Basic Study Open Access
Copyright ©The Author(s) 2016. Published by Baishideng Publishing Group Inc. All rights reserved.
World J Hepatol. Jul 28, 2016; 8(21): 902-914
Published online Jul 28, 2016. doi: 10.4254/wjh.v8.i21.902
Anti-hepatitis C virus potency of a new autophagy inhibitor using human liver slices model
Sylvie Lagaye, Jesintha Gaston, Stanislas Pol, Institut Pasteur, INSERM U1223, 75015 Paris, France
Sylvie Lagaye, Jesintha Gaston, Hong Shen, Institut Cochin, INSERM U1016, CNRS UMR8104, Université Paris Descartes (UMR S1016), 75014 Paris, France
Sonia Brun, Clarisse Dubray, Jerôme Courcambeck, Firas Bassisi, Philippe Halfon, Genoscience Pharma, 13006 Marseille, France
Ruzena Stranska, KU Leuven, Rega Institute, 3000 Leuven, Belgium
Claire Camus, Philippe Halfon, Laboratoire Alphabio, 13006 Marseille, France
Géraldine Rousseau, APHP, Groupe Hospitalier La Pitié Salpé­trière, Service de Chirurgie digestive et Hépato bilio pancréatique, 75013 Paris, France
Pierre-Philippe Massault, APHP, Groupe Hospitalier Cochin, Service de Chirurgie digestive, Hépato-biliaire et Endocrinienne, 75014 Paris, France
Stanislas Pol, Université Paris Descartes, 75014 Paris, France
Stanislas Pol, APHP, Groupe Hospitalier Cochin, Unité d’Hépatologie, 75014 Paris, France
Stanislas Pol, Institut Pasteur, Département de Recherche Translationnelle, INSERM UMS20, 75015 Paris, France
Author contributions: Lagaye S wrote the paper; Lagaye S, Brun S, Gaston J, Shen H, Stranska R, Camus C, Dubray C, Rousseau G, Massault PP, Courcambeck J and Bassisi F performed the experiments; Lagaye S, Brun S, Camus C, Halfon P and Pol S analyzed the data; Lagaye S, Halfon P and Pol S conceived and designed the experiments; Halfon P and Pol S contributed equally to the work.
Supported by The Institut National de la Santé et de la Recherche Médicale (INSERM, France); and the personal support of Professor Jean-François Delfraissy as Director of the French Agency, Agence Nationale de Recherches sur le Sida et les hépatites virales (ANRS).
Institutional review board statement: Institutional review board statement is not required for manuscript submission in our Institution.
Institutional animal care and use committee statement: No animal use in the experiments.
Conflict-of-interest statement: The authors have no conflict of interest to declare.
Data sharing statement: No data sharing.
Open-Access: This article is an open-access article which was selected by an in-house editor and fully peer-reviewed by external reviewers. It is distributed in accordance with the Creative Commons Attribution Non Commercial (CC BY-NC 4.0) license, which permits others to distribute, remix, adapt, build upon this work non-commercially, and license their derivative works on different terms, provided the original work is properly cited and the use is non-commercial. See: http://creativecommons.org/licenses/by-nc/4.0/
Correspondence to: Sylvie Lagaye, PhD, DSc, Senior Scientist, Institut Pasteur, INSERM U1223, 25-28 rue du Dr Roux, 75015 Paris, France. sylvie.lagaye@inserm.fr
Telephone: +33-1-40613424 Fax: +33-1-45688548
Received: March 24, 2016
Peer-review started: April 6, 2016
First decision: May 16, 2016
Revised: June 1, 2016
Accepted: June 27, 2016
Article in press: June 29, 2016
Published online: July 28, 2016

Abstract

AIM: To evaluate the antiviral potency of a new anti-hepatitis C virus (HCV) antiviral agent targeting the cellular autophagy machinery.

METHODS: Non-infected liver slices, obtained from human liver resection and cut in 350 μm-thick slices (2.7 × 106 cells per slice) were infected with cell culture-grown HCV Con1b/C3 supernatant (multiplicity of infection = 0.1) cultivated for up to ten days. HCV infected slices were treated at day 4 post-infection with GNS-396 for 6 d at different concentrations. HCV replication was evaluated by strand-specific real-time quantitative reverse transcription - polymerase chain reaction. The infectivity titers of supernatants were evaluated by foci formation upon inoculation into naive Huh-7.5.1 cells. The cytotoxic effect of the drugs was evaluated by lactate dehydrogenase leakage assays.

RESULTS: The antiviral efficacy of a new antiviral drug, GNS-396, an autophagy inhibitor, on HCV infection of adult human liver slices was evidenced in a dose-dependent manner. At day 6 post-treatment, GNS-396 EC50 was 158 nmol/L without cytotoxic effect (compared to hydroxychloroquine EC50 = 1.17 μmol/L).

CONCLUSION: Our results demonstrated that our ex vivo model is efficient for evaluation the potency of autophagy inhibitors, in particular a new quinoline derivative GNS-396 as antiviral could inhibit HCV infection in a dose-dependent manner without cytotoxic effect.

Key Words: Host antiviral therapy, Hepatitis C virus, Tissue culture, Autophagy, Quinoline derivative

Core tip: Hepatitis C virus (HCV) infection (or spread) is a serious public health challenge counting approximately 170 million people that are chronically infected worldwide. Efficient interferon-free treatments with new direct acting antivirals are expected to cure more than 90% of HCV-infected patients but they are not available in all the countries. Autophagy machinery is required to initiate HCV replication. Host antiviral therapy is an additional option for the treatment of HCV infection. The new autophagy inhibitor GNS-396 demonstrated significant efficacy and additive activity in inhibiting HCV replication and might be an additional option to treat HCV infected individuals.



INTRODUCTION

Approximately 170 million people worldwide are chronically infected with hepatitis C virus (HCV)[1]. Until recently, the most effective treatment against HCV infection was the combination of pegylated interferon-α 2a or b and ribavirin (PR) which achieved sustained virological response (SVR) in about 45% of individuals infected by HCV genotype 1, 65% by HCV genotype 4, 70% by HCV genotype 3 and more than 85% by HCV genotype 2[2,3]. The frequent side effects associated with PR and the rates of non response to PR includes partial or null virologic response and breakthrough or relapse after PR discontinuation. Thus, development of novel and more effective antiviral treatments were essential[4].

Two HCV NS3 protease inhibitors (PI), boceprevir (BOC) and telaprevir (TVR) have been approved and combined with PR, have increased the SVR to about 75% in therapy naïve HCV genotype 1 infected patients[5-9]. Over the past few years, other direct acting antivirals (DAAs) were developed[10-14] as second generation of PI with higher antiviral potency, HCV NS5A replication complex inhibitors and nucleotide analogue HCV NS5B polymerase inhibitors[13] as well as host-targeted indirect antivirals like cyclophilin inhibitors[15] and lambda interferon[15]. Interferon-free treatments with new DAAs are expected to cure more than 90% of HCV-infected patients[16]. But they are not available in all the countries[17]. At the present time, triple therapy combining PR with NS3 PI (TVR or BOC) is going to remain the main treatment for HCV patients[16-21]. That is why it appears important to continue research in limiting virus replication and the autophagy inhibition could be a new additional pathway because of recent evidences obtained regarding to an increased autophagic response in the liver of chronically HCV infected patients[22].

Autophagy is a catabolic process which degrades a cellular own component through the lysosomal machinery[23]. It has been shown that autophagy is activated during virus and bacterial infection[24] and that some viruses can use the autophagy system to facilitate their own replication[25-29]. Previously, several studies evidenced that HCV infection resulted in endoplasmic reticulum stress and autophagy responses, that HCV regulated the autophagy pathway, that the autophagy machinery was required to initiate HCV replication, and finally, that the suppression of autophagy inhibited HCV replication[30-35]. Interestingly, it has been demonstrated that HCV induces autophagosomes via a Class III PI3K-independent pathway and uses autophagosomal membranes as sites for its RNA replication[36].

The lysosomotropic anti-malarial drugs, chloroquine (CQ) and hydroxychloroquine (HCQ), belonging to the quinoline family, are among the autophagy inhibitors, which act by preventing the acidification of lysosomes, leading to the inhibition of both fusion of autophagosome with lysosome and lysosomal protein degradation[23]. In fact, CQ exerts an inhibitory effect for several RNA viruses including coronaviruses, flaviviruses and human immunodeficiency virus[37-39]. Recently, it has been shown that a treatment with CQ of HCV infected cells suppressed the replication of the virus in a dose-dependent manner by preventing the autophagic proteolysis[40].

In the present study, we used the established ex vivo model of primary human liver slices culture which allows to the de novo replication of primary viral isolates and production of high titer infectious HCV particles[41] to evaluate the potential antiviral potency GNS-396[42], a new autophagy inhibitor in comparison with a well-known autophagy inhibitor, HCQ. Presented results might be additional options to treat HCV infected individuals.

MATERIALS AND METHODS
Human liver tissue specimens

Adult human primary liver tissue samples were obtained from HCV and also hepatitis B virus, and human immunodeficiency virus seronegative patients who underwent liver resection surgery, mainly for liver metastasis in the absence of underlying liver disease. Experimental procedures were carried out in accordance with French laws and Regulations.

Liver slices preparation, culture and infection

Slices were prepared and cultured as described[41,43]. Briefly, uninfected human liver slices, obtained from human liver resection, were cut into 350 μm thick slices of (2.7 × 106 cells per slice) with a vibratome (Leica, VTS1200) and transferred to 0.4 μm organotypic culture inserts (Millipore) in 12-wells plates (1 slice/well) containing 2 mL of complete Dulbecco’s modified eagles’s medium (DMEM) culture media and maintained at 37 °C under a constant flow of humidified 95% O2/5% CO2 for up to 24 h before viral infection. Cell number for tissue slices was estimated at approximately 2.7 × 106 cells per slice based on a 14-cell thick slice (cell diameter approximately 25 μm)[41]. The complete culture medium consisted of DMEM with 4.5 g/L D-glucose and glutamine (Life Technologies) supplemented with 10% fetal calf serum (Life Technologies, 16000-044), 5% penicillin-streptomycin (Life Technologies, 10378-016), 1% amphotericin (Sigma Aldrich), 5 μg/mL insulin (Life Technologies, 51500-056), 0.4 μg/mL dexamethasone (Sigma Aldrich, D4902), 10 mmol/L HEPES (Life Technologies, 15630080), non-essential amino acids (Life Technologies), 20 mmol/Lsodium pyruvate (Life Technologies) and 50 μg/mL ascorbic acid (Sigma Aldrich). One day post-culture in twelve-transwell plates, human primary liver slices were inoculated with HCV Con1/C3 at a multiplicity of infection equal to 0.1 at 37 °C in the same culture conditions as described above, for overnight. The infectious clone Con1/C3 (genotype 1b) (JFH1-derived chimeric viruses whose structural proteins are encoded by the genotype 1b-HCV sequence Con1)[44] could efficiently infect human liver slices which maintain their hepatocyte differentiation and retain normal physiological and biochemical parameters for at least 10 d. The inoculum was then removed; the slices were washed three times with PBS and then supplemented with complete culture medium. Then, liver slices were cultured without medium replacements, as previously described[41].

HCV RNA transfection and virus production

To produce HCVcc, viral RNAs were transcribed in vitro and electroporated into Huh-7.5.1 cell line (kindly provided by Professor Francis V Chisari, The Scripps Research Institute, La Jolla, CA), as described previously[45]. The infectious titer of cell culture supernatants was evaluated by classical titration assay[45]. In brief, the HCV infection of Huh-7.5.1 cells was performed with serial 10-fold dilution of viral supernatants. Seventy-two hours later, the formation of infected cells foci were detected by staining with human HCV positive sera or mouse monoclonal antibodies directed against HCV core (Ozyme) and nonstructural (NS5A) (Virostat, clone1877) proteins. Titrations were performed in duplicate.

Quantification of HCV strands RNA by real-time quantitative reverse transcription- polymerase chain reaction

A strand-specific real-time quantitative reverse transcription-polymerase chain reaction technique to quantify the intracellular levels of positive and negative strand HCV RNA was performed as previously described[46-49]. The quantification of 28S rRNA was used as an internal standard to quantify HCV in total liver RNA, as previously described[46], (threshold of detection: 25 copies/reaction). Briefly, reverse transcription was carried out using oligo(dT) primer (Life Technologies) and Moloney murine leukemia virus reverse transcriptase (Promega) as recommended by the manufacturer. Real-time polymerase chain reactions were performed using the Light CyclerR (Roche Applied Science) and Fast Start DNA Master SYBR Green I kit (Life Science, Roche) according with the manufacturer’s protocol.

Reverse transcription was performed using primers located in the 5’ NCR region of HCV genome, tag-RC1 (5’-GGC CGT CAT GGT GGC GAA TAA GTC TAG CCA TGG CGT TAG TA-3’)[47] and RC21 (5’-CTC CCG GGG CAC TCG CAA GC-3’)[48] for the negative and positive strands, respectively, as described previously[46]. After a denaturation step performed at 70 °C for 8 min, the RNA template was incubated at 48 °C for 5 min in the presence of 200 ng of tag-RC1 primer and 1.25 mmol/L of each deoxynucleoside triphosphate (dNTP) (Promega) in a total volume of 12 μL. Reverse transcription was carried out for 60 min at 60 °C in the presence of 20 U RNaseOutTM (Life Technologies) and 7.5 U ThermoscriptTM reverse transcriptase (Life Technologies), in the buffer recommended by the manufacturer. An additional treatment was applied by adding 1 μL (2U) RNaseH (Life Technologies) for 20 min at 37 °C. The first round of nested PCR was performed with 2 μL of the cDNA obtained in a total volume of 50 μL, containing 3 U Taq polymerase (Promega), 0.5 mmol/L dNTP Mix (Promega), and 0.5 μmol/L RC1 (5’-GTC TAG CCA TGG CGT TAG TA-3’) and RC21 primers for positive-strand amplification, or tag (5’-GGC CGT CAT GGTGGC GAA TAA-3’) and RC21 primers for negative strand amplification. The PCR protocol consisted of 18 cycles of denaturation (94 °C for 1 min), annealing (55 °C for 45 s), and extension (72 °C for 2 min). The cDNA obtained was purified using the Quick-clean kit (Qiagen), according to the manufacturer’s instructions, and 2 μL of the purified product suspended in 10 μL nuclease free water (Promega) were then subjected to real-time PCR. The reaction was carried out using the DNA Fast Start SYBR Green Kit (Life Science, Roche), with LightcyclerTM instruments and technology (Roche Diagnostics). PCR amplifications were performed in a total volume of 20 μL, containing 3 mmol/L MgCl2, 2 μL DNA Master green (Life Science) and 50 ng of the 197 R (5’-CTTTCGCGACCCAACACTAC-3’) and 104 (5’-AGAGCCATAGTGGTCTGCGG-3’) primers[48,49]. The PCR protocol consisted of one step of initial denaturation for 10 min at 94 °C, followed by 40 cycles of denaturation (95 °C for 15 s), annealing (57 °C for 5 s), and extension (72 °C for 8 s). After amplification, the specificity of PCR products was checking by a melting curve analysis.

Western blotting

Western blotting was performed as following. Each liver slice was washed 3 times in PBS, incubated in Laemmli buffer[50] at 100 °C for 10 min. The lysate was passed through a 26 G needle, 10 times and kept at -80 °C. Before electrophoresis in pre-cast sodium dodecyl sulfate polyacrylamide gel 4%-12% (Life Technologies), the samples were incubated at 95 °C for 5 min. After electrophoresis, proteins were transferred to a 0.22 μm Protran membrane BA83 (Schleicher and Schuell) and HCV proteins were detected by Western blotting using mouse monoclonal antibodies to core (C7-50, 1:10000) (Ozyme), to NS5A (1:2000) (Virostat, clone 1877), to LC3 proteins (Sigma-Aldrich) and to glyceraldehyde-3-phosphate dehydrogenase (GAPDH) (Abcam), and to β-actin (Pierce biotechnology). Horseradish peroxidase-conjugated anti-mouse IgG (GeHealthCare Life Sciences) at the dilution of 1:50000 were used as secondary antibodies. The reactions were developed using enhanced chemiluminescence detection reagents (GeHealthCare Life Sciences), followed by exposure to X-OMAT film (GeHealthCare Life Sciences). LC3-II protein expression analysis was performed with Image J software.

Drugs inhibition of HCVсс Сon1/C3 replication and cytotoxicity assays

The HCVcc Con1/C3 inhibition either by pegylated-interferon α-2a (peg-INF) (Roche, Pegasys) or/and ribavirin (RBV) (Schering Plough, Rebetol) or TVR (Janssen-Cilag, Incivo) or BOC (Schering-Plough, Boceprevir) or SOF (Gilead Sciences Intl Ltd, Sofosbuvir) or GNS-396 (Figure 1) (Genoscience Pharma, Marseille, France) or HCQ (Figure 1) (Genoscience Pharma, Marseille, France) or 0.5% dimethylsufoxide (DMSO) (Sigma Aldrich) as a carrier control, and the cytotoxicity assays were performed as following. At day 4 post-infection with HCVcc Con1/C3 the human liver slices were treated by addition of different concentrations of the following drugs: peg-INF or/and RBV or TVR or BOC or SOF or HCQ (0.1, 1, 2.5, 5 μmol/L or a new quinoline derivative, GNS-396 (0.1, 1, 2.5, 5 μmol/L) alone or 0.5% DMSO as a carrier control, to culture medium, twice daily, up to day 10 post-infection. The infectivity (ffu/mL) was measured at day 2, day 4 or day 6 post-treatment depending on the experiment as described[41]. All the experiments were performed in triplicate. The cytoTox 96R Non-Radioactive Cytotoxicity Assay (Promega, G1780) was used to assess the potential cytotoxicity of the drugs. Results of lactate deshydrogenase (LDH) leakage were compared to the carrier control calculated (Figure 2) as described previously[51].

Figure 1
Figure 1 GNS-396 and hydroxychloroquine structures. HCQ: Hydroxychloroquine.
Figure 2
Figure 2 Cytotoxicity assays of the drugs used hydroxychloroquine (A) and GNS-396 (B). Percentages of lactate deshydrogenase (LDH) leakage are relative to DMSO control-treated liver slices. Drugs cytotoxicity is significant at % of control > 120. All experiments were performed on triplicate. Values are expressed as means ± SE, comparisons were performed using the Mann-Whitney rank-sum test (P < 0.001). HCQ: Hydroxychloroquine; DMSO: Dimethylsufoxide.
Evaluation of autophagy modulation and inhibition

Autophagy modulation was evaluated on HeLa cells treated with GNS-396, a new quinolone derivative. For tracking different stages of autophagy the tandem fusion of mRFP and EGFP fused to LC3 make a pH-sensitive sensor (mRFP-EGFP-LC3) that is used to monitor autophagy in live cells[52]. The EGFP tag is acid-sensitive while the mRFP tag is not. The double tagged LC3 can be used to label autophagosomes, amphisomes and autolysosomes. In autophagosomes, both tags emit yellow light. However, the fusion of autophagosomes to acidic lysosomes results in acidic autolysosomes where the green fluorescence from GFP is lost. Subsequently, the red fluorescence from mRFP is lost when the double tagged protein is degraded. The autophagic flux inhibition was shown using a SkBr3 mRFP1-EGFP-LC3 stable breast cancer cell line treated with 100 μmol/L GNS-396 or 100 μmol/L HCQ during 6 h. HCQ was used as a positive control of autophagy inhibition. Cell images were obtained using an epifluorescence microscope (Nikon, Eclipse Ci).

Autophagy inhibition was evaluated on HeLa cells treated with GNS-396, at different drug concentrations (1, 10, 100 μmol/L) during 4 h or 6 h in the presence or absence of bafilomycine A1 (100 nmol/L) (Sigma-Aldrich) added for the last 2 h. Bafilomycin A1 (BafA1) is an autophagy inhibitor as endosomal acidification inhibitor. It is a known inhibitor of the late phase of autophagy. Bafilomycin A1 prevents maturation of autophagic vacuoles by inhibiting fusion between autophagosomes and lysosomes. Bafilomycin A1 acts by inhibiting vacuolar H+ ATPase[53,54]. HCQ was used as a positive control of autophagy inhibition. The LC3-II protein expression in cell lysates was evaluated by Western-blot assay [anti-LC3 antibody (Sigma-Aldrich)] compared to either GAPDH protein expression [anti-GAPDH antibody (Abcam)] or β-actin protein expression [anti-actin antibody (Pierce biotechnology). LC3-II protein expression analysis was performed with Image J software.

Statistical analysis

At different days of the kinetics, the results were obtained from the mean of the three slices culture. Statistical tests were performed using the GraphPad Prism 5.0 software (GraphPad Software, La Jolla, CA, United States). Values are expressed as means ± standard errors of the mean. The data were compared using an unpaired two-tailed student’s t-test or the Mann-Whitney rank-sum test. P-value < 0.05 was considered significant.

RESULTS
Validation of GNS-396, a new quinoline derivative, as inhibitor of autophagy

We evaluated the effect of GNS-396 (Figure 1)[42], a new quinoline derivative, on autophagy by treatment of HeLa cells with various concentrations of GNS-396 during 6 h. HCQ was used as a positive control of autophagy inhibition. The microtubule-associated protein 1A/1B-light chain 3 (LC3) is a soluble ubiquitin-like protein with a molecular mass of approximately 17 kDa that exists ubiquitously in mammalian tissues and cultured cells, as an unconjugated form (LC3-I) or conjugated to autophagosomes membranes (LC3-II: lipidated form). During autophagy, a cytosolic form of LC3 (LC3-I) is conjugated to phosphatidylethanolamine to form LC3-phosphatidylethanolamine conjugate (LC3-II), which is recruited to autophagosomal membranes allowing for the closure of the autophagic vacuole. Autophagosomes fuse with lysosomes to form autolysosomes, and intra-autophagosomal components are degraded by lysosomal hydrolases. At the same time, LC3-II in autolysosomal lumen is degraded. Thus, lysosomal turnover of the autophagosomal marker LC3-II reflects autophagic activity. Analysis of LC3 intracellular expression by Western blotting demonstrated an increase of normalized LC3-II protein expression when HeLa cells were treated with GNS-396, in a dose-dependent manner (Figure 3A), reflecting the accumulation of autophagosomes in cells, and therefore an effective modulation of the autophagy. Consequently, GNS-396 is a dose-dependent autophagy modulator with a magnitude of normalized LC-3 II similar to which achieved with HCQ treatment, a well-known autophagic inhibitor (Figure 3B). Similar results were obtained on Huh7.5.1 cell line (data not shown).

Figure 3
Figure 3 Modulation and inhibition of autophagy by treatment with GNS-396 in HeLa cells. A and B: Autophagy modulation was evaluated using HeLa cells treated either with (A) GNS-396 or (B) HCQ at different concentrations (1, 10 and 100 μmol/L) during 6 h. HCQ was used as a positive control of autophagy modulation. Intracellular expression of proteins LC3 was evaluated by Western-blot assay and normalized for LC3-II; C and D: Autophagy inhibition by treatment with GNS-396 in HeLa cells. Autophagy inhibition was evaluated using HeLa cells treated either with (C) GNS-396 or (D) HCQ at different concentrations (1, 10 and 100 μmol/L) during 4 h in the presence or absence of 100 nmol/L bafilomycine A1. HCQ was used as a positive control of autophagy inhibition. LC3-II intracellular expression was evaluated by Western-blot assay and normalized. HCQ: Hydroxychloroquine; LC3: Microtubule-associated protein 1A/1B-light chain; DMEM: Dulbecco’s modified eagles’s medium; GAPDH: Glyceraldehyde-3-phosphate dehydrogenase; BafA1: Bafilomycin A1.

To evaluate if the observed accumulation of autophagosomes after GNS-396 treatment was a consequence of either a stimulated production of new autophagosomes (in this case, GNS-396 would be an autophagy inducer) or a result of autophagosome clearance blockage (in this case, GNS-396 would be an autophagy inhibitor), HeLa cells were treated with different concentrations of GNS-396 in the absence or presence of a lysosomal protease inhibitor, Bafilomycin A1, that increases lysosomal pH and blocks autophagosome-lysosome fusion (Figure 3C) and LC3 protein levels were measured. HCQ was used as a positive control of autophagy inhibition (Figure 3D). After 4 h exposure of HeLa cells to GNS-396 (100 μmol/L), the accumulation of LC3-II was observed (Figure 3C) which was not enhanced in the presence of BafA1, supporting the idea that GNS-396 inhibits autophagic flux as HCQ (Figure 3D). To confirm that GNS-396 is an autophagy inhibitor, the autophagic flux was monitored by fluorescence microscopy, using the mRFP-EGFP-LC3 tandem-tagged fluorescent protein in SkBr3 mRFP-EGFP-LC3 stable cell line (Figure 4). In green/red merged images, yellow dots (i.e., mRFP+EGFP+) indicate autophagosomes or non-acidic autolysosomes, while red dots (i.e., mRFP+EGFP-) indicate autolysosomes. The autophagy flux is increased when both yellow and red punctua (dots) are increased in cells while the autophagic flux is blocked when only yellow punctua (dots) are increased without an accompanying increase of red punctua in cells. SkBr3 mRFP-EGFP-LC3 stable cell line was treated during 6 h with either GNS-396 (Figure 4C) or HCQ, a well-known autophagic inhibitor (Figure 4B) (100 μmol/L). An accumulation of yellow punctua (dots) corresponding to autophagosomes or non-acidic autolysosomes was noticeable (Figure 4B and C), indicating that GNS-396 blocks the autophagic flux, and may act as lysosomotropic agent as HCQ.

Figure 4
Figure 4 Inhibition of autophagic flux by treatment with GNS-396 in SkBr3 mRFP-EGFP-LC3 stable cell line. Autophagic flux was monitored using the mRFP-EGFP-LC3 tandem-tagged fluorescent protein in SkBr3 mRFP-EGFP-LC3 stable cell line. A: SkBr3 mRFP-EGFP-LC3 stable cell line without any treatment is representative of basal autophagy; SkBr3 mRFP1-EGFP-LC3 stable cell line was treated either with (B) 100 μmol/L HCQ or (C) 100 μmol/L GNS-396 during 6 h. In green/red merged images, yellow punctua (i.e., mRFP+EGFP+) indicate autophagosomes or non-acidic autolysosomes, while red punctua (i.e., mRFP+EGFP) indicate autolysosomes. HCQ is used as a positive control of autophagy inhibition. HCQ: Hydroxychloroquine; DMEM: Dulbecco’s modified eagles’s medium; LC3: Microtubule-associated protein 1A/1B-light chain.
Modulation of autophagy and inhibition of HCV infection in human liver slices model by GNS-396 treatment

The level of LC3 and viral proteins expression were analysed by Western blotting after 1, 4, 6 and 10 d post-infection (Figure 5). HCV infection induced autophagy with an increase of protein LC3-II expression (Figure 5B) as compared to non-infected liver slices (Figure 5A), along with an increase of intracellular expression of the core and NS5A proteins consistent with the previous reports[22,36]. Intracellular expression of the viral proteins was decreased significantly at day 6 post-treatment with HCQ (1 μmol/L) or GNS-396 (1 μmol/L) (Figure 5D) in comparison with HCVcc infected liver slices not treated (Figure 5B). The HCQ- and GNS-396-treatment induced an accumulation of LC3-II protein in HCV infected liver slices treated with 1 μmol/L HCQ or 1 μmol/L GNS-396 (Figure 5D) in comparison either with not infected liver slices treated (Figure 5C) or not (Figure 5A), or with HCV infected liver slices without treatment (Figure 5B). At day 10, the normalized LC3-II protein expression increased when liver slices infected (Figure 5D) or not (Figure 5C) were treated either with GNS-396 (1 μmol/L) or HCQ (1 μmol/L). The GNS-396 and HCQ effects were tested on the de novo viral production of HCVcc Con1 infected liver slices (Figures 6 and 7). At day 4 post-infection, HCVcc Con1 infected liver slices were treated for 6 d with different concentrations either of GNS-396 or HCQ. From day 1 to day 6 post-treatment, the HCV RNA replication (Figure 6A and B) and the infectivity (Figure 7A and B) were inhibited in a dose-dependent manner. The addition of RBV with the new drug GNS-396 showed no significant difference in the viral inhibition (data not shown).

Figure 5
Figure 5 Inhibition of autophagy by treatment with GNS-396 (1 μmol/L) in primary adult human liver slices infected with cell culture-grown hepatitis C virus Con1 (multiplicity of infection = 1). A: Intracellular expression of LC3-I/LC3-II proteins in non-infected liver slices without treatment; B: Intracellular expression of LC3-I/LC3-II proteins and the normalization of intracellular protein LC3-II expression, for 10 d in non-infected liver slices with treatment either by GNS-396 (1 μmol/L) (G) or HCQ (1 μmol/L) (H) or without treatment (D: day); C: Expression of LC3-I/LC3-II proteins and HCV core and NS5A proteins in HCVcc Con1 infected liver slices either without treatment or (D) either with treatment by GNS-396 (1 μmol/L) or HCQ (1 μmol/L) and the normalization of Intracellular protein LC3-II expression for 10 d in HCVcc Con1 infected liver slices with treatment either by GNS-396 (1 μmol/L) (G) or HCQ (1 μmol/L) (H) or without treatment (D: day). LC3: Microtubule-associated protein 1A/1B-light chain; HCVcc: Cell culture-grown hepatitis C virus; NS5A: HCV nonstructural protein 5A.
Figure 6
Figure 6 Inhibition of hepatitis C virus RNA replication by treatment either with GNS-396 or hydroxychloroquine in a dose-dependent manner in primary adult human cell culture-grown hepatitis C virus Con1 infected liver slices. Human liver slices were infected overnight with HCVcc Con1 (MOI = 0.1). The supernatant is then removed, the human liver slices washed and cultured. The liver slices and culture supernatants were collected different times post-infection. At day 4 post-infection, the liver slices were treated with increasing concentrations either of GNS-396 (0.01, 0.1, 1, 5 μmol/L) (A, B) or HCQ (C, D) for 6 d (black arrow: Start of the treatment either with GNS-396 or HCQ). Human HCVcc Con1 infected liver slices were lysed to evaluate intracellular levels of positive- and negative-strand HCV RNA by specific strand RT-qPCR at 1, 4, 6, 8, 10 d post-infection. The results were compared using the two-paired Student’s test. Values are expressed as means ± standard errors: (A) HCV RNA replication by treatment with GNS-396: Positive strand (black line), P < 0.03; negative strand (grey line), P < 0.013, GNS-396 0.01 μmol/L (red line), P < 0.04; GNS-396 0.1 μmol/L (green line), P < 0.05; GNS-396 1 μmol/L (pink line), P < 0.05; GNS-396 5 μmol/L (blue line), P < 0.05; (C) HCV RNA replication by treatment with HCQ: Positive strand (black line), P < 0.03; negative strand (grey line), P < 0.015; HCQ 0.1 µmol/L (red line), P < 0.0001; HCQ 1 μmol/L (green line), P < 0.0001; HCQ 2.5 μmol/L (pink line), P < 0.01; HCQ 5 μmol/L (blue line), P < 0.03. The detection of negative strand of HCV RNA evidences active replication as well as the increase overtime of both positive and negative strands of HCV RNA; B: Inhibition of HCV replication (%) with GNS-396 treatment P < 0.0038; D: Inhibition of HCV replication (%) with HCQ treatment P < 0.0013. The replication was significantly inhibited in a dose-dependent manner in presence of increasing concentrations either of GNS-396 (B) or HCQ (D) for 6 d. HCVcc: Cell culture-grown hepatitis C virus; HCQ: Hydroxychloroquine; qRT-PCR: Quantitative technique consisting of reverse transcription followed by real-time polymerase chain reaction; MOI: Multiplicity of infection; CTRL: Control-treated liver slices.
Figure 7
Figure 7 Dose-dependent inhibition of primary-culture-derived virus infectivity in primary adult human cell culture–grown hepatitis C virus Con1 infected liver slices by treatment either with GNS-396 (A) or hydroxychloroquine (B). Kinetics of infectivity of culture supernatants from human liver slices infected by HCV Con1 (MOI = 0.1) and treated either GNS-396 (A) or with HCQ (B) or at day 4 post-infection for 6 d. A: Con1 (black line), P < 0.0001; GNS-396 0.01 μmol/L (green line), P < 0.0003; GNS-396 0.1 μmol/L (blue line), P < 0.019; GNS-396 1 μmol/L (red line), P < 0.05; GNS-396 5 μmol/L (purple line), P < 0.05; B: Con1 (black line), P < 0.0001; HCQ 0.1 μmol/L (red line), P < 0.0001; HCQ 1 μmol/L (green line), P < 0.0001; HCQ 2.5 μmol/L (red line), P < 0.0001; HCQ 5 μmol/L (purple line), P < 0.0003. Each curve represented the average of 2 independent infections performed in triplicate from 2 different donors. Values are expressed as means ± SE. The results were compared using the two-paired student’s test. HCV: Hepatitis C virus; HCQ: Hydroxychloroquine; MOI: Multiplicity of infection; CTRL: Control-treated liver slices.
EC50 analysis of HCV replication with GNS-396 treatment compared to that of validated antiviral drugs

The ability of various concentrations of different antiviral drugs to inhibit HCV replication was measured by detecting the replication of negative strands HCV RNA (Figure 6A and C) (Table 1). The calculated EC50 of different antiviral drugs is listed as Table 1 and compared to GNS-396. In summary, our model confirms that the antiviral activity of triple therapy was higher than that of the dual therapy by PR as extensively reported in clinical trials[5,6]. The new quinoline derivative GNS-396 has about 10-fold lower EC50 than HCQ (0.158 μmol/L as compared to 1.17 μmol/L) (Figure 6B and D). No significant cytotoxic effects were observed when evaluated by the lactate dehydrogenase leakage (LDH) assays (Figure 2A and B). A 50% cytotoxic concentration (CC50 value) of 25 μmol/L was obtained for GNS-396 in the human liver slices culture at day 6 post-treatment. Similar CC50 values were obtained in proliferating Huh-7-5-1 replicon cells (23 μmol/L).

Table 1 Inhibition of hepatitis C virus infectivity and 50% effective concentration of hepatitis C virus replication with direct active antivirals and autophagy inhibitors.
Drugs1Infectivity inhibition (%)Average (SD)2Replication inhibition (EC50)3Average (SD)4CC50 (SD)5
INF (2.6 to 260 nmol/L)6Up to 95%517 ng/mL7.240 ng/mL (± 4)
RBV (1 to 100 μmol/L)3% to 37%3146 μmol/L13400 μmol/L (± 21)
7TVR (0.01 to 50 μmol/L)62% to 89%40.395 μmol/L0.03840 μmol/L (± 3)
7BOC (0.01 to 50 μmol/L)61% to 95%50.417 μmol/L0.02441 μmol/L (± 5)
7SOF (0.01 to 50 μmol/L)75% to 95%40.147 μmol/L0.01723 μmol/L (± 2)
7HCQ (0.1 to 50 μmol/L)25% to 94%41.17 μmol/L0.02327 μmol/L (± 2)
7GNS-396 (0.01 to 5 μmol/L)6% to 93%30.158 μmol/L0.01425 μmol/L (± 2)
INF (2.6 to 260 nmol/L)/RBV 100 μmol/LUp to 98%610 ng/mL3.143 ng/mL (± 4)
7HCQ (0.1 to 5 μmol/L)/RBV 50 μmol/L27% to 85%20.456 μmol/L0.04431 μmol/L (± 3)
7GNS-396 (0.01 to 5 μmol/L)/RBV 100 μmol/L9% to 94%20.157 μmol/L0.01226 μmol/L (± 2)
7TVR (0.01 to 50 μmol/L)/RBV 100 μmol/LUp to 98%30.310 μmol/L0.02949 μmol/L (± 3)
7BOC (0.01 to 50 μmol/L)/RBV 100 μmol/LUp to 95%20.370 μmol/L0.03548 μmol/L (± 4)
7SOF (0.01 to 50 μmol/L)/RBV 100 μmol/LUp to 100%20.080 μmol/L0.02817 μmol/L (± 5)
7TVR 1 μmol/L/7BOC 1 μmol/LUp to 89%20.410 μmol/L0.03950 μmol/L (± 3)
7TVR 1 μmol/L/INF 26 nmol/L/RBV 100 μmol/LUp to 99%30.315 μmol/L0.03144 μmol/L (± 4)
7BOC 1 μmol/L/INF 26 nmol/L/RBV 100 μmol/LUp to 97%20.350 μmol/L0.03347 μmol/L (± 3)
7SOF 1 μmol/L/INF 26 nmol/L/RBV 100 μmol/LUp to 100%30.055 μmol/L0.02918 μmol/L (± 3)
DISCUSSION

Our study evidenced that: (1) the ex vivo model of human liver slices HCVcc Con1 infection may be efficiently used for the assay of the antiviral potency of a new inhibitor (GNS-396 compared to HCQ) which interfered with autophagy; and (2) GNS-396 was a potent autophagy inhibitor, acting as “lysosomotropic agent” which was able to inhibit HCV replication in primary human adult HCVcc infected liver slices culture.

The establishment of the ex vivo model (feasibility, rapidity, specificity, potency) was already described in detail in 2012[41] with comparison between primary human hepatocytes, Huh-7.5.1 cell line. The Huh-7 cell system has several limitations that includes the inability to study the effects of pharmacologic inhibitors targeting the non-structural proteins of the most prevalent and problematic viral strains (e.g., genotypes 1a and 1b). Moreover, the study of virus/host cell interactions is limited since the permissive cell lines are transformed and poorly differentiated. Firstly, the human liver slices culture maintains the original three-dimensional structure of the liver that allows cell crosstalk: The extra-cellular matrix and Kupffer cells essential for the normal function of the hepatocytes and the lobular structure. Secondly, the gene expression profiles in liver tissue slices were similar to that of the in vivo gene expression. Thirdly, primary hepatocytes preparations undergo treatment with collagenase (a treatment might have a negative effect on integrity of the proteins repertoire on the cell surface), but not the liver slices. Noteworthy, using established procedures, the tissue slices remained viable for, at least 10 d as it was shown by the secretion of albumin and urea. Moreover, the Huh-7 cell infection with primary isolates from patients are not very efficient. The infection of adult human liver slices culture allowed to achieve the robust replication of HCVcc genotype 2a, 1a and 1b genome and to obtain infectivity titers above 105 ffu/mL. In addition, we reported robust and productive infection using human primary isolates HCV genotype 1b. Stem cell-derived hepatocytes (hESC-Heps) displayed equivalence to primary adult hepatocytes. HESC-Heps were capable of supporting the full HCV life cycle (JFH1), including the release of infectious virions. Although supportive, hESC-Hep viral infection levels were not as great as those observed in Huh7 cells. Up to now, the hESC-Heps were not infected with primary isolates[55]. Currently, we are establishing a culture of liver slices for 21 d, which allows us to follow the variation of different parameters and in particular, complete inhibition of viral production (data not shown).

Previous studies have reported that autophagy proteins are required to initiate HCV replication and translation[28,30-36]. Some data demonstrated that the suppression of LC3 protein lipidation, a necessary step for the formation of autophagosomes could also suppress HCV replication[30]. CQ is a well-known autophagic inhibitor which is often used as an anti-malarial agent. HCQ is a “lysosomotropic” weak base that raises the lysosomal pH quickly[37]. Furthermore, many studies have reported the antiviral effect of CQ on other positive strand RNA viruses, such as polioviruses, coxsackieviruses, dengue viruses, coronaviruses (SARS-CoV virus)[24-29], HIV-1[56]. In our study, we demonstrated the antiviral effect of HCQ and the new quinoline derivative GNS-396 on HCVcc replication in a dose - dependent manner. Compared to the treatment with HCQ alone, HCQ inhibition was more pronounced in combination with RBV or with other direct antivirals, suggesting a synergistic effect of the combined drugs on HCVcc infection in human liver slices. This result is consistent with a previous study which demonstrated the antiviral effect of CQ in combination with peg-IFN in HCV infected Huh-7 cell line[33]. Similarly, on Huh-7 cells infected with HCVpp (genotype 1a and 3a), it has been shown that CQ reduced by 50% virus infectivity at 50 μmol/L concentration, when the antiviral effect was tested[57]. Recently, ferroquine (FQ), an antimalarial ferrocenic analog of CQ, has been described as a novel inhibitor of HCV. FQ potently inhibited HCV infection of hepatoma cell lines[58]. Compared to these investigations, our study using the quinoline derivative GNS-396, revealed an inhibition of the virus infectivity up to 93% respectively at day 6 post-treatment with lower drug amounts (EC50 = 0.158 μmol/L). This demonstrates that GNS-396 is a stronger antiviral than HCQ (EC50 = 1.17 μmol/L). EC50 is a measure of the effectiveness of the drug in inhibiting the biochemical function. In our study, we evaluated the EC50 of HCV replication at day 6 post-treatment. The lower EC50 value indicates the greater potency of inhibiting HCV replication. As shown in Table 1, the infectivity inhibition, consistent with the inhibition of HCV replication, demonstrated that the new drug evaluated in the human HCV infected liver slices culture model, had a potent antiviral effect compared to the well-known established antivirals. In combination with the other well established drugs like DAA or inhibitors of other host targets (cyclophilin), quinoline derivatives could be additional therapeutic options for HCV infected patients.

In conclusion, this study demonstrated the relevance of the human HCV infected liver slices culture in preclinical studies of the new anti-viral drugs. New host-targeted therapies inhibiting autophagy (GNS-396, HCQ) have demonstrated significant efficiency and additive activity in inhibiting HCV replication. The ex vivo model of culture of human HCV infected liver slices might allow further evaluation of the efficacy of new antiviral drugs in single or in combined therapy and their potential toxicity in particular for patients ”difficult to treat”. Moreover, the infection of human liver slices culture with primary viral isolates from patients that we succeed to establish[41], should allow highlighting the potential of early emergence of drug resistant viral variants during the anti-viral treatments.

ACKNOWLEDGMENTS

The authors are grateful to Professor Francis V Chisari, for the kind gift of Huh-7.5.1 cells and to Professor Ralf Bartenschlager for kindly providing the chimeric pFK-Con1/C3 plasmid. We are also grateful to the members of the Departments of Digestive Surgery in the Groupe Hospitalier La Pitié Salpétrière and Cochin-Hôtel Dieu as to the members of the Department of Hepatology in Groupe Hospitalier Cochin-Hôtel Dieu, APHP, Paris, France, for the technical assistance. We are deeply indebted to Doctor Vladimir A. Morozov and Doctor Matthew Albert for the critical reading of the manuscript.

COMMENTS
Background

Hepatitis C virus (HCV) infection (or spread) is a serious public health challenge counting approximately 170 million people that are chronically infected worldwide. Host antiviral therapy is an additional option for the treatment of HCV infection.

Research frontiers

Interferon-free treatments with new direct acting antivirals are expected to cure more than 90% of HCV-infected patients. But they are not available in all the countries. At the present time, triple therapy combining pegylated interferon-α 2a or b and ribavirin with NS3 protease inhibitors (telaprevir or boceprevir) is going to remain the main treatment for HCV patients. That is why it appears important to continue research in limiting virus replication and the autophagy inhibition could be a new additional pathway because of recent evidences obtained regarding to an increased autophagic response in the liver of chronically HCV infected patients.

Innovations and breakthroughs

This is the first study evaluating a new autophagy inhibitor as antiviral that could inhibit HCV infection in a dose-dependent manner without cytotoxic effect using the relevant ex vivo model of the human liver slices culture.

Applications

This study highlight the relevance of the ex vivo model of the human HCV infected liver slices culture in preclinical studies of the new anti-viral drugs in single or in combined therapy and their potential toxicity in particular for patients “difficult to treat”. Moreover, the infection of human liver slices culture with primary viral isolates from patients that the authors succeed to establish, should allow highlighting the potential of early emergence of drug resistant viral variants during the anti-viral treatments.

Terminology

Autophagy is a catabolic process which degrades a cellular own component through the lysosomal machinery. It has been shown that autophagy is activated during virus and bacterial infection and that some viruses can use the autophagy system to facilitate their own replication.

Peer-review

The manuscript is clear and comprehensive.

Footnotes

Manuscript source: Invited manuscript

Specialty Type: Gastroenterology and Hepatology

Country of Origin: France

Peer-Review Report Classification

Grade A (Excellent): 0

Grade B (Very good): B

Grade C (Good): C

Grade D (Fair): 0

Grade E (Poor): 0

P- Reviewer: Jin B, Irato P S- Editor: Ji FF L- Editor: A E- Editor: Li D

References
1.  Szabó E, Lotz G, Páska C, Kiss A, Schaff Z. Viral hepatitis: new data on hepatitis C infection. Pathol Oncol Res. 2003;9:215-221.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 67]  [Cited by in F6Publishing: 61]  [Article Influence: 2.9]  [Reference Citation Analysis (0)]
2.  Corouge M, Pol S. New treatments for chronic hepatitis C virus infection. Med Mal Infect. 2011;41:579-587.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 6]  [Cited by in F6Publishing: 7]  [Article Influence: 0.5]  [Reference Citation Analysis (0)]
3.  Mallet V, Gilgenkrantz H, Serpaggi J, Verkarre V, Vallet-Pichard A, Fontaine H, Pol S. Brief communication: the relationship of regression of cirrhosis to outcome in chronic hepatitis C. Ann Intern Med. 2008;149:399-403.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 235]  [Cited by in F6Publishing: 245]  [Article Influence: 15.3]  [Reference Citation Analysis (0)]
4.  Ashfaq UA, Javed T, Rehman S, Nawaz Z, Riazuddin S. An overview of HCV molecular biology, replication and immune responses. Virol J. 2011;8:161.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 104]  [Cited by in F6Publishing: 115]  [Article Influence: 8.8]  [Reference Citation Analysis (0)]
5.  López-Labrador FX. Hepatitis C Virus NS3/4A Protease Inhibitors. Recent Pat Antiinfect Drug Discov. 2008;3:157-167.  [PubMed]  [DOI]  [Cited in This Article: ]
6.  Asselah T, Marcellin P. New direct-acting antivirals’ combination for the treatment of chronic hepatitis C. Liver Int. 2011;31 Suppl 1:68-77.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 118]  [Cited by in F6Publishing: 123]  [Article Influence: 9.5]  [Reference Citation Analysis (0)]
7.  Poordad F, McCone J, Bacon BR, Bruno S, Manns MP, Sulkowski MS, Jacobson IM, Reddy KR, Goodman ZD, Boparai N. Boceprevir for untreated chronic HCV genotype 1 infection. N Engl J Med. 2011;364:1195-1206.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1948]  [Cited by in F6Publishing: 1951]  [Article Influence: 150.1]  [Reference Citation Analysis (0)]
8.  Bacon BR, Gordon SC, Lawitz E, Marcellin P, Vierling JM, Zeuzem S, Poordad F, Goodman ZD, Sings HL, Boparai N. Boceprevir for previously treated chronic HCV genotype 1 infection. N Engl J Med. 2011;364:1207-1217.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1287]  [Cited by in F6Publishing: 1288]  [Article Influence: 99.1]  [Reference Citation Analysis (0)]
9.  Jacobson IM, McHutchison JG, Dusheiko G, Di Bisceglie AM, Reddy KR, Bzowej NH, Marcellin P, Muir AJ, Ferenci P, Flisiak R. Telaprevir for previously untreated chronic hepatitis C virus infection. N Engl J Med. 2011;364:2405-2416.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1866]  [Cited by in F6Publishing: 1835]  [Article Influence: 141.2]  [Reference Citation Analysis (0)]
10.  Zeuzem S, Asselah T, Angus P, Zarski JP, Larrey D, Müllhaupt B, Gane E, Schuchmann M, Lohse A, Pol S. Efficacy of the protease inhibitor BI 201335, polymerase inhibitor BI 207127, and ribavirin in patients with chronic HCV infection. Gastroenterology. 2011;141:2047-2055; quiz e14.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 116]  [Cited by in F6Publishing: 111]  [Article Influence: 8.5]  [Reference Citation Analysis (0)]
11.  Jacobson IM, Gordon SC, Kowdley KV, Yoshida EM, Rodriguez-Torres M, Sulkowski MS, Shiffman ML, Lawitz E, Everson G, Bennett M. Sofosbuvir for hepatitis C genotype 2 or 3 in patients without treatment options. N Engl J Med. 2013;368:1867-1877.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 846]  [Cited by in F6Publishing: 820]  [Article Influence: 74.5]  [Reference Citation Analysis (0)]
12.  Lawitz E, Mangia A, Wyles D, Rodriguez-Torres M, Hassanein T, Gordon SC, Schultz M, Davis MN, Kayali Z, Reddy KR. Sofosbuvir for previously untreated chronic hepatitis C infection. N Engl J Med. 2013;368:1878-1887.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1322]  [Cited by in F6Publishing: 1287]  [Article Influence: 117.0]  [Reference Citation Analysis (0)]
13.  Sulkowski MS, Gardiner DF, Rodriguez-Torres M, Reddy KR, Hassanein T, Jacobson I, Lawitz E, Lok AS, Hinestrosa F, Thuluvath PJ. Daclatasvir plus sofosbuvir for previously treated or untreated chronic HCV infection. N Engl J Med. 2014;370:211-221.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 933]  [Cited by in F6Publishing: 889]  [Article Influence: 88.9]  [Reference Citation Analysis (0)]
14.  Poordad F, Schiff ER, Vierling JM, Landis C, Fontana RJ, Yang R, McPhee F, Hughes EA, Noviello S, Swenson ES. Daclatasvir with sofosbuvir and ribavirin for hepatitis C virus infection with advanced cirrhosis or post-liver transplantation recurrence. Hepatology. 2016;63:1493-1505.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 348]  [Cited by in F6Publishing: 341]  [Article Influence: 42.6]  [Reference Citation Analysis (0)]
15.  Hopkins S, DiMassimo B, Rusnak P, Heuman D, Lalezari J, Sluder A, Scorneaux B, Mosier S, Kowalczyk P, Ribeill Y. The cyclophilin inhibitor SCY-635 suppresses viral replication and induces endogenous interferons in patients with chronic HCV genotype 1 infection. J Hepatol. 2012;57:47-54.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 74]  [Cited by in F6Publishing: 72]  [Article Influence: 6.0]  [Reference Citation Analysis (0)]
16.  Schaefer EA, Chung RT. Anti-hepatitis C virus drugs in development. Gastroenterology. 2012;142:1340-1350.e1.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 66]  [Cited by in F6Publishing: 72]  [Article Influence: 6.0]  [Reference Citation Analysis (0)]
17.  Hill A, Khoo S, Fortunak J, Simmons B, Ford N. Minimum costs for producing hepatitis C direct-acting antivirals for use in large-scale treatment access programs in developing countries. Clin Infect Dis. 2014;58:928-936.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 155]  [Cited by in F6Publishing: 159]  [Article Influence: 15.9]  [Reference Citation Analysis (0)]
18.  Deuffic-Burban S, Schwarzinger M, Obach D, Mallet V, Pol S, Pageaux GP, Canva V, Deltenre P, Roudot-Thoraval F, Larrey D. Should we await IFN-free regimens to treat HCV genotype 1 treatment-naive patients? A cost-effectiveness analysis (ANRS 95141). J Hepatol. 2014;61:7-14.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 49]  [Cited by in F6Publishing: 53]  [Article Influence: 5.3]  [Reference Citation Analysis (0)]
19.  European Association for the Study of the Liver. EASL Recommendations on Treatment of Hepatitis C 2015. J Hepatol. 2015;63:199-236.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 877]  [Cited by in F6Publishing: 890]  [Article Influence: 98.9]  [Reference Citation Analysis (0)]
20.  AASLD/IDSA HCV Guidance Panel. Hepatitis C guidance: AASLD-IDSA recommendations for testing, managing, and treating adults infected with hepatitis C virus. Hepatology. 2015;62:932-954.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 912]  [Cited by in F6Publishing: 961]  [Article Influence: 106.8]  [Reference Citation Analysis (0)]
21.  Zopf S, Kremer AE, Neurath MF, Siebler J. Advances in hepatitis C therapy: What is the current state - what come’s next? World J Hepatol. 2016;8:139-147.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 54]  [Cited by in F6Publishing: 47]  [Article Influence: 5.9]  [Reference Citation Analysis (0)]
22.  Rautou PE, Cazals-Hatem D, Feldmann G, Mansouri A, Grodet A, Barge S, Martinot-Peignoux M, Duces A, Bièche I, Lebrec D. Changes in autophagic response in patients with chronic hepatitis C virus infection. Am J Pathol. 2011;178:2708-2715.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 50]  [Cited by in F6Publishing: 48]  [Article Influence: 3.7]  [Reference Citation Analysis (0)]
23.  Shintani T, Klionsky DJ. Autophagy in health and disease: a double-edged sword. Science. 2004;306:990-995.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2022]  [Cited by in F6Publishing: 1988]  [Article Influence: 99.4]  [Reference Citation Analysis (0)]
24.  Orvedahl A, Levine B. Eating the enemy within: autophagy in infectious diseases. Cell Death Differ. 2009;16:57-69.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 155]  [Cited by in F6Publishing: 169]  [Article Influence: 10.6]  [Reference Citation Analysis (0)]
25.  Jackson WT, Giddings TH, Taylor MP, Mulinyawe S, Rabinovitch M, Kopito RR, Kirkegaard K. Subversion of cellular autophagosomal machinery by RNA viruses. PLoS Biol. 2005;3:e156.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 621]  [Cited by in F6Publishing: 640]  [Article Influence: 33.7]  [Reference Citation Analysis (0)]
26.  Lee YR, Lei HY, Liu MT, Wang JR, Chen SH, Jiang-Shieh YF, Lin YS, Yeh TM, Liu CC, Liu HS. Autophagic machinery activated by dengue virus enhances virus replication. Virology. 2008;374:240-248.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 287]  [Cited by in F6Publishing: 285]  [Article Influence: 17.8]  [Reference Citation Analysis (0)]
27.  Wong J, Zhang J, Si X, Gao G, Mao I, McManus BM, Luo H. Autophagosome supports coxsackievirus B3 replication in host cells. J Virol. 2008;82:9143-9153.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 290]  [Cited by in F6Publishing: 296]  [Article Influence: 18.5]  [Reference Citation Analysis (0)]
28.  Dreux M, Chisari FV. Viruses and the autophagy machinery. Cell Cycle. 2010;9:1295-1307.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 140]  [Cited by in F6Publishing: 153]  [Article Influence: 11.8]  [Reference Citation Analysis (0)]
29.  Silva LM, Jung JU. Modulation of the autophagy pathway by human tumor viruses. Semin Cancer Biol. 2013;23:323-328.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 42]  [Cited by in F6Publishing: 43]  [Article Influence: 3.9]  [Reference Citation Analysis (0)]
30.  Sir D, Chen WL, Choi J, Wakita T, Yen TS, Ou JH. Induction of incomplete autophagic response by hepatitis C virus via the unfolded protein response. Hepatology. 2008;48:1054-1061.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 284]  [Cited by in F6Publishing: 285]  [Article Influence: 17.8]  [Reference Citation Analysis (0)]
31.  Dreux M, Gastaminza P, Wieland SF, Chisari FV. The autophagy machinery is required to initiate hepatitis C virus replication. Proc Natl Acad Sci USA. 2009;106:14046-14051.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 355]  [Cited by in F6Publishing: 377]  [Article Influence: 25.1]  [Reference Citation Analysis (0)]
32.  Dreux M, Chisari FV. Impact of the autophagy machinery on hepatitis C virus infection. Viruses. 2011;3:1342-1357.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 42]  [Cited by in F6Publishing: 44]  [Article Influence: 3.4]  [Reference Citation Analysis (0)]
33.  Mohl BP, Tedbury PR, Griffin S, Harris M. Hepatitis C virus-induced autophagy is independent of the unfolded protein response. J Virol. 2012;86:10724-10732.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 44]  [Cited by in F6Publishing: 44]  [Article Influence: 3.7]  [Reference Citation Analysis (0)]
34.  Shrivastava S, Bhanja Chowdhury J, Steele R, Ray R, Ray RB. Hepatitis C virus upregulates Beclin1 for induction of autophagy and activates mTOR signaling. J Virol. 2012;86:8705-8712.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 125]  [Cited by in F6Publishing: 129]  [Article Influence: 10.8]  [Reference Citation Analysis (0)]
35.  Huang H, Kang R, Wang J, Luo G, Yang W, Zhao Z. Hepatitis C virus inhibits AKT-tuberous sclerosis complex (TSC), the mechanistic target of rapamycin (MTOR) pathway, through endoplasmic reticulum stress to induce autophagy. Autophagy. 2013;9:175-195.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 98]  [Cited by in F6Publishing: 111]  [Article Influence: 9.3]  [Reference Citation Analysis (0)]
36.  Sir D, Kuo CF, Tian Y, Liu HM, Huang EJ, Jung JU, Machida K, Ou JH. Replication of hepatitis C virus RNA on autophagosomal membranes. J Biol Chem. 2012;287:18036-18043.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 135]  [Cited by in F6Publishing: 131]  [Article Influence: 10.9]  [Reference Citation Analysis (0)]
37.  Savarino A, Boelaert JR, Cassone A, Majori G, Cauda R. Effects of chloroquine on viral infections: an old drug against today’s diseases? Lancet Infect Dis. 2003;3:722-727.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 840]  [Cited by in F6Publishing: 867]  [Article Influence: 41.3]  [Reference Citation Analysis (0)]
38.  Vincent MJ, Bergeron E, Benjannet S, Erickson BR, Rollin PE, Ksiazek TG, Seidah NG, Nichol ST. Chloroquine is a potent inhibitor of SARS coronavirus infection and spread. Virol J. 2005;2:69.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1198]  [Cited by in F6Publishing: 1186]  [Article Influence: 62.4]  [Reference Citation Analysis (0)]
39.  Rolain JM, Colson P, Raoult D. Recycling of chloroquine and its hydroxyl analogue to face bacterial, fungal and viral infections in the 21st century. Int J Antimicrob Agents. 2007;30:297-308.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 261]  [Cited by in F6Publishing: 286]  [Article Influence: 16.8]  [Reference Citation Analysis (0)]
40.  Mizui T, Yamashina S, Tanida I, Takei Y, Ueno T, Sakamoto N, Ikejima K, Kitamura T, Enomoto N, Sakai T. Inhibition of hepatitis C virus replication by chloroquine targeting virus-associated autophagy. J Gastroenterol. 2010;45:195-203.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 91]  [Cited by in F6Publishing: 96]  [Article Influence: 6.9]  [Reference Citation Analysis (0)]
41.  Lagaye S, Shen H, Saunier B, Nascimbeni M, Gaston J, Bourdoncle P, Hannoun L, Massault PP, Vallet-Pichard A, Mallet V. Efficient replication of primary or culture hepatitis C virus isolates in human liver slices: a relevant ex vivo model of liver infection. Hepatology. 2012;56:861-872.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 36]  [Cited by in F6Publishing: 35]  [Article Influence: 2.9]  [Reference Citation Analysis (0)]
42.  Halfon P, Nallet J, Petit SJ, Bouzidi M, Joly F, Camus C, Benech P, Dubuisson , Courcambeck J, Wychowski C. Chloroquine and related compounds are Inhibitors of Hepatitis C Virus RNA-by inhibiting autophagy proteolysis. Hepatology. 2010;52:807.  [PubMed]  [DOI]  [Cited in This Article: ]
43.  Olinga P, Groothuis GM.  Use of human tissue slices in drug targeting research. Drug targeting. Organ-specific strategies. Series: Methods and Principles in Medicinal Chemistry. G Molema, DKF Meijer, editors series, Mannhold R, Kubinyi H, Timmerman H, eds Weinheim, Germany: Wiley-VCH 2001; 309-331.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 7]  [Cited by in F6Publishing: 7]  [Article Influence: 0.3]  [Reference Citation Analysis (0)]
44.  Pietschmann T, Kaul A, Koutsoudakis G, Shavinskaya A, Kallis S, Steinmann E, Abid K, Negro F, Dreux M, Cosset FL. Construction and characterization of infectious intragenotypic and intergenotypic hepatitis C virus chimeras. Proc Natl Acad Sci USA. 2006;103:7408-7413.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 602]  [Cited by in F6Publishing: 593]  [Article Influence: 32.9]  [Reference Citation Analysis (0)]
45.  Zhong J, Gastaminza P, Cheng G, Kapadia S, Kato T, Burton DR, Wieland SF, Uprichard SL, Wakita T, Chisari FV. Robust hepatitis C virus infection in vitro. Proc Natl Acad Sci USA. 2005;102:9294-9299.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1427]  [Cited by in F6Publishing: 1441]  [Article Influence: 75.8]  [Reference Citation Analysis (0)]
46.  Carrière M, Pène V, Breiman A, Conti F, Chouzenoux S, Meurs E, Andrieu M, Jaffray P, Grira L, Soubrane O. A novel, sensitive, and specific RT-PCR technique for quantitation of hepatitis C virus replication. J Med Virol. 2007;79:155-160.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 27]  [Cited by in F6Publishing: 27]  [Article Influence: 1.6]  [Reference Citation Analysis (0)]
47.  Komurian-Pradel F, Perret M, Deiman B, Sodoyer M, Lotteau V, Paranhos-Baccalà G, André P. Strand specific quantitative real-time PCR to study replication of hepatitis C virus genome. J Virol Methods. 2004;116:103-106.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 69]  [Cited by in F6Publishing: 69]  [Article Influence: 3.5]  [Reference Citation Analysis (0)]
48.  Besnard NC, Andre PM. Automated quantitative determination of hepatitis C virus viremia by reverse transcription-PCR. J Clin Microbiol. 1994;32:1887-1893.  [PubMed]  [DOI]  [Cited in This Article: ]
49.  Nozaki A, Kato N. Quantitative method of intracellular hepatitis C virus RNA using LightCycler PCR. Acta Med Okayama. 2002;56:107-110.  [PubMed]  [DOI]  [Cited in This Article: ]
50.  Laemmli UK. Cleavage of structural proteins during the assembly of the head of bacteriophage T4. Nature. 1970;227:680-685.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 185380]  [Cited by in F6Publishing: 187478]  [Article Influence: 3471.8]  [Reference Citation Analysis (0)]
51.  Braydich-Stolle L, Hussain S, Schlager JJ, Hofmann MC. In vitro cytotoxicity of nanoparticles in mammalian germline stem cells. Toxicol Sci. 2005;88:412-419.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 915]  [Cited by in F6Publishing: 714]  [Article Influence: 37.6]  [Reference Citation Analysis (0)]
52.  Kimura M, Taniguchi M, Mikami Y, Masuda T, Yoshida T, Mishina M, Shimizu T. Identification and characterization of zebrafish semaphorin 6D. Biochem Biophys Res Commun. 2007;363:762-768.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 8]  [Cited by in F6Publishing: 7]  [Article Influence: 0.4]  [Reference Citation Analysis (0)]
53.  Klionsky DJ, Abeliovich H, Agostinis P, Agrawal DK, Aliev G, Askew DS, Baba M, Baehrecke EH, Bahr BA, Ballabio A. Guidelines for the use and interpretation of assays for monitoring autophagy in higher eukaryotes. Autophagy. 2008;4:151-175.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1743]  [Cited by in F6Publishing: 1821]  [Article Influence: 113.8]  [Reference Citation Analysis (0)]
54.  Yamamoto A, Tagawa Y, Yoshimori T, Moriyama Y, Masaki R, Tashiro Y. Bafilomycin A1 prevents maturation of autophagic vacuoles by inhibiting fusion between autophagosomes and lysosomes in rat hepatoma cell line, H-4-II-E cells. Cell Struct Funct. 1998;23:33-42.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1015]  [Cited by in F6Publishing: 1055]  [Article Influence: 40.6]  [Reference Citation Analysis (0)]
55.  Zhou X, Sun P, Lucendo-Villarin B, Angus AG, Szkolnicka D, Cameron K, Farnworth SL, Patel AH, Hay DC. Modulating innate immunity improves hepatitis C virus infection and replication in stem cell-derived hepatocytes. Stem Cell Reports. 2014;3:204-214.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 40]  [Cited by in F6Publishing: 39]  [Article Influence: 3.9]  [Reference Citation Analysis (0)]
56.  Savarino A, Gennero L, Chen HC, Serrano D, Malavasi F, Boelaert JR, Sperber K. Anti-HIV effects of chloroquine: mechanisms of inhibition and spectrum of activity. AIDS. 2001;15:2221-2229.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 91]  [Cited by in F6Publishing: 94]  [Article Influence: 4.1]  [Reference Citation Analysis (0)]
57.  Ashfaq UA, Javed T, Rehman S, Nawaz Z, Riazuddin S. Lysosomotropic agents as HCV entry inhibitors. Virol J. 2011;8:163.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 44]  [Cited by in F6Publishing: 43]  [Article Influence: 3.3]  [Reference Citation Analysis (0)]
58.  Vausselin T, Calland N, Belouzard S, Descamps V, Douam F, Helle F, François C, Lavillette D, Duverlie G, Wahid A. The antimalarial ferroquine is an inhibitor of hepatitis C virus. Hepatology. 2013;58:86-97.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 40]  [Cited by in F6Publishing: 42]  [Article Influence: 3.8]  [Reference Citation Analysis (0)]