Review
EUROPEAN ASSOCIATION FOR THE STUDY OF THE LIVER I
JOURNAL OF HEPATOLOGY
Genomics and HCV infection: Progression of fibrosis and
treatment response
Emilie Estrabaud1'23'4'*, Michel Vidaud5, Patrick Marcellin1'2'3'4, Tarik Asselah1'2'3'4'*
1INSERM, UMR773, Team «Viral hepatitis », Centre de Recherche Bichat Beaujon, BP 416, F-75018 Paris, France; 2Université Denis Diderot Paris 7, site Bichat, BP 416, F-750Î8 Paris, France; 3Service d'Hépatologie, PMAD Hôpital Beaujon, 100 Bd du Général Leclerc, Clichy la Garenne, 92110 Clichy Cedex, France; 4Laboratory of Excellence Labex INFLAMEX, PRES Paris Sorbonne Cité, France; 5Service de Biochimie,
Hôpital Beaujon, Clichy, France
Summary
HCV infection is a global health problem that affects 170 million people worldwide. The severity of the disease varies from asymptomatic chronic infection to cirrhosis and hepatocellular carcinoma (HCC). Recently, the standard of care for genotype 1 patients has greatly improved with the addition of protease inhibitors (telaprevir or boceprevir) to pegylated interferon (PegIFN) and ribavirin (RBV). The prediction of fibrosis progression and the response to antiviral treatment are two major issues in the management of patients with chronic hepatitis C. Differential expression of mRNAs was first analyzed for both progression of fibrosis and treatment response. Specific polymorphisms, associated with either fibrosis or viral response, were identified thanks to major improvements in genome scanning technologies. Since 2009, several independent genome wide association studies (GWAS) have reported an association between genetic polymorphisms within the IL-28B promoter and both natural and treatment-induced clearance in genotype 1 infected patients. These different studies showed the strong association and the importance of IL-28B polymorphisms in the treatment response. Combining the different genetic factors could improve their predictive value and help identify patients at a high risk of progression of fibrosis as well as those with a lower chance of responding to treatment. The aim of this review was to discuss the genomic factors (mRNAs, miRNAs, and SNPs) and HCV infection with clinical implications for either progression of fibrosis or treatment response. Recent findings on the IL-28B polymorphism and its application in clinical practice will also be discussed. © 2012 European Association for the Study of the Liver. Published by Elsevier B.V. All rights reserved.
Introduction
Chronic hepatitis C is the most common cause of cirrhosis and the first indication for liver transplantation in Europe and the United States. The progression from early stage of fibrosis to cirrhosis varies widely among patients with chronic hepatitis C.
Identifying patients in whom fibrosis will progress rapidly as well as non-responders is crucial for disease prognosis. Some defined markers such as male gender, age, alcohol, and obesity may play a role in accelerating fibrosis and treatment failure. The current standard of care for HCV genotype 1 patients is triple therapy with the addition of protease inhibitors (boceprevir and telaprevir) to pegylated interferon (PegIFN) plus ribavirin (RBV). For genotype non-1 patients, treatment is still PegIFN and RBV. The prediction of non-response to treatment is mandatory to avoid side effects and reduce costs.
Since the sequencing of the entire human genome in 2001, major advances have been made in genotyping technologies, in particular, the decrease in the cost of genotyping, the large-scale discovery of single nucleotide polymorphisms (SNPs), the development of massive multiplexed genotyping and the efforts of SNPs consortiums and the HapMap project.
Since 2009, at least 4 different GWAS have investigated genomic markers associated with a response to PegIFN/RBV in patients with chronic HCV (CHC). The same SNPs located in the promoter region of IL-28B were highly associated with natural and treatment-induced viral clearance [1].
The aim of this review was to present and discuss the major genomic markers that have been associated with either fibrosis progression or treatment response in patients with CHC. The
Keywords: Personalized medicine; Genetic; IL-28B; Direct-acting antivirals; Cirrhosis; Biomarkers. Received 13 July 2011; received in revised form 11 May 2012; accepted 14 May 2012
* Corresponding authors. Addresses: Faculté de Medecine Xavier Bichat. INSERM U773.16 rue Henri Huchard. 75018 Paris. France. Tel.: +33 (0) 157277564; fax: +33 (0) 157277531 (E. Estrabaud)' Service d'Hépatologie & INSERM U773' Hôpital Beaujon' 100 Bd du Général Leclerc' 92110 Clichy' France. Tel.: +33 (0) 660957890; fax: +33 (o) 147309440 (t. Asselah).
E-mail addresses: emilie.estrabaud@inserm.fr (E. Estrabaud)' tarik.asselah@bjn.aphp.fr (T. Asselah).
Abbreviations: CHCchronic hepatitis C; CRS' cirrhosis risk score; EVR early virological response; GWAS'genome wide association study; HCV'hepatitis C virus; HSChepatic stellate cells; IFN' Interferon; IL' Interleukin; ISG' interferon stimulated gene; NR' non-response; PegIFN' pegylated-interferon; RVR' rapid virological response; SNP' single nucleotide polymorphism; SVR sustained virological response; IP10' inducible protein 10.
Key Points
• The monitoring of fibrosis is crucial for prognosis of chronic hepatitis C (CHC) and decision to treat. Liver biopsy remains the gold standard to distinguish between moderate and mild fibrosis. Several genetic factors have been identified to help diagnose fibrosis progression
• Genome wide association studies (GWAS) have identified previously unknown markers associated with fibrosis progression and antiviral response. Some polymorphisms have been associated with viral response and fibrosis progression, due to their ability to modulate expression of genes whose products play a role in the antiviral response or fibrosis progression
• The cirrhosis risk score (CRS) including a combination of 7 SNPs may distinguish patients at high risk
of developing cirrhosis. Several SNPs regulating expression of MMP9 and MMP1 have been associated with fibrosis and its progression. IL-28B is not associated with fibrosis stage
• Fibrosis progression is different for each patient; some patients are defined as "rapid fibrosers" and others as "slow fibrosers". The progression likely depends on the intensity of aggressive stimuli (HCV, alcohol, etc.), the duration of exposition to the agent, but also the host genetics and the environment (obesity, etc.)
• Fibrosis regression is different for each patient. The regression is directly associated with the control of the aggressive agent (HCV eradication, control of alcohol consomption, etc.), but also with the host genetics (capacity of fibrosis degradation and repair), and the environment. Some patients are defined as "rapid regressors" and some as "slow regressors"
• By scanning several hundreds of thousands SNPs, 4 independent GWAS reported single nucleotide polymorphisms (SNPs), near the IL-28B (IFNA3) region, that were associated with response to PeglFN/RBV treatment in genotype 1 patients. The association of IL-28B SNPs with viral response is less strong in HCV genotypes 2 and 3 than in genotypes 1 and 4
• The molecular mechanisms of IL-28B SNPs are unknown
• Interferon stimulated genes (ISGs) are highly expressed in the livers of non-responders at baseline compared
to sustained virological responders (SVR). ISG intrahepatic expression remains a better predictive factor of antiviral response in patients with chronic hepatitis C than the IL-28B genotype. However, IL-28B is non-invasive and easier to perform
• The SNP rs12979860 CC is associated with both SVR and RVR. RVR is a strong predictor of SVR whatever the IL-28B genotype. In patients with RVR, rs12979860 CC genotype is not associated with higher SVR rates
• IL-28B polymorphisms may be used in combination with some ISGs, viral genotype and other SNPs to improve prediction of treatment response. IL-28B SNP may be less predictive in triple therapy and future combination
combination of these different markers may help identify a strong predictive genetic signature for fibrosis progression and treatment response.
Genome wide association studies, power and limitations
When the GWAS technology was first described in 1996 [2] (Box 1), its main limitations were due to the lack of technology and the authors suggested geneticists to keep their samples for future experiments. Originally, the HapMap project characterized the pattern of genetic variations in 270 individuals of different ethnicities. The dataset then amounted to a total of 3.1 million SNPs, which is a very important resource for the selection of genetic markers and genotyping assays.
GWAS involves identification of statistical associations between a trait or disease and a genetic polymorphism (SNP), throughout the entire genome.
Linkage disequilibrium can be used to indirectly determine untyped variations around an SNP. The r2 value, which is commonly used to evaluate linkage disequilibriums, measures the correlation between alleles at a nearby genetic variant. If alleles of 2 SNPs are always found on the same chromosome, there is a perfect correlation and r2 = 1. In practice, this means that typing one of these 2 SNPs will provide complete allele information at the 2 sites. If r2 <1 between the 2 alleles, the sample size will need to be proportionally increased to detect a significant association [3].
So far only SNPs with a frequency of more than 5% of the minor allele can be detected effectively by linkage disequilibrium in GWAS. Thus, the HapMAp project does not provide perfect coverage of GWAS so that even in very large GWAS it is impossible to detect rare low frequency genetic variants. However, the technology will probably improve in the future, thus increasing coverage of low frequency variants.
Different methods of correcting the p values of associations have been described to counteract associations due to chance because of the many statistical tests. The most commonly used approach consists in the adjustment of the p value according to Bonferroni. The threshold of 0.05 is divided by the number of SNPs analyzed, which leads to an adjusted threshold of 10~6 to 10~8, for current available assays. However, several real associations may be lost by deleting all associations with a p value above this threshold. Furthermore, to improve the statistical relevance of GWAS, it is extremely important to replicate significant associations in at least one independent cohort of individuals (trait and control).
The GWAS technology has greatly improved in the last decade, however, the detection of rare variants would probably improve this approach even more [3].
Fibrosis
Liver fibrosis is defined as the excess accumulation of extracellular matrix proteins. Fibrogenesis is a complex dynamic process, mediated by necro-inflammation, and activation of stellate cells [4]. Fibrosis progression determines the prognosis and thus the need for treatment. It is therefore crucial to monitor fibrosis in patients with CHC. Several studies have assessed the identification of mRNAs and miRNAs expression during fibrosis (Table 1) and will be discussed in the first part of the review.
The natural history of HCV and factors associated with fibrosis progression are discussed in Box 2.
Principles of GWAS and its clinical and biological application
Patients with the trait . or the disease
Controls
IU FT11 IhFI PI I UHFI PI I S
-3,000,000 SNPs in the genome
Domain significantly associated with the trait
Chr location 22
GWAS uses dense maps of SNPs covering the entire genome to look for allele frequency differences between cases (disease or trait) and controls.
The study of the association between the isolated marker and a disease or trait is based on the selection of genes that are already known to play a role in the trait, while GWAS is based on whole genome scanning. Thus, the main advantage of GWAS is to identify both genes that are expected to play a role and genes that are not, providing new insights into the understanding of the physiopathology of the disease. This technique has revolutionized the identification of genetic markers associated with common disease or traits. Hundreds of thousands of SNPs are tested in a single GWAS for their association with a disease or trait in hundreds or thousands of people. A significant difference in frequency suggests that the corresponding region of the genome encodes for functional DNA variants that may influence the tested disease or trait.
The results obtained with GWAS can be used in two different ways: (i) to help to understand the physiopathology of the disease or trait tested and (ii) to improve personalized medicine. GWAS can identify markers that may be used to develop new therapeutic targets and be tested as new biomarkers to evaluate the risk in patients associated with this variant.
Variation of mRNAs expression and fibrosis
A deregulation of gene expression mainly affecting the IFN ab and y pathways ( STATI, STAT2, ISGF3G/IRF9, IFI27, GIP3, GIP2, OAS2, MXI, CXCL9, CXCLI0, CXCLII, and Viperin) has been reported in patients with CHC and mild fibrosis [5,6].
The transition from mild to moderate fibrosis is crucial in the decision to treat. The expression of 240 liver genes has been compared in 62 patients with mild fibrosis (F1) and moderate fibrosis (F2). Twenty-two genes were upregulated in F2 and mainly involved the cytoskeleton, growth factor cytokines, growth factor receptors, extra-cellular matrix remodeling, and the cell junction [7].
Liver steatosis is frequent in patients with CHC [8]. Three genes involved in the inflammatory pathway (SITPEC, SIGIRR, and TOLLIP) have been described as specifically associated with advanced steatosis in CHC [9].
One study investigated changes in liver mRNAs expression in 13 transplanted patients comparing patients in whom fibrosis progressed with those in whom it did not, before and after transplantation [10]. Fifteen of the 31 upregulated genes encoded for markers of myofibroblasts and myofibroblast-like cells. Liver stress injury and fibrosis development can cause an increase in myofibroblasts due to activation of HSCs and their conversion into the contractile phenotype [10]. It is inter-
esting to note that these data suggest that early fibrosis progression may be associated with a reduction in the pools of quiescent HSCs and with an increase in the number of myofi-broblast-like cells.
Variation in micro-RNAs expression and fibrosis
Micro-RNAs regulate up to 60% of cellular mRNA expression and stability [11-14].
In one study, mir-21 expression was found to be correlated to both HCV viral load and fibrosis [15]. Results are conflicting for mir-122 but it is probably only weakly or not associated with viral load [15,16]. Two studies have reported a correlation between mir-122 expression, liver damage, and stages of fibrosis [15,16] while another did not find any association with mir-122 expression in the serum of patients with CHC [17].
A recent study demonstrated that the different members of the mir-29 family were all downregulated in HCV infected patients. Moreover, freshly isolated HSCs expressed a high rate of mir-29, which was rapidly and markedly reduced after HSC activation [18]. Mir-29 targeted various types of collagen and, interestingly, mir-29 inhibition in mice has been shown to upreg-ulate collagen expression in the liver [19]. Thus, mir-29 downreg-ulation during HSCs activation might play a role in fibrosis by inducing direct accumulation of collagen in the liver.
Liver fibrosis: modification of liver architecture
Normal liver
Hepatocytes
Liver injury
Lymphocyte
Space of Disse
Quiescent stellate cell
Stimuli
Chronic infection, alcohol, etc...
Hepatic sinusoid Normal liver:
The hepatocytes form a parenchymal cell. Quiescent stellate cells are localized between the parenchyma and the sinusoid endothelial cells. Kupffer cells are localized below the endothelial cells in the hepatic sinusoid.
Endothelial cell Activated stellate cell
Deposition of scar matrix
Fibrosis:
Kupffer cell activation
Activation of HSCs to a myofibroblast phenotype.
HSCs activated secrete high amounts of extracelullar
matrix protein.
Activation of Kupffer cells.
Kupffer cells secrete fibrogenic mediators.
Lymphocytes infiltrate the parenchyma.
The accumulation of extracellular matrix proteins and marked modifications in liver architecture define fibrosis progression. In the normal liver, hepatocytes form parenchyma. Hepatic stellate cells (HSCs), that are key cells involved in fibrogenesis, are located between parenchymal cells (hepatocytes) and sinusoidal endothelial cells of the hepatic lobule. HSCs are quiescent and store vitamin A and retinyl palmitate in lipid droplets within the cytoplasm. Under physiological conditions, HSCs regulate vitamin A homeostasis by expressing specific receptors for retinol-binding protein (RBP) at the cell surface. Activation of retinol is regulated by endocytosis of the complex retinol/RBP. HSCs are activated by different stimuli such as chronic viral infection or alcohol intake. Activated HSCs proliferate and undergo a proliferative myofibroblast (MF) phenotype. Under pathological conditions, HSCs produce large amounts of extracellular matrix protein, including collagen, proteoglycan and adhesive glycoproteins, and release vitamin A.
Kupffer cells are the resident liver macrophages in the liver. They remove material from the portal circulation. Kupffer cells direct the destruction of hepatocytes by producing harmful soluble mediators and act as antigen-presenting cells during viral infections. Kupffer cells produce a significant amount of chemoattractant molecules for cytotoxic CD8 and regulatory T cells. They are one of the main sources of transforming growth factor pi production, which induces the transformation of HSCs into myofibroblasts.
When hepatocytes undergo necrosis from different stimuli, infiltrating lymphocytes surround the parenchyma.
Single nucleotide polymorphisms and fibrosis
identification ofSNPs that modulate gene expression associated with fibrosis and fibrosis progression
Several studies have tried to indentify SNPs associated with either fibrosis progression or treatment response (Table 2).
Matrix metalloproteinases (MMPs) play an important role in fibrosis progression. MMP-1, MMP-3, and MMP-9 gene polymorphisms have been shown to influence the transcriptional activity of their respective gene promoters. Interestingly, both MMP-1 2G
homozygote and MMP-9 C allele were more frequent in HCV patients with cirrhosis than in those without cirrhosis [20].
Monocyte chemotactic protein 1 (MCP-1) is upregulated in HSCs during CHC. MCP-1 harbors a functional polymorphism located in its promoter. Interestingly, the 2A homozygote genotype in this specific polymorphism was more frequent in patients with mild fibrosis [21].
Decreased vitamin D levels and genetic variations in the vitamin D receptor (VDR) gene have been described as an important modulator of multiple diseases, including hepatic disorders [22].
Table 1. Variations in mRNAs and miRNAs expression associated with fibrosis progression. (A) Markers identified by candidate gene strategy; (B) markers identified by scanning approach.
A References
Patients (n) Targets (n) Results
Asselah et al., [7]
240 cytoskeleton (KRT19 and SCG10)
growth factors cytokines (CXCL6, IL-8, IL-2, IL-1A and CXCL10)
growth factors receptors (CCR2, CXCR3 and CXCR4) extra-cellular matrix remodelling (TIMP1, MMP7 and MMP9) cell junction (ITGA2 and CLDN4)
miRNAs
Marquez et al., [15] 20 2 mir-122, mir-21
Morita et al., [16| 185 1 mir-122
Bihrer et al., [17] 68 + 19 1 mir-122
B References Patients (n) Targets (n) Results
Smith et al., [10] 13 (0, 3, 6 and 13,026 markers of myofibroblasts (MF) and MF like cells (CARP,
12 months MAGP2, NDRG4, COL12A1, MYOM1, CHRND, FHL1,
after liver MYBPC1, CASQ2, CHRNA1, AMPD1, CACNA1S, MYH2,
transplantation) MYL2 and NEB)
retinoid related proteins (RARRES3, STRA13, RXRB, RXRA,
RDH5, RBP4, RBP5, RODH and RODH-4)
Chiappini et al., [9] 43 22,300 inflammatory pathway (SITPEC, SIGIRR and TOLLIP)
miRNAs
Bandyopadhyay et al., [18] 22 + 4 346 mir-29 family
Table 2. Identification of SNPs associated with fibrosis and the response to antivirals in patients with chronic hepatitis C.
Genes Description of polymorphisms
MMP1 and 9 MMP1 2 G homozygote at position -1607 and MMP9 C allele at position -1562 are more frequent in cir-rhotic patients
CCL2 (MCP1) AA homozygote at position -2518 is more frequent in mild fibrosis
Fibrosis Cirrhosis risk score 7 SNPs (AZIN1: rs62522600; TLR4: rs4986791; TRPM5: rs886277; AP3S2: rs2290351; B008027: rs4290029; STXBP5: rs17740066; AQP2: rs2878771) have been all associated with fibrosis. AZIN1 SNP generates an alternative spliced from of AZIN1 that modifies the fibrogenic potential of HSCs TLR4 SNP have an identified role in fibrosis
PNPLA3 The rs738409 mutant GG is associated with higher risk of steatosis, fibrosis and fibrosis progression
Vitamin D receptor (VDR) Haplotype rs1544410 C, rs7975232 A and rs731236 A with fibrosis progression and cirrhosis
KIR/HLA The combination of KIR2DL3 and HLAC1 is more frequent in patients with high SVR rates
e s IL6 The SNP rs1800795 CC is associated with low IL6 production and high SVR rates
n o p s e IL10 Mutations at positions -819, -1082 and -592 are associated with modulation of IL-10 expression and different viral SVR rates
ral IFNG The mutation C764G is associated with higher SVR rates
> CCR5 CCR5A32 deletion is associated with resistance to HIV-1 and with poor SVR rates in HCV patients
IL28B rs1299860 CC genotype and rs8009917 TT are both associated with high SVR rates
VDR genotyping in 251 patients with chronic hepatitis C showed an association between the haplotype rs1544410 C, rs7975232 A, and rs731236 A with fibrosis progression and cirrhosis. Forty-five percent of the [CCA]-haplotype patients had rapid fibrosis progression and 21.1% had cirrhosis [23].
Cirrhosis risk score
A gene-centric disease association study of 24, 832 putative functional SNPs was performed to assess the association of SNPs with cirrhosis in a cohort of 433 patients with CHC [24]. One SNP located in the DEAD box polypeptide 5 was associated with an
increased risk of advanced fibrosis while the second SNP, located in the gene encoding carnitine palmitoyltransferase 1A (CPTAI), was associated with a decreased risk of advanced fibrosis [24].
In a second study, the authors confirmed all significant SNPs, and selected 361 markers to build a signature predicting cirrhosis, called the cirrhosis risk score (CRS) [25]. Interestingly, DDX5 and CPTIA were not selected for the final 7 SNPs for the CRS. Possible reasons were (i) lower odds ratios and frequencies in the risk group and (ii) decreased robustness and accuracy of these 2 SNPs in multivariate analysis compared to the 7 selected genes. Of the 7 CRS genes, antizyme-inhibitor-1 (AZINI) and Toll-like receptor 4 (TLR4) have been shown to play a role in fibrosis. A recent study has reported that the association of AZINI SNP with the rapid progression of fibrosis, leads to enhanced generation of a novel alternative splice form from AZIN1 that modifies the fibr-ogenic potential of HSCs [26]. All 7 SNPs were associated with the risk of cirrhosis with odds ratios ranging from 1.86 to 3.23. However, the AUC of each SNP was <0.6, showing that predictability was moderate when they were used individually [25]. The addition of clinical factors to the group of SNPs or each of them individually did not significantly improve the AUC.
Two major limitations are associated with the use of CRS to identify patients with rapid progression of fibrosis: (i) CRS cutoff values may only distinguish patients with a very high risk of cirrhosis (ii) CRS was identified in Caucasian patients so it may not be applicable to all ethnic groups [25].
In another study, paired liver biopsies from 271 untreated patients with CHC (F0 = 104, F1 =101, and F2 = 59) were fol-lowed-up for at least 60 months. Mean CRS was significantly higher is patients in whom fibrosis progressed, especially in patients with F0 at the initial biopsy [27].
CRS remained the only variable associated with fibrosis progression in multivariate analysis, including gender and alcohol intake [28].
Genetic studies have reported an association between advanced steatosis and specific SNPs located in genes encoding microsomal triglyceride transfer protein (MTP G493T) [29-31], peroxisome proliferator activated alpha (PPAR L162V) [32], methylenetetrahydrofolate reductase (MTHFR C677T) [33,34], cytokines playing a role in the inflammatory response such as interlekin-10 and -6 (IL-10 and IL-6) [35], transforming growth factor beta-1 (TGFB1) [35], tumor necrosis factor (TNF, -238 position) [36,37] and leptin receptor (LEPR) [35].
PNPLA3 and fibrosis
A non-synonymous sequence variation (rs738409 C/G) encoding an isoleucine to methionine substitution in the adiponutrin/pat-atin-like phospholipase-3 (PNPLA3) has been shown to be strongly associated with increased hepatic fat levels [38]. This SNP was then shown to be associated with disease severity, fibro-sis, and steatosis in non-alcoholic fatty liver disease (NAFLD) [39,40] and in alcoholic liver disease (ALD). Moreover, the same SNP was also associated with elevated liver enzymes in healthy subjects [41]. Interestingly, patients with CHC carrying the rs738409 mutant GG allele had a high risk of steatosis as well as fibrosis and fibrosis progression [42-44]. However, there are conflicting results reporting that PNPLA3 rs738409 GG mutant variant may be a prominent risk factor for HCC in patients with alcoholic cirrhosis, while its effects were negligible in patients with HCV cirrhosis [45].
IL-28B and fibrosis
The analysis of rs8099917 SNPs during liver fibrosis in chronic hepatitis C showed that the G allele, previously shown to be a risk of treatment failure, was associated with lower activity and less fibrosis with a trend towards a lower rate of fibrosis progression. Interestingly, when patients were stratified according to HCV genotype, the association was more significant in HCV genotype non-1 infected patients [46]. However, independent studies have reported that fibrosis progression was not associated with either rs12979860 or with rs8099917 [47,48].
The analysis of a larger cohort showed that PNPLA3 polymorphisms were strongly associated with an increased risk of steatosis in patients with HCV (excluding genotype 3) while the association with IL-28B SNPs was weak [49]. Steatosis was found in 22.5% and 39.6% of two independent cohorts of patients carrying the favorable IL-28B genotype vs. 47.6% and 67.4% of IL-28B non-favorable genotypes, respectively [50].
It has been suggested that levels of LDL cholesterol are significantly higher in CHC subjects carrying the rs12979860 CC genotype compared to CT and TT [51]. However, triglyceride levels were lower in patients carrying the IL-28B CC genotype. IL-28B CC may be associated with less pronounced lipid metabolism disturbances, as shown by serum lipoprotein levels and hepatic steatosis.
Altogether, the different studies have suggested that various SNPs, especially, AZINI and PNLPA3 are associated with fibrosis progression. Moreover, several extra-cellular-matrix and chemo-kine mRNAs are probably upregulated in patients with more advanced fibrosis. However, it is important to bear in mind that some studies have investigated fibrosis progression by analyzing paired biopsies while other studies were cross-sectional. Taking into account the duration of infection could provide useful information to improve the evaluation of fibrosis progression in cross-sectional liver biopsies.
Prediction of treatment response
The current standard of care for chronic hepatitis C and the factors associated with a non-response are shown in Box 3.
Variation in mRNAs expression and treatment response
An 8-gene subset accurately predicted treatment response (GIP2/ IFI15/ISG15, ATF5, IFIT1, MX1, USP18/UBP43, DUSP1, CEB1, and RPS28) using a number of independent classifier analyses [52]. Interestingly, another study evaluated the expression profile of a selection of genes in SVRs and NRs. IFI27 and CXLC9, a two-gene signature, predicted the response to treatment in 79% of the patients with a predictive accuracy of 100% for NRs and 70% for SVRs [53]. One study showed that SVR could be predicted, prior to treatment, by analyzing gene expression of signal transducer and activator of transcription-6 (STAT-6) and suppressor of cytokine signaling-1. Interestingly, even after 24 h of treatment, IFN-dependent gene expression can help predict the probability of achieving an SVR [54]. Many of the genes, found to be upregu-lated in non-responders and responders, encode molecules secreted in the serum (cytokines) [55,56]. Thus, they are a logical approach to the development of serum markers to predict treatment response.
Current treatment in chronic hepatitis C and predictive factors of non-response
Host factors
Viral factors
Treatment
Advanced age
Male gender
Ethnicity
Compliance
Insulin-resistance
Alcohol
Cirrhosis
Genetic
Viral load Genotype Quasispecies
Regimen
Dose and duration Side effects
Treatment for genotype non-1 patients, includes the combination of PeglFN/RBV [140-142]. In patients with genotype 1, the new standard of care includes the addition of a protease inhibitor (boceprevir or telaprevir) to PeglFN/ RBV. The goal of treatment is to obtain a sustained virological response (SVR) defined as undetectable HCV RNA in serum after 24 weeks of post-treatment follow-up. SVR results in the eradication of HCV infection and improvement of the histological outcome [143].
Brief description of factors associated with SVR
Responsiveness to antiviral therapy depends upon viral factors as well as host factors. Viral factors associated with response to treatment are genotype and viral load [144].
Patients who develop a rapid virological response, defined as HCV RNA negative at treatment week 4 have a greater chance to achieve SVR (higher than 85%). Patients who do not have any decrease in viral load will not respond to treatment.
Advanced age, male gender, African American ethnicity, poor adherence, cirrhosis, insulin resistance (and also steatosis, diabetes and alcohol consumption) are all events associated with a poor response to PeglFN/RBV treatment [8, 145]. The chance of achieving an SVR significantly decreases in patients with immune depression, transplantation, or HIV co-infection [146].
Variation of micro-RNAs expression and treatment response
Since cellular miRNAs regulate many cellular pathways, including the immune response, some miRNAs are probably involved in the modulation of antiviral response to PeglFN/RBV in CHC. IFNb has been shown to rapidly modulate the expression of numerous miRNAs, and 8 cellular IFNb-induced miRNAs have sequence-predicted targets within the HCV genome [57].
The level of expression of intra-hepatic mir-122 has been shown to be associated with HCV treatment response. Expression of mir-122 was significantly lower in primary non-responders compared to early responders [58]. Interestingly, the anti mir-122 (SPC3649 or miravirsen) molecules that inhibit HCV replication in chimpanzees [59] have recently been tested in a randomized double-blind study in treatment naive patients with genotype 1 [60].
However, an independent study investigating the expression profile of 470 cellular miRNAs in 99 CHCs, showed no significant
difference in mir-122 expression between responders and non-responders [61]. Eight miRNAs (mir-34b, mir-145, mir-143, mir-652, mir-18a, mir-27b, mir-422b, and mir-378) were differentially expressed in responders and non-responders. Moreover, mir-34b and mir-422 were consistently and significantly high and low in non-responders both 12 and 24 weeks after the end of the treatment [61].
Single nucleotide polymorphisms and treatment response
The main SNPs associated with SVR are summarized in Table 2.
Killer cell immunoglobulin-like receptors (KIR) activating and inhibiting receptors
Natural killer (NKs) cells are a subset of lymphocytes that interact directly with virus-infected cells, activate dentritic cells and secrete Th1-type cytokines to increase antiviral cytotoxic T cell response. NK responses are controlled by multiple activating and inhibitory signals such as the KIR receptors. The ligands for these receptors are human leukocyte antigen (HLA) class I. The wide genetic diversity of the KIR family and the HLA class I generate a large number of combinations between KIR and HLA.
The combination of KIR2DL3 and HLA-C1 was associated with both spontaneous clearance [62] and SVR [63,64] in patients with CHC. This specific combination is supposed to have a weaker inhibitory effect than other combinations leading to a stronger NK cell response.
KIRs are clonally expressed on NKs in a stochastic manner. Interestingly, Khakoo et al. reported a linear trend between the number of KIR2DL3-HLA-C1 interactions and the ORs resolving the infection. In this model, NK cell activity may be mediated through weak inhibitory KIR2DL3-HLA-C1 interactions.
HLA class II alleles have been shown to play a role in spontaneous clearance in patients with HCV infection [65]. Interestingly, a new interaction between KIR2DL3 and HLA class II DRB1*1201 was found to be associated with spontaneous clearance [66].
Prediction of treatment failure improved from 66% with IL-28B to 80% using both IL-28B and HLA-C2/HLA-C2 genes [67]. Moreover, the incorporation of two KIRs in a model including viral load and ethnicity (SVR score) provided complementary results with IL-28B across the CC, CT, and TT genotypes [68].
Genetic variants in the cytokine system and treatment response STAT3 is mainly activated in the liver by IL-6, a cytokine that has been involved in a variety of cell functions including stimulation of hepatocytes to produce acute-phase proteins. The polymorphism rs1800795 within the IL-6 promoter is associated with lower IL-6 levels compared to patients carrying the G allele. Interestingly, higher response rates were reported in patients carrying the rs1800795 G allele compared to C [69]. Yee et al. confirmed the importance of rs1800795 in the viral response and reported that several haplotypes containing rs1800797, rs1800796, rs1800795, and rs2069830 within IL-6, were associated with lower SVR rates [70]. IL-10 is a T-helper type 2 cytokine that plays a major role in T and B cell regulation. Peripheral blood mononuclear cells (PBMCs) from patients with CHC had increased IL-10 mRNA and protein expression. Moreover IL-10 has been shown to inhibit the production of IFNa by stimulation with viral infections [71]. SNPs within
the 11-10 promoter at positions -819 and -592 have been associated with different IL-10 levels. Interestingly, a strong relationship was found between ¡L-10 polymorphisms and response to IFNa treatment [72]. The frequency of -1082 GG genotypes (high IL-10) was higher in patients who did not eliminate the virus compared to controls. The higher level of IL-10 in these patients was associated with a higher risk of ineffective viral clearance and development of chronic infection in female patients [73]. Of all the 8 SNPs identified in the entire ¡FNy gene, the variant C764G was significantly associated with SVR. Functional analyses showed that the G allele conferred higher promoter activity and a stronger binding affinity for HSF1 [74].
¡L-28B and treatment response
Since 2009, several independent GWAS have reported a strong association between SNPs, located in the ¡L-28B promoter region with both natural and treatment-induced viral clearance (summarized in Box 4).
Almost 85% of the patients with East Asian ancestry harbor the C allele while only 40% of the patients with African-American ancestry carry genotype CC (Fig. 1) [75]. Thus, rs12979860 genotype diversity could explain the higher SVR rate in Asian patients.
Interestingly, the rs12979860 CC genotype was also associated with early improved viral kinetics, a higher rate of rapid virological response, and complete early virological response [76]. The association of rs12979860 with viral response has been confirmed in independent cohort studies [77,78]. Interestingly, ¡L-28B genotype was associated with treatment response in patients with CHC but not in those with acute hepatitis C [79]. Moreover, jaundice was more common in CC patients (64%) during acute infection than in CT (24%) or TT (6%). However, jaundice was only associated with an increased chance of spontaneous viral clearance in non-CC patients [80].
It has been suggested that the influence of rs12979860 genotype is a stronger predictive factor of SVR in patients with HCV-G1 than in HCV non-G1 patients [81].
HCV-G-2 and G-3 patients carrying the rs12979860 CC allele had a more rapid reduction in plasma HCV RNA, 3 days after beginning of treatment. However, in accordance with previous results [82], no significant association was found between rs12979860 and SVR in this cohort (98 genotype 2 and 241 genotype 3 patients). Interestingly, the assessment of ¡L-28B SNPs in patients with genotype 2 or 3 might help determine the suitable treatment duration [83].
In a recent study including 164 HCV genotype 4 patients from different ethnic groups, ¡L-28B rs12979860 CC was associated with a better treatment response rate (81.8% vs. 46.5% and 29.4% for CC, CT and TT respectively). No significant relationship was found between rs12979860 and the stage of fibrosis [47].
Interestingly, it has been suggested that ¡L-28B SNP influences HCV re-infection in patients receiving liver transplants. Rs12979860 CC donors were associated with rapid, complete early, and sustained virological response to PegIFN/RBV, compared to CT and TT [84]. While the rs8099917 favorable genotype was associated with a higher SVR rate in both recipients and donors, recipient rs12979860 status will probably have a weaker impact on viral response [84]. The use of both ¡L-28B genotype in
CO £=
¡2 Africa Europe Asia America
n = 428 n = 761 n = 797 n = 358
Fig. 1. Frequency of IL-28B rs12979860 protective allele in worldwide populations. rs12979860 CC is present in up to 80% of patients with Asian ancestry and only 40% of patients with African ancestry. Figure adapted from data reported in Thomas et al. [77].
donors and recipients and HCV core substitution predicted an SVR with 83% sensitivity and 82% specificity [85].
Which specific IL-28B SNPs for the prediction of SVR? Rs8099917 was identified in part by Rauch et al. and Tanaka because rs12979860 was not investigated, since this SNP is not present on either the Illumina Array or the Affymetrix 6.0 genotyping platform. Prediction of SVR with rs12979860 or rs8099917 will probably not matter in Caucasian patients. However, the frequency of these SNPs in other ethnicities may limit its use as predictive factor in the general population. For example, the use of IL-28B in the decision to treat patients with African ancestry, in which the frequency of the rs 12979860 favorable allele is only 40%, would exclude a large number of patients from being eligible for treatment. Therefore, IL-28B may be useful in combination with other predictive factors to avoid excluding patients based on ethnicity.
CCR5 and HCV spontaneous viral clearance
The chemokine receptor CCR5 is activated through the binding of RANTES. CCR5 is also a co-receptor of HIV-1. Interestingly, a common 32-base deletion in the CCR5 gene has been associated with resistance to HIV-1 infection [86-88]. The potential role of CCR5 deletion in resistance to HCV has been studied and suggested to adversely affect outcome of HCV infection [89-94]. However, the different reports are conflicting.
A recent study has investigated the association of both CCR5D32 and rs12979860 with spontaneous HCV clearance
[95]. The authors described an association of response rates with rs12979860 CC only in CCR5 WT homozygous while HCV clearance remained poor in CCR5D32 carriers even in CC patients. Because the frequency of the CCR5D32 homozygous deletion is rare, it is difficult to study the influence of this mutation, either alone or in combination with IL-28B genotype, on the outcome of HCV infection. RANTES is expressed and secreted by HSCs and induces migration, proliferation, and fibrogenic properties
[96]. Different studies have reported the overexpression of CCR5/RANTES in models of liver fibrosis and in patients with CHC [97]. Pharmaceutical companies have developed RANTES inhibitors/CCR5 antagonists which have been successfully tested in phase III studies in patients with HIV infection [98]. Interestingly, the administration of the RANTES inhibitor greatly improved liver fibrosis in mice and accelerated fibrosis regression [99].
IL-28B identification by GWAS
GWAS IL-28B SNP identified Wild type/risk allele Total population % of SVR Population studied % risk allele (within each ethnicity) Viral genotype
Ge et al. rs12979860 C/T 1137 C/C: 79 C/T: 38 T/T: 26 American Caucasian African American Hispanic 30 60 40 1
Tanaka et al. rs8099917 rs12980275 T/G A/G 314 T/T: 64 T/G: 13 G/G: 0 Japanese 15 12 1
Suppiah et al. rs8099917 T/G 848 T/T: 56 T/G: 36 G/G: 31 Australian Caucasian 27 1
Rauch et al. rs8099917 T/G 914 T/T: 76 T/G: 22 G/G: 1 Swiss 17 1, 2, 3 and 4
Since 2009, several independent GWAS have identified the same SNPs located in the promoter region of IL-28B that are associated with HCV clearance [75, 82, 124, 125, 147].
A GWAS reported, for the first time, an association of response to PeglFN/RBV with IL-28B polymorphisms in patients with CHC.1137 patients were included in the cohort. In the different SNPs scanned, rs12979860 was strongly associated with treatment-induced viral clearance in both European-American, African-American and Hispanics. In patients with European-American ancestry, the SVR rate reached almost 80% in patients with genotype CC. However, in African-American patients only 50-55% of patients with the C allele achieve an SVR [75].
Two SNPs (rs12980275 and rs8099917) were found to be significantly associated with viral response to PeglFN/RBV, in an independent GWAS including 154 HCV genotype 1 Japanese patients. It is interesting to note that individuals who are homozygous for the rs8099917 minor allele GG (also called risk allele) were only found in NR [125].
rs8099917 SNP was strongly associated with treatment response in 2 groups of patients. The rs8099917 G allele could predict the response to treatment with 57% sensitivity and 67% sensitivity [124]. The authors showed that IL-28B had a distinct haplotype block and there was no significant association in SNPs outside this block. The haplotype analysis identified a six-allele haplotype (GCCTAG: rs12980275, rs8105790, rs8103142, rs10853727, rs8109886 and rs8099917) associated with viral response. Indeed, this haplotype was present in 31.5% of non-responders compared to 18.8% of responders [124].
In another GWAS, the rs8099917 TT genotype was associated with progression to chronic HCV infection in both mono-infected patients and HIV co-infected patients [82].
Combination of the IL-28B genotype with other factors to predict treatment response
Host factors
High ISGs expression before treatment has been associated with a low SVR rate [52,100]. Since treatment is based on PeglFN intake, patients who already have high ISG expression may not be as stimulated as patients with low ISGs expression. Analysis of post-treatment biopsies in patients with an RVR revealed that PeglFN did not induce ISGs expression above pretreatment levels [100].
Both IFNa and k induce ISGs expression in HCV infected cells, in vitro [101]. However, the kinetics of IFNk-mediated STAT activation and induction of effector genes were different from those of IFNa, suggesting distinct mechanisms of IFNk- and IFNa-induced antiviral states.
Intra-hepatic ISGs expression may be a better predictor of SVR than IL-28B genotype [102,103]. Interestingly, global expression of ISGs is strongly associated with genetic variation of IL-28B [103]. Multivariate analysis of predictors of response showed that a set of 4 ISGs were better predictors than rs12979860 SNP [102]. Despite a small sample size, another study reported an association between rs12979860 SNP and 3 ISGs (ISG!5, IFI27, and IFI6) [104]. Moreover, it has been suggested that IL-28B genotype is associated with a specific cell-type modulation of ISGs expres-
sion ( MxA, PKR, OAS!, and ISG15) in hepatic cells and PBMCs [105].
Several studies have shown that elevated IP10 levels may be a prognostic marker of HCV treatment outcome in HCV genotype 1 infection [55,106-110]. The combination of serum IP-10 and rs12979860 significantly improved the prediction of SVR [111113]. Serum IP-10 was particularly informative in rs12979860 CT carriers, in whom high serum IP-10 levels resulted in a 64% SVR compared to 24% in patients with low IP-10 [112]. Serum IP-10 levels below 150pg/ml significantly predicted a greater reduction of HCV RNA and increased SVR rates in genotype 1 rs12979860 CC patients [111].
It has been suggested that mir-122 expression is associated with treatment outcome [58]. IL-28B genotype and mir-122 hepatic expression have been reported to be independently associated with viral response [102].
Vitamin D deficiency is common in patients with chronic liver disease. A recent study showed that vitamin D serum levels improve prediction of an SVR in association with the IL-28B rs12979860 polymorphism [114]. Vitamin D maybe a key regulator of the innate immune response [115]. However, this study was retrospective. Furthermore, it has not been shown if correcting a vitamin D deficiency before starting treatment may increase SVR rates.
Viral factors and treatment response
Two amino acids substitutions at positions 70 and 91 within the core protein and an accumulation of mutations in the interferon sensitivity determining region (ISDR) located in the NS5A coding region have been associated with viral response [116,117].
Interestingly, two studies [118,119] have shown that ¡L-28B polymorphisms as well as amino acid 70 substitution in the core protein were independently associated with SVR. The SVR rate in patients receiving telaprevir in combination with PegIFN/RBV, was high in those carrying the rs8099917 TT genotype whatever the core 70 substitutions [118].
Of all the host and viral factors achieving an RVR is probably the strongest predictor of SVR [120]. Early viral kinetics were improved and there was a greater probability of achieving an RVR with the rs12979860 CC genotype compared to CT and TT [76,121]. RVR was a strong predictor of SVR whatever the ¡L-28B, suggesting that achieving an RVR may compensate for the negative influence of ¡L-28B genotype. The CC ¡L-28B genotype was associated with high SVR rates in non-RVR patients [76]. However, in patients with an RVR the rate of SVR was high and there were no associations between ¡L-28B genotype and SVR [121]. It is not clear whether the C allele is associated with a higher SVR rate in HCV genotype 2 or 3 patients achieving an RVR [122,123].
¡L-28B polymorphisms may be used in combination with several factors; ISGs, viral genotype and other SNPs before, but also during treatment, to improve prediction of treatment response.
The activity oflFNk and ¡L-28B polymorphisms
The activities of IFNks are summarized in Box 5.
Although multiple SNPs located in the ¡L-28B promoter region have been identified and associated with SVR, none of them were causal variants. The results of studies assessing variations of circulating IFNks in the different ¡L-28B SNPs are contradictory [104,119,124-126].
The putative causal SNP rs8103142 (Lys70Arg) within the ¡L-28B coding region was identified in strong linkage disequilibrium with rs12979860. However, there was no significant difference from the Lys70Arg mutation on either ¡L-28B activity or expression [104].
Detailed genotyping was performed on the ¡L-28B coding region in a cohort of 389 HIV/HCV co-infected patients. Homozygotes for the haplotypes rs4803219, rs28416813, rs8103142, and rs4803217 were strongly linked to the rs12979860 CC genotype (r2 = 0.97). This specific haplotype was especially frequent in individuals with spontaneous clearance compared to those with CHC [127], suggesting a possible effect of these SNPs on viral clearance.
Interestingly, IFNks were highly activated in chimpanzees immediately after HCV infection while IFNaßs were slightly induced [128].
However, it is important to remember that while a common variant associated with a disease in a GWAS is generally linked to a causal effect that is nearby, this is not necessary the case. Thus rs12979860 may be in strong linkage disequilibrium with an unidentified causal SNP located in another gene.
¡L-28B and future therapies ¡FNk-based treatment
The strong association between ¡L-28B genotype and treatment response suggests that IFNk may be an attractive alternative to
IL-28B-CC 90% 71%
IL-28B-nonCC
80% 68%
PR T12PR Telaprevir
PR BOC/PR48 Boceprevir
Fig. 2. SVR in triple therapy according to IL-28B rs12979860. Telaprevir-based therapy improved both RVR and SVR across the different rs12979860 genotypes. In patients with the unfavorable allele, the addition of telaprevir doubled SVR rates. From 87% to 90% of the CC patients achieved an SVR with telaprevir compared to 64% with PegIFN/RBV [135]. Therefore, patients with all different rs12979860 genotypes benefit from telaprevir triple therapy. Among patients carrying the rs12979860 CC allele, 89% of treatment-naïve, and 82% of patients with a treatment-failure had an early response (HCV RNA undetectable at week 8) with boceprevir in addition to PegIFN/RBV. It is interesting to note that these early responder patients were eligible for shorter treatment [139]. Naïve genotype 1 patients with the favorable rs12979860 CC allele do not benefit (SVR) from boceprevir triple therapy compared to PegIFN/RBV.
IFNa. Indeed, IFNk has a limited effect on hematopoietic cells and the central nervous system, promising antiviral activity in the liver and limited receptor expression.
Zymogenetics and Bristol-Myers Squibb have developed PegIFN^ and are assessing its efficacy in the treatment of HCV. A phase 1b study was published in 2010 [129].
The phase IIb study reported a higher early virological response (EVR) following treatment with PegIFNk combined with RBV than with PegIFNa, in patients with genotype 1 and 4. Interestingly, at different weekly doses of PegIFNk (240 to 120 ig), cEVR rates were 55-56.3% compared to 37.9% with 180 ig of PegIFNa in genotype 1 and 4 patients. The tolerability and safety of PegIFNk were better than with PegIFNa, with less anemia and fewer flu-like symptoms [130]. These findings show that PegIFNk has an antiviral effect against HCV. However, further and longer studies on the effect of PegIFNk are needed. Interestingly, rs12979860 CC was associated with an increase in SVR in patients treated with PegIFN (Fig. 2).
IL-28B and triple therapies
In naïve and previously treated genotype 1 patients, the present standard of care is triple therapy with the protease inhibitor boceprevir or telaprevir combined with PegIFN and RBV [131-134]. IL-28B status and response to therapy have been evaluated for telaprevir and boceprevir used in combination with PegIFNa/RBV. In studies of boceprevir triple therapy in both naïve and experienced genotype 1 patients, IL-28B CC genotype was associated with a higher SVR rate [131,134]. Moreover, the favorable genotype was significantly associated with undetectable HCV RNA after 8 weeks of therapy in both naïve (89% for CC vs. 52% for CT and TT) and experienced patients (89-82% for the CC genotype vs. 52% and 51% for CT and TT) [131,134]. Therefore, IL-28B may be used to identify patients who are eligible for shorter therapy. In naïve genotype 1 patients treated with boceprevir, the IL-28B polymorphism was an independent predictor of SVR. SVR rates were higher in CC, CT, and TT patients receiving triple therapy. The differences in SVR rates in patients receiving either PegIFN/ RBV or triple therapy were more marked in CT and TT patients than in CC patients (40% and 30% in CT and TT vs. 3% in CC).
IFNs lambda: signaling and activity
IFN type III or IFNAare genetically distinct from type I IFNs and act through a specific receptor system [148, 149]. However, type I and type III IFN induce the same signal transduction and share identical biological activities. IFNsA belongs to a cytokine type 2 family including IL-10, IL-22 and IL-26. There are three IFNsA encoded by three genes clustered on human Chr19, IFNA1 (IL-29), IFNA2 (.IL-28A) and IFNA3 (IL-28B).
IFNsA expression is induced by LPS or double stranded RNA TLR stimulation [150]. Plasmacytoid dendritic cells are the main producers of IFNsA. Types I and III IFNs are co-produced in response to the same inducers suggesting a common mechanism of regulation. IFNsA binds to a receptor at the cell surface containing two subunits: IFNLR1 specific for IFNsA and IL-10R2 shared among the cytokine 2 family members. While IL-10R2 is ubiquitously expressed, IFNLR1 expression is limited to certain type of cells. A high level of expression of IFNLR1 has been reported in hepatocytes from liver biopsies [151, 152].
IFNsA stimulation results in the activation of IFN-stimulated gene expression (ISGs). IFNsA and type I IFNs binding to their receptors activates the protein kinases Jak1 and Tyk2, leading to activation of downstream STATs. Activated STAT1 and STAT2 heterodimerize with IRF9 and form the ISGF3 complex. The activated complex translocates into the nucleus to activate ISGs transcription by recognition of their IFN-stimulated response element (IRSE) within the promoter region [153, 154]. Moreover, all class II cytokines including IFNsA are able to activate STAT1 ad STAT3. The heterodimer STAT1/STAT3 then translocates into the nucleus and binds IFNA-activated sites (GAS) again resulting in ISGs transcription [155]. Hundreds of ISGs are produced in response to IFNs stimulation. Evidence has shown that even if different IFNs signal through the same pathway each IFN can induce the activation of different ISGs [156, 157]. In the HCV replicon system, Marcello et al. reported that while IFNA induced a steady increase in ISGs levels, type I IFNs peaked early and declined rapidly [101]. These results supported a distinct antiviral state of type I and III IFNs.
The effect of type III IFNs on immune cell function is not fully understood. It has been reported that IFNA decreases the production of Th2-type cytokines, potentially activating a Th1 immune pathway [158-161].
In vitro, the antiviral activity of IFNsA has been described for several viruses including HCV and HBV [101, 153, 162]. Recently, Zhang et al. showed that IL-28B signal through the Jak/STAT pathway in HCV replicating cells [163].
Telaprevir-based therapy improved both RVR and SVR across all different rs12979860 genotypes (Fig. 2) [135]. Although only Caucasians were genotyped for IL-28B SNP, there was an increase
in SVR rates across all IL-28B genotypes in genotype 1 naïve patients treated with telaprevir triple therapy. Patients with the CC genotype were most likely to achieve an RVR and to have
shorter treatment duration. The difference in SVR rates between patients receiving triple therapy and PegIFN/RBV was more marked in patients with the T allele than in those with the CC genotype [135]. There was no significant difference in SVR rates across the different IL-28B genotypes with telaprevir triple therapy in genotype 1 experienced patients [136].
Multivariate analysis identified rs8099917 genotype TT and substitution at aa 70 (Arg70) as significant determinants of SVR after a 12-week or 24-week regimen of triple therapy in patients with HCV genotype 1 [118]. The efficacy of triple therapy was high in patients with the favorable genotype with an SVR rate of 84%, whatever substitution of core aa 70. However, in patients with the unfavorable genotype, the presence of Arg or Gly at position 70 in the core region was associated with a 50% and 12% SVR respectively [118].
The INFORM-1 study reported that a dual combination of the nucleoside polymerase inhibitor mericitabine and the HCV protease inhibitor danoprevir produced a rapid and substantial decrease in viral load that was maintained throughout two weeks of treatment [137]. Finally, the mean reduction in HCV RNA serum levels with the IFN-free regimen was slightly higher in patients with CC polymorphism than in those with CT and TT [138].
Overall, the ability to predict the SVR in patients receiving triple therapy has not been fully evaluated. IL-28B may have limited predictive value in patients with prior failure to PegIFN/RBV. Moreover, in patients with the favorable IL-28B genotype, it may be difficult to not use triple therapy considering the overall increase SVR rates, reported in the different trials, and the possibility of a shorten treatment duration.
For genotype 4, the current treatment remains the combination of PegIFN/RBV, thus the IL-28B polymorphism may be an important factor associated with response. Further studies are needed to determine whether patients with genotype 4 and good predictors of response, including IL-28B CC, can benefit from shorter therapy.
Conclusions
Prediction of both progression of fibrosis and SVR are main issues in chronic hepatitis C. Major advances in genetics during the last decade, allow the identification of specific markers associated with either fibrosis progression or viral response. In particular, SNPs located within genes encoding MMP-I and MMP-9, MCPI, PNLPA3, and vitamin D receptor have been associated with rapid progression of fibrosis. Moreover, CRS comprising 7 different SNPs was able to discriminate patients with high risk of developing a rapid fibrosis progression from those who will remain at low stages of fibrosis. Since the description of IL-28B SNPs association with both natural and treatment-induced HCV clearance, many studies have investigated the use of these factors for the prediction of viral response. IL-28B has been strongly associated with viral response in all reports, including large cohorts of patients.
However, for the single patient, the use of IL-28B as a predictive factor of treatment outcome and decision to treat is not sufficient. IL-28B should probably be combined with other markers to increase baseline prediction of treatment outcome.
During treatment, RVR is the strongest predictor of SVR even compared to IL28 SNPs. For individual patients, RVR may represent global predictors (host and viral) for response. Interestingly, the rs12979860 CC genotype was highly associated with viral
response in patients who did not achieve an RVR [121]. Recent studies have suggested that in triple therapy IL-28B genotyping identifies patients who might be eligible for shorter therapy [135,139]. In genotype 1 experienced patients, previous response (relapse, partial response, non-response) appears to be a stronger predictor than IL-28B status. If strong predictive factors of treatment response are identified for triple therapy for HCV-G1, the duration of treatment may be shortened.
Conflict of interest
The Authors declared that they do not have anything to disclose regarding funding or conflict of interest with respect to this manuscript.
Financial support
EE is supported by a fellowship from the French National Agency for Research on AIDS and Viral Hepatitis (ANRS).
References
[1] Afdhal NH, McHutchison JG, Zeuzem S, Mangia A, Pawlotsky JM, Murray JS, et al. Hepatitis C pharmacogenetics: state of the art in 2010. Hepatology 2011;1:336-345.
[2] Risch N, Merikangas K. The future of genetic studies of complex human diseases. Science 1996;5281:1516-1517.
[3] Karlsen TH, Melum E, Franke A. The utility of genome-wide association studies in hepatology. Hepatology 2010;5:1833-1842.
[4] Marcellin P, Asselah T, Boyer N. Fibrosis and disease progression in hepatitis C. Hepatology 2002;5 (Suppl. 1):S47-S56.
[5] Bieche I, Asselah T, Laurendeau I, Vidaud D, Degot C, Paradis V, et al. Molecular profiling of early stage liver fibrosis in patients with chronic hepatitis C virus infection. Virology 2005;1:130-144.
[6] Helbig KJ, Lau DT, Semendric L, Harley HA, Beard MR. Analysis of ISG expression in chronic hepatitis C identifies viperin as a potential antiviral effector. Hepatology 2005;3:702-710.
[7] Asselah T, Bieche I, Laurendeau I, Paradis V, Vidaud D, Degott C, et al. Liver gene expression signature of mild fibrosis in patients with chronic hepatitis C. Gastroenterology 2005;6:2064-2075.
[8] Asselah T, Rubbia-Brandt L, Marcellin P, Negro F. Steatosis in chronic hepatitis C: why does it really matter? Gut 2006;1:123-130.
[9] Chiappini F, Barrier A, Saffroy R, Domart MC, Dagues N, Azoulay D, et al. Exploration of global gene expression in human liver steatosis by high-density oligonucleotide microarray. Lab Invest 2006;2:154-165.
[10] Smith MW, Walters KA, Korth MJ, Fitzgibbon M, Proll S, Thompson JC, et al. Gene expression patterns that correlate with hepatitis C and early progression to fibrosis in liver transplant recipients. Gastroenterology 2006;1:179-187.
[11] Bartel DP. MicroRNAs: genomics, biogenesis, mechanism, and function. Cell 2004;2:281-297.
[12] Meister G, Landthaler M, Dorsett Y, Tuschl T. Sequence-specific inhibition of microRNA- and siRNA-induced RNA silencing. RNA 2004;3:544-550.
[13] Meister G, Tuschl T. Mechanisms of gene silencing by double-stranded RNA. Nature 2004;7006:343-349.
[14] Napoli C, Lemieux C, Jorgensen R Introduction of a chimeric chalcone synthase gene into petunia results in reversible co-suppression of homologous genes in trans. Plant Cell 1990;4:279-289.
[15] Marquez RT, Bandyopadhyay S, Wendlandt EB, Keck K, Hoffer BA, Icardi MS, et al. Correlation between microRNA expression levels and clinical parameters associated with chronic hepatitis C viral infection in humans. Lab Invest 2010;12:1727-1736.
[16] Morita K, Taketomi A, Shirabe K, Umeda K, Kayashima H, Ninomiya M, et al. Clinical significance and potential of hepatic microRNA-122 expression in hepatitis C. Liver Int 2011;4:474-484.
[17] Bihrer V, Friedrich-Rust M, Kronenberger B, Forestier N, Haupenthal J, Shi Y, et al. Serum miR-122 as a Biomarker of Necroinflammation in Patients With Chronic Hepatitis C Virus Infection. Am J Gastroenterol 2011;109:1163-1669.
[22 [23
[24 [25 [26
[28 [29
[32 [33 [34 [35 [36 [37
Bandyopadhyay S, Friedman RC, Marquez RT, Keck K, Kong B, Icardi MS, [38
et al. Hepatitis C virus infection and hepatic stellate cell activation downregulate miR-29: miR-29 overexpression reduces hepatitis C viral abundance in culture. J Infect Dis 2011;12:1753-1762. [39
van Rooij E, Sutherland LB, Thatcher JE, DiMaio JM, Naseem RH, Marshall WS, et al. Dysregulation of microRNAs after myocardial infarction reveals a role of miR-29 in cardiac fibrosis. Proc Natl Acad Sci USA 2008;35:13027-13032. [40
Okamoto K, Mimura K, Murawaki Y, Yuasa I. Association of functional gene polymorphisms of matrix metalloproteinase (MMP)-1, MMP-3 and MMP-9 with the progression of chronic liver disease. J Gastroenterol Hepatol 2005;7:1102-1108. [41
Muhlbauer M, Bosserhoff AK, Hartmann A, Thasler WE, Weiss TS, Herfarth H, et al. A novel MCP-1 gene polymorphism is associated with hepatic MCP-1 expression and severity of HCV-related liver disease. Gastroenterology [42
2003;4:1085-1093.
Valdivielso JM, Fernandez E. Vitamin D receptor polymorphisms and diseases. Clin Chim Acta 2006;1-2:1-12. [43
Baur K, Mertens JC, Schmitt J, Iwata R, Stieger B, Eloranta JJ, et al. Combined effect of 25-OH vitamin D plasma levels and genetic Vitamin D Receptor (NR 1I1) variants on fibrosis progression rate in HCV patients. Liver Int 2011;32:635-643. [44
Huang H, Shiffman ML, Cheung RC, Layden TJ, Friedman S, Abar OT, et al. Identification of two gene variants associated with risk of advanced fibrosis in patients with chronic hepatitis C. Gastroenterology 2006;6:1679-1687. [45
Huang H, Shiffman ML, Friedman S, Venkatesh R, Bzowej N, Abar OT, et al. A 7 gene signature identifies the risk of developing cirrhosis in patients with chronic hepatitis C. Hepatology 2007;2:297-306.
Paris AJ, Snapir Z, Christopherson CD, Kwok SY, Lee UE, Ghiassi-Nejad Z, [46
et al. A polymorphism that delays fibrosis in hepatitis C promotes alternative splicing of AZIN1, reducing fibrogenesis. Hepatology 2011;54:2198-2207.
Marcolongo M, Young B, Dal Pero F, Fattovich G, Peraro L, Guido M, et al. A [47
seven-gene signature (cirrhosis risk score) predicts liver fibrosis progression in patients with initially mild chronic hepatitis C. Hepatology 2009;4:1038-1044. [48
Trepo E, Potthoff A, Pradat P, Bakshi R, Young B, Lagier R, et al. Role of a cirrhosis risk score for the early prediction of fibrosis progression in hepatitis C patients with minimal liver disease. J Hepatol 2011;1:38-44. Petit JM, Masson D, Minello A, Duvillard L, Galland F, Verges B, et al. Lack of [49
association between microsomal triglyceride transfer protein gene polymorphism and liver steatosis in HCV-infected patients. Mol Genet Metab 2006;2:196-198. [50
Zampino R, Ingrosso D, Durante-Mangoni E, Capasso R, Tripodi MF, Restivo L, et al. Microsomal triglyceride transfer protein (MTP) -493G/T gene polymorphism contributes to fat liver accumulation in HCV genotype 3 [51
infected patients. J Viral Hepat 2008;10:740-746.
Mirandola S, Osterreicher CH, Marcolongo M, Datz C, Aigner E, Schlabra-kowski A, et al. Microsomal triglyceride transfer protein polymorphism (-493G/T) is associated with hepatic steatosis in patients with chronic [52
hepatitis C. Liver Int 2009;4:557-565.
Verdi H, Koytak ES, Onder O, Ergul AA, Cinar K, Idilman R, et al. Peroxisome proliferator-activated receptor alpha L162V polymorphism in nonalcoholic [53
steatohepatitis and genotype 1 hepatitis C virus-related liver steatosis. J Investig Med 2005;7:353-359.
Adinolfi LE, Ingrosso D, Cesaro G, Cimmino A, D'Anto M, Capasso R, et al. Hyperhomocysteinemia and the MTHFR C677T polymorphism promote [54
steatosis and fibrosis in chronic hepatitis C patients. Hepatology 2005;5:995-1003.
Borgia G, Gentile I, Fortunato G, Borrelli F, Borelli S, de Caterina M, et al. Homocysteine levels and sustained virological response to pegylated- [55
interferon alpha2b plus ribavirin therapy for chronic hepatitis C: a prospective study. Liver Int 2009;2:248-252.
Iuliano AD, Feingold E, Wahed AS, Kleiner DE, Belle SH, Conjeevaram HS, [56
et al. Host genetics, steatosis and insulin resistance among African Americans and Caucasian Americans with hepatitis C virus genotype-1 infection. Intervirology 2009;1:49-56. [57
Sanchez-Munoz D, Romero-Gomez M, Gonzalez-Escribano MF, Torres B, Castellano-Megias VM, Gomez-Izquierdo L, et al. Tumour necrosis factor alpha polymorphisms are not involved in the development of steatosis in [58
chronic hepatitis C. Eur J Gastroenterol Hepatol 2004;8:761-765. Valenti L, Al-Serri A, Daly AK, Galmozzi E, Rametta R, Dongiovanni P, et al. Homozygosity for the patatin-like phospholipase-3/adiponutrin I148M [59
polymorphism influences liver fibrosis in patients with nonalcoholic fatty liver disease. Hepatology 2005;4:1209-1217.
Romeo S, Kozlitina J, Xing C, Pertsemlidis A, Cox D, Pennacchio LA, et al. Genetic variation in PNPLA3 confers susceptibility to nonalcoholic fatty liver disease. Nat Genet 2008;12:1461-1465.
Rotman Y, Koh C, Zmuda JM, Kleiner DE, Liang TJ. The association of genetic variability in patatin-like phospholipase domain-containing protein 3 (PNPLA3) with histological severity of nonalcoholic fatty liver disease. Hepatology 2010;3:894-903.
Valenti L, Al-Serri A, Daly AK, Galmozzi E, Rametta R, Dongiovanni P, et al. Homozygosity for the patatin-like phospholipase-3/adiponutrin I148M polymorphism influences liver fibrosis in patients with nonalcoholic fatty liver disease. Hepatology 2010;4:1209-1217.
Kollerits B, Coassin S, Kiechl S, Hunt SC, Paulweber B, Willeit J, et al. A common variant in the adiponutrin gene influences liver enzyme values. J Med Genet 2010;2:116-119.
Tian C, Stokowski RP, Kershenobich D, Ballinger DG, Hinds DA. Variant in PNPLA3 is associated with alcoholic liver disease. Nat Genet 2010;1: 21-23.
Trepo E, Pradat P, Potthoff A, Momozawa Y, Quertinmont E, Gustot T, et al. Impact of patatin-like phospholipase-3 (rs738409 C > G) polymorphism on fibrosis progression and steatosis in chronic hepatitis C. Hepatology 2011;1:60-69.
Stickel F, Buch S, Lau K, Zu Schwabedissen HM, Berg T, Ridinger M, et al. Genetic variation in the PNPLA3 gene is associated with alcoholic liver injury in Caucasians. Hepatology 2011;1:86-95.
Nischalke HD, Berger C, Luda C, Berg T, Muller T, Grunhage F, et al. The PNPLA3 rs738409 148M/M genotype is a risk factor for liver cancer in alcoholic cirrhosis but shows no or weak association in hepatitis C cirrhosis. PLoS One 2011;11:e27087.
Bochud PY, Bibert S, Kutalik Z, Patin E, Guergnon J, Nalpas B, et al. IL-28B alleles associated with poor hepatitis C virus (HCV) clearance protect against inflammation and fibrosis in patients infected with non-1 HCV genotypes. Hepatology 2012;2:384-394.
Asselah T, De Muynck S, Broet P, Masliah-Planchon J, Blanluet M, Bieche I, et al. IL-28B polymorphism is associated with treatment response in patients with genotype 4 chronic hepatitis C.J Hepatol 2011;3:527-532. Marabita F, Aghemo A, De Nicola S, Rumi MG, Cheroni C, Scavelli R, et al. Genetic variation in IL-28B gene is not associated with fibrosis progression in patients with chronic hepatitis C and known date of infection. Hepatology 2011;54:1127-1134.
Cai T, Dufour JF, Muellhaupt B, Gerlach T, Heim M, Moradpour D, et al. Viral
genotype-specific role of PNPLA3, PPARG, MTTP, and IL-28B in hepatitis C
virus-associated steatosis. J Hepatol 2011;55:529-535.
Tillmann HL, Patel K, Muir AJ, Guy CD, Li JH, Lao XQ, et al. Beneficial IL-28B
genotype associated with lower frequency of hepatic steatosis in patients
with chronic hepatitis C.J Hepatol 2011;55:1195-1200.
Li JH, Lao XQ, Tillmann HL, Rowell J, Patel K, Thompson A, et al. Interferon-
lambda genotype and low serum low-density lipoprotein cholesterol levels
in patients with chronic hepatitis C infection. Hepatology
2010;6:1904-1911.
Chen L, Borozan I, Feld J, Sun J, Tannis LL, Coltescu C, et al. Hepatic gene expression discriminates responders and nonresponders in treatment of chronic hepatitis C viral infection. Gastroenterology 2005;5:1437-1444. Asselah T, Bieche I, Narguet S, Sabbagh A, Laurendeau I, Ripault MP, et al. Liver gene expression signature to predict response to pegylated interferon plus ribavirin combination therapy in patients with chronic hepatitis C. Gut 2008;4:516-524.
Younossi ZM, Baranova A, Afendy A, Collantes R, Stepanova M, Manyam G, et al. Early gene expression profiles of patients with chronic hepatitis C treated with pegylated interferon-alfa and ribavirin. Hepatology 2009;3:763-774.
Butera D, Marukian S, Iwamaye AE, Hembrador E, Chambers TJ, Di Bisceglie AM, et al. Plasma chemokine levels correlate with the outcome of antiviral therapy in patients with hepatitis C. Blood 2005;4:1175-1182. Ji X, Cheung R, Cooper S, Li Q, Greenberg HB, He XS. Interferon alfa regulated gene expression in patients initiating interferon treatment for chronic hepatitis C. Hepatology 2003;3:610-621.
Pedersen IM, Cheng G, Wieland S, Volinia S, Croce CM, Chisari FV, et al. Interferon modulation of cellular microRNAs as an antiviral mechanism. Nature 2007;7164:919-922.
Sarasin-Filipowicz M, Krol J, Markiewicz I, Heim MH, Filipowicz W. Decreased levels of microRNA miR-122 in individuals with hepatitis C responding poorly to interferon therapy. Nat Med 2009;1:31-33. Lanford RE, Hildebrandt-Eriksen ES, Petri A, Persson R, Lindow M, Munk ME, et al. Therapeutic silencing of microRNA-122 in primates with chronic hepatitis C virus infection. Science 2010;5962:198-201.
[62 [63
[65 [66
[70 [71 [72 [73 [74
[75 [76
[77 [78
Janssen LH, Reesink WH, Zeuzem S, Läwitz E, Rodriguez-Torres M, Chen A, [81
et al. A randomized, double-blind, placebo (PLB) controlled safety and antiviral proof of concept study of miravirsen (MIR), an oligonucleotide targeting MIR-122, in treatment naive patients with genotype 1 (GT1) chronic HCV infection. AASLD, 2011. [82
Murakami Y, Tanaka M, Toyoda H, Hayashi K, Kuroda M, Tajima A, et al. Hepatic microRNA expression is associated with the response to interferon treatment of chronic hepatitis C. BMC Med Genomics 2010;48. Khakoo SI, Thio CL, Martin MP, Brooks CR, Gao X, Astemborski J, et al. HLA [83
and NK cell inhibitory receptor genes in resolving hepatitis C virus infection. Science 2004;5685:872-874.
Knapp S, Warshow U, Hegazy D, Brackenbury L, Guha IN, Fowell A, et al. Consistent beneficial effects of killer cell immunoglobulin-like receptor [84
2DL3 and group 1 human leukocyte antigen-C following exposure to hepatitis C virus. Hepatology 2010;4:1168-1175.
Vidal-Castineira JR, Lopez-Vazquez A, Diaz-Pena R, Alonso-Arias R, Marti- [85
nez-Borra J, Perez R, et al. Effect of killer immunoglobulin-like receptors in the response to combined treatment in patients with chronic hepatitis C virus infection. J Virol 2010;1:475-481.
Azocar J, Clavijo OP, Yunis EJ. MHC class II genes in HCV viral clearance of [86
hepatitis C infected Hispanic patients. Hum Immunol 2003;1:99-102.
Romero V, Azocar J, Zuniga J, Clavijo OP, Terreros D, Gu X, et al. Interaction
of NK inhibitory receptor genes with HLA-C and MHC class II alleles in
Hepatitis C virus infection outcome. Mol Immunol 2008;9:2429-2436.
Suppiah V, Gaudieri S, Armstrong NJ, O'Connor KS, Berg T, Weltman M, et al.
IL-28B, HLA-C, and KIR variants additively predict response to therapy in [87
chronic hepatitis C virus infection in a European Cohort: a cross-sectional
study. PLoS Med 2011;9:e1001092.
Golden-Mason L, Bambha KM, Cheng L, Howell CD, Taylor MW, Clark PJ, [88
et al. Natural killer inhibitory receptor expression associated with treatment failure and interleukin-28B genotype in patients with chronic hepatitis C. Hepatology 2011;5:1559-1569. [89
Nattermann J, Vogel M, Berg T, Danta M, Axel B, Mayr C, et al. Effect of the interleukin-6 C174G gene polymorphism on treatment of acute and chronic hepatitis C in human immunodeficiency virus coinfected patients. Hepatology 2007;4:1016-1025. [90 Yee LJ, Im K, Borg B, Yang H, Liang TJ. Interleukin-6 haplotypes and the response to therapy of chronic hepatitis C virus infection. Genes Immun 2009;4:365-372.
Payvandi F, Amrute S, Fitzgerald-Bocarsly P. Exogenous and endogenous IL- [91
10 regulate IFN-alpha production by peripheral blood mononuclear cells in response to viral stimulation. J Immunol 1998;12:5861-5868.
Edwards-Smith CJ, Jonsson JR, Purdie DM, Bansal A, Shorthouse C, Powell [92
EE. Interleukin-10 promoter polymorphism predicts initial response of
chronic hepatitis C to interferon alfa. Hepatology 1999;2:526-530.
Paladino N, Fainboim H, Theiler G, Schroder T, Munoz AE, Flores AC, et al. [93
Gender susceptibility to chronic hepatitis C virus infection associated with
interleukin 10 promoter polymorphism. J Virol 2006;18:9144-9150.
Huang Y, Yang H, Borg BB, Su X, Rhodes SL, Yang K, et al. A functional SNP of
interferon-gamma gene is important for interferon-alpha-induced and [94
spontaneous recovery from hepatitis C virus infection. Proc Natl Acad Sci
USA 2007;3:985-990.
Ge D, Fellay J, Thompson AJ, Simon JS, Shianna KV, Urban TJ, et al. Genetic [95
variation in IL-28B predicts hepatitis C treatment-induced viral clearance. Nature 2009;7262:399-401.
Thompson AJ, Muir AJ, Sulkowski MS, Ge D, Fellay J, Shianna KV, et al. Interleukin-28B polymorphism improves viral kinetics and is the strongest [96
pretreatment predictor of sustained virologic response in genotype 1 hepatitis C virus. Gastroenterology 2010;1,120-9 e18.
Thomas DL, Thio CL, Martin MP, Qi Y, Ge D, O'Huigin C, et al. Genetic [97
variation in IL-28B and spontaneous clearance of hepatitis C virus. Nature 2009;7265:798-801. [98
McCarthy JJ, Li JH, Thompson A, Suchindran S, Lao XQ, Patel K, et al. Replicated association between an IL-28B gene variant and a sustained [99
response to pegylated interferon and ribavirin. Gastroenterology 2010;7:2307-2314. [100
Nattermann J, Vogel M, Nischalke HD, Danta M, Mauss S, Stellbrink HJ, et al. Genetic variation in IL-28B and treatment-induced clearance of hepatitis C virus in HIV-positive patients with acute and chronic hepatitis C. J Infect [101
Dis 2011;5:595-601.
Tillmann HL, Thompson AJ, Patel K, Wiese M, Tenckhoff H, Nischalke HD, et al. A polymorphism near IL-28B is associated with spontaneous clearance of acute hepatitis C virus and jaundice. Gastroenterology [102
2010;5:1586-1592, 1592 e1.
Montes-Cano MA, Garcia-Lozano JR, Abad-Molina C, Romero-Gomez M, Barroso N, Aguilar-Reina J, et al. Interleukin-28B genetic variants and hepatitis virus infection by different viral genotypes. Hepatology 2010;1:33-37.
Rauch A, Kutalik Z, Descombes P, Cai T, Di Iulio J, Mueller T, et al. Genetic variation in IL-28B is associated with chronic hepatitis C and treatment failure: a genome-wide association study. Gastroenterology 2010;4:1338-1345, 1345 e1-7.
Lindh M, Lagging M, Farkkila M, Langeland N, Morch K, Nilsson S, et al. Interleukin 28B gene variation at rs12979860 determines early viral kinetics during treatment in patients carrying genotypes 2 or 3 of hepatitis C virus. J Infect Dis 2011;12:1748-1752.
Lange CM, Moradpour D, Doehring A, Lehr HA, Mullhaupt B, Bibert S, et al. Impact of donor and recipient IL-28B rs12979860 genotypes on hepatitis C virus liver graft reinfection. J Hepatol 2010;55:322-327. Fukuhara T, Taketomi A, Motomura T, Okano S, Ninomiya A, Abe T, et al. Variants in IL-28B in liver recipients and donors correlate with response to peg-interferon and ribavirin therapy for recurrent hepatitis C. Gastroen-terology 2010;5:1577-1585, 1585 e1-3.
Dean M, Carrington M, Winkler C, Huttley GA, Smith MW, Allikmets R, et al. Genetic restriction of HIV-1 infection and progression to AIDS by a deletion allele of the CKR5 structural gene. Hemophilia Growth and Development Study, Multicenter AIDS Cohort Study, Multicenter Hemophilia Cohort Study, San Francisco City Cohort, ALIVE Study. Science 1996;5283:1856-1862.
Michael NL, Louie LG, Rohrbaugh AL, Schultz KA, Dayhoff DE, Wang CE, et al. The role of CCR5 and CCR2 polymorphisms in HIV-1 transmission and disease progression. Nat Med 1997;10:1160-1162. Huang Y, Paxton WA, Wolinsky SM, Neumann AU, Zhang L, He T, et al. The role of a mutant CCR5 allele in HIV-1 transmission and disease progression. Nat Med 1996;11:1240-1243.
Woitas RP, Ahlenstiel G, Iwan A, Rockstroh JK, Brackmann HH, Kupfer B, et al. Frequency of the HIV-protective CC chemokine receptor 5-Delta32/ Delta32 genotype is increased in hepatitis C. Gastroenterology 2002;7:1721-1728.
Hellier S, Frodsham AJ, Hennig BJ, Klenerman P, Knapp S, Ramaley P, et al. Association of genetic variants of the chemokine receptor CCR5 and its ligands, RANTES and MCP-2, with outcome of HCV infection. Hepatology 2003;6:1468-1476.
Mangia A, Santoro R, D'Agruma L, Andriulli A. HCV chronic infection and CCR5-delta32/delta32. Gastroenterology 2003;3:868-869, author reply 869-870.
Zhang M, Goedert JJ, O'Brien TR. High frequency of CCR5-delta32 homo-
zygosity in HCV-infected, HIV-1-uninfected hemophiliacs results from
resistance to HIV-1. Gastroenterology 2003;3:867-868.
Promrat K, McDermott DH, Gonzalez CM, Kleiner DE, Koziol DE, Lessie M,
et al. Associations of chemokine system polymorphisms with clinical
outcomes and treatment responses of chronic hepatitis C. Gastroenterology
2003;2:352-360.
Goulding C, McManus R, Murphy A, MacDonald G, Barrett S, Crowe J, et al. The CCR5-delta32 mutation: impact on disease outcome in individuals with hepatitis C infection from a single source. Gut 2005;8:1157-1161. Nattermann J, Timm J, Nischalke HD, Olbrich A, Michalk M, Tilmann HL, et al. The predictive value of IL-28B gene polymorphism for spontaneous clearance in a single source outbreak cohort is limited in patients carrying the CCR5Delta32 mutation. J Hepatol 2011;203:595-601. Schwabe RF, Bataller R, Brenner DA. Human hepatic stellate cells express CCR5 and RANTES to induce proliferation and migration. Am J Physiol Gastrointest Liver Physiol 2003;5:G949-G958.
Seki E, De Minicis S, Gwak GY, Kluwe J, Inokuchi S, Bursill CA, et al. CCR1 and CCR5 promote hepatic fibrosis in mice. J Clin Invest 2009;7:1858-1870. Kuhmann SE, Hartley O. Targeting chemokine receptors in HIV: a status report. Annu Rev Pharmacol Toxicol 2008:425-461. Affo S, Bataller R. RANTES antagonism: a promising approach to treat chronic liver diseases. J Hepatol 2011;4:936-938.
Sarasin-Filipowicz M, Oakeley EJ, Duong FH, Christen V, Terracciano L, Filipowicz W, et al. Interferon signaling and treatment outcome in chronic hepatitis C. Proc Natl Acad Sci USA 2008;19:7034-7039. Marcello T, Grakoui A, Barba-Spaeth G, Machlin ES, Kotenko SV, MacDonald MR, et al. Interferons alpha and lambda inhibit hepatitis C virus replication with distinct signal transduction and gene regulation kinetics. Gastroen-terology 2006;6:1887-1898.
Dill MT, Duong FH, Vogt JE, Bibert S, Bochud PY, Terracciano L, et al. Interferon-induced gene expression is a stronger predictor of treatment
response than IL-28B genotype in patients with hepatitis C. Gastroenter- [122
ology 2010;3:1021-1031.
[103] Honda M, Sakai A, Yamashita T, Nakamoto Y, Mizukoshi E, Sakai Y, et al. Hepatic ISG expression is associated with genetic variation in interleukin
28B and the outcome of IFN therapy for chronic hepatitis C. Gastroenter- [123
ology 2010;2:499-509.
[104] Urban TJ, Thompson AJ, Bradrick SS, Fellay J, Schuppan D, Cronin KD, et al.
IL-28B genotype is associated with differential expression of intrahepatic [124
interferon-stimulated genes in patients with chronic hepatitis C. Hepatol-ogy 2010;6:1888-1896.
[105] Abe H, Hayes CN, Ochi H, Maekawa T, Tsuge M, Miki D, et al. IL28 variation [125 affects expression of interferon stimulated genes and peg-interferon and
ribavirin therapy. J Hepatol 2011;6:1094-1101.
[106] Diago M, Castellano G, Garcia-SamaniegoJ, Perez C, Fernandez I, Romero M,
et al. Association of pretreatment serum interferon gamma inducible [126
protein 10 levels with sustained virological response to peginterferon plus ribavirin therapy in genotype 1 infected patients with chronic hepatitis C. Gut 2006;3:374-379. [127
[107] Zeremski M, Markatou M, Brown QB, Dorante G, Cunningham-Rundles S, Talal AH. Interferon gamma-inducible protein 10: a predictive marker of successful treatment response in hepatitis C virus/HIV-coinfected patients. [128 J Acquir Immune Defic Syndr 2007;3:262-268.
[108] Romero AI, Lagging M, Westin J, Dhillon AP, Dustin LB, Pawlotsky JM, et al. Interferon (IFN)-gamma-inducible protein-10: association with histological
results, viral kinetics, and outcome during treatment with pegylated IFN- [129
alpha 2a and ribavirin for chronic hepatitis C virus infection. J Infect Dis 2006;7:895-903.
[109] Narumi S, Tominaga Y, Tamaru M, Shimai S, Okumura H, Nishioji K, et al. Expression of IFN-inducible protein-10 in chronic hepatitis. J Immunol [130 1997;11:5536-5544.
[110] Lagging M, Romero AI, Westin J, Norkrans G, Dhillon AP, Pawlotsky JM, et al. IP-10 predicts viral response and therapeutic outcome in diffi-cult-to-treat patients with HCV genotype 1 infection. Hepatology 2006;6: 1617-1625. [131
[111] Lagging M, Askarieh G, Negro F, Bibert S, Soderholm J, Westin J, et al. Response prediction in chronic hepatitis C by assessment of IP-10 and IL-28B-related single nucleotide polymorphisms. PLoS One 2011;2:e17232. [132
[112] Darling JM, Aerssens J, Fanning G, McHutchison JG, Goldstein DB, Thompson AJ, et al. Quantitation of pretreatment serum interferon-gamma-inducible protein-10 improves the predictive value of an IL-28B gene [133 polymorphism for hepatitis C treatment response. Hepatology 2011;1:14-22.
[113] Beinhardt S, Aberle JH, Strasser M. Dulic-Lakovic E, Maieron A, Kreil A, [134 et al., Serum Level of IP-10 Increases Predictive Value of IL-28B Polymorphisms for Spontaneous Clearance of Acute HCV Infectio. Gastroenterology 2011;142:78-85. [135
[114] Bitetto D, Fattovich G, Fabris C, Ceriani E, Falleti E, Fornasiere E, et al. Complementary role of vitamin D deficiency and the interleukin-28B rs12979860 C/T polymorphism in predicting antiviral response in chronic hepatitis C. Hepatology 2011;4:1118-1126. [136
[115] White JH. Vitamin D signaling, infectious diseases, and regulation of innate immunity. Infect Immun 2008;9:3837-3843.
[116] Akuta N, Suzuki F, Sezaki H, Suzuki Y, Hosaka T, Someya T, et al. Association
of amino acid substitution pattern in core protein of hepatitis C virus [137
genotype 1b high viral load and non-virological response to interferon-ribavirin combination therapy. Intervirology 2005;6:372-380.
[117] Enomoto N, Sakuma I, Asahina Y, Kurosaki M, Murakami T, Yamamoto C, et al. Comparison of full-length sequences of interferon-sensitive and resistant hepatitis C virus 1b. Sensitivity to interferon is conferred by [138 amino acid substitutions in the NS5A region. J Clin Invest 1995;1:224-230.
[118] Akuta N, Suzuki F, Hirakawa M, Kawamura Y, Yatsuji H, Sezaki H, et al. Amino acid substitution in hepatitis C virus core region and genetic variation near the interleukin 28B gene predict viral response to telaprevir [139 with peginterferon and ribavirin. Hepatology 2010;2:421-429.
[119] Hayes CN, Kobayashi M, Akuta N, Suzuki F, Kumada H, Abe H, et al. HCV substitutions and IL-28B polymorphisms on outcome of peg-interferon
plus ribavirin combination therapy. Gut 2011;2:261-267. [140
[120] Fried MW, Hadziyannis SJ, Shiffman ML, Messinger D, Zeuzem S. Rapid virological response is the most important predictor of sustained virolog-
ical response across genotypes in patients with chronic hepatitis C virus [141
infection. J Hepatol 2011;1:69-75.
[121] Mangia A, Thompson AJ, Santoro R, Piazzolla V, Copetti M, Minerva N, et al. Limited utility of IL-28B in the setting of response-guided treatment with
detailed on-treatment virological monitoring. Hepatology 2011;24: [142
772-780.
Mangia A, Thompson AJ, Santoro R, Piazzolla V, Tillmann HL, Patel K, et al.
An IL-28B polymorphism determines treatment response of hepatitis C
virus genotype 2 or 3 patients who do not achieve a rapid virologic
response. Gastroenterology 2010;3:821-827, 827 e1.
Sarrazin C, Susser S, Doehring A, Lange CM, Muller T, Schlecker C, et al.
Importance of IL-28B gene polymorphisms in hepatitis C virus genotype 2
and 3 infected patients. J Hepatol 2011;3:415-421.
Suppiah V, Moldovan M, Ahlenstiel G, Berg T, Weltman M, Abate ML, et al.
IL-28B is associated with response to chronic hepatitis C interferon-alpha
and ribavirin therapy. Nat Genet 2009;10:1100-1104.
Tanaka Y, Nishida N, Sugiyama M, Kurosaki M, Matsuura K, Sakamoto N,
et al. Genome-wide association of IL-28B with response to pegylated
interferon-alpha and ribavirin therapy for chronic hepatitis C. Nat Genet
2009;10:1105-1109.
Langhans B, Kupfer B, Braunschweiger I, Arndt S, Schulte W, Nischalke HD, et al. Interferon-lambda serum levels in hepatitis C. J Hepatol 2011;5:859-865.
di Iulio J, Ciuffi A, Fitzmaurice K, Kelleher D, Rotger M, Fellay J, et al. Estimating the net contribution of interleukin-28B variation to spontaneous hepatitis C virus clearance. Hepatology 2011;5:1446-1454. Thomas E, Gonzalez VD, Li Q, Modi AA, Chen W, Noureddin M, et al. HCV Infection Induces a Unique Hepatic Innate Immune Response Associated with Robust Production of Type III Interferons. Gastroenterology 2012;142:978-988.
Muir AJ, Shiffman ML, Zaman A, Yoffe B, de la Torre A, Flamm S, et al. Phase 1b study of pegylated interferon lambda 1 with or without ribavirin in patients with chronic genotype 1 hepatitis C virus infection. Hepatology 2010;3:822-832.
Zeuzem S, Arora S, Bacon B, Box T, Charlton M, Diago M, et al. Pegylated interferon-lambda shows superior viral response with improved safety and tolerability versus PegIFN alpha in HCV patients (G1/2/3/4): emerge phase IIB through week 12. In: 46th Annual Meeting of the European Association for the study of the Liver (EASL); 2011.
Poordad F, McCone Jr J, Bacon BR, Bruno S, Manns MP, Sulkowski MS, et al. Boceprevir for untreated chronic HCV genotype 1 infection. N Engl J Med 2011;13:1195-1206.
Jacobson IM, McHutchison JG, Dusheiko G, Di Bisceglie AM, Reddy KR,
Bzowej NH, et al. Telaprevir for previously untreated chronic hepatitis C
virus infection. N Engl J Med 2011;25:2405-2416.
Zeuzem S, Andreone P, Pol S, Lawitz E, Diago M, Roberts S, et al.
Telaprevir for retreatment of HCV infection. N Engl J Med 2011;25:
2417-2428.
Bacon BR, Gordon SC, Lawitz E, Marcellin P, Vierling JM, Zeuzem S, et al. Boceprevir for previously treated chronic HCV genotype 1 infection. N Engl J Med 2011;13:1207-1217.
Jacobson IM, Catlett I, Marcellin P, Bzowej NH, Muir AJ, Adda M, et al. Telaprevir substantially improved SVR rates across all IL-28B genotypes in the advance trial. In: 46th Annual Meeting of the European Association for the study of the Liver (EASL); 2011.
Pol S, Aerssens J, Zeuzem S. Similar SVR rates in IL-28B CC, CT or TT prior relapser partial- or null-responder patients treated with telaprevir/pegin-terferon/ribavirin: retrospective analysis of the REALIZE study. J Hepatol 2011 (Suppl. 1):S6.
Gane EJ, Roberts SK, Stedman CA, Angus PW, Ritchie B, Elston R, et al. Oral combination therapy with a nucleoside polymerase inhibitor (RG7128) and danoprevir for chronic hepatitis C genotype 1 infection (INFORM-1): a randomised, double-blind, placebo-controlled, dose-escalation trial. Lancet 2010;9751:1467-1475.
Chu TW, Kulkarni R, Gane EJ, Roberts SK, Stedman C, Angus PW, et al. Effect of IL-28B Genotype on Early Viral Kinetics during Interferon-Free Treatment of Patients with Chronic Hepatitis C. Gastroenterology 2012;142:790-795.
Poordad F. IL-28B polymorphism predicts virologic response in patients with hepatitis C genotype 1 treated with boceprevir combination therapy. In: 46th Annual Meeting of the European Association for the study of the Liver (EASL); 2011.
Fried MW, Shiffman ML, Reddy KR, Smith C, Marinos G, Goncales Jr FL, et al. Peginterferon alfa-2a plus ribavirin for chronic hepatitis C virus infection. N Engl J Med 2002;13:975-982.
Bronowicki JP, Ouzan D, Asselah T, Desmorat H, Zarski JP, Foucher J, et al. Effect of ribavirin in genotype 1 patients with hepatitis C responding to pegylated interferon alfa-2a plus ribavirin. Gastroenterology 2006;4:1040-1048.
Manns MP, McHutchison JG, Gordon SC, Rustgi VK, Shiffman M, Reindollar R, et al. Peginterferon alfa-2b plus ribavirin compared with interferon
alfa-2b plus ribavirin for initial treatment of chronic hepatitis C: a randomised trial. Lancet 2001;9286:958-965.
[143] Maylin S, Martinot-Peignoux M, Moucari R, Boyer N, Ripault MP, Cazals-Hatem D, et al. Eradication of hepatitis C virus in patients successfully treated for chronic hepatitis C. Gastroenterology 2008;3:821-829.
[144] Martinot-Peignoux M, Maylin S, Moucari R, Ripault MP, Boyer N, Cardoso AC, et al. Virological response at 4 weeks to predict outcome of hepatitis C treatment with pegylated interferon and ribavirin. Antivir Ther 2009;4: 501-511.
[145] Romero-Gomez M, Del Mar Viloria M, Andrade RJ, Salmeron J, Diago M, Fernandez-Rodriguez CM, et al. Insulin resistance impairs sustained response rate to peginterferon plus ribavirin in chronic hepatitis C patients. Gastroenterology 2005;3:636-641.
[146] Peffault de Latour R, Ribaud P, Robin M, Valla D, Marcellin P, Socie G, et al. Allogeneic hematopoietic cell transplant in HCV-infected patients. J Hepatol 2008;6:1008-1017.
[147] Asselah T, Essioux L, Marcellin P, Fried MW, Jensen DM, Germer S, et al. A chromosome 19 SNP (RS12979860) predicts outcome (EVR/SVR) in HCV patients treated with interferon, independent of pegylation or ribavirin. J Hepatol 2010;EASL:A1180.
[148] Kotenko SV, Gallagher G, Baurin VV, Lewis-Antes A, Shen M, Shah NK, et al. IFN-lambdas mediate antiviral protection through a distinct class II cytokine receptor complex. Nat Immunol 2003;1:69-77.
[149] Sheppard P, Kindsvogel W, Xu W, Henderson K, Schlutsmeyer S, Whitmore TE, et al. IL-28, IL-29 and their class II cytokine receptor IL-28R. Nat Immunol 2003;1:63-68.
[150] Coccia EM, Severa M, Giacomini E, Monneron D, Remoli ME, Julkunen I, et al. Viral infection and Toll-like receptor agonists induce a differential expression of type I and lambda interferons in human plasmacytoid and monocyte-derived dendritic cells. Eur J Immunol 2004;3:796-805.
[151] Pagliaccetti NE, Robek MD. Interferon-lambda in the immune response to hepatitis B virus and hepatitis C virus. J Interferon Cytokine Res 2010;8:585-590.
[152] Witte K, Gruetz G, Volk HD, Looman AC, Asadullah K, Sterry W, et al. Despite IFN-lambda receptor expression, blood immune cells, but not keratinocytes or melanocytes, have an impaired response to type III interferons: implications for therapeutic applications of these cytokines. Genes Immun 2009;8:702-714.
[153] Doyle SE, Schreckhise H, Khuu-Duong K, Henderson K, Rosier R, Storey H, et al. Interleukin-29 uses a type 1 interferon-like program to promote antiviral responses in human hepatocytes. Hepatology 2006;4: 896-906.
[154] Zhou Z, Hamming OJ, Ank N, Paludan SR, Nielsen AL, Hartmann R. Type III interferon (IFN) induces a type I IFN-like response in a restricted subset of cells through signaling pathways involving both the Jak-STAT pathway and the mitogen-activated protein kinases. J Virol 2007;14:7749-7758.
[155] Renauld JC. Class II cytokine receptors and their ligands: key antiviral and inflammatory modulators. Nat Rev Immunol 2003;8:667-676.
[156] Der SD, Zhou A, Williams BR, Silverman RH. Identification of genes differentially regulated by interferon alpha, beta, or gamma using oligo-nucleotide arrays. Proc Natl Acad Sci USA 1998;26:15623-15628.
[157] Rani MR, Foster GR, Leung S, Leaman D, Stark GR, Ransohoff RM. Characterization of beta-R1, a gene that is selectively induced by interferon beta (IFN-beta) compared with IFN-alpha. J Biol Chem 1996;37: 22878-22884.
[158] Dai R, Phillips RA, Zhang Y, Khan D, Crasta O, Ahmed SA Suppression of LPS-induced Interferon-gamma and nitric oxide in splenic lymphocytes by select estrogen-regulated microRNAs: a novel mechanism of immune modulation. Blood 2008;12:4591-4597.
[159] Jordan WJ, Eskdale J, Srinivas S, Pekarek V, Kelner D, Rodia M, et al. Human interferon lambda-1 (IFN-lambda1/IL-29) modulates the Th1/Th2 response. Genes Immun 2007;3:254-261.
[160] Srinivas S, Dai J, Eskdale J, Gallagher GE, Megjugorac NJ, Gallagher G. Interferon-lambda1 (interleukin-29) preferentially down-regulates inter-leukin-13 over other T helper type 2 cytokine responses in vitro. Immunology 2008;4:492-502.
[161] Morrow MP, Yan J, Pankhong P, Shedlock DJ, Lewis MG, Talbott K, et al. IL-28B/IFN-lambda 3 drives granzyme B loading and significantly increases CTL killing activity in macaques. Mol Ther 2010;9:1714-1723.
[162] Robek MD, Boyd BS, Chisari FV. Lambda interferon inhibits hepatitis B and C virus replication. J Virol 2005;6:3851-3854.
[163] Zhang L, Jilg N, Shao RX, Lin W, Fusco DN, Zhao H, et al. IL-28B inhibits hepatitis C virus replication through the JAK-STAT pathway. J Hepatol 2010;55:289-298.