmon D, Bonnet F, Cacoub P, et al. Causes of death among HIV-infected patients in France in 2010 (national survey): trends since 2000. AIDS 2014;28: 1181–91. pmid:24901259
  6. 6. Brouard C, Pillonel J, Sogni P, Chollet A, Lazarus JV, Pascal X, et al. Hepatitis B virus in drug users in France: prevalence and vaccination history, ANRS-Coquelicot Survey 2011–2013. Epidemiol Infect 2017: 1–11.
  7. 7. Wang Q, De Luca A, Smith C, Zangerle R, Sambatakou H, Bonnet F, et al. Chronic Hepatitis B and C Virus Infection and Risk for Non-Hodgkin Lymphoma in HIV-Infected Patients: A Cohort Study. Ann Intern Med 2017;166: 9–17. pmid:27750294
  8. 8. Hofstraat SHI, Falla AM, Duffell EF, Hahné SJM, Amato-Gauci AJ, Veldhuijzen IK, et al. Current prevalence of chronic hepatitis B and C virus infection in the general population, blood donors and pregnant women in the EU/EEA: a systematic review. Epidemiol Infect 2017;145: 2873–85. pmid:28891457
  9. 9. Coffie PA, Tchounga BK, Bado G, Kabran M, Minta DK, Wandeler G, et al. Prevalence of hepatitis B and delta according to HIV-type: a multi-country cross-sectional survey in West Africa. BMC Infect Dis 2017;17: 466. pmid:28676076
  10. 10. Chen M, Wong WW, Law MG, Kiertiburanakul S, Yunihastuti E, Merati TP, et al. Hepatitis B and C Co-Infection in HIV Patients from the TREAT Asia HIV Observational Database: Analysis of Risk Factors and Survival. PLoS One 2016;11: e0150512. pmid:26933963
  11. 11. European Association for the Study of the Liver. EASL 2017 Clinical Practice Guidelines on the management of hepatitis B virus infection. J Hepatol 2017;67: 370–398. pmid:28427875
  12. 12. Terrault NA, Lok ASF, McMahon BJ, Chang KM, Hwang JP, Jonas MM, et al. Update on prevention, diagnosis, and treatment of chronic hepatitis B: AASLD 2018 hepatitis B guidance. Hepatology 2018;67: 1560–1599. pmid:29405329
  13. 13. Toscano AL, Corrêa MC. Evolution of hepatitis B serological markers in HIV coinfected patients: a case study. Rev Saude Publica 2017;51: 24. pmid:28380208
  14. 14. Boyd A, Canini L, Gozlan J, Lascoux-Combe C, Miailhes P, Fonquernie L, et al. Development of anti-hepatitis B surface (HBs) antibodies after HBs antigen loss in HIV-hepatitis B virus co-infected patients. J Clin Virol 2017;95: 55–60. pmid:28869890
  15. 15. Martín-Carbonero L, Teixeira T, Poveda E, Plaza Z, Vispo E, González-Lahoz J, et al. Clinical and virological outcomes in HIV-infected patients with chronic hepatitis B on long-term nucleos(t)ide analogues. AIDS 2011;25: 73–9. pmid:21076274
  16. 16. Salpini R, Colagrossi L, Bellocchi MC, Surdo M, Becker C, Alteri C, et al. Hepatitis B surface antigen genetic elements critical for immune escape correlate with hepatitis B virus reactivation upon immunosuppression. Hepatology 2015;61: 823–33. pmid:25418031
  17. 17. Werle-Lapostolle B, Bowden S, Locarnini S, Wursthorn K, Petersen J, Lau G, et al. Persistence of cccDNA during the natural history of chronic hepatitis B and decline during adefovir dipivoxil therapy. Gastroenterology 2004;126: 1750–8. pmid:15188170
  18. 18. Peters L, Mocroft A, Grint D, Moreno S, Calmy A, Jevtovic D, et al. Uptake of tenofovir-based antiretroviral therapy among HIV-HBV-coinfected patients in the EuroSIDA study. Antivir Ther 2018. (in press)
  19. 19. Pugliese P, Cuzin L, Cabié A, Poizot-Martin I, Allavena C, Duvivier C, et al. A large French prospective cohort of HIV-infected patients: the Nadis Cohort. HIV Med 2009;10: 504–11. pmid:19486189
  20. 20. Pivert A, Servant-Delmas A, Lunel-Fabiani F, Le Guillou-Guillemette H, Laperche S, Ducancelle A. Correlation between the promoter basal core and precore mutations and HBsAg quantification in French blood donors infected with hepatitis B virus. J Med Virol 2015;87: 529–35. pmid:25170961
  21. 21. Zoutendijk R, Zaaijer HL, de Vries-Sluijs TE, Reijnders JG, Mulder JW, Kroon FP, et al. Hepatitis B surface antigen declines and clearance during long-term tenofovir therapy in patients coinfected with HBV and HIV. J Infect Dis 2012;206: 974–80. pmid:22782950
  22. 22. Gaudet CE. Review of doing Bayesian data analysis: a tutorial with R, JAGS, and Stan (second edition). Clin Neuropsychol 2017;31: 1268–1270.
  23. 23. Striki A, Manolakopoulos S, Deutsch M, Kourikou A, Kontos G, Kranidioti H, et al. Hepatitis B s antigen kinetics during treatment with nucleos(t)ides analogues in patients with hepatitis B e antigen-negative chronic hepatitis B. Liver Int 2017;37: 1642–1650. pmid:28345181
  24. 24. Avihingsanon A, Lewin SR, Kerr S, Chang JJ, Piyawat K, Napissanant N, et al. Efficacy of tenofovir disoproxil fumarate/emtricitabine compared with emtricitabine alone in antiretroviral-naive HIV-HBV coinfection in Thailand. Antivir Ther 2010;15: 917–22. pmid:20834105
  25. 25. Quiros-Roldan E, Calabresi A, Lapadula G, Tirelli V, Costarelli S, Cologni G, et al. Evidence of long-term suppression of hepatitis B virus DNA by tenofovir as rescue treatment in patients coinfected by HIV. Antivir Ther 2008;13: 341–8. pmid:18572746
  26. 26. Levrero M, Subic M, Villeret F, Zoulim F. Perspectives and limitations for nucleo(t)side analogs in future HBV therapies. Curr Opin Virol 2018; 30: 80–89. pmid:29777955
  27. 27. Petersen J, Heyne R, Mauss S, Schlaak J, Schiffelholz W, Eisenbach C, et al. Effectiveness and Safety of Tenofovir Disoproxil Fumarate in Chronic Hepatitis B: A 3-Year Prospective Field Practice Study in Germany. Dig Dis Sci 2016;61: 3061–3071. pmid:26576555
  28. 28. Huang AJ, Núñez M. Outcomes in HIV/HBV-Coinfected Patients in the Tenofovir Era Are Greatly Affected by Immune Suppression. J Int Assoc Provid AIDS Care 2015;14: 360–8. pmid:25999329
  29. 29. Hsu YC, Wei MT, Nguyen MH. Tenofovir alafenamide as compared to tenofovir disoproxil fumarate in the management of chronic hepatitis B with recent trends in patient demographics. Expert Rev Gastroenterol Hepatol 2017; 11: 999–1008. pmid:28965428
  30. 30. Viganò M, Loglio A, Grossi G, Lampertico P. Tenofovir alafenamide (TAF) treatment of HBV, what are the unanswered questions? Expert Rev Anti Infect Ther 2018;16: 153–161. pmid:29338458
  31. 31. Chu CM, Liaw YF. HBsAg seroclearance in asymptomatic carriers of high endemic areas: appreciably high rates during a long-term follow-up. Hepatology 2007;45: 1187–92. pmid:17465003
  32. 32. Liaw YF, Brunetto MR, Hadziyannis S. The natural history of chronic HBV infection and geographical differences. Antivir Ther 2010;15: 25–33.
  33. 33. Boyd A, Duchesne L, Lacombe K. Research gaps in viral hepatitis. J Int AIDS Soc 2018;21: e25054. pmid:29633564
  34. 34. Arendt E, Jaroszewicz J, Rockstroh J, Meyer-Olson D, Zacher BJ, Mederacke I, et al. Improved immune status corresponds with long-term decline of quantitative serum hepatitis B surface antigen in HBV/HIV co-infected patients. Viral Immunol 2012;25: 442–7. pmid:23131018
  35. 35. Kuhnhenn L, Jiang B, Kubesch A, Vermehren J, Knop V, Susser S, et al. Impact of HBV genotype and mutations on HBV DNA and qHBsAg levels in patients with HBeAg-negative chronic HBV infection. Aliment Pharmacol Ther 2018;47: 1523–1535. pmid:29637585
  36. 36. Jaroszewicz J, Reiberger T, Meyer-Olson D, Mauss S, Vogel M, Ingiliz P, et al. Hepatitis B surface antigen concentrations in patients with HIV/HBV co-infection. PLoS One 2012;7: e43143. pmid:22905219
  37. 37. Boyd A, Maylin S, Moh R, Mahjoub N, Gabillard D, Eholié SP, et al. Hepatitis B surface antigen quantification as a predictor of seroclearance during treatment in HIV-hepatitis B virus coinfected patients from Sub-Saharan Africa. J Gastroenterol Hepatol 2016;31: 634–44. pmid:26313291
  38. 38. Martinot-Peignoux M, Lapalus M, Asselah T, Marcellin P. HBsAg quantification: useful for monitoring natural history and treatment outcome. Liver Int 2014;34: 97–107. pmid:24373085