WO2013024158A1 - Combinaisons d'inhibiteurs de protéine kinase et d'interférons ou d'inhibiteurs de protéine kinase et d'antiviraux agissant directement pour le traitement et la prévention d'une infection par le vhc - Google Patents

Combinaisons d'inhibiteurs de protéine kinase et d'interférons ou d'inhibiteurs de protéine kinase et d'antiviraux agissant directement pour le traitement et la prévention d'une infection par le vhc Download PDF

Info

Publication number
WO2013024158A1
WO2013024158A1 PCT/EP2012/066109 EP2012066109W WO2013024158A1 WO 2013024158 A1 WO2013024158 A1 WO 2013024158A1 EP 2012066109 W EP2012066109 W EP 2012066109W WO 2013024158 A1 WO2013024158 A1 WO 2013024158A1
Authority
WO
WIPO (PCT)
Prior art keywords
interferon
ifn
hcv
protein kinase
combination
Prior art date
Application number
PCT/EP2012/066109
Other languages
English (en)
Inventor
Joachim Lupberger
Thomas Baumert
Original Assignee
INSERM (Institut National de la Santé et de la Recherche Médicale)
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 INSERM (Institut National de la Santé et de la Recherche Médicale) filed Critical INSERM (Institut National de la Santé et de la Recherche Médicale)
Publication of WO2013024158A1 publication Critical patent/WO2013024158A1/fr

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/517Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim ortho- or peri-condensed with carbocyclic ring systems, e.g. quinazoline, perimidine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/41Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with two or more ring hetero atoms, at least one of which being nitrogen, e.g. tetrazole
    • A61K31/425Thiazoles
    • A61K31/427Thiazoles not condensed and containing further heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/496Non-condensed piperazines containing further heterocyclic rings, e.g. rifampin, thiothixene or sparfloxacin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/506Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim not condensed and containing further heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/19Cytokines; Lymphokines; Interferons
    • A61K38/21Interferons [IFN]

Definitions

  • HCV Hepatitis C virus
  • liver disease including fibrosis and steatosis (Chisari, Nature, 2005, 435: 930-932).
  • About 20% of patients with chronic HCV infection develop liver cirrhosis, which progresses to hepatocellular carcinoma in 5% of the cases (Hoofnagle, Hepatology, 2002, 36: S21-S29; Blonski et al, Clin. Liver Dis., 2008, 12: 661-674; Jacobson et al, Clin. Gastroenterol. Hepatol, 2010, 8: 924-933; Castello et al., Clin. Immunol, 2010, 134: 237-250; McGivern et al., Oncogene, 2011, 30: 1969-1983).
  • Chronic HCV infection is the leading indication for liver transplantations (Seeff et al., Hepatology, 2002, 36: 1-2).
  • liver transplantation is not a cure for hepatitis C; viral recurrence being an invariable problem and the leading cause of graft loss (Brown, Nature, 2005, 436: 973-978; Watt et al, Am. J. Transplant, 2009, 9: 1707-1713).
  • Current therapies include administration of ribavirin and/or interferon-alpha (IFN-Cc), two non-specific anti-viral agents.
  • IFN-Cc interferon-alpha
  • HCV entry into target cells is a promising target for antiviral preventive and therapeutic strategies since it is essential for initiation, spread, and maintenance of infection (Timpe et al, Gut, 2008, 57: 1728-1737; Zeisel et al, Hepatology, 2008, 48: 299-307). Indeed, HCV initiates infection by attaching to molecules or receptors on the surface of hepatocytes. Current evidence suggests that HCV entry is a multistep process involving several host factors including heparan sulfate (Barth et al, J. Biol.
  • entry inhibitors are a promising novel class of antivirals for prevention and treatment of HCV infection (for review see Zeisel et al, J. Hepatol, 2011, 54: 566-576).
  • Entry inhibitors in preclinical or early clinically development include HCV-receptor- and HCV-envelop-specific antibodies as well as small molecules (Zeisel et al, J. Hepatol, 2011, 54: 566-576; Catanese et al, J. Virol, 2007, 81 : 8063-8071; Fafi-Kremer et al, J. Exp.
  • EP 1 256 348 discloses substances, including antibodies, with antiviral effects that inhibit binding of HCV E2 and CD81.
  • International patent application WO 2007/130646 describes in vitro and cell-based assays for identifying agents that interfere with HCV interactions with Claudin-1 thereby preventing HCV infection.
  • the present Applicants have generated monoclonal antibodies that efficiently inhibit HCV infection by targeting host entry factor Claudin-1 (EP 08 305 597 and WO 2010/034812).
  • HCV entry co-factors epidermal growth factor receptor (EGFR) and ephrin type-A receptor 2 (EphA2)
  • EGFR epidermal growth factor receptor
  • EphA2 ephrin type-A receptor 2
  • the present invention relates to systems and improved strategies for the prevention and/or treatment of HCV infection and HCV-related diseases. More specifically, the present Applicants have demonstrated that interferon-alpha in combination with erlotinib or with dasatinib (which are both protein kinase inhibitors) act in a highly synergistic manner on the inhibition of HCV infection (see Example 1), suggesting that a combination of a protein kinase inhibitor and an interferon may be an effective antiviral approach to prevent primary HCV infection, such as after liver transplantation, and might also restrain virus spread in chronically infected patients.
  • the present invention provides a combination of at least one protein kinase inhibitor and at least one interferon for use in the treatment or prevention of HCV infection.
  • the at least one interferon is a human interferon.
  • the human interferon may be a natural human interferon, a recombinant human interferon, a synthetic version of a human interferon, or derivatives thereof.
  • the at least one interferon of an inventive combination is selected from the group consisting of interferon-alpha (IFN-Cc), pegylated IFN-CC, albumin-IFN-cc, interferon-beta (IFN- ⁇ ), pegylated IFN- ⁇ , albumin- IFN- ⁇ , interferon-omega (IFN-CO), pegylated IFN-CO, albumin-IFN-CO, interferon- gamma (IFN- ⁇ ), pegylated IFN- ⁇ , albumin-IFN- ⁇ , interferon-lambda (IFN- ⁇ ), pegylated IFN- ⁇ , albumin-IFN- ⁇ , equivalents thereof, analogs thereof, derivatives thereof and any combination thereof.
  • the at least on interferon is interferon-alpha (IFN-Cc), pegylated IFN-CC, or albumin-IFN-CC.
  • the at least one interferon of an inventive combination is interferon-alpha-a (IFN-Cc2a) or interferon-alpha-b (IFN-Cc2b).
  • the present invention provides a combination of at least one protein kinase inhibitor and at least one direct acting antiviral for use in the treatment or prevention of HCV infection.
  • the at least one direct acting antiviral is a HCV protease inhibitor or a HCV polymerase inhibitor of an NS5A inhibitor.
  • the at least one direct acting antiviral is selected from the group consisting of telaprevir, boceprevir, danoprevir, TMC-435, daclatasvir, mericitabine and GS7977.
  • the at least one protein kinase inhibitor is a tyrosine kinase inhibitor.
  • the tyrosine kinase inhibitor may be a tyrosine kinase inhibitor that acts on the epidermal growth factor receptor (EGFR) or a tyrosine kinase inhibitor that acts on the ephrin type-A receptor 2 (EphA2).
  • Tyrosine kinase inhibitors that act on EGFR may be selected from the group consisting of erlotinib, gefitinib, vandetanib, lapatinib, neratinib, afatinib, equivalents thereof, and any combination thereof.
  • Tyrorine kinase inhibitors that act on EphA2 may be dasatinib.
  • the tyrosine kinase inhibitor is an anti-receptor tyrosine kinase antibody. In other embodiments, the tyrosine kinase inhibitor is a small molecule.
  • the protein kinase inhibitor and interferon or the protein kinase inhibitor and the direct acting antiviral act in a highly synergistic manner to inhibit HCV infection.
  • the interferon or the direct acting antiviral decreases the IC 50 for the inhibition of HCV infection by the protein kinase inhibitor by a factor of at least 10 fold or at least 25 fold, preferably at least 50 fold or at least 75 fold, more preferably at least 100 fold or 200 fold, and even more preferably more than 200 fold (e.g., more than 350 fold).
  • the protein kinase inhibitor decreases the IC 50 for the inhibition of HCV infection by the interferon or the direct acting antiviral by a factor of at least 10 fold or at least 25 fold, preferably at least 50 fold or at least 75 fold, more preferably at least 100 fold or 200 fold, and even more preferably more than 200 fold (e.g., more than 350 fold).
  • the combination index (CI) of the at least one protein kinase inhibitor and at least one interferon or direct acting antiviral is lower than 1 , preferably lower than 0.90, more preferably lower than 0.80, and even more preferably lower than 0.50.
  • the combinations of the present invention can find application in a variety of prophylactic and therapeutic treatments.
  • the combinations are provided for use in the prevention of HCV infection of a cell (e.g., a susceptible cell or a population of susceptible cells); for preventing or treating HCV infection or a HCV-related disease in a subject; for controlling chronic HCV infection; and for preventing HCV recurrence in a liver transplantation patient.
  • HCV infection may be due to HCV of a genotype selected from the group consisting of genotype 1 , genotype 2, genotype 3, genotype 4, genotype 5, genotype 6, and genotype 7, or more specifically of a subtype selected from the group consisting of subtype la, subtype lb, subtype 2a, subtype 2b, subtype 2c, subtype 3a, subtype 4a-f, subtype 5a, and subtype 6a.
  • a genotype selected from the group consisting of genotype 1 , genotype 2, genotype 3, genotype 4, genotype 5, genotype 6, and genotype 7, or more specifically of a subtype selected from the group consisting of subtype la, subtype lb, subtype 2a, subtype 2b, subtype 2c, subtype 3a, subtype 4a-f, subtype 5a, and subtype 6a.
  • the present invention provides a method of reducing the likelihood of a susceptible cell of becoming infected with HCV as a result of contact with HCV, which comprises contacting the susceptible cell with an effective amount of an inventive combination. Also provided is a method of reducing the likelihood of a subject's susceptible cells of becoming infected with HCV as a result of contact with HCV, which comprises administering to the subject an effective amount of an inventive combination or a pharmaceutical composition thereof.
  • the present invention also provides a method of treating or preventing HCV infection or a HCV- associated disease (e.g., a liver disease or pathology) in a subject in need thereof, which comprises administering to the subject an effective amount of an inventive combination or a pharmaceutical composition thereof.
  • the invention also provides a method for controlling chronic HCV infection in a subject in need thereof, which comprises administering to the subject an effective amount of an inventive combination or a pharmaceutical composition thereof. Also provided is a method of preventing HCV recurrence in a liver transplantation patient, which comprises administering to the patient an effective amount of an inventive combination or a pharmaceutical composition thereof.
  • Administration of an inventive combination to a subject may be by any suitable route, including, for example, parenteral, aerosol, oral and topical routes.
  • the inventive combination, or pharmaceutical composition thereof may be administered alone or in combination with a therapeutic agent, such as an anti-viral agent.
  • the HCV infection or HCV-related disease or HCV re-infection may be caused by a Hepatitis C virus that is resistant to a direct acting antiviral and/or that is transmitted by cell-cell transmission.
  • inventive combinations may be administered per se or as pharmaceutical compositions. Accordingly, in another aspect, the present invention provides for the use of an inventive combination for the manufacture of medicaments, pharmaceutical compositions, or pharmaceutical kits for the treatment and/or prevention of HCV infection and HCV-associated diseases.
  • the present invention provides a pharmaceutical composition
  • a pharmaceutical composition comprising an effective amount of an inventive combination (i.e., at least one protein kinase inhibitor and at least one interferon as described herein or at least one protein kinase inhibitor and at least one direct acting antiviral) and at least one pharmaceutically acceptable carrier or excipient.
  • the pharmaceutical composition is adapted for administration in combination with an additional therapeutic agent, such as an antiviral agent.
  • the pharmaceutical composition further comprises an additional therapeutic agent, such as an antiviral agent.
  • Antiviral agents suitable for use in methods and pharmaceutical compositions of the present invention include, but are not limited to, ribavirin, anti- HCV (monoclonal or polyclonal) antibodies, RNA polymerase inhibitors, protease inhibitors, IRES inhibitors, helicase inhibitors, antisense compounds, ribozymes, entry inhibitors, micro-RNA antagonists, cytokines, therapeutic vaccines, NS5A antagonists, polymerase inhibitors, cyclophilin A antagonists, and any combination thereof.
  • Figure 1 is a set of graphs illustrating the synergistic effects of interferon-alpha and the protein kinase inhibitors, erlotinib and dasatinib, on the inhibition of HCVcc infection.
  • Combination of EGFR inhibitor erlotinib with IFN-a potentiates the antiviral impact of IFN-a.
  • Huh7.5.1 cells were pre-incubated for 1 hour with serial concentrations of (A) IFN-a2a or (B) IFN-a2b and 0.1 ⁇ erlotinib or dasatinib before incubation with HCVcc Luc-Jcl in the presence of both compounds. Dotted lines at CI values of 0.9 and 1.1 indicate the boundaries of an additive interaction.
  • FIG. 2 is a set of graphs illustrating the synergistic effects of interferon-alpha and the protein kinase inhibitor, dasatinib, on the inhibition of HCVcc infection.
  • Huh7.5.1 cells were pre-incubated for 1 hour with serial concentrations of (A) IFN- a2a or (B) IFN-a2b and 0.1 ⁇ dasatinib before incubation with HCVcc Luc-Jcl in the presence of both compounds.
  • Combination of IFN-a with 0.1 ⁇ dasatinib resulted in a shift in the IC 50 of IFN-a2a and IFN-a2b up to 10 fold.
  • C-D Synergy was confirmed using three-dimensional analysis of the inhibitor- inhibitor combinations according to Prichard and Shipman. Surface amplitudes >20 % above the zero plane highlight inhibitor combinations acting synergistically. One representative experiment is shown.
  • FIG. 3 is a set of graphs showing the synergistic activity of combinations of protein kinase inhibitors and DAAs.
  • Huh7.5.1 cells were pre-incubated for 1 hour with serial concentrations of (A) a protease inhibitors: telaprevir, boceprevir, TMC- 435 or danoprevir, (B) a NS5A inhibitor: daclatasvir or (C) a polymerase inhibitor: mericitabine or GS-7977 and 0.1 ⁇ PKIs (erlotinib or dasatinib) before incubation with HCVcc Luc-Jcl in the presence of both compounds.
  • Dotted lines at combination values of 0.9 and 1.1 indicate the boundaries of an additive interaction.
  • Figure 4 is a set of graphs showing the synergistic activity of combinations of the protein kinase inhibitor, erlotinib and the DAA, telaprevir. Combinations were performed as in Figure 3.
  • Huh7.5.1 cells were pre-incubated for 1 hour with serial concentrations of protease inhibitor telaprevir and protein kinase inhibitors (PKIs) (erlotinib or dasatinib) before incubation with HCVcc Luc-Jcl in the presence of both compounds.
  • PKIs protein kinase inhibitors
  • HCVcc infection was analysed by quantification of luciferase reporter gene expression.
  • Figure 5 is a set of graphs showing the synergistic activity of combinations of the protein kinase inhibitor, erlotinib and the DAA, daclatasvir. Combinations were performed as in Figures 3 and 4.
  • (A) %HCV infection relative to cells infected in the absence of inhibitors is shown. Data are indicated as means ⁇ SEM from at least three independent experiments performed in triplicate are shown (HCV infection in the absence of inhibitors 100%).
  • Figure 6 is a set of graphs showing the synergistic activity of combinations of a protein kinase inhibitors, erlotinib or dasatinib and the DAA, GS-7977. Combinations were performed as in Figure 3. Combination of GS-7977 and (A) dasatinib or (B) erlotinib decreased the IC 50 of GS-7977 up to 210 fold. Means ⁇ SEM from at least three independent experiments performed in triplicate are shown. (C) Synergy was confirmed using the method of Prichard and Shipman. One representative experiment is shown.
  • Figure 7 is a set of graphs showing that protein kinase inhibitors inhibit cell-free entry of protease inhibitor-resistant variants without cross-resistance.
  • Huh7.5.1 cells were pre-incubated for 1 hour with serial concentrations of (A) telaprevir, (B) boceprevir, (C) erlotinib or respective isotype control reagents before incubation with HCVcc-Jcl-Luc containing the DAA-resistant mutations R155T and A156S, respectively, in the presence of each compound.
  • HCV infection was analyzed 72 hours post-incubation by luciferase reporter gene expression in cell lysates as described in Example 7. Means ⁇ SEM from at least three independent experiments performed in triplicate are shown.
  • Figure 8 is a set of graphs showing cell-cell transmission of protease inhibitor resistant variants and its inhibition by erlotinib.
  • NS5A+ HCV producer cells (Pi) were transfected with HCV RNA encoding for HCV Luc-Jcl A156S (A-C) or Jcl L36M R155K (D-F).
  • NS5A+ HCV producer cells and target GFP-expressing cells were co-cultivated with an anti-E2 mAb to block cell-free transmission.
  • Protease inhibitor- resistant HCV variant Luc-Jcl A156S (A-C) and Jcl L36M R155K (D-F) producer cells (Pi) cultured with uninfected target cells (T) were then incubated with erlotinib (B and E) or control IgG (A and D). HCV-infected target cells were quantified by flow cytometry. Cell-cell transmission of DAA-resistant variants, not affected by controls, is inhibited by erlotinib. Percentage of infected target cells is shown as histograms (C and F) and is represented as means ⁇ SD from three experiments performed in triplicate.
  • the term "subject” refers to a human or another mammal ⁇ e.g., primate, dog, cat, goat, horse, pig, mouse, rat, rabbit, and the like), that can be the host of Hepatitis C virus (HCV), but may or may not be infected with the virus, and/or may or may not suffer from a HCV-related disease.
  • HCV Hepatitis C virus
  • Non-human subjects may be transgenic or otherwise modified animals.
  • the subject is a human being. In such embodiments, the subject is often referred to as an individual".
  • the term "individual” does not denote a particular age, and thus encompasses newborns, children, teenagers, and adults.
  • HCV refers to any major HCV genotype, subtype, isolate and/or quasispecies.
  • HCV genotypes include, but are not limited to, genotypes 1, 2, 3, 4, 5, 6 and 7;
  • HCV subtypes include, but are not limited to, subtypes la, lb, 2a, 2b, 2c, 3a, 4a-f, 5a and 6a.
  • HCV infection refers to the introduction of HCV genetic information into a target cell, such as by fusion of the target cell membrane with HCV or an HCV envelope glycoprotein-positive cell.
  • HCV-related disease and “HCV-associated disease” are herein used interchangeably. They refer to any disease or disorder known or suspected to be associated with and/or caused, directly or indirectly, by HCV.
  • HCV-related (or HCV- associated) diseases include, but are not limited to, a wide variety of liver diseases, such as subclinical carrier state of acute hepatitis, chronic hepatitis, cirrhosis, and hepatocellular carcinoma.
  • liver diseases such as subclinical carrier state of acute hepatitis, chronic hepatitis, cirrhosis, and hepatocellular carcinoma.
  • the term includes symptoms and side effects of any HCV infection, including latent, persistent and sub-clinical infections, whether or not the infection is clinically apparent.
  • treatment is used herein to characterize a method or process that is aimed at (1) delaying or preventing the onset of a disease or condition (e.g., HCV infection or HCV-related disease); (2) slowing down or stopping the progression, aggravation, or deterioration of the symptoms of the disease or condition; (3) bringing about amelioration of the symptoms of the disease or condition; or (4) curing the disease or condition.
  • a treatment may be administered prior to the onset of the disease or condition, for a prophylactic or preventive action. Alternatively or additionally, a treatment may be administered after initiation of the disease or condition, for a therapeutic action.
  • a "pharmaceutical composition” is defined herein as comprising an effective amount of a combination of the invention, and at least one pharmaceutically acceptable carrier or excipient.
  • the term "effective amount” refers to any amount of a compound, agent, antibody, composition, or combination that is sufficient to fulfil its intended purpose(s), e.g., a desired biological or medicinal response in a cell, tissue, system or subject.
  • the purpose(s) may be: to prevent HCV infection, to prevent the onset of a HCV-related disease, to slow down, alleviate or stop the progression, aggravation or deterioration of the symptoms of a HCV-related disease (e.g., chronic hepatitis C, cirrhosis, and the like); to bring about amelioration of the symptoms of the disease, or to cure the HCV- related disease.
  • pharmaceutically acceptable carrier or excipient refers to a carrier medium which does not interfere with the effectiveness of the biological activity of the active ingredient(s) and which is not significantly toxic to the host at the concentration at which it is administered.
  • the term includes solvents, dispersion, media, coatings, antibacterial and antifungal agents, isotonic agents, and adsorption delaying agents, and the like.
  • solvents, dispersion, media, coatings, antibacterial and antifungal agents, isotonic agents, and adsorption delaying agents, and the like are well known in the art (see for example "Remington 's Pharmaceutical Sciences” ' , E.W. Martin, 18 th Ed., 1990, Mack Publishing Co.: Easton, PA, which is incorporated herein by reference in its entirety).
  • Susceptible cell and “HCV-susceptible cell” are used interchangeably. They refer to any cell that may be infected with HCV.
  • Susceptible cells include, but are not limited to, liver or hepatic cells, primary cells, hepatoma cells, CaCo2 cells, dendritic cells, placental cells, endometrial cells, lymph node cells, lymphoid cells (B and T cells), peripheral blood mononuclear cells, and monocytes/macrophages.
  • preventing, inhibiting or blocking HCV infection when used in reference to an inventive combination means reducing the amount of HCV genetic information introduced into a susceptible cell or susceptible cell population as compared to the amount of HCV genetic information that would be introduced in the absence of the combination.
  • isolated?' as used herein in reference to a protein or polypeptide, means a protein or polypeptide, which by virtue of its origin or manipulation is separated from at least some of the components with which it is naturally associated or with which it is associated when initially obtained.
  • isolated it is alternatively or additionally meant that the protein or polypeptide of interest is produced or synthesized by the hand of man.
  • protein protein
  • polypeptide amino acid sequences of a variety of lengths, either in their neutral (uncharged) forms or as salts, and either unmodified or modified by glycosylation, side-chain oxidation, or phosphorylation.
  • the amino acid sequence is a full-length native protein. In other embodiments, the amino acid sequence is a smaller fragment of the full-length protein.
  • the amino acid sequence is modified by additional substituents attached to the amino acid side chains, such as glycosyl units, lipids, or inorganic ions such as phosphates, as well as modifications relating to chemical conversions of the chains such as oxidation of sulfydryl groups.
  • the term “protein” (or its equivalent terms) is intended to include the amino acid sequence of the full-length native protein, or a fragment thereof, subject to those modifications that do not significantly change its specific properties.
  • the term “protein” encompasses protein isoforms, i.e., variants that are encoded by the same gene, but that differ in their pi or MW, or both.
  • Such isoforms can differ in their amino acid sequence ⁇ e.g., as a result of allelic variation, alternative splicing or limited proteolysis), or in the alternative, may arise from differential post-translational modification ⁇ e.g., glycosylation, acylation, phosphorylation) .
  • analog refers to a polypeptide that possesses a similar or identical function as the protein but need not necessarily comprise an amino acid sequence that is similar or identical to the amino acid sequence of the protein or a structure that is similar or identical to that of the protein.
  • a protein analog has an amino acid sequence that is at least 30%, more preferably, at least 35%, 40%>, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95% or 99% identical to the amino acid sequence of the protein.
  • fragment refers to a polypeptide comprising an amino acid sequence of at least 5 consecutive amino acid residues (preferably, at least about: 10, 15, 20, 25, 30, 35, 40, 50, 60, 70, 80, 90, 100, 125, 150, 175, 200, 250 or more amino acid residues) of the amino acid sequence of a protein.
  • the fragment of a protein may or may not possess a functional activity of the protein.
  • biologically active refers to a molecule that shares sufficient amino acid sequence identity or homology with the protein to exhibit similar or identical properties to the protein.
  • a biologically active fragment of an inventive antibody is a fragment that retains the ability of the antibody to bind to a HCV receptor.
  • homologous or “homology”
  • identity refers to the sequence similarity between two polypeptide molecules or between two nucleic acid molecules.
  • the percentage of homology between two sequences corresponds to the number of matching or homologous positions shared by the two sequences divided by the number of positions compared and multiplied by 100. Generally, a comparison is made when two sequences are aligned to give maximum homology.
  • Homologous amino acid sequences share identical or similar amino acid sequences. Similar residues are conservative substitutions for, or "allowed point mutations" of, corresponding amino acid residues in a reference sequence.
  • Consservative substitutions" of a residue in a reference sequence are substitutions that are physically or functionally similar to the corresponding reference residue, e.g. that have a similar size, shape, electric charge, chemical properties, including the ability to form covalent or hydrogen bonds, or the like.
  • the present invention provides combinations and methods for the treatment and prevention of HCV infection.
  • a combination according to the invention comprises at least one protein kinase inhibitor and at least one interferon, or at least one protein kinase inhibitor and at least one direct acting antiviral, and is intended for use in the treatment or the prevention of HCV infection.
  • the combinations of the present invention comprise at least one protein kinase inhibitor.
  • the term 'protein kinase inhibitor refers to any molecule that specifically blocks the action of one or more protein kinases. Protein kinase inhibitors are subdivided by the amino acids whose phosphorylation is inhibited. Most kinases act on both serine and threonine, the tyrosine kinases act on tyrosine, and a number (dual- specificity) kinases act on all three.
  • the term “serine/threonine kinase inhibitor” refers to a molecule that specifically blocks the action of one or more serine and/or threonine kinases.
  • the term “tyrosine kinase inhibitor” refers to a molecule that specifically blocks the action of one or more tyrosine kinases.
  • the present Applicants have shown that interferons and direct acting antivirals act in synergy with erlotinib or dasatinib to inhibit HCV infection. Both erlotinib and dasatinib are tyrosine kinase inhibitors. Therefore, in certain preferred embodiments, the at least one protein kinase inhibitor present in a combination according to the invention is a tyrosine kinase inhibitor.
  • Tyrosine kinase inhibitors are generally used in cancer therapy. Indeed, research indicates that mutations which make tyrosine kinases constantly active can be a contributing factor in the development of cancerous cells. So, when a tyrosine kinase inhibitor is administered, the cell communication and reproduction is reduced, and cancerous cell growth can be lowered to the point of stopping growth.
  • HCV entry cofactors EGFR (epidermal growth factor receptor) and EphA2 (ephrin type-A receptor A)
  • EGFR epidermal growth factor receptor
  • EphA2 ephrin type-A receptor A
  • erlotinib and dasatinib broadly impaired infection by all major HCV genotypes and viral escape variants in vitro and in the human liver-chimeric Alb- uPA/SCID mouse model (Lupberger et al, Nature Medicine, 2011, 17: 589-595). They showed that erlotinib and dasatinib interfere with CD81-CLDN1 co-receptor interactions and with glycoprotein-dependant viral fusion.
  • the at least one protein kinase inhibitor present in a combination of the invention is a tyrosine kinase inhibitor that acts on the epidermal growth factor receptor (EGFR).
  • EGFR epidermal growth factor receptor
  • the target protein EGFR is also sometimes referred to as Herl or ErbB-1.
  • tyrosine kinase inhibitors that act on EGFR include, but are not limited to, erlotinib, gefitinib, vandetanib, and lapatinib.
  • the at least one tyrosine kinase inhibitor that acts on EGFR is erlotinib.
  • Erlotinib is marketed under the tradename TARCEVA ® by Genentech and OSI pharmaceuticals in the United States and by Roche elsewhere. Erlotinib binds in a reversible fashion to the adenosine triphosphate (ATP) binding site of the epidermal growth factor receptor. For the signal to be transmitted, two members of the EGFR family need to come together to form a homodimer.
  • the at least one tyrosine kinase inhibitor that acts on EGFR is gefitinib.
  • Gefitinib (tradename IRES S A ® ) is marketed by AstraZeneca and Teva. In Europe, gefitinib is indicated in advanced non-small cell lung cancer in all lines of treatment for patients harboring EGFR mutations.
  • the at least one tyrosine kinase inhibitor that acts on EGFR is vandetanib.
  • Vandetanib also known as ZD6474 is being developed by AstraZeneca. It is an antagonist of the epidermal growth factor receptor (EGFR) and the vascular endothelial growth factor receptor (VEGFR).
  • EGFR epidermal growth factor receptor
  • VEGFR vascular endothelial growth factor receptor
  • Vandetanib became the first drug to be approved by the FDA for the treatment of late-stage (metastatic) medullatory thyroid cancer in adult patients who are ineligible for surgery.
  • the at least one tyrosine kinase inhibitor that acts on EGFR is lapatinib.
  • Lapatinib in the form of lapatinib ditosylate (tradenames TYKERB ® in the U.S. and TYVERB ® in Europe) is marketed by Glaxo SmithKline.
  • Lapatinib is a dual tyrosine kinase inhibitor, which inhibits the tyrosine kinase activity associated with EGFR and HER2/neu (Human EGFR type 2). In February 2010, lapatinib received accelerated approval as front-line therapy in triple positive breast cancer.
  • tyrosine kinase inhibitors that act on EGFR and that are suitable for use in the present invention include molecules that are currently under development for human use, including, but not limited to, neratinib (also known as HKI-272, being developed by Pfizer), which is under investigation for the treatment of breast cancer and other solid tumors; and afatinib (also known as BIBW 2992, being developed by Boehringer Ingelheim), which is a candidate drug against non-small cell lung carcinoma, both of which are dual inhibitors of EGFR and Her2.
  • neratinib also known as HKI-272, being developed by Pfizer
  • afatinib also known as BIBW 2992, being developed by Boehringer Ingelheim
  • tyrosine kinase inhibitors that act on EGFR include anti- EGFR antibodies, such as Cetuximab, Panitumumab, Matuzumab, Zalutumumab, Nimotuzumab, Necitumumab, and the like.
  • the at least one protein kinase inhibitor present in a combination of the invention is a tyrosine kinase inhibitor that acts on the ephrin type- A receptor 2 (EphA2).
  • EphA2 ephrin type- A receptor 2
  • Examples of such tyrosine kinase inhibitors include, for example, dasatinib.
  • Dasatinib (BMS-354825) is sold under the tradename SPRYCEL ® by Bristol- Myers Squibb. Dasatinib is a multi-targeted kinase inhibitor mainly developed for Bcr-Abl and Src family kinases, but which also inhibits multiple Eph kinases, including EphA2. Dasatinib is approved for use in patients with chronic myelogenous leukemia (CML) after imatinib treatment, and Philadelphia chromosome-positive acute lymphoblastic leukemia. It is being evaluated for use in numerous other cancers, including advanced prostate cancer. Other examples of tyrosine kinase inhibitors that act on EphA2 include anti- EphA2 antibodies, such as those developed by Medlmmune Inc.
  • the at least one protein kinase inhibitor present in a combination of the present invention is an antibody against a receptor tyrosine kinase (RTK) other than EGFR and EphA2.
  • RTK receptor tyrosine kinase
  • the receptor tyrosine kinases may belong to the insulin receptor family, PDGF receptor family, VEGF receptor family, HGF receptor family, Trk receptor family, AXL receptor family, LTK receptor family, TIE receptor family, ROR receptor family, DDR receptor family, RET receptor family, KLG receptor family, RYK receptor family, or MuSK receptor family.
  • anti-RTK monoclonal antibodies include, but are not limited to, anti-VEGF antibodies such as Bevacizumab and Ranibizumab; anti-Erb2 antibodies such as Trastuzumab; anti-HER2/neu antibodies such as Trastuzumab, Ertimaxomab, and Pertuzumab; anti-VEGFR2 antibodies such as Ramucirumab and Alacizumab pegol; anti-VEGF-A antibodies such as Ranibizumab and Bevacizumab; anti-PDGF-R antibodies such as Olaratumab; and anti-IGF-1 receptor antibodies such as Figitumumab; Robatumumal and Cixutumumab.
  • anti-VEGF antibodies such as Bevacizumab and Ranibizumab
  • anti-Erb2 antibodies such as Trastuzumab
  • anti-HER2/neu antibodies such as Trastuzumab, Ertimaxomab, and Pertuzumab
  • interferon refers to any interferon or interferon derivative ⁇ e.g., pegylated interferon) that can be used in the prevention or treatment of HCV infection and/or in the prevention or treatment of HCV-related diseases, in particular cirrhosis and liver cancer.
  • Interferons are a family of cytokines produced by eukaryotic cells in response to viral infection and other antigenic stimuli, which display broad-spectrum antiviral, antiproliferative and immunomodulatory effects.
  • Recombinant forms of interferons have been widely applied in the treatment of various conditions and diseases, such as viral infections ⁇ e.g., HCV, HBV and HIV), inflammatory disorders and diseases (e.g., multiple sclerosis, arthritis, cystic fibrosis), and tumors ⁇ e.g., liver cancer, lymphomas, myelomas, etc).
  • Interferons are classified as Type I, Type II and Type III, depending on the cell receptor to which they bind.
  • Type I interferons bind to a specific cell surface receptor complex known as the IFN-a receptor (IFNAR) that consists of two chains (IFNAR1 and IFNAR2).
  • IFNAR IFN-a receptor
  • the type I interferons present in humans are interferon-alpha (IFN-a), interferon-beta (IFN- ⁇ ) and interferon-omega (IFN-CO).
  • IFN-a interferon-alpha
  • IFN- ⁇ interferon-beta
  • IFN-CO interferon-omega
  • Treatments based on the use of IFN-a or pegylated IFN-a remain the cornerstone of therapy for chronic HCV infection.
  • a new form of IFN-a with an extended in vivo half- life, albumin- interferon or albinterferon a recombinant formulation of IFN-a genetically fused to the human blood protein albumin
  • Albumin-interferon has been shown to exhibit high antiviral activity and to offer safety/tolerability comparable to the current standard of care and fared well in phase III clinical trials in patients with chronic HCV infection (Zeuzem et al, Gastroenterology, 2010, 139: 1257-1266; Nelson et al, Gastroenterology, 2010, 139: 1267-1276).
  • Interferon- ⁇ has also been demonstrated to display antiviral activity against HCV and to be useful in the treatment of HCV infection, alone or in combination with ribavirin (Fukutomi et al, J. Hepatology, 2001, 34: 100-107; Sang Hoon Ahn et al, Gut and Liver, 2009, 3: 20-25).
  • Pegylated IFN- ⁇ is currently undergoing clinical testing in Japan for HCV patients who do not respond well to the conventional combination therapy of ribavirin and IFN-a (Toray Industries Inc., Press Release, Feb. 2009). Interferon-co also exhibits antiviral properties (Buckwold et al, Antiviral Res., 2007, 73: 118-125) and has been tested for anti-HCV effect using an implantable infusion pump for the continuous delivery and consistent dose of drug for 3 to 12 months (Rohloff et al, J. Diabetes Sci. Technol, 2008, 2: 461-467).
  • Type II interferons bind to the interferon-gamma receptor (IFNGR).
  • IFNGR interferon-gamma receptor
  • IFN- ⁇ interferon-gamma
  • IFN- ⁇ does have anti- viral and antitumor effects, however these effects are weaker when compared to INF-a.
  • Type III interferons signal through a receptor complex consisting of the interferon-lambda receptor (IFNLR1 or CRF2-12) and the interleukin 10 receptor 2 (IL10R2 or CRF2-4).
  • type III interferons include three interferon lambda (IFN- ⁇ ) proteins referred to as IFN- ⁇ , ⁇ - ⁇ 2 and ⁇ - ⁇ 3 also known as interleukin 29 (IL29), interleukin 28A (IL28A) and interleukin 28B (IL28B), respectively.
  • IFN- ⁇ interferon lambda
  • IL28A interleukin 28A
  • IL28B interleukin 28B
  • the at least one interferon molecule present in a combination according to the invention is selected from the group consisting of IFN-cc, IFN- ⁇ , IFN-co, IFN- ⁇ , IFN- ⁇ , analogs thereof and derivatives thereof.
  • the interferon present in the combination is selected from the group consisting of IFN-cc, analogs thereof and derivatives thereof.
  • the interferon present in the combination is selected from the group consisting of IFN-CO, analogs thereof and derivatives thereof.
  • the interferon present in the combination is selected from the group consisting of IFN- ⁇ , analogs thereof and derivatives thereof.
  • interferon As used herein, the terms “interferon”, “IFN and “IFN molecule” more specifically refer to a peptide or protein having an amino acid substantially identical (e.g., et least 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or even 100% identical) to all or a portion of the sequence of an interferon (e.g., a human interferon), such as IFN-CC, IFN- ⁇ , IFN-CO, IFN- ⁇ , and IFN- ⁇ that are known in the art.
  • an interferon e.g., a human interferon
  • Interferons suitable for use in the present invention include, but are not limited to, natural human interferons produced using human cells, recombinant human interferons produced from mammalian cells, E-co/z-produced recombinant human interferons, synthetic versions of human interferons and equivalents thereof.
  • Other suitable interferons include consensus interferons which are a type of synthetic interferons having an amino acid sequence that is a rough average of the sequence of all the known human IFN subtypes (for example, all the known IFN-CC subtypes, or all the known IFN- ⁇ subtypes, or all the known IFN-co subtypes, or all the known IFN- ⁇ subtypes, or all the known IFN- ⁇ subtypes).
  • interferons present in combinations according to the invention have been approved for human use. In other embodiments, interferons present in combinations according to the present are undergoing human clinical trials.
  • the terms "interferon”, “IFN”, and “IFN molecule” also include interferon derivatives, i.e., molecules of interferon (as described above) that have been modified or transformed.
  • a suitable transformation may be any modification that imparts a desirable property to the interferon molecule. Examples of desirable properties include, but are not limited to, prolongation of in vivo half-life, improvement of therapeutic efficacy, decrease of dosing frequency, increase of solubility/water solubility, increase of resistance against proteolysis, facilitation of controlled release, and the like.
  • pegylated interferons have been produced ⁇ e.g., pegylated IFN-a) and are currently used to treat hepatitis. Pegylated interferons exhibit longer half-lifes, which allows for less frequent administration of the drug. Pegylating an interferon molecule involves covalently binding the interferon to polyethylene glycol (PEG), an inert, non-toxic and biodegradable organic polymer. Therefore, in certain embodiments, the at least one interferon present in a combination according to the invention is a pegylated interferon. Interferons have also been produced as fusion proteins with human albumin ⁇ e.g., albumin-IFN-Cc).
  • the albumin- fusion platform takes advantage of the long half-life of human albumin to provide a treatment that allows the dosing frequency of IFN to be reduced in individuals with chronic hepatitis C. Therefore, in certain embodiments, the at least one interferon present in a combination according to the invention is an albumin- interferon fusion protein.
  • alpha interferon alpha interferon
  • interferon-alpha i.e., glycoproteins
  • IFN-a molecules suitable for use in the present invention include, but are not limited to, recombinant IFN-a-2b (such as INTRON-A ® interferon available from Schering Corporation); recombinant IFN-a-2a (such as ROFERON ® interferon available from Hoffman-La Roche); recombinant IFN-a-2C (such as BEROFOR ® alpha 2 interferon available from Boehringer Ingelheim Pharmaceutical, Inc); IFN-a-nl, a purified blend of natural alpha interferons (such as SUMERIFERON ® available from Sumitomo, Japan or WELLFERON ® interferon alpha-nl (INS) available from Glaxo-Wellcome Ltd); IFN-a-n3, a mixture of natural alpha interferons (such as ALFERON ® made by Interferon Sciences); human leukocyte interferon-a obtained from the leukocyte fraction of human blood following induction with Sendai virus (such as MULTI
  • interferon alpha molecules include IFN-CC derivatives, including, but not limited to, pegylated IFN-CC-2a (such as PEGASYS ® available from Hoffman- La Roche); pegylated IFN-CC-2b (such as PEGINTRON ® available from Schering Corporation); albumin IFN-CC-2b also known as albinterferon (such as ALBUFERON ® available from Human Genome Sciences), and equivalents thereof.
  • pegylated IFN-CC-2a such as PEGASYS ® available from Hoffman- La Roche
  • pegylated IFN-CC-2b such as PEGINTRON ® available from Schering Corporation
  • albumin IFN-CC-2b also known as albinterferon (such as ALBUFERON ® available from Human Genome Sciences), and equivalents thereof.
  • beta interferon refers to the family of highly homologous species-specific proteins (i.e., glycoproteins) that are known in the art and have the ability to induce resistance to viral antigens.
  • IFN- ⁇ molecules suitable for use in the present invention include, but are not limited to, recombinant ⁇ - ⁇ -la (such as, REBIF ® available from Pfizer or AVONEX ® available from Biogen Idex), recombinant IFN-p-lb (such as BETAFERON ® /BETASERON ® available from Bayer HealthCare or EXTAVIA ® , the generic form of BETAFERON, available from Novartis, or ZIFERON ® , an interferon- ⁇ lb biosimilar, available from Zistdaru Danesh Ltd), IFN- ⁇ molecules described in U.S. Pat. Nos. 4,820,638 and 5,795,779) and, equivalents thereof.
  • recombinant ⁇ - ⁇ -la such as, REBIF ® available from Pfizer or AVONEX ® available from Biogen Idex
  • IFN-p-lb such as BETAFERON ® /BETASERON ® available
  • interferon beta molecules include IFN- ⁇ derivatives, including, but not limited to, pegylated INF- ⁇ (such as TRK-560 being developed by Toray Industries, Inc.), pegylated ⁇ - ⁇ -la (such as BUBO 17 being developed by Biogen pie); pegylated ⁇ - ⁇ -lb (such as NUlOO and NU400 being developed by Nuron Biotech); albumin- IFN- ⁇ fusion proteins such as those described in U.S. Pat. No. 7,572,437, and equivalents thereof.
  • pegylated INF- ⁇ such as TRK-560 being developed by Toray Industries, Inc.
  • pegylated ⁇ - ⁇ -la such as BUBO 17 being developed by Biogen pout
  • pegylated ⁇ - ⁇ -lb such as NUlOO and NU400 being developed by Nuron Biotech
  • albumin- IFN- ⁇ fusion proteins such as those described in U.S. Pat. No. 7,572,437, and equivalents thereof.
  • interferon interferon-omega
  • interferon- ⁇ interferon- ⁇
  • IFN- co immunodeficiency-cose cleavage-associated kinase
  • Typical IFN-CO molecules suitable for use in the present invention include, but are not limited to, IFN-CO described in European patent No. EPO 170 204, ITCA being developed by Intarcia Therapeutics, Inc., and equivalents thereof.
  • interferon omega molecules include IFN-CO derivatives, including, but not limited to, pegylated INF-CO that can be obtained using a method described in U.S. Pat. Nos. 5,612,460; 5,711,944; 5,951,974 or 5,951,974; albumin-IFN-CO fusion proteins such as those described in U.S. Pat. No. 7,572,437, and equivalents thereof.
  • gamma interferon gamma interferon
  • interferon-gamma gamma interferon
  • interferon- 1 ⁇ gamma interferon
  • IFN- ⁇ Typical IFN- ⁇ molecules suitable for use in the present invention include, but are not limited to, IFN- ⁇ described in U.S. Pat. Nos.
  • interferon omega molecules include IFN- ⁇ derivatives, including, but not limited to, pegylated INF- ⁇ that can be obtained using a method described in U.S. Pat. Nos. 5,612,460; 5,711,944; 5,951,974 or 5,951,974 albumin-IFN-CO fusion proteins such as those described in U.S. Pat. No. 7,572,437, and equivalents thereof.
  • interferon interferon-lambda
  • interferon- ⁇ interferon- ⁇
  • IFN- ⁇ immunoglobulin- ⁇ molecules
  • Typical IFN- ⁇ molecules suitable for use in the present invention include, but are not limited to, IFN- ⁇ , ⁇ - ⁇ 2 and ⁇ - ⁇ 3 molecules described in international patent applications number WO02/086087, WO2004/037995 and WO/2005/023862 and equivalents thereof.
  • interferon omega molecules include IFN- ⁇ derivatives including, but not limited to, pegylated ⁇ - ⁇ -la (such as BMS-914143 being developed by Bristol-Myers Squibb), albumin- IFN-CO fusion proteins such as those described in U.S. Pat. No. 7,572,437, and equivalents thereof.
  • the terms "interferon” , "/FN”, and “/FN molecule” also include interferon- like molecules, i.e., molecules that have functional and/or structural features exhibited by or similar to known interferons or interferon analogs, such as those described above.
  • Certain combinations of the present invention comprise at least one protein kinase inhibitor and at least one direct acting antiviral.
  • direct acting antiviral refers to molecules that interfere with specific steps of the lifecycle of HCV and are thus useful in the prevention or treatment of HCV infection.
  • a direct acting antiviral agent suitable for use in a combination of the present invention may exert its effects by any mechanism that interferes with one or more specific steps of the lifecycle of HCV.
  • telaprevir, boceprevir, danoprevir and TMC-435 used by the present Applicants are protease inhibitors
  • daclatasvir is a NS5A inhibitor
  • mericitabine and GS7977 are polymerase inhibitors.
  • the at least one direct acting antiviral present in a combination according to the invention is a HCV protease inhibitor or a HCV polymerase inhibitor or an NS5A inhibitor.
  • Protease inhibitors suitable for use in the context of the present invention include NS3/4A protease inhibitors.
  • NS3/4A protease inhibitors that can be present in a combination of the present invention include, but are not limited to, VX- 950 (also known as telaprevir), ITMN-191 (also known as danoprevir), boceprevir, BMS-650032, VX-985, BI 201335, and TMC-435.
  • the at least one direct acting antiviral present in a combination according to the invention is NS3/4A protease inhibitor telaprevir, boceprevir, danoprevir or TMC- 435.
  • Telaprevir also known as VX-950
  • INCIVEK ® a registered trademark of Lucent Technologies Inc.
  • ITMN-191 also known as R7227 or danoprevir
  • Boceprevir initially developed by Schering-Plough, and then by Merck and marketed under the tradename VICTRELIS ®
  • BMS-650032 is being developed by Bristol- Myers-Squibb.
  • VX-985 is a NS3/4A protease inhibitor being developed by Vertex Pharmaceuticals, Inc. BI 201335 is being developed by Boehringer Ingelheim and is now in Phase III clinical trials in the United States. TMC-435, a NS3/4A protease inhibitor being developed by Medivir/Tibotec/ Johnson & Johnson, is also in Phase III clinical trials.
  • NS3/4A protease inhibitors that can be present in a combination according to the invention include, but are not limited to, NS3/4A protease inhibitors that are currently in phase II clinical trials such as GS 9256 and GS 9451 (being developed by Gilead), MK-7009 (also known as vaniprevir, being developed by Merck), ACH-1625 (being developed by Achillion), and ABT-450 (being developed by Abbott/Enanta); NS3/4A protease inhibitors that are currently in phase I clinical trials such as BMS-791325 (being developed by Bristol-Myers Squibb), VX-985 and VX-500 (being developed by Vertex pharmaceuticals), and PHX1766 (being developed by Phenomix); and NS3/4A protease inhibitors that are currently in preclinical trials such as VX-813 (being developed by Vertex), AVL-181 and AVL-192 (being developed by Avila Therapeutics), and ACH-2684 (being developed
  • Polymerase inhibitors suitable for use in the context of the present invention include NS5B polymerase inhibitors.
  • the NS5B an R A-dependent R A polymerase (RdRp) enzyme, is a highly conserved structure across all hepatitis C genotypes. It is therefore, an ideal target for drug therapy.
  • RdRp R A-dependent R A polymerase
  • Nucleoside inhibitors target the catalytic sites of the enzyme and act as chain terminators.
  • Non-nucleoside inhibitors are allosteric inhibitors.
  • the at least one direct acting antiviral present in a combination according to the invention is the HCV NS5B polymerase inhibitor, mericitabine or GS-7977.
  • Mericitabine (also known as RG7128 or RO5024048), is a prodrug of PSI-6130, an oral cytidine nucleoside analogue. It is being developed by Roche and Pharmasset. Mericitabine has shown in vitro activity against all of the most common HCV genotypes.
  • GS-7977 (also known as PSI-7977) is being developed by Gilead Sciences. It is currently in Phase III clinical trials. It is being studied as a treatment to be used in combination with ribavirin.
  • GS-78977 is a prodrug that is metabolized to the active antiviral agent 2'-deoxy-2'-a-fluoro-P-C-methyluridine-5 '-monophosphate.
  • NS5B polymerase inhibitors that can be present in a combination according to the invention include, but are not limited to, nucleoside/nucleotide polymerase inhibitors that are currently in Phase II clinical trials such as IDX184 (being developed by Idenix); non-nucleoside polymerase inhibitors that are currently in Phase II clinical trials such as VX-222 (initially developed by ViroChem, now owned by Vertex); PF-868554 (being developed by Pfizer); ABT-072 and ABT-333 (being developed by Abbott), GS 9190 (being developed by Gilead) and ANA598 (also known as setrobuvir, being developed by Anadys); nucleoside/nucleotide polymerase inhibitors that are currently in Phase I clinical trials such as BI 207127 (being developed by Boehringer Ingelheim), MK- 0608 (being developed by Isis/Merck), TMC649128 (being developed by Medivir/Tibotec),
  • NS5A inhibitors suitable for use in the context of the present invention include, in particular daclatasvir (also known as BMS-790052), which is developed by Bristol- Myers- Squibb.
  • a combination according to the present invention is such that (1) it is intended for use in the treatment or the prevention of HCV infection and (2) the at least one protein kinase inhibitor and at least one interferon or the least one protein kinase inhibitor and at least one direct acting antiviral act in a highly synergistic manner on the inhibition of HCV infection.
  • the interferon or the direct acting antiviral decreases the interferon or the direct acting antiviral
  • IC 50 for the inhibition of HCV infection by the protein kinase inhibitor by a factor of at least 10 fold or at least 25 fold, preferably at least 50 fold or at least 75 fold, more preferably at least 100 fold or 200 fold, and even more preferably more than 200 fold ⁇ e.g., more than 350 fold).
  • the concentration of protein kinase inhibitor necessary to obtain a 50% inhibition of HCV entry is at least 10 times or at least 25 times, preferably at least 50 times or at least 75 times, more preferably at least 100 times or 200 times, and even more preferably more than 200 times ⁇ e.g., more than 350 times) lower than the concentration of protein kinase inhibitor that would be necessary to obtain the same HCV entry inhibition in the absence of interferon or direct acting antiviral.
  • the protein kinase inhibitor decreases the IC 50 for the inhibition of HCV infection by the interferon or the direct acting antiviral by a factor of at least 10 fold or at least 25 fold, preferably at least 50 fold or at least 75 fold, more preferably at least 100 fold or 200 fold, and even more preferably more than 200 fold ⁇ e.g., more than 350 fold).
  • the concentration of interferon or direct acting antiviral necessary to obtain a 50% inhibition of HCV entry is at least 10 times or at least 25 times, preferably at least 50 times or at least 75 times, more preferably at least 100 times or 200 times, and even more preferably more than 200 times (e.g., more than 350 times) lower than the concentration of interferon or direct acting antiviral that would be necessary to obtain the same HCV entry inhibition in the absence of protein kinase inhibitor.
  • a combination of the present invention is characterized by a combination index (CI) that is lower than 1 (which is defined as a marked synergy).
  • a combination of the present invention is preferably characterized by a CI lower than 1, preferably lower than 0.90, more preferably by a CI lower than 0.85, and even more preferably by a CI lower than 0.50.
  • a combination of the present invention is characterized by a surface amplitude > 20% as determined by the method of Prichard and Shipman (Prichard et al, Antiviral Res., 1990, 14: 181-205).
  • the combinations according of the present invention may be used in therapeutic and prophylactic methods to treat and/or prevent HCV infection, or to treat and/or prevent a liver disease or a pathological condition affecting HCV-susceptible cells, such as liver cells, lymphoid cells, or monocytes/macrophages.
  • Methods of treatment of the present invention may be accomplished using an inventive combination or a pharmaceutical composition comprising an inventive combination (see below). These methods generally comprise administration of an effective amount of at least one interferon (or at least one direct acting antiviral) and at least one protein kinase inhibitor, or a pharmaceutical composition thereof, to a subject in need thereof.
  • the interferon (or direct acting antiviral) and protein kinase inhibitor may be administered concurrently (i.e., together or separately but at about the same time, e.g., within 5 minutes, 15 minutes or 30 minutes of each other), or alternatively, they may be administered sequentially (i.e., separately and at different times, e.g., different times of the same day or different times of the same week or different times of the same month, etc). Administration may be performed using any of the methods known to one skilled in the art.
  • the combination of protein kinase inhibitor and interferon or direct acting antiviral, or a composition thereof may be administered by various routes including, but not limited to, aerosol, parenteral, oral or topical route.
  • the combination, or pharmaceutical composition thereof will be administered in an effective amount, i.e. an amount that is sufficient to fulfill its intended purpose.
  • the exact amount of the combination or pharmaceutical composition to be administered will vary from subject to subject, depending on the age, sex, weight and general health condition of the subject to be treated, the desired biological or medical response (e.g., prevention of HCV infection or treatment of HCV-associated liver disease), and the like.
  • an effective amount is one that inhibits or prevents HCV from entering into a subject's susceptible cells and/or infecting a subject's cells, so as to prevent HCV infection, treat or prevent liver disease or another HCV-associated pathology in the subject.
  • the present invention provides a method for treating or preventing a liver disease or pathology in a subject, which comprises administering to the subject an effective amount of at least one protein kinase inhibitor and at least one interferon or at least one protein kinase inhibitor and at least one direct acting antiviral (as defined above) (or pharmaceutical composition thereof) which inhibits HCV from entering or infecting the subject's cells, so as to treat or prevent the liver disease or pathology in the subject.
  • the liver disease or pathology may be inflammation of the liver, liver fibrosis, cirrhosis, and/or hepatocellular carcinoma (i.e., liver cancer) associated with HCV infection.
  • the present invention also provides a method for treating or preventing a HCV- associated disease or condition (including a liver disease) in a subject, which comprises administering to the subject an effective amount of at least one interferon and at least one protein kinase inhibitor or at least one protein kinase inhibitor and at least one direct acting antiviral (as defined above) (or pharmaceutical composition thereof) which inhibits HCV from entering or infecting the subject's cells, so as to treat or prevent the HCV-associated disease or condition in the subject.
  • the combination (or pharmaceutical composition thereof) is administered to a subject diagnosed with acute hepatitis C.
  • the combination (or pharmaceutical composition thereof) is administered to a subject diagnosed with chronic hepatitis C.
  • Administration of an inventive combination, or pharmaceutical composition, according to such methods may result in amelioration of at least one of the symptoms experienced by the individual including, but not limited to, symptoms of acute hepatitis C such as decreased appetite, fatigue, abdominal pain, jaundice, itching, and flu-like symptoms; symptoms of chronic hepatitis C such as fatigue, marked weight loss, flu-like symptoms, muscle pain, joint pain, intermittent low-grade fevers, itching, sleep disturbances, abdominal pain, appetite changes, nausea, diarrhea, dyspepsia, cognitive changes, depression, headaches, and mood swings; symptoms of cirrhosis such as ascites, bruising and bleeding tendency, bone pain, varices (especially in the stomach and esophagus), steatorrhea, jaundice and hepatic encephalopathy; and symptoms of extrahepatic manifestations associated with HCV such as thyroiditis, porphyria cutanea tarda, cryoglobulinemia, glomerulonephritis, sicca syndrome, thrombocyto
  • administration of a combination or pharmaceutical composition thereof according to such methods may slow down, reduce, stop or alleviate the progression of HCV infection or an HCV-associated disease, or reverse the progression to the point of eliminating the infection or disease.
  • Administration of a combination or pharmaceutical composition of the present invention according to such methods may also result in reduction in the number of viral infections, reduction in the number of infectious viral particles, and/or reduction in the number of virally infected cells.
  • the effects of a treatment according to the invention may be monitored using any of the assays known in the art for the diagnosis of HCV infection and/or liver disease.
  • Such assays include, but are not limited to, serological blood tests, liver function tests to measure one or more of albumin, alanine transaminase (ALT), alkaline phosphatase (ALP), aspartate transaminase (AST), and gamma glutamyl transpeptidase (GGT), and molecular nucleic acid tests using different techniques such as polymerase chain reaction (PCR), transcription mediated amplification (TMA), or branched DNA (bDNA). Combinations and compositions of the present invention may also be used in immunization therapies. Accordingly, the present invention provides a method of reducing the likelihood of susceptible cells of becoming infected with HCV as a result of contact with HCV.
  • ALT alanine transaminase
  • ALP alkaline phosphatase
  • AST aspartate transaminase
  • GTT gamma glutamyl transpeptidase
  • PCR polymerase chain reaction
  • TMA transcription mediated
  • the method comprises contacting the susceptible cells with an effective amount of at least one interferon and at least one protein kinase inhibitor or at least one protein kinase inhibitor and at least one direct acting antiviral (as defined above) or a pharmaceutical composition thereof which inhibits HCV from entering or infecting the susceptible cells, so as to reduce the likelihood of the cells to become infected with HCV as a result of contact with HCV.
  • the present invention also provides a method of reducing the likelihood of a subject's susceptible cells of becoming infected with HCV as a result of contact with HCV. In this method, contacting the susceptible cells with the combination or composition may be performed by administrating a combination or a pharmaceutical composition thereof to the subject.
  • Reducing the likelihood of susceptible cells or of a subject of becoming infected with HCV means decreasing the probability of susceptible cells or a subject to become infected with HCV as a result of contact with HCV.
  • the decrease may be of any significant amount, e.g., at least a 2-fold decrease, more than a 2-fold decrease, at least a 10-fold decrease, more than a 10-fold decrease, at least a 100-fold decrease, or more than a 100-fold decrease.
  • the subject is infected with HCV prior to administration of the inventive composition. In other embodiments, the subject is not infected with HCV prior to administration of the inventive composition. In yet other embodiments, the subject is not infected with, but has been exposed to, HCV. In certain embodiments, the subject may be infected with HIV or HBV.
  • the methods of the present invention may be used to reduce the likelihood of a subject's susceptible cells of becoming infected with HCV as a result of liver transplant.
  • a diseased liver is removed from a HCV-infected patient, serum viral levels plummet.
  • virus levels rebound and can surpass pre-transplant levels within a few days (Powers et al., Liver TranspL, 2006, 12: 207-216).
  • Liver transplant patients may benefit from administration of a combination, or pharmaceutical composition, according the invention. Administration may be performed prior to liver transplant, during liver transplant, and/or following liver transplant.
  • Other subjects that may benefit from administration of a combination of interferon and protein kinase inhibitor according to the present invention include, but are not limited to, babies born to HCV-infected mothers, in particular if the mother is also HIV-positive; health-care workers who have been in contact with HCV- contaminated blood or blood contaminated medical instruments; drug users who have been exposed to HCV by sharing equipments for injecting or otherwise administering drugs; and people who have been exposed to HCV through tattooing, ear/body piercing and acupuncture with poor infection control procedures.
  • Other subjects that may benefit from administration of a combination according to the invention include, but are not limited to, subjects that exhibit one or more factors that are known to increase the rate of HCV disease progression. Such factors include, in particular, age, gender (males generally exhibit more rapid disease progression than females), alcohol consumption, HIV co-infection (associated with a markedly increased rate of disease progression), and fatty liver.
  • Still other subjects that may benefit from administration of a combination according to the invention include patients with HCV infections that are resistant to the standard of care or to other combinations of antivirals - antiviral resistance being a major challenge in HCV prevention and treatment (Pawlotsky et al., Hepatology, 2011, 53: 1742-1751).
  • the HCV infection or HCV-related disease to be treated by a combination according to the invention is caused by a Hepatitis C virus that is resistant to a direct acting antiviral.
  • a direct acting antiviral molecules together with clinical studies showing that these drugs may lead to the selection of resistant viruses if administered alone or in combination therapy, has raised concerns that resistance may undermine DAA-based therapy (Pawlotsky, Hepatology, 2011, 53: 1742-1751; Schaefer et al, Gastroenterology, 2012, 142: 1340- 1350 el341).
  • a protein kinase inhibitor efficiently prevents dissemination of DAA-resistant HCV variants resulting in more rapid and sustained virus elimination.
  • a combination of an anti-HCV entry agent and of a direct acting antiviral prevents DAA resistance allowing suppression of viral infection below the detection limit in a sustained manner.
  • a combination of an interferon and a protein kinase inhibitor or of at least one protein kinase inhibitor and at least one direct acting antiviral or a pharmaceutical composition thereof is administered alone according to a method of treatment of the present invention.
  • a combination of an interferon and a protein kinase inhibitor or of at least one protein kinase inhibitor and at least one direct acting antiviral or a pharmaceutical composition thereof is administered in combination with at least one additional therapeutic agent.
  • the combination or pharmaceutical composition may be administered prior to administration of the therapeutic agent, concurrently with the therapeutic agent, and/or following administration of the therapeutic agent.
  • Therapeutic agents that may be administered in combination with an inventive combination or pharmaceutical composition may be selected among a large variety of biologically active compounds that are known to have a beneficial effect in the treatment or prevention of HCV infection, or a HCV-associated disease or condition.
  • Such agents include, in particular, antiviral agents including, but not limited to, ribavirin, anti-HCV (monoclonal or polyclonal) antibodies, R A polymerase inhibitors, protease inhibitors, IRES inhibitors, helicase inhibitors, antisense compounds, ribozymes, micro-RNA antagonists, cytokines, therapeutic vaccines, NS5A antagonists, polymerase inhibitors, and any combination thereof.
  • An inventive combination (optionally after formulation with one or more appropriate pharmaceutically acceptable carriers or excipients), in a desired dosage can be administered to a subject in need thereof by any suitable route.
  • Various delivery systems are known and can be used to administer combinations of the present invention, including tablets, capsules, injectable solutions, encapsulation in liposomes, microparticles, microcapsules, etc.
  • Methods of administration include, but are not limited to, dermal, intradermal, intramuscular, intraperitoneal, intralesional, intravenous, subcutaneous, intranasal, pulmonary, epidural, ocular, and oral routes.
  • An inventive combination or composition may be administered by any convenient or other appropriate route, for example, by infusion or bolus injection, by absorption through epithelial or mucocutaneous linings (e.g., oral, mucosa, rectal and intestinal mucosa, etc). Administration can be systemic or local. Parenteral administration may be preferentially directed to the patient's liver, such as by catheterization to hepatic arteries or into a bile duct.
  • the interferon (or direct acting antiviral) and protein kinase inhibitor are administered sequentially (i.e., at different times or separately but at substantially the same time)
  • the interferon (or direct acting antiviral) and protein kinase inhibitor may be administered by the same route (e.g., intravenously) or by different routes (e.g., orally and intravenously).
  • the combination and therapeutic agent may be administered by the same route or different routes.
  • an inventive combination (or a composition thereof) of the present invention will be in a dosage such that the amount delivered is effective for the intended purpose.
  • the route of administration, formulation and dosage administered will depend upon the therapeutic effect desired, the severity of the HCV- related condition to be treated if already present, the presence of any infection, the age, sex, weight, and general health condition of the patient as well as upon the potency, bioavailability, and in vivo half-life of the interferon and protein kinase inhibitor used, the use (or not) of concomitant therapies, and other clinical factors. These factors are readily determinable by the attending physician in the course of the therapy.
  • the dosage to be administered can be determined from studies using animal models (e.g., chimpanzee or mice). Adjusting the dose to achieve maximal efficacy based on these or other methods are well known in the art and are within the capabilities of trained physicians. As studies are conducted using the inventive combination of an interferon and a protein kinase inhibitor, further information will emerge regarding the appropriate dosage levels and duration of treatment.
  • a treatment according to the present invention may consist of a single dose or multiple doses.
  • administration of an inventive combination, or composition thereof may be constant for a certain period of time or periodic and at specific intervals, e.g., hourly, daily, weekly (or at some other multiple day interval), monthly, yearly (e.g., in a time release form).
  • the delivery may occur at multiple times during a given time period, e.g., two or more times per week; two or more times per month, and the like.
  • the delivery may be continuous delivery for a period of time, e.g., intravenous delivery.
  • the amount of combination administered will preferably be in the range of about 1 ng/kg to about 100 mg/kg body weight of the subject, for example, between about 100 ng/kg and about 50 mg/kg body weight of the subject; or between about 1 g/kg and about 10 mg/kg body weight of the subject, or between about 100 M-g/kg and about 1 mg/kg body weight of the subject.
  • compositions comprising an effective amount of at least one interferon and at least one protein kinase inhibitor, or at least one protein kinase inhibitor and at least one direct acting antiviral as described herein and at least one pharmaceutically acceptable carrier or excipient.
  • the composition further comprises one or more additional biologically active agents.
  • the combinations and pharmaceutical compositions thereof may be administered in any amount and using any route of administration effective for achieving the desired prophylactic and/or therapeutic effect.
  • the optimal pharmaceutical formulation can be varied depending upon the route of administration and desired dosage. Such formulations may influence the physical state, stability, rate of in vivo release, and rate of in vivo clearance of the administered active ingredient.
  • the pharmaceutical compositions of the present invention may be formulated in dosage unit form for ease of administration and uniformity of dosage.
  • unit dosage form refers to a physically discrete unit of an interferon or of a protein kinase inhibitor or of both an interferon and a protein kinase inhibitor for the patient to be treated.
  • unit dosage form also refers to a physically discrete unit of a direct acting antiviral or of a protein kinase inhibitor or of both a direct acting antiviral and a protein kinas inhibitor for the patient to be treated. It will be understood, however, that the total daily dosage of the compositions will be decided by the attending physician within the scope of sound medical judgement.
  • sterile injectable preparations for example, sterile injectable aqueous or oleaginous suspensions may be formulated according to the known art using suitable dispersing or wetting agents, and suspending agents.
  • the sterile injectable preparation may also be a sterile injectable solution, suspension or emulsion in a non-toxic parenterally acceptable diluent or solvent, for example, as a solution in 2,3-butanediol.
  • acceptable vehicles and solvents that may be employed are water, Ringer's solution, U.S.P. and isotonic sodium chloride solution.
  • sterile, fixed oils are conventionally employed as a solution or suspending medium.
  • any bland fixed oil can be employed including synthetic mono- or di-glycerides.
  • Fatty acids such as oleic acid may also be used in the preparation of injectable formulations.
  • Sterile liquid carriers are useful in sterile liquid form compositions for parenteral administration.
  • Injectable formulations can be sterilized, for example, by filtration through a bacterial-retaining filter, or by incorporating sterilizing agents in the form of sterile solid compositions which can be dissolved or dispersed in sterile water or other sterile injectable medium prior to use.
  • Liquid pharmaceutical compositions which are sterile solutions or suspensions can be administered by, for example, intravenous, intramuscular, intraperitoneal or subcutaneous injection. Injection may be via single push or by gradual infusion. Where necessary or desired, the composition may include a local anesthetic to ease pain at the site of injection.
  • Injectable depot forms are made by forming micro-encapsulated matrices of the active ingredient in biodegradable polymers such as polylactide-polyglycolide. Depending upon the ratio of active ingredient to polymer and the nature of the particular polymer employed, the rate of ingredient release can be controlled. Examples of other biodegradable polymers include poly(orthoesters) and poly( anhydrides). Depot injectable formulations can also be prepared by entrapping the active ingredient in liposomes or microemulsions which are compatible with body tissues.
  • Liquid dosage forms for oral administration include, but are not limited to, pharmaceutically acceptable emulsions, microemulsions, solutions, suspensions, syrups, elixirs, and pressurized compositions.
  • the liquid dosage form may contain inert diluents commonly used in the art such as, for example, water or other solvent, solubilising agents and emulsifiers such as ethyl alcohol, isopropyl alcohol, ethyl carbonate, ethyl acetate, benzyl alcohol, benzyl benzoate, propylene glycol, 1,3-butylene glycol, dimethylformamide, oils (in particular, cotton seed, ground nut, corn, germ, olive, castor, and sesame oils), glycerol, tetrahydrofurfuryl alcohol, polyethylene glycols, and fatty acid esters of sorbitan and mixtures thereof.
  • inert diluents commonly used in the art such as, for example,
  • the oral compositions can also include adjuvants such as wetting agents, suspending agents, preservatives, sweetening, flavouring, and perfuming agents, thickening agents, colors, viscosity regulators, stabilizes or osmo-regulators.
  • suitable liquid carriers for oral administration include water (potentially containing additives as above, e.g., cellulose derivatives, such as sodium carboxymethyl cellulose solution), alcohols (including monohydric alcohols and polyhydric alcohols such as glycols) and their derivatives, and oils ⁇ e.g., fractionated coconut oil and arachis oil).
  • the liquid carrier can be halogenated hydrocarbon or other pharmaceutically acceptable propellant.
  • Solid dosage forms for oral administration include, for example, capsules, tablets, pills, powders, and granules.
  • an inventive combination may be mixed with at least one inert, physiologically acceptable excipient or carrier such as sodium citrate or dicalcium phosphate and one or more of: (a) fillers or extenders such as starches, lactose, sucrose, glucose, mannital, and silicic acid; (b) binders such as, for example, carboxymethylcellulose, alginates, gelatine, polyvinylpyrrolidone, sucrose, and acacia; (c) humectants such as glycerol; (d) disintegrating agents such as agar-agar, calcium carbonate, potato or tapioca starch, alginic acid, certain silicates, and sodium carbonate; (e) solution retarding agents such as paraffin; absorption accelerators such as quaternary ammonium compounds; (g) wetting agents such as, for example, cetyl alcohol
  • excipients suitable for solid formulations include surface modifying agents such as non-ionic and anionic surface modifying agents.
  • surface modifying agents include, but are not limited to, poloxamer 188, benzalkonium chloride, calcium stearate, cetostearyl alcohol, cetomacrogol emulsifying wax, sorbitan esters, colloidal silicon dioxide, phosphates, sodium dodecylsulfate, magnesium aluminum silicate, and triethanolamine.
  • the dosage form may also comprise buffering agents.
  • Solid compositions of a similar type may also be employed as fillers in soft and hard-filled gelatine capsules using such excipients as lactose or milk sugar as well as high molecular weight polyethylene glycols and the like.
  • the solid dosage forms of tablets, dragees, capsules, pills, and granules can be prepared with coatings and shells such as enteric coatings, release controlling coatings and other coatings well known in the pharmaceutical formulating art. They may optionally contain opacifying agents and can also be of a composition such that they release the active ingredient(s) only, or preferably, in a certain part of the intestinal tract, optionally, in a delaying manner.
  • Examples of embedding compositions which can be used include polymeric substances and waxes.
  • an inventive composition may be desirable to administer an inventive composition locally to an area in need of treatment (e.g., the liver). This may be achieved, for example, and not by way of limitation, by local infusion during surgery (e.g., liver transplant), topical application, by injection, by means of a catheter, by means of suppository, or by means of a skin patch or stent or other implant.
  • the composition is preferably formulated as a gel, an ointment, a lotion, or a cream which can include carriers such as water, glycerol, alcohol, propylene glycol, fatty alcohols, triglycerides, fatty acid esters, or mineral oil.
  • carriers such as water, glycerol, alcohol, propylene glycol, fatty alcohols, triglycerides, fatty acid esters, or mineral oil.
  • Other topical carriers include liquid petroleum, isopropyl palmitate, polyethylene glycol, ethanol (95%), polyoxyethylenemonolaurat (5%) in water, or sodium lauryl sulphate (5%) in water.
  • Other materials such as antioxidants, humectants, viscosity stabilizers, and similar agents may be added as necessary.
  • the inventive compositions may be disposed within transdermal devices placed upon, in, or under the skin.
  • transdermal devices include patches, implants, and injections which release the active ingredient by either passive or active release mechanisms.
  • Transdermal administrations include all administration across the surface of the body and the inner linings of bodily passage including epithelial and mucosal tissues. Such administrations may be carried out using the present compositions in lotions, creams, foams, patches, suspensions, solutions, and suppositories (rectal and vaginal).
  • Transdermal administration may be accomplished through the use of a transdermal patch containing an active ingredient (i.e., a combination of an interferon and a protein kinase inhibitor) and a carrier that is non-toxic to the skin, and allows the delivery of the ingredient for systemic absorption into the bloodstream via the skin.
  • the carrier may take any number of forms such as creams and ointments, pastes, gels, and occlusive devices.
  • the creams and ointments may be viscous liquid or semisolid emulsions of either the oil- in- water or water-in-oil type. Pastes comprised of absorptive powders dispersed in petroleum or hydrophilic petroleum containing the active ingredient may be suitable.
  • a variety of occlusive devices may be used to release the active ingredient into the bloodstream such as a semi-permeable membrane covering a reservoir containing the active ingredient with or without a carrier, or a matrix containing the active ingredient.
  • Suppository formulations may be made from traditional materials, including cocoa butter, with or without the addition of waxes to alter the suppository's melting point, and glycerine.
  • Water soluble suppository bases such as polyethylene glycols of various molecular weights, may also be used.
  • the pharmaceutical composition may further comprise vaccine carriers known in the art such as, for example, thyroglobulin, albumin, tetanus toxoid, and polyamino acids such as polymers of D-lysine and D-glutamate.
  • vaccine carriers known in the art such as, for example, thyroglobulin, albumin, tetanus toxoid, and polyamino acids such as polymers of D-lysine and D-glutamate.
  • the vaccine may also include any of a variety of well known adjuvants such as, for example, incomplete Freund's adjuvant, alum, aluminium phosphate, aluminium hydroxide, monophosphoryl lipid A (MPL, GlaxoSmithKline), a saponin, CpG oligonucleotides, montanide, vitamin A and various water-in-oil emulsions prepared from biodegradable oils such as squalene and/or tocopherol, Quil A, Ribi Detox, CRL-1005, L-121 and combinations thereof.
  • adjuvants such as, for example, incomplete Freund's adjuvant, alum, aluminium phosphate, aluminium hydroxide, monophosphoryl lipid A (MPL, GlaxoSmithKline), a saponin, CpG oligonucleotides, montanide, vitamin A and various water-in-oil emulsions prepared from biodegradable oils such as squalene and/or tocop
  • an inventive combination is the only active ingredient in a pharmaceutical composition of the present invention.
  • the pharmaceutical composition further comprises one or more biologically active agents.
  • suitable biologically active agents include, but are not limited to, vaccine adjuvants and therapeutic agents such as anti-viral agents (as described above), antiinflammatory agents, immunomodulatory agents, analgesics, antimicrobial agents, antibacterial agents, antibiotics, antioxidants, antiseptic agents, and combinations thereof.
  • an interferon (or a direct acting antiviral), a protein kinase inhibitor and additional therapeutic agent(s) may be combined in one or more preparations for simultaneous, separate or sequential administration of the different components. More specifically, an inventive composition may be formulated in such a way that the interferon (or the direct acting antiviral), protein kinase inhibitor and therapeutic agent(s) can be administered together or independently from one another. For example, an interferon (or a direct acting antiviral), a protein kinase inhibitor and a therapeutic agent can be formulated together in a single composition. Alternatively, they may be maintained ⁇ e.g., in different compositions and/or containers) and administered separately.
  • the present invention provides a pharmaceutical pack or kit comprising one or more containers (e.g., vials, ampoules, test tubes, flasks or bottles) containing one or more ingredients of an inventive pharmaceutical composition, allowing administration of a combination of the present invention.
  • containers e.g., vials, ampoules, test tubes, flasks or bottles
  • Different ingredients of a pharmaceutical pack or kit may be supplied in a solid (e.g., lyophilized) or liquid form. Each ingredient will generally be suitable as aliquoted in its respective container or provided in a concentrated form.
  • Pharmaceutical packs or kits may include media for the reconstitution of lyophilized ingredients. Individual containers of the kits will preferably be maintained in close confinement for commercial sale.
  • a pharmaceutical pack or kit includes one or more additional therapeutic agent(s) (e.g., one or more anti- viral agents, as described above).
  • additional therapeutic agent(s) e.g., one or more anti- viral agents, as described above.
  • a notice or package insert in the form prescribed by a governmental agency regulating the manufacture, use or sale of pharmaceutical or biological products, which notice reflects approval by the agency of manufacture, use or sale for human administration.
  • the notice of package insert may contain instructions for use of a pharmaceutical composition according to methods of treatment disclosed herein.
  • An identifier e.g., a bar code, radio frequency, ID tags, etc., may be present in or on the kit. The identifier can be used, for example, to uniquely identify the kit for purposes of quality control, inventory control, tracking movement between workstations, etc.
  • Interferon-alpha Interferon-alpha-2a was obtained from Roche; Interferon- alpha-2b was obtained from Merck. HCVcc Production and Infection. Cell-culture derived HCVcc (Luc-Jcl) were generated as previously described (Koutsoudakis et al, J. Virol, 2006, 80: 5308- 5320; Zeisel et al, Hepatology, 2007, 46: 1722-1731). For infection experiments, Huh7.5.1 cells were incubated with HCVcc infected as described previously (Zeisel et al., World J. Gastroenterol, 2007, 13: 4824-4830; Fofana et al., Gastroenterology, 2010, 139: 953-964; Lupberger et al. , Nature Medicine, 2011, 17: 589-595).
  • Huh7.5.1 cells were pre-incubated with erlotinib or dasatinib and with IFN-CC for 1 hour. The Huh7.5.1 cells were then incubated with HCVcc Luc-Jcl in the presence of the compounds. HCVcc infection was analyzed two days later by luciferase reporter gene expression as previously described (Krieger et al., Hepatology, 2010, 54: 1144-1157; Fofana et al., Gastroenterology, 2010, 139: 953-964; Koutsoudakis et al, J. Virol, 2006, 80: 5308- 5320).
  • the Combination Index was calculated as described (Zhao et al., Clin. Cancer Res., 2004, 10: 7994-8004).
  • a CI of less than 0.9 indicates synergy; a CI equal to 0.9-1.1 indicates additivity; and a CI of more than 1.1 indicates antagonism (Zhao et al, Clin. Cancer Res., 2004, 10: 7994-8004; Zhu et al, J. Infect. Dis., 2012, 205: 656-662).
  • IFN-a combined with clinical kinase inhibitors targeting HCV entry co-factors EGFR or EphA2 inhibit HCV infection in a synergistic manner.
  • the present Applicants have previously identified EGFR and EphA2 as co-factors for HCV entry; and demonstrated that the clinical kinase inhibitors of EGFR (erlotinib) and of EphA2 (dasatinib) inhibit infection and viral spread at IC 50 0.45-0.53 ⁇ (Lupberger et al, Nature Medicine, 2011, 17: 589- 595).
  • RTK receptor tyrosine kinase
  • combinations according to the invention will be further characterized by comparative analysis of neutralization in state-of-the-art in vitro models (Krieger et al., Hepatology, 2010, 51 : 1144-1157; Fofana et al, 2010, 139: 953-964).
  • the human liver-chimeric SCID/Alb-uPA mouse model will be used in a preclinical study.
  • This model is a well characterized preclinical model for the in vivo assessment of antivirals.
  • Pharmacokinetic and toxicity of selected combinations in uPA/SCID mice will be examined as previously described (Law et al, Nat. Med., 2008, 14: 25- 27; Vanwolleghem et al, Hepatology, 2008, 47: 1846-1855). Briefly, transplanted SCID/Alb-uPA mice will be infected with HCV-infected human serum intravenously and the effect of combinations of the invention on viral load will be assessed.
  • Treatment outcome will be evaluated clinically (toxicity), virologically (viral load), and morphologically (histopathology of transplanted hepatocytes and other tissues) as described recently (Vanwolleghem et al, Gastroenterology, 2007, 133: 1144-1155).
  • the safety profile will be further assessed in non-human primates.
  • HCV infection Treatment of HCV infection in subjects chronically infected patients.
  • the combinations will be evaluated for their ability to achieve reduction in viral load by > 1 logio from the baseline value.
  • Example 5 Inhibition of HCVcc Infection using an Inventive Combination of a Protein Kinase Inhibitor and a Direct Acting Antiviral
  • Protease inhibitors telaprevir, boceprevir, danoprevir and TMC-435
  • NS5A inhibitor daclatasvir
  • polymerase inhibitors mericitabine and GS-7977 (formally known as PSI-7977)
  • Second-generation protease inhibitors have beend demonstrated to have a higher genetic barrier for resistance. However, single amino acid substitutions are able to confer drug resistance in vivo. Importanly, it has been demonstrated that several telaprevir- and boceprevir-resistance mutations confer cross-resistance to these second-generation protease inhibitors (Sarrazin et al, J. Hepatol, 2012, 56(1): S88- 100). Combination of a second-generation protease inhibitor, TMC-435 or danoprevir, and a protein kinase inhibitor (CIs of 0.3 to 0.57 - see Figure 3A and Table 2), demonstrating the relevance of adding an entry- inhibitor as a concept to improve antiviral efficacy.
  • NS5A inhibitors A number of novel DAAs have reached early- to late stage clinical development, including NS5A inhibitors and polymerase inhibitors.
  • the first NS5A inhibitor, daclatasvir (Gao et al., Nature, 2010, 465: 96-100), has been shown to have potent antiviral activity against HCV genotype 1 in monotherapy. However, its genetic barrier to resistance is low and resistant variants developed rapidly without imposing a loss of in vivo viral fitness (Gao et al, Nature, 2010, 465: 96-100).
  • Combination of daclatasvir with erlotinib or dasatinib also resulted in a synergistic activity with CIs of 0.25 to 0.40 (see Figure 3C and Table 2). The most effective combinations was the combination of daclatasvir and erlotinib decreasing its IC 50 up to 60 fold ( Figure 5A- B).
  • GS-7977 is currently in clinical development and has been suggested as having the potential to become the cornerstone of an efficacious, all-oral combination regiment for many patients with chronic hepatitis C infection (Zeisel et al, Front Biosci., 2009, 14: 3274-3285; Zeisel et al, J. Hepatol, 2011, 54: 566-576).
  • GS-7977 investigated whether protein kinase inhibitors potentiate the antiviral activity of GS-7977.
  • the present Applicants first investigated the role of IFN signal transducers in HCV entry and infection using functional silencing of gene expression by STAT-specific siRNAs.
  • STAT3 is an important regulator in the STAT1 mediated IFN-a response (Ho et al., J. Biol. Chem., 2006, 281 : 14111-14118) and has been described as an interaction partner of EGFR in proliferative cells (Lo et al, Br. J. Cancer, 2006, 94: 184-188).
  • the impact of STAT3 on HCV entry and infection was studied using RNAi in HCV permissive Huh7.5.1 cells.
  • HCVpp HCV pseudoparticles
  • HCVcc cell-culture derived HCV
  • STAT3 inhibitor Cpdl88 Xu et al, PLoS One, 2009, 4:e4783
  • STAT3 is relevant for HCV infection, and most likely plays a mechanistic role for the observed synergy between protein kinase inhibitors and interferon-alpha. Since STAT3 is a co-factor for HCV infection, it is also an antiviral target, using compounds that inhibit STAT3 function or expression.
  • the drug-resistant mutations were introduced into the Jcl -Luc plasmid (Koutsoudakis et al, J. Virol, 2006, 80: 5308-5320; Pietschmann et al, Proc. Natl. Acad. Sci. USA, 2006, 103: 7408-7413) using in vitro site directed mutagenesis (Quickchange XL, Stratagene) as previously described (Zhu et al, J. Infect. Dis., 2012, 205: 656-662).
  • Nucleotide changes were made in the HCV Luc- Jcl construct to generate the A156S or L36M or R155K amino acid substitutions in the NS3 protein.
  • a one-step polymerase chain reaction (PCR) mutagenesis was performed using mutation primers.
  • the introduction of mutations into Jcl constructs was confirmed by DNA sequence analysis.
  • HCV Cell-Cell Transmission Cell-cell transmission of HCV was assessed as previously described (Witteveldt et al, J. Gen. Virol, 20069, 90: 48-58). Briefly, producer Huh7.5.1 cells were electroporated with HCV Jcl RNA and cultured with naive target Huh7.5-GFP cells in the presence of 10 ⁇ erlotinib or DMSO solvent/rat IgG control. An HCV anti-E2-neutralizing antibody (Witteveldt et al. , J. Gen. Virol, 20069, 90: 48-58) (25 ⁇ g/mL) was added to block cell-free transmission (Witteveldt et al, J. Gen.
  • DAA-resistant variants are efficiently transmitted by cell-cell transmission.
  • Cell-cell transmission is considered more rapid and efficient than cell-free spread because it obviates rate-limiting early steps in the virus life cycle, such as virion attachment (Timpe et al., Hepatology, 2008, 47: 17-24).
  • the cell-cell transmission of DAA-resistant variants may thus accelerate viral spread, leading to viral breakthrough and treatment failure.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Epidemiology (AREA)
  • Medicinal Chemistry (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Zoology (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Immunology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)

Abstract

La présente invention concerne des combinaisons pour l'utilisation dans le traitement ou la prévention d'une infection par le VHC. En particulier, l'invention concerne des combinaisons qui comprennent au moins un inhibiteur de protéine kinase et au moins un interféron ou un antiviral agissant directement, l'inhibiteur de protéine kinase et l'interféron ou l'inhibiteur de protéine kinase et l'antiviral agissant directement agissant d'une manière fortement synergique pour inhiber une infection par le VHC de cellules sensibles. L'invention concerne également des compositions pharmaceutiques et des trousses comprenant de telles combinaisons, et des procédés d'utilisation de ces compositions et trousses pour le traitement ou la prévention d'une infection par le VHC.
PCT/EP2012/066109 2011-08-17 2012-08-17 Combinaisons d'inhibiteurs de protéine kinase et d'interférons ou d'inhibiteurs de protéine kinase et d'antiviraux agissant directement pour le traitement et la prévention d'une infection par le vhc WO2013024158A1 (fr)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
EP11306051.1 2011-08-17
EP11306051 2011-08-17

Publications (1)

Publication Number Publication Date
WO2013024158A1 true WO2013024158A1 (fr) 2013-02-21

Family

ID=46704647

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/EP2012/066109 WO2013024158A1 (fr) 2011-08-17 2012-08-17 Combinaisons d'inhibiteurs de protéine kinase et d'interférons ou d'inhibiteurs de protéine kinase et d'antiviraux agissant directement pour le traitement et la prévention d'une infection par le vhc

Country Status (1)

Country Link
WO (1) WO2013024158A1 (fr)

Cited By (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8680106B2 (en) 2011-10-21 2014-03-25 AbbVic Inc. Methods for treating HCV
US8685984B2 (en) 2011-10-21 2014-04-01 Abbvie Inc. Methods for treating HCV
US8809265B2 (en) 2011-10-21 2014-08-19 Abbvie Inc. Methods for treating HCV
WO2014147246A1 (fr) * 2013-03-21 2014-09-25 INSERM (Institut National de la Santé et de la Recherche Médicale) Méthode et composition pharmaceutique pour l'utilisation dans le traitement de maladies hépatiques chroniques associées à une faible expression d'hepcidine
US8853176B2 (en) 2011-10-21 2014-10-07 Abbvie Inc. Methods for treating HCV

Citations (24)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0170204A2 (fr) 1984-08-01 1986-02-05 BOEHRINGER INGELHEIM INTERNATIONAL GmbH Séquences génétiques, interféron peptide du type I codé par celles-ci et les organismes les produisant
US4727138A (en) 1981-10-19 1988-02-23 Genentech, Inc. Human immune interferon
US4820638A (en) 1982-05-28 1989-04-11 Dr. Karl Thomae Gmbh Novel alpha interferon species produced by recombinant means
US4845196A (en) 1985-06-24 1989-07-04 G. D. Searle & Co. Modified interferon gammas
US4897471A (en) 1982-05-06 1990-01-30 Amgen Consensus human leukocyte interferon
US5574137A (en) 1983-12-16 1996-11-12 Genentech, Inc. Recombinant gamma interferons having enhanced stability and methods therefor
US5612460A (en) 1989-04-19 1997-03-18 Enzon, Inc. Active carbonates of polyalkylene oxides for modification of polypeptides
US5690925A (en) 1983-12-16 1997-11-25 Genentech, Inc. Recombinant gamma interferons having enhanced stability and methods therefor
US5711944A (en) 1993-11-10 1998-01-27 Enzon, Inc. Interferon polymer conjugates
EP0830559A1 (fr) 1995-05-24 1998-03-25 Trijicon, Inc. Dispositif de vision reflexe pour la vision de jour et de nuit
US5795779A (en) 1982-11-01 1998-08-18 Berlex Laboratories, Inc. Human interferon-β (IFN-β) produced in Chinese hamster ovary (CHO) cells
US5951974A (en) 1993-11-10 1999-09-14 Enzon, Inc. Interferon polymer conjugates
WO2002086087A2 (fr) 2001-04-20 2002-10-31 Zymogenetics, Inc. Famille proteique de cytokine
EP1256348A1 (fr) 2000-02-14 2002-11-13 Mitsubishi Pharma Corporation Remedes contre l'hepatite c
WO2004037995A2 (fr) 2002-10-23 2004-05-06 Zymogenetics, Inc. Procedes de traitement d'infection virale a l'aide de il-28 et il-29
WO2005023862A2 (fr) 2003-08-07 2005-03-17 Zymogenetics, Inc. Preparations homogenes d'il-28 et d'il-29
WO2005117885A1 (fr) * 2004-06-04 2005-12-15 Bioniche Life Sciences Inc. Utilisation de l’imatinib pour traiter les troubles et les infections virales du foie
WO2006005465A1 (fr) 2004-07-08 2006-01-19 Istituto Di Ricerche Di Biologia Molecolare P Angeletti Spa Proteine de liaison a des antigenes dirigees contre le recepteur capteur b1, lesquelles inhibent la replication du vhc
WO2007130646A2 (fr) 2006-05-04 2007-11-15 The Rockefeller University Co-récepteur de vhc et ses méthodes d'utilisation
US7572437B2 (en) 2002-07-01 2009-08-11 Yan Fu Long acting human interferon analogs
WO2010013466A1 (fr) * 2008-07-29 2010-02-04 国立大学法人東京大学 Inhibition de la réplication du virus de l'hépatite c par l'inhibiteur cdk
WO2010034812A1 (fr) 2008-09-25 2010-04-01 INSERM (Institut National de la Santé et de la Recherche Médicale) Anticorps anti-claudine 1 monoclonaux pour inhiber l'infection au virus de l'hépatite c
WO2010034670A2 (fr) * 2008-09-26 2010-04-01 INSERM (Institut National de la Santé et de la Recherche Médicale) Kinases de cellules hôtes comme cibles de thérapies antivirales contre l'infection par le virus de l'hépatite c
WO2011112516A1 (fr) * 2010-03-08 2011-09-15 Ico Therapeutics Inc. Traitement et prévention de l'infection par le virus de l'hépatite c en utilisant des oligonucléotides antisens de la kinase c-raf

Patent Citations (26)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4727138A (en) 1981-10-19 1988-02-23 Genentech, Inc. Human immune interferon
US4762791A (en) 1981-10-19 1988-08-09 Genentech, Inc. Human immune interferon
US4929554A (en) 1981-10-19 1990-05-29 Goeddel David V Human immune interferon
US4897471A (en) 1982-05-06 1990-01-30 Amgen Consensus human leukocyte interferon
US4820638A (en) 1982-05-28 1989-04-11 Dr. Karl Thomae Gmbh Novel alpha interferon species produced by recombinant means
US5795779A (en) 1982-11-01 1998-08-18 Berlex Laboratories, Inc. Human interferon-β (IFN-β) produced in Chinese hamster ovary (CHO) cells
US5574137A (en) 1983-12-16 1996-11-12 Genentech, Inc. Recombinant gamma interferons having enhanced stability and methods therefor
US5690925A (en) 1983-12-16 1997-11-25 Genentech, Inc. Recombinant gamma interferons having enhanced stability and methods therefor
EP0170204A2 (fr) 1984-08-01 1986-02-05 BOEHRINGER INGELHEIM INTERNATIONAL GmbH Séquences génétiques, interféron peptide du type I codé par celles-ci et les organismes les produisant
US4845196A (en) 1985-06-24 1989-07-04 G. D. Searle & Co. Modified interferon gammas
US5612460A (en) 1989-04-19 1997-03-18 Enzon, Inc. Active carbonates of polyalkylene oxides for modification of polypeptides
US5711944A (en) 1993-11-10 1998-01-27 Enzon, Inc. Interferon polymer conjugates
US5951974A (en) 1993-11-10 1999-09-14 Enzon, Inc. Interferon polymer conjugates
EP0830559A1 (fr) 1995-05-24 1998-03-25 Trijicon, Inc. Dispositif de vision reflexe pour la vision de jour et de nuit
EP1256348A1 (fr) 2000-02-14 2002-11-13 Mitsubishi Pharma Corporation Remedes contre l'hepatite c
WO2002086087A2 (fr) 2001-04-20 2002-10-31 Zymogenetics, Inc. Famille proteique de cytokine
US7572437B2 (en) 2002-07-01 2009-08-11 Yan Fu Long acting human interferon analogs
WO2004037995A2 (fr) 2002-10-23 2004-05-06 Zymogenetics, Inc. Procedes de traitement d'infection virale a l'aide de il-28 et il-29
WO2005023862A2 (fr) 2003-08-07 2005-03-17 Zymogenetics, Inc. Preparations homogenes d'il-28 et d'il-29
WO2005117885A1 (fr) * 2004-06-04 2005-12-15 Bioniche Life Sciences Inc. Utilisation de l’imatinib pour traiter les troubles et les infections virales du foie
WO2006005465A1 (fr) 2004-07-08 2006-01-19 Istituto Di Ricerche Di Biologia Molecolare P Angeletti Spa Proteine de liaison a des antigenes dirigees contre le recepteur capteur b1, lesquelles inhibent la replication du vhc
WO2007130646A2 (fr) 2006-05-04 2007-11-15 The Rockefeller University Co-récepteur de vhc et ses méthodes d'utilisation
WO2010013466A1 (fr) * 2008-07-29 2010-02-04 国立大学法人東京大学 Inhibition de la réplication du virus de l'hépatite c par l'inhibiteur cdk
WO2010034812A1 (fr) 2008-09-25 2010-04-01 INSERM (Institut National de la Santé et de la Recherche Médicale) Anticorps anti-claudine 1 monoclonaux pour inhiber l'infection au virus de l'hépatite c
WO2010034670A2 (fr) * 2008-09-26 2010-04-01 INSERM (Institut National de la Santé et de la Recherche Médicale) Kinases de cellules hôtes comme cibles de thérapies antivirales contre l'infection par le virus de l'hépatite c
WO2011112516A1 (fr) * 2010-03-08 2011-09-15 Ico Therapeutics Inc. Traitement et prévention de l'infection par le virus de l'hépatite c en utilisant des oligonucléotides antisens de la kinase c-raf

Non-Patent Citations (82)

* Cited by examiner, † Cited by third party
Title
BACON ET AL., NEW ENGL. J. MED., vol. 364, 2011, pages 1207 - 1217
BARTH ET AL., J. BIOL. CHEM., vol. 278, 2003, pages 41003 - 41012
BARTOSCH ET AL., J. EXP. MED., vol. 197, 2003, pages 633 - 642
BLONSKI ET AL., CLIN. LIVER DIS., vol. 12, 2008, pages 661 - 674
BRIMACOMBE ET AL., J. VIROL., vol. 85, 2011, pages 596 - 605
BROWN, NATURE, vol. 436, 2005, pages 973 - 978
BUCKWOLD ET AL., ANTIVIRAL RES., vol. 73, 2007, pages 118 - 125
CASTELLO ET AL., CLIN. IMMUNOL., vol. 134, 2010, pages 237 - 250
CATANESE ET AL., J. VIROL., vol. 81, 2007, pages 8063 - 8071
CHISARI, NATURE, vol. 435, 2005, pages 930 - 932
DAYHOFF ET AL.: "Atlas of Protein Sequence and Structure", vol. 22, 1978, NAT. BIOMED. RES. FOUNDATION, pages: 354 - 352
E.W. MARTIN: "Remington's Pharmaceutical Sciences", 1990, MACK PUBLISHING CO.
EVANS ET AL., NATURE, vol. 446, 2007, pages 801 - 805
FAFI-KREMER ET AL., J. EXP. MED., vol. 207, 2010, pages 2019 - 2031
FOFANA ET AL., GASTROENTEROLOGY, vol. 139, 2010, pages 953 - 964
FOFANA ET AL., GASTROENTEROLOGY, vol. 139, 2010, pages 953 - 964,EL-4
FOFANA ISABEL ET AL: "SYNERGY OF ENTRY INHIBITORS AND DIRECT ACTING ANTIVIRALS OR INTERFERON-ALFA IDENTIFIES NOVEL ANTIVIRAL COMBINATIONS FOR HEPATITIS C VIRUS INFECTION", HEPATOLOGY, vol. 54, no. Suppl. 1, October 2011 (2011-10-01), & 62ND ANNUAL MEETING OF THE AMERICAN-ASSOCIATION-FOR-THE-STUDY-OF-LIVE R-DISEASES (AASLD); SAN FRANCISCO, CA, USA; NOVEMBER 04 -08, 2011, pages 401A, XP009155976 *
FUKUTOMI ET AL., J. HEPATOLOGY, vol. 34, 2001, pages 100 - 107
GANE ET AL., LANCET, vol. 376, 2010, pages 1467 - 1475
GAO ET AL., NATURE, vol. 465, 2010, pages 96 - 100
GROVE ET AL., J. VIROL., vol. 81, 2007, pages 3162 - 3169
HEZODE ET AL., NEW ENGL. J. MED., vol. 360, 2009, pages 1839 - 1850
HO ET AL., J. BIOL. CHEM., vol. 281, 2006, pages 14111 - 14118
HOFINANN ET AL., NAT. REV. GASTROENTEROL. HEPATOL., vol. 8, 2011, pages 257 - 264
HOFINANN ET AL., NAT. REV; GASTROENTEROL. HEPATOL., vol. 8, 2011, pages 257 - 264
HOOFNAGLE, HEPATOLOGY, vol. 36, 2002, pages 21 - 529
JACOBSON ET AL., CLIN. GASTROENTEROL. HEPATOL., vol. 8, 2010, pages 924 - 933
JOACHIM LUPBERGER ET AL: "EGFR and EphA2 are host factors for hepatitis C virus entry and possible targets for antiviral therapy", NATURE MEDICINE, NATURE PUBLISHING GROUP, NEW YORK, NY, US, vol. 17, no. 5, 1 May 2011 (2011-05-01), pages 589 - 596, XP002666425, ISSN: 1078-8956, [retrieved on 20110424], DOI: 10.1038/NM.2341 *
KAPADIA ET AL., J. VIROL., vol. 81, 2007, pages 374 - 383
KECK ET AL., J. VIROL., vol. 79, 2005, pages 13199 - 13208
KOUTSOUDAKIS ET AL., J. VIROL., vol. 80, 2006, pages 5308 - 5320
KRIEGER ET AL., HEPATOLOGY, vol. 51, 2010, pages 1144 - 1157
KRIEGER ET AL., HEPATOLOGY, vol. 54, 2010, pages 1144 - 1157
KWO ET AL., LANCET, vol. 376, 2010, pages 705 - 716
LAUER ET AL., N. ENGL. J. MED., vol. 345, 2001, pages 41 - 52
LAW ET AL., NAT. MED., vol. 14, 2008, pages 25 - 27
LAW ET AL., NAT., vol. 14, 2008, pages 25 - 27
LO ET AL., BR. J. CANCER, vol. 94, 2006, pages 184 - 188
LUPBERGER ET AL., NATURE MEDICINE, vol. 17, 2011, pages 589 - 595
MATSUMURA ET AL., GASTROENTEROLOGY, vol. 137, 2009, pages 673 - 681
MCGIVERN ET AL., ONCOGENE, vol. 30, 2011, pages 1969 - 1983
MCHUTCHISON ET AL., NEW ENGL. J. MED., vol. 362, 2010, pages 1292 - 1303
NELSON ET AL., GASTROENTEROLOGY, vol. 139, 2010, pages 1267 - 1276
OSBURN ET AL., GASTROENTEROLOGY, vol. 138, 2009, pages 315 - 324
PAWLOTSKY ET AL., HEPATOLOGY, vol. 53, 2011, pages 1742 - 1751
PAWLOTSKY, HEPATOLOGY, vol. 53, 2011, pages 1742 - 1751
PAWLOTSKY, TRENDS MICROBIOL., vol. 12, 2004, pages 96 - 102
PEREIRA ET AL., NAT. REV. GASTROENTEROL. HEPATOL., vol. 6, 2009, pages 403 - 411
PESTKA ET AL., PROC. NATL. ACAD. SCI. USA, vol. 104, 2007, pages 6025 - 630
PIETSCHMANN ET AL., PROC. NATL. ACAD. SCI. USA, vol. 103, 2006, pages 7408 - 7413
PILERI ET AL., SCIENCE, vol. 282, 1998, pages 938 - 941
PLOSS ET AL., NATURE, vol. 457, 2009, pages 882 - 886
POORDAD ET AL., NEW ENGL. J. MED., vol. 364, 2011, pages 1195 - 1206
POWERS ET AL., LIVER TRANSPL., vol. 12, 2006, pages 207 - 216
PRICHARD ET AL., ANTIVIRAL RES., vol. 14, 1990, pages 181 - 205
ROHLOFF ET AL., J. DIABETES SCI. TECHNOL., vol. 2, 2008, pages 461 - 467
SANG HOON AHN ET AL., GUT AND LIVER, vol. 3, 2009, pages 20 - 25
SARRAZIN ET AL., GASTROENTEROL., vol. 138, 2010, pages 447 - 462
SARRAZIN ET AL., J. HEPATOL., vol. 56, no. 1, 2012, pages 88 - 100
SCARSELLI ET AL., EMBO J., vol. 21, 2002, pages 5017 - 5025
SCHAEFER ET AL., GASTROENTEROLOGY, vol. 142, 2012, pages 1340 - 1350 E1341
SEEFF ET AL., HEPATOLOGY, vol. 36, 2002, pages 1 - 2
SEEFF, SEMIN. GASTROINTEST., vol. 6, 1995, pages 20 - 27
STEPHEN J POLYAK ET AL: "A Summary of the 18th International Symposium on Hepatitis C Virus and Related Viruses", GASTROENTEROLOGY, vol. 142, no. 1, 12 September 2011 (2011-09-12), pages e1 - e5, XP028342851, ISSN: 0016-5085, [retrieved on 20111112], DOI: 10.1053/J.GASTRO.2011.11.002 *
SYDER ET AL., J. HEPATOL., vol. 54, 2011, pages 48 - 55
TIMPE ET AL., GUT, vol. 57, 2008, pages 1728 - 1737
TIMPE ET AL., HEPATOLOGY, vol. 47, 2008, pages 17 - 24
VANWOLLEGHEM ET AL., GASTROENTEROLOGY, vol. 133, 2007, pages 1144 - 1155
VANWOLLEGHEM ET AL., HEPATOLOGY, vol. 47, 2008, pages 1846 - 1855
WATT ET AL., AM. J. TRANSPLANT, vol. 9, 2009, pages 1707 - 1713
WITTEVELDT ET AL., J. GEN. VIROL., no. 90, pages 48 - 58
WITTEVELDT ET AL., J. GEN. VIROL., vol. 20069, no. 90, pages 48 - 58
XU ET AL., PLOS ONE, vol. 4, 2009, pages E4783
ZEISEL ET AL., FRONT BIOSCI., vol. 14, 2009, pages 3274 - 3285
ZEISEL ET AL., HEPATOLOGY, vol. 46, 2007, pages 1722 - 1731
ZEISEL ET AL., HEPATOLOGY, vol. 48, 2008, pages 299 - 307
ZEISEL ET AL., J. HEPATOL., vol. 54, 2011, pages 566 - 576
ZEISEL ET AL., WORLD J. GASTROENTEROL., vol. 13, 2007, pages 4824 - 4830
ZEUZEM ET AL., GASTROENTEROLOGY, vol. 139, 2010, pages 1257 - 1266
ZHAO ET AL., CLIN. CANCER RES., vol. 10, 2004, pages 7994 - 8004
ZHONG ET AL., PROC. NATL. ACAD. SCI. USA, vol. 102, 2005, pages 9294 - 2929
ZHU ET AL., J. INFECT. DIS., vol. 205, 2012, pages 656 - 662

Cited By (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8680106B2 (en) 2011-10-21 2014-03-25 AbbVic Inc. Methods for treating HCV
US8685984B2 (en) 2011-10-21 2014-04-01 Abbvie Inc. Methods for treating HCV
US8809265B2 (en) 2011-10-21 2014-08-19 Abbvie Inc. Methods for treating HCV
US8853176B2 (en) 2011-10-21 2014-10-07 Abbvie Inc. Methods for treating HCV
US8969357B2 (en) 2011-10-21 2015-03-03 Abbvie Inc. Methods for treating HCV
US8993578B2 (en) 2011-10-21 2015-03-31 Abbvie Inc. Methods for treating HCV
US9452194B2 (en) 2011-10-21 2016-09-27 Abbvie Inc. Methods for treating HCV
WO2014147246A1 (fr) * 2013-03-21 2014-09-25 INSERM (Institut National de la Santé et de la Recherche Médicale) Méthode et composition pharmaceutique pour l'utilisation dans le traitement de maladies hépatiques chroniques associées à une faible expression d'hepcidine

Similar Documents

Publication Publication Date Title
EP1750713B1 (fr) Utilisation de l"imatinib pour traiter les troubles et les infections virales du foie
Pawlotsky New antiviral agents for hepatitis C
US8575195B2 (en) Treatment of hepatitis C virus related diseases using hydroxychloroquine or a combination of hydroxychloroquine and an anti-viral agent
Poordad et al. Emerging therapeutic options in hepatitis C virus infection
WO2013024155A1 (fr) Combinaisons d'anticorps anti-facteur d'entrée de vhc et d'agents antiviraux à action directe pour le traitement et la prévention de l'infection par le vhc
WO2013024158A1 (fr) Combinaisons d'inhibiteurs de protéine kinase et d'interférons ou d'inhibiteurs de protéine kinase et d'antiviraux agissant directement pour le traitement et la prévention d'une infection par le vhc
JP2014532657A (ja) C型肝炎ウイルスを処置するための方法および組成物
TW200816990A (en) Identification and characterization of HCV replicon variants with reduced susceptibility to HCV-796, and methods related thereto
US20070258946A1 (en) Combination Therapy for Treating Hepatitis C Virus Infection
Jesudian et al. Emerging therapeutic targets for hepatitis C virus infection
CA2566677A1 (fr) Therapie combinatoire pour traiter l'infection par le virus de l'hepatite
US20170224765A1 (en) Treatments of hepatitis c virus infection
RU2392963C2 (ru) Синтетические варианты гипергликозилированного протеазо-резистентного полипептида, пероральные композиции и способы применения таких вариантов
De Bruijne et al. New developments in the antiviral treatment of hepatitis C
Poordad et al. Developments in hepatitis C therapy during 2000–2002
JP7381493B2 (ja) Hcvを治療するための併用療法
Gaetano et al. The era of Direct-Acting Antivirals: The evolving Role of Interferon and Ribavirin for the Treatment of chronic Hepatitis c
JP2018519306A (ja) Hcvを処置するための方法
Leav et al. Safety and pharmacokinetics of a novel human monoclonal antibody directed against the E2 glycoprotein of hepatitis C virus (MBL-HCV1) in healthy volunteers
Sodhi et al. New Horizons and Perspectives in the Management of Chronic Hepatitis C
WO2013024156A2 (fr) Combinaisons d'anticorps contre le facteur d'entrée du vhc et d'interférons pour le traitement et la prévention d'une infection par le vhc
Jaimie Aslanidis Examining the pathological nature of Hepatitis C and current drug therapies used in an Australian general practice context.

Legal Events

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

Ref document number: 12748461

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 12748461

Country of ref document: EP

Kind code of ref document: A1