51
|
Huang CI, Huang CF, Yeh ML, Lin YH, Liang PC, Liu SYV, Hsieh MH, Lin ZY, Chen SC, Huang JF, Chuang WL, Dai CY, Yu ML. Role of IL-28B genetic variants in HCV-related liver disease severity in patients with different viral genotypes. Medicine (Baltimore) 2018; 97:e9782. [PMID: 29517696 PMCID: PMC5882450 DOI: 10.1097/md.0000000000009782] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/04/2017] [Revised: 12/04/2017] [Accepted: 01/10/2018] [Indexed: 12/25/2022] Open
Abstract
Reports of the role of host interleukin 28B (IL-28B) genetic variants in liver disease severity in patients with chronic hepatitis C (CHC) have obtained conflicting results. The impact of IL-28B in Asian patients with different viral genotypes remains elusive.We try to elucidate the effect of IL-28B genetic variants in a large Asian cohort with different viral genotypes.The association between the IL-28B rs8099917 genotype and liver fibrosis was investigated in 1288 patients with biopsy-proven CHC.Patients with hepatitis C virus genotype 1 (HCV-1) infection comprised 59.4% of the population. The remaining 40.6% (518 patients) did not have HCV-1 infection. Of the 1084 patients with the IL-28 genotype, 85.6% (928 patients) had the TT genotype. Univariate analysis revealed that, compared to patients without advanced liver fibrosis, patients with advanced liver fibrosis (Metavir fibrosis score 3-4) had an older age, a lower platelet count, a higher α-fetoprotein level, a higher alanine aminotransferase level, a higher incidence of diabetes, and a higher frequency of rs8099917 non-TT genotype carriage.Logistic regression analysis revealed that factors significantly associated with advanced liver fibrosis included age (odds ratio [OR]/95% confidence interval [CI]: 1.023/1.009-1.037, P = .001), diabetes (OR/CI: 1.736/1.187-2.539, P = .004), α-fetoprotein (OR/CI: 1.007/1.002-1.012, P = .009), platelet count (OR/CI: 0.991/0.988-0.993, P < .001), and carriage of the rs8099917 non-TT genotype (OR/CI: 0.585/0.400-0.856, P = .006). When patients were classified by viral genotype, factors that had significant independent associations with advanced liver fibrosis in patients with HCV-1 infection included diabetes (OR/CI: 2.379/1.452-3.896, P = .001), α-fetoprotein (OR/CI: 1.023/1.012-1.035, P < .001), platelet count (OR/CI: 0.99/0.987-0.994, P < .001), and carriage of the rs8099917 non-TT genotype (OR/CI: 0.529/0.328-0.854, P = .009). In patients who had advanced liver fibrosis but not HCV-1 infection, factors that had significant independent associations with advanced liver fibrosis included age (OR/CI: 1.039/1.016-1.063, P = .001) and platelet count (OR/CI: 0.99/0.986-0.995, P < .001); additionally, IL-28B genetic variants were not associated with liver disease severity.Unfavorable IL-28B genetic variants were associated with advanced liver disease. The genetic effect is limited to patients with HCV-1 infection.
Collapse
Affiliation(s)
- Ching-I Huang
- Graduate Institute of Clinical Medicine, College of Medicine, Kaohsiung Medical University
- Hepatobiliary Division, Department of Internal Medicine, Kaohsiung Medical University Hospital
| | - Chung-Feng Huang
- Graduate Institute of Clinical Medicine, College of Medicine, Kaohsiung Medical University
- Hepatobiliary Division, Department of Internal Medicine, Kaohsiung Medical University Hospital
- Faculty of Internal Medicine, College of Medicine, Kaohsiung Medical University
| | - Ming-Lun Yeh
- Hepatobiliary Division, Department of Internal Medicine, Kaohsiung Medical University Hospital
- Faculty of Internal Medicine, College of Medicine, Kaohsiung Medical University
| | - Yi-Hung Lin
- Hepatobiliary Division, Department of Internal Medicine, Kaohsiung Medical University Hospital
- Department of Internal Medicine, Kaohsiung Municipal Hsiao-Kang Hospital, Kaohsiung Medical University Hospital
| | - Po-Cheng Liang
- Hepatobiliary Division, Department of Internal Medicine, Kaohsiung Medical University Hospital
- Department of Internal Medicine, Kaohsiung Municipal Hsiao-Kang Hospital, Kaohsiung Medical University Hospital
| | - Shang-Yin Vanson Liu
- Department of Marine Biotechnology and Resources, National Sun Yat-Sen University
| | - Meng-Hsuan Hsieh
- Hepatobiliary Division, Department of Internal Medicine, Kaohsiung Medical University Hospital
- Faculty of Internal Medicine, College of Medicine, Kaohsiung Medical University
- Department of Preventive Medicine, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
| | - Zu-Yau Lin
- Hepatobiliary Division, Department of Internal Medicine, Kaohsiung Medical University Hospital
- Faculty of Internal Medicine, College of Medicine, Kaohsiung Medical University
| | - Shinn-Cherng Chen
- Hepatobiliary Division, Department of Internal Medicine, Kaohsiung Medical University Hospital
- Faculty of Internal Medicine, College of Medicine, Kaohsiung Medical University
| | - Jee-Fu Huang
- Graduate Institute of Clinical Medicine, College of Medicine, Kaohsiung Medical University
- Hepatobiliary Division, Department of Internal Medicine, Kaohsiung Medical University Hospital
- Faculty of Internal Medicine, College of Medicine, Kaohsiung Medical University
| | - Wan-Long Chuang
- Graduate Institute of Clinical Medicine, College of Medicine, Kaohsiung Medical University
- Hepatobiliary Division, Department of Internal Medicine, Kaohsiung Medical University Hospital
- Faculty of Internal Medicine, College of Medicine, Kaohsiung Medical University
| | - Chia-Yen Dai
- Graduate Institute of Clinical Medicine, College of Medicine, Kaohsiung Medical University
- Hepatobiliary Division, Department of Internal Medicine, Kaohsiung Medical University Hospital
- Faculty of Internal Medicine, College of Medicine, Kaohsiung Medical University
- Department of Preventive Medicine, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
| | - Ming-Lung Yu
- Graduate Institute of Clinical Medicine, College of Medicine, Kaohsiung Medical University
- Hepatobiliary Division, Department of Internal Medicine, Kaohsiung Medical University Hospital
- Faculty of Internal Medicine, College of Medicine, Kaohsiung Medical University
| |
Collapse
|
52
|
Morozov VA, Lagaye S. Hepatitis C virus: Morphogenesis, infection and therapy. World J Hepatol 2018; 10:186-212. [PMID: 29527256 PMCID: PMC5838439 DOI: 10.4254/wjh.v10.i2.186] [Citation(s) in RCA: 87] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/04/2017] [Revised: 01/11/2018] [Accepted: 02/07/2018] [Indexed: 02/06/2023] Open
Abstract
Hepatitis C virus (HCV) is a major cause of liver diseases including liver cirrhosis and hepatocellular carcinoma. Approximately 3% of the world population is infected with HCV. Thus, HCV infection is considered a public healthy challenge. It is worth mentioning, that the HCV prevalence is dependent on the countries with infection rates around 20% in high endemic countries. The review summarizes recent data on HCV molecular biology, the physiopathology of infection (immune-mediated liver damage, liver fibrosis and lipid metabolism), virus diagnostic and treatment. In addition, currently available in vitro, ex vivo and animal models to study the virus life cycle, virus pathogenesis and therapy are described. Understanding of both host and viral factors may in the future lead to creation of new approaches in generation of an efficient therapeutic vaccine.
Collapse
Affiliation(s)
- Vladimir Alexei Morozov
- Center for HIV and Retrovirology, Department of Infectious Diseases, Robert Koch Institute, Berlin 13353, Germany
| | - Sylvie Lagaye
- Department of Immunology, Institut Pasteur, INSERM U1223, Paris 75015, France
| |
Collapse
|
53
|
Beilstein F, Lemasson M, Pène V, Rainteau D, Demignot S, Rosenberg AR. Lysophosphatidylcholine acyltransferase 1 is downregulated by hepatitis C virus: impact on production of lipo-viro-particles. Gut 2017; 66:2160-2169. [PMID: 27582510 DOI: 10.1136/gutjnl-2016-311508] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/21/2016] [Revised: 08/02/2016] [Accepted: 08/08/2016] [Indexed: 12/18/2022]
Abstract
OBJECTIVE HCV is intimately linked with the liver lipid metabolism, devoted to the efflux of triacylglycerols stored in lipid droplets (LDs) in the form of triacylglycerol-rich very-low-density lipoproteins (VLDLs): (i) the most infectious HCV particles are those of lowest density due to association with triacylglycerol-rich lipoproteins and (ii) HCV-infected patients frequently develop hepatic steatosis (increased triacylglycerol storage). The recent identification of lysophosphatidylcholine acyltransferase 1 (LPCAT1) as an LD phospholipid-remodelling enzyme prompted us to investigate its role in liver lipid metabolism and HCV infectious cycle. DESIGN Huh-7.5.1 cells and primary human hepatocytes (PHHs) were infected with JFH1-HCV. LPCAT1 depletion was achieved by RNA interference. Cells were monitored for LPCAT1 expression, lipid metabolism and HCV production and infectivity. The density of viral particles was assessed by isopycnic ultracentrifugation. RESULTS Upon HCV infection, both Huh-7.5.1 cells and PHH had decreased levels of LPCAT1 transcript and protein, consistent with transcriptional downregulation. LPCAT1 depletion in either naive or infected Huh-7.5.1 cells resulted in altered lipid metabolism characterised by LD remodelling, increased triacylglycerol storage and increased secretion of VLDL. In infected Huh-7.5.1 cells or PHH, LPCAT1 depletion increased production of the viral particles of lowest density and highest infectivity. CONCLUSIONS We have identified LPCAT1 as a modulator of liver lipid metabolism downregulated by HCV, which appears as a viral strategy to increase the triacylglycerol content and hence infectivity of viral particles. Targeting this metabolic pathway may represent an attractive therapeutic approach to reduce both the viral titre and hepatic steatosis.
Collapse
Affiliation(s)
- Frauke Beilstein
- Sorbonne Universités, UPMC Univ. Paris 06, Inserm, Université Paris Descartes, Sorbonne Paris Cité, UMR_S 1138, Centre de Recherche des Cordeliers, Paris, France.,EPHE, Ecole Pratique des Hautes Etudes, PSL Research University, Laboratoire de Pharmacologie Cellulaire et Moléculaire, Paris, France
| | - Matthieu Lemasson
- Université Paris Descartes, EA 4474 «Hepatitis C Virology», Paris, France
| | - Véronique Pène
- Université Paris Descartes, EA 4474 «Hepatitis C Virology», Paris, France
| | | | - Sylvie Demignot
- Sorbonne Universités, UPMC Univ. Paris 06, Inserm, Université Paris Descartes, Sorbonne Paris Cité, UMR_S 1138, Centre de Recherche des Cordeliers, Paris, France.,EPHE, Ecole Pratique des Hautes Etudes, PSL Research University, Laboratoire de Pharmacologie Cellulaire et Moléculaire, Paris, France
| | - Arielle R Rosenberg
- Université Paris Descartes, EA 4474 «Hepatitis C Virology», Paris, France.,AP-HP, Groupe Hospitalier Cochin, Service de Virologie, Paris, France
| |
Collapse
|
54
|
Gauthiez E, Habfast-Robertson I, Rüeger S, Kutalik Z, Aubert V, Berg T, Cerny A, Gorgievski M, George J, Heim MH, Malinverni R, Moradpour D, Müllhaupt B, Negro F, Semela D, Semmo N, Villard J, Bibert S, Bochud PY. A systematic review and meta-analysis of HCV clearance. Liver Int 2017; 37:1431-1445. [PMID: 28261910 DOI: 10.1111/liv.13401] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/13/2017] [Accepted: 02/20/2017] [Indexed: 02/13/2023]
Abstract
While hepatitis C exemplifies the role of host genetics in infectious diseases outcomes, there is no comprehensive overview of polymorphisms influencing spontaneous and/or treatment-induced hepatitis C virus clearance. We performed a systematic review and meta-analysis of host polymorphisms associated with these phenotypes. Literature search was conducted using combinations of keywords in three databases. Studies were reviewed and relevant data systematically extracted for subsequent meta-analyses. Polymorphisms from candidate gene studies were tested in two cohorts of HCV-infected patients with available genomic data. The literature search yielded 8'294 citations, among which 262 studies were selected. In the meta-analysis of 27 HLA studies, the most significant associations with spontaneous hepatitis C virus clearance included DQB1*02, DQB1*03, DRB1*04 and DRB1*11. In the meta-analysis of 16 studies of KIR genes and their HLA-ligands, KIR2DS3 was associated with both spontaneous and treatment-induced clearance, and the HLA-C2 ligand with failure to spontaneously clear the virus. In a pooled analysis of 105 candidate genes and two genome-wide association studies, we observed associations of single nucleotide polymorphisms from nine genes (EIF2AK2, IFNAR2, ITPA, MBL2, MX1, OASL, SPP1, TGFB1, TNK2) with response to interferon-based therapy. Meta-analysis of 141 studies confirmed the association of IFNL3/4 polymorphisms with spontaneous and treatment-induced hepatitis C virus clearance, even in previously underpowered groups, such as hepatitis C virus genotypes 2/3-infected patients. This study may contribute to a better understanding of hepatitis C virus immunopathogenesis and highlights the complex role of host genetics in hepatitis C virus clearance.
Collapse
Affiliation(s)
- Emeline Gauthiez
- Infectious Diseases Service, Department of Medicine, University Hospital and University of Lausanne, Lausanne, Switzerland
| | | | - Sina Rüeger
- Institute for Social and Preventive Medicine and Department of Medical Genetics, University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Zoltan Kutalik
- Institute for Social and Preventive Medicine and Department of Medical Genetics, University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Vincent Aubert
- Division of Immunology and Allergy, Centre Hospitalier Universitaire Vaudois, Lausanne, Switzerland
| | - Thomas Berg
- Department of Hepatology, Universitätsklinikum Leipzig, Leipzig, Germany
| | | | - Meri Gorgievski
- Division of Infectious Diseases, University Hospital of Bern, Bern, Switzerland
| | - Jacob George
- Storr Liver Centre, Westmead Millennium Institute, University of Sydney and Westmead Hospital, Sydney, NSW, Australia
| | - Markus H Heim
- Department of Gastroenterology, University Hospital, Basel, Switzerland
| | | | - Darius Moradpour
- Division of Gastroenterology and Hepatology, University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Beat Müllhaupt
- Division of Gastroenterology and Hepatology, University Hospital of Zurich, Zurich, Switzerland
| | - Francesco Negro
- Division of Clinical Pathology and Division of Gastroenterology and Hepatology, University Hospitals, Geneva, Switzerland
| | - David Semela
- Division of Gastroenterology, Canton Hospital, St. Gall, Switzerland
| | - Nasser Semmo
- Hepatology, Department of clinical Research, University of Bern, Bern, Switzerland
| | - Jean Villard
- Transplantation Immunology Unit, Division of Immunology and Allergy, Department Medicine and Laboratory Medicine, Geneva University Hospital and Medical School, Geneva, Switzerland
| | - Stéphanie Bibert
- Infectious Diseases Service, Department of Medicine, University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Pierre-Yves Bochud
- Infectious Diseases Service, Department of Medicine, University Hospital and University of Lausanne, Lausanne, Switzerland
| | | |
Collapse
|
55
|
Ellwanger JH, Kaminski VDL, Valverde-Villegas JM, Simon D, Lunge VR, Chies JAB. Immunogenetic studies of the hepatitis C virus infection in an era of pan-genotype antiviral therapies - Effective treatment is coming. INFECTION GENETICS AND EVOLUTION 2017; 66:376-391. [PMID: 28811194 DOI: 10.1016/j.meegid.2017.08.011] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/11/2017] [Revised: 08/10/2017] [Accepted: 08/11/2017] [Indexed: 02/08/2023]
Abstract
What are the factors that influence human hepatitis C virus (HCV) infection, hepatitis status establishment, and disease progression? Firstly, one has to consider the genetic background of the host and HCV genotypes. The immunogenetic host profile will reflect how each infected individual deals with infection. Secondly, there are environmental factors that drive susceptibility or resistance to certain viral strains. These will dictate (I) the susceptibility to infection; (II) whether or not an infected person will promote viral clearance; (III) the immune response and the response profile to therapy; and (IV) whether and how long it would take to the development of HCV-associated diseases, as well as their severity. Looking at this scenario, this review addresses clinical aspects of HCV infection, following by an update of molecular and cellular features of the immune response against the virus. The evasion mechanisms used by HCV are presented, considering the potential role of exosomes in infection. Genetic factors influencing HCV infection and pathogenesis are the main topics of the article. Shortly, HLAs, MBLs, TLRs, ILs, and IFNLs genes have relevant roles in the susceptibility to HCV infection. In addition, ILs, IFNLs, as well as TLRs genes are important modulators of HCV-associated diseases. The viral aspects that influence HCV infection are presented, followed by a discussion about evolutionary aspects of host and HCV interaction. HCV and HIV infections are close related. Thus, we also present a discussion about HIV/HCV co-infection, focusing on cellular and molecular aspects of this interaction. Pharmacogenetics and treatment of HCV infection are the last topics of this review. The understanding of how the host genetics interacts with viral and environmental factors is crucial for the development of new strategies to prevent HCV infection, even in an era of potential development of pan-genotypic antivirals.
Collapse
Affiliation(s)
- Joel Henrique Ellwanger
- Laboratório de Imunobiologia e Imunogenética, Departamento de Genética, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil
| | - Valéria de Lima Kaminski
- Laboratório de Imunobiologia e Imunogenética, Departamento de Genética, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil
| | - Jacqueline María Valverde-Villegas
- Laboratório de Imunobiologia e Imunogenética, Departamento de Genética, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil
| | - Daniel Simon
- Laboratório de Genética Molecular Humana, Universidade Luterana do Brasil (ULBRA), Canoas, Brazil
| | - Vagner Ricardo Lunge
- Laboratório de Diagnóstico Molecular, Universidade Luterana do Brasil (ULBRA), Canoas, Brazil
| | - José Artur Bogo Chies
- Laboratório de Imunobiologia e Imunogenética, Departamento de Genética, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil.
| |
Collapse
|
56
|
Abstract
Hepatitis C virus (HCV) infection is a prevalent condition associated with numerous extrahepatic manifestations. Epidemiologic studies have found that HCV is associated with increased cardiovascular morbidity and mortality, in particular with carotid atherosclerosis, cerebrovascular events, and coronary heart disease. The mechanisms involved encompass a chronic systemic inflammatory state, insulin resistance, and a potential, direct infection of the vascular endothelium. Sustained virologic response with interferon-based regimens is associated with reduced cardiovascular events, although this must be validated with newer direct-acting antivirals. This clear association between HCV and cardiovascular events may have significant economical and public health implications.
Collapse
Affiliation(s)
- Nicolas Goossens
- Division of Gastroenterology and Hepatology, Geneva University Hospitals, 4 Rue Gabrielle-Perret-Gentil, Geneva 4 1211, Switzerland
| | - Francesco Negro
- Division of Gastroenterology and Hepatology, Geneva University Hospitals, 4 Rue Gabrielle-Perret-Gentil, Geneva 4 1211, Switzerland; Division of Clinical Pathology, Geneva University Hospitals, 4 Rue Gabrielle-Perret-Gentil, Geneva 4 1211, Switzerland.
| |
Collapse
|
57
|
Abstract
PURPOSE OF REVIEW Direct-acting antiviral agents (DAAs) have markedly improved the prognosis of hepatitis C virus (HCV)-genotype 3 (GT3), a highly prevalent infection worldwide. However, in patients with hepatic fibrosis, cirrhosis, or hepatocellular carcinoma (HCC), GT3 infection presents a treatment challenge compared with other genotypes. The dependence of the HCV life cycle on host lipid metabolism suggests the possible utility of targeting host cellular factors for combination anti-HCV therapy. We discuss current and emergent DAA regimens for HCV-GT3 treatment. We then summarize recent research findings on the reliance of HCV entry, replication, and virion assembly on host lipid metabolism. RECENT FINDINGS Current HCV treatment guidelines recommend the use of daclatasvir plus sofosbuvir (DCV/SOF) or sofosbuvir plus velpatasvir (SOF/VEL) for the management of GT3 based upon clinical efficacy [≥88% overall sustained virological response (SVR)] and tolerability. Potential future DAA options, such as SOF/VEL co-formulated with GS-9857, also look promising in treating cirrhotic GT3 patients. However, HCV resistance to DAAs will likely continue to impact the therapeutic efficacy of interferon-free treatment regimens. Disruption of HCV entry by targeting required host cellular receptors shows potential in minimizing HCV resistance and broadening therapeutic options for certain subpopulations of GT3 patients. The use of cholesterol biosynthesis and transport inhibitors may also improve health outcomes for GT3 patients when used synergistically with DAAs. Due to the morbidity and mortality associated with HCV-GT3 infection compared to other genotypes, efforts should be made to address current limitations in the therapeutic prevention and management of HCV-GT3 infection.
Collapse
|
58
|
Yang CHT, Yoo ER, Ahmed A. The Role of Direct-acting Antivirals in the Treatment of Children with Chronic Hepatitis C. J Clin Transl Hepatol 2017; 5:59-66. [PMID: 28507928 PMCID: PMC5411358 DOI: 10.14218/jcth.2016.00053] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/24/2016] [Revised: 12/13/2016] [Accepted: 01/05/2017] [Indexed: 12/13/2022] Open
Abstract
In the United States, chronic infection with the hepatitis C virus (HCV) affects an estimated 0.1-2% of the pediatric population, who are consequently at risk for major complications, including cirrhosis, hepatocellular carcinoma, and death. The current standard of treatment for chronic hepatitis C (CHC) in children is pegylated-interferon-alpha (PEG-IFN) in combination with ribavirin. PEG-IFN/ribavirin therapy is approved for children ages 3 and older; however, it is often held from use until adulthood because of its extensive list of potential side effects and high likelihood of causing adverse symptoms. While CHC is usually indolent in children and adolescents, immediately treating and curbing the spread of HCV before adulthood is important, as there can be transmission to other individuals via sexual activity and infected females can later vertically transmit the infection during pregnancy, the latter representing the most common means of transmission for children in the United States. The recent development of direct-acting antivirals has shown promising results in clinical trials for use in children and has dramatically increased the rates of sustained virological response in adults while improving side effect profiles as compared to interferon-based treatments. Given the usually indolent course of CHC in children, significant side effects of the currently-approved PEG-IFN/ribavirin therapy, and likely availability of all-oral interferon-free regimens for children within a few years, deferring treatment in clinically-stable children with CHC in anticipation of upcoming superior treatment modalities may be justified.
Collapse
Affiliation(s)
- Christine Hong Ting Yang
- Division of Pediatric Gastroenterology, Stanford University School of Medicine, Stanford, CA, USA
| | - Eric R. Yoo
- Department of Medicine, University of Illinois College of Medicine, Chicago, IL, USA
| | - Aijaz Ahmed
- Division of Gastroenterology and Hepatology, Stanford University School of Medicine, Stanford, CA, USA
- *Correspondence to: Aijaz Ahmed, Division of Gastroenterology and Hepatology, Stanford University School of Medicine, 750 Welch Road, Suite #210, Stanford, CA 94304, USA. Tel: +1-650-498-5691, Fax: +1-650-498-5692, E-mail:
| |
Collapse
|
59
|
Cornberg M, Petersen J, Schober A, Mauss S, Böker KHW, Link R, Günther R, Serfert Y, Pfeiffer-Vornkahl H, Manns MP, Sarrazin C, Hüppe D, Berg T, Niederau C. Real-world use, effectiveness and safety of anti-viral treatment in chronic hepatitis C genotype 3 infection. Aliment Pharmacol Ther 2017; 45:688-700. [PMID: 28078723 DOI: 10.1111/apt.13925] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2016] [Revised: 11/27/2016] [Accepted: 12/09/2016] [Indexed: 12/12/2022]
Abstract
BACKGROUND Treatment of chronic hepatitis C genotype 3 (GT3) is more challenging compared with other genotypes. Since 2014, several new treatment regimens have been approved but sometimes based on limited data. AIM To validate the use, effectiveness and safety of anti-viral treatment in chronic hepatitis C genotype 3 infection under real-word conditions. METHODS The German Hepatitis C-Registry is a large national non-interventional real-world study for patients with chronic hepatitis C. A total of 1322 GT3 patients were enrolled (211 untreated and 1111 treated patients). RESULTS Between February 2014 and September 2015, five different treatment strategies have been used (PegIFN+RBV, PegIFN+RBV+SOF, SOF+RBV, DCV+SOF±RBV, LDV/SOF±RBV). Treatment uptake and use of treatment concepts changed markedly and rapidly during the study influenced by new approvals, guideline recommendations, and label updates. PegIFN-based therapies constantly declined while DCV-based therapies increased with one interruption after the approval of LDV/SOF, which was frequently used until new guidelines recommended not using this combination for GT3. Per-protocol SVR ranged from 80.9% in the PegIFN+RBV group to 96.1% in PegIFN+RBV+SOF treated patients. Treatment-experienced patients with cirrhosis showed a suboptimal SVR of 68% for SOF+RBV but a high SVR of 90-95% for DCV+SOF±RBV. The safety analysis showed more adverse events and a stronger decline of haemoglobin for RBV containing regimens. CONCLUSIONS Real-world data can validate the effectiveness and safety for treatment regimens that had previously been approved with limited data, in particular for specific subgroups of patients. The present study demonstrates how rapid new scientific data, new treatment guidelines, new drug approvals and label changes are implemented into routine clinical practice today.
Collapse
Affiliation(s)
- M Cornberg
- Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, Hannover, Germany and German Center for Infection Research (DZIF), Partner-site Hannover-Braunschweig, Germany
| | - J Petersen
- IFI-Institute for Interdisciplinary Medicine, Hamburg, Germany
| | - A Schober
- Hepatologische Praxis, Göttingen, Germany
| | - S Mauss
- Center for HIV and Hepatogastroenterology, Duesseldorf, Germany
| | | | - R Link
- MVZ Offenburg, Offenburg, Germany
| | - R Günther
- Universitätsklinikum Schleswig-Holstein (UKSH), Campus Kiel, Kiel, Germany
| | - Y Serfert
- Leberstiftungs-GmbH Deutschland, Hannover, Germany
| | | | - M P Manns
- Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, Hannover, Germany and German Center for Infection Research (DZIF), Partner-site Hannover-Braunschweig, Germany
| | - C Sarrazin
- St.-Josefs-Hospital Wiesbaden and University Hospital Frankfurt, Frankfurt, Germany
| | - D Hüppe
- Hepatologische Schwerpunktpraxis Herne, Herne, Germany
| | - T Berg
- University Hospital Leipzig, Leipzig, Germany
| | - C Niederau
- St. Josef-Hospital Katholisches Klinikum Oberhausen, Oberhausen, Germany
| |
Collapse
|
60
|
McMahon BJ, Bruden D, Townshend-Bulson L, Simons B, Spradling P, Livingston S, Gove J, Hewitt A, Plotnik J, Homan C, Espera H, Negus S, Snowball M, Barbour Y, Bruce M, Gounder P. Infection With Hepatitis C Virus Genotype 3 Is an Independent Risk Factor for End-Stage Liver Disease, Hepatocellular Carcinoma, and Liver-Related Death. Clin Gastroenterol Hepatol 2017; 15:431-437.e2. [PMID: 27765729 PMCID: PMC5316337 DOI: 10.1016/j.cgh.2016.10.012] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/28/2016] [Revised: 10/07/2016] [Accepted: 10/09/2016] [Indexed: 02/07/2023]
Abstract
BACKGROUND & AIMS Few studies have examined factors associated with disease progression in hepatitis C virus (HCV) infection. We examined the association of 11 risk factors with adverse outcomes in a population-based prospective cohort observational study of Alaska Native/American Indian persons with chronic HCV infection. METHODS We collected data from a population-based cohort study of liver-related adverse outcomes of infection in American Indian/Alaska Native persons with chronic HCV living in Alaska, recruited from 1995 through 2012. We calculated adjusted hazard ratios (aHR) and 95% confidence intervals (CIs) for end-stage liver disease (ESLD; presence of ascites, esophageal varices, hepatic encephalopathy, or coagulopathy), hepatocellular carcinoma (HCC), and liver-related death using a Cox proportional hazards model. RESULTS We enrolled 1080 participants followed up for 11,171 person-years (mean, 10.3 person-years); 66%, 19%, and 14% were infected with HCV genotypes 1, 2, and 3, respectively. On multivariate analysis, persons infected with HCV genotype 3 had a significantly increased risk of developing all 3 adverse outcomes. Their aHR for ESLD was 2.1 (95% CI, 1.5-3.0), their aHR for HCC was 3.1 (95% CI, 1.4-6.6), and their aHR for liver-related death was 2.4 (95% CI, 1.5-4.0) compared with genotype 1. Heavy alcohol use was an age-adjusted risk factor for ESLD (aHR, 2.2; 95% CI, 1.6-3.2), and liver-related death (aHR, 2.9; 95% CI, 1.8-4.6). Obesity was a risk factor for ESLD (aHR, 1.4; 95% CI, 1.0-1.9), and diabetes was a risk factor for ESLD (aHR, 1.5; 95% CI, 1.1-2.2). Male sex was a risk factor for HCC (aHR, 3.6; 95% CI, 1.6-8.2). CONCLUSIONS In a population-based cohort study of American Indian/Alaska Native persons with chronic HCV infection, we found those infected with HCV genotype 3 to be at high risk for ESLD, HCC, and liver-related death.
Collapse
Affiliation(s)
- Brian J McMahon
- Liver Disease and Hepatitis Program, Alaska Native Tribal Health Consortium, Anchorage, Alaska; Arctic Investigations Program, Division of Preparedness and Emerging Infections, National Center for Emerging and Zoonotic Infectious Diseases, Centers for Disease Control, Anchorage, Alaska.
| | - Dana Bruden
- Arctic Investigations Program, Division of Preparedness and Emerging Infections, National Center for Emerging and Zoonotic Infectious Diseases, Centers for Disease Control, Anchorage, Alaska
| | - Lisa Townshend-Bulson
- Liver Disease and Hepatitis Program, Alaska Native Tribal Health Consortium, Anchorage, Alaska
| | - Brenna Simons
- Liver Disease and Hepatitis Program, Alaska Native Tribal Health Consortium, Anchorage, Alaska
| | - Phillip Spradling
- Division of Viral Hepatitis, Centers for Human Immunodeficiency Virus, Tuberculosis Prevention, Sexually Transmitted Diseases, and Viral Hepatitis, Centers for Disease Control, Atlanta, Georgia
| | - Stephen Livingston
- Liver Disease and Hepatitis Program, Alaska Native Tribal Health Consortium, Anchorage, Alaska
| | - James Gove
- Liver Disease and Hepatitis Program, Alaska Native Tribal Health Consortium, Anchorage, Alaska
| | - Annette Hewitt
- Liver Disease and Hepatitis Program, Alaska Native Tribal Health Consortium, Anchorage, Alaska
| | - Julia Plotnik
- Liver Disease and Hepatitis Program, Alaska Native Tribal Health Consortium, Anchorage, Alaska
| | - Chriss Homan
- Liver Disease and Hepatitis Program, Alaska Native Tribal Health Consortium, Anchorage, Alaska
| | - Hannah Espera
- Liver Disease and Hepatitis Program, Alaska Native Tribal Health Consortium, Anchorage, Alaska
| | - Susan Negus
- Liver Disease and Hepatitis Program, Alaska Native Tribal Health Consortium, Anchorage, Alaska
| | - Mary Snowball
- Liver Disease and Hepatitis Program, Alaska Native Tribal Health Consortium, Anchorage, Alaska
| | - Youssef Barbour
- Liver Disease and Hepatitis Program, Alaska Native Tribal Health Consortium, Anchorage, Alaska
| | - Michael Bruce
- Arctic Investigations Program, Division of Preparedness and Emerging Infections, National Center for Emerging and Zoonotic Infectious Diseases, Centers for Disease Control, Anchorage, Alaska
| | - Prabhu Gounder
- Arctic Investigations Program, Division of Preparedness and Emerging Infections, National Center for Emerging and Zoonotic Infectious Diseases, Centers for Disease Control, Anchorage, Alaska
| |
Collapse
|
61
|
Pineda JA, Morano-Amado LE, Granados R, Macías J, Téllez F, García-Deltoro M, Ríos MJ, Collado A, Delgado-Fernández M, Suárez-Santamaría M, Serrano M, Miralles-Álvarez C, Neukam K. Week 4 response predicts sustained virological response to all-oral direct-acting antiviral-based therapy in cirrhotic patients with hepatitis C virus genotype 3 infection. Clin Microbiol Infect 2017; 23:409.e5-409.e8. [PMID: 28137633 DOI: 10.1016/j.cmi.2016.12.034] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2016] [Revised: 12/29/2016] [Accepted: 12/31/2016] [Indexed: 10/20/2022]
Abstract
OBJECTIVE The aim of this study was to determine the predictive capacity of response at treatment week (TW) 4 for the achievement of sustained virological response 12 weeks after the scheduled end of therapy date (SVR12) to treatment against hepatitis C virus (HCV) genotype 3 (GT3) infection with all-oral direct-acting antiviral (DAA) -based regimens. PATIENTS AND METHODS From a prospective multicohort study, HCV GT3-infected patients who completed a course of currently recommended DAA-based therapy at 33 Spanish hospitals and who had reached the SVR12 evaluation time-point were selected. TW4 HCV-RNA levels were categorized as target-not-detected (TND), below the lower limit of quantification (LLOQTD) and ≥LLOQ. RESULTS A total of 123 patients were included, 86 (70%) received sofosbuvir/ daclatasvir±ribavirin, 27 (22%) received sofosbuvir/ ledipasvir/ ribavirin and 10 (8.1%) received sofosbuvir/ ribavirin, respectively. In all, 114 (92.7%) of the 123 patients presented SVR12 in an on-treatment approach, but nine (7.3%) patients relapsed, all of them had presented cirrhosis at baseline. In those who achieved TND, LLOQTD and ≥LLOQ, SVR12 was observed in 81/83 (98%; 95% CI 91.5%-99.7%), 24/28 (85.7%; 95% CI 67.3%-96%) and 9/12 (75%; 95% CI 42.8%-94.5%), respectively; p(linear association) 0.001. Corresponding numbers for subjects with cirrhosis were: 52/54 (96.3%; 95% CI 87.3%-95.5%), 14/18 (77.8%; 95% CI 52.4%-93.6%) and 7/10 (70%; 95% CI 34.8%-93.3%); p 0.004. CONCLUSIONS TW4-response indicates the probability of achieving SVR12 to currently used DAA-based therapy in HCV genotype 3-infected individuals with cirrhosis. This finding may be useful to tailor treatment strategy in this setting.
Collapse
Affiliation(s)
- J A Pineda
- Unit of Infectious Diseases and Microbiology, Hospital Universitario de Valme, Seville, Spain
| | - L E Morano-Amado
- Unit of Infectious Pathology, Hospital Universitario Alvaro Cunqueiro, Vigo, Spain
| | - R Granados
- Unit of Infectious Diseases, Hospital Universitario de Gran Canaria Dr Negrín, Las Palmas de Gran Canaria, Spain
| | - J Macías
- Unit of Infectious Diseases and Microbiology, Hospital Universitario de Valme, Seville, Spain; Instituto de Biomedicina de Sevilla (IBiS), Seville, Spain
| | - F Téllez
- Unit of Infectious Diseases and Microbiology, Hospital La Línea, AGS Campo de Gibraltar, La Linea de la Concepcion, Spain
| | - M García-Deltoro
- Unit of Infectious Diseases, Consorcio Hospital General Universitario de Valencia, Valencia, Spain
| | - M J Ríos
- Unit of Infectious Diseases, Hospital Virgen Macarena, Seville, Spain
| | - A Collado
- Unit of Infectious Diseases, Hospital Universitario Torrecárdenas, Almeria, Spain
| | | | - M Suárez-Santamaría
- Fundación Biomédica del Complejo Hospitalario Universitario de Vigo (CHUVI), Vigo, Spain
| | - M Serrano
- Unit of Infectious Diseases, Hospital Universitario de Gran Canaria Dr Negrín, Las Palmas de Gran Canaria, Spain
| | - C Miralles-Álvarez
- Unit of Infectious Pathology, Hospital Universitario Alvaro Cunqueiro, Vigo, Spain
| | - K Neukam
- Unit of Infectious Diseases and Microbiology, Hospital Universitario de Valme, Seville, Spain; Instituto de Biomedicina de Sevilla (IBiS), Seville, Spain.
| | | | | |
Collapse
|
62
|
Federico A, Dallio M, Loguercio C. Silymarin/Silybin and Chronic Liver Disease: A Marriage of Many Years. Molecules 2017; 22:molecules22020191. [PMID: 28125040 PMCID: PMC6155865 DOI: 10.3390/molecules22020191] [Citation(s) in RCA: 280] [Impact Index Per Article: 35.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2016] [Revised: 01/13/2017] [Accepted: 01/18/2017] [Indexed: 02/06/2023] Open
Abstract
Silymarin is the extract of Silybum marianum, or milk thistle, and its major active compound is silybin, which has a remarkable biological effect. It is used in different liver disorders, particularly chronic liver diseases, cirrhosis and hepatocellular carcinoma, because of its antioxidant, anti-inflammatory and antifibrotic power. Indeed, the anti-oxidant and anti-inflammatory effect of silymarin is oriented towards the reduction of virus-related liver damages through inflammatory cascade softening and immune system modulation. It also has a direct antiviral effect associated with its intravenous administration in hepatitis C virus infection. With respect to alcohol abuse, silymarin is able to increase cellular vitality and to reduce both lipid peroxidation and cellular necrosis. Furthermore, silymarin/silybin use has important biological effects in non-alcoholic fatty liver disease. These substances antagonize the progression of non-alcoholic fatty liver disease, by intervening in various therapeutic targets: oxidative stress, insulin resistance, liver fat accumulation and mitochondrial dysfunction. Silymarin is also used in liver cirrhosis and hepatocellular carcinoma that represent common end stages of different hepatopathies by modulating different molecular patterns. Therefore, the aim of this review is to examine scientific studies concerning the effects derived from silymarin/silybin use in chronic liver diseases, cirrhosis and hepatocellular carcinoma.
Collapse
Affiliation(s)
- Alessandro Federico
- Department of Clinical and Experimental Medicine, Second University of Naples, 80131 Naples, Italy.
| | - Marcello Dallio
- Department of Clinical and Experimental Medicine, Second University of Naples, 80131 Naples, Italy.
| | - Carmelina Loguercio
- Department of Clinical and Experimental Medicine, Second University of Naples, 80131 Naples, Italy.
| |
Collapse
|
63
|
Ramirez S, Mikkelsen LS, Gottwein JM, Bukh J. Robust HCV Genotype 3a Infectious Cell Culture System Permits Identification of Escape Variants With Resistance to Sofosbuvir. Gastroenterology 2016; 151:973-985.e2. [PMID: 27453546 DOI: 10.1053/j.gastro.2016.07.013] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2016] [Revised: 07/01/2016] [Accepted: 07/12/2016] [Indexed: 01/15/2023]
Abstract
BACKGROUND & AIMS Direct-acting antivirals (DAAs) effectively eradicate chronic hepatitis C virus (HCV) infection, although HCV genotype 3a is less responsive to these drugs. We aimed to develop genotype 3a infectious cultures and study the effects of inhibitors of NS5A and NS5B and resistance to sofosbuvir-the only nucleotide analog approved for treatment of chronic HCV infection. METHODS The developed HCV genotype 3a full-length genome (DBN3a), with a strain-DBN coding sequence, modified NS5B consensus sequence, pS52 untranslated regions, and coding mutations from a culture-efficient JFH1-based core-NS5A (DBN) recombinant, was transfected into Huh7.5 cells. The efficacy of selected DAAs was determined in dose-response assays, in which the number of HCV-infected cells was measured after incubation with different concentrations of the specific DAA. Long-term culture of infected Huh7.5 cells with increasing concentrations of sofosbuvir was used to promote selection of HCV-resistant variants. RESULTS We engineered a DBN3a variant with 17 substitutions (DBN3acc) that had replication and propagation kinetics in Huh7.5 cells comparable with prototype J6/JFH1. The adaptive mutations also produced culture-efficient DBN-based recombinants with NS5B from HCV genotype 3a strains S52 and DH11. Compared with genotype 1a, genotype 3a was less sensitive to daclatasvir, ledipasvir, and elbasvir, but equally sensitive to ombitasvir, velpatasvir, beclabuvir, dasabuvir, MK-3682, and sofosbuvir. Exposure of Huh7.5 cells infected with DBN3a to sofosbuvir led to identification of an escape variant with substitutions in NS5B, including the resistance-associated substitution S282T. This variant showed increased infectivity of Huh7.5 cells, compared with DBN3a, and was genetically stable in cell cultures without sofosbuvir. Sofosbuvir, MK-3682, dasabuvir, or combinations of sofosbuvir and ledipasvir or sofosbuvir and velpatasvir had decreased efficacy against infection with the DBN3a sofosbuvir escape variant. CONCLUSIONS We developed a system for highly efficient culture of HCV genotype 3a. Genotype 1a has a high genetic barrier to resistance for sofosbuvir, whereas resistance to this DAA can be induced in genotype 3a. We therefore isolated HCV genotype 3a variants with reduced sensitivity to sofosbuvir, with increased fitness and with cross-resistance to other NS5B inhibitors. These findings indicate that sofosbuvir escape variants could compromise the effectiveness of nucleotide analogs against HCV. GenBank accession numbers: KX280712-KX280716.
Collapse
Affiliation(s)
- Santseharay Ramirez
- Copenhagen Hepatitis C Program (CO-HEP), Department of Infectious Diseases and Clinical Research Centre, Hvidovre Hospital and Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Lotte S Mikkelsen
- Copenhagen Hepatitis C Program (CO-HEP), Department of Infectious Diseases and Clinical Research Centre, Hvidovre Hospital and Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Judith M Gottwein
- Copenhagen Hepatitis C Program (CO-HEP), Department of Infectious Diseases and Clinical Research Centre, Hvidovre Hospital and Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Jens Bukh
- Copenhagen Hepatitis C Program (CO-HEP), Department of Infectious Diseases and Clinical Research Centre, Hvidovre Hospital and Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.
| |
Collapse
|
64
|
Aslam R, Raza SM, Naeemi H, Mubarak B, Afzal N, Khaliq S. SOCS3 mRNA expression and polymorphisms as pretreatment predictor of response to HCV genotype 3a IFN-based treatment. SPRINGERPLUS 2016; 5:1826. [PMID: 27818864 PMCID: PMC5074986 DOI: 10.1186/s40064-016-3506-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/22/2016] [Accepted: 10/10/2016] [Indexed: 01/10/2023]
Abstract
Aim Suppressor of Cytokine Signaling 3 (SOCS3) gene belongs to SOCS family as one of the negative regulators of cytokine signaling and IFN response that function via the JAK-STAT pathway in antiviral response. SOCS3 expression and genetic polymorphism influences the pathogenesis and outcome of antiviral treatment in hepatitis C virus (HCV) infected patients. This study was designed for analysis of SOCS3 gene expression and polymorphism in Pakistani HCV patients. Methods This descriptive study was conducted on 250 diagnosed HCV genotype 3a infected subjects. The study population was divided into two major groups on the basis of therapeutic response i.e. sustained virological response (SVR) and non-responders/relapsers (NR). SOCS3 gene mRNA expression analysis was done by using Real time PCR technique, whereas ARMS PCR technique was used for analysis of SOCS3 gene polymorphisms i.e. 8464 A/C (rs12952093), −4874 A/G (rs4969170) and −1383 A/G, (rs4969168). Results Gene expression analysis of SOCS3 showed that there was statistically significant increase of 2.275-fold and 3.72-fold in relative gene expression for SVR and NR as compared to normal healthy samples (p < 0.001). The distribution of rs4969168, rs4969170 and rs12952093 genotype frequencies between SVR versus NR group were not statistically significant, only the allelic frequency of rs4969170 was statistically significant (p ≤ 0.0001) with therapeutic response. Conclusion The gene expression analysis of SOCS3 showed a clear difference in mRNA expression of SOCS3 as a possible indicator of therapeutic response rather than polymorphism of SOCS3 gene in our studied population.
Collapse
Affiliation(s)
- Rabia Aslam
- Department of Immunology, University of Health Sciences Lahore, Khayaban-e-Jamia Punjab, Lahore, Pakistan
| | - Syed Mohsin Raza
- Department of Physiology & Cell Biology, University of Health Sciences Lahore, Khayaban-e-Jamia Punjab, Lahore, Pakistan ; Allied Health Sciences, University of Health Sciences Lahore, Lahore, Pakistan
| | - Humeira Naeemi
- Department of Immunology, University of Health Sciences Lahore, Khayaban-e-Jamia Punjab, Lahore, Pakistan
| | - Bushra Mubarak
- Department of Immunology, University of Health Sciences Lahore, Khayaban-e-Jamia Punjab, Lahore, Pakistan
| | - Nadeem Afzal
- Department of Immunology, University of Health Sciences Lahore, Khayaban-e-Jamia Punjab, Lahore, Pakistan
| | - Saba Khaliq
- Department of Physiology & Cell Biology, University of Health Sciences Lahore, Khayaban-e-Jamia Punjab, Lahore, Pakistan ; Department of Immunology, University of Health Sciences Lahore, Khayaban-e-Jamia Punjab, Lahore, Pakistan
| |
Collapse
|
65
|
Maasoumy B, Vermehren J. Diagnostics in hepatitis C: The end of response-guided therapy? J Hepatol 2016; 65:S67-S81. [PMID: 27641989 DOI: 10.1016/j.jhep.2016.07.023] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/25/2016] [Revised: 07/20/2016] [Accepted: 07/21/2016] [Indexed: 02/07/2023]
Abstract
On-treatment hepatitis C virus (HCV) RNA has been used to predict response to interferon (IFN)-based therapy. The concept of response-guided treatment (RGT) was established to determine optimal treatment duration and to early identify patients not responding to futile therapies. RGT helped to improve sustained virologic response (SVR) rates and lower the rates of adverse effects. RGT was of particular importance for telaprevir- and boceprevir-based triple therapies. RGT strategies are dependent on highly sensitive and reproducible HCV RNA quantification. However, different HCV RNA assays are used in routine clinical practice and these differ significantly in their performance characteristics. The development of IFN-free therapies has fundamentally changed the role of on-treatment HCV RNA for SVR prediction. Given the high efficacy and excellent tolerability of IFN-free regimens, the interest in treatment individualization has decreased. However, shorter treatment durations may still be desirable, particularly with respect to the high costs of current IFN-free direct-acting antiviral agents (DAAs). Moreover, some difficult-to-treat patients remain, e.g., those infected with HCV genotype 3 in whom the current standard of care may not always be sufficient to achieve SVR, especially in treatment-experienced patients with cirrhosis. Here, a RGT extension may be feasible. However, current data on the predictive value of on-treatment HCV RNA are limited and have shown conflicting results. As more potent DAAs become available, the role of response prediction may diminish further. Currently, shorter treatment duration is only based on baseline HCV RNA whereas no RGT strategy is recommended for any of the approved DAA regimens available.
Collapse
Affiliation(s)
- Benjamin Maasoumy
- Medizinische Hochschule Hannover, Klinik für Gastroenterologie, Hepatologie und Endokrinologie, Hannover, Germany
| | - Johannes Vermehren
- Universitätsklinikum Frankfurt, Medizinische Klinik 1, Frankfurt am Main, Germany.
| |
Collapse
|
66
|
Bukh J. The history of hepatitis C virus (HCV): Basic research reveals unique features in phylogeny, evolution and the viral life cycle with new perspectives for epidemic control. J Hepatol 2016; 65:S2-S21. [PMID: 27641985 DOI: 10.1016/j.jhep.2016.07.035] [Citation(s) in RCA: 161] [Impact Index Per Article: 17.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/25/2016] [Accepted: 07/29/2016] [Indexed: 12/11/2022]
Abstract
The discovery of hepatitis C virus (HCV) in 1989 permitted basic research to unravel critical components of a complex life cycle for this important human pathogen. HCV is a highly divergent group of viruses classified in 7 major genotypes and a great number of subtypes, and circulating in infected individuals as a continuously evolving quasispecies destined to escape host immune responses and applied antivirals. Despite the inability to culture patient viruses directly in the laboratory, efforts to define the infectious genome of HCV resulted in development of experimental recombinant in vivo and in vitro systems, including replicons and infectious cultures in human hepatoma cell lines. And HCV has become a model virus defining new paradigms in virology, immunology and biology. For example, HCV research discovered that a virus could be completely dependent on microRNA for its replication since microRNA-122 is critical for the HCV life cycle. A number of other host molecules critical for HCV entry and replication have been identified. Thus, basic HCV research revealed important molecules for development of host targeting agents (HTA). The identification and characterization of HCV encoded proteins and their functional units contributed to the development of highly effective direct acting antivirals (DAA) against the NS3 protease, NS5A and the NS5B polymerase. In combination, these inhibitors have since 2014 permitted interferon-free therapy with cure rates above 90% among patients with chronic HCV infection; however, viral resistance represents a challenge. Worldwide control of HCV will most likely require the development of a prophylactic vaccine, and numerous candidates have been pursued. Research characterizing features critical for antibody-based virus neutralization and T cell based virus elimination from infected cells is essential for this effort. If the world community promotes an ambitious approach by applying current DAA broadly, continues to develop alternative viral- and host- targeted antivirals to combat resistant variants, and invests in the development of a vaccine, it would be possible to eradicate HCV. This would prevent about 500 thousand deaths annually. However, given the nature of HCV, the millions of new infections annually, a high chronicity rate, and with over 150 million individuals with chronic infection (which are frequently unidentified), this effort remains a major challenge for basic researchers, clinicians and communities.
Collapse
Affiliation(s)
- Jens Bukh
- Copenhagen Hepatitis C Program (CO-HEP), Department of Infectious Diseases and Clinical Research Centre, Hvidovre Hospital and Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark.
| |
Collapse
|
67
|
Abstract
Non-alcoholic fatty liver disease is associated with hepatocellular carcinoma. In the March 10 issue of Nature, Greten and colleagues report that this metabolic disruption affects tumor surveillance by depleting CD4+ T helper cells through lipotoxic mechanisms associated with NAFLD.
Collapse
|
68
|
Poordad F, Landis CS, Asatryan A, Jackson DF, Ng TI, Fu B, Lin C, Yao B, Kort J. High antiviral activity of NS5A inhibitor ABT-530 with paritaprevir/ritonavir and ribavirin against hepatitis C virus genotype 3 infection. Liver Int 2016; 36:1125-32. [PMID: 26778412 PMCID: PMC5067610 DOI: 10.1111/liv.13067] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2015] [Accepted: 01/07/2016] [Indexed: 12/28/2022]
Abstract
BACKGROUND & AIMS ABT-530 is a next-generation hepatitis C virus (HCV) NS5A inhibitor with potent pangenotypic antiviral activity in vitro. Paritaprevir is an NS3/4A protease inhibitor codosed with ritonavir that displays in vitro activity against HCV genotypes 1-4 and 6. METHODS Efficacy, pharmacokinetics and safety of ABT-530 with paritaprevir/ritonavir and ribavirin were evaluated in this phase 2, open-label, multicentre study in treatment-naïve non-cirrhotic patients with genotype 3 infection. Ten patients, all genotype 3a, received 120 mg ABT-530 and 150/100 mg paritaprevir/ritonavir once daily with ribavirin for 12 weeks. RESULTS Nine (90%) patients achieved a sustained virological response at post-treatment weeks 12 and 24. One patient experienced virological failure at treatment week 6. Sequence analyses for HCV variants in samples from this patient identified A166S in NS3 at baseline and after breakthrough, as well as A30K at baseline and linked S24F+M28K+A30K variants in NS5A after breakthrough. Neither genotype 3 NS3 A166S nor NS5A A30K variant confers any resistance to paritaprevir or ABT-530 respectively. However, genotype 3 NS5A S24F+M28K+A30K-linked variant confers a >5000-fold increase in ABT-530 EC50 relative to that of the wild-type replicon. This patient's ABT-530 exposure was comparable to the cohort, while paritaprevir and ritonavir exposures were the lowest of all patients. No serious or severe adverse events and adverse events leading to early discontinuation were reported. CONCLUSIONS Results from this study show that ABT-530 holds promise as part of a direct-acting antiviral treatment regimen for HCV genotype 3 infection.
Collapse
Affiliation(s)
- Fred Poordad
- Texas Liver InstituteUniversity of Texas Health Science CenterSan AntonioTXUSA
| | | | | | | | | | - Bo Fu
- AbbVie Inc.North ChicagoILUSA
| | | | | | | |
Collapse
|
69
|
Cuypers L, Ceccherini-Silberstein F, Van Laethem K, Li G, Vandamme AM, Rockstroh JK. Impact of HCV genotype on treatment regimens and drug resistance: a snapshot in time. Rev Med Virol 2016; 26:408-434. [PMID: 27401933 DOI: 10.1002/rmv.1895] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2016] [Revised: 06/11/2016] [Accepted: 06/15/2016] [Indexed: 12/11/2022]
Abstract
The introduction of highly potent direct-acting antivirals (DAAs) has revolutionized hepatitis C virus treatment. Nevertheless, viral eradication worldwide remains a challenge also in the era of DAA treatment, because of the high associated costs, high numbers of undiagnosed patients, high re-infection rates in some risk groups and suboptimal drug efficacies associated with host and viral factors as well as advanced stages of liver disease. A correct determination of the HCV genotype allows administration of the most appropriate antiviral regimen. Additionally, HCV genetic sequencing improves our understanding of resistance-associated variants, either naturally occurring before treatment, acquired by transmission at HCV infection, or emerging after virological failure. Because treatment response rates, and the prevalence and development of drug resistance variants differ for each DAA regimen and HCV genotype, this review summarizes treatment opportunities per HCV genotype, and focuses on viral genetic sequencing to guide clinical decision making. Although approval of the first pan-genotypic DAA-only regimen is expected soon, HCV genetic sequencing will remain important because when DAA therapies fail, genotyping and resistance testing to select a new active DAA combination will be essential. Copyright © 2016 John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Lize Cuypers
- KU Leuven - University of Leuven, Department of Microbiology and Immunology, Rega Institute for Medical Research, Clinical and Epidemiological Virology, Leuven, Belgium
| | | | - Kristel Van Laethem
- KU Leuven - University of Leuven, Department of Microbiology and Immunology, Rega Institute for Medical Research, Clinical and Epidemiological Virology, Leuven, Belgium
| | - Guangdi Li
- KU Leuven - University of Leuven, Department of Microbiology and Immunology, Rega Institute for Medical Research, Clinical and Epidemiological Virology, Leuven, Belgium.,Department of Metabolism and Endocrinology, Metabolic Syndrome Research Center, Key Laboratory of Diabetes Immunology, Ministry of Education, National Clinical Research Center for Metabolic Diseases, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Anne-Mieke Vandamme
- KU Leuven - University of Leuven, Department of Microbiology and Immunology, Rega Institute for Medical Research, Clinical and Epidemiological Virology, Leuven, Belgium.,Center for Global Health and Tropical Medicine, Microbiology Unit, Institute for Hygiene and Tropical Medicine, University Nova de Lisboa, Lisbon, Portugal
| | | |
Collapse
|
70
|
Grassi G, Di Caprio G, Fimia GM, Ippolito G, Tripodi M, Alonzi T. Hepatitis C virus relies on lipoproteins for its life cycle. World J Gastroenterol 2016; 22:1953-1965. [PMID: 26877603 PMCID: PMC4726671 DOI: 10.3748/wjg.v22.i6.1953] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/25/2015] [Revised: 10/19/2015] [Accepted: 12/21/2015] [Indexed: 02/06/2023] Open
Abstract
Hepatitis C virus (HCV) infects over 150 million people worldwide. In most cases, HCV infection becomes chronic causing liver disease ranging from fibrosis to cirrhosis and hepatocellular carcinoma. Viral persistence and pathogenesis are due to the ability of HCV to deregulate specific host processes, mainly lipid metabolism and innate immunity. In particular, HCV exploits the lipoprotein machineries for almost all steps of its life cycle. The aim of this review is to summarize current knowledge concerning the interplay between HCV and lipoprotein metabolism. We discuss the role played by members of lipoproteins in HCV entry, replication and virion production.
Collapse
|
71
|
Sukowati CHC, El-Khobar KE, Ie SI, Anfuso B, Muljono DH, Tiribelli C. Significance of hepatitis virus infection in the oncogenic initiation of hepatocellular carcinoma. World J Gastroenterol 2016; 22:1497-1512. [PMID: 26819517 PMCID: PMC4721983 DOI: 10.3748/wjg.v22.i4.1497] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/28/2015] [Revised: 08/06/2015] [Accepted: 10/12/2015] [Indexed: 02/06/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is one of the most common causes of cancer-related death worldwide. Chronic infection of hepatitis B virus (HBV) and/or hepatitis C virus (HCV) is a major risk factor in the development of the HCC, independently from excessive alcohol abuse and metabolic disease. Since the biology of HBV and HCV is different, their oncogenic effect may go through different mechanisms, direct and/or indirect. Viral hepatitis infection is associated with cellular inflammation, oxidative stress, and DNA damage, that may lead to subsequent hepatic injuries such as chronic hepatitis, fibrosis, cirrhosis, and finally HCC. Direct oncogenic properties of these viruses are related with their genotypic characteristics and the ability of viral proteins to interact with host proteins, thus altering the molecular pathways balance of the cells. In addition, the integration of HBV DNA, especially the gene S and X, in a particular site of the host genome can disrupt chromosomal stability and may activate various oncogenic mechanisms, including those in hematopoietic cells. Recently, several studies also had demonstrated that viral hepatitis could trigger the population of hepatic cancer stem cells. This review summarize available pre-clinical and clinical data in literature regarding oncogenic properties of HBV and HCV in the early initiation of HCC.
Collapse
MESH Headings
- Animals
- Carcinoma, Hepatocellular/epidemiology
- Carcinoma, Hepatocellular/genetics
- Carcinoma, Hepatocellular/metabolism
- Carcinoma, Hepatocellular/pathology
- Cell Transformation, Viral
- Gene Expression Regulation, Neoplastic
- Gene Expression Regulation, Viral
- Genotype
- Hepacivirus/genetics
- Hepacivirus/pathogenicity
- Hepatitis B virus/genetics
- Hepatitis B virus/pathogenicity
- Hepatitis B, Chronic/complications
- Hepatitis B, Chronic/virology
- Hepatitis C, Chronic/complications
- Hepatitis C, Chronic/virology
- Host-Pathogen Interactions
- Humans
- Liver Neoplasms/epidemiology
- Liver Neoplasms/genetics
- Liver Neoplasms/metabolism
- Liver Neoplasms/virology
- Neoplastic Stem Cells/metabolism
- Neoplastic Stem Cells/pathology
- Neoplastic Stem Cells/virology
- Oncogenes
- Risk Factors
Collapse
|
72
|
von Delft A, Humphreys IS, Brown A, Pfafferott K, Lucas M, Klenerman P, Lauer GM, Cox AL, Gaudieri S, Barnes E. The broad assessment of HCV genotypes 1 and 3 antigenic targets reveals limited cross-reactivity with implications for vaccine design. Gut 2016; 65:112-23. [PMID: 26092843 PMCID: PMC4717358 DOI: 10.1136/gutjnl-2014-308724] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/27/2014] [Accepted: 04/20/2015] [Indexed: 12/16/2022]
Abstract
OBJECTIVE Developing a vaccine that is cross-reactive between HCV genotypes requires data on T cell antigenic targets that extends beyond genotype-1. We characterised T cell immune responses against HCV genotype-3, the most common infecting genotype in the UK and Asia, and assessed within genotype and between genotype cross-reactivity. DESIGN T cell targets were identified in 140 subjects with either acute, chronic or spontaneously resolved HCV genotype-3 infection using (1) overlapping peptides and (2) putative human leucocyte antigens (HLA)-class-I wild type and variant epitopes through the prior assessment of polymorphic HCV genomic sites associated with host HLA, in IFNγ-ELISpot assays. CD4+/CD8+ T cell subsets were defined and viral variability at T cell targets was determined through population analysis and viral sequencing. T cell cross-reactivity between genotype-1 and genotype-3 variants was assessed. RESULTS In resolved genotype-3 infection, T cells preferentially targeted non-structural proteins at a high magnitude, whereas in chronic disease T cells were absent or skewed to target structural proteins. Additional responses to wild type but not variant HLA predicted peptides were defined. Major sequence viral variability was observed within genotype-3 and between genotypes 1 and 3 HCV at T cell targets in resolved infection and at dominant epitopes, with limited T cell cross-reactivity between viral variants. Overall 41 CD4/CD8+ genotype-3 T cell targets were identified with minimal overlap with those described for HCV genotype-1. CONCLUSIONS HCV T cell specificity is distinct between genotypes with limited T cell cross-reactivity in resolved and chronic disease. Therefore, viral regions targeted in natural HCV infection may not serve as attractive targets for a vaccine that aims to protect against multiple HCV genotypes.
Collapse
Affiliation(s)
| | | | | | | | - Michaela Lucas
- Institute of Immunology and Infectious Diseases, Murdoch University, Perth, Western Australia, Australia,School of Medicine and Pharmacology, Harry Perkins Institute, University of Western Australia, Western Australia, Australia,School of Pathology and Laboratory Medicine, University of Western Australia, Western Australia, Australia
| | | | | | - Andrea L Cox
- John Hopkins University, Baltimore, Maryland, USA
| | - Silvana Gaudieri
- Institute of Immunology and Infectious Diseases, Murdoch University, Perth, Western Australia, Australia,School of Anatomy, Physiology and Human Biology, University of Western Australia, Perth, Western Australia, Australia
| | | |
Collapse
|
73
|
Foster GR, Afdhal N, Roberts SK, Bräu N, Gane EJ, Pianko S, Lawitz E, Thompson A, Shiffman ML, Cooper C, Towner WJ, Conway B, Ruane P, Bourlière M, Asselah T, Berg T, Zeuzem S, Rosenberg W, Agarwal K, Stedman CAM, Mo H, Dvory-Sobol H, Han L, Wang J, McNally J, Osinusi A, Brainard DM, McHutchison JG, Mazzotta F, Tran TT, Gordon SC, Patel K, Reau N, Mangia A, Sulkowski M. Sofosbuvir and Velpatasvir for HCV Genotype 2 and 3 Infection. N Engl J Med 2015; 373:2608-17. [PMID: 26575258 DOI: 10.1056/nejmoa1512612] [Citation(s) in RCA: 645] [Impact Index Per Article: 64.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
BACKGROUND In phase 2 trials, treatment with the combination of the nucleotide polymerase inhibitor sofosbuvir and the NS5A inhibitor velpatasvir resulted in high rates of sustained virologic response in patients chronically infected with hepatitis C virus (HCV) genotype 2 or 3. METHODS We conducted two randomized, phase 3, open-label studies involving patients who had received previous treatment for HCV genotype 2 or 3 and those who had not received such treatment, including patients with compensated cirrhosis. In one trial, patients with HCV genotype 2 were randomly assigned in a 1:1 ratio to receive sofosbuvir-velpatasvir, in a once-daily, fixed-dose combination tablet (134 patients), or sofosbuvir plus weight-based ribavirin (132 patients) for 12 weeks. In a second trial, patients with HCV genotype 3 were randomly assigned in a 1:1 ratio to receive sofosbuvir-velpatasvir for 12 weeks (277 patients) or sofosbuvir-ribavirin for 24 weeks (275 patients). The primary end point for the two trials was a sustained virologic response at 12 weeks after the end of therapy. RESULTS Among patients with HCV genotype 2, the rate of sustained virologic response in the sofosbuvir-velpatasvir group was 99% (95% confidence interval [CI], 96 to 100), which was superior to the rate of 94% (95% CI, 88 to 97) in the sofosbuvir-ribavirin group (P=0.02). Among patients with HCV genotype 3, the rate of sustained virologic response in the sofosbuvir-velpatasvir group was 95% (95% CI, 92 to 98), which was superior to the rate of 80% (95% CI, 75 to 85) in the sofosbuvir-ribavirin group (P<0.001). The most common adverse events in the two studies were fatigue, headache, nausea, and insomnia. CONCLUSIONS Among patients with HCV genotype 2 or 3 with or without previous treatment, including those with compensated cirrhosis, 12 weeks of treatment with sofosbuvir-velpatasvir resulted in rates of sustained virologic response that were superior to those with standard treatment with sofosbuvir-ribavirin. (Funded by Gilead Sciences; ASTRAL-2 ClinicalTrials.gov number, NCT02220998; and ASTRAL-3, NCT02201953.).
Collapse
Affiliation(s)
- Graham R Foster
- From Queen Mary University of London (G.R.F.), University College London (W.R.), King's College Hospital (W.R.), and Institute of Liver Studies (K.A.) - all in London; Beth Israel Deaconess Medical Center, Boston (N.A.); Alfred Health and Monash University (S.K.R.) and St. Vincent's Hospital (A.T.), Melbourne, VIC, and Monash Health and Monash University, Clayton, VIC (S.P.) - all in Australia; James J. Peters Veterans Affairs Medical Center, Bronx (N.B.), and Icahn School of Medicine at Mount Sinai, New York (N.B.) - both in New York; Auckland Clinical Studies, Auckland (E.J.G.), and Christchurch Clinical Studies Trust and University of Otago, Christchurch (C.A.M.S.) - both in New Zealand; Texas Liver Institute, University of Texas Health Science Center, San Antonio (E.L.); Liver Institute of Virginia, Richmond (M.L.S.); University of Ottawa, Ottawa (C.C.), and Vancouver Infectious Diseases Centre, Vancouver, BC (B.C.) - both in Canada; Kaiser Permanente (W.J.T.), Ruane Medical (P.R.), and Cedars-Sinai Medical Center (T.T.T.), Los Angeles, and Gilead Sciences, Foster City (H.M., H.D.-S., L.H., J.W., J.M., A.O., D.M.B., J.G.M.) - all in California; Hôpital Saint Joseph, Marseilles (M.B.), and Service d'Hépatologie, Hôpital Beaujon, INSERM UMR 1149, Université Paris Diderot, Clichy (T.A.) - both in France; University Hospital Leipzig, Leipzig (T.B.), and Johann Wolfgang Goethe University, Frankfurt (S.Z.) - both in Germany; Santa Maria Annunziata Hospital, Florence (F.M.), and Casa Sollievo della Sofferenza Hospital, San Giovanni Rotondo (A.M.) - both in Italy; Henry Ford Health System, Detroit (S.C.G.); Duke University School of Medicine, Durham, NC (K.P.); Rush University Medical Center, Chicago (N.R.); and Johns Hopkins University, Baltimore (M.S.)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
74
|
Transcriptomic Analysis of Chronic Hepatitis B and C and Liver Cancer Reveals MicroRNA-Mediated Control of Cholesterol Synthesis Programs. mBio 2015; 6:e01500-15. [PMID: 26646011 PMCID: PMC4676282 DOI: 10.1128/mbio.01500-15] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Chronic hepatitis B (CHB), chronic hepatitis C (CHC), and associated hepatocellular carcinoma (HCC) are characterized by cholesterol imbalance and dyslipidemia; however, the key regulatory drivers of these phenotypes are incompletely understood. Using gene expression microarrays and high-throughput sequencing of small RNAs, we performed integrative analysis of microRNA (miRNA) and gene expression in nonmalignant and matched cancer tissue samples from human subjects with CHB or CHC and HCC. We also carried out follow-up functional studies of specific miRNAs in a cell-based system. These studies led to four major findings. First, pathways affecting cholesterol homeostasis were among the most significantly overrepresented among genes dysregulated in chronic viral hepatitis and especially in tumor tissue. Second, for each disease state, specific miRNA signatures that included miRNAs not previously associated with chronic viral hepatitis, such as miR-1307 in CHC, were identified. Notably, a few miRNAs, including miR-27 and miR-224, were components of the miRNA signatures of all four disease states: CHB, CHC, CHB-associated HCC, and CHC-associated HCC. Third, using a statistical simulation method (miRHub) applied to the gene expression data, we identified candidate master miRNA regulators of pathways controlling cholesterol homeostasis in chronic viral hepatitis and HCC, including miR-21, miR-27, and miR-33. Last, we validated in human hepatoma cells that both miR-21 and miR-27 significantly repress cholesterol synthesis and that miR-27 does so in part through regulation of the gene that codes for the rate-limiting enzyme 3-hydroxy-3-methyl-glutaryl-coenzyme A (HMG-CoA) reductase (HMGCR). Hepatitis B virus (HBV) and hepatitis C virus (HCV) are phylogenetically unrelated hepatotropic viruses that persistently infect hundreds of millions of people world-wide, often leading to chronic liver disease and hepatocellular carcinoma (HCC). Chronic hepatitis B (CHB), chronic hepatitis C (CHC), and associated HCC often lead to cholesterol imbalance and dyslipidemia. However, the regulatory mechanisms underlying the dysregulation of lipid pathways in these disease states are incompletely understood. MicroRNAs (miRNAs) have emerged as critical modulators of lipid homeostasis. Here we use a blend of genomic, molecular, and biochemical strategies to identify key miRNAs that drive the lipid phenotypes of chronic viral hepatitis and HCC. These findings provide a panoramic view of the miRNA landscape in chronic viral hepatitis, which could contribute to the development of novel and more-effective miRNA-based therapeutic strategies.
Collapse
|
75
|
Mattos AAD, Marcon PDS, Araújo FSBD, Coral GP, Tovo CV. HEPATOCELLULAR CARCINOMA IN A NON-CIRRHOTIC PATIENT WITH SUSTAINED VIROLOGICAL RESPONSE AFTER HEPATITIS C TREATMENT. Rev Inst Med Trop Sao Paulo 2015; 57:519-522. [PMID: 27049708 PMCID: PMC4727140 DOI: 10.1590/s0036-46652015000600011] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2014] [Accepted: 04/01/2015] [Indexed: 02/08/2023] Open
Abstract
Chronic infection by hepatitis C virus (HCV) is one of the main risk factors for the development of liver cirrhosis and hepatocellular carcinoma. However, the emergence of hepatocellular carcinoma (HCC) in non-cirrhotic HCV patients, especially after sustained virological response (SVR) is an unusual event. Recently, it has been suggested that HCV genotype 3 may have a particular oncogenic mechanism, but the factors involved in these cases as well as the profile of these patients are still not fully understood. Thus, we present the case of a non-cirrhotic fifty-year-old male with HCV infection, genotype 3a, who developed HCC two years after treatment with pegylated-interferon and ribavirin, with SVR, in Brazil.
Collapse
|
76
|
Everson GT, Towner WJ, Davis MN, Wyles DL, Nahass RG, Thuluvath PJ, Etzkorn K, Hinestrosa F, Tong M, Rabinovitz M, McNally J, Brainard DM, Han L, Doehle B, McHutchison JG, Morgan T, Chung RT, Tran TT. Sofosbuvir With Velpatasvir in Treatment-Naive Noncirrhotic Patients With Genotype 1 to 6 Hepatitis C Virus Infection: A Randomized Trial. Ann Intern Med 2015; 163:818-26. [PMID: 26551051 DOI: 10.7326/m15-1000] [Citation(s) in RCA: 85] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Effective, pangenotypic treatments for hepatitis C virus (HCV) infection are needed. OBJECTIVE To assess the safety and efficacy of sofosbuvir with velpatasvir in patients infected with HCV genotypes 1 to 6. DESIGN Randomized, phase 2, open-label study. (ClinicalTrials.gov: NCT01858766). SETTING 48 U.S. sites. PATIENTS 377 treatment-naive noncirrhotic patients. In part A, patients infected with HCV genotypes 1 to 6 were randomly assigned to sofosbuvir, 400 mg, with velpatasvir, 25 or 100 mg, for 12 weeks. In part B, patients with genotype 1 or 2 HCV infection were randomly assigned to sofosbuvir, 400 mg, and velpatasvir, 25 or 100 mg, with or without ribavirin for 8 weeks. MEASUREMENTS Sustained virologic response at 12 weeks (SVR12). RESULTS In part A, SVR12 rates were 96% (26 of 27) with velpatasvir, 25 mg, and 100% (28 of 28) with velpatasvir, 100 mg, for genotype 1; 93% (25 of 27) in both groups for genotype 3; and 96% (22 of 23) with velpatasvir, 25 mg, and 95% (21 of 22) with velpatasvir, 100 mg, for genotypes 2, 4, 5, and 6. In part B, for genotype 1, SVR12 rates were 87% (26 of 30) with velpatasvir, 25 mg; 83% (25 of 30) with velpatasvir, 25 mg, plus ribavirin; 90% (26 of 29) with velpatasvir, 100 mg; and 81% (25 of 31) with velpatasvir, 100 mg, plus ribavirin. For genotype 2, SVR12 rates were 77% (20 of 26) with velpatasvir, 25 mg; 88% (22 of 25) with velpatasvir, 25 mg, plus ribavirin; 88% (23 of 26) with velpatasvir, 100 mg; and 88% (23 of 26) with velpatasvir, 100 mg, plus ribavirin. Adverse events included fatigue (21%), headache (20%), and nausea (12%). One patient committed suicide. LIMITATION The study was open-label, no inferential statistics were planned, and sample sizes were small. CONCLUSION Twelve weeks of sofosbuvir, 400 mg, and velpatasvir, 100 mg, was well-tolerated and resulted in high SVR in patients infected with HCV genotypes 1 to 6. PRIMARY FUNDING SOURCE Gilead Sciences.
Collapse
Affiliation(s)
- Gregory T. Everson
- From University of Colorado Denver, Aurora, Colorado; Kaiser Permanente and Cedars-Sinai Medical Center, Los Angeles, California; Digestive CARE, South Florida Center of Gastroenterology, Wellington, Florida; University of California, San Diego, La Jolla, California; ID CARE, Hillsborough, New Jersey; Mercy Medical Center, Baltimore, Maryland; Borland-Groover Clinic, Jacksonville, Florida; Orlando Immunology Center, Orlando, Florida; Huntington Medical Research Institutes, Pasadena, California
- University of Pittsburgh, Pittsburgh, Pennsylvania; Gilead Sciences, Foster City, California; VA Long Beach Healthcare System, Long Beach, California; and Massachusetts General Hospital, Boston, Massachusetts
| | - William J. Towner
- From University of Colorado Denver, Aurora, Colorado; Kaiser Permanente and Cedars-Sinai Medical Center, Los Angeles, California; Digestive CARE, South Florida Center of Gastroenterology, Wellington, Florida; University of California, San Diego, La Jolla, California; ID CARE, Hillsborough, New Jersey; Mercy Medical Center, Baltimore, Maryland; Borland-Groover Clinic, Jacksonville, Florida; Orlando Immunology Center, Orlando, Florida; Huntington Medical Research Institutes, Pasadena, California
- University of Pittsburgh, Pittsburgh, Pennsylvania; Gilead Sciences, Foster City, California; VA Long Beach Healthcare System, Long Beach, California; and Massachusetts General Hospital, Boston, Massachusetts
| | - Mitchell N. Davis
- From University of Colorado Denver, Aurora, Colorado; Kaiser Permanente and Cedars-Sinai Medical Center, Los Angeles, California; Digestive CARE, South Florida Center of Gastroenterology, Wellington, Florida; University of California, San Diego, La Jolla, California; ID CARE, Hillsborough, New Jersey; Mercy Medical Center, Baltimore, Maryland; Borland-Groover Clinic, Jacksonville, Florida; Orlando Immunology Center, Orlando, Florida; Huntington Medical Research Institutes, Pasadena, California
- University of Pittsburgh, Pittsburgh, Pennsylvania; Gilead Sciences, Foster City, California; VA Long Beach Healthcare System, Long Beach, California; and Massachusetts General Hospital, Boston, Massachusetts
| | - David L. Wyles
- From University of Colorado Denver, Aurora, Colorado; Kaiser Permanente and Cedars-Sinai Medical Center, Los Angeles, California; Digestive CARE, South Florida Center of Gastroenterology, Wellington, Florida; University of California, San Diego, La Jolla, California; ID CARE, Hillsborough, New Jersey; Mercy Medical Center, Baltimore, Maryland; Borland-Groover Clinic, Jacksonville, Florida; Orlando Immunology Center, Orlando, Florida; Huntington Medical Research Institutes, Pasadena, California
- University of Pittsburgh, Pittsburgh, Pennsylvania; Gilead Sciences, Foster City, California; VA Long Beach Healthcare System, Long Beach, California; and Massachusetts General Hospital, Boston, Massachusetts
| | - Ronald G. Nahass
- From University of Colorado Denver, Aurora, Colorado; Kaiser Permanente and Cedars-Sinai Medical Center, Los Angeles, California; Digestive CARE, South Florida Center of Gastroenterology, Wellington, Florida; University of California, San Diego, La Jolla, California; ID CARE, Hillsborough, New Jersey; Mercy Medical Center, Baltimore, Maryland; Borland-Groover Clinic, Jacksonville, Florida; Orlando Immunology Center, Orlando, Florida; Huntington Medical Research Institutes, Pasadena, California
- University of Pittsburgh, Pittsburgh, Pennsylvania; Gilead Sciences, Foster City, California; VA Long Beach Healthcare System, Long Beach, California; and Massachusetts General Hospital, Boston, Massachusetts
| | - Paul J. Thuluvath
- From University of Colorado Denver, Aurora, Colorado; Kaiser Permanente and Cedars-Sinai Medical Center, Los Angeles, California; Digestive CARE, South Florida Center of Gastroenterology, Wellington, Florida; University of California, San Diego, La Jolla, California; ID CARE, Hillsborough, New Jersey; Mercy Medical Center, Baltimore, Maryland; Borland-Groover Clinic, Jacksonville, Florida; Orlando Immunology Center, Orlando, Florida; Huntington Medical Research Institutes, Pasadena, California
- University of Pittsburgh, Pittsburgh, Pennsylvania; Gilead Sciences, Foster City, California; VA Long Beach Healthcare System, Long Beach, California; and Massachusetts General Hospital, Boston, Massachusetts
| | - Kyle Etzkorn
- From University of Colorado Denver, Aurora, Colorado; Kaiser Permanente and Cedars-Sinai Medical Center, Los Angeles, California; Digestive CARE, South Florida Center of Gastroenterology, Wellington, Florida; University of California, San Diego, La Jolla, California; ID CARE, Hillsborough, New Jersey; Mercy Medical Center, Baltimore, Maryland; Borland-Groover Clinic, Jacksonville, Florida; Orlando Immunology Center, Orlando, Florida; Huntington Medical Research Institutes, Pasadena, California
- University of Pittsburgh, Pittsburgh, Pennsylvania; Gilead Sciences, Foster City, California; VA Long Beach Healthcare System, Long Beach, California; and Massachusetts General Hospital, Boston, Massachusetts
| | - Federico Hinestrosa
- From University of Colorado Denver, Aurora, Colorado; Kaiser Permanente and Cedars-Sinai Medical Center, Los Angeles, California; Digestive CARE, South Florida Center of Gastroenterology, Wellington, Florida; University of California, San Diego, La Jolla, California; ID CARE, Hillsborough, New Jersey; Mercy Medical Center, Baltimore, Maryland; Borland-Groover Clinic, Jacksonville, Florida; Orlando Immunology Center, Orlando, Florida; Huntington Medical Research Institutes, Pasadena, California
- University of Pittsburgh, Pittsburgh, Pennsylvania; Gilead Sciences, Foster City, California; VA Long Beach Healthcare System, Long Beach, California; and Massachusetts General Hospital, Boston, Massachusetts
| | - Myron Tong
- From University of Colorado Denver, Aurora, Colorado; Kaiser Permanente and Cedars-Sinai Medical Center, Los Angeles, California; Digestive CARE, South Florida Center of Gastroenterology, Wellington, Florida; University of California, San Diego, La Jolla, California; ID CARE, Hillsborough, New Jersey; Mercy Medical Center, Baltimore, Maryland; Borland-Groover Clinic, Jacksonville, Florida; Orlando Immunology Center, Orlando, Florida; Huntington Medical Research Institutes, Pasadena, California
- University of Pittsburgh, Pittsburgh, Pennsylvania; Gilead Sciences, Foster City, California; VA Long Beach Healthcare System, Long Beach, California; and Massachusetts General Hospital, Boston, Massachusetts
| | - Mordechai Rabinovitz
- From University of Colorado Denver, Aurora, Colorado; Kaiser Permanente and Cedars-Sinai Medical Center, Los Angeles, California; Digestive CARE, South Florida Center of Gastroenterology, Wellington, Florida; University of California, San Diego, La Jolla, California; ID CARE, Hillsborough, New Jersey; Mercy Medical Center, Baltimore, Maryland; Borland-Groover Clinic, Jacksonville, Florida; Orlando Immunology Center, Orlando, Florida; Huntington Medical Research Institutes, Pasadena, California
- University of Pittsburgh, Pittsburgh, Pennsylvania; Gilead Sciences, Foster City, California; VA Long Beach Healthcare System, Long Beach, California; and Massachusetts General Hospital, Boston, Massachusetts
| | - John McNally
- From University of Colorado Denver, Aurora, Colorado; Kaiser Permanente and Cedars-Sinai Medical Center, Los Angeles, California; Digestive CARE, South Florida Center of Gastroenterology, Wellington, Florida; University of California, San Diego, La Jolla, California; ID CARE, Hillsborough, New Jersey; Mercy Medical Center, Baltimore, Maryland; Borland-Groover Clinic, Jacksonville, Florida; Orlando Immunology Center, Orlando, Florida; Huntington Medical Research Institutes, Pasadena, California
- University of Pittsburgh, Pittsburgh, Pennsylvania; Gilead Sciences, Foster City, California; VA Long Beach Healthcare System, Long Beach, California; and Massachusetts General Hospital, Boston, Massachusetts
| | - Diana M. Brainard
- From University of Colorado Denver, Aurora, Colorado; Kaiser Permanente and Cedars-Sinai Medical Center, Los Angeles, California; Digestive CARE, South Florida Center of Gastroenterology, Wellington, Florida; University of California, San Diego, La Jolla, California; ID CARE, Hillsborough, New Jersey; Mercy Medical Center, Baltimore, Maryland; Borland-Groover Clinic, Jacksonville, Florida; Orlando Immunology Center, Orlando, Florida; Huntington Medical Research Institutes, Pasadena, California
- University of Pittsburgh, Pittsburgh, Pennsylvania; Gilead Sciences, Foster City, California; VA Long Beach Healthcare System, Long Beach, California; and Massachusetts General Hospital, Boston, Massachusetts
| | - Lingling Han
- From University of Colorado Denver, Aurora, Colorado; Kaiser Permanente and Cedars-Sinai Medical Center, Los Angeles, California; Digestive CARE, South Florida Center of Gastroenterology, Wellington, Florida; University of California, San Diego, La Jolla, California; ID CARE, Hillsborough, New Jersey; Mercy Medical Center, Baltimore, Maryland; Borland-Groover Clinic, Jacksonville, Florida; Orlando Immunology Center, Orlando, Florida; Huntington Medical Research Institutes, Pasadena, California
- University of Pittsburgh, Pittsburgh, Pennsylvania; Gilead Sciences, Foster City, California; VA Long Beach Healthcare System, Long Beach, California; and Massachusetts General Hospital, Boston, Massachusetts
| | - Brian Doehle
- From University of Colorado Denver, Aurora, Colorado; Kaiser Permanente and Cedars-Sinai Medical Center, Los Angeles, California; Digestive CARE, South Florida Center of Gastroenterology, Wellington, Florida; University of California, San Diego, La Jolla, California; ID CARE, Hillsborough, New Jersey; Mercy Medical Center, Baltimore, Maryland; Borland-Groover Clinic, Jacksonville, Florida; Orlando Immunology Center, Orlando, Florida; Huntington Medical Research Institutes, Pasadena, California
- University of Pittsburgh, Pittsburgh, Pennsylvania; Gilead Sciences, Foster City, California; VA Long Beach Healthcare System, Long Beach, California; and Massachusetts General Hospital, Boston, Massachusetts
| | - John G. McHutchison
- From University of Colorado Denver, Aurora, Colorado; Kaiser Permanente and Cedars-Sinai Medical Center, Los Angeles, California; Digestive CARE, South Florida Center of Gastroenterology, Wellington, Florida; University of California, San Diego, La Jolla, California; ID CARE, Hillsborough, New Jersey; Mercy Medical Center, Baltimore, Maryland; Borland-Groover Clinic, Jacksonville, Florida; Orlando Immunology Center, Orlando, Florida; Huntington Medical Research Institutes, Pasadena, California
- University of Pittsburgh, Pittsburgh, Pennsylvania; Gilead Sciences, Foster City, California; VA Long Beach Healthcare System, Long Beach, California; and Massachusetts General Hospital, Boston, Massachusetts
| | - Timothy Morgan
- From University of Colorado Denver, Aurora, Colorado; Kaiser Permanente and Cedars-Sinai Medical Center, Los Angeles, California; Digestive CARE, South Florida Center of Gastroenterology, Wellington, Florida; University of California, San Diego, La Jolla, California; ID CARE, Hillsborough, New Jersey; Mercy Medical Center, Baltimore, Maryland; Borland-Groover Clinic, Jacksonville, Florida; Orlando Immunology Center, Orlando, Florida; Huntington Medical Research Institutes, Pasadena, California
- University of Pittsburgh, Pittsburgh, Pennsylvania; Gilead Sciences, Foster City, California; VA Long Beach Healthcare System, Long Beach, California; and Massachusetts General Hospital, Boston, Massachusetts
| | - Raymond T. Chung
- From University of Colorado Denver, Aurora, Colorado; Kaiser Permanente and Cedars-Sinai Medical Center, Los Angeles, California; Digestive CARE, South Florida Center of Gastroenterology, Wellington, Florida; University of California, San Diego, La Jolla, California; ID CARE, Hillsborough, New Jersey; Mercy Medical Center, Baltimore, Maryland; Borland-Groover Clinic, Jacksonville, Florida; Orlando Immunology Center, Orlando, Florida; Huntington Medical Research Institutes, Pasadena, California
- University of Pittsburgh, Pittsburgh, Pennsylvania; Gilead Sciences, Foster City, California; VA Long Beach Healthcare System, Long Beach, California; and Massachusetts General Hospital, Boston, Massachusetts
| | - Tram T. Tran
- From University of Colorado Denver, Aurora, Colorado; Kaiser Permanente and Cedars-Sinai Medical Center, Los Angeles, California; Digestive CARE, South Florida Center of Gastroenterology, Wellington, Florida; University of California, San Diego, La Jolla, California; ID CARE, Hillsborough, New Jersey; Mercy Medical Center, Baltimore, Maryland; Borland-Groover Clinic, Jacksonville, Florida; Orlando Immunology Center, Orlando, Florida; Huntington Medical Research Institutes, Pasadena, California
- University of Pittsburgh, Pittsburgh, Pennsylvania; Gilead Sciences, Foster City, California; VA Long Beach Healthcare System, Long Beach, California; and Massachusetts General Hospital, Boston, Massachusetts
| |
Collapse
|
77
|
Pianko S, Flamm SL, Shiffman ML, Kumar S, Strasser SI, Dore GJ, McNally J, Brainard DM, Han L, Doehle B, Mogalian E, McHutchison JG, Rabinovitz M, Towner WJ, Gane EJ, Stedman CA, Reddy KR, Roberts SK. Sofosbuvir Plus Velpatasvir Combination Therapy for Treatment-Experienced Patients With Genotype 1 or 3 Hepatitis C Virus Infection: A Randomized Trial. Ann Intern Med 2015; 163:809-17. [PMID: 26551263 DOI: 10.7326/m15-1014] [Citation(s) in RCA: 73] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Effective treatment options are needed for patients with genotype 1 or 3 hepatitis C virus (HCV) infection in whom previous therapy has failed. OBJECTIVE To assess the efficacy and safety of sofosbuvir plus velpatasvir, with and without ribavirin, in treatment-experienced patients. DESIGN Randomized, phase 2, open-label study. (ClinicalTrials.gov: NCT01909804). SETTING 58 sites in Australia, New Zealand, and the United States. PATIENTS Treatment-experienced adults with genotype 3 HCV infection without cirrhosis (cohort 1) and with compensated cirrhosis (cohort 2) and patients with genotype 1 HCV infection that was unsuccessfully treated with a protease inhibitor with peginterferon and ribavirin (50% could have compensated cirrhosis) (cohort 3). INTERVENTION All patients received 12 weeks of treatment that included 400 mg of sofosbuvir once daily. Patients in each cohort were randomly assigned to 25 mg of velpatasvir once daily with or without ribavirin or 100 mg of velpatasvir once daily with or without ribavirin. MEASUREMENTS Proportion of patients with sustained virologic response at week 12 after treatment (SVR12). RESULTS In cohort 1, SVR12 rates were 85% with 25 mg of velpatasvir, 96% with 25 mg of velpatasvir plus ribavirin, 100% with 100 mg of velpatasvir, and 100% with 100 mg of velpatasvir plus ribavirin. In cohort 2, SVR12 rates were 58% with 25 mg of velpatasvir, 84% with 25 mg of velpatasvir plus ribavirin, 88% with 100 mg of velpatasvir, and 96% with 100 mg of velpatasvir plus ribavirin. In cohort 3, SVR12 rates were 100% with 25 mg of velpatasvir, 97% with 25 mg of velpatasvir plus ribavirin, 100% with 100 mg of velpatasvir, and 96% with 100 mg of velpatasvir plus ribavirin. The most common adverse events were headache, fatigue, and nausea. LIMITATION Treatment assignments were not blinded, and no inferential statistics were planned. CONCLUSION Treatment with 400 mg of sofosbuvir plus 100 mg of velpatasvir for 12 weeks was well-tolerated and highly effective in treatment-experienced patients with genotype 1 or 3 HCV infection. PRIMARY FUNDING SOURCE Gilead Sciences.
Collapse
Affiliation(s)
- Stephen Pianko
- From Monash Health and Monash University, Clayton, Victoria, Australia; Northwestern University, Chicago, Illinois; Liver Institute of Virginia, Richmond, Virginia; Weill Cornell Medical College, New York, New York; Royal Prince Alfred Hospital and University of Sydney, Camperdown, New South Wales, Australia; Kirby Institute, University of New South Wales, and St. Vincent's Hospital, Sydney, New South Wales, Australia; Gilead Sciences, Foster City, California
- University of Pittsburgh, Pittsburgh, Pennsylvania; Kaiser Permanente Medical Center, Los Angeles, California; Auckland Clinical Studies, Auckland, New Zealand; Christchurch Clinical Studies Trust and University of Otago, Christchurch, New Zealand; University of Pennsylvania, Philadelphia, Pennsylvania; and Alfred Health and Monash University, Melbourne, Victoria, Australia
| | - Steven L. Flamm
- From Monash Health and Monash University, Clayton, Victoria, Australia; Northwestern University, Chicago, Illinois; Liver Institute of Virginia, Richmond, Virginia; Weill Cornell Medical College, New York, New York; Royal Prince Alfred Hospital and University of Sydney, Camperdown, New South Wales, Australia; Kirby Institute, University of New South Wales, and St. Vincent's Hospital, Sydney, New South Wales, Australia; Gilead Sciences, Foster City, California
- University of Pittsburgh, Pittsburgh, Pennsylvania; Kaiser Permanente Medical Center, Los Angeles, California; Auckland Clinical Studies, Auckland, New Zealand; Christchurch Clinical Studies Trust and University of Otago, Christchurch, New Zealand; University of Pennsylvania, Philadelphia, Pennsylvania; and Alfred Health and Monash University, Melbourne, Victoria, Australia
| | - Mitchell L. Shiffman
- From Monash Health and Monash University, Clayton, Victoria, Australia; Northwestern University, Chicago, Illinois; Liver Institute of Virginia, Richmond, Virginia; Weill Cornell Medical College, New York, New York; Royal Prince Alfred Hospital and University of Sydney, Camperdown, New South Wales, Australia; Kirby Institute, University of New South Wales, and St. Vincent's Hospital, Sydney, New South Wales, Australia; Gilead Sciences, Foster City, California
- University of Pittsburgh, Pittsburgh, Pennsylvania; Kaiser Permanente Medical Center, Los Angeles, California; Auckland Clinical Studies, Auckland, New Zealand; Christchurch Clinical Studies Trust and University of Otago, Christchurch, New Zealand; University of Pennsylvania, Philadelphia, Pennsylvania; and Alfred Health and Monash University, Melbourne, Victoria, Australia
| | - Sonal Kumar
- From Monash Health and Monash University, Clayton, Victoria, Australia; Northwestern University, Chicago, Illinois; Liver Institute of Virginia, Richmond, Virginia; Weill Cornell Medical College, New York, New York; Royal Prince Alfred Hospital and University of Sydney, Camperdown, New South Wales, Australia; Kirby Institute, University of New South Wales, and St. Vincent's Hospital, Sydney, New South Wales, Australia; Gilead Sciences, Foster City, California
- University of Pittsburgh, Pittsburgh, Pennsylvania; Kaiser Permanente Medical Center, Los Angeles, California; Auckland Clinical Studies, Auckland, New Zealand; Christchurch Clinical Studies Trust and University of Otago, Christchurch, New Zealand; University of Pennsylvania, Philadelphia, Pennsylvania; and Alfred Health and Monash University, Melbourne, Victoria, Australia
| | - Simone I. Strasser
- From Monash Health and Monash University, Clayton, Victoria, Australia; Northwestern University, Chicago, Illinois; Liver Institute of Virginia, Richmond, Virginia; Weill Cornell Medical College, New York, New York; Royal Prince Alfred Hospital and University of Sydney, Camperdown, New South Wales, Australia; Kirby Institute, University of New South Wales, and St. Vincent's Hospital, Sydney, New South Wales, Australia; Gilead Sciences, Foster City, California
- University of Pittsburgh, Pittsburgh, Pennsylvania; Kaiser Permanente Medical Center, Los Angeles, California; Auckland Clinical Studies, Auckland, New Zealand; Christchurch Clinical Studies Trust and University of Otago, Christchurch, New Zealand; University of Pennsylvania, Philadelphia, Pennsylvania; and Alfred Health and Monash University, Melbourne, Victoria, Australia
| | - Gregory J. Dore
- From Monash Health and Monash University, Clayton, Victoria, Australia; Northwestern University, Chicago, Illinois; Liver Institute of Virginia, Richmond, Virginia; Weill Cornell Medical College, New York, New York; Royal Prince Alfred Hospital and University of Sydney, Camperdown, New South Wales, Australia; Kirby Institute, University of New South Wales, and St. Vincent's Hospital, Sydney, New South Wales, Australia; Gilead Sciences, Foster City, California
- University of Pittsburgh, Pittsburgh, Pennsylvania; Kaiser Permanente Medical Center, Los Angeles, California; Auckland Clinical Studies, Auckland, New Zealand; Christchurch Clinical Studies Trust and University of Otago, Christchurch, New Zealand; University of Pennsylvania, Philadelphia, Pennsylvania; and Alfred Health and Monash University, Melbourne, Victoria, Australia
| | - John McNally
- From Monash Health and Monash University, Clayton, Victoria, Australia; Northwestern University, Chicago, Illinois; Liver Institute of Virginia, Richmond, Virginia; Weill Cornell Medical College, New York, New York; Royal Prince Alfred Hospital and University of Sydney, Camperdown, New South Wales, Australia; Kirby Institute, University of New South Wales, and St. Vincent's Hospital, Sydney, New South Wales, Australia; Gilead Sciences, Foster City, California
- University of Pittsburgh, Pittsburgh, Pennsylvania; Kaiser Permanente Medical Center, Los Angeles, California; Auckland Clinical Studies, Auckland, New Zealand; Christchurch Clinical Studies Trust and University of Otago, Christchurch, New Zealand; University of Pennsylvania, Philadelphia, Pennsylvania; and Alfred Health and Monash University, Melbourne, Victoria, Australia
| | - Diana M. Brainard
- From Monash Health and Monash University, Clayton, Victoria, Australia; Northwestern University, Chicago, Illinois; Liver Institute of Virginia, Richmond, Virginia; Weill Cornell Medical College, New York, New York; Royal Prince Alfred Hospital and University of Sydney, Camperdown, New South Wales, Australia; Kirby Institute, University of New South Wales, and St. Vincent's Hospital, Sydney, New South Wales, Australia; Gilead Sciences, Foster City, California
- University of Pittsburgh, Pittsburgh, Pennsylvania; Kaiser Permanente Medical Center, Los Angeles, California; Auckland Clinical Studies, Auckland, New Zealand; Christchurch Clinical Studies Trust and University of Otago, Christchurch, New Zealand; University of Pennsylvania, Philadelphia, Pennsylvania; and Alfred Health and Monash University, Melbourne, Victoria, Australia
| | - Lingling Han
- From Monash Health and Monash University, Clayton, Victoria, Australia; Northwestern University, Chicago, Illinois; Liver Institute of Virginia, Richmond, Virginia; Weill Cornell Medical College, New York, New York; Royal Prince Alfred Hospital and University of Sydney, Camperdown, New South Wales, Australia; Kirby Institute, University of New South Wales, and St. Vincent's Hospital, Sydney, New South Wales, Australia; Gilead Sciences, Foster City, California
- University of Pittsburgh, Pittsburgh, Pennsylvania; Kaiser Permanente Medical Center, Los Angeles, California; Auckland Clinical Studies, Auckland, New Zealand; Christchurch Clinical Studies Trust and University of Otago, Christchurch, New Zealand; University of Pennsylvania, Philadelphia, Pennsylvania; and Alfred Health and Monash University, Melbourne, Victoria, Australia
| | - Brian Doehle
- From Monash Health and Monash University, Clayton, Victoria, Australia; Northwestern University, Chicago, Illinois; Liver Institute of Virginia, Richmond, Virginia; Weill Cornell Medical College, New York, New York; Royal Prince Alfred Hospital and University of Sydney, Camperdown, New South Wales, Australia; Kirby Institute, University of New South Wales, and St. Vincent's Hospital, Sydney, New South Wales, Australia; Gilead Sciences, Foster City, California
- University of Pittsburgh, Pittsburgh, Pennsylvania; Kaiser Permanente Medical Center, Los Angeles, California; Auckland Clinical Studies, Auckland, New Zealand; Christchurch Clinical Studies Trust and University of Otago, Christchurch, New Zealand; University of Pennsylvania, Philadelphia, Pennsylvania; and Alfred Health and Monash University, Melbourne, Victoria, Australia
| | - Erik Mogalian
- From Monash Health and Monash University, Clayton, Victoria, Australia; Northwestern University, Chicago, Illinois; Liver Institute of Virginia, Richmond, Virginia; Weill Cornell Medical College, New York, New York; Royal Prince Alfred Hospital and University of Sydney, Camperdown, New South Wales, Australia; Kirby Institute, University of New South Wales, and St. Vincent's Hospital, Sydney, New South Wales, Australia; Gilead Sciences, Foster City, California
- University of Pittsburgh, Pittsburgh, Pennsylvania; Kaiser Permanente Medical Center, Los Angeles, California; Auckland Clinical Studies, Auckland, New Zealand; Christchurch Clinical Studies Trust and University of Otago, Christchurch, New Zealand; University of Pennsylvania, Philadelphia, Pennsylvania; and Alfred Health and Monash University, Melbourne, Victoria, Australia
| | - John G. McHutchison
- From Monash Health and Monash University, Clayton, Victoria, Australia; Northwestern University, Chicago, Illinois; Liver Institute of Virginia, Richmond, Virginia; Weill Cornell Medical College, New York, New York; Royal Prince Alfred Hospital and University of Sydney, Camperdown, New South Wales, Australia; Kirby Institute, University of New South Wales, and St. Vincent's Hospital, Sydney, New South Wales, Australia; Gilead Sciences, Foster City, California
- University of Pittsburgh, Pittsburgh, Pennsylvania; Kaiser Permanente Medical Center, Los Angeles, California; Auckland Clinical Studies, Auckland, New Zealand; Christchurch Clinical Studies Trust and University of Otago, Christchurch, New Zealand; University of Pennsylvania, Philadelphia, Pennsylvania; and Alfred Health and Monash University, Melbourne, Victoria, Australia
| | - Mordechai Rabinovitz
- From Monash Health and Monash University, Clayton, Victoria, Australia; Northwestern University, Chicago, Illinois; Liver Institute of Virginia, Richmond, Virginia; Weill Cornell Medical College, New York, New York; Royal Prince Alfred Hospital and University of Sydney, Camperdown, New South Wales, Australia; Kirby Institute, University of New South Wales, and St. Vincent's Hospital, Sydney, New South Wales, Australia; Gilead Sciences, Foster City, California
- University of Pittsburgh, Pittsburgh, Pennsylvania; Kaiser Permanente Medical Center, Los Angeles, California; Auckland Clinical Studies, Auckland, New Zealand; Christchurch Clinical Studies Trust and University of Otago, Christchurch, New Zealand; University of Pennsylvania, Philadelphia, Pennsylvania; and Alfred Health and Monash University, Melbourne, Victoria, Australia
| | - William J. Towner
- From Monash Health and Monash University, Clayton, Victoria, Australia; Northwestern University, Chicago, Illinois; Liver Institute of Virginia, Richmond, Virginia; Weill Cornell Medical College, New York, New York; Royal Prince Alfred Hospital and University of Sydney, Camperdown, New South Wales, Australia; Kirby Institute, University of New South Wales, and St. Vincent's Hospital, Sydney, New South Wales, Australia; Gilead Sciences, Foster City, California
- University of Pittsburgh, Pittsburgh, Pennsylvania; Kaiser Permanente Medical Center, Los Angeles, California; Auckland Clinical Studies, Auckland, New Zealand; Christchurch Clinical Studies Trust and University of Otago, Christchurch, New Zealand; University of Pennsylvania, Philadelphia, Pennsylvania; and Alfred Health and Monash University, Melbourne, Victoria, Australia
| | - Edward J. Gane
- From Monash Health and Monash University, Clayton, Victoria, Australia; Northwestern University, Chicago, Illinois; Liver Institute of Virginia, Richmond, Virginia; Weill Cornell Medical College, New York, New York; Royal Prince Alfred Hospital and University of Sydney, Camperdown, New South Wales, Australia; Kirby Institute, University of New South Wales, and St. Vincent's Hospital, Sydney, New South Wales, Australia; Gilead Sciences, Foster City, California
- University of Pittsburgh, Pittsburgh, Pennsylvania; Kaiser Permanente Medical Center, Los Angeles, California; Auckland Clinical Studies, Auckland, New Zealand; Christchurch Clinical Studies Trust and University of Otago, Christchurch, New Zealand; University of Pennsylvania, Philadelphia, Pennsylvania; and Alfred Health and Monash University, Melbourne, Victoria, Australia
| | - Catherine A.M. Stedman
- From Monash Health and Monash University, Clayton, Victoria, Australia; Northwestern University, Chicago, Illinois; Liver Institute of Virginia, Richmond, Virginia; Weill Cornell Medical College, New York, New York; Royal Prince Alfred Hospital and University of Sydney, Camperdown, New South Wales, Australia; Kirby Institute, University of New South Wales, and St. Vincent's Hospital, Sydney, New South Wales, Australia; Gilead Sciences, Foster City, California
- University of Pittsburgh, Pittsburgh, Pennsylvania; Kaiser Permanente Medical Center, Los Angeles, California; Auckland Clinical Studies, Auckland, New Zealand; Christchurch Clinical Studies Trust and University of Otago, Christchurch, New Zealand; University of Pennsylvania, Philadelphia, Pennsylvania; and Alfred Health and Monash University, Melbourne, Victoria, Australia
| | - K. Rajender Reddy
- From Monash Health and Monash University, Clayton, Victoria, Australia; Northwestern University, Chicago, Illinois; Liver Institute of Virginia, Richmond, Virginia; Weill Cornell Medical College, New York, New York; Royal Prince Alfred Hospital and University of Sydney, Camperdown, New South Wales, Australia; Kirby Institute, University of New South Wales, and St. Vincent's Hospital, Sydney, New South Wales, Australia; Gilead Sciences, Foster City, California
- University of Pittsburgh, Pittsburgh, Pennsylvania; Kaiser Permanente Medical Center, Los Angeles, California; Auckland Clinical Studies, Auckland, New Zealand; Christchurch Clinical Studies Trust and University of Otago, Christchurch, New Zealand; University of Pennsylvania, Philadelphia, Pennsylvania; and Alfred Health and Monash University, Melbourne, Victoria, Australia
| | - Stuart K. Roberts
- From Monash Health and Monash University, Clayton, Victoria, Australia; Northwestern University, Chicago, Illinois; Liver Institute of Virginia, Richmond, Virginia; Weill Cornell Medical College, New York, New York; Royal Prince Alfred Hospital and University of Sydney, Camperdown, New South Wales, Australia; Kirby Institute, University of New South Wales, and St. Vincent's Hospital, Sydney, New South Wales, Australia; Gilead Sciences, Foster City, California
- University of Pittsburgh, Pittsburgh, Pennsylvania; Kaiser Permanente Medical Center, Los Angeles, California; Auckland Clinical Studies, Auckland, New Zealand; Christchurch Clinical Studies Trust and University of Otago, Christchurch, New Zealand; University of Pennsylvania, Philadelphia, Pennsylvania; and Alfred Health and Monash University, Melbourne, Victoria, Australia
| |
Collapse
|
78
|
Affiliation(s)
- Patricia A Thibault
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Joyce A Wilson
- Department of Microbiology &Immunology, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| |
Collapse
|
79
|
Gondeau C, Pageaux GP, Larrey D. Hepatitis C virus infection: Are there still specific problems with genotype 3? World J Gastroenterol 2015; 21:12101-13. [PMID: 26576095 PMCID: PMC4641128 DOI: 10.3748/wjg.v21.i42.12101] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2015] [Revised: 08/07/2015] [Accepted: 09/30/2015] [Indexed: 02/06/2023] Open
Abstract
Hepatitis C virus (HCV) infection is one of the most common causes of chronic liver disease and the main indication for liver transplantation worldwide. As promising specific treatments have been introduced for genotype 1, clinicians and researchers are now focusing on patients infected by non-genotype 1 HCV, particularly genotype 3. Indeed, in the golden era of direct-acting antiviral drugs, genotype 3 infections are no longer considered as easy to treat and are associated with higher risk of developing severe liver injuries, such as cirrhosis and hepatocellular carcinoma. Moreover, HCV genotype 3 accounts for 40% of all HCV infections in Asia and is the most frequent genotype among HCV-positive injecting drug users in several countries. Here, we review recent data on HCV genotype 3 infection/treatment, including clinical aspects and the underlying genotype-specific molecular mechanisms.
Collapse
|
80
|
Foster GR, Pianko S, Brown A, Forton D, Nahass RG, George J, Barnes E, Brainard DM, Massetto B, Lin M, Han B, McHutchison JG, Subramanian GM, Cooper C, Agarwal K. Efficacy of sofosbuvir plus ribavirin with or without peginterferon-alfa in patients with hepatitis C virus genotype 3 infection and treatment-experienced patients with cirrhosis and hepatitis C virus genotype 2 infection. Gastroenterology 2015; 149:1462-70. [PMID: 26248087 DOI: 10.1053/j.gastro.2015.07.043] [Citation(s) in RCA: 178] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/28/2015] [Revised: 07/22/2015] [Accepted: 07/25/2015] [Indexed: 12/19/2022]
Abstract
BACKGROUND & AIMS We conducted an open-label, randomized, phase 3 trial to determine the efficacy and safety of sofosbuvir and ribavirin, with and without peginterferon-alfa, in treatment-experienced patients with cirrhosis and hepatitis C virus (HCV) genotype 2 infection and treatment-naïve or treatment-experienced patients with HCV genotype 3 infection. METHODS The study was conducted at 80 sites in Europe, North America, Australia, and New Zealand Patients were randomly assigned (1:1:1) to groups given sofosbuvir and ribavirin for 16 weeks (n = 196); sofosbuvir and ribavirin for 24 weeks (n = 199); or sofosbuvir, peginterferon-alfa, and ribavirin for 12 weeks (n = 197). The primary end point was the percentage of patients with HCV RNA <15 IU/mL 12 weeks after stopping therapy (sustained virologic response [SVR12]). From October 2013 until April 2014, we enrolled and treated 592 patients-48 with genotype 2 HCV and compensated cirrhosis who had not achieved SVR with previous treatments and 544 with genotype 3 HCV (279 treatment-naïve and 265 previously treated). Overall, 219 patients (37%) had compensated cirrhosis. The last post-treatment week 12 patient visit was in January 2015. RESULTS Rates of SVR12 among patients with genotype 2 HCV were 87% and 100%, for those receiving 16 and 24 weeks of sofosbuvir and ribavirin, respectively, and 94% for those receiving sofosbuvir, peginterferon, and ribavirin for 12 weeks. Rates of SVR12 among patients with genotype 3 HCV were 71% and 84% in those receiving 16 and 24 weeks of sofosbuvir and ribavirin, respectively, and 93% in those receiving sofosbuvir, peginterferon, and ribavirin. On-treatment virologic failure occurred in 3 patients with HCV genotype 3a receiving sofosbuvir and ribavirin for 24 weeks. The most common adverse events were fatigue, headache, insomnia, and nausea. Overall, 1% of patients discontinued treatment due to adverse events. CONCLUSIONS Among patients with genotype 3 HCV infection, including a large proportion of treatment-experienced patients with cirrhosis, the combination of sofosbuvir, peginterferon, and ribavirin for 12 weeks produces high rates of SVR. Treatment-experienced patients with cirrhosis and genotype 2 HCV infection had high rates of SVR in all groups. EudraCT ID 2013-002641-11.
Collapse
Affiliation(s)
- Graham R Foster
- Queen Mary University of London, Barts Health, United Kingdom.
| | - Stephen Pianko
- Monash Health and Monash University, Melbourne, Victoria, Australia
| | - Ashley Brown
- Imperial College Healthcare, National Health Service Trust, London, United Kingdom
| | - Daniel Forton
- St George's University of London, London, United Kingdom
| | | | - Jacob George
- Storr Liver Centre, Westmead Millennium Institute, University of Sydney and Westmead Hospital, Sydney, New South Wales, Australia
| | - Eleanor Barnes
- Nuffield Department of Medicine, Oxford NHIR BRC and representing STOP-HCV, United Kingdom
| | | | | | - Ming Lin
- Gilead Sciences, Foster City, California
| | - Bin Han
- Gilead Sciences, Foster City, California
| | | | | | - Curtis Cooper
- The Ottawa Hospital, University of Ottawa, Ottawa, Ontario, Canada
| | - Kosh Agarwal
- Institute of Liver Studies, King's College Hospital, London, United Kingdom
| | | |
Collapse
|
81
|
Colpitts CC, Baumert TF. Viral hepatitis: A new HCV cell culture model for the next clinical challenges. Nat Rev Gastroenterol Hepatol 2015; 12:611-3. [PMID: 26441247 DOI: 10.1038/nrgastro.2015.170] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Despite advances in hepatitis C treatment, substantial clinical hurdles remain to achieve universal cure and global control of infection. Saeed et al. identified SEC14L2 as a host factor permitting replication of clinical HCV isolates in cell culture, providing a novel system to model infection of patient-derived viruses.
Collapse
Affiliation(s)
- Che C Colpitts
- Inserm, U1110, Institut de Recherche sur les Maladies Virales et Hépatiques, Université de Strasbourg, 67000 Strasbourg, France
| | - Thomas F Baumert
- Inserm, U1110, Institut de Recherche sur les Maladies Virales et Hépatiques, Université de Strasbourg, 67000 Strasbourg, France
| |
Collapse
|
82
|
Del Campo JA, Romero-Gómez M. Modulation of host lipid metabolism by hepatitis C virus: Role of new therapies. World J Gastroenterol 2015; 21:10776-10782. [PMID: 26478669 PMCID: PMC4600579 DOI: 10.3748/wjg.v21.i38.10776] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/21/2015] [Revised: 07/07/2015] [Accepted: 09/14/2015] [Indexed: 02/06/2023] Open
Abstract
It is well established that hepatitis C virus (HCV) infection and replication relies on host lipid metabolism. HCV proteins interact and associate with lipid droplets to facilitate virion assembly and production. Besides, circulating infective particles are associated with very low-density lipoprotein. On the other hand, higher serum lipid levels have been associated with sustained viral response to pegylated interferon and ribavirin therapy in chronic HCV infection, suggesting a relevant role in viral clearance for host proteins. Host and viral genetic factors play an essential role in chronic infection. Lipid metabolism is hijacked by viral infection and could determine the success of viral replication. Recently development of direct acting antiviral agents has shown a very high efficacy (> 90%) in sustained viral response rates even for cirrhotic patients and most of the viral genotypes. HCV RNA clearance induced by Sofosbuvir has been associated with an increased concentration and size of the low-density lipoprotein particles. In this review, host genetic factors, viral factors and the interaction between them will be depicted to clarify the major issues involved in viral infection and lipid metabolism.
Collapse
|
83
|
Weiskirchen R, Tacke F. Small heterodimer partner (SHP) links hepatitis C and liver fibrosis: a small protein on the big stage. Liver Int 2015; 35:2218-21. [PMID: 26037155 DOI: 10.1111/liv.12881] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Affiliation(s)
- Ralf Weiskirchen
- Institute of Molecular Pathobiochemistry, Experimental Gene Therapy and Clinical Chemistry, University Hospital RWTH Aachen, Aachen, Germany
| | - Frank Tacke
- Department of Medicine III, University Hospital RWTH Aachen, Aachen, Germany
| |
Collapse
|
84
|
Pawlotsky JM, Flisiak R, Sarin SK, Rasenack J, Piratvisuth T, Chuang WL, Peng CY, Foster GR, Shah S, Wedemeyer H, Hézode C, Zhang W, Wong KA, Li B, Avila C, Naoumov NV. Alisporivir plus ribavirin, interferon free or in combination with pegylated interferon, for hepatitis C virus genotype 2 or 3 infection. Hepatology 2015; 62:1013-1023. [PMID: 26118427 DOI: 10.1002/hep.27960] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/06/2015] [Revised: 06/17/2015] [Accepted: 06/24/2015] [Indexed: 12/14/2022]
Abstract
UNLABELLED Alisporivir is a cyclophilin inhibitor with pan-genotypic anti-hepatitis C virus (HCV) activity and a high barrier to viral resistance. The VITAL-1 study assessed alisporivir as interferon (IFN)-free therapy in treatment-naïve patients infected with HCV genotype 2 or 3. Three hundred forty patients without cirrhosis were randomized to: arm 1, alisporivir (ALV) 1,000 mg once-daily (QD); arm 2, ALV 600 mg QD and ribavirin (RBV); arm 3, ALV 800 mg QD and RBV; arm 4, ALV 600 mg QD and pegylated IFN (Peg-IFN); or arm 5, Peg-IFN and RBV. Patients receiving IFN-free ALV regimens who achieved rapid virological response (RVR) continued the same treatment throughout, whereas those with detectable HCV RNA at week 4 received ALV, RBV, and Peg-IFN from weeks 6 to 24. Overall, 300 patients received ALV-based regimens. In arm 1 to arm 4, the intent-to-treat rates of sustained virological response (SVR) 24 weeks after treatment (SVR24) were from 80% to 85%, compared with 58% (n = 23 of 40) with Peg-IFN/RBV. Per-protocol analysis showed higher SVR24 rates in patients who received ALV/RBV, IFN-free after RVR (92%; n = 56 of 61) than with ALV alone after RVR (72%; n = 13 of 18) or with Peg-IFN/RBV (70%; n = 23 of 33). Both RVRs and SVRs to ALV IFN-free regimens were numerically higher in genotype 3- than in genotype 2-infected patients. Viral breakthrough was infrequent (3%; n = 7 of 258). IFN-free ALV treatment showed markedly better safety/tolerability than IFN-containing regimens. CONCLUSIONS ALV plus RBV represents an effective IFN-free option for a proportion of patients with HCV genotype 2 or 3 infections, with high SVR rates for patients with early viral clearance. Further investigations of ALV in IFN-free combination regimens with direct-acting antiviral drugs deserve exploration in future trials.
Collapse
Affiliation(s)
- Jean-Michel Pawlotsky
- National Reference Center for Viral Hepatitis B, C and Delta, Department of Virology, Hôpital Henri Mondor, Université Paris-Est, Créteil, France
- INSERM U955, Créteil, France
| | | | - Shiv K Sarin
- Institute of Liver and Biliary Sciences, New Delhi, India
| | - Jens Rasenack
- Klinikum der Albert-Ludwigs-Universität, Freiburg, Germany
| | - Teerha Piratvisuth
- Prince of Songkla University, Songklanagarind Hospital, Hat-Yai, Songkhla, Thailand
| | - Wan-Long Chuang
- Kaohsiung Medical University, Chang-Ho Memorial Hospital, Kaohsiung, Taiwan
| | - Cheng-Yuan Peng
- School of Medicine, China Medical University, Taichung, Taiwan
| | - Graham R Foster
- Bart's and The London School of Medicine, Queen Mary's University of London, London, United Kingdom
| | | | | | - Christophe Hézode
- INSERM U955, Créteil, France
- Department of Hepatology and Gastroenterology, Hôpital Henri Mondor, Université Paris-Est, Créteil, France
| | - Wei Zhang
- Beijing Novartis Pharma Co Ltd, Shanghai, China
| | - Kelly A Wong
- Novartis Institute of Biomedical Research, Emeryville, CA
| | - Bin Li
- Novartis Institute of Biomedical Research, Cambridge, MA
| | | | | |
Collapse
|
85
|
Martini A, Fattovich G, Guido M, Bugianesi E, Biasiolo A, Ieluzzi D, Gallotta A, Fassina G, Merkel C, Gatta A, Negro F, Pontisso P. HCV genotype 3 and squamous cell carcinoma antigen (SCCA)-IgM are independently associated with histological features of NASH in HCV-infected patients. J Viral Hepat 2015; 22:800-8. [PMID: 25611978 DOI: 10.1111/jvh.12394] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/03/2014] [Accepted: 12/07/2014] [Indexed: 12/13/2022]
Abstract
Nonalcoholic steatohepatitis (NASH) enhances the risk of progressive liver disease. In chronic hepatitis C (CHC), liver steatosis is frequent, especially in genotype 3, but its clinical significance is debated. As squamous cell carcinoma antigen (SCCA)-IgM has been associated with advanced liver disease and risk of tumour development, we evaluated its occurrence in CHC and the possible relation with NASH at liver biopsy. Using a validated ELISA, serum SCCA-IgM was measured in 91 patients with CHC at the time of liver biopsy performed before antiviral treatment, at the end of treatment and 6 months thereafter, and in 93 HCV-negative patients with histological diagnosis of nonalcoholic fatty liver disease, as controls. SCCA-IgM was detected in 33% of CHC patients and in 4% of controls. This biomarker was found more elevated in CHC patients with histological NASH, and at multivariate analysis, SCCA-IgM and HCV genotype 3 were independently associated with NASH [OR (95% CI): 6.94 (1.21-40) and 27.02 (4.44-166.6)]. As predictors of NASH, HCV genotype 3 and SCCA-IgM had a specificity and a sensitivity of 97% and 44%, and of 95% and 27%, respectively. PPV and NPV were 80% and 86% for HCV genotype 3 vs 73% and 72% for SCCA-IgM. In patients with sustained virologic response to therapy, SCCA-IgM levels decreased significantly, while these remained unchanged in nonresponders. In conclusion, SCCA-IgM is detectable in one-third of patients with CHC and significantly correlates with histological NASH.
Collapse
Affiliation(s)
- A Martini
- Department of Medicine, University of Padua, Padua, Italy
| | - G Fattovich
- Division of Gastroenterology and Endoscopy, Azienda Ospedaliera Universitaria Integrata, Verona, Italy.,Department of Medicine, University of Verona, Verona, Italy
| | - M Guido
- Department of Medicine, University of Padua, Padua, Italy
| | - E Bugianesi
- Division of Gastroenterology and Hepatology, Department of Medical Sciences, University of Torino, Torino, Italy
| | - A Biasiolo
- Department of Medicine, University of Padua, Padua, Italy
| | - D Ieluzzi
- Division of Gastroenterology and Endoscopy, Azienda Ospedaliera Universitaria Integrata, Verona, Italy
| | | | | | - C Merkel
- Department of Medicine, University of Padua, Padua, Italy
| | - A Gatta
- Department of Medicine, University of Padua, Padua, Italy
| | - F Negro
- Division of Clinical Pathology, University Hospitals of Geneva, Geneva, Switzerland.,Division Gastroenterology and Hepatology, University Hospitals of Geneva, Geneva, Switzerland
| | - P Pontisso
- Department of Medicine, University of Padua, Padua, Italy
| |
Collapse
|
86
|
Rüeger S, Bochud PY, Dufour JF, Müllhaupt B, Semela D, Heim MH, Moradpour D, Cerny A, Malinverni R, Booth DR, Suppiah V, George J, Argiro L, Halfon P, Bourlière M, Talal AH, Jacobson IM, Patin E, Nalpas B, Poynard T, Pol S, Abel L, Kutalik Z, Negro F. Impact of common risk factors of fibrosis progression in chronic hepatitis C. Gut 2015; 64:1605-15. [PMID: 25214320 DOI: 10.1136/gutjnl-2014-306997] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/19/2014] [Accepted: 08/22/2014] [Indexed: 12/15/2022]
Abstract
OBJECTIVE The natural course of chronic hepatitis C varies widely. To improve the profiling of patients at risk of developing advanced liver disease, we assessed the relative contribution of factors for liver fibrosis progression in hepatitis C. DESIGN We analysed 1461 patients with chronic hepatitis C with an estimated date of infection and at least one liver biopsy. Risk factors for accelerated fibrosis progression rate (FPR), defined as ≥ 0.13 Metavir fibrosis units per year, were identified by logistic regression. Examined factors included age at infection, sex, route of infection, HCV genotype, body mass index (BMI), significant alcohol drinking (≥ 20 g/day for ≥ 5 years), HIV coinfection and diabetes. In a subgroup of 575 patients, we assessed the impact of single nucleotide polymorphisms previously associated with fibrosis progression in genome-wide association studies. Results were expressed as attributable fraction (AF) of risk for accelerated FPR. RESULTS Age at infection (AF 28.7%), sex (AF 8.2%), route of infection (AF 16.5%) and HCV genotype (AF 7.9%) contributed to accelerated FPR in the Swiss Hepatitis C Cohort Study, whereas significant alcohol drinking, anti-HIV, diabetes and BMI did not. In genotyped patients, variants at rs9380516 (TULP1), rs738409 (PNPLA3), rs4374383 (MERTK) (AF 19.2%) and rs910049 (major histocompatibility complex region) significantly added to the risk of accelerated FPR. Results were replicated in three additional independent cohorts, and a meta-analysis confirmed the role of age at infection, sex, route of infection, HCV genotype, rs738409, rs4374383 and rs910049 in accelerating FPR. CONCLUSIONS Most factors accelerating liver fibrosis progression in chronic hepatitis C are unmodifiable.
Collapse
Affiliation(s)
- S Rüeger
- Department of Medical Genetics, University of Lausanne, Lausanne, Switzerland Institute of Social and Preventive Medicine, University Hospital and University of Lausanne, Lausanne, Switzerland Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - P-Y Bochud
- Infectious Diseases Service, University Hospital and University of Lausanne, Lausanne, Switzerland
| | - J-F Dufour
- Department of Hepatology, University of Berne, Berne, Switzerland
| | - B Müllhaupt
- Department of Gastroenterology and Hepatology, University Hospital of Zurich, Zurich, Switzerland
| | - D Semela
- Department of Gastroenterology, Canton Hospital St Gallen, St Gallen, Switzerland
| | - M H Heim
- Department of Gastroenterology and Hepatology, University Hospital of Basel, Basel, Switzerland
| | - D Moradpour
- Department of Gastroenterology and Hepatology, University Hospital and University of Lausanne, Lausanne, Switzerland
| | - A Cerny
- Epatologia, Clinica Moncucco, Lugano, Switzerland
| | | | - D R Booth
- Institute for Immunology and Allergy Research, Westmead Millennium Institute, University of Sydney, Sydney, Australia
| | - V Suppiah
- Institute for Immunology and Allergy Research, Westmead Millennium Institute, University of Sydney, Sydney, Australia Storr Liver Unit, Westmead Millennium Institute and Westmead Hospital, University of Sydney and University of Sydney Medical Foundation, Sydney, Australia
| | - J George
- Storr Liver Unit, Westmead Millennium Institute and Westmead Hospital, University of Sydney and University of Sydney Medical Foundation, Sydney, Australia
| | - L Argiro
- Laboratoire d'Immunologie et de Génétique des Maladies Parasitaires, INSERM-UMR 906/Université de la Méditerranée, Marseilles, France
| | - P Halfon
- Laboratoire Alphabio, Hôpital Ambroise Paré, Marseilles, France
| | - M Bourlière
- Service d'Hépato-gastroentérologie, Hôpital Saint-Joseph, Marseilles, France
| | - A H Talal
- Division of Gastroenterology and Hepatology, Weill Cornell Medical College, New York, USA
| | - I M Jacobson
- Division of Gastroenterology and Hepatology, Weill Cornell Medical College, New York, USA
| | - E Patin
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM, U980, Imagine Institute, Paris, France University Paris Descartes, Paris, France
| | - B Nalpas
- University Paris Descartes, Paris, France Département d'Hépatologie, INSERM Unité 1016, Groupe Hospitalier Cochin-Hôtel Dieu-Broca, Paris, France
| | - T Poynard
- Université Pierre et Marie Curie, Service d'Hépato-gastroentérologie, Hôpital Pitié-Salpêtrière (AP-HP), Paris, France
| | - S Pol
- University Paris Descartes, Paris, France Département d'Hépatologie, INSERM Unité 1016, Groupe Hospitalier Cochin-Hôtel Dieu-Broca, Paris, France
| | - L Abel
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM, U980, Imagine Institute, Paris, France University Paris Descartes, Paris, France St Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, New York, USA
| | - Z Kutalik
- Institute of Social and Preventive Medicine, University Hospital and University of Lausanne, Lausanne, Switzerland Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - F Negro
- Clinical Pathology, University Hospitals of Geneva, Geneva, Switzerland Department of Gastroenterology and Hepatology, University Hospitals of Geneva, Geneva, Switzerland
| |
Collapse
|
87
|
Hepatitis C Virus Deletion Mutants Are Found in Individuals Chronically Infected with Genotype 1 Hepatitis C Virus in Association with Age, High Viral Load and Liver Inflammatory Activity. PLoS One 2015; 10:e0138546. [PMID: 26405760 PMCID: PMC4583497 DOI: 10.1371/journal.pone.0138546] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2015] [Accepted: 09/01/2015] [Indexed: 01/08/2023] Open
Abstract
Hepatitis C virus (HCV) variants characterized by genomic deletions in the structural protein region have been sporadically detected in liver and serum of hepatitis C patients. These defective genomes are capable of autonomous RNA replication and are packaged into infectious viral particles in cells co-infected with the wild-type virus. The prevalence of such forms in the chronically HCV-infected population and the impact on the severity of liver disease or treatment outcome are currently unknown. In order to determine the prevalence of HCV defective variants and to study their association with clinical characteristics, a screening campaign was performed on pre-therapy serum samples from a well-characterized cohort of previously untreated genotype 1 HCV-infected patients who received treatment with PEG-IFNα and RBV. 132 subjects were successfully analyzed for the presence of defective species exploiting a long-distance nested PCR assay. HCV forms with deletions predominantly affecting E1, E2 and p7 proteins were found in a surprising high fraction of the subjects (25/132, 19%). Their presence was associated with patient older age, higher viral load and increased necroinflammatory activity in the liver. While the presence of circulating HCV carrying deletions in the E1-p7 region did not appear to significantly influence sustained virological response rates to PEG-IFNα/RBV, our study indicates that the presence of these subgenomic HCV mutants could be associated with virological relapse in patients who did not have detectable viremia at the end of the treatment.
Collapse
|
88
|
Lin MS, Lin HS, Chung CM, Lin YS, Chen MY, Chen PH, Hu JH, Chou WN, Huang JC, Huang TJ. Serum aminotransferase ratio is independently correlated with hepatosteatosis in patients with HCV: a cross-sectional observational study. BMJ Open 2015; 5:e008797. [PMID: 26369802 PMCID: PMC4577874 DOI: 10.1136/bmjopen-2015-008797] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/18/2015] [Revised: 08/06/2015] [Accepted: 08/21/2015] [Indexed: 12/19/2022] Open
Abstract
OBJECTIVES The incidence of non-alcoholic fatty liver disease (NAFLD) is significant in hepatitis C virus (HCV) carriers due to multiple mechanisms, and this worsens the progression of chronic liver diseases, such as cirrhosis and hepatocellular carcinoma, and death. The purpose of this study was to examine whether the alanine aminotransferase/aspartate aminotransferase (ALT/AST) ratio correlates with the status of hepatosteatosis. DESIGN A cross-sectional observational study. SETTING Community-based annual examination in northern Taiwan. PARTICIPANTS A total of 1354 participants (age 20 years or over) were enrolled after excluding participants with HCV seronegative, laboratory or questionnaires loss, moderate alcohol consumption, liver cirrhosis, tumours and postlobectomy. OUTCOME MEASURES Fatty liver was diagnosed according to echogenic findings. NAFLD included grades 1-3 fatty liver and high-degree NAFLD defined grades 2-3 fatty liver. RESULTS 580 males and 774 females with a mean age of 47.2 (SD=16.1) years were cross-sectionally studied. The participants with NAFLD have significantly higher levels of ALT/AST ratio, fasting glucose, triglyceride and systolic/diastolic blood pressure than non-NAFLD participants. The association between NAFLD and ALT/AST was significant even when adjusting for the metabolic syndrome (aOR 1.90; 95% CI 1.37 to 2.65; p<0.001). In patients with a high degree of NAFLD, the ALT/AST ratio was still a significant predictor for hepatosteatosis (aOR 2.44; 95% CI 1.58 to 3.77; p<0.001). CONCLUSIONS The ALT/AST ratio could be a strong risk of hepatosteatosis in patients with chronic HCV infection.
Collapse
Affiliation(s)
- Ming-Shyan Lin
- Division of Cardiology, Chang Gung Memorial Hospital, Yunlin, Taiwan
| | - Huang-Shen Lin
- Division of Infectious Diseases, Chang Gung Memorial Hospital, Chiayi, Taiwan
| | - Chang-Ming Chung
- Division of Cardiology, Chang Gung Memorial Hospital, Chiayi, Taiwan
| | - Yu-Sheng Lin
- Division of Cardiology, Chang Gung Memorial Hospital, Chiayi, Taiwan
| | - Mei-Yen Chen
- School of Nursing, Chang Gung University of Science and Technology, Chiayi, Taiwan
| | - Po-Han Chen
- Division of Orthopedic, Chang Gung Memorial Hospital, Yunlin, Taiwan
| | - Jing-Hong Hu
- Division of Gastroenterology and Hepatology, Chang Gung Memorial Hospital, Yunlin, Taiwan
| | - Wen-Nan Chou
- Division of Gastroenterology and Hepatology, Chang Gung Memorial Hospital, Yunlin, Taiwan
| | - Jui-Chu Huang
- Department of Internal Medicine, Chang Gung Memorial Hospital, Yunlin, Taiwan
| | - Tung-Jung Huang
- Department of Internal Medicine, Chang Gung Memorial Hospital, Yunlin, Taiwan
| |
Collapse
|
89
|
Dyal HK, Aguilar M, Bhuket T, Liu B, Holt EW, Torres S, Cheung R, Wong RJ. Concurrent Obesity, Diabetes, and Steatosis Increase Risk of Advanced Fibrosis Among HCV Patients: A Systematic Review. Dig Dis Sci 2015; 60:2813-24. [PMID: 26138651 DOI: 10.1007/s10620-015-3760-3] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/19/2015] [Accepted: 06/10/2015] [Indexed: 12/12/2022]
Abstract
BACKGROUND Rising rates of obesity, diabetes mellitus (DM), and nonalcoholic fatty liver disease among patients with chronic hepatitis C virus infection (HCV) may contribute to more rapid disease progression. AIM To evaluate the impact of concurrent obesity, DM, and steatosis on disease progression among HCV patients. METHODS A systematic review using structured keyword search of MEDLINE and EMBASE from January 1, 2001, to July 1, 2014, was performed to identify original articles evaluating the association of obesity, DM, and steatosis with advanced fibrosis (AF) among adults with chronic HCV. Studies involving HCV patients coinfected with human immunodeficiency virus, hepatitis B virus, hepatocellular carcinoma, or other chronic liver diseases were excluded. Quality assessment utilized Newcastle-Ottawa Scale. RESULTS Twenty cohort studies met inclusion criteria for analyses. Obesity was associated with increased risk of AF in seven studies with effect size ranging from OR 1.08 to 7.69. However, four studies did not demonstrate a significant association between obesity and AF. The presence of advanced steatosis among HCV patients was associated with increased risk of AF in 12 studies (OR 1.80-14.3). Concurrent DM was associated with increased risk of AF in six studies (OR 2.25-9.24). Thirteen studies were good quality, and seven studies were fair quality. CONCLUSION Concurrent DM and steatosis are associated with increased risk of AF among chronic HCV patients. The majority of studies demonstrated significant associations of obesity with AF. Targeted interventions to optimize management of obesity-related diseases among HCV patients may help mitigate HCV disease progression.
Collapse
Affiliation(s)
- Harleen K Dyal
- Division of Gastroenterology and Hepatology, Highland Hospital, Alameda Health System - Highland Hospital Campus, Highland Care Pavilion 5th Floor, Endoscopy Unit, 1411 East 31st Street, Oakland, CA, 94602, USA,
| | | | | | | | | | | | | | | |
Collapse
|
90
|
Cheetham TC, Niu F, Chiang K, Yuan Y, Kalsekar A, Hechter R, Hay JW, Nyberg L. Factors Associated with Failure to Achieve SVR in Hepatitis C Genotype 3 Patients Within an Integrated Care Delivery System. J Manag Care Spec Pharm 2015; 21:641-7. [PMID: 26233536 PMCID: PMC10398146 DOI: 10.18553/jmcp.2015.21.8.641] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
BACKGROUND Achievement of sustained virologic response (SVR) and factors associated with treatment failure in hepatitis C virus (HCV) genotype 3 have been described in tertiary and referral care settings, with rates of SVR reported to range between 72% and 89%. Fewer data exist on SVR outside of these settings. OBJECTIVE To describe rates of SVR and characterize factors associated with achievement of SVR within an integrated health care delivery system. METHODS A retrospective cohort study of genotype 3 HCV patients treated with dual therapy (pegylated interferon-alpha plus ribavirin) was conducted at Kaiser Permanente Southern California. Adult patients diagnosed with HCV and testing positive for HCV-RNA genotype 3 were identified from electronic medical records. Data were collected on patient demographics, baseline health status, and comorbid conditions. A multivariate logistic regression model was used to determine the association between baseline patient factors and SVR. RESULTS A total of 484 HCV genotype 3 patients met the eligibility criteria. The median age was 49 years, and 65.7% were male. Overall, 252 (52.1%) achieved SVR. Aged ≥ 45 years and male gender were associated with lower rates of SVR; cirrhosis and chronic diseases (diabetes and chronic obstructive pulmonary disease) were also associated with lower rates of SVR. CONCLUSIONS SVR was lower in patients within an integrated care delivery system than in those in tertiary and referral centers. Males and older patients had lower rates of SVR, as well as patients with cirrhosis, diabetes, and chronic obstructive pulmonary disease.
Collapse
Affiliation(s)
- T Craig Cheetham
- Southern California Permanente Medical Group, 100 S. Los Robles Ave., 2nd Fl., Pasadena, CA 91101-2453.
| | | | | | | | | | | | | | | |
Collapse
|
91
|
Lawitz E, Jacobson IM, Nelson DR, Zeuzem S, Sulkowski MS, Esteban R, Brainard D, McNally J, Symonds WT, McHutchison JG, Dieterich D, Gane E. Development of sofosbuvir for the treatment of hepatitis C virus infection. Ann N Y Acad Sci 2015; 1358:56-67. [PMID: 26235748 DOI: 10.1111/nyas.12832] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
The nucleotide analog NS5B polymerase inhibitor sofosbuvir was approved by the U.S. Food and Drug Administration (FDA) in December 2013 for the treatment of chronic hepatitis C virus (HCV) infection in combination with ribavirin or peginterferon and ribavirin. Sofosbuvir was developed to meet an urgent medical need for shorter, safer, simplified, more effective HCV treatment regimens and to reduce or eliminate the need for peginterferon. New treatment regimens were especially required for patient populations with limited treatment options, including patients who had failed prior HCV therapy, those with compensated and decompensated cirrhosis, and those who were either intolerant of or had contraindications to interferon. Sofosbuvir plus ribavirin for patients with genotype 2 or 3 HCV infection was the first approved all-oral treatment option. Sofosbuvir is also the backbone of the first regimen available for patients awaiting liver transplantation to prevent HCV recurrence, as well as the first oral interferon-free regimen for patients coinfected with HCV and HIV. This paper describes the development of sofosbuvir up to its original FDA approval.
Collapse
Affiliation(s)
- Eric Lawitz
- Texas Liver Institute, University of Texas Health Science Center, San Antonio, Texas
| | - Ira M Jacobson
- Icahn School of Medicine at Mount Sinai, New York, New York
| | | | - Stefan Zeuzem
- Medical Center, Johann Wolfgang Goethe University, Frankfurt am Main, Germany
| | - Mark S Sulkowski
- Johns Hopkins University School of Medicine, Baltimore, Maryland
| | | | | | | | | | | | | | - Edward Gane
- Auckland City Hospital, Auckland, New Zealand
| |
Collapse
|
92
|
Eradikation des Hepatitis-C-Virus und Verhinderung klinischer Endpunkte. GASTROENTEROLOGE 2015. [DOI: 10.1007/s11377-015-0995-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
|
93
|
De Clercq E. Development of antiviral drugs for the treatment of hepatitis C at an accelerating pace. Rev Med Virol 2015; 25:254-67. [DOI: 10.1002/rmv.1842] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2015] [Revised: 05/04/2015] [Accepted: 05/05/2015] [Indexed: 02/06/2023]
Affiliation(s)
- Erik De Clercq
- Rega Institute for Medical Research; KU Leuven; Leuven Belgium
| |
Collapse
|
94
|
Abstract
Hepatitis C virus (HCV) is a leading etiology of hepatocellular carcinoma (HCC). The interaction of HCV with its human host is complex and multilayered; stemming in part from the fact that HCV is a RNA virus with no ability to integrate in the host's genome. Direct and indirect mechanisms of HCV-induced HCC include activation of multiple host pathways such as liver fibrogenic pathways, cellular and survival pathways, interaction with the immune and metabolic systems. Host factors also play a major role in HCV-induced HCC as evidenced by genomic studies identifying polymorphisms in immune, metabolic, and growth signaling systems associated with increased risk of HCC. Despite highly effective direct-acting antiviral agents, the morbidity and incidence of liver-related complications of HCV, including HCC, is likely to persist in the near future. Clinical markers to selectively identify HCV subjects at higher risk of developing HCC have been reported however they require further validation, especially in subjects who have experienced sustained virological response. Molecular biomarkers allowing further refinement of HCC risk are starting to be implemented in clinical platforms, allowing objective stratification of risk and leading to individualized therapy and surveillance for HCV individuals. Another role for molecular biomarker-based stratification could be enrichment of HCC chemoprevention clinical trials leading to smaller sample size, shorter trial duration, and reduced costs.
Collapse
Affiliation(s)
- Nicolas Goossens
- Division of Liver Diseases, Department of Medicine, Liver Cancer Program, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, USA
- Division of Gastroenterology and Hepatology, Geneva University Hospital, Geneva, Switzerland
| | - Yujin Hoshida
- Division of Liver Diseases, Department of Medicine, Liver Cancer Program, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, USA
| |
Collapse
|
95
|
Janardhan SV, Reau NS. Should NS5A inhibitors serve as the scaffold for all-oral anti-HCV combination therapies? ACTA ACUST UNITED AC 2015; 7:11-20. [PMID: 25926761 PMCID: PMC4403691 DOI: 10.2147/hmer.s79584] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Chronic hepatitis C virus (HCV) infection represents a global health problem that affects up to 130–150 million people worldwide. The HCV treatment landscape has been transformed recently by the introduction of direct-acting antiviral (DAA) agents that target viral proteins, including the NS3 protease, the NS5B polymerase, and the NS5A protein. Treatment with multiple DAAs in combination has been shown to result in high rates of sustained virologic response, without the need for pegylated interferon, and a shorter duration of therapy compared with interferon-based regimens; however, the optimal combination of DAAs has yet to be determined. The class of NS5A inhibitors has picomolar potency with pangenotypic activity, and recent clinical studies have shown these inhibitors to be an important component of DAA combination regimens. This review discusses the rational design of an optimal anti-HCV DAA cocktail, with a focus on the role of NS5A in the HCV life cycle, the attributes of the NS5A class of inhibitors, and the potential for NS5A inhibitors to act as a scaffold for DAA-only treatment regimens.
Collapse
Affiliation(s)
- Sujit V Janardhan
- Center for Liver Diseases, Section of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, University of Chicago, Chicago, IL, USA
| | - Nancy S Reau
- Center for Liver Diseases, Section of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, University of Chicago, Chicago, IL, USA
| |
Collapse
|
96
|
Bridge SH, Sheridan DA, Felmlee DJ, Crossey MME, Fenwick FI, Lanyon CV, Dubuc G, Seidah NG, Davignon J, Thomas HC, Taylor-Robinson SD, Toms GL, Neely RDG, Bassendine MF. PCSK9, apolipoprotein E and lipoviral particles in chronic hepatitis C genotype 3: evidence for genotype-specific regulation of lipoprotein metabolism. J Hepatol 2015; 62:763-70. [PMID: 25463543 DOI: 10.1016/j.jhep.2014.11.016] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2014] [Revised: 10/28/2014] [Accepted: 11/12/2014] [Indexed: 02/07/2023]
Abstract
BACKGROUND & AIMS Hepatitis C virus (HCV) associates with lipoproteins to form "lipoviral particles" (LVPs) that can facilitate viral entry into hepatocytes. Initial attachment occurs via heparan sulphate proteoglycans and low-density lipoprotein receptor (LDLR); CD81 then mediates a post-attachment event. Proprotein convertase subtilisin kexin type 9 (PCSK9) enhances the degradation of the LDLR and modulates liver CD81 levels. We measured LVP and PCSK9 in patients chronically infected with HCV genotype (G)3. PCSK9 concentrations were also measured in HCV-G1 to indirectly examine the role of LDLR in LVP clearance. METHODS HCV RNA, LVP (d<1.07g/ml) and non-LVP (d>1.07g/ml) fractions, were quantified in patients with HCV-G3 (n=39) by real time RT-PCR and LVP ratios (LVPr; LVP/(LVP+non-LVP)) were calculated. Insulin resistance (IR) was assessed using the homeostasis model assessment of IR (HOMA-IR). Plasma PCSK9 concentrations were measured by ELISA in HCV-G3 and HCV-G1 (n=51). RESULTS In HCV-G3 LVP load correlated inversely with HDL-C (r=-0.421; p=0.008), and apoE (r=-0.428; p=0.013). The LVPr varied more than 35-fold (median 0.286; range 0.027 to 0.969); PCSK9 was the strongest negative predictor of LVPr (R(2)=16.2%; p=0.012). HOMA-IR was not associated with LVP load or LVPr. PCSK9 concentrations were significantly lower in HCV-G3 compared to HCV-G1 (p<0.001). PCSK9 did not correlate with LDL-C in HCV-G3 or G1. CONCLUSIONS The inverse correlation of LVP with apoE in HCV-G3, compared to the reverse in HCV-G1 suggests HCV genotype-specific differences in apoE mediated viral entry. Lower PCSK9 and LDL concentrations imply upregulated LDLR activity in HCV-G3.
Collapse
Affiliation(s)
- Simon H Bridge
- Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, United Kingdom; Faculty of Health and Life Sciences, Northumbria University, Newcastle upon Tyne, United Kingdom.
| | - David A Sheridan
- Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, United Kingdom; Institute of Translational & Stratified Medicine, Plymouth University Peninsula School of Medicine & Dentistry, United Kingdom
| | - Daniel J Felmlee
- Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, United Kingdom; Inserm U1110, University of Strasbourg and Center for Liver and Digestive Diseases, Strasbourg University Hospitals, 3 Rue Koeberlé, F-67000 Strasbourg, France
| | - Mary M E Crossey
- Liver Unit, Department of Medicine, Imperial College London, St Mary's Hospital Campus, Praed Street, London, United Kingdom
| | - Fiona I Fenwick
- Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Clare V Lanyon
- Faculty of Health and Life Sciences, Northumbria University, Newcastle upon Tyne, United Kingdom
| | - Geneviève Dubuc
- Hyperlipidemia and Atherosclerosis Research Group, Clinical Research Institute of Montréal (IRCM), Montréal, Canada; University of Montréal, Montréal, Canada
| | - Nabil G Seidah
- Laboratory of Biochemical Neuroendocrinology, Clinical Research Institute of Montréal, Montréal, Canada; University of Montréal, Montréal, Canada
| | - Jean Davignon
- Hyperlipidemia and Atherosclerosis Research Group, Clinical Research Institute of Montréal (IRCM), Montréal, Canada; University of Montréal, Montréal, Canada
| | - Howard C Thomas
- Liver Unit, Department of Medicine, Imperial College London, St Mary's Hospital Campus, Praed Street, London, United Kingdom
| | - Simon D Taylor-Robinson
- Liver Unit, Department of Medicine, Imperial College London, St Mary's Hospital Campus, Praed Street, London, United Kingdom
| | - Geoffrey L Toms
- Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - R Dermot G Neely
- Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, United Kingdom; Department of Clinical Biochemistry, Newcastle upon Tyne Hospitals NHS Foundation Trust, Royal Victoria Infirmary, United Kingdom
| | - Margaret F Bassendine
- Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, United Kingdom.
| |
Collapse
|
97
|
Impairment of type I but not type III IFN signaling by hepatitis C virus infection influences antiviral responses in primary human hepatocytes. PLoS One 2015; 10:e0121734. [PMID: 25826356 PMCID: PMC4380495 DOI: 10.1371/journal.pone.0121734] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2014] [Accepted: 02/13/2015] [Indexed: 12/15/2022] Open
Abstract
Peginterferon lambda-1a (Lambda), a type III interferon (IFN), acts through a unique receptor complex with limited cellular expression outside the liver which may result in a differentiated tolerability profile compared to peginterferon alfa (alfa). In Phase 2b clinical studies, Lambda administered in combination with ribavirin (RBV) was efficacious in patients with hepatitis C virus (HCV) infection representing genotypes 1 through 4, and was associated with more rapid declines in HCV RNA compared to alfa plus RBV. To gain insights into potential mechanisms for this finding, we investigated the effects of HCV replication on IFN signaling in primary human hepatocytes (PHH) and in induced hepatocyte-like cells (iHLCs). HCV infection resulted in rapid down-regulation of the type I IFN-α receptor subunit 1 (IFNAR1) transcript in hepatocytes while the transcriptional level of the unique IFN-λ receptor subunit IL28RA was transiently increased. In line with this observation, IFN signaling was selectively impaired in infected cells upon stimulation with alfa but not in response to Lambda. Importantly, in contrast to alfa, Lambda was able to induce IFN-stimulated gene (ISG) expression in HCV-infected hepatocytes, reflecting the onset of innate responses. Moreover, global transcriptome analysis in hepatocytes indicated that Lambda stimulation prolonged the expression of various ISGs that are potentially beneficial to antiviral defense mechanisms. Collectively, these observed effects of HCV infection on IFN receptor expression and signaling within infected hepatocytes provide a possible explanation for the more pronounced early virologic responses observed in patients treated with Lambda compared to alfa.
Collapse
|
98
|
Lawitz E, Poordad F, Brainard DM, Hyland RH, An D, Dvory-Sobol H, Symonds WT, McHutchison JG, Membreno FE. Sofosbuvir with peginterferon-ribavirin for 12 weeks in previously treated patients with hepatitis C genotype 2 or 3 and cirrhosis. Hepatology 2015; 61:769-75. [PMID: 25322962 PMCID: PMC4365682 DOI: 10.1002/hep.27567] [Citation(s) in RCA: 100] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2014] [Accepted: 10/15/2014] [Indexed: 02/06/2023]
Abstract
UNLABELLED Sofosbuvir (SOF) in combination with ribavirin (RBV) for 12 or 24 weeks is the current standard of care for patients infected with hepatitis C virus (HCV) genotypes 2 and 3, respectively. However, in clinical trials treatment-experienced patients, particularly those with cirrhosis, had suboptimal sustained virological response (SVR) rates. We assessed the efficacy and safety of sofosbuvir plus peginterferon and ribavirin (SOF+Peg-IFN+RBV) administered for 12 weeks to treatment-experienced patients with HCV genotypes 2 and 3, with and without cirrhosis. We enrolled 47 patients in this open-label, nonrandomized, uncontrolled phase 2 study. The primary endpoint was the proportion of patients with SVR at 12 weeks after cessation of study treatment (SVR12). The overall rate of SVR12 was 89% (95% confidence interval [CI]: 77-97). Rates of SVR12 were higher in patients with genotype 2 than in those with genotype 3, 96% (95% CI: 78-100) and 83% (95% CI: 62-95), respectively. Rates of SVR12 were similar in patients with and without cirrhosis: for genotype 2, 93% of patients with cirrhosis and 100% of patients without cirrhosis achieved SVR12, and for genotype 3, the SVR12 rate was 83% in patients both with and without cirrhosis. One patient discontinued study treatment because of an adverse event and four patients experienced serious adverse events. The most common adverse events were influenza-like illness, fatigue, anemia, and neutropenia. CONCLUSION In treatment-experienced patients with HCV genotypes 2 and 3, 12-week administration of SOF+Peg-IFN+RBV provided high SVR rates, irrespective of cirrhosis status. No safety concerns were identified.
Collapse
Affiliation(s)
- Eric Lawitz
- Texas Liver InstituteSan Antonio, TX
- University of Texas Health Science CenterSan Antonio, TX
| | - Fred Poordad
- Texas Liver InstituteSan Antonio, TX
- University of Texas Health Science CenterSan Antonio, TX
| | | | | | - Di An
- Gilead SciencesFoster City, CA
| | | | | | | | - Fernando E Membreno
- Texas Transplant Institute, Methodist Specialty and Transplant HospitalSan Antonio, TX
| |
Collapse
|
99
|
Meissner EG, Lee YJ, Osinusi A, Sims Z, Qin J, Sturdevant D, McHutchison J, Subramanian M, Sampson M, Naggie S, Patel K, Remaley AT, Masur H, Kottilil S. Effect of sofosbuvir and ribavirin treatment on peripheral and hepatic lipid metabolism in chronic hepatitis C virus, genotype 1-infected patients. Hepatology 2015; 61:790-801. [PMID: 25203718 PMCID: PMC4340816 DOI: 10.1002/hep.27424] [Citation(s) in RCA: 119] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/18/2014] [Accepted: 09/03/2014] [Indexed: 12/23/2022]
Abstract
UNLABELLED Hepatitis C virus (HCV) modulates intrahepatic cholesterol biosynthetic pathways to promote viral replication. Chronic HCV infection is associated with altered metabolism, including dyslipidemia and insulin resistance (IR), which contributes to disease progression and influences response to therapy. To further understand the impact of HCV infection on host metabolism, we examined changes in serum lipid profiles and intrahepatic expression of lipid-related genes during interferon (IFN)-free treatment of chronic HCV, genotype 1 infection with sofosbuvir and ribavirin (RBV), and explored associations with treatment outcome. Serum lipids (total cholesterol, low-density lipoprotein [LDL], high-density lipoprotein [HDL], and triglycerides [TGs]) and hemoglobin A1C (HbA1C) were measured during treatment, while gene expression of lipid-related genes was assessed using paired pre- and end-of-treatment (EOT) liver biopsies from 8 patients (n=7 sustained virologic response [SVR]; n=1 relapse) and unpaired EOT liver biopsies from 25 patients (n=17 SVR; n=8 relapse). Serum LDL concentration and particle size increased early in therapy, whereas TG concentration and very-low-density lipoprotein particle size decreased concomitantly, irrespective of treatment outcome. Whereas LDL increased in patients regardless of treatment outcome, average LDL concentration was lower at baseline and post-treatment in patients who relapsed. Analysis of paired liver biopsies revealed altered expression of genes associated with lipid transport, assembly, and signaling. In unpaired EOT liver biopsies, intrahepatic expression of fatty acid metabolism and lipid transport genes was lower in patients who experienced treatment relapse. CONCLUSION Clearance of HCV using an IFN-free antiviral regimen results in rapid changes in peripheral and intrahepatic metabolic pathways, implicating a direct effect of HCV replication on lipid homeostasis.
Collapse
Affiliation(s)
- Eric G. Meissner
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Yu-Jin Lee
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Anu Osinusi
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
- Division of Infectious Diseases, Institute of Human Virology, University of Maryland Medical School, Baltimore, MD, USA
| | - Zayani Sims
- Critical Care Medicine Department, Clinical Center, National Institutes of Health, Bethesda, MD, USA
| | - Jing Qin
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Dan Sturdevant
- Genomics Unit, Research Technologies Section, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA
| | | | | | - Maureen Sampson
- Lipoprotein Metabolism Section, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Susanna Naggie
- Duke University, Duke Clinical Research Institute, Durham, NC, USA
| | - Keyur Patel
- Duke University, Duke Clinical Research Institute, Durham, NC, USA
| | - Alan T. Remaley
- Lipoprotein Metabolism Section, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Henry Masur
- Critical Care Medicine Department, Clinical Center, National Institutes of Health, Bethesda, MD, USA
| | - Shyam Kottilil
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
100
|
Abstract
Hepatitis C virus (HCV) is a global health burden with an estimated 170-200 million peoples chronically infected worldwide. HCV infection remains as an independent risk factor for chronic hepatitis, liver cirrhosis, hepatocellular carcinoma, and a major reason for liver transplantation. Discovery of direct acting antiviral (DAA) drugs have shown promising results with more than 90% success rate in clearing the HCV RNA in patients, although long-term consequences remain to be evaluated. microRNAs (miRNAs) are important players in establishment of HCV infection and target crucial host cellular factors needed for productive HCV replication and augmented cell growth. Altered expression of miRNAs is involved in the pathogenesis associated with HCV infection by controlling signaling pathways such as immune response, proliferation and apoptosis. miRNA is emerging as a means of communication between various cell types inside the liver. There is likely possibility of developing circulating miRNAs as biomarkers of disease progression and can also serve as diagnostic tool with potential of early therapeutic intervention in HCV associated end stage liver disease. This review focuses on recent studies highlighting the contribution of miRNAs in HCV life cycle and their coordinated regulation in HCV mediated liver disease progression.
Collapse
Affiliation(s)
| | - Robert Steele
- Departments of Pathology, Saint Louis University, St. Louis, Missouri, USA
| | - Ranjit Ray
- Departments of Internal Medicine, Saint Louis University, St. Louis, Missouri, USA
| | - Ratna B Ray
- Departments of Pathology, Saint Louis University, St. Louis, Missouri, USA
| |
Collapse
|