Open access peer-reviewed chapter

Strategies to Preclude Hepatitis C Virus Entry

Written By

Thierry Burnouf, Ching-Hsuan Liu and Liang-Tzung Lin

Reviewed: 01 September 2016 Published: 08 March 2017

DOI: 10.5772/65470

From the Edited Volume

Advances in Treatment of Hepatitis C and B

Edited by Naglaa Allam

Chapter metrics overview

1,485 Chapter Downloads

View Full Metrics

Abstract

Without a preventive vaccine, hepatitis C virus (HCV) remains an important pathogen worldwide with millions of carriers at risk of end-stage liver diseases. Despite the introduction of novel direct-acting antivirals (DAAs), resistance problems, challenges with the difficult-to-treat populations and high costs limit the widespread application of these drugs. Antivirals with alternative mechanism(s) of action, such as by restricting viral entry or cell-to-cell spread, could help expand the scope of antiviral strategies for the management of hepatitis C. Transfusion-associated HCV infection remains another issue in endemic and resource-limited areas around the world. This chapter describes some of the latest developments in antiviral strategies to preclude HCV entry, such as through monoclonal antibodies and small molecules, as well as measures to enhance the safety of therapeutic plasma products in blood transfusion.

Keywords

  • hepatitis C virus
  • viral entry
  • antivirals
  • entry inhibitors
  • monoclonal antibodies
  • small molecules
  • therapeutic plasma products

1. Introduction

Hepatitis C virus (HCV) is a major pathogen that predisposes about 170–300 million people worldwide to risks of end-stage liver diseases (ESLD), including cirrhosis and hepatocellular carcinoma (HCC). The hepatotropic virus remains one of the top indications for liver transplantation in treating ESLD [1]. While a preventive vaccine remains unavailable, the recent introduction of direct-acting antivirals (DAAs) has revolutionized the treatment for hepatitis C, phasing out the decade-old interferon (IFN)-based regimens. The majority of DAAs, however, focus on targeting viral replication such as via inhibition of the HCV NS3/4A protease, the NS5A cofactor, and the NS5B polymerase [2]. Although the DAAs have significantly improved the rate of sustained virological response (SVR) in the most prevalent genotype 1 patients, several challenges persist in real-world setting including high cost, drug-drug interactions, emergence of drug resistance, hard-to-treat populations (e.g., human immunodeficiency virus [HIV] coinfection, ESLD, and transplant patients), and management of DAA failures [35]. With the advent of hepatitis C treatment in larger populations and borrowing from the experience with HIV cocktail therapy, it is becoming clear that developing therapeutic strategies with different modes of action would be necessary to address the various limitations of current DAAs. In addition, HCV transmission due to transfusion of contaminated blood products remains an issue in endemic areas around the world. This is particularly the case in resource-limited countries that face inadequate supply of safe blood products or have poorly controlled blood screening practices, leading to significant risk of transfusion-associated HCV infection [6]. Measures to enhance the safety of therapeutic plasma products such as through the implementation of viral inactivation treatments are therefore a necessity to reduce such risk.

The multistep process of HCV entry makes it an attractive target since it is the foremost fundamental prerequisite in establishing an infection. Following successful entry, the viral life cycle initiates to produce more virions, and with this development the underlying disease begins its progression. Blocking HCV infection by targeting its entry therefore has important implications for both prophylactic and therapeutic purposes since it abolishes the viral life cycle. As a prophylactic treatment, it can be used to prevent infection or reinfection. This is particularly useful in liver transplant setting of hepatitis C wherein the liver allograft is inevitably reinfected [7, 8]. As a therapeutic treatment, precluding HCV entry via de novo infection or cell-to-cell transmission helps to restrict viral spread in an infected person which could slow the progression of the disease. In addition, incorporation of strategies to block HCV entry into existing DAA treatments is expected to maximize the treatment response rate, even producing a synergistic effect [9], as with the experience of using multiple inhibitors in HIV cocktail therapy to concomitantly target various stages of the viral life cycle. Since more steps are being targeted in such a multipronged approach, the inclusion of entry inhibitors to existing DAAs could impose a higher genetic barrier to drug-resistance development. Such tactic not only aids in disrupting persistent HCV infection but could also help to ultimately achieve viral clearance. These aspects therefore make the development of HCV entry blocking strategies highly advantageous in both expanding the scope of antiviral treatments against hepatitis C and providing new insights into antiviral management. This chapter describes some of the latest development of strategies in precluding HCV entry for the management of hepatitis C.

Advertisement

2. Overview of HCV entry

Owing to the development of infectious HCV culture systems (e.g., cell-culture-derived HCV, HCVcc) and viral pseudoparticles bearing HCV glycoproteins (e.g., HCV pseudoparticles, HCVpp), a scenario of how HCV entry occurs has slowly emerged over the last decade of research. It is widely recognized that the HCV particle undergoes a series of intimate and well-orchestrated interactions with various receptors/coreceptors on the hepatocyte host cell surface as well as in the tight junctions, which ultimately lead to the attachment, internalization, and fusion of the virion with the cellular membrane. A number of these receptor interactions are thought to be attributed to the highly lipidated nature of the HCV virion. Specifically, HCV exists as a lipo-viro particle (LVP) with a lipid composition that includes the apolipoproteins and resembles that of very low-density lipoproteins (VLDLs) and low-density lipoproteins (LDLs) [1015]. The association with lipids on the viral particle is thought to contribute to the shielding of HCV glycoproteins from neutralization by the host antibody-mediated response. In addition, the presence of the apolipoproteins on the virion has a large influence on the production of infectious HCV and also its tissue tropism [13, 1622].

Following circulation in the blood, the HCV viral particles reach the liver and begin the interactions with molecules at the surface of the hepatocytes (Figure 1). The initial contacts are with nonspecific receptor(s) including the glycosaminoglycan (GAG) heparan sulfate moieties [2325] that can be found on the transmembrane core proteins syndecans [26, 27]. These early interactions facilitate the attachment of the HCV virion and its accumulation on the hepatocytes for subsequent binding to more specific receptors. Although the LDL receptor (LDLR) has also been suggested as a potential initial attachment factor [2830], recent evidence suggests that it may play a more essential role in viral replication [31, 32]. Nevertheless, these initial interactions have been shown to be mediated via apolipoprotein E (apoE) on the virion [29, 3336]. The capturing process of the HCV particle is finalized by its interaction with the scavenger receptor class B type I (SR-BI) [37, 38], which is able to associate with the virion’s lipoproteins [37, 39] as well as the HCV E2 glycoprotein [40, 41]. Binding of HCV with SR-BI induces lipoprotein rearrangements that help prime the virion for subsequent binding to other host cell factors and promote entry. This process is proposed to occur via SR-BI’s lipid transfer activity between the viral particle and the plasma membrane [37, 42] and/or by direct interaction with the hypervariable region 1 (HVR1) domain on E2 [37, 43], which ultimately leads to conformational change and the exposure of functional glycoprotein epitopes for additional receptor binding. Closely following this event is the engagement of the HCV particle with the tetraspanin receptor CD81 [44, 45], which is an important entry factor for the virus [41, 46, 47]. HCV binding to CD81 is proposed to induce a dynamic lateral diffusion of virus-receptor complexes toward the tight junction area for further interactions with additional entry factors and viral internalization [22, 48]. Specifically, CD81 forms a coreceptor complex with the tight junction protein claudin-1 (CLDN1) [49, 50] and is engaged in late events of HCV entry [51]. This re-localization and virus-receptor complex association with CLDN1 involves multiple signaling pathways (e.g., Rho GTPases, PI3K/AKT, and ERK/MAPK) [52, 53], includes the activation of host cell kinases such as the epidermal growth factor receptor (EGFR) and ephrin receptor A2 (EphA2) [54, 55], and is influenced by the absence of the CD81-associated partner EWI-2wint on the hepatocytes [56, 57]. The EWI-2wint molecule is normally bound to CD81 on most cell type surfaces and inhibits its diffusion which is required to promote HCV entry; however, it is not expressed in the hepatocytes, and hence its absence has been suggested to contribute to the restricted tropism of the virus [56]. Following interaction with the CD81/CLDN1 complex, the HCV particle is presumed to then interact with the tight junction protein occludin (OCLN) prior to viral internalization [58]. Additional proteins that take part in influencing virion entry into the hepatocyte include the transferrin receptor 1 (TfR1) [59] and the cholesterol transporter Niemann-Pick C1-like 1 (NPC1L1) [60], although their specific role and interplay with other entry factors in the HCV entry process remain to be defined. The HCV particle finally enters the cell via clathrin-mediated endocytosis [61]. The HCV-receptor complexes then migrate to endosomal compartments [62, 63] where acidification occurs to induce membrane fusion, which allows the release of viral RNA into the host cytosol.

Figure 1.

Overview of HCV entry.

The above sequential and multistep entry process consequently yields the successful release of the HCV genome into the host cytoplasm for direct translation and the ensuing launch of viral replication. The roles played by several of these entry factors including SR-BI, CD81, CLDN1, and OCLN not only mediate HCV entry but also presumably help to define tissue and species tropism of the virus [6467]. The understanding of how HCV achieves viral entry has led to the possibility of antiviral targeting. From docking to virus internalization, essentially all steps are targetable to prevent HCV infection of the host cell. In addition, given the association of HCV with lipoproteins and the viral particle’s interaction with lipoprotein and lipid receptors (LDLR, SR-BI, and NPC1L1), the lipidic nature of HCV virion also offers various methods of pharmacological intervention. Finally, many of the entry factors including CD81, SR-BI, CLDN1, OCLN, and NPC1L1 also play a role in mediating HCV cell-to-cell transmission between intercellular junctions [6871], and therefore targeting these molecules could help restrict both cell-free entry and cell-to-cell spread of HCV.

Advertisement

3. Current development in inhibition of HCV entry

3.1. Use of monoclonal antibodies to target host cell receptors or viral antigens

Recent insight into the molecular interactions of HCV at the cellular membrane has significantly enhanced the understanding of the HCV entry paradigm and revealed potential targets for drug intervention, including the use of monoclonal antibodies (mAbs) to mask HCV entry receptors/coreceptors or viral antigens. As described below and summarized in Table 1, the use of mAbs targeting CD81, SR-BI, CLDN1, or the HCV E2 has been shown to have prophylactic/therapeutic effects against HCV infection in both cell culture and animal models.

CandidatesEffect(s)Stage of DevelopmentReference
mAbs Against Host Entry Factors
Anti-CD81 mAbsInhibit CD81-E2 interactionMouse model[7577]
Anti-SR-BI mAbsInhibit SR-BI-E2 interactionMouse model[8082]
Anti-CLDN1 mAbsInhibit E2-CD81-CLDN1 associationMouse model[8487]
Passive Immunotherapy Against HCV
Anti-E2 mAbsNeutralize circulating virionPhase II[8996]
Polyclonal IgGNeutralize circulating virionPhase III[190, 192, 193]
Small Molecule Inhibitors
Heparin, heparin-derived compoundsHeparan sulfate competitorsCell culture[24, 25]
HeparinasesHeparan sulfate enzymeCell culture[25]
EGCGCompete with heparan sulfate; alter viral shape; inhibit cell-to-cell spreadCell culture[99101]
DelphinidinAlter viral shapeCell culture[101]
SSb2Inhibit attachment & viral fusionCell culture[102]
GAInactivate virionCell culture[103]
Hydrolysable tannins CHLA & PUGInactivate virion; inhibit attachment & cell-to-cell spreadCell culture[104]
LODInactivate virion; inhibit attachmentCell culture[105]
DHMDInactivate virion; inhibit attachmentCell culture[106]
CurcuminDecrease viral envelope fluidity; inhibit cell-to-cell spreadCell culture[107]
CV-NE1/E2 glycan-binding proteinCell culture[109]
GriffithsinE1/E2 glycan-binding proteinMouse model[110]
MBLE1/E2 glycan-binding proteinCell culture[111]
Recombinant L-ficolinE1/E2 glycan-binding proteinCell culture[112]
BA-LNCsE2 glycan-binding proteinCell culture[114]
Oleanolic acidE2 glycan-binding proteinCell culture[115]
CD81-derived peptidesInteract with E2Cell culture[116, 117]
CLDN1-derived peptide (CL58)Interact with E1 & E2Cell culture[81]
E2-derived peptideInterfere with E1/E2 hetero-dimerizationCell culture[119]
TerfenadineCD81 competitorCell culture[120]
ITX 5061SR-BI inhibitorPhase Ib[121124]
AspirinDown regulates CLDN1Cell culture[125]
ErlotinibEGFR inhibitor; inhibit cell-to-cell spreadMouse model[54]
DasatinibEphA2 inhibitor; inhibit cell-to-cell spreadCell culture[54]
Tipifarnib, sorafenibRas, Raf inhibitorCell culture[126]
FerristatinTfR1 inhibitorCell culture[59]
EzetimibeNPC1L1 inhibitorMouse model[60]
PF-429242Down regulate NPC1L1 & LDLRCell culture[127]
PhenothiazinesModulate host cell membraneCell culture[128]
ChlorpromazineClathrin-coated pit formation inhibitorCell culture[61]
ArbidolTrap virion in clathrin-coated vesiclesCell culture[129]
Bafilomycin A1, concanamycin ADisturb acidic endosomal compartmentsCell culture[130]
Chloroquine, ammonium chlorideDisturb acidic endosomal compartmentsCell culture[131]
RAFI dUY11Inhibit viral fusionCell culture[132]
FerroquineInhibit cell-to-cell spreadCell culture[133]
Triazine-based compoundsInhibit post- binding step & cell-to-cell spreadCell culture[134, 135]
SilibininInhibit viral fusion & cell-to-cell spreadClinical Legalon® SIL[136140]
TamoxifenInhibit attachment & post-binding stepCell culture[142]
HCV II-1 (GS-563253)Inhibit attachment & post-binding stepCell culture[143]
EI-1 (BJ486K)Inhibit post-binding stepCell culture[144]

Table 1.

Antiviral strategies to preclude HCV entry.

3.1.1. Anti-CD81 monoclonal antibodies

CD81 is the first putative receptor identified for HCV entry [72, 73] and plays an important role in the virus infection. The molecule is a member of the tetraspanin superfamily with four transmembrane domains and two extracellular loops and is expressed in most human tissues [74]. Commercial CD81 mAb JS-81 has been applied in human liver-chimeric mouse model and shown prophylactic protection but no postexposure effect inhibiting HCV infection [75]; nonetheless, this experimental test inspired subsequent studies of anti-CD81 mAbs as antiviral agents. Of the newly generated antibodies, mAb QV-6A8-F2-C4 produced by genetic immunization could efficiently inhibit HCVcc infection and pan-genotypic HCVpp entry in a similar range as mAb JS-81 [76]. The antibody also appeared to block neutralizing antibody-resistant HCV cell-to-cell transmission and viral dissemination in a dose-dependent manner, with a less cytotoxic or antiproliferative property than JS-81 in vitro. In a recent study, another mAb K04 generated with hybridoma technique not only showed inhibitory effect against HCVpp and HCVcc infection in hepatoma cells and primary human hepatocytes (PHH), but also surprisingly blocked HCV infections in both prophylactic setting and postinfection stage in human liver-chimeric mice [77]. This is probably due to the improved intrinsic binding affinity of mAb K04 to CD81 large extracellular loop (LEL) and a different binding epitope as compared to mAb JS81. However, treatment-associated reductions in body weight and human serum albumin levels were observed in this study. Further research will be needed to determine the minimal dose of antibodies needed to provide protection and to evaluate the toxicology of anti-CD81 mAbs for long-term development.

3.1.2. Anti-SR-BI monoclonal antibodies

SR-BI is a member of the CD36 family primarily expressed in liver and non-placental steroidogenic tissues which facilitates selective cholesterol uptake [78]. The molecule has been proposed to be a horseshoe-like glycoprotein with a large extracellular loop anchored to the plasma membrane at both N- and C-termini with short extensions into the cytoplasm [79]. It was first identified as the alternative E2 receptor on HepG2 cells which efficiently recognize soluble E2 proteins but do not express CD81 on their surface [40]. As described above, both CD81 and SR-BI are considered necessary for HCV entry, since the overexpression of CD81 on HepG2 cells restores HCVpp entry in these originally poorly permissive cells [41]. Monoclonal antibodies targeting SR-BI that inhibited HCV infections include mAb C167, mAb16-71, mAb8, and mAb151. For HCV inhibitory activities in vitro, mAb C167 effectively prevented infection in hepatoma cells with HCVcc and ex vivo virus recovered from HCVcc-infected chimpanzees [80]; mAb16-71 exhibited preventive effect against HCVcc infection in both hepatoma cells and PHH [81]; mAb8 and mAb151 also prevented HCVcc infection in reporter Huh-7 cells [82]. Additionally, mAb16-71, mAb8, and mAb151 all showed their ability in blocking HCV cell-to-cell spread in vitro and in vivo. Human liver-chimeric mouse models challenged with serum-derived HCV isolates of different genotypes revealed the anti-HCV property in vivo of the three antibodies in both prophylactic and postexposure settings. Specifically, mAb16-71 showed complete blockage of infection and intrahepatic spread of HCV isolates with a prophylactic treatment, but had no effect on chronically infected chimeric mice; mAb151, on the other hand, appeared to be effective against an HCV variant escaped from adaptive immune response in a liver transplant patient and displayed better antiviral activity in inhibiting viral spread and amplification in the postexposure setup.

3.1.3. Anti-CLDN1 monoclonal antibodies

The CLDN1 tight junction protein has four transmembrane domains and is highly expressed in the liver [83]. Its role in HCV entry is proposed to occur in the post-binding steps [64]. Anti-CLDN1 antibodies directed against the CLDN1 extracellular loops were found effective in neutralizing HCV infection in hepatoma cells through disrupting CD81-CLDN1 association and therefore inhibiting E2 binding to the cell surface [84]. A CLDN1 mAb OM-7D3-B3 targeting CLDN1 extracellular loop was found to be effective in inhibiting HCV isolates in vitro [85]. Further experiments in human liver-chimeric mouse models confirmed its potency in preventing HCV infection and eliminating persistent infection in vivo [86]. Pretreatment of another anti-CLDN1 mAb 3A2 targeting CLDN1 extracellular loop also showed protective effect in a chimeric mouse model [87]. Safety profiles of these antibodies were also assessed regarding the levels of human albumin, aspartate transaminase, alanine transaminase and total bilirubin, and potential side effects on the other organs and tight junction integrity. Further studies were suggested to assess potential immune-mediated adverse effects to ensure its relevance for clinical use [86, 87].

3.1.4. Anti-HCV E2 monoclonal antibodies

Another approach to developing entry-inhibiting mAbs is to target the glycoproteins on the HCV virion surface. Albeit HCV glycoproteins exhibit high variability and are protected by glycosylation and lipids on the viral particle, neutralizing mAbs have been designed to target more conserved and accessible regions, specifically on the E2 glycoprotein [88]. Effects of E2 mAbs have been shown in vitro and in vivo [8994]. Clinical trials have been carried out to assess the protective function of human anti-E2 mAbs HCV-AbXTL68 and MBL-HCV1 in liver transplant settings of HCV-positive patients. With a higher dose and daily infusion of HCV-AbXTL68, HCV RNA in patient serum showed transient reduction in the first week post-transplantation but not yet below the detectable limits [95]. MBL-HCV1, on the other hand, successfully suppressed the viral load from 7 to 28 days after transplantation in genotype 1a-infected patients with multiple infusions. Although the primary endpoint at day 42 was not met, the viral rebound was significantly delayed, and the magnitude of the viral load reduction was greater than the previous HCV-AbXTL68 therapy [96]. The result indicates that mAbs may be a promising class of entry inhibitors that adsorbs circulating virions to protect the new liver from reinfection after transplantation. A study of combination therapy with DAAs to prevent allograft HCV infection is currently underway [96].

Current obstacles to the development of mAbs as therapeutic antiviral agents include the high cost of production, storage, and administration, which can only be done by injection so far [88]. Nevertheless, the associated immune responses such as antibody-dependent cell-mediated cytotoxicity (ADCC) and complement-dependent cytotoxicity (CDC) may help to clear the viruses and infected cells [88, 97]. Antibodies that directly block host cell entry factors are more likely to be effective for the diverse circulating viral strains; however, due to the distribution and multiple functions of such molecules, the blockage may cause potential adverse side effects [97]. As for antibodies targeting viral antigens, designing suitable candidates may be a challenging issue due to the heterogeneity of the HCV glycoproteins [98], but such antibodies may provide a safer option for the synergistic therapy with other antivirals of different modes of action to suppress the development of resistance, particularly at the early post-transplantation stage [96]. Additional neutralizing antibodies against other entry factors have also been reported to antagonize HCV infection in vitro, such as anti-TfR1 [59] and anti-NPC1L1 [60] antibodies, suggesting they could also be potentially developed for treatments against hepatitis C.

3.2. Small-molecule inhibitors of HCV entry

In addition to the mAbs, great efforts have been put into identifying small molecules with potent antiviral effects against HCV entry. The source of such entry inhibitors includes clinically approved medications, synthetic molecules, and natural product-based compounds. These small molecules could be further evaluated for development as drug candidates or drug leads. Below is a panel of small molecules that have been investigated with their activities inhibiting HCV entry (Table 1).

3.2.1. Small molecules inhibiting viral attachment

The attachment step represents the primary interaction of an HCV virion with its host cell surface. Since the GAG heparan sulfate moieties dominate the capturing of HCV virions, the heparan sulfate homologue heparin and its derivatives as well as the enzyme heparinases which degrade the molecule were all shown to inhibit the viral binding to hepatoma cells [24, 25]. (-)-Epigallocatechin-3-gallate (EGCG), a green tea catechin, was speculated to exert its inhibitory effect on viral attachment [99] by competing with heparan sulfate for HCV binding [100] or altering the viral shape [101]. Delphinidin, an anthocyanidin extracted from plant pigment, was also demonstrated to inactivate HCVpp by altering its shape and was particularly potent when added concurrently with the viral inoculation [101]. The natural terpenoid saikosaponin b2 (SSb2), isolated from the root of Bupleurum kaoi, was observed to specifically block HCV particle binding and early viral entry without affecting other stages of the viral life cycle [102]. SSb2 could inactivate cell-free HCV particles and was suggested to target the glycoprotein E2 in mediating its antiviral effect against HCV infection. Several other natural compounds including the gallic acid (GA) extracted from Limonium sinense [103], the hydrolyzable tannins chebulagic acid (CHLA) and punicalagin (PUG) [104], and the hepatoprotective plant Phyllanthus urinaria-derived monolactone loliolide (LOD) [105] and butenolide (4R,6S)-2-dihydromenisdaurilide (DHMD) [106] were also found to efficiently inactivate cell-free HCV viral particles and impede viral attachment. Another natural compound curcumin extracted from turmeric was shown to decrease the fluidity of viral envelope and therefore prevent the binding and fusion [107], possibly by inserting into the membrane in a manner similar to cholesterol [108].

3.2.2. Small molecules blocking viral glycoproteins

A variety of broad-spectrum antiviral agents have exhibited their ability to interact with the glycans on viral glycoproteins. In the case of HCV, glycan-binding proteins interfere with the association between the E1/E2 heterodimer and the host cell receptor CD81. Lectins such as cyanovirin-N (CV-N) and griffithsin, isolated from cyanobacterium Nostoc ellipsosporum and the red alga Griffithsia sp., respectively, were reported to have such effect. CV-N was shown to interact with N-linked glycans of HCV glycoproteins and disrupt E1/E2 binding to CD81 [109]. The inhibitory effect of griffithsin on HCV entry was also quenched when N-linked high-mannose oligosaccharides were present, indicating a pattern similar to CV-N of affecting the glycoproteins-CD81 interaction [110]; pretreatment of griffithsin was shown to delay the viral infection in chimeric mouse model. Humoral lectins of the innate immune systems including the mannan-binding lectin (MBL) and L-ficolin were also considered to have analogous effect neutralizing HCV particles. MBL [111] and recombinant oligomeric L-ficolin [112] were found to interact with the glycans on the E1/E2 heterodimer in a calcium-dependent manner, thereby inhibiting the viral entry. Notably, the MBL-associated complement system was activated upon its binding to HCV E1/E2, suggesting the use of humoral lectins as viral entry inhibitors may also help facilitate viral clearance. However, the detailed mechanism and specific target of the humoral lectins remain to be defined. The boronic acid (BA)-modified nanoparticles were also found to suppress HCV entry in a way that acted similar to lectins [113], with the incorporation of lipid nanocapsule (BA-LNC) techniques enhancing their stability and solubility [114]. Chemically modified oleanolic acid, a triterpene compound originally extracted from Dipsacus asperoides, was found able to interrupt the E2-CD81 interaction by binding to E2 [115].

Besides the glycan-binding proteins, molecules imitating HCV host entry factors or viral glycoproteins were also developed in the attempt to block the viral entry. An imidazole-based scaffold presenting CD81 helix D amino acid side chains [116] and stapled peptides based on CD81 LEL [117] were designed to antagonize the E2-CD81 interaction by mimicking the putative E2-binding region of CD81. A CLDN1-derived peptide, CL58, was also found to inhibit HCV entry in the post-attachment stage by interacting with HCV E1 and E2 [118]. As for viral glycoprotein-based molecules, an E2-derived peptide was found able to block E1/E2-mediated fusion by targeting E1 and therefore interfere with the hetero-dimerization of the glycoproteins [119].

3.2.3. Small molecules targeting host entry factors and CD81-triggered signaling pathway

In addition to the therapeutic antibodies mentioned in the previous section, several small molecules have been suggested to exert their inhibitory activity of HCV entry by targeting cellular receptors/coreceptors. Terfenadine, an antihistamine, was found able to prevent HCV infection by competing with the CD81 antibody JS81 binding to the LEL of CD81 protein on the hepatoma cell surface [120]. ITX 5061, a clinical stage compound originally characterized as a p38 MAPK inhibitor, was identified with its capability of antagonizing SR-BI [121] and further validated for its potency of inhibiting HCV entry at post-binding step [122]. The anti-HCV effect of ITX 5061 was found additive to synergistic in combination with several standard-of-care therapeutics, and the resistant mutant was defined on the viral glycoprotein E2 [123]. A latest phase 1b clinical trial [124] revealed that the ITX 5061-treated patients, especially the genotype 1-infected patients, had a significant reduction in HCV RNA through the first week after liver transplantation and viral evolution were restricted; however, the viral RNA levels became comparable in both ITX 5061-treated and untreated patients, suggesting the need to incorporate other antiviral agents using different modes of actions to eliminate HCV infection. Aspirin, alternatively, inhibited HCV entry by downregulating CLDN1 [125].

Since the receptor tyrosine kinases are also involved in the HCV entry process, two clinically approved protein kinase inhibitors were evaluated for their ability to abrogate the viral entry. Both erlotinib, an EGFR inhibitor, and dasatinib, an EphA2 inhibitor, could successfully block HCV entry in a dose-dependent manner as well as the cell-to-cell transmission. Specifically, erlotinib was shown to inhibit the membrane fusion of hepatoma cells overexpressing HCV glycoproteins. In vivo treatment of erlotinib resulted in a significant suppression of the viral load in PHH-chimeric mouse model with HCV infection [54]. Furthermore, inhibitors of EGFR downstream kinases Ras (tipifarnib) and Raf (sorafenib) were also assessed and found effective in blocking HCV entry [126].

Inhibitors of other entry factors were also shown to be effective in hampering the viral entry. Pretreatment of ferristatin, a TfR1 inhibitor that binds to the molecule and causes its internalization and degradation, was shown to decrease HCVcc infection in vitro [59]. The NPC1L1 internalization inhibitor ezetimibe, which is also an FDA-approved cholesterol-lowering medication, diminished HCVcc foci formation before and during the viral challenge. Daily oral administration of ezetimibe starting two weeks before infection also delayed the viral growth of a genotype 1 clinical isolate in PHH-chimeric mouse model [60]. PF-429242, an SKI-1/S1P inhibitor, potentially impeded HCV entry by downregulating NPC1L1 and LDLR expression [127]. On the other hand, phenothiazines, a group of synthesized nitrogen- and sulfur-containing tricyclic compounds, inhibited HCV fusion into the cell by modulating the host cell membrane. Insertion of phenothiazines into the cholesterol-rich membrane increased its fluidity, thus possibly decreasing the local inhomogeneity of the cell required for the viral fusion [128].

3.2.4. Inhibition of clathrin-mediated endocytosis and viral fusion

Since HCV fusion has been discovered to be facilitated by clathrin-mediated endocytosis and requires an acidic environment, several reagents were assessed for their effectiveness in preventing HCV entry through blocking such pathways. Chlorpromazine, an inhibitor of clathrin-coated pit formation, was shown to inhibit both HCVpp and HCVcc infection in vitro in the validation of clathrin-mediated endocytosis pathway of HCV fusion to the host cell membrane [61]. Arbidol, a broad-spectrum antiviral agent that blocks viral entry and has been licensed in some regions for influenza, was described to trap the HCV virion in clathrin-coated vesicles, thereby hindering the release of viral genome and the following infection [129]. It was also suggested that arbidol could generally cause the intracellular accumulation of clathrin-coated structures and restrain the formation of clathrin-coated pits on the cell surface [129], possibly due to its tropism for lipid bilayers.

Small molecules disturbing the acidic endosomal compartments were also identified as HCV entry inhibitors in the discovery of the low pH-triggered entry. These include bafilomycin A1 and concanamycin A, which are inhibitors of vacuolar H+-ATPases [130]. Weak bases such as chloroquine and ammonium chloride were also found to inhibit the low pH-dependent conformational change required for the viral fusion, based on their ability to penetrate lysosomes and increase the pH [131]. Finally, dUY11, one of the rigid amphipathic fusion inhibitors (RAFIs), was suggested to inhibit HCV entry by interacting with the hydrophobic structures in virions and preventing the formation of negative curvature required for viral fusion [132]. Curcumin [107] is also able to affect the fusion step as previously mentioned.

3.2.5. Small molecules inhibiting cell-to-cell transmission

Besides inhibiting the HCV entry in de novo infection, blocking cell-to-cell spread of the viral particles is also important as this mode of transmission facilitates efficient spread of the virus in the liver escaping from neutralizing antibodies [68, 69]. Ferroquine was speculated to interact with HCV glycoprotein E1 and abrogate cell-to-cell spread of the virus [133]. Triazine-based compounds indicated to be closely related to the amino acids on the glycoprotein could also selectively inhibit genotype 1 HCV entry at the post-attachment step along with cell-to-cell transmission [134, 135]. Several molecules also block cell-to-cell spread in addition to their activities in hindering HCV viral entry. For instance, besides impeding viral attachment, CHLA and PUG exhibit pronounced antiviral effects at the postinfection stage, especially in restricting HCV foci expansion [104]. Others include EGCG [99], curcumin [107], erlotinib, and dasatinib [54]. Silibinin, the major component of Silybum marianum that has been designated as an orphan drug for the prevention of recurrent hepatitis C in liver transplant patients [136], was also suggested to possess a prominent effect blocking transmission of the viral particles between intercellular junctions [137, 138], although other studies have proposed that it may slow down clathrin-mediated endocytosis [139] as well as inhibit viral membrane fusion [140]. This could be useful since DAA-resistant HCV variants have been suggested to escape via cell-to-cell transmission route [141]. Therefore, the choice of inhibitors exhibiting mechanistic effect against both HCV cell-to-cell spread and cell-free entry, or a combination of such two types of inhibitors, should facilitate viral clearance.

3.2.6. Additional candidate entry inhibitors

Some other molecules were found able to prevent the infection at different steps of HCV entry. The estrogen receptor modulator tamoxifen [142] and HCV infectivity inhibitor 1 (HCV II-1 or GS-563253) [143] were shown to inhibit the HCV infection at both attachment and post-binding steps. HCV II-1 was also found capable of impeding infectious virion propagation [143]. HCV entry inhibitor 1 (EI-1 or BJ486K), a flavonoid ladanein, was shown to interrupt the viral entry at post-attachment stage [144]. The exact mechanisms of these molecules require further investigations. Other compounds such as serum amyloid A [145, 146], p7 ion channel-derived peptide H2-3 [147], amphipathic DNA polymers [148], lactoferrins [149], tellimagrandin I and its derivatives [150], indole derivatives [151], and imidazo[1,2α][1,8]naphthyridine derivatives [152] were found able to inhibit HCV entry with mechanisms that remain to be clarified.

3.3. Control of HCV infection risks in human blood-derived therapeutic products

Many viruses can contaminate human blood. HCV, along with HIV and HBV are a major cause of infectious complications of blood product transfusion therapy. HCV contamination in patients by transfusion of blood components such as red blood cells, platelets or clinical plasma, as well as industrial fractionated plasma products, has been well documented. At the time of the “tainted blood scandal,” numerous recipients of blood components and hemophiliacs receiving plasma-derived factor VIII concentrates were contaminated through transfusion of nonvirally inactivated products prepared from blood products that were not HCV-tested. HCV transmission through blood transfusion is a major medical issue, as infection can lead to high risk of liver cirrhosis and eventually cancer complications.

3.3.1. HCV safety nets for blood components

There are now over 100 million whole blood donations collected each year in the world. Collected blood is most often separated by “blood establishments” into red blood cell concentrates, platelet concentrates, and plasma that are transfused at nearby hospitals. Plasma, which can be obtained from whole blood collection or drawn by specialized apheresis procedures, can also be used as raw material for the production of “industrial” plasma protein products. These protein drugs include immunoglobulins G (IgGs), various coagulation factors, albumin, and many others. Industrial plasma products are manufactured from pools of plasma of several thousand liters, making them statistically more susceptible to contamination by HCV and other viruses as one highly infectious donation would contaminate the whole plasma pool and potentially the derived products.

Today in developed economies benefiting from strict regulatory oversight, several measures are in place to decrease the possibility for patients to acquire HCV by transfusion. Blood transfusion HCV safety nets for blood components rely on complementary measures encompassing (a) epidemiological control of the population, (b) individual screening of candidate blood donors to defer those identified as presenting potential risk factors, and (c) individual blood donation testing to identify and eliminate donations reactive to anti-HCV antibodies and/or HCV RNA nucleic acid test (NAT) [153]. In technology-advanced countries applying such procedures, this has allowed to decrease the risk of acquiring HCV by transfusion of single blood components down to approximately 1 per 1.8 million. The remaining risk reflects the inevitable presence of “window-phase” donations for which all markers to detect donor infection by HCV, either indirect or direct, are found nonreactive [154, 155]. Understandably, HCV transmission risks are substantially higher in less developed economies (a) lacking a safe blood donor base, (b) relying on paid or "replacement" donors to increase the blood supply, (c) with a deficient blood collection system, and (d) with a lack of reliable viral testing procedures [6]. The ultimate barrier to avoiding HCV transmission risks from blood products collected during the window-phase period relies on the implementation of dedicated viral reduction treatments. Those have been developed for industrial plasma protein products, plasma for transfusion, and platelet concentrates. Until now, however, no treatment is available commercially for whole blood and red blood cell concentrates.

3.3.2. HCV reduction treatment of industrial plasma protein products

Development and implementation of dedicated viral/HCV reduction treatments of industrial plasma protein products took place in the 1980s and early 1990s [156]. In the early 1980s, albumin, a relatively heat-stable protein, was the only plasma product subjected to specific HCV inactivation by heat treatment at 60 °C for 10 h in the liquid state (a process called pasteurization), in the presence of fatty acid stabilizers. From the mid-1980s to the early 1990s, heat treatment of freeze-dried coagulation factors at 60?68 ?C for 24?96 h or 80 ?C for 72 h were developed to inactivate HIV and HCV concomitantly [156]. Although pasteurization has successfully been adapted to several plasma products (such as antithrombin and alpha 1-antitrypsin), a milestone in the safety of industrial plasma products was the development of the solvent/detergent (S/D) incubation procedure at 20–37 °C [157] designed to dissolve the lipid envelope of viruses, including HCV, without affecting plasma protein functions. This technique is still largely used for a wide range of industrial plasma products owing to well-proven efficacy and a safety profile established by years of industrial and clinical practices [158]. Other HCV viral inactivation treatments include low pH incubation and caprylic acid precipitation/incubation of immunoglobulin products [159]. An additional milestone to enhance plasma protein product safety is nanofiltration, a procedure of filtration of protein solutions on 15–35 nm nanopore membrane devices designed to entrap and remove viruses [160]. This dedicated virus removal methodology is well established, including for HCV, and is currently applied to most plasma products [156]. Thanks to the implementation of such reduction treatments, most often combined in a complementary manner at different stages of the manufacturing process, no case of HCV transmission by industrial plasma products has been reported since 1993 [154].

3.3.3. HCV reduction treatment applied to plasma and platelet concentrates for transfusion

3.3.3.1. Plasma

Several viral inactivation treatments of clinical plasma are licensed in various countries [161]. The S/D technology was adapted to 100–500 l of pooled industrial plasma in the early 1990s [162] and demonstrated, prior to HCV identification, to efficiently inactivate non-A-non-B hepatitis virus [163]. The removal of the S/D agents is typically achieved by oil extraction and column hydrophobic interaction chromatography [162]. A miniaturized version of the S/D process using a different detergent (Triton X-45 instead of Triton X-100) has been developed allowing its implementation in single-use equipment, thereby facilitating its application in developing countries, such as Egypt, currently lacking industrial capacity [164]. The efficacy of such method to inactivate HCV has been specifically demonstrated using an in vitro culture assay [165].

A procedure consisting in adding methylene blue and illuminating acellular plasma was made available in the early 1990s [166]. The method leads to inactivation of free HCV particles through photochemical alteration of nucleic acids and incapacity of replication [154, 167].

Two other photoinactivation procedures of plasma have been licensed more recently. One combines the addition of psoralen S-59 (amotosalen) with ultraviolet light A illumination [168]. The other is based on the addition of riboflavin followed by UV irradiation [169]. These small molecules can penetrate membranes and intercalate with helical regions of HCV nucleic acids. Subsequent UV illumination irreversibly alters nucleic acids, making HCV particles unable to replicate [154, 170].

3.3.3.2. Platelets

Development of HCV inactivation methods in cellular blood products in general, and platelet concentrates in particular, has been more challenging due to the difficulty to inactivate intracellular viruses without affecting cell function for transfusion. The two photoinactivation methods applied to plasma could nevertheless be adapted to the inactivation of HCV and other viruses in platelet concentrates [170172].

3.3.3.3. Cryoprecipitate

Cryoprecipitate, obtained by a freeze-thaw process of plasma, is rich in factor VIII, von Willebrand factor, and fibrinogen. This plasma fraction is still largely used in many developing countries for substitution therapy in hemophilia A, von Willebrand factor disease, or fibrinogen deficiency, respectively. The frequency of treatment of patients with congenital deficiency exposes them to a high risk of infection in countries such as Egypt with a close to 10% HCV incidence [173, 174]. Similar mini-pool methods of HCV inactivation used for clinical plasma are applied to cryoprecipitates [164].

3.3.3.4. Red blood cell concentrates and whole blood

No methodology is licensed yet for HCV inactivation in red blood cell concentrates or whole blood. However, the riboflavin/UV pathogen reduction technology is being adapted to the treatment of whole blood [175] and has been shown recently to contribute to lower the risk of malaria transmission in a clinical study in Ghana [176]. It is still uncertain whether a pathogen reduction technology can be developed to substantially inactivate HCV in whole blood or red blood cell concentrates without detrimentally affecting their transfusion quality and functionality or immunogenic potential.

3.4. Therapeutic apheresis and passive immunotherapy

Additional methods of precluding HCV infection are to remove circulating virus through therapeutic apheresis or attempting to neutralize HCV infectivity by administering plasma-derived anti-HCV immunoglobulins. These strategies are aimed at reducing the infectious viral load and have been explored in clinical trials.

3.4.1. Therapeutic apheresis for the removal of HCV virions

Therapeutic apheresis is the process of transiently circulating the blood outside the body and removing the components causing particular diseases by membrane separation and adsorption separation technologies. In the case of HCV, immunoadsorption apheresis was first applied to treat the chronic hepatitis C-related cryoglobulinemia that causes autoimmune symptoms [177]. The technique of heparin-induced extracorporeal LDL precipitation (HELP) apheresis, which could eliminate apolipoprotein B-containing lipoproteins, was then discovered to reduce HCV viral load [178]; however, the decline was found not correlated with LDL reduction in plasma and appeared to be transient due to the high turnover rate of HCV [179]. Studies using combination therapy of antiviral agents and double-filtration plasmapheresis (DFPP) that selectively removes substances with high molecular weight including HCV particles and therefore, happened to display better effects of suppressing the viral kinetics and therefore have been substantially explored during the past decade. Patients who underwent the prophylactic combination treatment of low-dose IFN, ribavirin, and DFPP had no evidence of HCV recurrence or fibrosing cholestatic hepatitis exacerbation for more than 1 year after liver transplantation [180]. Combination of DFPP and IFN also achieved impressive SVR in difficult-to-treat patients (i.e., relapsed, nonresponder, or HIV-coinfected patients) [181184] and may also be safe for the elderly population [185]. However, the approach of apheresis for decreasing HCV viral load requires specialty equipment and possesses potential risk of adverse events (e.g., blood pressure lowering, puncture site hematoma, or infection) [181, 185].

3.4.2. Passive immunotherapy using plasma-derived polyclonal HCV immunoglobulins

Passive immunotherapy, also known as antibody therapy, is a very well-established treatment based on the administration of polyclonal hyperimmune immunoglobulins extracted from plasma or mAbs prepared by genetic engineering technologies. One application of passive immunity is to prevent or treat infections due to viruses or to reduce the pathologies associated with bacterial or venom toxins. Human immunoglobulins for passive immunotherapy are fractionated from the plasma of immunized donors having high-titer antibodies against a particular organism or antigen. For the fractionation process, plasma donations from hundreds or thousands of donors are pooled and subjected to various purification and viral inactivation steps, as described in this chapter, to isolate an essentially pure Ig preparation [159, 186]. Current human plasma-derived hyperimmune globulin products are used for the prophylaxis and treatment of viral diseases due to hepatitis B virus (HBV), rabies virus, cytomegalovirus, hepatitis A virus, or respiratory syncytial virus [187]. Human plasma-derived polyclonal hepatitis B immunoglobulin for intravenous use has been made available commercially for over 20 years in some countries. These licensed preparations are efficacious to predictably prevent HBV recurrence after liver transplantation and vertical HBV transmission from mother to child and are used as prophylactic treatment to prevent infection following contact with HBV-contaminated body fluids [188].

The possibility to use polyclonal HCV immunoglobulin to treat or prevent HCV infection has been proposed for many years [189], but no commercial preparation is available yet as it is not proven whether such immunoglobulin can prevent HCV infection or control viremia in infected patients. The rationale in polyclonal HCV immunoglobulins made from large pool of plasma units is to have a preparation that contains neutralizing antibodies to various strains of HCV [189]. However, the presence of neutralizing antibodies has been unclear initially as their presence in plasma was just considered to reflect the occurrence of an infection. Data have suggested that HCV-neutralizing antibodies exist in anti-HCV-positive plasma, but the anti-HCV antibody titer does not correlate with neutralizing capacity [190]. In vitro and animal experiments in a mice model have nevertheless suggested the presence of neutralizing antibodies in polyclonal IgG from a patient with a long-standing HCV infection [191]. A clinical study was initiated in the USA to evaluate the capacity of polyclonal plasma-derived HCV immunoglobulins to “prevent post-transplantation HCV infection of the liver graft and related progression of HCV-related liver disease.” This clinical trial was “designed to evaluate a polyclonal human hepatitis C immune globulin given during and post liver transplantation for preventing or reducing the impact of recurrent HCV infection” [192]. However the trial was terminated in 2012 after treatment of seven patients (five receiving the immunoglobulins and two standard-of-care treatment alone) and no data reported. A new trial has begun in 2013 and was recently completed [193]. It is unclear whether plasma-derived polyclonal HCV immunoglobulin will be developed. If this occurs, clear donor screening and donation testing criteria should be defined to determine the specifications of the plasma donations suitable for fractionation, as well as the fractionation methodology itself to exclude any infectious risks from the fractionation of plasma donations. It should be noted that several mAbs for clinical use in HCV-infected patients have been proposed and one has undergone a clinical trial [190]. The future will indicate whether any HCV immunoglobulin, either polyclonal or monoclonal, has a role to play in the control of HCV infection.

Advertisement

4. Prospects of targeting HCV entry in clinical setting

Treatment options against hepatitis C have significantly improved owing to recent advances in the development of anti-HCV therapeutics. Nevertheless, there is still much room for improvement due to potential drug resistance and possibility of viral rebound, which usually require long periods of monitoring and analysis to uncover. More importantly, there is currently no immunization or prophylactic treatment against hepatitis C. Introducing novel antivirals with a different mode of action, such as targeting viral entry using mAbs or small molecules, not only helps expand the spectrum of anti-HCV drugs but also in developing novel treatment modalities. Many of the mAbs targeting HCV receptors/coreceptors as well as small-molecule inhibitors of HCV entry impede both viral attachment and cell-to-cell transmission; this is useful in providing protection against de novo infection and at the same time in helping restrict viral spread. The inclusion of viral entry inhibitors to current DAAs has already been shown to produce synergistic treatment effect [9]. Furthermore, taking a multistep targeting approach would help elevate the genetic barrier against selection of resistant variants, thus facilitating viral clearance. Finally, the advantage of developing entry inhibitors is its potential prophylactic application against hepatitis C, which is particularly useful in protecting liver allografts from recurrent HCV infection. Other protective measures of hepatitis C transmission in clinical scenarios include implementation of viral inactivation methods for the removal of HCV infectivity in therapeutic plasma products [165]. In addition, therapeutic apheresis [180] and protective anti-HCV immunoglobulins [192, 193] have also been suggested for prevention of HCV reinfection in liver transplant patients. In the absence of an approved hepatitis C vaccine, these approaches could be explored as preventive and prophylactic measures against HCV infection. With the above-described strategies to preclude HCV entry, it is foreseeable, in a not-too-distant future, that these tactics under development will help provide a better management of chronic and recurrent hepatitis C, particularly in liver transplant setting.

Advertisement

Acknowledgments

CHL is a recipient of the 2016 CanHepC (Canadian Network on Hepatitis C) Summer Research Scholarship and the MOST 105 College Research Scholarship (105-2815-C-038-018-B). TB (NSC 102-2320-B-038-041-MY3) and LTL (NSC101-2320-B-038-038-MY2; MOST103-2320-B-038-031-MY3) are supported by funding from the Ministry of Science and Technology of Taiwan.

References

  1. 1. U.S. Health Resources & Services Administration. Organ Procurement and Transplantation Network [Internet]. 2016. Available from: https://optn.transplant.hrsa.gov/data/view-data-reports/ [Accessed: 2016-08-15]
  2. 2. Feeney ER, Chung RT. Antiviral treatment of hepatitis C. BMJ. 2014;348:g3308. DOI: 10.1136/bmj.g3308
  3. 3. Zoulim F, Liang TJ, Gerbes AL, Aghemo A, Deuffic-Burban S, Dusheiko G, et al. Hepatitis C virus treatment in the real world: optimising treatment and access to therapies. Gut. 2015;64(11):1824-33. DOI: 10.1136/gutjnl-2015-310421
  4. 4. Hoofnagle JH, Sherker AH. Therapy for hepatitis C—the costs of success. N Engl J Med. 2014;370(16):1552-3. DOI: 10.1056/NEJMe1401508
  5. 5. Liang TJ, Ghany MG. Current and future therapies for hepatitis C virus infection. N Engl J Med. 2013;368(20):1907-17. DOI: 10.1056/NEJMra1213651
  6. 6. Thursz M, Fontanet A. HCV transmission in industrialized countries and resource-constrained areas. Nat Rev Gastroenterol Hepatol. 2014;11(1):28-35. DOI: 10.1038/nrgastro.2013.179
  7. 7. Verna EC, Brown RS, Jr. Hepatitis C and liver transplantation: enhancing outcomes and should patients be retransplanted. Clin Liver Dis. 2008;12(3):637-59, ix-x. DOI: 10.1016/j.cld.2008.03.010
  8. 8. Firpi RJ, Clark V, Soldevila-Pico C, Morelli G, Cabrera R, Levy C, et al. The natural history of hepatitis C cirrhosis after liver transplantation. Liver Transpl. 2009;15(9):1063-71. DOI: 10.1002/lt.21784
  9. 9. Xiao F, Fofana I, Thumann C, Mailly L, Alles R, Robinet E, et al. Synergy of entry inhibitors with direct-acting antivirals uncovers novel combinations for prevention and treatment of hepatitis C. Gut. 2015;64(3):483-94. DOI: 10.1136/gutjnl-2013-306155
  10. 10. Merz A, Long G, Hiet MS, Brugger B, Chlanda P, Andre P, et al. Biochemical and morphological properties of hepatitis C virus particles and determination of their lipidome. J Biol Chem. 2011;286(4):3018-32. DOI: 10.1074/jbc.M110.175018
  11. 11. Catanese MT, Uryu K, Kopp M, Edwards TJ, Andrus L, Rice WJ, et al. Ultrastructural analysis of hepatitis C virus particles. Proc Natl Acad Sci U S A. 2013;110(23):9505-10. DOI: 10.1073/pnas.1307527110
  12. 12. Andre P, Komurian-Pradel F, Deforges S, Perret M, Berland JL, Sodoyer M, et al. Characterization of low- and very-low-density hepatitis C virus RNA-containing particles. J Virol. 2002;76(14):6919-28. DOI: 10.1128/JVI.76.14.6919-6928.2002
  13. 13. Chang KS, Jiang J, Cai Z, Luo G. Human apolipoprotein e is required for infectivity and production of hepatitis C virus in cell culture. J Virol. 2007;81(24):13783-93. DOI: 10.1128/JVI.01091-07
  14. 14. Meunier JC, Russell RS, Engle RE, Faulk KN, Purcell RH, Emerson SU. Apolipoprotein c1 association with hepatitis C virus. J Virol. 2008;82(19):9647-56. DOI: 10.1128/JVI.00914-08
  15. 15. Nielsen SU, Bassendine MF, Burt AD, Martin C, Pumeechockchai W, Toms GL. Association between hepatitis C virus and very-low-density lipoprotein (VLDL)/LDL analyzed in iodixanol density gradients. J Virol. 2006;80(5):2418-28. DOI: 10.1128/JVI.80.5.2418-2428.2006
  16. 16. Jiang J, Luo G. Apolipoprotein E but not B is required for the formation of infectious hepatitis C virus particles. J Virol. 2009;83(24):12680-91. DOI: 10.1128/JVI.01476-09
  17. 17. Benga WJ, Krieger SE, Dimitrova M, Zeisel MB, Parnot M, Lupberger J, et al. Apolipoprotein E interacts with hepatitis C virus nonstructural protein 5A and determines assembly of infectious particles. Hepatology. 2010;51(1):43-53. DOI: 10.1002/hep.23278
  18. 18. Huang H, Sun F, Owen DM, Li W, Chen Y, Gale M, Jr., et al. Hepatitis C virus production by human hepatocytes dependent on assembly and secretion of very low-density lipoproteins. Proc Natl Acad Sci U S A. 2007;104(14):5848-53. DOI: 10.1073/pnas.0700760104
  19. 19. Gastaminza P, Cheng G, Wieland S, Zhong J, Liao W, Chisari FV. Cellular determinants of hepatitis C virus assembly, maturation, degradation, and secretion. J Virol. 2008;82(5):2120-9. DOI: 10.1128/JVI.02053-07
  20. 20. Icard V, Diaz O, Scholtes C, Perrin-Cocon L, Ramiere C, Bartenschlager R, et al. Secretion of hepatitis C virus envelope glycoproteins depends on assembly of apolipoprotein B positive lipoproteins. PLoS One. 2009;4(1):e4233. DOI: 10.1371/journal.pone.0004233
  21. 21. Hueging K, Doepke M, Vieyres G, Bankwitz D, Frentzen A, Doerrbecker J, et al. Apolipoprotein E codetermines tissue tropism of hepatitis C virus and is crucial for viral cell-to-cell transmission by contributing to a postenvelopment step of assembly. J Virol. 2014;88(3):1433-46. DOI: 10.1128/JVI.01815-13
  22. 22. Douam F, Lavillette D, Cosset FL. The mechanism of HCV entry into host cells. Prog Mol Biol Transl Sci. 2015;129:63-107. DOI: 10.1016/bs.pmbts.2014.10.003
  23. 23. Barth H, Schafer C, Adah MI, Zhang F, Linhardt RJ, Toyoda H, et al. Cellular binding of hepatitis C virus envelope glycoprotein E2 requires cell surface heparan sulfate. J Biol Chem. 2003;278(42):41003-12. DOI: 10.1074/jbc.M302267200
  24. 24. Barth H, Schnober EK, Zhang F, Linhardt RJ, Depla E, Boson B, et al. Viral and cellular determinants of the hepatitis C virus envelope-heparan sulfate interaction. J Virol. 2006;80(21):10579-90. DOI: 10.1128/JVI.00941-06
  25. 25. Koutsoudakis G, Kaul A, Steinmann E, Kallis S, Lohmann V, Pietschmann T, et al. Characterization of the early steps of hepatitis C virus infection by using luciferase reporter viruses. J Virol. 2006;80(11):5308-20. DOI: 10.1128/JVI.02460-05
  26. 26. Shi Q, Jiang J, Luo G. Syndecan-1 serves as the major receptor for attachment of hepatitis C virus to the surfaces of hepatocytes. J Virol. 2013;87(12):6866-75. DOI: 10.1128/JVI.03475-12
  27. 27. Lefevre M, Felmlee DJ, Parnot M, Baumert TF, Schuster C. Syndecan 4 is involved in mediating HCV entry through interaction with lipoviral particle-associated apolipoprotein E. PLoS One. 2014;9(4):e95550. DOI: 10.1371/journal.pone.0095550
  28. 28. Agnello V, Abel G, Elfahal M, Knight GB, Zhang QX. Hepatitis C virus and other flaviviridae viruses enter cells via low density lipoprotein receptor. Proc Natl Acad Sci U S A. 1999;96(22):12766-71. DOI: 10.1073/pnas.96.22.12766
  29. 29. Owen DM, Huang H, Ye J, Gale M, Jr. Apolipoprotein E on hepatitis C virion facilitates infection through interaction with low-density lipoprotein receptor. Virology. 2009;394(1):99-108. DOI: 10.1016/j.virol.2009.08.037
  30. 30. Molina S, Castet V, Fournier-Wirth C, Pichard-Garcia L, Avner R, Harats D, et al. The low-density lipoprotein receptor plays a role in the infection of primary human hepatocytes by hepatitis C virus. J Hepatol. 2007;46(3):411-9. DOI: 10.1016/j.jhep.2006.09.024
  31. 31. Albecka A, Belouzard S, Op de Beeck A, Descamps V, Goueslain L, Bertrand-Michel J, et al. Role of low-density lipoprotein receptor in the hepatitis C virus life cycle. Hepatology. 2012;55(4):998-1007. DOI: 10.1002/hep.25501
  32. 32. Syed GH, Tang H, Khan M, Hassanein T, Liu J, Siddiqui A. Hepatitis C virus stimulates low-density lipoprotein receptor expression to facilitate viral propagation. J Virol. 2014;88(5):2519-29. DOI: 10.1128/JVI.02727-13
  33. 33. Jiang J, Cun W, Wu X, Shi Q, Tang H, Luo G. Hepatitis C virus attachment mediated by apolipoprotein E binding to cell surface heparan sulfate. J Virol. 2012;86(13):7256-67. DOI: 10.1128/JVI.07222-11
  34. 34. Jiang J, Wu X, Tang H, Luo G. Apolipoprotein E mediates attachment of clinical hepatitis C virus to hepatocytes by binding to cell surface heparan sulfate proteoglycan receptors. PLoS One. 2013;8(7):e67982. DOI: 10.1371/journal.pone.0067982
  35. 35. Xu Y, Martinez P, Seron K, Luo G, Allain F, Dubuisson J, et al. Characterization of hepatitis C virus interaction with heparan sulfate proteoglycans. J Virol. 2015;89(7):3846-58. DOI: 10.1128/JVI.03647-14
  36. 36. Hishiki T, Shimizu Y, Tobita R, Sugiyama K, Ogawa K, Funami K, et al. Infectivity of hepatitis C virus is influenced by association with apolipoprotein E isoforms. J Virol. 2010;84(22):12048-57. DOI: 10.1128/JVI.01063-10
  37. 37. Dao Thi VL, Granier C, Zeisel MB, Guerin M, Mancip J, Granio O, et al. Characterization of hepatitis C virus particle subpopulations reveals multiple usage of the scavenger receptor BI for entry steps. J Biol Chem. 2012;287(37):31242-57. DOI: 10.1074/jbc.M112.365924
  38. 38. Catanese MT, Ansuini H, Graziani R, Huby T, Moreau M, Ball JK, et al. Role of scavenger receptor class B type I in hepatitis C virus entry: kinetics and molecular determinants. J Virol. 2010;84(1):34-43. DOI: 10.1128/JVI.02199-08
  39. 39. Maillard P, Huby T, Andreo U, Moreau M, Chapman J, Budkowska A. The interaction of natural hepatitis C virus with human scavenger receptor SR-BI/Cla1 is mediated by ApoB-containing lipoproteins. FASEB J. 2006;20(6):735-7. DOI: 10.1096/fj.05-4728fje
  40. 40. Scarselli E, Ansuini H, Cerino R, Roccasecca RM, Acali S, Filocamo G, et al. The human scavenger receptor class B type I is a novel candidate receptor for the hepatitis C virus. EMBO J. 2002;21(19):5017-25. DOI: 10.1093/emboj/cdf529
  41. 41. Bartosch B, Vitelli A, Granier C, Goujon C, Dubuisson J, Pascale S, et al. Cell entry of hepatitis C virus requires a set of co-receptors that include the CD81 tetraspanin and the SR-B1 scavenger receptor. J Biol Chem. 2003;278(43):41624-30. DOI: 10.1074/jbc.M305289200
  42. 42. Zahid MN, Turek M, Xiao F, Thi VL, Guerin M, Fofana I, et al. The postbinding activity of scavenger receptor class B type I mediates initiation of hepatitis C virus infection and viral dissemination. Hepatology. 2013;57(2):492-504. DOI: 10.1002/hep.26097
  43. 43. Bankwitz D, Vieyres G, Hueging K, Bitzegeio J, Doepke M, Chhatwal P, et al. Role of hypervariable region 1 for the interplay of hepatitis C virus with entry factors and lipoproteins. J Virol. 2014;88(21):12644-55. DOI: 10.1128/JVI.01145-14
  44. 44. Zeisel MB, Koutsoudakis G, Schnober EK, Haberstroh A, Blum HE, Cosset FL, et al. Scavenger receptor class B type I is a key host factor for hepatitis C virus infection required for an entry step closely linked to CD81. Hepatology. 2007;46(6):1722-31. DOI: 10.1002/hep.21994
  45. 45. Kapadia SB, Barth H, Baumert T, McKeating JA, Chisari FV. Initiation of hepatitis C virus infection is dependent on cholesterol and cooperativity between CD81 and scavenger receptor B type I. J Virol. 2007;81(1):374-83. DOI: 10.1128/JVI.01134-06
  46. 46. Koutsoudakis G, Herrmann E, Kallis S, Bartenschlager R, Pietschmann T. The level of CD81 cell surface expression is a key determinant for productive entry of hepatitis C virus into host cells. J Virol. 2007;81(2):588-98. DOI: 10.1128/JVI.01534-06
  47. 47. Molina S, Castet V, Pichard-Garcia L, Wychowski C, Meurs E, Pascussi JM, et al. Serum-derived hepatitis C virus infection of primary human hepatocytes is tetraspanin CD81 dependent. J Virol. 2008;82(1):569-74. DOI: 10.1128/JVI.01443-07
  48. 48. Feneant L, Levy S, Cocquerel L. CD81 and hepatitis C virus (HCV) infection. Viruses. 2014;6(2):535-72. DOI: 10.3390/v6020535
  49. 49. Harris HJ, Davis C, Mullins JG, Hu K, Goodall M, Farquhar MJ, et al. Claudin association with CD81 defines hepatitis C virus entry. J Biol Chem. 2010;285(27):21092-102. DOI: 10.1074/jbc.M110.104836
  50. 50. Harris HJ, Farquhar MJ, Mee CJ, Davis C, Reynolds GM, Jennings A, et al. CD81 and claudin 1 coreceptor association: role in hepatitis C virus entry. J Virol. 2008;82(10):5007-20. DOI: 10.1128/JVI.02286-07
  51. 51. Farquhar MJ, Hu K, Harris HJ, Davis C, Brimacombe CL, Fletcher SJ, et al. Hepatitis C virus induces CD81 and claudin-1 endocytosis. J Virol. 2012;86(8):4305-16. DOI: 10.1128/JVI.06996-11
  52. 52. Brazzoli M, Bianchi A, Filippini S, Weiner A, Zhu Q, Pizza M, et al. CD81 is a central regulator of cellular events required for hepatitis C virus infection of human hepatocytes. J Virol. 2008;82(17):8316-29. DOI: 10.1128/JVI.00665-08
  53. 53. Liu Z, Tian Y, Machida K, Lai MM, Luo G, Foung SK, et al. Transient activation of the PI3K-AKT pathway by hepatitis C virus to enhance viral entry. J Biol Chem. 2012;287(50):41922-30. DOI: 10.1074/jbc.M112.414789
  54. 54. Lupberger J, Zeisel MB, Xiao F, Thumann C, Fofana I, Zona L, et al. EGFR and EphA2 are host factors for hepatitis C virus entry and possible targets for antiviral therapy. Nat Med. 2011;17(5):589-95. DOI: 10.1038/nm.2341
  55. 55. Diao J, Pantua H, Ngu H, Komuves L, Diehl L, Schaefer G, et al. Hepatitis C virus induces epidermal growth factor receptor activation via CD81 binding for viral internalization and entry. J Virol. 2012;86(20):10935-49. DOI: 10.1128/JVI.00750-12
  56. 56. Rocha-Perugini V, Montpellier C, Delgrange D, Wychowski C, Helle F, Pillez A, et al. The CD81 partner EWI-2wint inhibits hepatitis C virus entry. PLoS One. 2008;3(4):e1866. DOI: 10.1371/journal.pone.0001866
  57. 57. Potel J, Rassam P, Montpellier C, Kaestner L, Werkmeister E, Tews BA, et al. EWI-2wint promotes CD81 clustering that abrogates Hepatitis C Virus entry. Cell Microbiol. 2013;15(7):1234-52. DOI: 10.1111/cmi.12112
  58. 58. Sourisseau M, Michta ML, Zony C, Israelow B, Hopcraft SE, Narbus CM, et al. Temporal analysis of hepatitis C virus cell entry with occludin directed blocking antibodies. PLoS Pathog. 2013;9(3):e1003244. DOI: 10.1371/journal.ppat.1003244
  59. 59. Martin DN, Uprichard SL. Identification of transferrin receptor 1 as a hepatitis C virus entry factor. Proc Natl Acad Sci U S A. 2013;110(26):10777-82. DOI: 10.1073/pnas.1301764110
  60. 60. Sainz B, Jr., Barretto N, Martin DN, Hiraga N, Imamura M, Hussain S, et al. Identification of the Niemann-Pick C1-like 1 cholesterol absorption receptor as a new hepatitis C virus entry factor. Nat Med. 2012;18(2):281-5. DOI: 10.1038/nm.2581
  61. 61. Blanchard E, Belouzard S, Goueslain L, Wakita T, Dubuisson J, Wychowski C, et al. Hepatitis C virus entry depends on clathrin-mediated endocytosis. J Virol. 2006;80(14):6964-72. DOI: 10.1128/JVI.00024-06
  62. 62. Meertens L, Bertaux C, Dragic T. Hepatitis C virus entry requires a critical postinternalization step and delivery to early endosomes via clathrin-coated vesicles. J Virol. 2006;80(23):11571-8. DOI: 10.1128/JVI.01717-06
  63. 63. Coller KE, Berger KL, Heaton NS, Cooper JD, Yoon R, Randall G. RNA interference and single particle tracking analysis of hepatitis C virus endocytosis. PLoS Pathog. 2009;5(12):e1000702. DOI: 10.1371/journal.ppat.1000702
  64. 64. Evans MJ, von Hahn T, Tscherne DM, Syder AJ, Panis M, Wolk B, et al. Claudin-1 is a hepatitis C virus co-receptor required for a late step in entry. Nature. 2007;446(7137):801-5. DOI: 10.1038/nature05654
  65. 65. Ploss A, Evans MJ, Gaysinskaya VA, Panis M, You H, de Jong YP, et al. Human occludin is a hepatitis C virus entry factor required for infection of mouse cells. Nature. 2009;457(7231):882-6. DOI: 10.1038/nature07684
  66. 66. Dorner M, Horwitz JA, Robbins JB, Barry WT, Feng Q, Mu K, et al. A genetically humanized mouse model for hepatitis C virus infection. Nature. 2011;474(7350):208-11. DOI: 10.1038/nature10168
  67. 67. Da Costa D, Turek M, Felmlee DJ, Girardi E, Pfeffer S, Long G, et al. Reconstitution of the entire hepatitis C virus life cycle in nonhepatic cells. J Virol. 2012;86(21):11919-25. DOI: 10.1128/JVI.01066-12
  68. 68. Timpe JM, Stamataki Z, Jennings A, Hu K, Farquhar MJ, Harris HJ, et al. Hepatitis C virus cell-cell transmission in hepatoma cells in the presence of neutralizing antibodies. Hepatology. 2008;47(1):17-24. DOI: 10.1002/hep.21959
  69. 69. Brimacombe CL, Grove J, Meredith LW, Hu K, Syder AJ, Flores MV, et al. Neutralizing antibody-resistant hepatitis C virus cell-to-cell transmission. J Virol. 2011;85(1):596-605. DOI: 10.1128/JVI.01592-10
  70. 70. Catanese MT, Loureiro J, Jones CT, Dorner M, von Hahn T, Rice CM. Different requirements for scavenger receptor class B type I in hepatitis C virus cell-free versus cell-to-cell transmission. J Virol. 2013;87(15):8282-93. DOI: 10.1128/JVI.01102-13
  71. 71. Barretto N, Sainz B, Jr., Hussain S, Uprichard SL. Determining the involvement and therapeutic implications of host cellular factors in hepatitis C virus cell-to-cell spread. J Virol. 2014;88(9):5050-61. DOI: 10.1128/JVI.03241-13
  72. 72. Pileri P, Uematsu Y, Campagnoli S, Galli G, Falugi F, Petracca R, et al. Binding of hepatitis C virus to CD81. Science. 1998;282(5390):938-41. DOI: 10.1126/science.282.5390.938
  73. 73. Flint M, Maidens C, Loomis-Price LD, Shotton C, Dubuisson J, Monk P, et al. Characterization of hepatitis C virus E2 glycoprotein interaction with a putative cellular receptor, CD81. J Virol. 1999;73(8):6235-44.
  74. 74. Levy S, Todd SC, Maecker HT. CD81 (TAPA-1): a molecule involved in signal transduction and cell adhesion in the immune system. Annu Rev Immunol. 1998;16:89-109. DOI: 10.1146/annurev.immunol.16.1.89
  75. 75. Meuleman P, Hesselgesser J, Paulson M, Vanwolleghem T, Desombere I, Reiser H, et al. Anti-CD81 antibodies can prevent a hepatitis C virus infection in vivo. Hepatology. 2008;48(6):1761-8. DOI: 10.1002/hep.22547
  76. 76. Fofana I, Xiao F, Thumann C, Turek M, Zona L, Tawar RG, et al. A novel monoclonal anti-CD81 antibody produced by genetic immunization efficiently inhibits Hepatitis C virus cell-cell transmission. PLoS One. 2013;8(5):e64221. DOI: 10.1371/journal.pone.0064221
  77. 77. Ji C, Liu Y, Pamulapati C, Bohini S, Fertig G, Schraeml M, et al. Prevention of hepatitis C virus infection and spread in human liver chimeric mice by an anti-CD81 monoclonal antibody. Hepatology. 2015;61(4):1136-44. DOI: 10.1002/hep.27603
  78. 78. Acton S, Rigotti A, Landschulz KT, Xu S, Hobbs HH, Krieger M. Identification of scavenger receptor SR-BI as a high density lipoprotein receptor. Science. 1996;271(5248):518-20. DOI: 10.1126/science.271.5248.518
  79. 79. Krieger M. Scavenger receptor class B type I is a multiligand HDL receptor that influences diverse physiologic systems. J Clin Invest. 2001;108(6):793-7. DOI: 10.1172/JCI14011
  80. 80. Catanese MT, Graziani R, von Hahn T, Moreau M, Huby T, Paonessa G, et al. High-avidity monoclonal antibodies against the human scavenger class B type I receptor efficiently block hepatitis C virus infection in the presence of high-density lipoprotein. J Virol. 2007;81(15):8063-71. DOI: 10.1128/JVI.00193-07
  81. 81. Meuleman P, Catanese MT, Verhoye L, Desombere I, Farhoudi A, Jones CT, et al. A human monoclonal antibody targeting scavenger receptor class B type I precludes hepatitis C virus infection and viral spread in vitro and in vivo. Hepatology. 2012;55(2):364-72. DOI: 10.1002/hep.24692
  82. 82. Lacek K, Vercauteren K, Grzyb K, Naddeo M, Verhoye L, Slowikowski MP, et al. Novel human SR-BI antibodies prevent infection and dissemination of HCV in vitro and in humanized mice. J Hepatol. 2012;57(1):17-23. DOI: 10.1016/j.jhep.2012.02.018
  83. 83. Furuse M, Fujita K, Hiiragi T, Fujimoto K, Tsukita S. Claudin-1 and -2: novel integral membrane proteins localizing at tight junctions with no sequence similarity to occludin. J Cell Biol. 1998;141(7):1539-50. DOI: 10.1083/jcb.141.7.1539
  84. 84. Krieger SE, Zeisel MB, Davis C, Thumann C, Harris HJ, Schnober EK, et al. Inhibition of hepatitis C virus infection by anti-claudin-1 antibodies is mediated by neutralization of E2-CD81-claudin-1 associations. Hepatology. 2010;51(4):1144-57. DOI: 10.1002/hep.23445
  85. 85. Fofana I, Krieger SE, Grunert F, Glauben S, Xiao F, Fafi-Kremer S, et al. Monoclonal anti-claudin 1 antibodies prevent hepatitis C virus infection of primary human hepatocytes. Gastroenterology. 2010;139(3):953-64, 64 e1-4. DOI: 10.1053/j.gastro.2010.05.073
  86. 86. Mailly L, Xiao F, Lupberger J, Wilson GK, Aubert P, Duong FH, et al. Clearance of persistent hepatitis C virus infection in humanized mice using a claudin-1-targeting monoclonal antibody. Nat Biotechnol. 2015;33(5):549-54. DOI: 10.1038/nbt.3179
  87. 87. Fukasawa M, Nagase S, Shirasago Y, Iida M, Yamashita M, Endo K, et al. Monoclonal antibodies against extracellular domains of claudin-1 block hepatitis C virus infection in a mouse model. J Virol. 2015;89(9):4866-79. DOI: 10.1128/JVI.03676-14
  88. 88. Flego M, Ascione A, Cianfriglia M, Vella S. Clinical development of monoclonal antibody-based drugs in HIV and HCV diseases. BMC Med. 2013;11:4. DOI: 10.1186/1741-7015-11-4
  89. 89. Broering TJ, Garrity KA, Boatright NK, Sloan SE, Sandor F, Thomas WD, Jr., et al. Identification and characterization of broadly neutralizing human monoclonal antibodies directed against the E2 envelope glycoprotein of hepatitis C virus. J Virol. 2009;83(23):12473-82. DOI: 10.1128/JVI.01138-09
  90. 90. Eren R, Landstein D, Terkieltaub D, Nussbaum O, Zauberman A, Ben-Porath J, et al. Preclinical evaluation of two neutralizing human monoclonal antibodies against hepatitis C virus (HCV): a potential treatment to prevent HCV reinfection in liver transplant patients. J Virol. 2006;80(6):2654-64. DOI: 10.1128/JVI.80.6.2654-2664.2006
  91. 91. Ilan E, Arazi J, Nussbaum O, Zauberman A, Eren R, Lubin I, et al. The hepatitis C virus (HCV)-Trimera mouse: a model for evaluation of agents against HCV. J Infect Dis. 2002;185(2):153-61. DOI: 10.1086/338266
  92. 92. Morin TJ, Broering TJ, Leav BA, Blair BM, Rowley KJ, Boucher EN, et al. Human monoclonal antibody HCV1 effectively prevents and treats HCV infection in chimpanzees. PLoS Pathog. 2012;8(8):e1002895. DOI: 10.1371/journal.ppat.1002895
  93. 93. Owsianka A, Tarr AW, Juttla VS, Lavillette D, Bartosch B, Cosset FL, et al. Monoclonal antibody AP33 defines a broadly neutralizing epitope on the hepatitis C virus E2 envelope glycoprotein. J Virol. 2005;79(17):11095-104. DOI: 10.1128/JVI.79.17.11095-11104.2005
  94. 94. Sabo MC, Luca VC, Prentoe J, Hopcraft SE, Blight KJ, Yi M, et al. Neutralizing monoclonal antibodies against hepatitis C virus E2 protein bind discontinuous epitopes and inhibit infection at a postattachment step. J Virol. 2011;85(14):7005-19. DOI: 10.1128/JVI.00586-11
  95. 95. Schiano TD, Charlton M, Younossi Z, Galun E, Pruett T, Tur-Kaspa R, et al. Monoclonal antibody HCV-AbXTL68 in patients undergoing liver transplantation for HCV: results of a phase 2 randomized study. Liver Transpl. 2006;12(9):1381-9. DOI: 10.1002/lt.20876
  96. 96. Chung RT, Gordon FD, Curry MP, Schiano TD, Emre S, Corey K, et al. Human monoclonal antibody MBL-HCV1 delays HCV viral rebound following liver transplantation: a randomized controlled study. Am J Transplant. 2013;13(4):1047-54. DOI: 10.1111/ajt.12083
  97. 97. Marasco WA, Sui J. The growth and potential of human antiviral monoclonal antibody therapeutics. Nat Biotechnol. 2007;25(12):1421-34. DOI: 10.1038/nbt1363
  98. 98. Ball JK, Tarr AW, McKeating JA. The past, present and future of neutralizing antibodies for hepatitis C virus. Antiviral Res. 2014;105:100-11. DOI: 10.1016/j.antiviral.2014.02.013
  99. 99. Ciesek S, von Hahn T, Colpitts CC, Schang LM, Friesland M, Steinmann J, et al. The green tea polyphenol, epigallocatechin-3-gallate, inhibits hepatitis C virus entry. Hepatology. 2011;54(6):1947-55. DOI: 10.1002/hep.24610
  100. 100. Colpitts CC, Schang LM. A small molecule inhibits virion attachment to heparan sulfate- or sialic acid-containing glycans. J Virol. 2014;88(14):7806-17. DOI: 10.1128/JVI.00896-14
  101. 101. Calland N, Sahuc ME, Belouzard S, Pene V, Bonnafous P, Mesalam AA, et al. Polyphenols inhibit Hepatitis C virus entry by a new mechanism of action. J Virol. 2015;89(19):10053-63. DOI: 10.1128/JVI.01473-15
  102. 102. Lin LT, Chung CY, Hsu WC, Chang SP, Hung TC, Shields J, et al. Saikosaponin b2 is a naturally occurring terpenoid that efficiently inhibits hepatitis C virus entry. J Hepatol. 2015;62(3):541-8. DOI: 10.1016/j.jhep.2014.10.040
  103. 103. Hsu WC, Chang SP, Lin LC, Li CL, Richardson CD, Lin CC, et al. Limonium sinense and gallic acid suppress hepatitis C virus infection by blocking early viral entry. Antiviral Res. 2015;118:139-47. DOI: 10.1016/j.antiviral.2015.04.003
  104. 104. Lin LT, Chen TY, Lin SC, Chung CY, Lin TC, Wang GH, et al. Broad-spectrum antiviral activity of chebulagic acid and punicalagin against viruses that use glycosaminoglycans for entry. BMC Microbiol. 2013;13:187. DOI: 10.1186/1471-2180-13-187
  105. 105. Chung CY, Liu CH, Burnouf T, Wang GH, Chang SP, Jassey A, et al. Activity-based and fraction-guided analysis of Phyllanthus urinaria identifies loliolide as a potent inhibitor of hepatitis C virus entry. Antiviral Res. 2016;130:58-68. DOI: 10.1016/j.antiviral.2016.03.012
  106. 106. Chung CY, Liu CH, Wang GH, Jassey A, Li CL, Chen L, et al. (4R,6S)-2-Dihydromenisdaurilide is a Butenolide that Efficiently Inhibits Hepatitis C Virus Entry. Sci Rep. 2016;6:29969. DOI: 10.1038/srep29969
  107. 107. Anggakusuma, Colpitts CC, Schang LM, Rachmawati H, Frentzen A, Pfaender S, et al. Turmeric curcumin inhibits entry of all hepatitis C virus genotypes into human liver cells. Gut. 2014;63(7):1137-49. DOI: 10.1136/gutjnl-2012-304299
  108. 108. Barry J, Fritz M, Brender JR, Smith PE, Lee DK, Ramamoorthy A. Determining the effects of lipophilic drugs on membrane structure by solid-state NMR spectroscopy: the case of the antioxidant curcumin. J Am Chem Soc. 2009;131(12):4490-8. DOI: 10.1021/ja809217u
  109. 109. Helle F, Wychowski C, Vu-Dac N, Gustafson KR, Voisset C, Dubuisson J. Cyanovirin-N inhibits hepatitis C virus entry by binding to envelope protein glycans. J Biol Chem. 2006;281(35):25177-83. DOI: 10.1074/jbc.M602431200
  110. 110. Meuleman P, Albecka A, Belouzard S, Vercauteren K, Verhoye L, Wychowski C, et al. Griffithsin has antiviral activity against hepatitis C virus. Antimicrob Agents Chemother. 2011;55(11):5159-67. DOI: 10.1128/AAC.00633-11
  111. 111. Brown KS, Keogh MJ, Owsianka AM, Adair R, Patel AH, Arnold JN, et al. Specific interaction of hepatitis C virus glycoproteins with mannan binding lectin inhibits virus entry. Protein Cell. 2010;1(7):664-74. DOI: 10.1007/s13238-010-0088-9
  112. 112. Hamed MR, Brown RJ, Zothner C, Urbanowicz RA, Mason CP, Krarup A, et al. Recombinant human L-ficolin directly neutralizes hepatitis C virus entry. J Innate Immun. 2014;6(5):676-84. DOI: 10.1159/000362209
  113. 113. Khanal M, Vausselin T, Barras A, Bande O, Turcheniuk K, Benazza M, et al. Phenylboronic-acid-modified nanoparticles: potential antiviral therapeutics. ACS Appl Mater Interfaces. 2013;5(23):12488-98. DOI: 10.1021/am403770q
  114. 114. Khanal M, Barras A, Vausselin T, Feneant L, Boukherroub R, Siriwardena A, et al. Boronic acid-modified lipid nanocapsules: a novel platform for the highly efficient inhibition of hepatitis C viral entry. Nanoscale. 2015;7(4):1392-402. DOI: 10.1039/c4nr03875d
  115. 115. Yu F, Wang Q, Zhang Z, Peng Y, Qiu Y, Shi Y, et al. Development of oleanane-type triterpenes as a new class of HCV entry inhibitors. J Med Chem. 2013;56(11):4300-19. DOI: 10.1021/jm301910a
  116. 116. VanCompernolle SE, Wiznycia AV, Rush JR, Dhanasekaran M, Baures PW, Todd SC. Small molecule inhibition of hepatitis C virus E2 binding to CD81. Virology. 2003;314(1):371-80. DOI: 10.1016/S0042-6822(03)00406-9
  117. 117. Cui HK, Qing J, Guo Y, Wang YJ, Cui LJ, He TH, et al. Stapled peptide-based membrane fusion inhibitors of hepatitis C virus. Bioorg Med Chem. 2013;21(12):3547-54. DOI: 10.1016/j.bmc.2013.02.011
  118. 118. Si Y, Liu S, Liu X, Jacobs JL, Cheng M, Niu Y, et al. A human claudin-1-derived peptide inhibits hepatitis C virus entry. Hepatology. 2012;56(2):507-15. DOI: 10.1002/hep.25685
  119. 119. Chi X, Niu Y, Cheng M, Liu X, Feng Y, Zheng F, et al. Identification of a potent and broad-spectrum hepatitis C virus fusion inhibitory peptide from the E2 stem domain. Sci Rep. 2016;6:25224. DOI: 10.1038/srep25224
  120. 120. Holzer M, Ziegler S, Albrecht B, Kronenberger B, Kaul A, Bartenschlager R, et al. Identification of terfenadine as an inhibitor of human CD81-receptor HCV-E2 interaction: synthesis and structure optimization. Molecules. 2008;13(5):1081-110. DOI: 10.3390/molecules13051081
  121. 121. Masson D, Koseki M, Ishibashi M, Larson CJ, Miller SG, King BD, et al. Increased HDL cholesterol and apoA-I in humans and mice treated with a novel SR-BI inhibitor. Arterioscler Thromb Vasc Biol. 2009;29(12):2054-60. DOI: 10.1161/ATVBAHA.109.191320
  122. 122. Syder AJ, Lee H, Zeisel MB, Grove J, Soulier E, Macdonald J, et al. Small molecule scavenger receptor BI antagonists are potent HCV entry inhibitors. J Hepatol. 2011;54(1):48-55. DOI: 10.1016/j.jhep.2010.06.024
  123. 123. Zhu H, Wong-Staal F, Lee H, Syder A, McKelvy J, Schooley RT, et al. Evaluation of ITX 5061, a scavenger receptor B1 antagonist: resistance selection and activity in combination with other hepatitis C virus antivirals. J Infect Dis. 2012;205(4):656-62. DOI: 10.1093/infdis/jir802
  124. 124. Rowe IA, Tully DC, Armstrong MJ, Parker R, Guo K, Barton D, et al. Effect of scavenger receptor class B type I antagonist ITX5061 in patients with hepatitis C virus infection undergoing liver transplantation. Liver Transpl. 2016;22(3):287-97. DOI: 10.1002/lt.24349
  125. 125. Yin P, Zhang L. Aspirin inhibits hepatitis C virus entry by downregulating claudin-1. J Viral Hepat. 2016;23(1):62-4. DOI: 10.1111/jvh.12446
  126. 126. Zona L, Lupberger J, Sidahmed-Adrar N, Thumann C, Harris HJ, Barnes A, et al. HRas signal transduction promotes hepatitis C virus cell entry by triggering assembly of the host tetraspanin receptor complex. Cell Host Microbe. 2013;13(3):302-13. DOI: 10.1016/j.chom.2013.02.006
  127. 127. Blanchet M, Sureau C, Guevin C, Seidah NG, Labonte P. SKI-1/S1P inhibitor PF-429242 impairs the onset of HCV infection. Antiviral Res. 2015;115:94-104. DOI: 10.1016/j.antiviral.2014.12.017
  128. 128. Chamoun-Emanuelli AM, Pecheur EI, Simeon RL, Huang D, Cremer PS, Chen Z. Phenothiazines inhibit hepatitis C virus entry, likely by increasing the fluidity of cholesterol-rich membranes. Antimicrob Agents Chemother. 2013;57(6):2571-81. DOI: 10.1128/AAC.02593-12
  129. 129. Blaising J, Levy PL, Polyak SJ, Stanifer M, Boulant S, Pecheur EI. Arbidol inhibits viral entry by interfering with clathrin-dependent trafficking. Antiviral Res. 2013;100(1):215-9. DOI: 10.1016/j.antiviral.2013.08.008
  130. 130. Tscherne DM, Jones CT, Evans MJ, Lindenbach BD, McKeating JA, Rice CM. Time- and temperature-dependent activation of hepatitis C virus for low-pH-triggered entry. J Virol. 2006;80(4):1734-41. DOI: 10.1128/JVI.80.4.1734-1741.2006
  131. 131. Ashfaq UA, Javed T, Rehman S, Nawaz Z, Riazuddin S. Lysosomotropic agents as HCV entry inhibitors. Virol J. 2011;8:163. DOI: 10.1186/1743-422X-8-163
  132. 132. St Vincent MR, Colpitts CC, Ustinov AV, Muqadas M, Joyce MA, Barsby NL, et al. Rigid amphipathic fusion inhibitors, small molecule antiviral compounds against enveloped viruses. Proc Natl Acad Sci U S A. 2010;107(40):17339-44. DOI: 10.1073/pnas.1010026107
  133. 133. Vausselin T, Calland N, Belouzard S, Descamps V, Douam F, Helle F, et al. The antimalarial ferroquine is an inhibitor of hepatitis C virus. Hepatology. 2013;58(1):86-97. DOI: 10.1002/hep.26273
  134. 134. Baldick CJ, Wichroski MJ, Pendri A, Walsh AW, Fang J, Mazzucco CE, et al. A novel small molecule inhibitor of hepatitis C virus entry. PLoS Pathog. 2010;6(9):e1001086. DOI: 10.1371/journal.ppat.1001086
  135. 135. Coburn GA, Fisch DN, Moorji SM, de Muys JM, Murga JD, Paul D, et al. Novel small-molecule inhibitors of hepatitis C virus entry block viral spread and promote viral clearance in cell culture. PLoS One. 2012;7(4):e35351. DOI: 10.1371/journal.pone.0035351
  136. 136. European Medicines Agency. Silibinin-C-2',3-dihydrogensuccinate, disodium salt for the prevention of recurrent hepatitis C in liver transplant recipients [Internet]. 2010. Available from: http://www.ema.europa.eu/docs/en_GB/document_library/Orphan_designation/2011/01/WC500101019.pdf [Accessed: 2016-08-15]
  137. 137. Liu CH, Lin CC, Hsu WC, Chung CY, Lin CC, Jassey A, et al. Highly bioavailable silibinin nanoparticles inhibit HCV infection. Gut. 2016. DOI: 10.1136/gutjnl-2016-312019
  138. 138. Wagoner J, Negash A, Kane OJ, Martinez LE, Nahmias Y, Bourne N, et al. Multiple effects of silymarin on the hepatitis C virus lifecycle. Hepatology. 2010;51(6):1912-21. DOI: 10.1002/hep.23587
  139. 139. Blaising J, Levy PL, Gondeau C, Phelip C, Varbanov M, Teissier E, et al. Silibinin inhibits hepatitis C virus entry into hepatocytes by hindering clathrin-dependent trafficking. Cell Microbiol. 2013;15(11):1866-82. DOI: 10.1111/cmi.12155
  140. 140. Wagoner J, Morishima C, Graf TN, Oberlies NH, Teissier E, Pecheur EI, et al. Differential in vitro effects of intravenous versus oral formulations of silibinin on the HCV life cycle and inflammation. PLoS One. 2011;6(1):e16464. DOI: 10.1371/journal.pone.0016464
  141. 141. Xiao F, Fofana I, Heydmann L, Barth H, Soulier E, Habersetzer F, et al. Hepatitis C virus cell-cell transmission and resistance to direct-acting antiviral agents. PLoS Pathog. 2014;10(5):e1004128. DOI: 10.1371/journal.ppat.1004128
  142. 142. Murakami Y, Fukasawa M, Kaneko Y, Suzuki T, Wakita T, Fukazawa H. Selective estrogen receptor modulators inhibit hepatitis C virus infection at multiple steps of the virus life cycle. Microbes Infect. 2013;15(1):45-55. DOI: 10.1016/j.micinf.2012.10.003
  143. 143. Bush CO, Pokrovskii MV, Saito R, Morganelli P, Canales E, Clarke MO, et al. A small-molecule inhibitor of hepatitis C virus infectivity. Antimicrob Agents Chemother. 2014;58(1):386-96. DOI: 10.1128/AAC.02083-13
  144. 144. Haid S, Novodomska A, Gentzsch J, Grethe C, Geuenich S, Bankwitz D, et al. A plant-derived flavonoid inhibits entry of all HCV genotypes into human hepatocytes. Gastroenterology. 2012;143(1):213-22 e5. DOI: 10.1053/j.gastro.2012.03.036
  145. 145. Lavie M, Voisset C, Vu-Dac N, Zurawski V, Duverlie G, Wychowski C, et al. Serum amyloid A has antiviral activity against hepatitis C virus by inhibiting virus entry in a cell culture system. Hepatology. 2006;44(6):1626-34. DOI: 10.1002/hep.21406
  146. 146. Cai Z, Cai L, Jiang J, Chang KS, van der Westhuyzen DR, Luo G. Human serum amyloid A protein inhibits hepatitis C virus entry into cells. J Virol. 2007;81(11):6128-33. DOI: 10.1128/JVI.02627-06
  147. 147. Hong W, Lang Y, Li T, Zeng Z, Song Y, Wu Y, et al. A p7 ion channel-derived peptide inhibits hepatitis C virus infection in vitro. J Biol Chem. 2015;290(38):23254-63. DOI: 10.1074/jbc.M115.662452
  148. 148. Matsumura T, Hu Z, Kato T, Dreux M, Zhang YY, Imamura M, et al. Amphipathic DNA polymers inhibit hepatitis C virus infection by blocking viral entry. Gastroenterology. 2009;137(2):673-81. DOI: 10.1053/j.gastro.2009.04.048
  149. 149. El-Fakharany EM, Sanchez L, Al-Mehdar HA, Redwan EM. Effectiveness of human, camel, bovine and sheep lactoferrin on the hepatitis C virus cellular infectivity: comparison study. Virol J. 2013;10:199. DOI: 10.1186/1743-422X-10-199
  150. 150. Tamura S, Yang GM, Yasueda N, Matsuura Y, Komoda Y, Murakami N. Tellimagrandin I, HCV invasion inhibitor from Rosae Rugosae Flos. Bioorg Med Chem Lett. 2010;20(5):1598-600. DOI: 10.1016/j.bmcl.2010.01.084
  151. 151. Han Z, Liang X, Wang Y, Qing J, Cao L, Shang L, et al. The discovery of indole derivatives as novel hepatitis C virus inhibitors. Eur J Med Chem. 2016;116:147-55. DOI: 10.1016/j.ejmech.2016.03.062
  152. 152. Wang H, Wang S, Cheng L, Chen L, Wang Y, Qing J, et al. Discovery of imidazo[1,2-alpha][1,8]naphthyridine derivatives as potential HCV entry inhibitor. ACS Med Chem Lett. 2015;6(9):977-81. DOI: 10.1021/acsmedchemlett.5b00159
  153. 153. WHO. Guidelines on good manufacturing practices for blood establishments. Annex 4. WHO Technical Report Series. 2011;961:148-214.
  154. 154. Busch MP, Glynn SA, Stramer SL, Strong DM, Caglioti S, Wright DJ, et al. A new strategy for estimating risks of transfusion-transmitted viral infections based on rates of detection of recently infected donors. Transfusion. 2005;45(2):254-64. DOI: 10.1111/j.1537-2995.2004.04215.x
  155. 155. Dodd RY. Current risk for transfusion transmitted infections. Curr Opin Hematol. 2007;14(6):671-6. DOI: 10.1097/MOH.0b013e3282e38e8a
  156. 156. Burnouf T, Radosevich M. Reducing the risk of infection from plasma products: specific preventative strategies. Blood Rev. 2000;14(2):94-110. DOI: 10.1054/blre.2000.0129
  157. 157. Horowitz B, Wiebe ME, Lippin A, Stryker MH. Inactivation of viruses in labile blood derivatives. I. Disruption of lipid-enveloped viruses by tri(n-butyl)phosphate detergent combinations. Transfusion. 1985;25(6):516-22. DOI: 10.1046/j.1537-2995.1985.25686071422.x
  158. 158. Dichtelmuller HO, Biesert L, Fabbrizzi F, Gajardo R, Groner A, von Hoegen I, et al. Robustness of solvent/detergent treatment of plasma derivatives: a data collection from Plasma Protein Therapeutics Association member companies. Transfusion. 2009;49(9):1931-43. DOI: TRF02222 [pii]10.1111/j.1537-2995.2009.02222.x
  159. 159. Burnouf T. Modern plasma fractionation. Transfus Med Rev. 2007;21(2):101-17. DOI: 10.1016/j.tmrv.2006.11.001
  160. 160. Burnouf T, Radosevich M. Nanofiltration of plasma-derived biopharmaceutical products. Haemophilia. 2003;9(1):24-37. DOI: 10.1046/j.1365-2516.2003.00701.x
  161. 161. Reesink HW, Panzer S, McQuilten ZK, Wood EM, Marks C, Wendel S, et al. Pathogen inactivation of platelet concentrates. Vox Sanguinis. 2010;99(1):85-95. DOI: 10.1111/j.1423-0410.2010.01319.x
  162. 162. Horowitz B, Bonomo R, Prince AM, Chin SN, Brotman B, Shulman RW. Solvent/detergent-treated plasma: a virus-inactivated substitute for fresh frozen plasma. Blood. 1992;79(3):826-31.
  163. 163. Piet MP, Chin S, Prince AM, Brotman B, Cundell AM, Horowitz B. The use of tri(n-butyl)phosphate detergent mixtures to inactivate hepatitis viruses and human immunodeficiency virus in plasma and plasma's subsequent fractionation. Transfusion. 1990;30(7):591-8. DOI: 10.1046/j.1537-2995.1990.30790385516.x
  164. 164. El-Ekiaby M, Sayed MA, Caron C, Burnouf S, El-Sharkawy N, Goubran H, et al. Solvent-detergent filtered (S/D-F) fresh frozen plasma and cryoprecipitate minipools prepared in a newly designed integral disposable processing bag system. Transfus Med. 2010;20(1):48-61. DOI: 10.1111/j.1365-3148.2009.00963.x
  165. 165. Chou ML, Burnouf T, Chang SP, Hung TC, Lin CC, Richardson CD, et al. TnBPTriton X-45 treatment of plasma for transfusion efficiently inactivates hepatitis C virus. PLoS One. 2015;10(2):e0117800. DOI: 10.1371/journal.pone.0117800
  166. 166. Mohr H, Pohl U, Lambrecht B, Wieding JU, Schmitt H. [Methylene blue/light treatment of virus inactivated human plasma: production and clinical experience]. Infusionsther Transfusionsmed. 1993;20 Suppl 2:19-24.
  167. 167. Steinmann E, Gravemann U, Friesland M, Doerrbecker J, Muller TH, Pietschmann T, et al. Two pathogen reduction technologies--methylene blue plus light and shortwave ultraviolet light--effectively inactivate hepatitis C virus in blood products. Transfusion. 2013;53(5):1010-8. DOI: 10.1111/j.1537-2995.2012.03858.x
  168. 168. de Alarcon P, Benjamin R, Dugdale M, Kessler C, Shopnick R, Smith P, et al. Fresh frozen plasma prepared with amotosalen HCl (S-59) photochemical pathogen inactivation: transfusion of patients with congenital coagulation factor deficiencies. Transfusion. 2005;45(8):1362-72. DOI: 10.1111/j.1537-2995.2005.00216.x
  169. 169. Hornsey VS, Drummond O, Morrison A, McMillan L, MacGregor IR, Prowse CV. Pathogen reduction of fresh plasma using riboflavin and ultraviolet light: effects on plasma coagulation proteins. Transfusion. 2009;49(10):2167-72. DOI: 10.1111/j.1537-2995.2009.02272.x
  170. 170. Ruane PH, Edrich R, Gampp D, Keil SD, Leonard RL, Goodrich RP. Photochemical inactivation of selected viruses and bacteria in platelet concentrates using riboflavin and light. Transfusion. 2004;44(6):877-85. DOI: 10.1111/j.1537-2995.2004.03355.x
  171. 171. Dodd RY, Moroff G, Wagner S, Dabay MH, Dorfman E, George V, et al. Inactivation of viruses in platelet suspensions that retain their in vitro characteristics: comparison of psoralen-ultraviolet A and merocyanine 540-visible light methods. Transfusion. 1991;31(6):483-90. DOI: 10.1046/j.1537-2995.1991.31691306242.x
  172. 172. Lin L, Cook DN, Wiesehahn GP, Alfonso R, Behrman B, Cimino GD, et al. Photochemical inactivation of viruses and bacteria in platelet concentrates by use of a novel psoralen and long-wavelength ultraviolet light. Transfusion. 1997;37(4):423-35. DOI: 10.1046/j.1537-2995.1997.37497265344.x
  173. 173. Attia MA. Prevalence of hepatitis B and C in Egypt and Africa. Antivir Ther. 1998;3(Suppl 3):1-9.
  174. 174. Reker C, Islam KM. Risk factors associated with high prevalence rates of hepatitis C infection in Egypt. Int J Infect Dis. 2014. DOI: 10.1016/j.ijid.2014.02.003
  175. 175. Marschner S, Goodrich R. Pathogen reduction technology treatment of platelets, plasma and whole blood using riboflavin and UV light. Transfus Med Hemother. 2011;38(1):8-18. DOI: 10.1159/000324160
  176. 176. Allain JP, Owusu-Ofori AK, Assennato SM, Marschner S, Goodrich RP, Owusu-Ofori S. Effect of Plasmodium inactivation in whole blood on the incidence of blood transfusion-transmitted malaria in endemic regions: the African Investigation of the Mirasol System (AIMS) randomised controlled trial. Lancet. 2016;387(10029):1753-61. DOI: 10.1016/S0140-6736(16)00581-X
  177. 177. Stefanutti C, Di Giacomo S, Mareri M, De Lorenzo F, D'Alessandri G, Angelico F, et al. Immunoadsorption apheresis (Selesorb) in the treatment of chronic hepatitis C virus-related type 2 mixed cryoglobulinemia. Transfus Apher Sci. 2003;28(3):207-14. DOI: 10.1016/S1473-0502(03)00055-7
  178. 178. Schettler V, Monazahian M, Wieland E, Ramadori G, Grunewald RW, Thomssen R, et al. Reduction of hepatitis C virus load by H.E.L.P.-LDL apheresis. Eur J Clin Invest. 2001;31(2):154-5. DOI: 10.1046/j.1365-2362.2001.00758.x
  179. 179. Schettler V, Monazahian M, Wieland E, Thomssen R, Muller GA. Effect of heparin-induced extracorporeal low-density lipoprotein precipitation (HELP) apheresis on hepatitis C plasma virus load. Ther Apher. 2001;5(5):384-6. DOI: 10.1046/j.1526-0968.2001.00374.x
  180. 180. Taniguchi M, Furukawa H, Shimamura T, Suzuki T, Yamashita K, Ota M, et al. Impact of double-filtration plasmapheresis in combination with interferon and ribavirin in living donor liver transplant recipients with hepatitis C. Transplantation. 2006;81(12):1747-9. DOI: 10.1097/01.tp.0000226075.04938.43
  181. 181. Fujiwara K, Kaneko S, Kakumu S, Sata M, Hige S, Tomita E, et al. Double filtration plasmapheresis and interferon combination therapy for chronic hepatitis C patients with genotype 1 and high viral load. Hepatol Res. 2007;37(9):701-10. DOI: 10.1111/j.1872-034X.2007.00117.x
  182. 182. Ishikawa T, Higuchi K, Kubota T, Seki K, Honma T, Yoshida T, et al. Complete early virological response was highly achieved by double filtration plasmapheresis plus IFN-beta induction therapy for HCV-1b patients with relapse or no response after previous IFN therapy. Ther Apher Dial. 2011;15(4):400-5. DOI: 10.1111/j.1744-9987.2011.00965.x
  183. 183. Mednikov RV, Rabinovich VI, Kizlo SN, Belyakov NA, Sokolov AA. Double Filtration Plasmapheresis in Treatment of Patients With Co-Infection of Hepatitis C and Human Immunodeficiency Virus. Ther Apher Dial. 2016. DOI: 10.1111/1744-9987.12396
  184. 184. Sugimoto K, Kim SR, El-Shamy A, Imoto S, Fujioka H, Kim KI, et al. Outcome of double-filtration plasmapheresis plus interferon treatment in nonresponders to pegylated interferon plus ribavirin combination therapy. Dig Dis. 2013;31(5-6):434-9. DOI: 10.1159/000355241
  185. 185. Kaneko S, Sata M, Ide T, Yamashita T, Hige S, Tomita E, et al. Efficacy and safety of double filtration plasmapheresis in combination with interferon therapy for chronic hepatitis C. Hepatol Res. 2010;40(11):1072-81. DOI: 10.1111/j.1872-034X.2010.00708.x
  186. 186. Radosevich M, Burnouf T. Intravenous immunoglobulin G: trends in production methods, quality control and quality assurance. Vox Sang. 2010;98(1):12-28. DOI: 10.1111/j.1423-0410.2009.01226.x
  187. 187. Sawyer LA. Antibodies for the prevention and treatment of viral diseases. Antiviral Res. 2000;47(2):57-77. DOI: 10.1016/S0166-3542(00)00111-X
  188. 188. Congly SE, Burak KW, Coffin CS. Hepatitis B immunoglobulin for prevention of hepatitis B virus infection and recurrence after liver transplantation. Expert Rev Clin Immunol. 2011;7(4):429-36. DOI: 10.1586/eci.11.30
  189. 189. Piazza M, Sagliocca L, Tosone G, Guadagnino V, Stazi MA, Orlando R, et al. Prophylaxis of hepatitis C with intramuscular immunoglobulin: clinical and economic appraisal. BioDrugs. 1999;12(4):291-300. DOI: 10.2165/00063030-199912040-00006
  190. 190. Borgia G. Specific immunoglobulin against HCV: new perspectives. IDrugs. 2004;7(6):570-4.
  191. 191. Vanwolleghem T, Bukh J, Meuleman P, Desombere I, Meunier JC, Alter H, et al. Polyclonal immunoglobulins from a chronic hepatitis C virus patient protect human liver-chimeric mice from infection with a homologous hepatitis C virus strain. Hepatology. 2008;47(6):1846-55. DOI: 10.1002/hep.22244
  192. 192. ClinicalTrials.gov: A service of the U.S. National Institutes of Health. Randomized Phase II Study of Hepatitis C Immune Globulin Intravenous (Human), Civacir(TM), in Liver Transplantation [Internet]. 2007. updated May 7 2012. Available from: https://clinicaltrials.gov/ct2/show/study/NCT00473824 [Accessed: 2016-08-15]
  193. 193. ClinicalTrials.gov: A service of the U.S. National Institutes of Health. Civacir® Polyclonal Immune Globulin (IgG) to Prevent Hepatitis C Virus (HCV) Recurrence in Liver Transplant Patients [Internet]. 2013. updated April 20 2016. Available from: https://www.clinicaltrials.gov/ct2/show/NCT01804829 [Accessed: 2016-08-15]

Written By

Thierry Burnouf, Ching-Hsuan Liu and Liang-Tzung Lin

Reviewed: 01 September 2016 Published: 08 March 2017