WO2011128278A1 - Single nucleotide polymorphisms that predict hcv treatment outcomes - Google Patents

Single nucleotide polymorphisms that predict hcv treatment outcomes Download PDF

Info

Publication number
WO2011128278A1
WO2011128278A1 PCT/EP2011/055579 EP2011055579W WO2011128278A1 WO 2011128278 A1 WO2011128278 A1 WO 2011128278A1 EP 2011055579 W EP2011055579 W EP 2011055579W WO 2011128278 A1 WO2011128278 A1 WO 2011128278A1
Authority
WO
WIPO (PCT)
Prior art keywords
interferon
patients
svr
treatment
evr
Prior art date
Application number
PCT/EP2011/055579
Other languages
French (fr)
Inventor
Tom Chu
Laurent Essioux
Soren Germer
Uri Lopatin
Nancy Shulman
James Thommes
Original Assignee
F. Hoffmann-La Roche Ag
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by F. Hoffmann-La Roche Ag filed Critical F. Hoffmann-La Roche Ag
Priority to JP2013504214A priority Critical patent/JP5695181B2/en
Priority to RU2012145777/10A priority patent/RU2567663C2/en
Priority to ES11717518.2T priority patent/ES2525290T3/en
Priority to MX2012011804A priority patent/MX2012011804A/en
Priority to CA2795911A priority patent/CA2795911A1/en
Priority to CN201180018656.0A priority patent/CN102844448B/en
Priority to KR20127029599A priority patent/KR101493137B1/en
Priority to BR112012025854A priority patent/BR112012025854A2/en
Priority to EP11717518.2A priority patent/EP2558592B1/en
Publication of WO2011128278A1 publication Critical patent/WO2011128278A1/en
Priority to HK13107392.4A priority patent/HK1180018A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/68Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving nucleic acids
    • C12Q1/6876Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes
    • C12Q1/6883Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes for diseases caused by alterations of genetic material
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/14Antivirals for RNA viruses
    • A61P31/18Antivirals for RNA viruses for HIV
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/68Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving nucleic acids
    • C12Q1/6876Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes
    • C12Q1/6888Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes for detection or identification of organisms
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/70Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving virus or bacteriophage
    • C12Q1/701Specific hybridization probes
    • C12Q1/706Specific hybridization probes for hepatitis
    • C12Q1/707Specific hybridization probes for hepatitis non-A, non-B Hepatitis, excluding hepatitis D
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q2600/00Oligonucleotides characterized by their use
    • C12Q2600/106Pharmacogenomics, i.e. genetic variability in individual responses to drugs and drug metabolism
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q2600/00Oligonucleotides characterized by their use
    • C12Q2600/156Polymorphic or mutational markers
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y10TECHNICAL SUBJECTS COVERED BY FORMER USPC
    • Y10TTECHNICAL SUBJECTS COVERED BY FORMER US CLASSIFICATION
    • Y10T436/00Chemistry: analytical and immunological testing
    • Y10T436/14Heterocyclic carbon compound [i.e., O, S, N, Se, Te, as only ring hetero atom]
    • Y10T436/142222Hetero-O [e.g., ascorbic acid, etc.]
    • Y10T436/143333Saccharide [e.g., DNA, etc.]

Definitions

  • the present invention relates to methods that useful for predicting the response of hepatitis C virus (HCV) infected patients to pharmacological treatment.
  • HCV hepatitis C virus
  • SVR sustained viro logical response
  • Rapid viro logical response (RVR, undetectable HCV RNA at week 4) is a strong predictor of SVR; conversely, failure to achieve an early viro logical response (EVR, greater than a two log decline in HCV RNA at week 12) is a strong predictor of nonresponse, independent of pretreatment
  • Treatment decisions could be personalized based on the likelihood of patients to respond to the standard of care. For example patients with the lowest likelihood of achieving an SVR with the current standard of care might defer treatment until direct acting antiviral agents are available. Conversely, patients with a high likelihood of achieving an SVR might prefer to initiate therapy immediately with a treatment regimen that is a known entity.
  • SNPs single nucleotide polymorphisms
  • the present invention is based on the discovery of an association between several SNP genotypes on human chromosome four and SVR in patients treated with interferon-based regimens.
  • the invention provides for a method for predicting sustained virological response of a human subject infected with HCV to interferon treatment comprising, providing a sample from said human subject, detecting the presence of a single nucleotide polymorphism within chromosome 4 and determining that said subject has a high likelihood of sustained virological response to interferon treatment if said single nucleotide polymorphism is present, wherein said single nucleotide polymorphism is selected from the group consisting of a G at rsl0009948, a G at rsl0023606, and a T at rs7673763.
  • Figure 1 Genome-wide association results for (a) SVR (b) EVR) and (c) SVR after adjustment for rs 12979860 by chromosome in the overall genotype 1 population.
  • Figure 2. Quantile-quantile plot of the distribution of test statistics for (a) SVR and (b) EVR). Grey circles denote expected p-values and blue circles show observed p values.
  • Viro logical endpoints included “early viro logical response” (EVR), defined as >2-log drop in serum HCV R A from baseline to week 12 (by Cobas Amplicor HCV Monitor Test, v2.0, limit of quantitation 600 IU/mL), complete EVR (cEVR) defined as undetectable HCV RNA in serum (by Cobas Amplicor HCV Test v2.0, limit of detection 50 IU/mL) or and
  • SVR sustained viro logical response
  • sample refers to a sample of tissue or fluid isolated from an individual, including, but not limited to, for example, tissue biopsy, plasma, serum, whole blood, spinal fluid, lymph fluid, the external sections of the skin, respiratory, intestinal and
  • samples of in vitro cell culture constituents including, but not limited to, conditioned medium resulting from the growth of cells in culture medium, putatively virally infected cells, recombinant cells, and cell components).
  • interferon and “interferon-alpha” are used herein interchangeably and refer to the family of highly homologous species-specific proteins that inhibit viral replication and cellular proliferation and modulate immune response.
  • suitable interferons include, but are not limited to, recombinant interferon alpha-2b such as Intron® A interferon available from Schering Corporation, Kenilworth, N.J., recombinant interferon alpha-2a such as Roferon®-A interferon available from Hoffmann-La Roche, Nutley, N.J., recombinant interferon alpha-2C such as Berofor® alpha 2 interferon available from Boehringer Ingelheim Pharmaceutical, Inc.,
  • interferon alpha-nl a purified blend of natural alpha interferons such as Sumiferon® available from Sumitomo, Japan or as Wellferon® interferon alpha-nl (INS) available from the Glaxo-Wellcome Ltd., London, Great Britain, or a consensus alpha interferon such as those described in U.S. Pat. Nos.
  • Interferon alpha-n3 a mixture of natural alpha interferons made by Interferon Sciences and available from the Purdue Frederick Co., Norwalk, Conn., under the Alferon Tradename.
  • the use of interferon alpha-2a or alpha-2b is preferred.
  • Interferons can include pegylated interferons as defined below.
  • pegylated interferon means polyethylene glycol modified conjugates of interferon alpha, preferably interferon alfa-2a and alfa-2b.
  • suitable pegylated interferon alpha include, but are not limited to, Pegasys® and Peg-Intron®.
  • ribavirin refers to the compound, l-((2R,3R,4S,5R)-3,4-Dihydroxy-5- hydroxymethyl-tetrahydro-furan-2-yl)-lH-[l,2,4]triazole-3-carboxylic acid amide which is a synthetic, non-interferon-inducing, broad spectrum antiviral nucleoside analog and available under the names, Virazole® and Copegus® .
  • Direct acting antiviral agents exert specific antiviral effects independent of immune function.
  • Examples of direct acting antiviral agents for HCV include but are limited to protease inhibitors, polymerase inhibitors, NS5A inhibitors, IRES inhibitors and helicase inhibitors.
  • the current recommended first line treatment for patients with chronic hepatitis C is pegylated interferon alpha in combination with ribavirin for 48 weeks in patients carrying genotype 1 or 4 virus and for 24 weeks in patients carrying genotype 2 or 3 virus.
  • Combined treatment with ribavirin was found to be more effective than interferon alpha monotherapy in patients who relapsed after one or more courses of interferon alpha therapy, as well as in previously untreated patients.
  • ribavirin exhibits significant side effects including teratogenicity and carcinogenicity.
  • ribavirin causes hemolytic anemia requiring dose reduction or discontinuation of ribavirin therapy in approximately 10 to 20% of patients, which may be related to the accumulation of ribavirin triphosphate in erythrocytes. Therefore, to reduce treatment cost and the incidence of adverse events, it is desirable to tailor the treatment to a shorter duration while not compromising efficacy.
  • a shortened "duration of treatment" for genotype 1 patients with pegylated interferon alpha with ribovirin would be, for example, 24 weeks.
  • a shortened duration of treatment for genotype 1 patients with pegylated interferon alpha with ribavirin in combination with a direct acting antiviral agent could be as short as 8 weeks, 12 weeks, or 16 weeks.
  • allele and “allelic variant” refer to alternative forms of a gene including introns, exons, intron/exon junctions and 3' and/or 5' untranslated regions that are associated with a gene or portions thereof. Generally, alleles occupy the same locus or position on homologous chromosomes. When a subject has two identical alleles of a gene, the subject is said to be homozygous for the gene or allele. When a subject has two different alleles of a gene, the subject is said to be heterozygous for the gene. Alleles of a specific gene can differ from each other in a single nucleotide, or several nucleotides, and can include substitutions, deletions, and insertions of nucleotides.
  • polymorphism refers to the coexistence of more than one form of a nucleic acid, including exons and introns, or portion (e.g., allelic variant) thereof.
  • a portion of a gene of which there are at least two different forms, i.e., two different nucleotide sequences, is referred to as a polymorphic region of a gene.
  • a polymorphic region can be a single nucleotide, i.e. "single nucleotide polymorphism" or "SNP", the identity of which differs in different alleles.
  • a polymorphic region can also be several nucleotides long.
  • polymorphisms Numerous methods for the detection of polymorphisms are known and may be used in conjunction with the present invention. Generally, these include the identification of one or more mutations in the underlying nucleic acid sequence either directly (e.g., in situ hybridization) or indirectly (identifying changes to a secondary molecule, e.g., protein sequence or protein binding).
  • One well-known method for detecting polymorphisms is allele specific hybridization using probes overlapping the mutation or polymorphic site and having about 5, 10, 20, 25, or 30 nucleotides around the mutation or polymorphic region.
  • probes overlapping the mutation or polymorphic site and having about 5, 10, 20, 25, or 30 nucleotides around the mutation or polymorphic region.
  • a kit e.g., several probes capable of hybridizing specifically to allelic variants, such as single nucleotide polymorphisms, are provided for the user or even attached to a solid phase support, e.g., a bead or chip.
  • rsl2979860 refers to a SNP identified by its accession number in the database of SNPs (dbSNP, www.ncbi. nlm. nih. gov/ SNP/) and is located on human chromosome 19 in the promoter region of the IL28b gene.
  • Interferon lambda is a type III interferon that triggers a signalling pathway that overlaps with the Jak/Stat pathway of type I interferons (including interferon-alpha).
  • HCV RNA is translated via an internal ribosomal entry site and is partially eIF2a- independent.
  • exogenous pegylated interferon can, however, "rescue” patients who are less sensitive to endogenous interferon.
  • the genetic polymorphism may play a role in ISG expression. This leads us to speculate that it may eventually be possible to selectively target the interferon pathway to produce a more favourable ISG profile that results in HC V eradication.
  • SNP in chromosome 4 is associated with SVR, though not with EVR.
  • the simplest explanation might require a gene in this region to be involved in a phenomenon that is exclusively associated with SVR such as hepatocyte turnover. Additional host-host or host-viral gene interactions may be involved in the mechanism by which both loci exert their effects, and require further careful evaluation.
  • SOC standard of care
  • the IL28b genotype will be determined before therapy is initiated and that patients will be stratified not only by HCV genotype - as currently recommended 1 - but also by human genotype.
  • the initial treatment regimen will be based upon both viral and human genotype. The putative role of ribavirin in this context will need to be addressed in prospective trials.
  • HCV R A encodes specific proteins that may inhibit the induction of type I interferons.
  • the NS3-4A protease of HCV blocks dsR A-induced interferon production by interfering with phosphorylation of interferon regulatory factor-3 (IRF-3).
  • the NS3-4A protease is a dual therapeutic target, whose inhibition may block viral replication and restore IRF-3 control of HCV RNA replication.
  • Protease inhibitors such as Telaprevir, which have robust antiviral effects when administered in combination with a second small molecule or the standard of care, also inhibit the protease functions by which HCV impairs host interferon response. It will be important to observe whether treatment outcomes are similar when direct acting antiviral agents are combined with peginterferon plus ribavirin in patients with interferon-refractory and interferon-sensitive IL28b phenotypes.
  • interferon-refractory patients will respond to triple therapy (direct acting antiviral agent plus peginterferon plus ribavirin) as if they were on monotherapy with the direct acting antiviral agent alone. If this is the case then patients with the interferon-refractory IL28b phenotype may be much more susceptible to the selection of resistance mutations during treatment with a drug such as telaprevir. 31 These possibilities suggest scenarios where patients with the interferon-susceptible SNP (rs 12979860) might benefit from abbreviated treatment with peginterferon and ribavirin, and those with an interferon-refractory genotype might be candidates for extended treatment durations and/or more intensive treatment regimens..
  • interferon- free combination regimens of direct acting antiviral agents may be more appropriate for patients with an interferon-refractory genotype.
  • Our unique dataset including a registration trial of peginterferon alfa-2a plus ribavirin therapy for patients who were previous non-responders to standard of care with a pegylated interferon, highlights the correlation of the rsl2979860 SNP with EVR rather than SVR. Stated differently, this SNP defines patient's responsiveness to interferon rather than the ultimate response to therapy and thus helps identify patients likely or unlikely to undergo an SVR because of their likelihood to achieve an EVR. It is unlikely that it predicts SVR independently of EVR. This has tremendous implications for use of direct acting antiviral agents together with interferon.
  • Patients predicted to have acceptable endogenous interferon responsiveness may be excellent candidates for drugs that target viral functions-such as protease inhibitors which also have an inhibitory role on endogenous interferon response- and poorer candidates for drugs that decrease the amounts of viral PAMP (pathogen associated molecular pattern, e.g. polymerase inhibitors) as these may serve to impair the patients capacity to facilitate their own cure via their target viral functions-such as protease inhibitors which also have an inhibitory role on endogenous interferon response- and poorer candidates for drugs that decrease the amounts of viral PAMP (pathogen associated molecular pattern, e.g. polymerase inhibitors) as these may serve to impair the patients capacity to facilitate their own cure via their
  • viral PAMP pathogen associated molecular pattern, e.g. polymerase inhibitors
  • Viro logical endpoints included early viro logical response (EVR), defined as undetectable HCV RNA in serum (by Cobas Amplicor HCV Test v2.0, limit of detection 50 IU/mL) or >2-log drop in serum HCV RNA from baseline to week 12 (by Cobas Amplicor HCV Monitor Test, v2.0, limit of quantitation 600 IU/mL) and sustained virological response (SVR), defined as undetectable HCV RNA ( ⁇ 50 IU/mL) at the end of a 24-week untreated follow-up period.
  • ELR early viro logical response
  • SVR sustained virological response
  • the responder group was composed of all genotype- 1 patients with SVR from the interferon na ' ive study population.
  • Non-responders were composed of 1) all genotype- 1 non-SVR patients from the study population re-challenged by pegylated-Interferon treatment, 2) genotype- 1 non-SVR patients form the interferon na ' ive study population treated with pegylated interferon with ribavirin.
  • the EVR group was composed of all genotype- 1 patients with EVR from the interferon na ' ive study population.
  • the non-EVR group was composed of l)all patients from the patients population re-challenged by pegylated-Interferon and 2) by the genotype -1 non EVR patients treated with pegylated interferon + ribavirin from the treatment na ' ive study population.
  • Samples were genotyped for 1,016,423 markers on the Illumina Infinium ® HD Assay Super, using HumanOmnil Quad (vl .O) chips and iScan scanner.
  • Initial quality control was performed to zero-out SNPs for discordant calls between replicate samples, excess heterozygosity, low call rate ( ⁇ 0.95), cluster separation, GenTrain score, intensities and cluster width. A total of 25 samples failed initial quality checks or subsequent genotyping attempts.
  • 1,002,139 SNPs with genotype calls were generated. The number of SNPs per chromosome, distribution of allelic frequencies and Hardy Weinberg equilibrium were calculated for the self- reported Caucasian population.
  • Statistical analysis was performed to zero-out SNPs for discordant calls between replicate samples, excess heterozygosity, low call rate ( ⁇ 0.95), cluster separation, GenTrain score, intensities and cluster width. A total of 25 samples failed initial quality checks or subsequent genotyping attempts.
  • 1,002,139 SNPs with genotype calls were generated. The number of S
  • virological response EMR or SVR
  • baseline variables age, body mass index [BMI], HCV RNA level and ALT quotient, entered as continuous variables, and sex, HCV genotype, histological diagnosis [presence or absence of cirrhosis] and race entered as categorical variables
  • Logistic regression PROC LOGISTIC, SAS v9.2 was used to test for associations between individual SNPs and response/non-response after adjustment for baseline BMI, sex, age, viral load, ALT quotient and principal components analysis (PCA) components.
  • PCA principal components analysis
  • ancestry was based on PCA as proposed by Purcell et al. 16 using an ancestry dataset created using the overall population ("pgt").
  • the overall population was supplemented with HapMap Phase III founders from 11 ethnically diverse subject sets and analysed in SAS JMP Genomics (SAS Institute Inc., Cary, NC, USA).
  • PCA components were compared with and without outliers. Outliers were defined as individuals whose ancestry was at least 6 standard deviations from the mean on one of the top ten inferred axes of variation.
  • SNPs that were on the X and Y chromosomes or in mitochondrial DNA were removed along with SNPs that were not genotyped in HapMap phase III founders (release number 27), or were in regions with known high linkage disequilibrium (Chr5, 44-51.5Mb; Chr6, 24-36Mb; Chr8, 8- 12Mb; Chrl 1, 42-58Mb; Chrl7, 40-43Mb). Remaining SNPs were thinned using PLINK 16 with a window size of 1000, r 2 ⁇ 0.25 and a window shift of 100. The association between viro logical response (EVR and SVR) and individual SNPs was tested by logistic regression analysis. Adjustments were made for baseline characteristics (BMI, sex, age, baseline HCV RNA level, ALT quotient and PCA components). Significance was assessed by likelihood ratio test (LRT).
  • LRT likelihood ratio test
  • the null model was defined as follows:
  • Viro logical response BMI + sex + age + HCV RNA level + ALT quotient + PCA components.
  • Viro logical response BMI + sex + age + HCV RNA level + ALT quotient + PCA components.
  • Viro logical response BMI + sex + age + HCV RNA level + ALT quotient + PCA components + SNP Maximum likelihood estimates for each alternative model (i.e. with SNPs) were compared to the corresponding null model (i.e., without SNPs) using LRT, which has a chi-square distribution and degrees of freedom equal to the number of differing parameters. SNPs were entered into the model as continuous variables (0, 1, 2). PCA components represented the top five components in the analysis populations. Similar models were run in the self-reported Caucasian population and in the non-genotype 1 population.
  • Linkage disequilibrium was calculated among significant SNPs (p ⁇ 10 ⁇ 5 ) in the IL-28 region in the Caucasian "pgt" population.
  • An LD plot of r 2 was created in Haploview v4.1, 17 with LD blocks being inferred by the method of Gabriel et al. 18
  • genotype 1 patients included 627 patients with known EVR status (215 responders [34.3%] and 412 nonresponders [65.7%]) and 516 patients with known SVR status (128 responders [24.8%] and 388 non-responders
  • Genome wide association results for SVR and EVR are presented by chromosome in Figure 1.
  • a series of highly significant p values were identified in the IL-28 region on chromosome 19.
  • Quantile-quantile plots showed that the expected and observed p values conform closely with the exception of a few large deviations for the p values associated with chromosome 19 ( Figure 2).
  • the logistic regression analysis for SVR and EVR revealed 12 and 19 SNPs, respectively, with p ⁇ 10 "5 (TABLE 2).
  • Ferenci P Fried MW, Shiffman ML, Smith CI, Marinos G, Goncales FL, Jr., Haussinger D, Diago M, Carosi G, Dhumeaux D, Craxi A, Chaneac M, Reddy KR. Predicting sustained viro logical responses in chronic hepatitis C patients treated with peginterferon alfa-2a (40 KD)/ribavirin. J Hepatol 2005;43:425-433.
  • Tanaka Y Nishida N, Sugiyama M, Kurosaki M, Matsuura K, Sakamoto N, Nakagawa M, Korenaga M, Hino K, Hige S, Ito Y, Mita E, Tanaka E, Mochida S, Murawaki Y, Honda M, Sakai A, Hiasa Y, Nishiguchi S, Koike A, Sakaida I, Imamura M, Ito K, Yano K, Masaki N, Sugauchi F, Izumi N, Tokunaga K, Mizokami M. Genome-wide
  • Esteban M Hepatitis C and evasion of the interferon system: a PKR paradigm. Cell Host Microbe 2009;6:495-497.
  • An OR >1 indicates that the probability of SVR or EVR increases as the number of copies of the minor allele increases. Conversely, an OR indicates that the probability of SVR or EVR decreases as the number of copies of the minor allele increases.
  • OR > 1 indicates that as the number of copies of the minor allele increases, an individual will have an increased probability of response
  • OR ⁇ 1 indicates that as the number of copies of the minor allele increases, an individual will have a decreased probability of response

Landscapes

  • Chemical & Material Sciences (AREA)
  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Organic Chemistry (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Engineering & Computer Science (AREA)
  • Zoology (AREA)
  • Wood Science & Technology (AREA)
  • Analytical Chemistry (AREA)
  • Genetics & Genomics (AREA)
  • Immunology (AREA)
  • General Health & Medical Sciences (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Molecular Biology (AREA)
  • Microbiology (AREA)
  • Biotechnology (AREA)
  • Biophysics (AREA)
  • Biochemistry (AREA)
  • Physics & Mathematics (AREA)
  • General Engineering & Computer Science (AREA)
  • Communicable Diseases (AREA)
  • Virology (AREA)
  • Pathology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Medicinal Chemistry (AREA)
  • Animal Behavior & Ethology (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • AIDS & HIV (AREA)
  • Tropical Medicine & Parasitology (AREA)
  • Oncology (AREA)
  • Measuring Or Testing Involving Enzymes Or Micro-Organisms (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Peptides Or Proteins (AREA)
  • Medicinal Preparation (AREA)

Abstract

The present invention is based on the discovery of associations that exist between single nucleotide polymorphisms (SNPs) on chromosome 4 and virological outcomes in a diverse population of patients with hepatitis C virus (HCV) who received interferon-based treatment.

Description

SINGLE NUCLEOTIDE POLYMORPHISMS THAT PREDICT HCV TREATMENT
OUTCOMES
The present invention relates to methods that useful for predicting the response of hepatitis C virus (HCV) infected patients to pharmacological treatment.
Background of the invention
The standard of care for chronic hepatitis C is the combination of peginterferon plus ribavirin.1 Overall sustained viro logical response (SVR) rates after treatment with the standard of care are approximately 50%,2"4 although it is difficult to predict whether an individual patient will achieve an SVR.
The probability of achieving an SVR varies with a collection of patient and viral factors. For example, younger patients, Caucasian and Asian patients, and individuals without advanced hepatic fibrosis are more likely to clear HCV infection after treatment.5"8 Similarly, patients infected with HCV genotypes 2 or 3, rather than genotype 1, and those with low baseline HCV RNA levels in serum have the best chance of a cure.2"4' 6' 8
More precise prediction of SVR is currently possible only after the start of treatment. Regardless of HCV genotype, individuals who clear HCV RNA after 4 or 12 weeks of treatment have a much better chance of achieving an SVR than those with persistent viremia.9 Rapid viro logical response (RVR, undetectable HCV RNA at week 4) is a strong predictor of SVR; conversely, failure to achieve an early viro logical response (EVR, greater than a two log decline in HCV RNA at week 12) is a strong predictor of nonresponse, independent of pretreatment
characteristics.10 The ability to prospectively differentiate between potential responders and non-responders to the standard of care could have a great impact on the care of patients with chronic hepatitis C.
Treatment decisions could be personalized based on the likelihood of patients to respond to the standard of care. For example patients with the lowest likelihood of achieving an SVR with the current standard of care might defer treatment until direct acting antiviral agents are available. Conversely, patients with a high likelihood of achieving an SVR might prefer to initiate therapy immediately with a treatment regimen that is a known entity.
In addition to host and viral factors, host genetic diversity also influences the response to treatment with the standard of care.11 Recent evidence from genome-wide association studies suggests that single nucleotide polymorphisms (SNPs) in the promoter region of the IL-28b gene exert a strong influence on the probability of SVR in patients treated with peginterferon plus ribavirin.12"14 The objectives of this analysis were to explore SNPs associated with both early virological response (EVR) and SVR in a diverse cohort of patients that includes treatment-naive individuals and nonresponders to a previous course of peginterferon alfa-2b (12KD) plus ribavirin who received peginterferon alfa-2a (40KD) monotherapy or combination therapy with either peginterferon alfa-2a (40KD) or conventional interferon plus ribavirin.
Summary of the invention
The present invention is based on the discovery of an association between several SNP genotypes on human chromosome four and SVR in patients treated with interferon-based regimens. In one embodiment, the invention provides for a method for predicting sustained virological response of a human subject infected with HCV to interferon treatment comprising, providing a sample from said human subject, detecting the presence of a single nucleotide polymorphism within chromosome 4 and determining that said subject has a high likelihood of sustained virological response to interferon treatment if said single nucleotide polymorphism is present, wherein said single nucleotide polymorphism is selected from the group consisting of a G at rsl0009948, a G at rsl0023606, and a T at rs7673763.
Brief description of the drawings
Figure 1. Genome-wide association results for (a) SVR (b) EVR) and (c) SVR after adjustment for rs 12979860 by chromosome in the overall genotype 1 population. Figure 2. Quantile-quantile plot of the distribution of test statistics for (a) SVR and (b) EVR). Grey circles denote expected p-values and blue circles show observed p values.
Figure 3. Linkage disequilibrium of significant SNPs (p<10~5) in the IL-28 region of the
Caucasian genotype 1 population. Detailed description of the invention
Definitions
To facilitate the understanding of this invention, a number of terms are defined below. Terms defined herein have meanings as commonly understood by a person of ordinary skill in the areas relevant to the present invention. Terms such as "a", "an" and "the" are not intended to refer to only a singular entity, but include the general class of which a specific example may be used for illustration. The terminology herein is used to describe specific embodiments of the invention, but their usage does not delimit the invention, except as outlined in the claims.
The term "response" to treatment with interferon is a desirable response to the administration of an agent. Viro logical endpoints included "early viro logical response" (EVR), defined as >2-log drop in serum HCV R A from baseline to week 12 (by Cobas Amplicor HCV Monitor Test, v2.0, limit of quantitation 600 IU/mL), complete EVR (cEVR) defined as undetectable HCV RNA in serum (by Cobas Amplicor HCV Test v2.0, limit of detection 50 IU/mL) or and
"sustained viro logical response" (SVR), defined as undetectable HCV RNA (<50 IU/mL) at the end of a 24-week untreated follow-up period.
The terms "sample" or "biological sample" refers to a sample of tissue or fluid isolated from an individual, including, but not limited to, for example, tissue biopsy, plasma, serum, whole blood, spinal fluid, lymph fluid, the external sections of the skin, respiratory, intestinal and
genitourinary tracts, tears, saliva, milk, blood cells, tumors, organs. Also included are samples of in vitro cell culture constituents (including, but not limited to, conditioned medium resulting from the growth of cells in culture medium, putatively virally infected cells, recombinant cells, and cell components).
The terms "interferon" and "interferon-alpha" are used herein interchangeably and refer to the family of highly homologous species-specific proteins that inhibit viral replication and cellular proliferation and modulate immune response. Typical suitable interferons include, but are not limited to, recombinant interferon alpha-2b such as Intron® A interferon available from Schering Corporation, Kenilworth, N.J., recombinant interferon alpha-2a such as Roferon®-A interferon available from Hoffmann-La Roche, Nutley, N.J., recombinant interferon alpha-2C such as Berofor® alpha 2 interferon available from Boehringer Ingelheim Pharmaceutical, Inc.,
Ridgefield, Conn., interferon alpha-nl, a purified blend of natural alpha interferons such as Sumiferon® available from Sumitomo, Japan or as Wellferon® interferon alpha-nl (INS) available from the Glaxo-Wellcome Ltd., London, Great Britain, or a consensus alpha interferon such as those described in U.S. Pat. Nos. 4,897,471 and 4,695,623 (especially Examples 7, 8 or 9 thereof) and the specific product available from Amgen, Inc., Newbury Park, Calif, or interferon alpha-n3 a mixture of natural alpha interferons made by Interferon Sciences and available from the Purdue Frederick Co., Norwalk, Conn., under the Alferon Tradename. The use of interferon alpha-2a or alpha-2b is preferred. Interferons can include pegylated interferons as defined below.
The terms "pegylated interferon", "pegylated interferon alpha" and "peginterferon" are used herein interchangeably and means polyethylene glycol modified conjugates of interferon alpha, preferably interferon alfa-2a and alfa-2b. Typical suitable pegylated interferon alpha include, but are not limited to, Pegasys® and Peg-Intron®.
The term "ribavirin" refers to the compound, l-((2R,3R,4S,5R)-3,4-Dihydroxy-5- hydroxymethyl-tetrahydro-furan-2-yl)-lH-[l,2,4]triazole-3-carboxylic acid amide which is a synthetic, non-interferon-inducing, broad spectrum antiviral nucleoside analog and available under the names, Virazole® and Copegus® .
"Direct acting antiviral agents" exert specific antiviral effects independent of immune function. Examples of direct acting antiviral agents for HCV include but are limited to protease inhibitors, polymerase inhibitors, NS5A inhibitors, IRES inhibitors and helicase inhibitors.
The current recommended first line treatment for patients with chronic hepatitis C is pegylated interferon alpha in combination with ribavirin for 48 weeks in patients carrying genotype 1 or 4 virus and for 24 weeks in patients carrying genotype 2 or 3 virus. Combined treatment with ribavirin was found to be more effective than interferon alpha monotherapy in patients who relapsed after one or more courses of interferon alpha therapy, as well as in previously untreated patients. However, ribavirin exhibits significant side effects including teratogenicity and carcinogenicity. Furthermore, ribavirin causes hemolytic anemia requiring dose reduction or discontinuation of ribavirin therapy in approximately 10 to 20% of patients, which may be related to the accumulation of ribavirin triphosphate in erythrocytes. Therefore, to reduce treatment cost and the incidence of adverse events, it is desirable to tailor the treatment to a shorter duration while not compromising efficacy. A shortened "duration of treatment" for genotype 1 patients with pegylated interferon alpha with ribovirin would be, for example, 24 weeks. A shortened duration of treatment for genotype 1 patients with pegylated interferon alpha with ribavirin in combination with a direct acting antiviral agent could be as short as 8 weeks, 12 weeks, or 16 weeks.
As used herein, the terms "allele" and "allelic variant" refer to alternative forms of a gene including introns, exons, intron/exon junctions and 3' and/or 5' untranslated regions that are associated with a gene or portions thereof. Generally, alleles occupy the same locus or position on homologous chromosomes. When a subject has two identical alleles of a gene, the subject is said to be homozygous for the gene or allele. When a subject has two different alleles of a gene, the subject is said to be heterozygous for the gene. Alleles of a specific gene can differ from each other in a single nucleotide, or several nucleotides, and can include substitutions, deletions, and insertions of nucleotides.
As used herein, the term "polymorphism" refers to the coexistence of more than one form of a nucleic acid, including exons and introns, or portion (e.g., allelic variant) thereof. A portion of a gene of which there are at least two different forms, i.e., two different nucleotide sequences, is referred to as a polymorphic region of a gene. A polymorphic region can be a single nucleotide, i.e. "single nucleotide polymorphism" or "SNP", the identity of which differs in different alleles. A polymorphic region can also be several nucleotides long.
Numerous methods for the detection of polymorphisms are known and may be used in conjunction with the present invention. Generally, these include the identification of one or more mutations in the underlying nucleic acid sequence either directly (e.g., in situ hybridization) or indirectly (identifying changes to a secondary molecule, e.g., protein sequence or protein binding).
One well-known method for detecting polymorphisms is allele specific hybridization using probes overlapping the mutation or polymorphic site and having about 5, 10, 20, 25, or 30 nucleotides around the mutation or polymorphic region. For use in a kit, e.g., several probes capable of hybridizing specifically to allelic variants, such as single nucleotide polymorphisms, are provided for the user or even attached to a solid phase support, e.g., a bead or chip.
The single nucleotide polymorphism, "rsl2979860" refers to a SNP identified by its accession number in the database of SNPs (dbSNP, www.ncbi. nlm. nih. gov/ SNP/) and is located on human chromosome 19 in the promoter region of the IL28b gene. The results of this analysis of data from two large multinational studies (described in the
Examples section) confirm and extend the remarkable association between SNPs in the IL-28 region and SVR after treatment with peginterferon plus ribavirin that was first reported by Ge et al.12 In the present analysis, the top SNP (rsl297980) was associated with both EVR (p=5.0 x 10" 26) and SVR (p=l .4 x 10~21). The inclusion of patients who achieved an SVR after treatment not only with peginterferon alfa-2a (40KD) plus ribavirin, but also with peginterferon alfa-2a (40KD) monotherapy and the combination of conventional interferon plus ribavirin, as well as the well- pedigreed nature of the large subgroup of non-responders are unique strengths of the study.
This study allows for the generation of several unique suggestions. First, we suggest that the effect of rs 12979860 is exerted most likely on EVR rather than on SVR. An association with EVR, but not SVR, was seen in previous non-responders to therapy and in patients treated with peginterferon alfa-2a (40KD) monotherapy, or conventional interferon plus ribavirin. We interpret this to mean that the mechanism through which this SNP acts is associated with the rapid first-phase decline in serum HCV RNA levels rather than with the slower second phase decline or hepatocyte turnover. The first phase decline in HCV RNA is a well described phenomenon that occurs shortly after the initiation of interferon-based therapy and has been suggested to be an indicator of a particular patient's sensitivity to interferon.19
Next, the particular chip used in this study allowed us to rank the significance of a series of SNPs
12 1 20 21
in addition to rs 12979860, all of which were identified in previous studies. " ' ' Although several highly significant markers were identified in the IL28b region, none of them were significantly associated with SVR after adjustment for rsl297980. Only one SNP (rs8099917) was statistically significantly associated with EVR after adjustment for rs 12979860; however, the association was such that it would not have been deemed significant after adjustment for multiple comparisons. In addition, by adjusting for rs 12979860 we were able to begin exploring potential gene-gene interactions that may affect the probability of achieving a virological response. As noted above, none of the other highly significant SNPs in the IL28b region identified in the primary analysis were retained after this process; however, a series of previously unreported SNPs located on chromosome 4 were revealed.
12 1 20 23
Consistent results obtained in several retrospective studies " ' " including this one, allow us to say with confidence that SNPs in the promoter region of IL28b are highly associated with the outcome of treatment with interferon-based therapy. It has been suggested that changes in one of the multiple promoters in this region alter hepatic expression of interferon lambda. Interferon lambda is a type III interferon that triggers a signalling pathway that overlaps with the Jak/Stat pathway of type I interferons (including interferon-alpha).24-26 We hypothesise that in patients with a "permissive" SNP, HCV infection induces local production of interferons -including type III interferons (such as IL28). This results in interferon-induced gene expression in the liver. This hypothesis is consistent with prior reports that non-responders to interferon-based therapy have increased hepatic expression of interferon-stimulated genes (ISGs) in their liver prior to the initiation of therapy, and are in an apparent "pre-activated" state.27 This apparent paradoxical relationship between the level of endogenous ISG induction and the subsequent responsiveness to interferon-based therapy has not been clearly explained. Some investigators have speculated that some of the downstream effects of ISG induction on an axis similar to protein kinase PKR might suppress endogenous protein production via phosphorylation of eukaryotic initiation factor 2 alpha (eIF2a) and result in hepatocytes that, being refractory to further stimulation by exogenous interferon, are unable to further attenuate protein production.28 HCV RNA is translated via an internal ribosomal entry site and is partially eIF2a- independent. Thus, replication of HCV RNA and assembly of further virions can continue unabated despite the "pre- activated" state.28 This hypothesis explains why a poorly responsive GWAS genotype is associated with a lower viral load than the more treatable genotype in our analysis, in spite of previous analyses that suggest increasing baseline viral load is an indicator of poor treatment outcomes.6 Endogenous production of interferon partially impairs viral replication, but simultaneously impairs host cell protein synthesis in these patients. There may be a threshold above which host cells cannot further decrease protein production or increase stimulation of ISGs. Thus, exogenous interferon cannot augment the host response. Paradoxically,
administration of exogenous pegylated interferon can, however, "rescue" patients who are less sensitive to endogenous interferon. The genetic polymorphism may play a role in ISG expression. This leads us to speculate that it may eventually be possible to selectively target the interferon pathway to produce a more favourable ISG profile that results in HC V eradication.
It is more challenging to explain the possible mechanism by which a SNP in chromosome 4 is associated with SVR, though not with EVR. The simplest explanation might require a gene in this region to be involved in a phenomenon that is exclusively associated with SVR such as hepatocyte turnover. Additional host-host or host-viral gene interactions may be involved in the mechanism by which both loci exert their effects, and require further careful evaluation. The discovery of an interferon treatment susceptibility locus in the IL28b region has implications for therapy with the current standard of care (SOC), i.e. peginterferon plus ribavirin. In the near future, one might envisage that in some patients the IL28b genotype will be determined before therapy is initiated and that patients will be stratified not only by HCV genotype - as currently recommended1 - but also by human genotype. In contrast to the current treatment paradigm, the initial treatment regimen will be based upon both viral and human genotype. The putative role of ribavirin in this context will need to be addressed in prospective trials.
This discovery also has implications for the ongoing development programs of direct acting antiviral agents for HCV therapy. Direct acting antiviral agents exert specific antiviral effects independent of immune function, but it is thought that at least some of the drug classes in development may benefit from an additive (if not synergistic) innate immunomodulatory effect. Indeed it is likely that interferon-based therapy will remain the backbone of the treatment because it is needed to prevent the emergence of resistant HCV.29 HCV R A encodes specific proteins that may inhibit the induction of type I interferons. For example the NS3-4A protease of HCV blocks dsR A-induced interferon production by interfering with phosphorylation of interferon regulatory factor-3 (IRF-3).30 Thus, the NS3-4A protease is a dual therapeutic target, whose inhibition may block viral replication and restore IRF-3 control of HCV RNA replication. Protease inhibitors such as Telaprevir, which have robust antiviral effects when administered in combination with a second small molecule or the standard of care, also inhibit the protease functions by which HCV impairs host interferon response. It will be important to observe whether treatment outcomes are similar when direct acting antiviral agents are combined with peginterferon plus ribavirin in patients with interferon-refractory and interferon-sensitive IL28b phenotypes. It is possible that interferon-refractory patients will respond to triple therapy (direct acting antiviral agent plus peginterferon plus ribavirin) as if they were on monotherapy with the direct acting antiviral agent alone. If this is the case then patients with the interferon-refractory IL28b phenotype may be much more susceptible to the selection of resistance mutations during treatment with a drug such as telaprevir.31 These possibilities suggest scenarios where patients with the interferon-susceptible SNP (rs 12979860) might benefit from abbreviated treatment with peginterferon and ribavirin, and those with an interferon-refractory genotype might be candidates for extended treatment durations and/or more intensive treatment regimens.. Alternatively, interferon- free combination regimens of direct acting antiviral agents may be more appropriate for patients with an interferon-refractory genotype. Our unique dataset, including a registration trial of peginterferon alfa-2a plus ribavirin therapy for patients who were previous non-responders to standard of care with a pegylated interferon, highlights the correlation of the rsl2979860 SNP with EVR rather than SVR. Stated differently, this SNP defines patient's responsiveness to interferon rather than the ultimate response to therapy and thus helps identify patients likely or unlikely to undergo an SVR because of their likelihood to achieve an EVR. It is unlikely that it predicts SVR independently of EVR. This has tremendous implications for use of direct acting antiviral agents together with interferon.
Utilization of baseline genetic predictors for interferon responsiveness allows tailoring of direct acting antiviral therapies, as well as standard of care with interferon. Patients who are defined as poorly interferon responsive, i.e. two T alleles in rsl2979860, may be poor candidates for small molecule therapeutics that rely on additive or synergistic effects of endogenous or exogenous interferon mediated pathways - particularly if these are provided as a single agent in addition to SOC. The concern would be that these patients would have an increased risk of developing drug resistant mutations as a result of effective monotherapy. Conversely, patients with an interferon responsive phenotype, i.e. two C alleles in rsl2979860, would make superior candidates for shorter courses of therapy with SOC alone, or in
combination with small molecules. Additional considerations that genetic predisposition to interferon responsiveness allow is "tuning" of combination direct acting antiviral agents.
Patients predicted to have acceptable endogenous interferon responsiveness may be excellent candidates for drugs that target viral functions-such as protease inhibitors which also have an inhibitory role on endogenous interferon response- and poorer candidates for drugs that decrease the amounts of viral PAMP (pathogen associated molecular pattern, e.g. polymerase inhibitors) as these may serve to impair the patients capacity to facilitate their own cure via their
endogenous interferon responsiveness. Similarly, patients predicted to have poor interferon responsiveness might be candidates for "quad" therapy as a first line of therapy (2 direct acting antiviral agents, added to peginterferon with ribavirin), as compared to a direct acting antiviral agent alone, or triple therapy (SOC with one DAA). Examples
Methods
Patients Sample collection and viro logical end-points
Samples were analysed from a subset of patients with chronic hepatitis C enrolled in 2 large, randomised, multinational, phase III trials.3' 15 In one study interferon-naive patients were randomised to 48 weeks of treatment with peginterferon alfa-2a (40KD) alone or in combination with ribavirin, or conventional interferon alfa-2b plus ribavirin.3 Only non-responders to a previous 12-week course of peginterferon alfa-2b (12KD) plus ribavirin were eligible for the second trial, which randomised patients to either 48 or 72 weeks of treatment with either a standard or induction dose regimen of peginterferon alfa-2a (40KD) (all patients received standard dose ribavirin).15 The study design, inclusion and exclusion criteria and primary results of these trials are published elsewhere.3' 15
Blood samples collected from patients who consented to participate in genetic analyses were stored in the Roche Clinical Sample repository. DNA was extracted at the Roche Clinical Sample repository and normalized to 50 ng/μΕ. Samples initially underwent a quality check using a Y-chromosome specific TaqMan Assay (Applied Biosystems, Foster City, CA) to assess both DNA quality and gender concordance with clinical data.
Viro logical endpoints included early viro logical response (EVR), defined as undetectable HCV RNA in serum (by Cobas Amplicor HCV Test v2.0, limit of detection 50 IU/mL) or >2-log drop in serum HCV RNA from baseline to week 12 (by Cobas Amplicor HCV Monitor Test, v2.0, limit of quantitation 600 IU/mL) and sustained virological response (SVR), defined as undetectable HCV RNA (<50 IU/mL) at the end of a 24-week untreated follow-up period.
In the GWAS analysis for SVR, the responder group was composed of all genotype- 1 patients with SVR from the interferon na'ive study population. Non-responders were composed of 1) all genotype- 1 non-SVR patients from the study population re-challenged by pegylated-Interferon treatment, 2) genotype- 1 non-SVR patients form the interferon na'ive study population treated with pegylated interferon with ribavirin.
All responders from the study population re-challenged by the pegylated interferon population were therefore excluded from our analysis. In the GWAS analysis for EVR, the EVR group was composed of all genotype- 1 patients with EVR from the interferon na'ive study population. The non-EVR group was composed of l)all patients from the patients population re-challenged by pegylated-Interferon and 2) by the genotype -1 non EVR patients treated with pegylated interferon + ribavirin from the treatment na'ive study population.
Additional exploratory analysis were conducted in the IL28B region SNP the Self-reported Caucasian in non-genotype 1 patients, and in each trial separately, (see details in the result section).
Genotypic data analysis
Samples were genotyped for 1,016,423 markers on the Illumina Infinium® HD Assay Super, using HumanOmnil Quad (vl .O) chips and iScan scanner. Initial quality control was performed to zero-out SNPs for discordant calls between replicate samples, excess heterozygosity, low call rate (<0.95), cluster separation, GenTrain score, intensities and cluster width. A total of 25 samples failed initial quality checks or subsequent genotyping attempts. After quality control, 1,002,139 SNPs with genotype calls were generated. The number of SNPs per chromosome, distribution of allelic frequencies and Hardy Weinberg equilibrium were calculated for the self- reported Caucasian population. Statistical analysis
Associations between virological response (EVR or SVR) and baseline variables (age, body mass index [BMI], HCV RNA level and ALT quotient, entered as continuous variables, and sex, HCV genotype, histological diagnosis [presence or absence of cirrhosis] and race entered as categorical variables) were evaluated in a univariate logistic regression model. Logistic regression (PROC LOGISTIC, SAS v9.2) was used to test for associations between individual SNPs and response/non-response after adjustment for baseline BMI, sex, age, viral load, ALT quotient and principal components analysis (PCA) components.
The analysis of ancestry was based on PCA as proposed by Purcell et al.16 using an ancestry dataset created using the overall population ("pgt"). The overall population was supplemented with HapMap Phase III founders from 11 ethnically diverse subject sets and analysed in SAS JMP Genomics (SAS Institute Inc., Cary, NC, USA). PCA components were compared with and without outliers. Outliers were defined as individuals whose ancestry was at least 6 standard deviations from the mean on one of the top ten inferred axes of variation. SNPs that were on the X and Y chromosomes or in mitochondrial DNA were removed along with SNPs that were not genotyped in HapMap phase III founders (release number 27), or were in regions with known high linkage disequilibrium (Chr5, 44-51.5Mb; Chr6, 24-36Mb; Chr8, 8- 12Mb; Chrl 1, 42-58Mb; Chrl7, 40-43Mb). Remaining SNPs were thinned using PLINK16 with a window size of 1000, r2<0.25 and a window shift of 100. The association between viro logical response (EVR and SVR) and individual SNPs was tested by logistic regression analysis. Adjustments were made for baseline characteristics (BMI, sex, age, baseline HCV RNA level, ALT quotient and PCA components). Significance was assessed by likelihood ratio test (LRT).
The null model was defined as follows:
Viro logical response = BMI + sex + age + HCV RNA level + ALT quotient + PCA components. In the null model the genetic effect of SNPs was assumed to be zero.
The alternative model was defined as:
Viro logical response = BMI + sex + age + HCV RNA level + ALT quotient + PCA components + SNP Maximum likelihood estimates for each alternative model (i.e. with SNPs) were compared to the corresponding null model (i.e., without SNPs) using LRT, which has a chi-square distribution and degrees of freedom equal to the number of differing parameters. SNPs were entered into the model as continuous variables (0, 1, 2). PCA components represented the top five components in the analysis populations. Similar models were run in the self-reported Caucasian population and in the non-genotype 1 population.
Linkage disequilibrium (LD) was calculated among significant SNPs (p<10~5) in the IL-28 region in the Caucasian "pgt" population. An LD plot of r2 was created in Haploview v4.1,17 with LD blocks being inferred by the method of Gabriel et al.18
Results Samples were available for a total of 406 treatment naive patients and 426 patients who had not responded to a previous course of treatment with peginterferon alfa-2b (12KD). The analysis of EVR was based on data from 800 patients, including 363 individuals (45%) who achieved an EVR and 437 individuals (55%) who did not achieve an EVR. The analysis of SVR was based on data from 663 patients, including 245 patients (37%) who achieved an SVR and 418 (63%) who did not achieve an SVR. The baseline characteristics of patients included in the analysis of EVR and SVR are shown in TABLE 1.
The univariate logistic regression models of baseline factors showed that HCV genotype (p=1.50 x 10~25), age (5.80 x 10~18), ALT quotient (p=7.50 x 10"10), race (p=9.20 x 10~7), BMI (p=4.70 x 10"5) and histological diagnosis (p=1.30 x 10"5) were significantly associated with EVR, and that HCV genotype (p=4.30 x 10~27), age (p=5.50 x 10~18), ALT quotient (p=2.10 x 10"8), race (p=2.10 x 10"8), histological diagnosis (p=5.40 x 10"7), BMI (p=7.20 x 10"6) and HCV RNA level (p=0.0022) were significantly associated with SVR. Of note sex was not significantly associated with either EVR or SVR and HCV RNA level was not significantly associated with SVR. GWAS results in the overall genotype 1 population
A total of 4 samples analysed for genotype were excluded because of high kinship coefficients (indicating a high degree of relatedness). The analysis of data from genotype 1 patients included 627 patients with known EVR status (215 responders [34.3%] and 412 nonresponders [65.7%]) and 516 patients with known SVR status (128 responders [24.8%] and 388 non-responders
[75.2%]).
Genome wide association results for SVR and EVR are presented by chromosome in Figure 1. A series of highly significant p values were identified in the IL-28 region on chromosome 19. Quantile-quantile plots showed that the expected and observed p values conform closely with the exception of a few large deviations for the p values associated with chromosome 19 (Figure 2). The logistic regression analysis for SVR and EVR revealed 12 and 19 SNPs, respectively, with p<10"5 (TABLE 2).
The top 6 SNPs associated with SVR and EVR, respectively, were identical and fell in the IL-28 region of chromosome 19. The top two SNPs for SVR and EVR were rsl2979860 (p=1.4xl0~21 and p=5.0xl0"26, respectively) and rsl2980275 (p=5.8xl0~18 and p=4.9xl0"23, respectively). Of the remaining six SNPs associated with SVR (TABLE 2), none were associated with EVR, none were located on chromosome 19 and four (rs943897, rs 17671102, rs4961441 and rs 1892723) may be spurious associations because of the small number of observations in the rare
homozygote class (BB <10 individuals). Of the remaining 13 SNPs associated with EVR
(TABLE 2), none were associated with SVR, only one (rs4803223) was located on chromosome 19, and two (rsl 189800 and rs4975629) may be spurious associations.
Logistic regression analyses of all SNPs with p<10"5 for any model of SVR and EVR are shown in TABLE 3 for the overall population and for the subgroup of self-reported Caucasians. When the analyses were repeated after adjustment for the effect of rsl 2979860 none of the markers were significantly associated with SVR and only one marker was significantly associated with EVR (rs8099917, p=0.0130).
Subsequent investigation of linkage disequilibrium among SNPs in the IL-28 region presented in TABLE 3 demonstrated one cluster with a high degree of linkage disequilibrium (between rsl2979860 and rsl2980275, r2=0.98) and a second cluster of SNPs downstream comprised of rsl2980275, rsl2979860, rs8109886 and rs8099917 (Figure 3). The genome wide results for SVR after adjustment for rsl 2979860 are shown in Figure lc. The results of logistic regression analysis for SVR after adjustment for rsl 2979860 and inclusion of additional non-responders revealed an additional 10 SNPs with p<10"5 (Table 4). Interestingly, three of the five most prominent of which (rsl0009948, rsl0023606, and rs7673763) are located on chromosome 4. Curiously, these were associated exclusively with SVR, and not with EVR. Exploratory analysis of rsl 2979860 in the Caucasian self-reported population
In order to better characterize and understand the association between IL28B, we first explore the association between rsl 2979860 in Non-genotype 1 in the self-reported Caucasian population. With the same logistic regression model as in the GWAS, a borderline significant association between SVR and the number of rsl2979860 C allele is seen (OR=1.88, 95% CI= [0.97; 3.67], p= 0.06) ,at the 5% nominal type 1 error. Similarly, a significant association between EVR and the number of rsl2979860 C allele (OR=2.27, 95% CI [ 1.12; 4.70], p=0.02).
In addition, we also describe the association between rsl 2979860 in the two trials separately. In the na'ive-treated population, an association between EVR and rsl 2979860 C allele is seen with an OR of nearly 5 (OR=4.98 95% CI [2.35; 10.53], p=2.6X10~5). The association is constantly seen in all the three treatment strata.
In the patient population who failed on pegylated-interferon therapy, no association was found with sustained viral response. However an association with rs 12979860 persists when comparing EVR (N=185) and non-EVR (N=154) (OR=1.91 [1.22; 2.96], p=0.003). The association was independent from the viral load at baseline, gender, age, treatment and ALT Quotient.
All of the compositions and/or methods disclosed and claimed herein can be made and executed without undue experimentation in light of the present disclosure. While the compositions and methods of this invention have been described in terms of preferred embodiments, it will be apparent to those of skill in the art that variations may be applied to the compositions and/or methods and in the steps or in the sequence of steps of the method described herein without departing from the concept, spirit and scope of the invention. All such similar substitutes and modifications apparent to those skilled in the art are deemed to be within the spirit, scope and concept of the invention as defined by the appended claims. References
1. Ghany MG, Strader DB, Thomas DL, Seeff LB. Diagnosis, management, and treatment of hepatitis C: An update. Hepatology 2009;49: 1335-1374.
2. Manns MP, McHutchison JG, Gordon SC, Rustgi VK, Shiffman M, Reindollar R,
Goodman ZD, Koury K, Ling M, Albrecht JK. Peginterferon alfa-2b plus ribavirin compared with interferon alfa-2b plus ribavirin for initial treatment of chronic hepatitis C: a randomised trial. Lancet 2001;358:958-965.
3 Fried MW, Shiffman ML, Reddy KR, Smith C, Marinos G, Goncales FL, Jr., Haussinger D, Diago M, Carosi G, Dhumeaux D, Craxi A, Lin A, Hoffman J, Yu J. Peginterferon alfa-2a plus ribavirin for chronic hepatitis C virus infection. N Engl J Med 2002;347:975-
982.
4 Hadziyannis SJ, Sette H, Jr., Morgan TR, Balan V, Diago M, Marcellin P, Ramadori G, Bodenheimer H, Jr., Bernstein D, Rizzetto M, Zeuzem S, Pockros PJ, Lin A, Ackrill AM. Peginterferon-alpha2a and ribavirin combination therapy in chronic hepatitis C: a randomized study of treatment duration and ribavirin dose. Ann Intern Med
2004;140:346-355. Conjeevaram HS, Fried MW, Jeffers LJ, Terrault NA, Wiley-Lucas TE, Afdhal N, Brown RS, Belle SH, Hoofhagle JH, Kleiner DE, Howell CD. Peginterferon and ribavirin treatment in African American and Caucasian American patients with hepatitis C genotype 1. Gastroenterology 2006;131 :470-477.
Dienstag JL, McHutchison JG. American Gastroenterological Association technical review on the management of hepatitis C. Gastroenterology 2006;130:231-264.
Missiha S, Heathcote J, Arenovich T, Khan K. Impact of asian race on response to combination therapy with peginterferon alfa-2a and ribavirin in chronic hepatitis C. Am J Gastroenterol 2007; 102:2181-2188.
Reddy KR, Messinger D, Popescu M, Hadziyannis SJ. Peginterferon alpha-2a (40 kDa) and ribavirin: comparable rates of sustained viro logical response in sub-sets of older and younger HCV genotype 1 patients. J Viral Hepat 2009;16:724-731.
Ferenci P, Fried MW, Shiffman ML, Smith CI, Marinos G, Goncales FL, Jr., Haussinger D, Diago M, Carosi G, Dhumeaux D, Craxi A, Chaneac M, Reddy KR. Predicting sustained viro logical responses in chronic hepatitis C patients treated with peginterferon alfa-2a (40 KD)/ribavirin. J Hepatol 2005;43:425-433.
Martinot-Peignoux M, Maylin S, Moucari R, Ripault MP, Boyer N, Cardoso AC, Giuily N, Castelnau C, Pouteau M, Stern C, Auperin A, Bedossa P, Asselah T, Marcellin P. Viro logical response at 4 weeks to predict outcome of hepatitis C treatment with pegylated interferon and ribavirin. Antivir Ther 2009;14:501-511.
Asselah T, Bieche I, Sabbagh A, Bedossa P, Moreau R, Valla D, Vidaud M, Marcellin P. Gene expression and hepatitis C virus infection. Gut 2009;58:846-858.
Ge D, Fellay J, Thompson AJ, Simon JS, Shianna KV, Urban TJ, Heinzen EL, Qiu P, Bertelsen AH, Muir AJ, Sulkowski M, McHutchison JG, Goldstein DB. Genetic variation in IL28B predicts hepatitis C treatment-induced viral clearance. Nature 2009;461 :399- 401.
Suppiah V, Moldovan M, Ahlenstiel G, Berg T, Weltman M, Abate ML, Bassendine M, Spengler U, Dore GJ, Powell E, Riordan S, Sheridan D, Smedile A, Fragomeli V, Muller T, Bahlo M, Stewart GJ, Booth DR, George J. IL28B is associated with response to chronic hepatitis C interferon-alpha and ribavirin therapy. Nat Genet 2009;41 : 1100-1104. Tanaka Y, Nishida N, Sugiyama M, Kurosaki M, Matsuura K, Sakamoto N, Nakagawa M, Korenaga M, Hino K, Hige S, Ito Y, Mita E, Tanaka E, Mochida S, Murawaki Y, Honda M, Sakai A, Hiasa Y, Nishiguchi S, Koike A, Sakaida I, Imamura M, Ito K, Yano K, Masaki N, Sugauchi F, Izumi N, Tokunaga K, Mizokami M. Genome-wide
association of IL28B with response to pegylated interferon-al ha and ribavirin therapy for chronic hepatitis C. Nat Genet 2009;41 : 1105-1109.
Jensen DM, Marcellin P, Freilich B, Andreone P, Di BA, Brandao-Mello CE, Reddy KR, Craxi A, Martin AO, Teuber G, Messinger D, Thommes JA, Tietz A. Re-treatment of patients with chronic hepatitis C who do not respond to peginterferon-alpha2b: a randomized trial. Ann Intern Med 2009;150:528-540.
Purcell S, Neale B, Todd-Brown K, Thomas L, Ferreira MA, Bender D, Mailer J, Sklar P, de Bakker PI, Daly MJ, Sham PC. PLINK: a tool set for whole-genome association and population-based linkage analyses. Am J Hum Genet 2007;81 :559-575.
Barrett JC, Fry B, Mailer J, Daly MJ. Haploview: analysis and visualization of LD and haplotype maps. Bioinformatics 2005;21 :263-265.
Gabriel SB, Schaff er SF, Nguyen H, Moore JM, Roy J, Blumenstiel B, Higgins J, Defelice M, Lochner A, Faggart M, Liu-Cordero SN, Rotimi C, Adeyemo A, Cooper R, Ward R, Lander ES, Daly MJ, Altshuler D. The structure of haplotype blocks in the human genome. Science 2002;296:2225-2229.
Neumann AU, Lam NP, Dahari H, Gretch DR, Wiley TE, Layden TJ, Perelson AS.
Hepatitis C viral dynamics in vivo and the antiviral efficacy of interferon-alpha therapy. Science 1998;282: 103-107.
Thomas DL, Thio CL, Martin MP, Qi Y, Ge D, O'huigin C, Kidd J, Kidd K, Khakoo SI, Alexander G, Goedert JJ, Kirk GD, Donfield SM, Rosen HR, Tobler LH, Busch MP, McHutchison JG, Goldstein DB, Carrington M. Genetic variation in IL28B and spontaneous clearance of hepatitis C virus. Nature 2009;461 :798-801.
Rauch A, Kutalik Z, Descombes P, Cai T, Di I J, Mueller T, Bochud M, Battegay M, Bernasconi E, Borovicka J, Colombo S, Cerny A, Dufour JF, Furrer H, Gunthard HF, Heim M, Hirschel B, Malinverni R, Moradpour D, Mullhaupt B, Witteck A, Beckmann JS, Berg T, Bergmann S, Negro F, Telenti A, Bochud PY. Genetic Variation in IL28B Is Associated With Chronic Hepatitis C and Treatment Failure: A Genome-wide
Association Study. Gastroenterology 2010.
McCarthy JJ, Li JH, Thompson A, Suchindran S, Lao XQ, Patel K, Tillmann HL, Muir AJ, McHutchison JG. Replicated association between an interleukin-28B gene variant and a sustained response to pegylated interferon and ribavirin [manuscript in press].
Gastroenterology 2010. Montes-Cano M, et al. IL28B genetic variants and hepatitis virus infection by different viral genotypes [manuscript in press]. Hepatology 2010.
Marcello T, Grakoui A, Barba-Spaeth G, Machlin ES, Kotenko SV, MacDonald MR, Rice CM. Interferons alpha and lambda inhibit hepatitis C virus replication with distinct signal transduction and gene regulation kinetics. Gastroenterology 2006;131 : 1887-1898. Robek MD, Boyd BS, Chisari FV. Lambda interferon inhibits hepatitis B and C virus replication. J Virol 2005;79:3851-3854.
Siren J, Pirhonen J, Julkunen I, Matikainen S. IFN-alpha regulates TLR-dependent gene expression of IFN-alpha, IFN-beta, IL-28, and IL-29. J Immunol 2005;174: 1932-1937. Asselah T, Bieche I, Narguet S, Sabbagh A, Laurendeau I, Ripault MP, Boyer N, Martinot-Peignoux M, Valla D, Vidaud M, Marcellin P. Liver gene expression signature to predict response to pegylated interferon plus ribavirin combination therapy in patients with chronic hepatitis C. Gut 2008;57:516-524.
Esteban M. Hepatitis C and evasion of the interferon system: a PKR paradigm. Cell Host Microbe 2009;6:495-497.
Asselah T, Benhamou Y, Marcellin P. Protease and polymerase inhibitors for the treatment of hepatitis C. Liver Int 2009;29 Suppl 1 :57-67.
Foy E, Li K, Wang C, Sumpter R, Jr., Ikeda M, Lemon SM, Gale M, Jr. Regulation of interferon regulatory factor-3 by the hepatitis C virus serine protease. Science
2003;300: 1145-1148.
Reesink HW, Zeuzem S, Weegink CJ, Forestier N, van VA, van de Wetering de Rooij, McNair L, Purdy S, Kauffman R, Alam J, Jansen PL. Rapid decline of viral RNA in hepatitis C patients treated with VX-950: a phase lb, placebo-controlled, randomized study. Gastroenterology 2006; 131 :997-1002.
Gane E, Roberts S, Stedman C, Angus P, Ritchie B, Elston R, Ipe D, Morcos P, Najera I, Chu T, Berrey M, Bradford W, Laughlin M, Shulman N, Smith P. Combination therapy with a nucleoside polymerase (R7128) and protease (R7227/ITMN-191) inhibitor in HCV: safety, pharmacokinetics , and viro logic results from INFORM- 1 [abstract 193]. Hepatology 2009;50 (Supplement): 394-5 A.
Askarieh G, Alsio A, Pugnale P, Negro F, Ferrari C, Neumann AU, Pawlotsky JM, Schalm SW, Zeuzem S, Norkrans G, Westin J, Soderholm J, Hellstrand K, Lagging M. Systemic and intrahepatic interferon-gamma- inducible protein 10 kDa predicts the first- phase decline in hepatitis C virus RNA and overall viral response to therapy in chronic hepatitis C. Hepatology 2009. Lanford RE, Hildebrandt-Eriksen ES, Petri A, Persson R, Lindow M, Munk ME,
Kauppinen S, Orum H. Therapeutic silencing of microRNA-122 in primates with chronic hepatitis C virus infection. Science 2010;327: 198-201.
TABLE 1
Figure imgf000021_0001
a. Histological diagnosis was missing for 2 EVR/SVR non-responders
TABLE 2
Results of the logistic regression analysis of SVR and EVR in the overall genotype 1 population showing all SNPs with p<10~5 in the primary analysis
Figure imgf000022_0001
Figure imgf000023_0001
Figure imgf000024_0001
95% CI = 95% confidence interval; MAF = minor allele frequency; OR = odds ratio; SNP = single nucleotide polymorphism
a. An OR >1 indicates that the probability of SVR or EVR increases as the number of copies of the minor allele increases. Conversely, an OR indicates that the probability of SVR or EVR decreases as the number of copies of the minor allele increases.
TABLE 3
Results of the logistic regression analysis in the overall genotype 1 population showing all SNPs in the IL28 region with p<10 for either SVR or EVR in the primary analysis
Figure imgf000025_0001
TABLE 4
Top findings (SNP with p < 10-5) from logistic regression on SVR in the overall genotype- 1 population using the supplemental model which includes adjustment for rsl2979860 in the population with additional non-responders.
Figure imgf000026_0001
OR > 1 indicates that as the number of copies of the minor allele increases, an individual will have an increased probability of response
OR < 1 indicates that as the number of copies of the minor allele increases, an individual will have a decreased probability of response
a. Missing Gene and Location values were not in the NCBI database

Claims

Claims
A method for predicting sustained viro logical response of a human subject infected with HCV to interferon treatment comprising:
providing a sample from said human subject, detecting the presence of a single
nucleotide polymorphism within chromosome 4 and determining that said subject has a high likelihood of sustained viro logical response to interferon treatment if said single nucleotide polymorphism is present, wherein said single nucleotide polymorphism is selected from the group consisting of a G at rsl0009948, a G at rsl0023606, and a T at rs7673763.
The method of claim 1 wherein said subject is infected with Genotype- 1 HCV.
The method of claims 1-2 wherein said interferon treatment comprises treatment selected from the group of peginterferon alfa-2a monotherapy, peginterferon alfa-2a with ribavirin, or interferon alfa-2b with ribavirin.
PCT/EP2011/055579 2010-04-13 2011-04-11 Single nucleotide polymorphisms that predict hcv treatment outcomes WO2011128278A1 (en)

Priority Applications (10)

Application Number Priority Date Filing Date Title
JP2013504214A JP5695181B2 (en) 2010-04-13 2011-04-11 Single nucleotide polymorphism predicting HCV treatment outcome
RU2012145777/10A RU2567663C2 (en) 2010-04-13 2011-04-11 Single nucleotide polymorphisms for predicting results of treatment of hepatic c virus infection
ES11717518.2T ES2525290T3 (en) 2010-04-13 2011-04-11 Single nucleotide polymorphisms that predict the results of HCV treatment
MX2012011804A MX2012011804A (en) 2010-04-13 2011-04-11 Single nucleotide polymorphisms that predict hcv treatment outcomes.
CA2795911A CA2795911A1 (en) 2010-04-13 2011-04-11 Single nucleotide polymorphisms that predict hcv treatment outcomes
CN201180018656.0A CN102844448B (en) 2010-04-13 2011-04-11 The single nucleotide polymorphism of prediction HCV therapy result
KR20127029599A KR101493137B1 (en) 2010-04-13 2011-04-11 Single nucleotide polymorphisms that predict hcv treatment outcomes
BR112012025854A BR112012025854A2 (en) 2010-04-13 2011-04-11 polymorphism of a single nucleotide that provides results for hcv treatment.
EP11717518.2A EP2558592B1 (en) 2010-04-13 2011-04-11 Single nucleotide polymorphisms that predict hcv treatment outcomes
HK13107392.4A HK1180018A1 (en) 2010-04-13 2013-06-25 Single nucleotide polymorphisms that predict hcv treatment outcomes hcv

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US32350210P 2010-04-13 2010-04-13
US61/323,502 2010-04-13

Related Child Applications (2)

Application Number Title Priority Date Filing Date
EP13152518.0A Previously-Filed-Application EP2589669B1 (en) 2010-04-13 2011-04-11 Single nucleotide polymorphisms that predict HCV treatment outcomes
EP13152522.2A Previously-Filed-Application EP2589670B1 (en) 2010-04-13 2011-04-11 Single nucleotide polymorphisms that predict HCV treatment outcomes

Publications (1)

Publication Number Publication Date
WO2011128278A1 true WO2011128278A1 (en) 2011-10-20

Family

ID=44275673

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/EP2011/055579 WO2011128278A1 (en) 2010-04-13 2011-04-11 Single nucleotide polymorphisms that predict hcv treatment outcomes

Country Status (12)

Country Link
US (1) US20120094389A1 (en)
EP (3) EP2589670B1 (en)
JP (1) JP5695181B2 (en)
KR (1) KR101493137B1 (en)
CN (1) CN102844448B (en)
BR (1) BR112012025854A2 (en)
CA (1) CA2795911A1 (en)
ES (3) ES2525300T3 (en)
HK (1) HK1180018A1 (en)
MX (1) MX2012011804A (en)
RU (1) RU2567663C2 (en)
WO (1) WO2011128278A1 (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2013079424A1 (en) * 2011-11-28 2013-06-06 F. Hoffmann-La Roche Ag A single nucleotide polymorphism on chromosome 15 that predicts hcv treatment responses

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4695623A (en) 1982-05-06 1987-09-22 Amgen Consensus human leukocyte interferon
US20050014159A1 (en) * 2003-07-15 2005-01-20 Jui-Lin Chen Method for detecting a propensity of an individual to response effectively to treatment of interferon-alpha and ribavirin combined therapy
US20050260567A1 (en) * 2004-04-29 2005-11-24 Roche Palo Alto Llc NS5A nucleotide sequence variation as a marker for interferon response
WO2009046369A2 (en) * 2007-10-05 2009-04-09 Medtronic, Inc. Use of a specific dosage regimen of ifn-alpha and ribavirin for treating hepatitis c

Family Cites Families (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6824768B2 (en) * 1998-12-18 2004-11-30 Schering Corporation Ribavirin-pegylated interferon alfa induction HCV combination therapy

Patent Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4695623A (en) 1982-05-06 1987-09-22 Amgen Consensus human leukocyte interferon
US4897471A (en) 1982-05-06 1990-01-30 Amgen Consensus human leukocyte interferon
US20050014159A1 (en) * 2003-07-15 2005-01-20 Jui-Lin Chen Method for detecting a propensity of an individual to response effectively to treatment of interferon-alpha and ribavirin combined therapy
US20050260567A1 (en) * 2004-04-29 2005-11-24 Roche Palo Alto Llc NS5A nucleotide sequence variation as a marker for interferon response
WO2009046369A2 (en) * 2007-10-05 2009-04-09 Medtronic, Inc. Use of a specific dosage regimen of ifn-alpha and ribavirin for treating hepatitis c

Non-Patent Citations (38)

* Cited by examiner, † Cited by third party
Title
ASKARIEH G, ALSIO A, PUGNALE P, NEGRO F, FERRARI C, NEUMANN AU, PAWLOTSKY JM, SCHALM SW, ZEUZEM S, NORKRANS G: "Systemic and intrahepatic interferon-gamma-inducible protein 10 kDa predicts the first- phase decline in hepatitis C virus RNA and overall viral response to therapy in chronic hepatitis C", HEPATOLOGY, 2009
ASSELAH T, BENHAMOU Y, MARCELLIN P: "Protease and polymerase inhibitors for the treatment of hepatitis C", LIVER INT, vol. 29, no. 1, 2009, pages 57 - 67
ASSELAH T, BIECHE I, NARGUET S, SABBAGH A, LAURENDEAU I, RIPAULT MP, BOYER N, MARTINOT-PEIGNOUX M, VALLA D, VIDAUD M: "Liver gene expression signature to predict response to pegylated interferon plus ribavirin combination therapy in patients with chronic hepatitis C.", GUT, vol. 57, 2008, pages 516 - 524
ASSELAH T, BIECHE I, SABBAGH A, BEDOSSA P, MOREAU R, VALLA D, VIDAUD M, MARCELLIN P: "Gene expression and hepatitis C virus infection", GUT, vol. 58, 2009, pages 846 - 858
BARRETT JC, FRY B, MALLER J, DALY MJ: "Haploview: analysis and visualization of LD and haplotype maps", BIOINFORMATICS, vol. 21, 2005, pages 263 - 265
CONJEEVARAM HS, FRIED MW, JEFFERS LJ, TERRAULT NA, WILEY-LUCAS TE, AFDHAL N, BROWN RS, BELLE SH, HOOFNAGLE JH, KLEINER DE: "Peginterferon and ribavirin treatment in African American and Caucasian American patients with hepatitis C genotype 1", GASTROENTEROLOGY, vol. 131, 2006, pages 470 - 477
DIENSTAG JL, MCHUTCHISON JG: "American Gastroenterological Association technical review on the management of hepatitis C", GASTROENTEROLOGY, vol. 130, 2006, pages 231 - 264
ESTEBAN M: "Hepatitis C and evasion of the interferon system: a PKR paradigm", CELL HOST MICROBE, vol. 6, 2009, pages 495 - 497
FERENCI P, FRIED MW, SHIFFMAN ML, SMITH CI, MARINOS G, GONCALES FL, JR., HAUSSINGER D, DIAGO M, CAROSI G, DHUMEAUX D: "Predicting sustained virological responses in chronic hepatitis C patients treated with peginterferon alfa-2a (40 KD)/ribavirin", J HEPATOL, vol. 43, 2005, pages 425 - 433
FOY E, LI K, WANG C, SUMPTER R, JR., IKEDA M, LEMON SM, GALE M, JR.: "Regulation of interferon regulatory factor-3 by the hepatitis C virus serine protease", SCIENCE, vol. 300, 2003, pages 1145 - 1148
FRIED MW, SHIFFMAN ML, REDDY KR, SMITH C, MARINOS G, GONCALES FL, JR., HAUSSINGER D, DIAGO M, CAROSI G, DHUMEAUX D: "Peginterferon alfa-2a plus ribavirin for chronic hepatitis C virus infection", N ENGL J MED, vol. 347, 2002, pages 975 - 982
GABRIEL SB, SCHAFFUER SF, NGUYEN H, MOORE JM, ROY J, BLUMENSTIEL B, HIGGINS J, DEFELICE M, LOCHNER A, FAGGART M: "The structure of haplotype blocks in the human genome", SCIENCE, vol. 296, 2002, pages 2225 - 2229
GANE E, ROBERTS S, STEDMAN C, ANGUS P, RITCHIE B, ELSTON R, IPE D, MORCOS P, NAJERA I, CHU T: "Combination therapy with a nucleoside polymerase (R7128) and protease (R7227/ITMN-191) inhibitor in HCV: safety, pharmacokinetics , and virologic results from INFORM- [abstract 193", HEPATOLOGY, vol. 50, 2009, pages 394 - 5A
GE D, FELLAY J, THOMPSON AJ, SIMON JS, SHIANNA KV, URBAN TJ, HEINZEN EL, QIU P, BERTELSEN AH, MUIR AJ: "Genetic variation in IL28B predicts hepatitis C treatment-induced viral clearance", NATURE, vol. 461, 2009, pages 399 - 401
GHANY MG, STRADER DB, THOMAS DL, SEEFFLB.: "Diagnosis, management, and treatment of hepatitis C: An update", HEPATOLOGY, vol. 49, 2009, pages 1335 - 1374
HADZIYANNIS SJ, SETTE H, JR., MORGAN TR, BALAN V, DIAGO M, MARCELLIN P, RAMADORI G, BODENHEIMER H, JR., BERNSTEIN D, RIZZETTO M: "Peginterferon-alpha2a and ribavirin combination therapy in chronic hepatitis C: a randomized study of treatment duration and ribavirin dose", ANN INTERN MED, vol. 140, 2004, pages 346 - 355
JENSEN DM, MARCELLIN P, FREILICH B, ANDREONE P, DI BA, BRANDAO-MELLO CE, REDDY KR, CRAXI A, MARTIN AO, TEUBER G: "Re-treatment of patients with chronic hepatitis C who do not respond to peginterferon-alpha2b: a randomized trial", ANN INTERN MED, vol. 150, 2009, pages 528 - 540
LANFORD RE, HILDEBRANDT-ERIKSEN ES, PETRI A, PERSSON R, LINDOW M, MUNK ME, KAUPPINEN S, ORUM H: "Therapeutic silencing ofmicroRNA-122 in primates with chronic hepatitis C virus infection", SCIENCE, vol. 327, 2010, pages 198 - 201
MANNS MP, MCHUTCHISON JG, GORDON SC, RUSTGI VK, SHIFFMAN M, REINDOLLAR R, GOODMAN ZD, KOURY K, LING M, ALBRECHT JK: "Peginterferon alfa-2b plus ribavirin compared with interferon alfa-2b plus ribavirin for initial treatment of chronic hepatitis C: a randomised trial", LANCET, vol. 358, 2001, pages 958 - 965
MARCELLO T, GRAKOUI A, BARBA-SPAETH G, MACHLIN ES, KOTENKO SV, MACDONALD MR, RICE CM: "Interferons alpha and lambda inhibit hepatitis C virus replication with distinct signal transduction and gene regulation kinetics", GASTROENTEROLOGY, vol. 131, 2006, pages 1887 - 1898
MARTINOT-PEIGNOUX M, MAYLIN S, MOUCARI R, RIPAULT MP, BOYER N, CARDOSO AC, GIUILY N, CASTELNAU C, POUTEAU M, STERN C: "Virological response at 4 weeks to predict outcome of hepatitis C treatment with pegylated interferon and ribavirin", ANTIVIR THER, vol. 14, 2009, pages 501 - 511
MCCARTHY JJ, LI JH, THOMPSON A, SUCHINDRAN S, LAO XQ, PATEL K, TILLMANN HL, MUIR AJ, MCHUTCHISON JG: "Replicated association between an interleukin-28B gene variant and a sustained response to pegylated interferon and ribavirin [manuscript in press]", GASTROENTEROLOGY, 2010
MISSIHA S, HEATHCOTE J, ARENOVICH T, KHAN K: "Impact of asian race on response to combination therapy with peginterferon alfa-2a and ribavirin in chronic hepatitis C", AM J GASTROENTEROL, vol. 102, 2007, pages 2181 - 2188
MONTES-CANO M ET AL.: "IL28B genetic variants and hepatitis virus infection by different viral genotypes [manuscript in press]", HEPATOLOGY, 2010
NEUMANN AU, LAM NP, DAHARI H, GRETCH DR, WILEY TE, LAYDEN TJ, PERELSON AS: "Hepatitis C viral dynamics in vivo and the antiviral efficacy of interferon-alpha therapy", SCIENCE, vol. 282, 1998, pages 103 - 107
PURCELL S, NEALE B, TODD-BROWN K, THOMAS L, FERREIRA MA, BENDER D, MALLER J, SKLAR P, DE BAKKER PI, DALY MJ: "PLINK: a tool set for whole-genome association and population-based linkage analyses", AM J HUM GENET, vol. 81, 2007, pages 559 - 575
RAUCH A, KUTALIK Z, DESCOMBES P, CAI T, DI IJ, MUELLER T, BOCHUD M, BATTEGAY M, BERNASCONI E, BOROVICKA J: "Genetic Variation in IL28B Is Associated With Chronic Hepatitis C and Treatment Failure: A Genome-wide Association Study", GASTROENTEROLOGY, 2010
RAUCH ANDRI ET AL: "Genetic variation in IL28B is associated with chronic hepatitis C and treatment failure: a genome-wide association study", GASTROENTEROLOGY, ELSEVIER, PHILADELPHIA, PA, vol. 138, no. 4, 1 April 2010 (2010-04-01), pages 1338 - 1345,13, XP002601554, ISSN: 0016-5085, DOI: DOI:10.1053/J.GASTRO.2009.12.056 *
REDDY KR, MESSINGER D, POPESCU M, HADZIYANNIS SJ: "Peginterferon alpha-2a (40 kDa) and ribavirin: comparable rates of sustained virological response in sub-sets of older and younger HCV genotype 1 patients.", J VIRAL HEPAT, vol. 16, 2009, pages 724 - 731
REESINK HW, ZEUZEM S, WEEGINK CJ, FORESTIER N, VAN VA, VAN DE WETERING DE ROOIJ, MCNAIR L, PURDY S, KAUFFMAN R, ALAM J: "Rapid decline of viral RNA in hepatitis C patients treated with VX-950: a phase Ib, placebo-controlled, randomized study", GASTROENTEROLOGY, vol. 131, 2006, pages 997 - 1002
ROBEK MD, BOYD BS, CHISARI FV: "Lambda interferon inhibits hepatitis B and C virus replication", J VIROL, vol. 79, 2005, pages 3851 - 3854
SIREN J, PIRHONEN J, JULKUNEN I, MATIKAINEN S: "IFN-alpha regulates TLR-dependent gene expression of IFN-alpha, IFN-beta, IL-28, and IL-29", J IMMUNOL, vol. 174, 2005, pages 1932 - 1937
SUPPIAH V, MOLDOVAN M, AHLENSTIEL G, BERG T, WELTMAN M, ABATE ML, BASSENDINE M, SPENGLER U, DORE GJ, POWELL E: "IL28B is associated with response to chronic hepatitis C interferon-alpha and ribavirin therapy", NAT GENET, vol. 41, 2009, pages 1100 - 1104
SUPPIAH VIJAYAPRAKASH ET AL: "IL28B is associated with response to chronic hepatitis C interferon-alpha and ribavirin therapy", NATURE GENETICS, NATURE PUBLISHING GROUP, US, vol. 41, no. 10, 1 October 2009 (2009-10-01), pages 1100 - 1104, XP002601552, ISSN: 1546-1718, [retrieved on 20090913], DOI: DOI:10.1038/NG.447 *
TANAKA Y, NISHIDA N, SUGIYAMA M, KUROSAKI M, MATSUURA K, SAKAMOTO N, NAKAGAWA M, KORENAGA M, HINO K, HIGE S: "Genome-wide association of IL28B with response to pegylated interferon-alpha and ribavirin therapy for chronic hepatitis C.", NAT GENET, vol. 41, 2009, pages 1105 - 1109
TANAKA YASUHITO ET AL: "Genome-wide association of IL28B with response to pegylated interferon-alpha and ribavirin therapy for chronic hepatitis C", NATURE GENETICS, NATURE PUBLISHING GROUP, US, vol. 41, no. 10, 1 October 2009 (2009-10-01), pages 1105 - 1109, XP002602520, ISSN: 1546-1718, [retrieved on 20090913], DOI: DOI:10.1038/NG.449 *
THOMAS DL, THIO CL, MARTIN MP, QI Y, GE D, O'HUIGIN C, KIDD J, KIDD K, KHAKOO SI, ALEXANDER G: "Genetic variation in IL28B and spontaneous clearance of hepatitis C virus", NATURE, vol. 461, 2009, pages 798 - 801
YEN Y-H ET AL: "Mutations in the interferon sensitivity-determining region (nonstructural 5A amino acid 2209-2248) in patients with hepatitis C-1b infection and correlating response to combined therapy of pegylated interferon and ribavirin", ALIMENTARY PHARMACOLOGY & THERAPEUTICS, BLACKWELL SCIENTIFIC PUBLICATIONS LTD., CAMBRIDGE, GB, vol. 27, no. 1, 1 January 2008 (2008-01-01), pages 72 - 79, XP002582304, ISSN: 0269-2813, [retrieved on 20071025], DOI: DOI:10.1111/J.1365-2036.2007.03560.X *

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2013079424A1 (en) * 2011-11-28 2013-06-06 F. Hoffmann-La Roche Ag A single nucleotide polymorphism on chromosome 15 that predicts hcv treatment responses
CN103987862A (en) * 2011-11-28 2014-08-13 弗·哈夫曼-拉罗切有限公司 A single nucleotide polymorphism on chromosome 15 that predicts HCV treatment responses
RU2606759C2 (en) * 2011-11-28 2017-01-10 Ф. Хоффманн-Ля Рош Аг Single nucleotide polymorphism on chromosome 15 enabling prediction of hcv treatment responses

Also Published As

Publication number Publication date
CN102844448B (en) 2015-11-25
ES2528899T3 (en) 2015-02-13
CA2795911A1 (en) 2011-10-20
HK1180018A1 (en) 2013-10-11
CN102844448A (en) 2012-12-26
ES2525290T3 (en) 2014-12-19
JP2013523168A (en) 2013-06-17
EP2558592B1 (en) 2014-10-22
BR112012025854A2 (en) 2017-01-10
EP2558592A1 (en) 2013-02-20
EP2589670A1 (en) 2013-05-08
EP2589669A1 (en) 2013-05-08
MX2012011804A (en) 2012-12-17
EP2589669B1 (en) 2014-10-22
ES2525300T3 (en) 2014-12-19
KR101493137B1 (en) 2015-02-12
JP5695181B2 (en) 2015-04-01
RU2012145777A (en) 2014-05-20
KR20130009842A (en) 2013-01-23
RU2567663C2 (en) 2015-11-10
US20120094389A1 (en) 2012-04-19
EP2589670B1 (en) 2014-12-17

Similar Documents

Publication Publication Date Title
Asselah et al. IL28B polymorphism is associated with treatment response in patients with genotype 4 chronic hepatitis C
Honda et al. Hepatic ISG expression is associated with genetic variation in interleukin 28B and the outcome of IFN therapy for chronic hepatitis C
US20120094284A1 (en) Prediction of Early Virological Response in HCV Treatment
Derbala et al. The predictive value of IL28B rs12979860, rs11881222 and rs8099917 polymorphisms and IP-10 in the therapeutic response of Egyptian genotype 4 patients
EP2459210A1 (en) Methods for diagnosing or predicting hepatitis c outcome in hcv infected patients
AU2011287642B2 (en) Prediction of HCV viral kinetics in interferon-free treatment
Abe et al. Inverse association of IL28B genotype and liver mRNA expression of genes promoting or suppressing antiviral state
EP2785875B1 (en) A single nucleotide polymorphism on chromosome 15 that predicts hcv treatment responses
EP2558592B1 (en) Single nucleotide polymorphisms that predict hcv treatment outcomes
Wu et al. IFNL4 ss469415590 polymorphism contributes to treatment decisions in patients with chronic hepatitis C virus genotype 1b, but not 2a, infection
Shaker et al. Is rs8099917 polymorphism of IL-28B gene a good predictor of response to therapy of HCV than rs12979860? An Egyptian study
MOHAMED et al. Interleukin 28b Single Nucleotide Polymorphism (rs8099917) as a Predictive Factor of Response to Pegylated-IFN/Ribavirintherapy in Chronic HCV Patients
Shehata et al. Interleukin 28B polymorphism as a predictive factor to treatment response in chronic hepatitis c genotype 4 Egyptian patients
Efat et al. Vitamin D receptor polymorphisms as predictors of response to interferon/ribavirin-based therapy in Egyptians with chronic hepatitis C infection
Hai et al. Relationship between ITPA genotype and outcome of extended therapy in HCV patients with a LVR to PEG-IFN and RBV
Fellay et al. analysis (Table IV). Regardless of the drug adherence, end of treatment responserates of patients infected with genotype 2 were around 94–99%, but the sustained virological response rates of the patients who received
Hosaka et al. oRIGINAL ARTIci. E

Legal Events

Date Code Title Description
WWE Wipo information: entry into national phase

Ref document number: 201180018656.0

Country of ref document: CN

121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 11717518

Country of ref document: EP

Kind code of ref document: A1

WWE Wipo information: entry into national phase

Ref document number: 2011717518

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 2259/MUMNP/2012

Country of ref document: IN

ENP Entry into the national phase

Ref document number: 2795911

Country of ref document: CA

WWE Wipo information: entry into national phase

Ref document number: MX/A/2012/011804

Country of ref document: MX

WWE Wipo information: entry into national phase

Ref document number: 2013504214

Country of ref document: JP

NENP Non-entry into the national phase

Ref country code: DE

ENP Entry into the national phase

Ref document number: 20127029599

Country of ref document: KR

Kind code of ref document: A

ENP Entry into the national phase

Ref document number: 2012145777

Country of ref document: RU

Kind code of ref document: A

REG Reference to national code

Ref country code: BR

Ref legal event code: B01A

Ref document number: 112012025854

Country of ref document: BR

ENP Entry into the national phase

Ref document number: 112012025854

Country of ref document: BR

Kind code of ref document: A2

Effective date: 20121009