EP3271371A1 - Dérivés nucléotidiques qui sont des inhibiteurs du vhc pour une utilisation dans le traitement de l'hépatite c - Google Patents

Dérivés nucléotidiques qui sont des inhibiteurs du vhc pour une utilisation dans le traitement de l'hépatite c

Info

Publication number
EP3271371A1
EP3271371A1 EP16759215.3A EP16759215A EP3271371A1 EP 3271371 A1 EP3271371 A1 EP 3271371A1 EP 16759215 A EP16759215 A EP 16759215A EP 3271371 A1 EP3271371 A1 EP 3271371A1
Authority
EP
European Patent Office
Prior art keywords
compound
pharmaceutically acceptable
acceptable salt
amount
prodrug
Prior art date
Legal status (The legal status 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 status listed.)
Withdrawn
Application number
EP16759215.3A
Other languages
German (de)
English (en)
Other versions
EP3271371A4 (fr
Inventor
Gennadiy KALAYANOV
Pedro Pinho
Paul TARGETT-ADAMS
Staffan Torssell
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Medivir AB
Original Assignee
Medivir AB
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 Medivir AB filed Critical Medivir AB
Publication of EP3271371A1 publication Critical patent/EP3271371A1/fr
Publication of EP3271371A4 publication Critical patent/EP3271371A4/fr
Withdrawn legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07HSUGARS; DERIVATIVES THEREOF; NUCLEOSIDES; NUCLEOTIDES; NUCLEIC ACIDS
    • C07H19/00Compounds containing a hetero ring sharing one ring hetero atom with a saccharide radical; Nucleosides; Mononucleotides; Anhydro-derivatives thereof
    • C07H19/02Compounds containing a hetero ring sharing one ring hetero atom with a saccharide radical; Nucleosides; Mononucleotides; Anhydro-derivatives thereof sharing nitrogen
    • C07H19/04Heterocyclic radicals containing only nitrogen atoms as ring hetero atom
    • C07H19/06Pyrimidine radicals
    • C07H19/10Pyrimidine radicals with the saccharide radical esterified by phosphoric or polyphosphoric acids
    • 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
    • A61K31/40Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with one nitrogen as the only ring hetero atom, e.g. sulpiride, succinimide, tolmetin, buflomedil
    • A61K31/407Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with one nitrogen as the only ring hetero atom, e.g. sulpiride, succinimide, tolmetin, buflomedil condensed with other heterocyclic ring systems, e.g. ketorolac, physostigmine
    • 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
    • A61K31/41641,3-Diazoles
    • A61K31/41781,3-Diazoles not condensed 1,3-diazoles and containing further heterocyclic rings, e.g. pilocarpine, nitrofurantoin
    • 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
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/439Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom the ring forming part of a bridged ring system, e.g. quinuclidine
    • 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
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/47Quinolines; Isoquinolines
    • A61K31/4709Non-condensed quinolines 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
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/47Quinolines; Isoquinolines
    • A61K31/472Non-condensed isoquinolines, e.g. papaverine
    • A61K31/4725Non-condensed isoquinolines, e.g. papaverine 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
    • A61K31/498Pyrazines or piperazines ortho- and peri-condensed with carbocyclic ring systems, e.g. quinoxaline, phenazine
    • 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
    • A61K31/513Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim having oxo groups directly attached to the heterocyclic ring, e.g. cytosine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/7042Compounds having saccharide radicals and heterocyclic rings
    • A61K31/7052Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides
    • A61K31/706Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides containing six-membered rings with nitrogen as a ring hetero atom
    • A61K31/7064Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides containing six-membered rings with nitrogen as a ring hetero atom containing condensed or non-condensed pyrimidines
    • A61K31/7068Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides containing six-membered rings with nitrogen as a ring hetero atom containing condensed or non-condensed pyrimidines having oxo groups directly attached to the pyrimidine ring, e.g. cytidine, cytidylic acid
    • A61K31/7072Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides containing six-membered rings with nitrogen as a ring hetero atom containing condensed or non-condensed pyrimidines having oxo groups directly attached to the pyrimidine ring, e.g. cytidine, cytidylic acid having two oxo groups directly attached to the pyrimidine ring, e.g. uridine, uridylic acid, thymidine, zidovudine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/04Peptides having up to 20 amino acids in a fully defined sequence; Derivatives thereof
    • A61K38/12Cyclic peptides, e.g. bacitracins; Polymyxins; Gramicidins S, C; Tyrocidins A, B or C
    • A61K38/13Cyclosporins
    • 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
    • A61K38/20Interleukins [IL]
    • 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]
    • 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
    • A61K38/55Protease inhibitors
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/14Antivirals for RNA viruses

Definitions

  • the present invention relates to a specific nucleotide derivative which is an inhibitor of the polymerase of hepatitis C virus (HCV) for use in the treatment or prophylaxis of hepatitis C, including HCV genotype 3 virus infection.
  • HCV hepatitis C virus
  • the invention further relates to combination therapy for HCV GT3 and all other genotypes with the nucleotide derivative and certain specified antiviral agents.
  • HCV is a single stranded, positive-sense RNA virus belonging to the Flaviviridae family of viruses in the hepacivirus genus.
  • the NS5B region of the RNA polygene encodes an RNA dependent RNA polymerase (RdRp), which is essential to viral replication.
  • RdRp RNA dependent RNA polymerase
  • a majority of infected individuals develop chronic hepatitis because HCV replicates preferentially in hepatocytes but is not directly cytopathic.
  • the lack of a vigorous T-lymphocyte response and the high propensity of the virus to mutate appear to promote a high rate of chronic infection.
  • Chronic hepatitis can progress to liver fibrosis, leading to cirrhosis, end-stage liver disease and HCC (hepatocellular carcinoma), making it the leading cause of liver transplantations.
  • HCV genotype 1 is the predominant genotype in Europe and in the US.
  • the extensive genetic heterogeneity of HCV has important diagnostic and clinical implications, perhaps explaining difficulties in vaccine development and the lack of response to current therapy.
  • HCV HCV
  • Transmission of HCV can occur through contact with contaminated blood or blood products, for example following blood transfusion or intravenous drug use.
  • the introduction of diagnostic tests used in blood screening has led to a downward trend in post-transfusion HCV incidence.
  • the existing infections will continue to present a serious medical and economic burden for decades.
  • the first generation HCV therapies were based on (pegylated) interferon-alpha (IFN-a) in combination with ribavirin.
  • IFN-a interferon-alpha
  • This combination therapy yields a sustained virologic response in more than 40% of patients infected by genotype 1 viruses and about 80% of those infected by genotypes 2 and 3.
  • this combination therapy has significant side effects and is poorly tolerated in many patients.
  • Major side effects include influenza-like symptoms, hematologic abnormalities and neuropsychiatric symptoms.
  • the second generation of HCV treatments added the HCV protease inhibitors telepravir or boceprevir, allowing treatment times to be shortened, but generating a significant number of serious side-effects.
  • nucleoside/nucleotide HCV polymerase inhibitors such as sofosbuvir
  • Sofosbuvir for example has been approved by the FDA and EMA for treatment of HCV genotypes 1 and 4.
  • Response rates in the sofosbuvir - ribavirin group were lower among patients with genotype 3 infection than amongst those with genotype 2 infection (56% vs. 97%).
  • the NS5B RdRp is absolutely essential for replication of the single-stranded, positive sense HCV RNA genome which makes it an attractive target for the development of antiviral compounds.
  • NS5B inhibitors There are two major classes of NS5B inhibitors: non-nucleoside inhibitors (NNIs) and nucleoside analogues.
  • NNIs non-nucleoside inhibitors
  • nucleoside analogues The NNIs bind to allosteric regions of the protein whereas the nucleoside inhibitors are anabolized to the corresponding nucleotide and act as alternative substrate for the polymerase.
  • the formed nucleotide is then incorporated in the nascent RNA polymer chain and can terminate the growth of the polymer chain.
  • both nucleoside and non-nucleoside inhibitors of NS5B are known.
  • the inhibition mechanism of nucleoside inhibitors involves phosphorylation of the nucleoside to the corresponding triphosphate.
  • the phosphorylation is commonly mediated by host cell kinases and is an absolute requirement for the nucleoside to be active as an alternative substrate for the NS5B polymerase.
  • the first phosphorylation step i.e. conversion of the nucleoside to the nucleoside 5'-monophosphate is the rate limiting step.
  • Subsequent conversion of the monophosphate to the di- and tri-phosphate usually proceed facile and are usually not rate limiting.
  • a strategy for increasing nucleoside triphosphate production is to use cell permeable nucleoside prodrugs of the monophosphate, i.e.
  • nucleoside carrying a masked phosphate moiety a "prodrug moiety" which are susceptible to intracellular enzymatic activation leading to a nucleoside monophosphate.
  • the thus formed monophosphate is subsequently converted to the active triphosphate by cellular kinases.
  • HCV inhibitors that may overcome the disadvantages of current HCV therapy such as side effects e.g. toxicity, limited efficacy, lack of pan-genotypic coverage, the emerging of resistance, and compliance failures, as well as improve the sustained viral response.
  • the present invention provides a new HCV inhibiting compound for use in the treatment of hepatitis C genotype 3 virus infection and which has useful properties regarding one or more of the following parameters: antiviral efficacy; pan-genotypic coverage; favourable profile of resistance development; lack of toxicity and genotoxicity; favourable pharmacokinetics and pharmacodynamics; and ease of formulation and administration.
  • an HCV inhibiting compound for use in the treatment of hepatitis C genotype 3 virus infection of the present invention need not demonstrate an improvement in every respect over all known compounds but may instead provide a balance of properties which in combination mean that the HCV inhibiting compound is a valuable alternative pharmaceutical agent.
  • Compounds for use according the invention may also be attractive due to the fact that they lack activity against other viruses, i.e. are selective, in particular against HIV. HIV infected patients often suffer from co-infections such as HCV. Treatment of such patients with an HCV inhibitor that also inhibits HIV may lead to the emergence of resistant HIV strains. Description of the Invention
  • the present invention provides the diastereomeric Compound 1 A:
  • a further aspect of the invention provides the diastereomeric Compound 1A for use in the treatment or prophylaxis of hepatitis C genotype 2a, 3a, 4a, 5a or 6a virus infection
  • the invention relates to a diastereomeric compound of formula (1 A) for use in the treatment of hepatitis C genotype 3a.
  • the compound of formula (1 A) may optionally be provided in the form of a pharmaceutically acceptable salt and/or solvate.
  • the compound for use according to the invention is provided in the form of a pharmaceutically acceptable salt.
  • the compound for use according to the invention is provided in the form of a pharmaceutically acceptable solvate.
  • the compound for use according to the invention is provided in its free form.
  • the active metabolite of the diastereomeric compound of formula (1 A) is believed to be the triphosphate,having the structure:
  • the invention provides a compound of formula (1 A)
  • the invention for use in the treatment of hepatitis C genotype 2a, 3a, 4a, 5a or 6a infection.
  • the invention relates to a compound of formula (1 A) for use in the treatment of hepatitis C genotype 3a. Consequently, there is provided a compound of formula (1A) for use in the treatment or prophylaxis of HCV genotype 3 infection.
  • the invention provides compounds of formula (1 A) for use in the treatment of HCV genotype 3 infection in humans.
  • the invention provides compounds of formula (1 A) for use in the prophylaxis of HCV genotype 3 infection in humans.
  • a compound of formula (1A) in the manufacture of a medicament for the treatment or prophylaxis of HCV genotype 3 infection, especially a medicament for the treatment of HCV genotype 3 infection.
  • the invention provides pharmaceutical compositions comprising a compound of formula (1 A) in association with a pharmaceutically acceptable adjuvant, diluent, excipient or carrier for use in the treatment of hepatitis C genotype 3 infection.
  • the pharmaceutical composition will typically contain an antivirally effective amount (e.g. for humans) of the compound of formula (1A), although sub-therapeutic amounts of the compound of formula (1A) may nevertheless be of value when intended for use in combination with other agents or in multiple doses.
  • One aspect of the invention relates to a pharmaceutical composition that comprises, in a pharmaceutically acceptable vehicle, carrier or diluent: a first compound, with the formula Compound 1A
  • HCV antivirals selected from:
  • the invention thus provides antiviral compositions and HCV antiviral treatment regimes comprising:
  • Compound 1 A in each case includes its pharmaceutically acceptable salts.
  • Asunaprevir (INN), has shown to be effective in inhibiting HCV replication.
  • the aforementioned compound can be obtained using methods known to those skilled in the art, including, for example, those methods disclosed in WO2003/099274, and WO2009/085659, which are hereby incorporated by reference in its entirety.
  • asunaprevir is believed to inhibit the HCV protease, in particular the NS3/4A protease.
  • Pharmaceutically acceptable salts of asunaprevir can be utilized in the compositions described herein.
  • Daclatasvir (INN), has been demonstrated to be effective in inhibiting HCV replication. Daclatasvir can be obtained using methods known to those skilled in the art, such as those methods described in WO2008/021927 & 21928 and WO2009/20828, which is hereby incorporated by reference in its entirety. Although this invention is not limited by any particular theory, it is believed that daclatasvir is an NS5A inhibitor where NS5A is a protein involved in the replication of the hepatitis C virus.
  • Beclabuvir (INN) has been demonstrated to be effective in inhibiting HCV replication. Beclauvir can be obtained using methods known to those skilled in the art, such as those methods described in WO2014/014885 and WO2007/136982, which is hereby incorporated by reference in its entirety. Although this invention is not limited by any particular theory, it is believed that beclabuvir is a non-nucleoside inhibitor of the HCV NS5A polymerase.
  • the composition can further include a pharmaceutically acceptable excipient, diluent and/or carrier, such as those described herein.
  • a pharmaceutically acceptable excipient, diluent and/or carrier such as those described herein.
  • an antiviral effective daily amount of Compound 1 A within the combination treatment of the invention would be from about 0.01 to about 700 mg/kg, or about 0.5 to about 400 mg/kg, or about 1 to about 250 mg/kg, or about 2 to about 200 mg/kg, or about 10 to about 150 mg/kg, or about 0.1 to about 50 mg/kg, or about 1 to about 20 mg/kg, or about 2 to about 10 mg/kg, or about 5 to about 8 mg/kg body weight. It may be appropriate to administer the required dose as one, two, three, four or more sub-doses at appropriate intervals throughout the day.
  • Said sub-doses may be formulated as unit dosage forms, for example, containing about 1 to about 5000 mg Compound 1 , or about 50 to about 3000 mg Compound 1 , or about 100 to about 1000 mg Compound 1 , or about 200 to about 600 mg Compound 1 , or about 100 to about 400 mg Compound 1 , or about 7 to about 3500 mg Compound 1 , or about 70 to about 1400 mg Compound 1 A, or about 140 to about 700 mg Compound 1 , or about 340 to about 580 mg of Compound 1 A per unit dosage form.
  • the unit dosage form is formulated for QD dosing with one of the above ranges.
  • compositions can include an amount of asunaprevir, or a pharmaceutically acceptable salt thereof in the range of about 50 mg to about 500 mg QD or BID. In other embodiments, the composition can include an amount of asunaprevir, or a pharmaceutically acceptable salt thereof in the range of about 100 mg to about 300 mg QD or BID. In still other embodiments, the composition can include an amount of asunaprevir, or a pharmaceutically acceptable salt thereof, at a dosage of about 100 mg 150 or 200 mg BID or preferably QD.
  • the composition can include an amount of daclatasvir, or a pharmaceutically acceptable salt thereof in the range of about 10 mg to about 225 mg BID or QD. In other embodiments, the composition can include an amount of daclatasvir, or a pharmaceutically acceptable salt thereof, in the range of about 15 mg to about 100 mg BID or preferably QD. In still other embodiments, the composition can include an amount of daclatasvir or a pharmaceutically acceptable salt thereof in the range of about 30-60 mg QD. In yet still other embodiments, a loading dose of 30 - 100 mg such as 30, 50 or 60 mg declatasvir or pharmaceutically acceptable salt thereof can be administered for the initial 1 -3 days of therapy.
  • compositions can include an amount of beclabuvir, or a pharmaceutically acceptable salt thereof in the range of about 25 mg to about 225 mg BID or QD. In other embodiments, the composition can include an amount of beclabuvir, or a pharmaceutically acceptable salt thereof, in the range of about 50 mg to about 100 mg QD or typically BID. In still other embodiments, the composition can include an amount of beclabuvir or a pharmaceutically acceptable salt thereof in the range of about 75 mg BID or QD.
  • a potential advantage of making a combination of Compound 1 A; and asunaprevir, daclatasvir and/or beclabuvir or pharmaceutically acceptable salts thereof may be a reduction in the required amounts of one or more compounds that are effective in treating a disease condition disclosed herein (for example, HCV) as compared to monotherapy treatment of an otherwise comparable patient population using any one component alone needed to achieve the same viral load reduction when administered as a monotherapy.
  • a disease condition disclosed herein for example, HCV
  • Compound 1A, and asunaprevir, daclatasvir and/or beclabuvir at dosages reflecting monotherapy dosage regimes for the respective component(s), and thereby provide an enhanced antiviral efficacy and/or onset of action.
  • Additional advantages of utilizing a combination of Compound 1 A and asunaprevir, daclatasvir and/or beclabuvir or pharmaceutically acceptable salts thereof may include little to no cross resistance between the respective components; different routes for elimination of the respective components; little, to no significant effects on cytochrome P 450; and/or little to no pharmacokinetic interactions between the respective components.
  • the percentages of Compound 1A, and asunaprevir, daclatasvir and/or beclabuvir or pharmaceutically acceptable salts thereof present in the composition can also vary.
  • the composition can include an amount of Compound 1A, or a pharmaceutically acceptable salt thereof, in the range of about 1 % to about 98% (weight/weight) based on the sum of the amount of Compound 1 A, or a pharmaceutically acceptable salt thereof and the amount of and asunaprevir, daclatasvir and/or beclabuvir or pharmaceutically acceptable salts thereof, in the composition.
  • Additional embodiments include, but are not limited to, an amount of Compound 1A, or a pharmaceutically acceptable salt thereof in the range of about 5% to about 80%, about 10% to about 70%, about 15% to about 60%, about 20% to about 50% and about 30% to about 40% (weight/weight) based on the sum of the amount of Compound 1A, or a pharmaceutically acceptable salt thereof, and the amount of and asunaprevir, daclatasvir and/or beclabuvir or pharmaceutically acceptable salts thereof in the composition.
  • the composition can include an amount of asunaprevir, or a pharmaceutically acceptable salt or prodrug thereof in the range of about 1 % to about 98% (weight/weight) based on the sum of the amount of asunaprevir and the other components in the composition.
  • additional embodiments include, but are not limited to, an amount of asunaprevir, or a pharmaceutically acceptable salt thereof in the range of about 5% to about 80%, about 10% to about 70%, about 15% to about 60%, about 20% to about 50% and about 30% to about 40 (weight/ weight) based on the sum of the amount of asunaprevir and the other components in the composition.
  • the composition can include an amount of daclatasvir, or a pharmaceutically acceptable salt or prodrug thereof in the range of about 1 % to about 98% (weight/weight) based on the sum of the amount of daclatasvir, or a pharmaceutically acceptable salt or prodrug thereof, and the amount of the other components in the composition.
  • Examples of additional embodiments include, but are not limited, to, an amount of daclatasvir, or a pharmaceutical y acceptable salt thereof, in the range of about 5% to about 80%, about 10% to about 70%, about 15% to about 60%, about 20% to about 50% and about 30% to about 40% (weight/weight) based on the sum of the amount of daclatasvir, or a pharmaceutically acceptable salt thereof, and the amount of the other components in the composition.
  • the composition can include an amount of beclabuvir, or a pharmaceutically acceptable salt or prodrug thereof in the range of about 1 % to about 98% (weight/weight) based on the sum of the amount of beclabuvir, or a pharmaceutically acceptable salt or prodrug thereof, and the amount of the other components in the composition.
  • Examples of additional embodiments include, but are not limited, to, an amount of beclabuvir, or a pharmaceutically acceptable salt thereof, in the range of about 5% to about 80%, about 10% to about 70%, about 15% to about 60%, about 20% to about 50% and about 30% to about 40% (weight/weight) based on the sum of the amount of beclabuvir, or a pharmaceutically acceptable salt thereof, and the amount of the other components in the composition.
  • an antiviral effective daily amount of Compound 1 A within the combination treatment of the invention would be from about 0.01 to about 700 mg/kg, or about 0.5 to about 400 mg/kg, or about 1 to about 250 mg/kg, or about 2 to about 200 mg/kg, or about 10 to about 150 mg/kg, or about 0.1 to about 50 mg/kg, or about 1 to about 20 mg/kg, or about 2 to about 10 mg/kg, or about 5 to about 8 mg/kg body weight. It may be appropriate to administer the required dose as one, two, three, four or more sub-doses at appropriate intervals throughout the day.
  • Said sub-doses may be formulated as unit dosage forms, for example, containing about 1 to about 5000 mg Compound 1A, or about 50 to about 3000 mg Compound 1A, or about 100 to about 1000 mg Compound 1 A, or about 200 to about 600 mg Compound 1A, or about 100 to about 400 mg Compound 1A, or about 7 to about 3500 mg Compound 1A, or about 70 to about 1400 mg Compound 1 A, or about 140 to about 700 mg Compound 1 A, or about 340 to about 580 mg of Compound 1 A per unit dosage form.
  • the unit dosage form is formulated for QD dosing with one of the above ranges.
  • compositions can include an amount of asunaprevir, or a pharmaceutically acceptable salt thereof in the range of about 50 mg to about 500 mg QD or BID. In other embodiments, the composition can include an amount of asunaprevir, or a pharmaceutically acceptable salt thereof in the range of about 100 mg to about 300 mg QD or BID. In still other embodiments, the composition can include an amount of asunaprevir, or a pharmaceutically acceptable salt thereof, at a dosage of about 100mg 150 or 200 mg BID or preferably QD.
  • the composition can include an amount of daclatasvir, or a pharmaceutically acceptable salt thereof in the range of about 10 mg to about 225 mg BID or QD. In other embodiments, the composition can include an amount of daclatasvir, or a pharmaceutically acceptable salt thereof, in the range of about 15 mg to about 100 mg BID or preferably QD. In still other embodiments, the composition can include an amount of daclatasvir or a pharmaceutically acceptable salt thereof in the range of about 30-60 mg QD. In yet still other embodiments, a loading dose of 30 - 100 mg such as 30, 50 or 60 mg declatasvir or pharmaceutically acceptable salt thereof can be administered for the initial 1 -3 days of therapy.
  • compositions can include an amount of beclabuvir, or a pharmaceutically acceptable salt thereof in the range of about 25 mg to about 225 mg BID or QD. In other embodiments, the composition can include an amount of beclabuvir, or a pharmaceutically acceptable salt thereof, in the range of about 50 mg to about 100 mg QD or typically BID. In still other embodiments, the composition can include an amount of beclabuvir or a pharmaceutically acceptable salt thereof in the range of about 75 mg BID or QD.
  • a potential advantage of making a combination of Compound 1 A; and asunaprevir, daclatasvir and/or beclabuvir or pharmaceutically acceptable salts thereof may be a reduction in the required amounts of one or more compounds that are effective in treating a disease condition disclosed herein (for example, HCV) as compared to monotherapy treatment of an otherwise comparable patient population using any one component alone needed to achieve the same viral load reduction when administered as a monotherapy.
  • a disease condition disclosed herein for example, HCV
  • the percentages of Compound 1A, and asunaprevir, daclatasvir and/or beclabuvir or pharmaceutically acceptable salts thereof present in the composition can also vary.
  • the composition can include an amount of Compound 1A, or a pharmaceutically acceptable salt thereof, in the range of about 1 % to about 98% (weight/weight) based on the sum of the amount of Compound 1 A, or a pharmaceutically acceptable salt thereof and the amount of and asunaprevir, daclatasvir and/or beclabuvir or pharmaceutically acceptable salts thereof, in the composition.
  • Additional embodiments include, but are not limited to, an amount of Compound 1A, or a pharmaceutically acceptable salt thereof in the range of about 5% to about 80%, about 10% to about 70%, about 15% to about 60%, about 20% to about 50% and about 30% to about 40% (weight/weight) based on the sum of the amount of Compound 1A, or a pharmaceutically acceptable salt thereof, and the amount of and asunaprevir, daclatasvir and/or beclabuvir or pharmaceutically acceptable salts thereof in the composition.
  • the composition can include an amount of asunaprevir, or a pharmaceutically acceptable salt or prodrug thereof in the range of about 1 % to about 98% (weight/weight) based on the sum of the amount of asunaprevir and the other components in the composition.
  • additional embodiments include, but are not limited to, an amount of asunaprevir, or a pharmaceutically acceptable salt thereof in the range of about 5% to about 80%, about 10% to about 70%, about 15% to about 60%, about 20% to about 50% and about 30% to about 40 (weight/ weight) based on the sum of the amount of asunaprevir and the other components in the composition.
  • the composition can include an amount of daclatasvir, or a pharmaceutically acceptable salt or prodrug thereof in the range of about 1 % to about 98% (weight/weight) based on the sum of the amount of daclatasvir, or a pharmaceutically acceptable salt or prodrug thereof, and the amount of the other components in the composition.
  • Examples of additional embodiments include, but are not limited, to, an amount of daclatasvir, or a pharmaceutical y acceptable salt thereof, in the range of about 5% to about 80%, about 10% to about 70%, about 15% to about 60%, about 20% to about 50% and about 30% to about 40% (weight/weight) based on the sum of the amount of daclatasvir, or a pharmaceutically acceptable salt thereof, and the amount of the other components in the composition.
  • the composition can include an amount of beclabuvir, or a pharmaceutically acceptable salt or prodrug thereof in the range of about 1 % to about 98% (weight/weight) based on the sum of the amount of beclabuvir, or a pharmaceutically acceptable salt or prodrug thereof, and the amount of the other components in the composition.
  • Examples of additional embodiments include, but are not limited, to, an amount of beclabuvir, or a pharmaceutically acceptable salt thereof, in the range of about 5% to about 80%, about 10% to about 70%, about 15% to about 60%, about 20% to about 50% and about 30% to about 40% (weight/weight) based on the sum of the amount of beclabuvir, or a pharmaceutically acceptable salt thereof, and the amount of the other components in the composition.
  • Some embodiments described herein relate to a method for ameliorating or treating a disease condition that, can include administering an amount of Compound I, or a pharmaceutically acceptable salt thereof, and an amount of asunaprevir, daclatasvir and/or beclabuvir or pharmaceutically acceptable salts thereof wherein the disease condition can be a hepatitis C virus infection, liver fibrosis, and/or impaired liver function.
  • Compound IA or a pharmaceutically acceptable salt thereof, asunaprevir, daclatasvir and/or beclabuvir or pharmaceutically acceptable salts thereof can be used to ameliorate and/or treat a disease condition.
  • Compounds 1 A and asunaprevir, daclatasvir and/or beclabuvir or pharmaceutically acceptable salts thereof can be present in the same dosage form such as the compositions described herein.
  • Compound 1 A, asunaprevir, daclatasvir and/or beclabuvir or pharmaceutically acceptable salts thereof can be administered as separate dosage forms.
  • Compound 1A can be administered in one tablet, asunaprevir (if present in the regime), can be administered in a second tablet, daclatasvir (if present) can be administered in a separate tablet and beclabuvir (if present) can be administeredin yet another tablet.
  • the dosage forms can be the same (e.g., as both pills) or different (e.g., two compounds can be formulated in a pill and the other compound can fee formulated as art injectable).
  • Compound IA or a pharmaceutically acceptable salt thereof; and asunaprevir, daclatasvir and/or beclabuvir or pharmaceutically acceptable salts thereof can vary.
  • the dosage forms can be administered simultaneously or sequentially.
  • the dosage form that contains Compound 1A, or pharmaceutically acceptable salt thereof can be administered before, after, in-between, concurrently or sequentially with asunaprevir (if present), delcatasvir (if present) and beclubavir (if present in the regime)
  • Compounds IA; and asunaprevir, daclatasvir and/or beclabuvir or pharmaceutically acceptable salts thereof, or pharmaceutically acceptable salts or prodrugs thereof can be administered concurrently.
  • the term "concurrently means effective concentrations of all two, three or four compounds are present in a subject.
  • Compound 1 A, and asunaprevir, daclatasvir and/or beclabuvir or pharmaceutically acceptable salts thereof can be administered in the same dosage form or separate dosage forms.
  • Compound 1A, and asunaprevir, daclatasvir and/or beclabuvir or pharmaceutically acceptable salts thereof can be administered sequentially.
  • the term "sequentially" means administering one compound for a first time period, then administering a second compound for a second time period, optionally followed by administering a third compound for a third period, in which the first, second, and if applicable third time periods do not overlap.
  • One aspect of the invention relates to a pharmaceutical composition that comprises, in a pharmaceutically acceptable vehicle, carrier or diluent: a first compound, with the formula
  • the invention in another aspect relates to a composition
  • a composition comprising, in a pharmaceutically acceptable vehicle, carrier or diluent Compound 1 , and Compound 2 and/or Compound 3, or pharmaceutically acceptable salts or prodrugs thereof, wherein the composition additionally comprises one or more therapeutic agents.
  • the one or more therapeutic agents are ribavirin and ritonavir.
  • Simeprevir, (INN), (Compound 2) has shown to be effective in inhibiting HCV replication.
  • the aforementioned compound can be obtained using methods known to those skilled in the art, including, for example, those methods disclosed in WO2013/061285, WO2008/092955 and WO2013/041655, which are hereby incorporated by reference in its entirety.
  • this invention is not limited by any particular theory, Compound 2 is believed to inhibit the HCV protease, in particular the NS3/4A protease.
  • Pharmaceutically acceptable salts and prodrugs of Compound 2 can be utilized in the compositions described herein.
  • JNJ56914845 also known as GSK2336805
  • Compound 3 has been demonstrated to be effective in inhibiting HCV replication.
  • Compound 3 is believed to have the structure:
  • Compound 3 is an NS5A inhibitor where NS5A is a protein involved in the replication of the hepatitis C virus.
  • one aspect of the invention provides pharmaceutical compositions comprising, in a pharmaceutically acceptable vehicle, carrier or diluent, Compound 1 A and simeprevir.
  • a further aspect of the invention provides pharmaceutical compositions comprising, in a pharmaceutically acceptable vehicle, carrier or diluent, Compound 1A and GSK2336805.
  • a further aspect of the invention provides pharmaceutical compositions comprising, in a pharmaceutically acceptable vehicle, carrier or diluent, Compound 1 A and simeprevir and GSK2336805.
  • an antiviral effective daily amount of Compound 1 A within the combination treatment of the invention would be from about 0.01 to about 700 mg/kg, or about 0.5 to about 400 mg/kg, or about 1 to about 250 mg/kg, or about 2 to about 200 mg/kg, or about 10 to about 150 mg/kg, or about 0.1 to about 50 mg/kg, or about 1 to about 20 mg/kg, or about 2 to about 10 mg/kg, or about 5 to about 8 mg/kg body weight. It may be appropriate to administer the required dose as one, two, three, four or more sub-doses at appropriate intervals throughout the day.
  • Said sub-doses may be formulated as unit dosage forms, for example, containing about 1 to about 5000 mg Compound 1A, or about 50 to about 3000 mg Compound 1A, or about 100 to about 1000 mg Compound 1 A, or about 200 to about 600 mg Compound 1A, or about 100 to about 400 mg Compound 1A, or about 7 to about 3500 mg Compound 1A, or about 70 to about 1400 mg Compound 1 A, or about 140 to about 700 mg Compound 1 A, or about 340 to about 580 mg of Compound 1 A per unit dosage form.
  • the unit dosage form is formulated for QD dosing with one of the above ranges.
  • compositions can include an amount of Compound 2, or a pharmaceutically acceptable salt or prodrug thereof in the range of about 50 mg to about 300 mg. In other embodiments, the composition can include an amount of Compound 2, or a pharmaceutically acceptable salt or prodrug thereof in the range of about 100 mg to about 200 mg. In still other embodiments, the composition can include an amount of Compound 2, or a pharmaceutically acceptable salt or prodrug thereof, in an amount of 100, 125 or 150 mg.
  • compositions can include an amount of Compound 3, or a pharmaceutically acceptable salt or prodrug thereof in the range of about 10 mg to about 100 mg. In other embodiments, the composition can include an amount of Compound 3, or a pharmaceutically acceptable salt or prodrug thereof, in the range of about 20 mg to about 60 mg. In still other embodiments, the composition can include an amount of Compound 3 or a pharmaceutically acceptable salt or prodrug thereof in the range of about 30-50 mg.
  • a potential advantage of making a combination of Compound 1 A and Compound 2, and/or 3, or pharmaceutically acceptable salts or prodrugs thereof may be a reduction in the required amounts of one or more compounds that are effective in treating a disease condition disclosed herein (for example, HCV) as compared to monotherapy treatment of an otherwise comparable patient population using either Compound 1 A, 2 or 3 alone.
  • the amount of Compound 1A or a pharmaceutically acceptable salt thereof in the composition can be less compared to the amount of Compound I or a pharmaceutically acceptable salt thereof needed to achieve the same viral load reduction when administered as a monotherapy.
  • the amount of Compound 2 or a pharmaceutically acceptable salt or prodrug thereof in the composition can be less compared to the amount of Compound 2, or a pharmaceutically acceptable salt or prodrug thereof, needed to achieve the same viral load reduction when administered as a monotherapy. In some embodiments, the amount of Compound 3 or a pharmaceutically acceptable salt or prodrug thereof in the composition can be less compared to the amount of Compound 3 or a pharmaceutically acceptable salt or prodrug thereof needed to achieve the same viral load reduction when administered as a monotherapy.
  • the sum of the amount of Compound 1A or a pharmaceutically acceptable salt and the amount of Compound 2, or a pharmaceutically acceptable salt or prodrug thereof and/or the amount of Compound 3, or a pharmaceutically acceptable salt or prodrug thereof is less than expected or predicted based on the additive combination of Compound 1 A or a pharmaceutically acceptable salt alone, Compound 2, or a pharmaceutically acceptable salt or prodrug thereof alone, and/or Compound 3, or a pharmaceutically acceptable salt or prodrug thereof alone for treating the disease condition such as HCV.
  • Additional advantages of utilizing a combination of Compound 1A and Compound, 2, and/or 3, or pharmaceutically acceptable salts or prodrugs thereof may include little to no cross resistance between Compound 1 A, and Compounds 2 and/or 3, or pharmaceutically acceptable salts or prodrugs thereof; different routes tor elimination of Compound 1A, and Compounds 2, and/or 3 or pharmaceutically acceptable salts or prodrugs thereof; little to no overlapping toxicities between Compound 1 A, and Compound 2, and/or 3 or pharmaceutically acceptable salts or prodrugs thereof; little, to no significant effects on cytochrome P 450; and/or little to no pharmacokinetic interactions between Compound 1A, and Compound 2, and/or 3 or pharmaceutically acceptable salts or prodrugs thereof.
  • the percentages of Compound 1 A, and Compound 2, and/or 3 or pharmaceutically acceptable salts or prodrugs thereof present in the composition can also vary.
  • the composition can include an amount of Compound 1 A, or a pharmaceutically acceptable salt thereof, in the range of about 1 % to about 98% (weight/weight) based on the sum of the amount of Compound 1 A, or a pharmaceutically acceptable salt thereof and the amount of Compounds 2 and/or 3 or pharmaceutically acceptable salts or prodrugs thereof, in the composition.
  • Additional embodiments include, but are not limited to, an amount of Compound 1 A, or a pharmaceutically acceptable salt thereof in the range of about 5% to about 80%, about 10% to about 70%, about 15% to about 60%, about 20% to about 50% and about 30% to about 40% (weight/weight) based on the sum of the amount of Compound 1A, or a pharmaceutically acceptable salt thereof, and the amount of Compounds 2 and/or 3, or pharmaceutically acceptable safe or prodrugs thereof in the composition.
  • the composition can include an amount of Compound 2, or a pharmaceutically acceptable salt or prodrug thereof in the range of about 1 % to about 98% (weight/weight) based on the sum of the amount of Compound 2, or a pharmaceutically acceptable salt or prodrug thereof and the amount of Compounds 1 and/or 3, or pharmaceutically acceptable salts or prodrugs thereof, in the composition.
  • Examples of additional embodiments include, but are not limited to, an amount of Compound 2, or a pharmaceutically acceptable salt or prodrug thereof in the range of about 5% to about 80%, about 10% to about 70%, about 15% to about 60%, about 20% to about 50% and about 30% to about 40 (weight/ weight) based on the sum of the amount of Compound 2, or a pharmaceutically acceptable salt or prodrug thereof, and the amount of Compounds 1 and/or 3, or pharmaceutically acceptable salts or prodrugs thereof in the composition.
  • the composition can include an amount of Compound 3, or a pharmaceutically acceptable salt or prodrug thereof in the range of about 1 % to about 98% (weight/weight) based on the sum of the amount of Compound 3, or a pharmaceutically acceptable salt or prodrug thereof, and the amount of Compounds 1 and/or 2, or pharmaceutically acceptable salts or prodrugs thereof, in the composition.
  • Examples of additional embodiments include, but are not limited, to, an amount of Compound 3, or a pharmaceutical y acceptable salt or prodrug thereof, in the range of about 5% to about 80%, about 10% to about 70%, about 15% to about 60%, about 20% to about 50% and about 30% to about 40% (weight/weight) based on the sum of the amount of Compound 3, or a pharmaceutically acceptable salt or prodrug thereof, and the amount of Compounds 1 and/or 2, or pharmaceutically acceptable salts or prodrugs thereof in the composition.
  • the composition can further include a pharmaceutically acceptable excipient, diluent and/or carrier, such as those described herein.
  • a pharmaceutically acceptable excipient, diluent and/or carrier such as those described herein.
  • an antiviral effective daily amount of Compound 1 A within the combination treatment of the invention would be from about 0.01 to about 700 mg/kg, or about 0.5 to about 400 mg/kg, or about 1 to about 250 mg/kg, or about 2 to about 200 mg/kg, or about 10 to about 150 mg/kg, or about 0.1 to about 50 mg/kg, or about 1 to about 20 mg/kg, or about 2 to about 10 mg/kg, or about 5 to about 8 mg/kg body weight. It may be appropriate to administer the required dose as one, two, three, four or more sub-doses at appropriate intervals throughout the day.
  • Said sub-doses may be formulated as unit dosage forms, for example, containing about 1 to about 5000 mg Compound 1A, or about 50 to about 3000 mg Compound 1A, or about 100 to about 1000 mg Compound 1 A, or about 200 to about 600 mg Compound 1A, or about 100 to about 400 mg Compound 1A, or about 7 to about 3500 mg Compound 1A, or about 70 to about 1400 mg Compound 1 A, or about 140 to about 700 mg Compound 1 A, or about 340 to about 580 mg of Compound 1 A per unit dosage form.
  • the unit dosage form is formulated for QD dosing with one of the above ranges.
  • compositions can include an amount of Compound 2, or a pharmaceutically acceptable salt or prodrug thereof in the range of about 50 mg to about 300 mg. In other embodiments, the composition can include an amount of Compound 2, or a pharmaceutically acceptable salt or prodrug thereof in the range of about 100 mg to about 200 mg. In still other embodiments, the composition can include an amount of Compound 2, or a pharmaceutically acceptable salt or prodrug thereof, in an amount of 100, 125 or 150 mg.
  • compositions can include an amount of Compound 3, or a pharmaceutically acceptable salt or prodrug thereof in the range of about 10 mg to about 100 mg. In other embodiments, the composition can include an amount of Compound 3, or a pharmaceutically acceptable salt or prodrug thereof, in the range of about 20 mg to about 60 mg. In still other embodiments, the composition can include an amount of Compound 3 or a pharmaceutically acceptable salt or prodrug thereof in the range of about 30-50 mg.
  • a potential advantage of making a combination of Compound 1 A and Compound 2, and/or 3, or pharmaceutically acceptable salts or prodrugs thereof may be a reduction in the required amounts of one or more compounds that are effective in treating a disease condition disclosed herein (for example, HCV) as compared to monotherapy treatment of an otherwise comparable patient population using either Compound 1 A, 2 or 3 alone.
  • the amount of Compound 1 ; or a pharmaceutically acceptable salt thereof in the composition can be less compared to the amount of Compound 1 A or a pharmaceutically acceptable salt thereof needed to achieve the same viral load reduction when administered as a monotherapy.
  • the amount of Compound 2 or a pharmaceutically acceptable salt or prodrug thereof in the composition can be less compared to the amount of Compound 2, or a pharmaceutically acceptable salt or prodrug thereof, needed to achieve the same viral load reduction when administered as a monotherapy. In some embodiments, the amount of Compound 3 or a pharmaceutically acceptable salt or prodrug thereof in the composition can be less compared to the amount of Compound 3 or a pharmaceutically acceptable salt or prodrug thereof needed to achieve the same viral load reduction when administered as a monotherapy.
  • the sum of the amount of Compound 1A or a pharmaceutically acceptable salt and the amount of Compound 2, or a pharmaceutically acceptable salt or prodrug thereof and/or the amount of Compound 3, or a pharmaceutically acceptable salt or prodrug thereof is less than expected or predicted based on the additive combination of Compound 1 A or a pharmaceutically acceptable salt alone, Compound 2, or a pharmaceutically acceptable salt or prodrug thereof alone, and/or Compound 3, or a pharmaceutically acceptable salt or prodrug thereof alone for treating the disease condition such as HCV.
  • the percentages of Compound 1 A, and Compound 2, and/or 3 or pharmaceutically acceptable salts or prodrugs thereof present in the composition can also vary.
  • the composition can include an amount of Compound 1 A, or a pharmaceutically acceptable salt thereof, in the range of about 1 % to about 98% (weight/weight) based on the sum of the amount of Compound 1 A, or a pharmaceutically acceptable salt thereof and the amount of Compounds 2 and/or 3 or pharmaceutically acceptable salts or prodrugs thereof, in the composition.
  • Additional embodiments include, but are not limited to, an amount of Compound 1 A, or a pharmaceutically acceptable salt thereof in the range of about 5% to about 80%, about 10% to about 70%, about 15% to about 60%, about 20% to about 50% and about 30% to about 40% (weight/weight) based on the sum of the amount of Compound 1A, or a pharmaceutically acceptable salt or prodrug thereof, and the amount of Compounds 2 and/or 3, or pharmaceutically acceptable safe or prodrugs thereof in the composition.
  • the composition can include an amount of Compound 2, or a pharmaceutically acceptable salt or prodrug thereof in the range of about 1 % to about 98% (weight/weight) based on the sum of the amount of Compound 2, or a pharmaceutically acceptable salt or prodrug thereof and the amount of Compounds 1 A and/or 3, or pharmaceutically acceptable salts or prodrugs thereof, in the composition.
  • Examples of additional embodiments include, but are not limited to, an amount of Compound 2, or a pharmaceutically acceptable salt or prodrug thereof in the range of about 5% to about 80%, about 10% to about 70%, about 15% to about 60%, about 20% to about 50% and about 30% to about 40 (weight/ weight) based on the sum of the amount of Compound 2, or a pharmaceutically acceptable salt or prodrug thereof, and the amount of Compounds 1 A and/or 3, or pharmaceutically acceptable salts or prodrugs thereof in the composition.
  • the composition can include an amount of Compound 3, or a pharmaceutically acceptable salt or prodrug thereof in the range of about 1 % to about 98% (weight/weight) based on the sum of the amount of Compound 3, or a pharmaceutically acceptable salt or prodrug thereof, and the amount of Compounds 1 A and/or 2, or pharmaceutically acceptable salts or prodrugs thereof, in the composition.
  • Examples of additional embodiments include, but are not limited, to, an amount of Compound 3, or a pharmaceutically acceptable salt or prodrug thereof, in the range of about 5% to about 80%, about 10% to about 70%, about 15% to about 60%, about 20% to about 50% and about 30% to about 40% (weight/weight) based on the sum of the amount of Compound 3, or a pharmaceutically acceptable salt or prodrug thereof, and the amount of Compounds 1 A and/or 2, or pharmaceutically acceptable salts or prodrugs thereof in the composition in an appropriate container, and labelled for treatment of an indicated condition.
  • Some embodiments described herein relate to a method for ameliorating or treating a disease condition that, can include administering an amount of Compound 1 A, or a pharmaceutically acceptable salt thereof, and an amount of Compound 2, or a pharmaceutically acceptable salt or prodrug thereof and/or an amount of Compound 3, or a pharmaceutically acceptable salt or prodrug thereof wherein the disease condition can be a hepatitis C virus infection, liver fibrosis, and/or impaired liver function.
  • compositions described herein can be present in the same dosage form such as the compositions described herein.
  • Compound 1A, and 2 and/or 3, or pharmaceutically acceptable salts or prodrugs thereof can be administered as separate dosage forms.
  • Compound 1 A can be administered in one tablet
  • Compound 2, or a pharmaceutically acceptable salt or prodrug thereof can be administered In a second tablet
  • Compound 3, or a pharmaceutically acceptable salt or prodrug thereof can be administered in a third tablet.
  • the dosage forms can be the same (e.g., as both pills) or different (e.g., two compounds can be formulated in a pill and the other compound can fee formulated as art injectable).
  • compositions 1 A, and 2 and/or 3, or pharmaceutically acceptable salts or prodrugs thereof can vary.
  • the dosage forms can be administered simultaneously or sequentially.
  • the dosage form that contains Compound 1A, or pharmaceutically acceptable salt thereof can be administered before, after, in-between, concurrently or sequentially with Compounds 2 and/or 3, or pharmaceutically acceptable salts or prodrugs thereof.
  • the dosage form that contains Compound 2, or a pharmaceutically acceptable salt or prodrug thereof can be administered before, after, in-between, concurrently or sequentially with Compound 1 A, or Compounds IA and 3, or pharmaceutically acceptable salts or prodrugs thereof.
  • the dosage form that contains Compound 3, or a pharmaceutically acceptable salt or prodrug thereof can be administered before, after, inbetween, concurrently or sequentially with Compound 1 A and 2, or pharmaceutically acceptable salts or prodrugs thereof.
  • Compounds 1 A; and Compound 2 and/or 3, or pharmaceutically acceptable salts or prodrugs thereof can be administered concurrently.
  • the term “concurrently means effective concentrations of all three compounds are present in a subject.
  • Compound 1 A, and Compound 2 and/or 3, or pharmaceutically acceptable salts or prodrugs thereof can be administered in the same dosage form or separate dosage forms.
  • the term "sequentially” means administering one compound for a first time period, then administering a second compound for a second time period, optionally followed by administering a third compound for a third period, in which the first, second, and if applicable third time periods do not overlap.
  • Additional therapeutic agents can also be administered to the subject having the disease condition.
  • a non-limiting list of additional therapeutic agents includes those previously described herein.
  • the additional agent(s) can be administered in the same dosage form as Compound 1 A or a pharmaceutically acceptable salt and/or Compound 2, or a pharmaceutically acceptable salt or prodrug thereof and/or Compound 3 or a pharmaceutically acceptable salt or prodrug thereof.
  • the additional therapeutic agent(s) can be included in a composition that includes Compound 1A, or a pharmaceutically acceptable salt, without Compounds 2 and/or 3, or pharmaceutically acceptable salts or prodrugs thereof; or a composition that includes Compound 2 or a pharmaceutically acceptable salt or prodrug thereof, without Compounds 1 A or without Compounds 1A and 3, or pharmaceutically acceptable salts or prodrugs thereof; or a composition that includes Compound 3, or a pharmaceutically acceptable salt or prodrug thereof, without Compounds 1 A or without Compounds 1 A and 2, or pharmaceutically acceptable salts or prodrugs thereof.
  • One aspect of the invention relates to a pharmaceutical composition that comprises, in a pharmaceutically acceptable vehicle, carrier or diluent: a first compound, with the formula
  • compositions or HCV treatment regimes comprising i) Compound 1 A, paritaprevir, ombitasvir
  • the invention relates to the use of such compositions for ameliorating or treating a disease condition in a patient population, and/or for the preparation of a medicament for ameliorating or treating such a disease condition.
  • the disease condition can be selected from a hepatitis C virus infection, liver fibrosis, and impaired liver function.
  • the invention relates to the use of a composition comprising Compound 1 A, and a Compound 2a and/or a Compound 3a, or pharmaceutically acceptable salts or prodrugs thereof, for ameliorating or treating hepatitis C virus infection, liver fibrosis, and impaired liver function.
  • the invention in another aspect relates to a method for ameliorating or treating a disease condition in a patient population that comprises administering a therapeutically effective amount of a first compound, or a pharmaceutically acceptable salt or prodrug thereof wherein the first compound is Compound 1 A; and a therapeutically effective amount of a second compound, or a pharmaceutically acceptable salt or prodrug thereof, wherein the second compound is a Compound 2a; and/or a therapeutically effective amount of a third compound, or a pharmaceutically acceptable salt or prodrug thereof wherein the third compound is a Compound 3a; to a subject suffering from the disease conditions.
  • the disease condition can be selected from & hepatitis C virus infection, liver fibrosis, and impaired liver function.
  • the invention relates to the use of Compound 1A or a pharmaceutically acceptable salt or prodrug thereof for ameliorating or treating a disease condition in a patient population and/or for the preparation of a medicament for ameliorating or treating such a disease condition, wherein Compound 1A or a pharmaceutically acceptable salt thereof is manufactured for use in combination with a Compound 2a or a pharmaceutically acceptable salt or prodrug thereof; and wherein Compound 1A and a Compound 2a or pharmaceutically acceptable salts or prodrugs thereof are manufactured for use in combination with a Compound 3a or a pharmaceutical acceptable salt or prodrug thereof wherein the disease condition is selected from hepatitis C virus infection, liver fibrosis, and impaired liver function,
  • the method or use for ameliorating or treating a disease condition in a patient population comprises administering, one or more additional therapeutic agents.
  • the one or more additional therapeutic agents are ribavirin and/or ritonavir.
  • Paritaprevir, (INN) one of the possibilities for Compound 2a has shown to be effective in inhibiting HCV replication.
  • the aforementioned compound can be obtained using methods known to those skilled in the art, including, for example, those methods disclosed in WO201 1/1 12558, and WO2013/106631 , which are hereby incorporated by reference in its entirety.
  • this invention is not limited by any particular theory, Compound 2a are believed to inhibit the HCV protease, in particular the NS3/4A protease.
  • compositions described herein Paritaprevir is preferably co-dosed with a pharmacokinetic booster of ritonavir, and each recital of Compound 2a herein shall be understood as including an optional boosting dose, for example 25-100 mg, such as 50 mg of ritonavir.
  • Ombitasvir one of the possibilities for Compound 3a has been demonstrated to be effective in inhibiting HCV replication.
  • Compound 3a can be obtained using methods known to those skilled in the art, such as those methods described in WO2012/051361 , which is hereby incorporated by reference in its entirety.
  • Each Compound 3a is an NS5A inhibitor where NS5A is a protein involved in the replication of the hepatitis C virus.
  • an antiviral effective daily amount of Compound 1 A within the combination treatment of the invention would be from about 0.01 to about 700 mg/kg, or about 0.5 to about 400 mg/kg, or about 1 to about 250 mg/kg, or about 2 to about 200 mg/kg, or about 10 to about 150 mg/kg, or about 0.1 to about 50 mg/kg, or about 1 to about 20 mg/kg, or about 2 to about 10 mg/kg, or about 5 to about 8 mg/kg body weight. It may be appropriate to administer the required dose as one, two, three, four or more sub-doses at appropriate intervals throughout the day.
  • Said sub-doses may be formulated as unit dosage forms, for example, containing about 1 to about 5000 mg Compound 1A, or about 50 to about 3000 mg Compound 1A, or about 100 to about 1000 mg Compound 1 A, or about 200 to about 600 mg Compound 1A, or about 100 to about 400 mg Compound 1A, or about 7 to about 3500 mg Compound 1A, or about 70 to about 1400 mg Compound 1 A, or about 140 to about 700 mg Compound 1 A, or about 340 to about 580 mg of Compound 1 A per unit dosage form.
  • the unit dosage form is formulated for QD dosing with one of the above ranges.
  • compositions can include an amount of a Compound 2a, or a pharmaceutically acceptable salt or prodrug thereof in the range of about 50 mg to about 300 mg.
  • the composition can include an amount of a Compound 2a, or a pharmaceutically acceptable salt or prodrug thereof in the range of about 75 mg to about 200 mg.
  • the composition can include an amount of a Compound 2a, or a pharmaceutically acceptable salt or prodrug thereof, in the range of about 100-150 mg.
  • the each dosage unit can include a booster of ritonavir, for example 25-100 mg, such as 50 mg.
  • compositions can include an amount of a Compound 3a, or a pharmaceutically acceptable salt or prodrug thereof in the range of about 5 mg to about 100 mg. In other embodiments, the composition can include an amount of a Compound 3a, or a pharmaceutically acceptable salt or prodrug thereof, in the range of about 12.5 mg to about 75 mg. In still other embodiments, the composition can include an amount of a Compound 3a or a pharmaceutically acceptable salt or prodrug thereof in the range of about 25 mg.
  • a potential advantage of making a combination of Compound 1A and a Compound 2a, and/or a compound 3a, or pharmaceutically acceptable salts or prodrugs thereof may be a reduction in the required amounts of one or more compounds that are effective in treating a disease condition disclosed herein (for example, HCV) as compared to monotherapy treatment of an otherwise comparable patient population using either Compound 1A, 2a or 3a alone.
  • the amount of Compound 1A ; or a pharmaceutically acceptable salt thereof in the composition can be less compared to the amount of Compound 1A or a pharmaceutically acceptable salt thereof needed to achieve the same viral load reduction when administered as a monotherapy.
  • the amount of a Compound 2a or a pharmaceutically acceptable salt or prodrug thereof in the composition can be less compared to the amount of a Compound 2a, or a pharmaceutically acceptable salt or prodrug thereof, needed to achieve the same viral load reduction when administered as a monotherapy. In some embodiments, the amount of a Compound 3a or a pharmaceutically acceptable salt or prodrug thereof in the composition can be less compared to the amount of a Compound 3a or a pharmaceutically acceptable salt or prodrug thereof needed to achieve the same viral load reduction when administered as a monotherapy.
  • the sum of the amount of Compound 1A or a pharmaceutically acceptable salt and the amount of a Compound 2a, or a pharmaceutically acceptable salt or prodrug thereof and/or the amount of a Compound 3a, or a pharmaceutically acceptable salt or prodrug thereof is less than expected or predicted based on the additive combination of Compound 1 A or a pharmaceutically acceptable salt or prodrug thereof, alone, a Compound 2a, or a pharmaceutically acceptable salt or prodrug thereof alone, and/or a Compound 3a, or a pharmaceutically acceptable salt or prodrug thereof alone for treating the disease condition such as HCV.
  • the percentages of Compound 1A, and Compound 2a, and/or 3a or pharmaceutically acceptable salts or prodrugs thereof present in the composition can also vary.
  • the composition can include an amount of Compound 1 A, or a pharmaceutically acceptable salt thereof, in the range of about 1 % to about 98% (weight/weight) based on the sum of the amount of Compound 1 A, or a pharmaceutically acceptable salt thereof and the amount of Compounds 2a and/or 3a or pharmaceutically acceptable salts or prodrugs thereof, in the composition.
  • Additional embodiments include, but are not limited to, an amount of Compound 1A, or a pharmaceutically acceptable salt thereof in the range of about 5% to about 80%, about 10% to about 70%, about 15% to about 60%, about 20% to about 50% and about 30% to about 40% (weight/weight) based on the sum of the amount of Compound 1A, or a pharmaceutically acceptable salt or prodrug thereof, and the amount of Compounds 2a and/or 3a, or pharmaceutically acceptable safe or prodrugs thereof in the composition.
  • the composition can include an amount of a Compound 2a, or a pharmaceutically acceptable salt or prodrug thereof in the range of about 1 % to about 98% (weight/weight) based on the sum of the amount of Compound 2a, or a pharmaceutically acceptable salt or prodrug thereof and the amount of Compounds 1 A and/or 3a, or pharmaceutically acceptable salts or prodrugs thereof, in the composition.
  • Examples of additional embodiments include, but are not limited to, an amount of a Compound 2a, or a pharmaceutically acceptable salt or prodrug thereof in the range of about 5% to about 80%, about 10% to about 70%, about 15% to about 60%, about 20% to about 50% and about 30% to about 40 (weight/ weight) based on the sum of the amount of Compound 2a, or a pharmaceutically acceptable salt or prodrug thereof, and the amount of Compounds 1 A and/or 3a, or pharmaceutically acceptable salts or prodrugs thereof in the composition.
  • the composition can include an amount of a Compound 3a, or a pharmaceutically acceptable salt or prodrug thereof in the range of about 1 % to about 98% (weight/weight) based on the sum of the amount of Compound 3a, or a pharmaceutically acceptable salt or prodrug thereof, and the amount of Compounds 1 A and/or 2a, or pharmaceutically acceptable salts or prodrugs thereof, in the composition.
  • Examples of additional embodiments include, but are not limited, to, an amount of a Compound 3a, or a pharmaceutically acceptable salt or prodrug thereof, in the range of about 5% to about 80%, about 10% to about 70%, about 15% to about 60%, about 20% to about 50% and about 30% to about 40% (weight/weight) based on the sum of the amount of Compound 3a, or a pharmaceutically acceptable salt or prodrug thereof, and the amount of Compounds 1A and/or 2a, or pharmaceutically acceptable salts or prodrugs thereof in the composition.
  • compositions described herein can be present in the same dosage form such as the compositions described herein.
  • Compound 1 A, and 2a and/or 3a, or pharmaceutically acceptable salts or prodrugs thereof can be administered as separate dosage forms.
  • Compound 1 A can be administered in one tablet
  • a Compound 2a, or a pharmaceutically acceptable salt or prodrug thereof can be administered In a second tablet
  • a Compound 3a, or a pharmaceutically acceptable salt or prodrug thereof can be administered in a third tablet.
  • the dosage forms can be the same (e.g., both or all as pills) or different (e.g., two compounds can be formulated in a pill and the other compound can be formulated as art injectable).
  • Administration of Compound 1 A or a pharmaceutically acceptable salt thereof; and a Compound 2a, or a pharmaceutically acceptable salt or prodrug thereof, and/or a Compound 3a, or a pharmaceutically acceptable salt or prodrug thereof can vary.
  • the dosage forms can be administered simultaneously or sequentially.
  • the dosage form that contains Compound 1A, or pharmaceutically acceptable salt thereof can be administered before, after, in-between, concurrently or sequentially with Compounds 2a and/or 3a, or pharmaceutically acceptable salts or prodrugs thereof.
  • the dosage form that contains Compound 2a, or a pharmaceutically acceptable salt or prodrug thereof can be administered before, after, in-between, concurrently or sequentially with Compound 1A, or Compounds 1 A and 3a, or pharmaceutically acceptable salts or prodrugs thereof.
  • the dosage form that contains Compound 3a, or a pharmaceutically acceptable salt or prodrug thereof can be administered before, after, inbetween, concurrently or sequentially with Compound 1A or Compound 1 A and 2a, or pharmaceutically acceptable salts or prodrugs thereof.
  • Compounds 1 A; and Compound 2a and/or 3a, or pharmaceutically acceptable salts or prodrugs thereof can be administered concurrently.
  • the term “concurrently means effective concentrations of all three compounds are present in a subject.
  • Compound 1 A, and Compound 2a and/or 3a, or pharmaceutically acceptable salts or prodrugs thereof can be administered in the same dosage form or separate dosage forms.
  • Compound 1A, and Compound 2a, and/or Compound 3a, or pharmaceutically acceptable salts or prodrugs hereof can be administered sequentially.
  • the term “sequentially” means administering one compound for a first time period, then administering a second compound for a second time period, optionally followed by administering a third compound for a third period, in which the first, second, and if applicable third time periods do not overlap.
  • One aspect of the invention relates to a pharmaceutical composition that comprises, in a pharmaceutically acceptable vehicle, carrier or diluent: a first compound, with the formula Compound 1A or a pharmaceutically acceptable salt thereof, and a second compound, or a pharmaceutically acceptable salt or prodrug thereof wherein the second compound is alisporavir ("Compound 2b");
  • the invention relates to a composition comprising, in a pharmaceutically acceptable vehicle, carrier or diluent Compound 1 A, and Compound 2b, or pharmaceutically acceptable salts or prodrugs thereof, wherein the composition additionally comprises one or more therapeutic agents.
  • the one or more therapeutic agents are ribavirin and ritonavir.
  • the invention relates to the use of such compositions for ameliorating or treating a disease condition in a patient population, and/or for the preparation of a medicament for ameliorating or treating such a disease condition.
  • the disease condition can be selected from a hepatitis C virus infection, liver fibrosis, and impaired liver function.
  • the invention relates to the use of a composition comprising Compound 1A, and Compound 2b, or pharmaceutically acceptable salts or prodrugs thereof, for ameliorating or treating hepatitis C virus infection, liver fibrosis, and impaired liver function.
  • the invention in another aspect relates to a method for ameliorating or treating a disease condition in a patient population that comprises administering a therapeutically effective amount of a first compound, or a pharmaceutically acceptable salt or prodrug thereof wherein the first compound is Compound 1 A; and a therapeutically effective amount of a second compound, or a pharmaceutically acceptable salt or prodrug thereof, wherein the second compound is Compound 2b; to a subject suffering from the disease conditions.
  • the disease condition can be selected from & hepatitis C virus infection, liver fibrosis, and impaired liver function.
  • the invention relates to the use of Compound 1A or a pharmaceutically acceptable salt or prodrug thereof for ameliorating or treating a disease condition in a patient population and/or for the preparation of a medicament for ameliorating or treating such a disease condition, wherein Compound 1 A or a pharmaceutically acceptable salt thereof is manufactured for use in combination with Compound 2b or a pharmaceutically acceptable salt or prodrug thereof; wherein the disease condition is selected from hepatitis C virus infection, liver fibrosis, and impaired liver function,
  • the method or use for ameliorating or treating a disease condition in a patient population comprises administering, one or more additional therapeutic agents.
  • the one or more additional therapeutic agents are ribavirin and/or ritonavir,
  • Alisporavir (INN) (Compound 2b) is a host cyclophilin A inhibitor (otherwise known as peptidyl prolylc/sfransisomerase inhibitors) which has been demonstrated to be effective in HCV treatment in phase II clinical trials. Alisporavir belongs to the cyclosporin class of macromolecules.
  • Compound 2b can be obtained using methods known to those skilled in the art, such as those methods described in WO2013/167703 and WO2006/038088 which are hereby incorporated by reference in its entirety.
  • an antiviral effective daily amount of Compound 1A within the combination treatment of the invention would be from about 0.01 to about 700 mg/kg, or about 0.5 to about 400 mg/kg, or about 1 to about 250 mg/kg, or about 2 to about 200 mg/kg, or about 10 to about 150 mg/kg, or about 0.1 to about 50 mg/kg, or about 1 to about 20 mg/kg, or about 2 to about 10 mg/kg, or about 5 to about 8 mg/kg body weight.
  • Said sub-doses may be formulated as unit dosage forms, for example, containing about 1 to about 5000 mg Compound 1 , or about 50 to about 3000 mg Compound 1 , or about 100 to about 1000 mg Compound 1A, or about 200 to about 600 mg Compound 1A, or about 100 to about 400 mg Compound 1 A, or about 7 to about 3500 mg Compound 1 A, or about 70 to about 1400 mg Compound 1 A, or about 140 to about 700 mg Compound 1A, or about 340 to about 580 mg of Compound 1 A per unit dosage form.
  • the unit dosage form is formulated for QD dosing with one of the above ranges.
  • compositions can include an amount of Compound 2b, or a pharmaceutically acceptable salt or prodrug thereof in the range of about 100-800 mg BID or QD In other embodiments, the composition can include an amount of Compound 2b, or a pharmaceutically acceptable salt or prodrug thereof, in the range of about 200-400 mg. In still other embodiments, the composition can include an amount of Compound 2b or a pharmaceutically acceptable salt or prodrug thereof in the range of about 200, 250, 300, 350 or 400 mg.
  • a potential advantage of making a combination of Compound 1A and Compound 2b, or pharmaceutically acceptable salts or prodrugs thereof may be a reduction in the required amounts of one or more compounds that are effective in treating a disease condition disclosed herein (for example, HCV) as compared to monotherapy treatment of an otherwise comparable patient population using either Compound 1A, or 2b alone.
  • the amount of Compound 1A ; or a pharmaceutically acceptable salt thereof in the composition can be less compared to the amount of Compound 1 A or a pharmaceutically acceptable salt thereof needed to achieve the same viral load reduction when administered as a monotherapy.
  • the amount of Compound 2b or a pharmaceutically acceptable salt or prodrug thereof in the composition can be less compared to the amount of Compound 2b, or a pharmaceutically acceptable salt or prodrug thereof, needed to achieve the same viral load reduction when administered as a monotherapy.
  • the sum of the amount of Compound 1A or a pharmaceutically acceptable salt and the amount of Compound 2b, or a pharmaceutically acceptable salt or prodrug thereof is less than expected or predicted based on the additive combination of Compound 1A or a pharmaceutically acceptable salt or prodrug thereof, alone, or Compound 2b, or a pharmaceutically acceptable salt or prodrug thereof alone, for treating the disease condition such as HCV.
  • Additional advantages of utilizing a combination of Compound 1A and Compound, 2b, or pharmaceutically acceptable salts or prodrugs thereof may include little to no cross resistance between Compound 1 A, and Compound 2b, or pharmaceutically acceptable salts or prodrugs thereof; different routes tor elimination of Compound 1 , and Compound 2b, or pharmaceutically acceptable salts or prodrugs thereof; little to no overlapping toxicities between Compound 1 A, and Compound 2b, or pharmaceutically acceptable salts or prodrugs thereof; little, to no significant effects on cytochrome P 450; and/or little to no pharmacokinetic interactions between Compound 1 A, and Compound 2b, or pharmaceutically acceptable salts or prodrugs thereof.
  • the percentages of Compound 1 A, and Compound 2b, or pharmaceutically acceptable salts or prodrugs thereof present in the composition can also vary.
  • the composition can include an amount of Compound 1 A, or a pharmaceutically acceptable salt thereof, in the range of about 1 % to about 98% (weight/weight) based on the sum of the amount of Compound 1 A, or a pharmaceutically acceptable salt thereof and the amount of Compounds 2b or pharmaceutically acceptable salts or prodrugs thereof, in the composition.
  • Additional embodiments include, but are not limited to, an amount of Compound 1 A, or a pharmaceutically acceptable salt thereof in the range of about 5% to about 80%, about 10% to about 70%, about 15% to about 60%, about 20% to about 50% and about 30% to about 40% (weight/weight) based on the sum of the amount of Compound 1 A, or a pharmaceutically acceptable salt or prodrug thereof, and the amount of Compounds 2b, or pharmaceutically acceptable safe or prodrugs thereof in the composition.
  • the composition can include an amount of Compound 2b, or a pharmaceutically acceptable salt or prodrug thereof in the range of about 1 % to about 98% (weight/weight) based on the sum of the amount of Compound 2b, or a pharmaceutically acceptable salt or prodrug thereof and the amount of Compounds 1 A, or pharmaceutically acceptable salts or prodrugs thereof, in the composition.
  • Examples of additional embodiments include, but are not limited to, an amount of Compound 2b, or a pharmaceutically acceptable salt or prodrug thereof in the range of about 5% to about 80%, about 10% to about 70%, about 15% to about 60%, about 20% to about 50% and about 30% to about 40 (weight/ weight) based on the sum of the amount of Compound 2b, or a pharmaceutically acceptable salt or prodrug thereof, and the amount of Compounds 1 A, or pharmaceutically acceptable salts thereof in the composition.
  • Some embodiments described herein relate to a method for ameliorating or treating a disease condition that, can include administering an amount of Compound 1 A, or a pharmaceutically acceptable salt thereof, and an amount of Compound 2b, or a pharmaceutically acceptable salt or prodrug thereof wherein the disease condition can be a hepatitis C virus infection, liver fibrosis, and/or impaired liver function.
  • compositions described herein can be present in the same dosage form such as the compositions described herein.
  • Compound 1 A, and 2b, or pharmaceutically acceptable salts or prodrugs thereof can be administered as separate dosage forms.
  • Compound 1 A, or a pharmaceutically acceptable salt thereof can be administered in one tablet
  • Compound 2b, or a pharmaceutically acceptable salt or prodrug thereof can be administered In a second tablet.
  • the dosage forms can be the same(e.g., as both pills) or different (e.g. one can be formulated in a pill and the other compound can fee formulated as an injectable).
  • Administration of Compound 1 A or a pharmaceutically acceptable salt thereof; and Compound 2b, or a pharmaceutically acceptable salt or prodrug thereof can vary.
  • the dosage forms can be administered simultaneously or sequentially.
  • the dosage form that contains Compound 1A, or pharmaceutically acceptable salt thereof can be administered before, after, in-between, concurrently or sequentially with Compounds 2b, or pharmaceutically acceptable salts or prodrugs thereof.
  • the dosage form that contains Compound 2b, or a pharmaceutically acceptable salt or prodrug thereof can be administered before, after, in-between, concurrently or sequentially with Compound 1 A, or pharmaceutically acceptable salts or prodrugs thereof.
  • Compounds 1 A; and Compound 2b, or pharmaceutically acceptable salts or prodrugs thereof can be administered concurrently.
  • the term “concurrently means effective concentrations of both compounds are present in a subject.
  • Compound 1A, and Compound 2b, or pharmaceutically acceptable salts or prodrugs thereof can be administered in the same dosage form or separate dosage forms.
  • Compound 1 A, and Compound 2b, or pharmaceutically acceptable salts or prodrugs hereof can be administered sequentially.
  • the term “sequentially” means administering one compound for a first time period, then administering a second compound for a second time period, in which the first and second time periods do not overlap.
  • One aspect of the invention relates to a pharmaceutical composition that comprises, in a pharmaceutically acceptable vehicle, carrier or diluent: a first compound, with the formula
  • the invention in another aspect relates to a composition
  • a composition comprising, in a pharmaceutically acceptable vehicle, carrier or diluent Compound 1 A, and Compound 2c, or pharmaceutically acceptable salts or prodrugs thereof, wherein the composition additionally comprises one or more therapeutic agents.
  • the one or more therapeutic agents are ribavirin and ritonavir.
  • the invention relates to the use of such compositions for ameliorating or treating a disease condition in a patient population, and/or for the preparation of a medicament for ameliorating or treating such a disease condition.
  • the disease condition can be selected from a hepatitis C virus infection, liver fibrosis, and impaired liver function.
  • the invention relates to the use of a composition comprising Compound 1A, and Compound 2c, or pharmaceutically acceptable salts or prodrugs thereof, for ameliorating or treating hepatitis C virus infection, liver fibrosis, and impaired liver function.
  • the invention in another aspect relates to a method for ameliorating or treating a disease condition in a patient population that comprises administering a therapeutically effective amount of a first compound, or a pharmaceutically acceptable salt or prodrug thereof wherein the first compound is Compound 1 A; and a therapeutically effective amount of a second compound, or a pharmaceutically acceptable salt or prodrug thereof, wherein the second compound is Compound 2c; to a subject suffering from the disease conditions.
  • the disease condition can be selected from & hepatitis C virus infection, liver fibrosis, and impaired liver function.
  • the invention relates to the use of Compound 1A or a pharmaceutically acceptable salt or prodrug thereof for ameliorating or treating a disease condition in a patient population and/or for the preparation of a medicament for ameliorating or treating such a disease condition, wherein Compound 1A or a pharmaceutically acceptable salt thereof is manufactured for use in combination with Compound 2c or a pharmaceutically acceptable salt or prodrug thereof; wherein the disease condition is selected from hepatitis C virus infection, liver fibrosis, and impaired liver function,
  • the method or use for ameliorating or treating a disease condition in a patient population comprises administering, one or more additional therapeutic agents.
  • the one or more additional therapeutic agents are ribavirin and/or ritonavir.
  • EDP-239 (Compound 2c) has been demonstrated to be effective in inhibiting HCV replication in phase II clinical trials. The formula of Compound 2c is believed to be
  • Compound 2c can be obtained using methods known to those skilled in the art, such as those methods described in WO2013/059281 and WO2010/099527, which are hereby incorporated by reference in its entirety. Although this invention is not limited by any particular theory, it is believed that Compound 2c is an NS5A inhibitor where NS5A is a protein involved in the replication of the hepatitis C virus.
  • the composition can further include a pharmaceutically acceptable exipient, diluent and/or carrier, such as those described herein.
  • an antiviral effective daily amount of Compound 1 A within the combination treatment of the invention would be from about 0.01 to about 700 mg/kg, or about 0.5 to about 400 mg/kg, or about 1 to about 250 mg/kg, or about 2 to about 200 mg/kg, or about 10 to about 150 mg/kg, or about 0.1 to about 50 mg/kg, or about 1 to about 20 mg/kg, or about 2 to about 10 mg/kg, or about 5 to about 8 mg/kg body weight. It may be appropriate to administer the required dose as one, two, three, four or more sub-doses at appropriate intervals throughout the day.
  • Said sub-doses may be formulated as unit dosage forms, for example, containing about 1 to about 5000 mg Compound 1A, or about 50 to about 3000 mg Compound 1A, or about 100 to about 1000 mg Compound 1 A, or about 200 to about 600 mg Compound 1 A, or about 100 to about 400 mg Compound 1 A, or about 7 to about 3500 mg Compound 1 A, or about 70 to about 1400 mg Compound 1 A, or about 140 to about 700 mg Compound 1 A, or about 340 to about 580 mg of Compound 1 A per unit dosage form.
  • the unit dosage form is formulated for QD dosing with one of the above ranges.
  • compositions can include an amount of Compound 2c, or a pharmaceutically acceptable salt or prodrug thereof in the range of about 25 mg to about 225 mg. In other embodiments, the composition can include an amount of Compound 2c, or a pharmaceutically acceptable salt or prodrug thereof, in the range of about 50 mg to about 150 mg. In still other embodiments, the composition can include an amount of Compound 2c or a pharmaceutically acceptable salt or prodrug thereof in the range of about 50- 100 mg. In yet still other embodiments, the a loading dose of 100-200 mg, such as 150 mg can be administered for the initial 1 -3 days of therapy.
  • a potential advantage of making a combination of Compound 1 A and Compound 2c, or pharmaceutically acceptable salts or prodrugs thereof may be a reduction in the required amounts of one or more compounds that are effective in treating a disease condition disclosed herein (for example, HCV) as compared to monotherapy treatment of an otherwise comparable patient population using either Compound 1 A, or 2c alone.
  • the amount of Compound 1A ; or a pharmaceutically acceptable salt thereof in the composition can be less compared to the amount of Compound 1 A or a pharmaceutically acceptable salt thereof needed to achieve the same viral load reduction when administered as a monotherapy.
  • the amount of Compound 2c or a pharmaceutically acceptable salt or prodrug thereof in the composition can be less compared to the amount of Compound 2c, or a pharmaceutically acceptable salt or prodrug thereof, needed to achieve the same viral load reduction when administered as a monotherapy.
  • the sum of the amount of Compound 1A or a pharmaceutically acceptable salt and the amount of Compound 2c, or a pharmaceutically acceptable salt or prodrug thereof is less than expected or predicted based on the additive combination of Compound 1A or a pharmaceutically acceptable salt thereof, alone, or Compound 2c, or a pharmaceutically acceptable salt or prodrug thereof alone, for treating the disease condition such as HCV.
  • the percentages of Compound 1 A, and Compound 2c, or pharmaceutically acceptable salts or prodrugs thereof present in the composition can also vary.
  • the composition can include an amount of Compound 1 A, or a pharmaceutically acceptable salt thereof, in the range of about 1 % to about 98% (weight/weight) based on the sum of the amount of Compound 1A, or a pharmaceutically acceptable salt thereof and the amount of Compound 2c or pharmaceutically acceptable salts or prodrugs thereof, in the composition.
  • Additional embodiments include, but are not limited to, an amount of Compound 1 A, or a pharmaceutically acceptable salt thereof in the range of about 5% to about 80%, about 10% to about 70%, about 15% to about 60%, about 20% to about 50% and about 30% to about 40% (weight/weight) based on the sum of the amount of Compound 1 A, or a pharmaceutically acceptable salt or prodrug thereof, and the amount of Compound 2c, or pharmaceutically acceptable safe or prodrugs thereof in the composition.
  • the composition can include an amount of Compound 2c, or a pharmaceutically acceptable salt or prodrug thereof in the range of about 1 % to about 98% (weight/weight) based on the sum of the amount of Compound 2c, or a pharmaceutically acceptable salt or prodrug thereof and the amount of Compounds 1A, or pharmaceutically acceptable salts or prodrugs thereof, in the composition.
  • Examples of additional embodiments include, but are not limited to, an amount of Compound 2c, or a pharmaceutically acceptable salt or prodrug thereof in the range of about 5% to about 80%, about 10% to about 70%, about 15% to about 60%, about 20% to about 50% and about 30% to about 40 (weight/ weight) based on the sum of the amount of Compound 2c, or a pharmaceutically acceptable salt or prodrug thereof, and the amount of Compounds 1 , or pharmaceutically acceptable salts or prodrugs thereof in the composition.
  • Some embodiments described herein relate to a method for ameliorating or treating a disease condition that, can include administering an amount of Compound 1 A, or a pharmaceutically acceptable salt thereof, and an amount of Compound 2c, or a pharmaceutically acceptable salt or prodrug thereof wherein the disease condition can be a hepatitis C virus infection, liver fibrosis, and/or impaired liver function.
  • compositions described herein can be present in the same dosage form such as the compositions described herein.
  • Compound 1A, and 2c, or pharmaceutically acceptable salts or prodrugs thereof can be administered as separate dosage forms.
  • Compound 1 A, or a pharmaceutically acceptable salt thereof can be administered in one tablet
  • Compound 2c, or a pharmaceutically acceptable salt or prodrug thereof can be administered In a second tablet.
  • the dosage forms can be the same(e.g., both as pills) or different (e.g. one can be formulated in a pill and the other compound can be formulated as an injectable).
  • Administration of Compound 1 AI or a pharmaceutically acceptable salt thereof; and Compound 2c, or a pharmaceutically acceptable salt or prodrug thereof can vary.
  • the dosage forms can be administered simultaneously or sequentially.
  • the dosage form that contains Compound 1A, or pharmaceutically acceptable salt thereof can be administered before, after, in-between, concurrently or sequentially with Compounds 2c, or pharmaceutically acceptable salts or prodrugs thereof.
  • the dosage form that contains Compound 2c, or a pharmaceutically acceptable salt or prodrug thereof can be administered before, after, in-between, concurrently or sequentially with Compound 1 A, or pharmaceutically acceptable salts or prodrugs thereof.
  • Compounds 1 A; and Compound 2c, or pharmaceutically acceptable salts or prodrugs thereof can be administered concurrently.
  • the term “concurrently means effective concentrations of both compounds are present in a subject.
  • Compound 1 A, and Compound 2c, or pharmaceutically acceptable salts or prodrugs thereof can be administered in the same dosage form or separate dosage forms.
  • Compound 1 A, and Compound 2c, or pharmaceutically acceptable salts or prodrugs hereof can be administered sequentially.
  • the term “sequentially” means administering one compound for a first time period, then administering a second compound for a second time period, in which the first and second time periods do not overlap.
  • One aspect of the invention relates to a pharmaceutical composition that comprises, in a pharmaceutically acceptable vehicle, carrier or diluent: a first compound, with the formula
  • the invention in another aspect relates to a composition
  • a composition comprising, in a pharmaceutically acceptable vehicle, carrier or diluent Compound 1 A, and Compound 2d and/or Compound 3d, or pharmaceutically acceptable salts or prodrugs thereof, wherein the composition additionally comprises one or more therapeutic agents.
  • the one or more therapeutic agents are ribavirin and ritonavir.
  • one aspect of the invention provides pharmaceutical compositions comprising, in a pharmaceutically acceptable vehicle, carrier or diluent, Compound 1 A and sovaprevir.
  • a further aspect of the invention provides pharmaceutical compositions comprising, in a pharmaceutically acceptable vehicle, carrier or diluent, Compound 1A and odalasvir.
  • a further aspect of the invention provides pharmaceutical compositions comprising, in a pharmaceutically acceptable vehicle, carrier or diluent, Compound 1 A and sovaprevir and odalasvir.
  • the invention relates to the use of such compositions for ameliorating or treating a disease condition in a patient population, and/or for the preparation of a medicament for ameliorating or treating such a disease condition.
  • the disease condition can be selected from a hepatitis C virus infection, liver fibrosis, and impaired liver function.
  • the invention relates to the use of a composition comprising Compound 1A, and Compound 2d and/or Compound 3d, or pharmaceutically acceptable salts or prodrugs thereof, for ameliorating or treating hepatitis C virus infection, liver fibrosis, and impaired liver function.
  • the invention in another aspect relates to a method for ameliorating or treating a disease condition in a patient population that comprises administering a therapeutically effective amount of a first compound, or a pharmaceutically acceptable salt or prodrug thereof wherein the first compound is Compound 1 A; and a therapeutically effective amount of a second compound, or a pharmaceutically acceptable salt or prodrug thereof, wherein the second compound is Compound 2d; and/or a therapeutically effective amount of a third compound, or a pharmaceutically acceptable salt or prodrug thereof wherein the third compound is Compound 3d; to a subject suffering from the disease conditions.
  • the disease condition can be selected from & hepatitis C virus infection, liver fibrosis, and impaired liver function.
  • the invention relates to the use of Compound 1A or a pharmaceutically acceptable salt or prodrug thereof for ameliorating or treating a disease condition in a patient population and/or for the preparation of a medicament for ameliorating or treating such a disease condition, wherein Compound 1A or a pharmaceutically acceptable salt thereof is manufactured for use in combination with Compound 2d or a pharmaceutically acceptable salt or prodrug thereof; and wherein Compound 1 A and Compound 2d or pharmaceutically acceptable salts or prodrugs thereof are manufactured for use in combination with Compound 3d or a pharmaceutical acceptable salt or prodrug thereof wherein the disease condition is selected from hepatitis C virus infection, liver fibrosis, and impaired liver function,
  • the method or use for ameliorating or treating a disease condition in a patient population comprises administering, one or more additional therapeutic agents.
  • the one or more additional therapeutic agents are ribavirin and/or ritonavir, Sovaprevir, (INN), (Compound 2d) has shown to be effective in inhibiting HCV replication.
  • the aforementioned compound can be obtained using methods known to those skilled in the art, including, for example, those methods disclosed in WO2008/008502, and WO2014/152928, which are hereby incorporated by reference in its entirety.
  • this invention is not limited by any particular theory, Compound 2d is believed to inhibit the HCV protease, in particular the NS3/4A protease.
  • Pharmaceutically acceptable salts and prodrugs of Compound 2d can be utilized in the compositions described herein.
  • Odalasvir (PINN, also known as ACH-3102) (Compound 3d) has been demonstrated to be effective in inhibiting HCV replication.
  • Compound 3d can be obtained using methods known to those skilled in the art, such as those methods described in WO2012/166716, which is hereby incorporated by reference in its entirety.
  • this invention is not limited by any particular theory, it is believed that Compound 3d is an NS5A inhibitor where NS5A is a protein involved in the replication of the hepatitis C virus.
  • the composition can further include a pharmaceutically acceptable exipient, diluent and/or carrier, such as those described herein.
  • a pharmaceutically acceptable exipient, diluent and/or carrier such as those described herein.
  • an antiviral effective daily amount of Compound 1 A within the combination treatment of the invention would be from about 0.01 to about 700 mg/kg, or about 0.5 to about 400 mg/kg, or about 1 to about 250 mg/kg, or about 2 to about 200 mg/kg, or about 10 to about 150 mg/kg, or about 0.1 to about 50 mg/kg, or about 1 to about 20 mg/kg, or about 2 to about 10 mg/kg, or about 5 to about 8 mg/kg body weight.
  • Said sub-doses may be formulated as unit dosage forms, for example, containing about 1 to about 5000 mg Compound 1A, or about 50 to about 3000 mg Compound 1A, or about 100 to about 1000 mg Compound 1 A, or about 200 to about 600 mg Compound 1A, or about 100 to about 400 mg Compound 1A, or about 7 to about 3500 mg Compound 1A, or about 70 to about 1400 mg Compound 1 A, or about 140 to about 700 mg Compound 1 A, or about 340 to about 580 mg of Compound 1 A per unit dosage form.
  • the unit dosage form is formulated for QD dosing with one of the above ranges.
  • compositions can include an amount of Compound 2d, or a pharmaceutically acceptable salt or prodrug thereof in the range of about 50 mg to about 1000 mg. In other embodiments, the composition can include an amount of Compound 2d, or a pharmaceutically acceptable salt or prodrug thereof in the range of about 100 mg to about 800 mg. In still other embodiments, the composition can include an amount of Compound 2, or a pharmaceutically acceptable salt or prodrug thereof, in the range of about 200 mg to about 400 mg. In an embodiment, the composition can include an amount of Compound 2d, or a pharmaceutically acceptable salt or prodrug thereof in the range of about 250-350 mg.
  • compositions can include an amount of Compound 3d, or a pharmaceutically acceptable salt or prodrug thereof in the range of about 25 mg to about 225 mg.
  • the composition can include an amount of Compound 3, or a pharmaceutically acceptable salt or prodrug thereof, in the range of about 50 mg to about 100 mg.
  • the composition can include an amount of Compound 3d or a pharmaceutically acceptable salt or prodrug thereof in the range of about 50 mg.
  • the a loading dose of 100-200 mg, such as 150 mg can be administered for the initial 1 -3 days of therapy.
  • a potential advantage of making a combination of Compound 1 A and Compound 2d, and/or 3d, or pharmaceutically acceptable salts or prodrugs thereof may be a reduction in the required amounts of one or more compounds that are effective in treating a disease condition disclosed herein (for example, HCV) as compared to monotherapy treatment of an otherwise comparable patient population using either Compound 1 A, 2d or 3d alone.
  • the amount of Compound 1A ; or a pharmaceutically acceptable salt thereof in the composition can be less compared to the amount of Compound 1A or a pharmaceutically acceptable salt thereof needed to achieve the same viral load reduction when administered as a monotherapy.
  • the amount of Compound 2d or a pharmaceutically acceptable salt or prodrug thereof in the composition can be less compared to the amount of Compound 2d, or a pharmaceutically acceptable salt or prodrug thereof, needed to achieve the same viral load reduction when administered as a monotherapy. In some embodiments, the amount of Compound 3d or a pharmaceutically acceptable salt or prodrug thereof in the composition can be less compared to the amount of Compound 3d or a pharmaceutically acceptable salt or prodrug thereof needed to achieve the same viral load reduction when administered as a monotherapy.
  • the sum of the amount of Compound 1A or a pharmaceutically acceptable salt and the amount of Compound 2d, or a pharmaceutically acceptable salt or prodrug thereof and/or the amount of Compound 3d, or a pharmaceutically acceptable salt or prodrug thereof is less than expected or predicted based on the additive combination of Compound 1 A or a pharmaceutically acceptable salt, alone, Compound 2d, or a pharmaceutically acceptable salt or prodrug thereof alone, and/or Compound 3d, or a pharmaceutically acceptable salt or prodrug thereof alone for treating the disease condition such as HCV.
  • the percentages of Compound 1A, and Compound 2d, and/or 3d or pharmaceutically acceptable salts or prodrugs thereof present in the composition can also vary.
  • the composition can include an amount of Compound 1 A, or a pharmaceutically acceptable salt thereof, in the range of about 1 % to about 98% (weight/weight) based on the sum of the amount of Compound 1 A, or a pharmaceutically acceptable salt thereof and the amount of Compounds 2d and/or 3d or pharmaceutically acceptable salts or prodrugs thereof, in the composition.
  • Additional embodiments include, but are not limited to, an amount of Compound 1A, or a pharmaceutically acceptable salt thereof in the range of about 5% to about 80%, about 10% to about 70%, about 15% to about 60%, about 20% to about 50% and about 30% to about 40% (weight/weight) based on the sum of the amount of Compound 1A, or a pharmaceutically acceptable salt, and the amount of Compounds 2d and/or 3d, or pharmaceutically acceptable safe or prodrugs thereof in the composition.
  • the composition can include an amount of Compound 2d, or a pharmaceutically acceptable salt or prodrug thereof in the range of about 1 % to about 98% (weight/weight) based on the sum of the amount of Compound 2d, or a pharmaceutically acceptable salt or prodrug thereof and the amount of Compounds 1 A and/or 3d, or pharmaceutically acceptable salts or prodrugs thereof, in the composition.
  • Examples of additional embodiments include, but are not limited to, an amount of Compound 2d, or a pharmaceutically acceptable salt or prodrug thereof in the range of about 5% to about 80%, about 10% to about 70%, about 15% to about 60%, about 20% to about 50% and about 30% to about 40 (weight/ weight) based on the sum of the amount of Compound 2d, or a pharmaceutically acceptable salt or prodrug thereof, and the amount of Compounds 1 A and/or 3d, or pharmaceutically acceptable salts or prodrugs thereof in the composition.
  • the composition can include an amount of Compound 3d, or a pharmaceutically acceptable salt or prodrug thereof in the range of about 1 % to about 98% (weight/weight) based on the sum of the amount of Compound 3d, or a pharmaceutically acceptable salt or prodrug thereof, and the amount of Compounds 1 A and/or 2d, or pharmaceutically acceptable salts or prodrugs thereof, in the composition.
  • Examples of additional embodiments include, but are not limited, to, an amount of Compound 3A, or a pharmaceutically acceptable salt or prodrug thereof, in the range of about 5% to about 80%, about 10% to about 70%, about 15% to about 60%, about 20% to about 50% and about 30% to about 40% (weight/weight) based on the sum of the amount of Compound 3d, or a pharmaceutically acceptable salt or prodrug thereof, and the amount of Compounds 1 A and/or 2d, or pharmaceutically acceptable salts or prodrugs thereof in the composition.
  • Some embodiments described herein relate to a method for ameliorating or treating a disease condition that, can include administering an amount of Compound 1 A, or a pharmaceutically acceptable salt thereof, and an amount of Compound 2d, or a pharmaceutically acceptable salt or prodrug thereof and/or an amount of Compound 3d, or a pharmaceutically acceptable salt or prodrug thereof wherein the disease condition can be a hepatitis C virus infection, liver fibrosis, and/or impaired liver function.
  • compositions described herein can be present in the same dosage form such as the compositions described herein.
  • Compound 1 A, and 2d and/or 3d, or pharmaceutically acceptable salts or prodrugs thereof can be administered as separate dosage forms.
  • Compound 1 A can be administered in one tablet
  • Compound 2d, or a pharmaceutically acceptable salt or prodrug thereof can be administered in a second tablet
  • Compound 3d, or a pharmaceutically acceptable salt or prodrug thereof can be administered in a third tablet.
  • the dosage forms can be the same (e.g., as both pills) or different (e.g., two compounds can be formulated in a pill and the other compound can fee formulated as art injectable).
  • Administration of Compound 1 A or a pharmaceutically acceptable salt thereof; and Compound 2d, or a pharmaceutically acceptable salt or prodrug thereof, and/or Compound 3d, or a pharmaceutically acceptable salt or prodrug thereof can vary.
  • the dosage forms can be administered simultaneously or sequentially.
  • the dosage form that contains Compound 1A, or pharmaceutically acceptable salt thereof can be administered before, after, in-between, concurrently or sequentially with Compounds 2d and/or 3d, or pharmaceutically acceptable salts or prodrugs thereof.
  • the dosage form that contains Compound 2d, or a pharmaceutically acceptable salt or prodrug thereof can be administered before, after, in-between, concurrently or sequentially with Compound 1A, or Compounds 1 A and 3d, or pharmaceutically acceptable salts or prodrugs thereof.
  • the dosage form that contains Compound 3d, or a pharmaceutically acceptable salt or prodrug thereof can be administered before, after, inbetween, concurrently or sequentially with Compound 1A or Compound 1 A and 2d, or pharmaceutically acceptable salts or prodrugs thereof.
  • Compounds 1 A; and Compound 2d and/or 3d, or pharmaceutically acceptable salts or prodrugs thereof can be administered concurrently.
  • the term “concurrently means effective concentrations of all three compounds are present in a subject.
  • Compound 1 A, and Compound 2d and/or 3d, or pharmaceutically acceptable salts or prodrugs thereof can be administered in the same dosage form or separate dosage forms.
  • Compound 1A, and Compound 2d, and/or 3d, or pharmaceutically acceptable salts or prodrugs hereof can be administered sequentially.
  • the term “sequentially” means administering one compound for a first time period, then administering a second compound for a second time period, optionally followed by administering a third compound for a third period, in which the first, second, and if applicable third time periods do not overlap.
  • a pharmaceutical composition that comprises, in a pharmaceutically acceptable vehicle, carrier or diluent: a first compound, with the formula
  • the invention in another aspect relates to a composition
  • a composition comprising, in a pharmaceutically acceptable vehicle, carrier or diluent Compound 1 A, and Compound 2e and/or Compound 3e, or pharmaceutically acceptable salts or prodrugs thereof, wherein the composition additionally comprises one or more therapeutic agents.
  • the one or more therapeutic agents are ribavirin and ritonavir.
  • one aspect of the invention provides pharmaceutical compositions comprising, in a pharmaceutically acceptable vehicle, carrier or diluent, Compound 1 A and grazoprevir.
  • a further aspect of the invention provides pharmaceutical compositions comprising, in a pharmaceutically acceptable vehicle, carrier or diluent, Compound 1A and elbasvir.
  • a further aspect of the invention provides pharmaceutical compositions comprising, in a pharmaceutically acceptable vehicle, carrier or diluent, Compound 1 A and grazoprevir and elbasvir.
  • the invention relates to the use of such compositions for ameliorating or treating a disease condition in a patient population, and/or for the preparation of a medicament for ameliorating or treating such a disease condition.
  • the disease condition can be selected from a hepatitis C virus infection, liver fibrosis, and impaired liver function.
  • the invention relates to the use of a composition comprising Compound 1A, and Compound 2e and/or Compound 3e, or pharmaceutically acceptable salts or prodrugs thereof, for ameliorating or treating hepatitis C virus infection, liver fibrosis, and impaired liver function.
  • the invention in another aspect relates to a method for ameliorating or treating a disease condition in a patient population that comprises administering a therapeutically effective amount of a first compound, or a pharmaceutically acceptable salt or prodrug thereof wherein the first compound is Compound 1 A; and a therapeutically effective amount of a second compound, or a pharmaceutically acceptable salt or prodrug thereof, wherein the second compound is Compound 2e; and/or a therapeutically effective amount of a third compound, or a pharmaceutically acceptable salt or prodrug thereof wherein the third compound is Compound 3e; to a subject suffering from the disease conditions.
  • the disease condition can be selected from & hepatitis C virus infection, liver fibrosis, and impaired liver function.
  • the invention relates to the use of Compound 1A or a pharmaceutically acceptable salt thereof for ameliorating or treating a disease condition in a patient population and/or for the preparation of a medicament for ameliorating or treating such a disease condition, wherein Compound 1 A or a pharmaceutically acceptable salt thereof is manufactured for use in combination with Compound 2e or a pharmaceutically acceptable salt or prodrug thereof; and wherein Compound 1 A and Compound 2e or pharmaceutically acceptable salts or prodrugs thereof are manufactured for use in combination with Compound 3e or a pharmaceutical acceptable salt or prodrug thereof wherein the disease condition is selected from hepatitis C virus infection, liver fibrosis, and impaired liver function,
  • the method or use for ameliorating or treating a disease condition in a patient population comprises administering, one or more additional therapeutic agents.
  • the one or more additional therapeutic agents are ribavirin and ritonavir, Grazoprevir, (Compound 2e) has shown to be effective in inhibiting HCV replication.
  • the aforementioned compound can be obtained using methods known to those skilled in the art, including, for example, those methods disclosed in WO2010/01 1566 and WO2013/024465, which are hereby incorporated by reference in its entirety.
  • this invention is not limited by any particular theory, Compound 2e is believed to inhibit the HCV protease, in particular the NS3/4A protease.
  • Pharmaceutically acceptable salts and prodrugs of Compound 2e can be utilized in the compositions described herein.
  • Compound 3e has been demonstrated to be effective in inhibiting HCV replication.
  • Compound 3e can be obtained using methods known to those skilled in the art, such as those methods described in WO2012/040923, which is hereby incorporated by reference in its entirety. Although this invention is not limited by any particular theory, it is believed that Compound 3e is an NS5A inhibitor a protein involved in the replication of the hepatitis C virus.
  • the composition can further include a pharmaceutically acceptable exipient, diluent and/or carrier, such as those described herein.
  • an antiviral effective daily amount of Compound 1 A within the combination treatment of the invention would be from about 0.01 to about 700 mg/kg, or about 0.5 to about 400 mg/kg, or about 1 to about 250 mg/kg, or about 2 to about 200 mg/kg, or about 10 to about 150 mg/kg, or about 0.1 to about 50 mg/kg, or about 1 to about 20 mg/kg, or about 2 to about 10 mg/kg, or about 5 to about 8 mg/kg body weight. It may be appropriate to administer the required dose as one, two, three, four or more sub-doses at appropriate intervals throughout the day.
  • Said sub-doses may be formulated as unit dosage forms, for example, containing about 1 to about 5000 mg Compound 1A, or about 50 to about 3000 mg Compound 1A, or about 100 to about 1000 mg Compound 1 A, or about 200 to about 600 mg Compound 1A, or about 100 to about 400 mg Compound 1A, or about 7 to about 3500 mg Compound 1A, or about 70 to about 1400 mg Compound 1 A, or about 140 to about 700 mg Compound 1 A, or about 340 to about 580 mg of Compound 1 A per unit dosage form.
  • the unit dosage form is formulated for QD dosing with one of the above ranges.
  • compositions can include an amount of Compound 2e, or a pharmaceutically acceptable salt or prodrug thereof in the range of about 10 mg to about 250 mg. In other embodiments, the composition can include an amount of Compound 2e, or a pharmaceutically acceptable salt or prodrug thereof in the range of about 25 mg to about 150 mg. In still other embodiments, the composition can include an amount of Compound 2e, or a pharmaceutically acceptable salt or prodrug thereof, in the range of about 50 mg to about 100 mg. In an embodiment, the composition can include an amount of Compound 2e, or a pharmaceutically acceptable salt or prodrug thereof in the range of about 100 mg.
  • compositions can include an amount of Compound 3e, or a pharmaceutically acceptable salt or prodrug thereof in the range of about 10 mg to about 250 mg. In other embodiments, the composition can include an amount of Compound 3e, or a pharmaceutically acceptable salt or prodrug thereof, in the range of about 25 mg to about 100 mg. In still other embodiments, the composition can include an amount of Compound 3e or a pharmaceutically acceptable salt or prodrug thereof in the range of about 40 mg to about 60 mg. In yet still other embodiments, the composition can include an amount of Compound 3e, or a pharmaceutically acceptable salt or prodrug thereof in the amount of about 50 mg.
  • a potential advantage of making a combination of Compound 1 A and Compound 2e, and/or 3e, or pharmaceutically acceptable salts or prodrugs thereof may be a reduction in the required amounts of one or more compounds that are effective in treating a disease condition disclosed herein (for example, HCV) as compared to monotherapy treatment of an otherwise comparable patient population using either Compound 1 A, 2e or 3e alone.
  • the amount of Compound 1A ; or a pharmaceutically acceptable salt thereof in the composition can be less compared to the amount of Compound 1A or a pharmaceutically acceptable salt thereof needed to achieve the same viral load reduction when administered as a monotherapy.
  • the amount of Compound 2e or a pharmaceutically acceptable salt or prodrug thereof in the composition can be less compared to the amount of Compound 2e, or a pharmaceutically acceptable salt or prodrug thereof, needed to achieve the same viral load reduction when administered as a monotherapy. In some embodiments, the amount of Compound 3e or a pharmaceutically acceptable salt or prodrug thereof in the composition can be less compared to the amount of Compound 3e or a pharmaceutically acceptable salt or prodrug thereof needed to achieve the same viral load reduction when administered as a monotherapy.
  • the sum of the amount of Compound 1A or a pharmaceutically acceptable salt and the amount of Compound 2e, or a pharmaceutically acceptable salt or prodrug thereof and/or the amount of Compound 3e, or a pharmaceutically acceptable salt or prodrug thereof is less than expected or predicted based on the additive combination of Compound 1 A or a pharmaceutically acceptable salt thereof, alone, Compound 2e, or a pharmaceutically acceptable salt or prodrug thereof alone, and/or Compound 3e, or a pharmaceutically acceptable salt or prodrug thereof alone for treating the disease condition such as HCV.
  • the percentages of Compound 1A, and Compound 2e, and/or 3e or pharmaceutically acceptable salts or prodrugs thereof present in the composition can also vary.
  • the composition can include an amount of Compound 1 A, or a pharmaceutically acceptable salt thereof, in the range of about 1 % to about 98% (weight/weight) based on the sum of the amount of Compound 1 A, or a pharmaceutically acceptable salt thereof and the amount of Compounds 2e and/or 3e or pharmaceutically acceptable salts or prodrugs thereof, in the composition.
  • Additional embodiments include, but are not limited to, an amount of Compound 1A, or a pharmaceutically acceptable salt thereof in the range of about 5% to about 80%, about 10% to about 70%, about 15% to about 60%, about 20% to about 50% and about 30% to about 40% (weight/weight) based on the sum of the amount of Compound 1A, or a pharmaceutically acceptable salt or prodrug thereof, and the amount of Compounds 2e and/or 3e, or pharmaceutically acceptable safe or prodrugs thereof in the composition.
  • the composition can include an amount of Compound 2e, or a pharmaceutically acceptable salt or prodrug thereof in the range of about 1 % to about 98% (weight/weight) based on the sum of the amount of Compound 2e, or a pharmaceutically acceptable salt or prodrug thereof and the amount of Compounds 1 A and/or 3e, or pharmaceutically acceptable salts or prodrugs thereof, in the composition.
  • Examples of additional embodiments include, but are not limited to, an amount of Compound 2e, or a pharmaceutically acceptable salt or prodrug thereof in the range of about 5% to about 80%, about 10% to about 70%, about 15% to about 60%, about 20% to about 50% and about 30% to about 40 (weight/ weight) based on the sum of the amount of Compound 2e, or a pharmaceutically acceptable salt or prodrug thereof, and the amount of Compounds 1 A and/or 3e, or pharmaceutically acceptable salts or prodrugs thereof in the composition.
  • the composition can include an amount of Compound 3e, or a pharmaceutically acceptable salt or prodrug thereof in the range of about 1 % to about 98% (weight/weight) based on the sum of the amount of Compound 3e, or a pharmaceutically acceptable salt or prodrug thereof, and the amount of Compounds 1 A and/or 2e, or pharmaceutically acceptable salts or prodrugs thereof, in the composition.
  • Examples of additional embodiments include, but are not limited, to, an amount of Compound 3e, or a pharmaceutically acceptable salt or prodrug thereof, in the range of about 5% to about 80%, about 10% to about 70%, about 15% to about 60%, about 20% to about 50% and about 30% to about 40% (weight/weight) based on the sum of the amount of Compound 3e, or a pharmaceutically acceptable salt or prodrug thereof, and the amount of Compounds 1 A and/or 2e, or pharmaceutically acceptable salts or prodrugs thereof in the composition.
  • Some embodiments described herein relate to a method for ameliorating or treating a disease condition that, can include administering an amount of Compound 1 A, or a pharmaceutically acceptable salt thereof, and an amount of Compound 2e, or a pharmaceutically acceptable salt or prodrug thereof and/or an amount of Compound 3e, or a pharmaceutically acceptable salt or prodrug thereof wherein the disease condition can be a hepatitis C virus infection, liver fibrosis, and/or impaired liver function.
  • Various dosages forms of Compound 1A or a pharmaceutically acceptable salt thereof, and/or Compound 2e, or a pharmaceutically acceptable salt or prodrug thereof and/or Compound 3e, or a pharmaceutically acceptable salt or prodrug thereof can be used to ameliorate and/or treat a disease condition.
  • Compounds 1 A and 2e and/or 3e or pharmaceutically acceptable salts or prodrugs thereof can be present in the same dosage form such as the compositions described herein.
  • Compound 1 A, and 2e and/or 3e, or pharmaceutically acceptable salts or prodrugs thereof can be administered as separate dosage forms.
  • Compound 1 A, or a pharmaceutically acceptable salt thereof can be administered in one tablet
  • Compound 2e, or a pharmaceutically acceptable salt or prodrug thereof can be administered in a second tablet
  • Compound 3e, or a pharmaceutically acceptable salt or prodrug thereof can be administered in a third tablet.
  • the dosage forms can be the same (e.g., both or all as pills) or different (e.g., two compounds can be formulated in a pill and the other compound can fee formulated as art injectable).
  • Administration of Compound 1 A or a pharmaceutically acceptable salt thereof; and Compound 2e, or a pharmaceutically acceptable salt or prodrug thereof, and/or Compound 3e, or a pharmaceutically acceptable salt or prodrug thereof can vary.
  • the dosage forms can be administered simultaneously or sequentially.
  • the dosage form that contains Compound 1A, or pharmaceutically acceptable salt thereof can be administered before, after, in-between, concurrently or sequentially with Compounds 2e and/or 3e, or pharmaceutically acceptable salts or prodrugs thereof.
  • the dosage form that contains Compound 2e, or a pharmaceutically acceptable salt or prodrug thereof can be administered before, after, in-between, concurrently or sequentially with Compound 1A, or Compounds 1 A and 3e, or pharmaceutically acceptable salts or prodrugs thereof.
  • the dosage form that contains Compound 3e, or a pharmaceutically acceptable salt or prodrug thereof can be administered before, after, inbetween, concurrently or sequentially with Compound 1A or Compound 1 A and 2e, or pharmaceutically acceptable salts or prodrugs thereof.
  • Compounds 1 A; and Compound 2e and/or 3e, or pharmaceutically acceptable salts or prodrugs thereof can be administered concurrently.
  • the term “concurrently means effective concentrations of all three compounds are present in a subject.
  • Compound 1 A, and Compound 2e and/or 3e, or pharmaceutically acceptable salts or prodrugs thereof can be administered in the same dosage form or separate dosage forms.
  • Compound 1A, and Compound 2e, and/or 3e, or pharmaceutically acceptable salts or prodrugs hereof can be administered sequentially.
  • the term “sequentially” means administering one compound for a first time period, then administering a second compound for a second time period, optionally followed by administering a third compound for a third period, in which the first, second, and if applicable third time periods do not overlap.
  • the invention relates to compounds for use in the treatment of HCV genotype 3a infection.
  • Representative genotypes in the context of treatment or prophylaxis in accordance with the invention include genotype 3a, such as wild type genotype 3a and mutant strains of the genotype 3a, for example the S282T and L159/320F mutants.
  • the invention provides a method for the treatment of HCV infection, in particular of the genotype 3a, such as wild type genotype 3a and mutant strains of the genotype 3a, for example the S282T and L159/320F mutants
  • Additional representative HCV genotypes in the context of the invention include the major HCV genotypes, i.e. genotype 1 a, 1 b, 2a, 4a, 5a and 6a.
  • genotype 1 b prevalent in Europe
  • 1 a prevalent in North America
  • the invention further relates to the treatment or prophylaxis of HCV infection caused by the genotypes 2a, 4a, 5a, 6a, especially the treatment of HCV infection caused by the genotypes 2a, 4a, 5a, 6a.
  • the invention also provides a method for the treatment or prophylaxis of HCV infection, of the genotypes 2a, 4a, 5a, 6a, especially the treatment of HCV infection caused by the genotypes 2a, 4a, 5a, 6a.
  • compositions of the invention have pan-genotypic coverage against each of the 6 genotypes, that is the EC 50 of the compound of the invention does not differ markedly between genotypes, thereby simplifying treatment.
  • the combination therapies of the invention are generally applicable to all HCV genotypes, especially genotype 1 a and genotype 1 b.
  • the compound of the invention have several chiral centers and may exist and be isolated in optically active and racemic forms, andmay exhibit polymorphism. It is to be understood that any racemic, optically active, diastereomeric, polymorphic or stereoisomeric form or mixtures thereof, of a compound provided herein is within the scope of this invention.
  • the absolute configuration of such compounds can be determined using methods known in the art such as, for example, X-ray diffraction or NMR and/or implication from starting materials of known stereochemistry and/or stereoselective synthesis methods.
  • compositions in accordance with the invention will preferably comprise substantially stereoisomerically pure preparations of the indicated stereoisomer.
  • Pure stereoisomeric forms of the compounds and intermediates as mentioned herein are defined as isomers substantially free of other enantiomeric or diastereomeric forms of the same basic molecular structure of said compounds or intermediates.
  • stereoisomerically pure concerns compounds or intermediates having a stereoisomeric excess of at least 80% (i.e. minimum 90% of one isomer and maximum 10% of the other possible isomers) up to a stereoisomeric excess of 100% (i.e. 100% of one isomer and none of the other), more in particular, compounds or intermediates having a stereoisomeric excess of 90% up to 100%, even more in particular having a stereoisomeric excess of 94% up to 100% and most in particular having a stereoisomeric excess of 97% up to 100%.
  • enantiomerically pure and “diastereomerically pure” should be understood in a similar way, but then having regard to the enantiomeric excess, and the diastereomeric excess, respectively, of the mixture in question.
  • diastereomeric salt effected by reaction with an optically active acid or base followed by selective crystallization of the formed diastereomeric salt.
  • optically active acid or base examples include tartaric acid, dibenzoyltartaric acid, ditoluoyltartaric acid and camphorsulfonic acid.
  • enantiomers may be separated by chromatographic techniques using chiral stationary phases. Pure stereochemically isomeric forms may also be obtained by synthesis from stereochemically pure forms of the appropriate starting materials, provided that the reaction occurs
  • Diastereomeric racemates of the compounds of the invention can be separated by conventional methods. Appropriate physical separation methods that may advantageously be employed are, for example, selective crystallization and chromatography, e.g. column chromatography.
  • the phosphorus atom may represent a chiral centre.
  • the chirality at this centre is designated "R” or "S” according to the Cahn-lngold-Prelog priority rules.
  • R or "S”
  • both the R- and S-isomers are meant to be included as well as a mixture of both stereoisomers.
  • compounds of formula (I) or any subgroup of formula (I) are pure stereoisomers at the phosphorus atom.
  • Preferred are compounds having the S- configuration at the phosphorus atom. These stereoisomers are designated S P .
  • the present invention also includes isotope-labelled compounds of formula (I) or any subgroup of formula (I), wherein one or more of the atoms is replaced by an isotope of that atom, i.e. an atom having the same atomic number as, but an atomic mass different from, the one(s) typically found in nature.
  • isotopes that may be incorporated into the compounds of formula (I) or any subgroup of formula (I), include but are not limited to isotopes of hydrogen, such as 2 H and 3 H (also denoted D for deuterium and T for tritium, respectively), carbon, such as 11 C, 13 C and 14 C, nitrogen, such as 13 N and 15 N, oxygen, such as 15 0, 17 0 and 18 0, phosphorus, such as 31 P and 32 P, sulfur, such as 35 S, fluorine, such as 18 F, chlorine, such as 36 CI, bromine such as 75 Br, 76 Br, 77 Br and 82 Br, and iodine, such as 123 l, 124 l, 125 l and 131 1.
  • isotopes of hydrogen such as 2 H and 3 H (also denoted D for deuterium and T for tritium, respectively)
  • carbon such as 11 C, 13 C and 14 C
  • nitrogen such as 13 N and 15 N
  • oxygen such as 15 0, 17 0 and 18
  • isotope included in an isotope-labelled compound will depend on the specific application of that compound. For example, for drug or substrate tissue distribution assays, compounds wherein a radioactive isotope such as 3 H or 14 C is incorporated will generally be most useful. For radio-imaging applications, for example positron emission tomography (PET) a positron emitting isotope such as 11 C, 18 F, 13 N or 15 0 will be useful.
  • PET positron emission tomography
  • a heavier isotope such as deuterium, i.e. 2 H, may provide greater metabolic stability to a compound of formula (I) or any subgroup of formula (I), which may result in, for example, an increased in vivo half life of the compound or reduced dosage requirements.
  • Isotope-labelled compounds of formula (I) or any subgroup of formula (I) can be prepared by processes analogous to those described in the Schemes and/or Examples herein below by using the appropriate isotope-labelled reagent or starting material instead of the corresponding non-isotope-labelled reagent or starting material, or by conventional techniques known to those skilled in the art.
  • the pharmaceutically acceptable addition salts comprise the therapeutically active non-toxic acid and base addition salt forms of the compounds of formula (I). Of interest are the free, i.e. non-salt forms of the compounds of formula (I).
  • the pharmaceutically acceptable acid addition salts can conveniently be obtained by treating the base form with such appropriate acid.
  • Appropriate acids comprise, for example, inorganic acids such as hydrohalic acids, e.g. hydrochloric or hydrobromic acid, sulfuric, nitric, phosphoric and the like acids; or organic acids such as, for example, acetic, propionic, hydroxyacetic, lactic, pyruvic, oxalic (i.e. ethanedioic), malonic, succinic (i.e. butanedioic acid), maleic, fumaric, malic (i.e.
  • salt forms can be converted by treatment with an appropriate base into the free base form.
  • the compounds of formula (I) containing an acidic proton may also be converted into their nontoxic metal or amine addition salt forms by treatment with appropriate organic and inorganic bases.
  • Appropriate base salt forms comprise, for example, the ammonium salts, the alkali and earth alkaline metal salts, e.g.
  • solvates covers any pharmaceutically acceptable solvates that the compounds of formula (I) as well as the salts thereof, are able to form.
  • Such solvates are for example hydrates, alcoholates, e.g. ethanolates, propanolates, and the like, especially hydrates.
  • the names of compounds used in this application are generated using ChemDraw Ultra 12.0.
  • the stereochemistry of a structure or a portion of a structure is not indicated with for example bold or dashed lines, the structure or portion of that structure is to be interpreted as encompassing all stereoisomers of it.
  • the triphosphate (15c) is then achieved by reaction with a pyrophosphate for instance tributylamine pyrophosphate followed by treatment with ammonia.
  • the triphosphate is subjected to the appropriate ion exchange procedure, for instance, if the potassium salt form is desired, the residue is passed through a column Dowex ® -K + .
  • the useful in vivo dosage to be administered and the particular mode of administration will vary depending upon the age, weight, the severity of the affliction, the mammalian species treated, the particular compounds employed, and the specific use for which these compounds are employed. (See e.g. Fingl et al 1975, in “The Pharmacological Basis of Therapeutics " , which is hereby incorporated herein by reference in its entirety, with particular reference to Ch. 1 , p. 1 ).
  • the determination of effective dosage levels that is the dosage levels necessary to achieve the desired result, can be accomplished by one skilled in the art using routine pharmacological methods. Typically, human clinical applications of products are commenced at lower dosage levels, with dosage level being increased until the desired effect is achieved.
  • acceptable in vitro studies can be used to establish useful doses and routes of administration of the compositions identified by the present methods using established pharmacological methods.
  • the dosage may be a single one or a series of two or more given in the course of one or more days, as is needed by the subject.
  • the compounds will be administered tor a period of continuous therapy, for example for a week or more, or for months or years,
  • human dosages for compounds have been established for at least some conditions, those same dosages, or dosages that are between about 10% and 200%, more preferably between about 50% and 150% of the established human dosage will be used.
  • a suitable human dosage can be inferred from ED 50 or ID 50 values, or other appropriate values derived from in vitro or in vivo studies, as qualified by toxicity studies and efficacy studies in animals. It will often be convenient to use the same dose as monotherapy for the respective components of the combination, with the aim of achieving a more profound antiviral effect, onset of action, reduction in side effects or reduction in resistance.
  • dosages may be calculated as the free base or the free acid as the case may be.
  • dosages may be calculated as the free base or the free acid as the case may be.
  • Dosage amount and interval may be adjusted individually to provide plasma levels of the active moiety which are sufficient to maintain the modulating effects, or minimal effective concentration (MEC).
  • MEC minimal effective concentration
  • the MEC will vary for each compound but can be estimated from in vitro data. Dosages necessary to achieve the MEC will depend on individual characteristics and route of administration. However, conventional MS or HPLC assays or bioassays can be used to determine plasma concentrations.
  • Dosage intervals can also be determined using MEC value.
  • Compositions should be administered using a regimen which maintains plasma levels above the MEC for 10-90% of the time, preferably between 30-90% and most preferably between 50-90%.
  • the effective local concentration of the drug may not be related to plasma concentration.
  • the attending physician would know how to and when to terminate, interrupt, or adjust administration due to toxicity or organ dysfunctions. Conversely, the attending physician would also know to adjust treatment, to higher levels if the clinical response was not adequate (precluding toxicity).
  • the magnitude of an administrated dose in the management of the disorder of interest will vary with the severity of the condition to be treated and the route of administration. The severity of the condition may, for example, be evaluated, in part, by standard prognostic evaluation methods. Further, the dose and perhaps dose frequency will also vary according to the age, body weight, and response of the individual patient. A program comparable to that discussed above may be used in veterinary medicine.
  • dosage levels In non-human animal studies, applications of potential products are commenced at higher dosage levels, with dosage being decreased until the desired effect is no longer achieved or adverse side effects disappear.
  • the dosage may range broadly, depending upon the desired effects and the therapeutic indication. Alternatively dosages may he based and calculated upon the surface area of the patient, as understood by those of skill in the art.
  • the toxicology of a particular compound, or of a subset of the compounds, sharing certain chemical moieties may be established by determining in vitro toxicity towards a cell line, such as a mammalian, and preferably human, cell line v
  • a cell line such as a mammalian, and preferably human, cell line v
  • the results of such studies are often predictive of toxicity in animals, such as mammals, or more specifically, humans.
  • the toxicity of particular compounds in an animal model such as mice, rats, rabbits, or monkeys, may he determined using known methods.
  • the efficacy of a particular compound may be established using several recognized methods, such as in vitro methods, animal models, or human clinical trials.
  • acceptable animal models may be used to establish efficacy of chemicals to treat such conditions.
  • the skilled artisan can be guided by the state of the art to choose an appropriate model, dose, and route of administration, and regime.
  • human clinical trials can also be used to determine the efficacy of a compound or composition in humans.
  • compositions and methods described herein can be administered to individuals who have been diagnosed with an HCV infection. Any of the compositions and methods described herein can be administered to individuals who have failed previous treatment for HCV infection (treatment failure patients, including non-responders and relapsers).
  • Treatment failure patients including non-responders and relapsers.
  • Individuals who have been clinically diagnosed as infected with HCV are of particular interest in many embodiments. Individual who are infected with HCV are identified as having HCV RNA in their blood, and/or having anti-HCV antibody in their serum. Such individuals include anti HCV ELISA-positive individuals, and individuals with a positive recombinant immunoblot assay (RIBA). Such individuals may also, but need not, have elevated serum ALT levels.
  • RIBA positive recombinant immunoblot assay
  • Treatment failure patients include non-responders (i.e. individuals in whom the HCV titer was not significantly or sufficiently reduced by a previous treatment for HCV, e.g. a previous IFN-alpha monotherapy, a previous IFN-alpha and ribavirin combination therapy, or a previous pegylated IFN-alpha and ribavirin combination therapy); and relapsers (i.e.
  • HCV-positive individuals have an HCV titer of at least about 10 5 , at least about 5 x 10 5 or at least about 10 6 or at least about 2x10 6 genome copies of HCV per milliliter of serum.
  • the patient may be infected with any HCV genotype (genotype I, including la and lb, 2, 3, 4, 6, etc, and subtypes (e.g. 2a, 2b, 3a etc)), particularly difficult to treat genotypes such as HCV genotype 1 , and particular HCV subtypes and quasispecies.
  • the patient is infected with HCV genotype 1 a.
  • the patient is infected with genotype 1 b.
  • the HCV-positive individuals are those who exhibit severe fibrosis or early cirrhosis (non-decompensated Childs-Pugh class A or less), or more advanced cirrhosis (decompensated, Childs-Pugh class B or C) due to chronic HCV infection and who are viremic despite prior anti-viral treatment with IFN-alpha-based therapies or who cannot tolerate IFN-based therapies, or who have a contraindication to such therapies.
  • HCV-positive individuals with stage 3 or 4 liver fibrosis according to the METAVIR scoring system are suitable for treatment with the compositions and methods described herein.
  • individuals suitable for treatment with the compositions and methods described herein are patients with decompensated cirrhosis with clinical manifestations, including patients with far-advanced liver cirrhosis, including those awaiting liver transplantation.
  • individuals suitable for treatment with the compositions md methods described herein include patients with milder degrees of fibrosis including those with early fibrosis (stages 1 and 2 in the METAVIR, Ludwig, and Schemer scoring systems; or stages 1 , 2, or in the Ishak scoring system).
  • the present invention concerns a pharmaceutical composition
  • a pharmaceutical composition comprising a therapeutically effective amount of a compound of formula (1 A), and a pharmaceutically acceptable carrier.
  • a therapeutically effective amount in this context is an amount sufficient to stabilize or to reduce viral infection, and in particular HCV infection, in infected subjects (e.g. humans).
  • the "therapeutically effective amount” will vary depending on individual requirements in each particular case. Features that influence the dose are e.g. the severity of the disease to be treated, age, weight, general health condition etc. of the subject to be treated, route and form of administration.
  • the invention relates to the use of a compound of formula (1 A), for the treatment of "treatment naive" patients, i.e. patients infected with HCV that are not previously treated against the infection.
  • the invention relates to the use of a compound of formula (1 A), the treatment of "treatment experienced” patients, i.e. patients infected with HCV that are previously treated against the infection and have subsequently relapsed.
  • the invention relates to the use of a compound of formula (1 A), the treatment of "non-responders", i.e. patients infected with HCV that are previously treated but have failed to respond to the treatment.
  • the present invention concerns a pharmaceutical composition
  • a pharmaceutical composition comprising a prophylactically effective amount of a compound of formula (1A) as specified herein, and a pharmaceutically acceptable carrier.
  • a prophylactically effective amount in this context is an amount sufficient to act in a prophylactic way against HCV infection, in subjects being at risk of being infected.
  • this invention relates to a process of preparing a pharmaceutical composition as specified herein, which comprises intimately mixing a pharmaceutically acceptable carrier with a therapeutically or prophylactically effective amount of a compound of formula (1 A), as specified herein. Therefore, the compounds of the present invention may be formulated into various pharmaceutical forms for administration purposes. As appropriate compositions there may be cited all compositions usually employed for systemically administering drugs.
  • compositions of this invention an effective amount of the particular compound, optionally in addition salt form or solvate, as the active ingredient is combined in intimate admixture with a pharmaceutically acceptable carrier, which carrier may take a wide variety of forms depending on the form of preparation desired for administration.
  • a pharmaceutically acceptable carrier which carrier may take a wide variety of forms depending on the form of preparation desired for administration.
  • These pharmaceutical compositions are desirable in unitary dosage form suitable, particularly, for administration orally, rectally, percutaneously, or by parenteral injection.
  • any of the usual pharmaceutical media may be employed such as, for example, water, glycols, oils, alcohols and the like in the case of oral liquid preparations such as suspensions, syrups, elixirs, emulsions and solutions; or solid carriers such as starches, sugars, kaolin, lubricants, binders, disintegrating agents and the like in the case of powders, pills, capsules, and tablets. Because of their ease in administration, tablets and capsules represent the most advantageous oral dosage unit forms, in which case solid pharmaceutical carriers are obviously employed.
  • the carrier will usually comprise sterile water, at least in large part, though other ingredients, for example, to aid solubility, may be included.
  • Injectable solutions may be prepared in which the carrier comprises saline solution, glucose solution or a mixture of saline and glucose solution.
  • Injectable suspensions may also be prepared in which case appropriate liquid carriers, suspending agents and the like may be employed. Also included are solid form preparations intended to be converted, shortly before use, to liquid form preparations.
  • the carrier optionally comprises a penetration enhancing agent and/or a suitable wetting agent, optionally combined with suitable additives of any nature in minor proportions, which additives do not introduce a significant deleterious effect on the skin.
  • the compounds of the present invention may also be administered via oral inhalation or insufflation in the form of a solution, a suspension or a dry powder using any art-known delivery system.
  • Unit dosage form refers to physically discrete units suitable as unitary dosages, each unit containing a predetermined quantity of active ingredient calculated to produce the desired therapeutic effect in association with the required pharmaceutical carrier.
  • unit dosage forms are tablets (including scored or coated tablets), capsules, pills, suppositories, powder packets, wafers, injectable solutions or suspensions and the like, and segregated multiples thereof.
  • the compounds of a combination treatment, or pharmaceutically acceptable salts or prodrugs thereof are formulated in an aqueous buffer.
  • Suitable aqueous buffers include, but are not limited to, acetate, succinate, citrate, and phosphate buffers varying in strengths from about 5 mM to about 100 mM.
  • the aqueous buffer includes reagents that provide for an isotonic solution. Such reagents include, but are not limited to, sodium chloride; and sugars e.g., mannitol, dextrose, sucrose, and the like,
  • the aqueous buffer further includes a non-ionic surfactant such a polysorbate 20 or 80,
  • the compounds, or pharmaceutically acceptable salts or prodrugs thereof of a combination therapy are formulated in a conventional SEDDS (Self emulsifying Drug Delivery System) vehicle, in particular those of Pouten Type IIIA, NIB or IV.
  • SEDDS Self emulsifying Drug Delivery System
  • the formulations may further include a preservative.
  • Suitable preservatives include, but are not limited to, a benzyl alcohol, phenol, chlorobutanol, benzalkonium chloride, and the like.
  • the formulation is stored at about 4 ⁇ .
  • Formulations may also be lyophilized, in which case they generally include cryoprotectants such as sucrose, trehalose, lactose, maltose, mannitol, and the like. Lyophilized formulations can be stored over extended periods of time, even at ambient temperatures.
  • the compounds of formula (1 A) show activity against HCV and can be used in the treatment and/or prophylaxis of HCV infection or diseases associated with HCV.
  • the compounds of formula (1A) can be used in the treatment of HCV infection or diseases associated with HCV.
  • Diseases associated with HCV include progressive liver fibrosis, inflammation and necrosis leading to cirrhosis, end-stage liver disease, and HCC.
  • a number of the compounds of this invention may be active against mutated strains of HCV. Additionally, many of the compounds of this invention may show a favourable pharmacokinetic profile and have attractive properties in terms of bioavailability, including an acceptable half-life, AUC (area under the curve) and peak values and lacking unfavourable phenomena such as insufficient quick onset and tissue retention.
  • the compounds of formula (1 A), the pharmaceutically acceptable addition salts or solvates thereof are useful in the treatment of warm-blooded animals, in particular humans, infected with HCV.
  • the compounds of formula (1A) are further useful for the prophylaxis of HCV infections.
  • the present invention furthermore relates to a method of treating a warm-blooded animal, in particular human, infected by HCV, or being at risk of infection by HCV, said method comprising the administration of an anti-HCV effective amount of a compound of formula (I).
  • the compounds of the present invention may therefore be used as a medicine, in particular as an anti HCV medicine.
  • Said use as a medicine or method of treatment comprises the systemic administration to HCV infected subjects or to subjects susceptible to HCV infection of an amount effective to combat the conditions associated with HCV infection.
  • the present invention also relates to the use of the present compounds in the manufacture of a medicament for the treatment or the prevention of HCV infection.
  • the present invention relates to the use of the compounds of formula (1 A) in the manufacture of a medicament for the treatment of HCV infection.
  • an antiviral effective daily amount would be from about 0.01 to about 700 mg/kg, or about 0.5 to about 400 mg/kg, or about 1 to about 250 mg/kg, or about 2 to about 200 mg/kg, or about 10 to about 150 mg/kg, or about 0.1 to about 50 mg/kg, or about 1 to about 20 mg/kg, or about 2 to about 10 mg/kg, or about 5 to about 8 mg/kg body weight. It may be appropriate to administer the required dose as one, two, three, four or more sub-doses at appropriate intervals throughout the day.
  • Said sub-doses may be formulated as unit dosage forms, for example, containing about 1 to about 5000 mg, or about 50 to about 3000 mg, or about 100 to about 1000 mg, or about 200 to about 600 mg, or about 100 to about 400 mg , or about 7 to about 3500 mg, or about 70 to about 1400 mg, or about 140 to about 700 mg, or about 340 to about 580 mg of active ingredient per unit dosage form.
  • the invention also relates to a combination of a compound of formula (1A), a pharmaceutically acceptable salt or solvate thereof, and another antiviral compound, in particular another anti- HCV compound.
  • the term "combination” may relate to a product containing (a) a compound of formula (1A) and (b) optionally another anti-HCV compound, as a combined preparation for simultaneous, separate or sequential use in treatment of HCV infections.
  • Anti-HCV compounds that can be used in such combinations include HCV polymerase inhibitors, HCV protease inhibitors, inhibitors of other targets in the HCV life cycle, and an immunomodulatory agents, and combinations thereof.
  • HCV polymerase inhibitors include, NM283 (valopicitabine), R803, JTK-109, JTK-003, HCV-371 , HCV-086, HCV-796 and R-1479, R-7128, MK-0608, VCH-759, PF-868554, GS9190, XTL-2125, NM-107, GSK625433, R-1626, BILB-1941 , ANA-598, IDX-184, IDX-375, INX-189, MK-3281 , MK-1220, ABT-333, PSI-7851 , PSI-6130, GS-7977 (sofosbuvir), VCH-916.
  • Inhibitors of HCV proteases include BILN-2061 , VX-950 (telaprevir), GS-9132 (ACH-806), SCH- 503034 (boceprevir), TMC435350 (simeprevir), TMC493706, ITMN-191 , MK-7009, BI-12202, BILN-2065, BI-201335, BMS-605339, R-7227, VX-500, BMS650032, VBY-376, VX-813, SCH-6, PHX-1766, ACH-1625, IDX-136, IDX-316.
  • An example of an HCV NS5A inhibitor is BMS790052, A-831 , A-689, NIM-81 1 and DEBIO-025 are examples of NS5B cyclophilin inhibitors.
  • Inhibitors of other targets in the HCV life cycle including NS3 helicase; metalloprotease inhibitors; antisense oligonucleotide inhibitors, such as ISIS-14803 and AVI-4065; siRNA's such as SIRPLEX-140-N; vector-encoded short hairpin RNA (shRNA); DNAzymes; HCV specific ribozymes such as heptazyme, RPI.13919; entry inhibitors such as HepeX-C, HuMax-HepC; alpha glucosidase inhibitors such as celgosivir, UT-231 B and the like; KPE-02003002; and BIVN 401 .
  • siRNA's such as SIRPLEX-140-N
  • shRNA vector-encoded short hairpin RNA
  • DNAzymes HCV specific ribozymes such as heptazyme, RPI.13919
  • entry inhibitors such as HepeX-C, HuMax-HepC
  • Immunomodulatory agents include, natural and recombinant interferon isoform compounds, including a-interferon, ⁇ -interferon, ⁇ -interferon, and ⁇ -interferon, such as Intron A®, Roferon- A®, Canferon-A300®, Advaferon®, Infergen®, Humoferon®, Sumiferon MP®, Alfaferone®, IFN- beta®, and Feron®; polyethylene glycol derivatized (pegylated) interferon compounds, such as PEG interferon-a-2a (Pegasys®), PEG interferon-a-2b (PEG-lntron®), and pegylated IFN- a-con1 ; long acting formulations and derivatizations of interferon compounds such as the albumin-fused interferon albuferon a; compounds that stimulate the synthesis of interferon in cells, such as resiquimod; interleukins; compounds that enhance the development of
  • antiviral agents include, ribavirin, amantadine, viramidine, nitazoxanide; telbivudine; NOV- 205; taribavirin; inhibitors of internal ribosome entry; broad-spectrum viral inhibitors, such as IMPDH inhibitors, and mycophenolic acid and derivatives thereof, and including, but not limited to, VX-497 (merimepodib), VX-148, and/or VX-944); or combinations of any of the above.
  • interferon-a IFN-a
  • pegylated interferon- ⁇ or ribavirin as well as therapeutics based on antibodies targeted against HCV epitopes
  • small interfering RNA Si RNA
  • ribozymes DNAzymes
  • DNAzymes antisense RNA
  • small molecule antagonists of for instance NS3 protease, NS3 helicase and NS5B polymerase for instance NS3 protease, NS3 helicase and NS5B polymerase.
  • combinations of a compound of formula (I) as specified herein and an anti-HIV compound preferably are those HIV inhibitors that have a positive effect on drug metabolism and/or pharmacokinetics that improve bioavailability.
  • An example of such an HIV inhibitor is ritonavir.
  • this invention further provides a combination comprising (a) a compound of formula (I) or a pharmaceutically acceptable salt or solvate thereof; and (b) ritonavir or a pharmaceutically acceptable salt thereof.
  • the compound ritonavir, its pharmaceutically acceptable salts, and methods for its preparation are described in WO 94/14436. US 6,037,157, and references cited therein: US 5,484,801 , US 08/402,690, WO 95/07696, and WO 95/09614, disclose preferred dosage forms of ritonavir.
  • the invention also concerns a process for preparing a combination as described herein, comprising the step of combining a compound of formula (I) and another agent, such as an antiviral, including an anti-HCV or anti-HIV agent, in particular those mentioned above.
  • the said combinations may find use in the manufacture of a medicament for treating HCV infection in a mammal infected therewith, said combination in particular comprising a compound of formula (I), as specified above and interferon-a (IFN-a), pegylated interferon-a, or ribavirin.
  • the invention provides a method of treating a mammal, in particular a human, infected with HCV comprising the administration to said mammal of an effective amount of a combination as specified herein.
  • said treating comprises the systemic administration of the said combination, and an effective amount is such amount that is effective in treating the clinical conditions associated with HCV infection.
  • the above-mentioned combinations are formulated in the form of a pharmaceutical composition that includes the active ingredients described above and a carrier, as described above.
  • Each of the active ingredients may be formulated separately and the formulations may be co-administered, or one formulation containing both and if desired further active ingredients may be provided.
  • the combinations may also be formulated as a combined preparation for simultaneous, separate or sequential use in HCV therapy.
  • the said composition may take any of the forms described above.
  • both ingredients are formulated in one dosage form such as a fixed dosage combination.
  • the present invention provides a pharmaceutical composition
  • a pharmaceutical composition comprising (a) a therapeutically effective amount of a compound of formula (I), including a possible stereoisomeric form thereof, or a pharmaceutically acceptable salt thereof, or a pharmaceutically acceptable solvate thereof, and (b) a therapeutically effective amount of ritonavir or a pharmaceutically acceptable salt thereof, and (c) a carrier.
  • the individual components of the combinations of the present invention can be administered separately at different times during the course of therapy or concurrently in divided or single combination forms.
  • the present invention is meant to embrace all such regimes of simultaneous or alternating treatment and the term "administering" is to be interpreted accordingly.
  • the separate dosage forms are administered simultaneously.
  • the combinations of the present invention contain an amount of ritonavir, or a pharmaceutically acceptable salt thereof, that is sufficient to clinically improve the bioavailability of the compound of formula (I) relative to the bioavailability when said compound of formula (I) is administered alone.
  • the combinations of the present invention contains an amount of ritonavir, or a pharmaceutically acceptable salt thereof, which is sufficient to increase at least one of the pharmacokinetic variables of the compound of formula (I) selected from t 1/2 , Cmin, C max , C ss , AUC at 12 hours, or AUC at 24 hours, relative to said at least one pharmacokinetic variable when the compound of formula (I) is administered alone.
  • the combinations of this invention can be administered to humans in dosage ranges specific for each component comprised in said combinations, e.g. the compound of formula (I) as specified above, and ritonavir or a pharmaceutically acceptable salt, may have dosage levels in the range of 0.02 to 5.0 g/day.
  • the weight ratio of the compound of formula (I) to ritonavir may be in the range of from about 30:1 to about 1 :15, or about 15: 1 to about 1 : 10, or about 15: 1 to about 1 : 1 , or about 10: 1 to about 1 : 1 , or about 8: 1 to about 1 : 1 , or about 5: 1 to about 1 : 1 , or about 3: 1 to about 1 :1 , or about 2:1 to 1 :1 .
  • the compound formula (I) and ritonavir may be co-administered once or twice a day, preferably orally, wherein the amount of the compound of formula (I) per dose is as described above; and the amount of ritonavir per dose is from 1 to about 2500 mg, or about 50 to about 1500 mg, or about 100 to about 800 mg, or about 100 to about 400 mg, or 40 to about 100 mg of ritonavir.
  • the reaction mixture was distilled at 47-50 °C under reduced pressure (250 - 50 mBar) from a 60 °C bath.
  • toluene 100 mL
  • the distillation at 48-51 °C under reduced pressure 150 - 50 mBar
  • tBME t- butylmethylether
  • Step b) (S)-lsopropyl 2-(((S)-(perfluorophenoxy)(phenoxy)phosphoryl)amino)propanoate (I-52) Phenyl dichlorophosphate (62.88 g, 0.298 mol, 1 .0 eq) was added under nitrogen to a solution of L-alanine isopropylester hydrochloride (50.0 g, 0.298 mol) in DCM (310 mL) at 0 °C - the addition was completed by wash with DCM (39 mL).
  • the mixture was stirred for one hour at -15 to -20 °C, then heated to -8 °C and a solution of pentafluorophenol (60.38 g, 0.328 mol, 1 .1 eq) and triethylamine (33.19 g, 0.328 mol, 1 .1 eq) in DCM (78 mL) was added over a period of 42 minutes with cooling keeping the temperature not higher than 0 °C - the addition was completed by wash with DCM (39 mL). The mixture was stirred for one hour at 0 °C and then over night at +5 °C. The formed precipitate was removed by filtration, and the filter cake washed with DCM (95 mL).
  • the precipitate was isolated by filtration.
  • the filter cake was washed with ethyl acetate:n-heptane (1 :9, 80 mL) and dried to constant under reduced pressure (below 0.1 mBar) without heating, which gave the title compound (75.64 g, 56%) as a white crystalline material.
  • Step f) 1 -((2R.3S,4R.5R)-3-chloro-3-fluoro-4-((triisopropylsilvnoxy)-5- (((triisopropylsilyl)oxy)methyl)tetrahvdrofuran-2-yl)pyrimidine-2,4(1 H,3H)-dione (1 f)
  • Step g) 1 -((2R,3S,4R,5R)-3-chloro-3-fluoro-4-hvdroxy-5-(hvdroxymethyl)tetrahvdrofuran-2- yl)pyrimidine-2,4(1 H,3H)-dione (1 a)
  • Step b) Lithium ((2R,3R,4S,5R)-4-chloro-5-(2,4-dioxo-3,4-dihvdropyrimidin-1 (2H)-yl)-4-fluoro-3- hvdroxytetrahvdrofuran-2-yl)methyl triphosphate (3b)
  • Solvent A 95%water:5%acetonitrile: 0.05M ammonium bicarbonate
  • Solvent B 95%water:5%acetonitrile: 0.8M ammonium bicarbonate
  • Triethylamine tri hydrofluoride (20.5 g, 126 mmol) was added to a stirred solution of compound 19a (17.0 g, 31 .4 mmol) in acetonitrile (1 15 mL) and THF (23 mL). The mixture was stirred for 72 h at rt, 20 h at 50 °C and then at rt overnight. The solution was concentrated on silica (60 g) and purified by silica gel chromatography eluted with a gradient of isohexane and EtOAc, which gave the title compound (68.0 g, 85%).
  • Triethylamine (10.8 g, 107 mmol) was added to a stirred solution of compound 19b (6.80 g, 26.8 mmol) under ice cooling followed by drop wise addition of benzoyl chloride (9.41 g, 66.9 mmol). The mixture was allowed to attain rt and stirred overnight. EtOH (5 mL) was added and the mixture was stirred for 30 minutes, then concentrated in vacuo. Water was added and the mixture was extracted with EtOAc (x3). The organic phase was washed with water and brine, dried (Na 2 S0 4 ), filtered and concentrated under reduced pressure. The product was purified by silica gel chromatography eluted with a gradient of isohexane and EtOAc, which gave the title compound (10.1 g, 86%).
  • Step f) ((2R,3R,4S,5R)-3-(benzoyloxy)-4-chloro-5-(2,4-dioxo-3,4-dihvdropyrimidin-1 (2H)-yl)-4- fluorotetrahvdrofuran-2-yl)methyl benzoate (19f)
  • TMSOTf (6.12 g, 27.5 mmol) was slowly added under N 2 to the solution. After the addition, the reaction mixture was heated to 80 °C for 5 h and then at 65 °C for 16 h.
  • Step e) 1 -((2R,3S,4R,5R)-3-chloro-3-fluoro-4-hvdroxy-5-(hvdroxymethyl)tetrahvdrofuran-2- yl)pyrimidine-2,4(1 H,3l-l)-dione (19a)
  • Deoxy-D-ribose (400.0 g, 2.98 mol) was dissolved in water (1 .6 kg) under nitrogen and the solution cooled to 3 - 7 °C.
  • Bromine 800 g, 10.0 mol, 3.36 eq.
  • the reaction mixture was gently warmed to 20 - 25 ⁇ and then stirred for approximately 20 hours.
  • the reaction mixture was cooled to -5 to -7°C and a solution of sodium hydroxide (27.65%, 720 g, 1 .67 eq.) was added while keeping the reaction temperature at -3 to -7 °C.
  • the water was distilled off at reduced pressure using a scrubber (cooled, 14% sodium hydroxide, 0.9 L), finally at p ⁇ 5 mbar and 50 °C.
  • 2-propanol was added portion wise to the residue followed by azeotropic distillation at reduced pressure.
  • the final water content was determined by KF titration to be less than 1 %.
  • 2-Propanol 400 mL was added to the residue and the mixture followed by filtration. The filter cake was washed with 2-propanol (1 L). The solvent was distilled off at reduced pressure. Toluene (400 mL) was added and distillation was resumed in order to remove residual 2- propanol and possibly more water.
  • the progress of the reaction was monitored as follows: A sample of the reaction mixture was diluted 10 times with dry DMF, N;0- bis(trimethylsilyl)trifluoroacetamide (0.25 mL) was added to 0.5 mL of the sample in DMF and analyzed by GC. If the reaction was not complete the necessary amount of TIPS-CI was calculated and added and the stirring continued for another 20 hours.
  • the reaction mixture was transferred into aqueous 12.5% ammonium chloride (1 .7 kg) and the mixture was warmed to room temperature.
  • the aqueous layer (Aq. 1 ) was separated and the organic phase was washed with purified water (1 L).
  • the aqueous wash (Aq. 2) was separated and the organic phase was secured.
  • Aq.1 was washed with heptanes (0.6 kg).
  • the aqueous phase was separated and then discarded.
  • Aq. 2 was added to the organic phase and the mixture was stirred for 1 minute.
  • the aqueous phase was separated and discarded.
  • the two organic phases were combined and concentrated at reduced pressure at 50 °C. Heptanes (0.7 kg) was added to the residue and the resulting suspension was filtered.
  • the filter cake was washed with heptanes (0.2 kg), the combined filtrate was concentrated at reduced pressure at 50 °C, which gave 506 g crude product.
  • the crude product was dissolved in a mixture of heptanes and toluene (0.5 L, 3:1 ) and purified by column chromatography on silica gel (silica gel 60, 2.5 kg and heptanes/toluene 3:1 v/v).
  • the column was eluted with heptanes/toluene (3:1 , 5.0 L), heptanes/toluene (2:1 , 2.5 L), heptanes/toluene (3:1 , 2.5 L) and toluene (7.5 L).
  • Fractions of ⁇ 1 L were collected and fractions holding pure compound 2c were combined and concentrated and fractions holding mixtures of compound 2c and di-fluoro compound were combined and re-purified.
  • the mother organic phase was concentrated to dryness at reduced pressure at 55 °C and then added to the heptane wash.
  • the thus combined organic phases were washed with 5% aqueous sodium chloride.
  • the phases were separated and the aqueous phase washed with heptanes (0.2 L), then discarded.
  • the organic phase was concentrated at reduced pressure which gave 440 g of crude product.
  • a 3 L reaction flask set up with mechanical stirrer, thermometer and an addition funnel was filled with nitrogen.
  • the flask was charged with ethyl acetate (1000 g) and cooled to 10 °C.
  • Lithium tri-ferf-butoxyaluminium hydride (30% solution in THF, 35 g, 0.05 eq.) was added. Stirring at 10 °C was continued for 5-10 minutes and then compound 28f (370.0 g, 0.88 mol) was added. Further lithium tri-ferf-butoxyaluminium hydride (30% solution in THF, 933.8 g, 1 .10 mol, 1.25 eq.) was added over a period of 70 minutes while keeping the reaction temperature at 10 °C.
  • the reaction was quenched by pouring the reaction mixture onto a quench mixture (1 .45 kg (10% NaCI - 10% NH 4 CI in 3M HCI)) keeping the temperature at 10 - 15 °C.
  • the resulting suspension was warmed to 20 - 25 °C.
  • the aqueous was separated and discarded and the organic phase was washed with acidic water (1 .0 L + 10 mL of 3M HCI) followed by a wash with 25% sodium chloride (250 ml_).
  • the organic phase was concentrated to dryness, finally at p ⁇ 35 mbar and 45 °C.
  • reaction flask set up for mechanical stirring, temperature measurement and condenser was filled with nitrogen and charged with toluene (740 mL), compound 28g (41 1 .5 g, 0.88 mol) and thionyl chloride (174.0 g, 1 .46 mol, 1 .66 equivalents).
  • the reaction flask was placed on a water bath, pre-heated to 50 °C and DMF (0.50 mL) was added.
  • the top of the condenser was connected a cooled scrubber (700 g of 27.65% sodium hydroxide) and a steady flow of nitrogen was applied. The reaction started shortly after the DMF was added and it was followed by HPLC.
  • Chlorobenzene 200 g was added to the residue and the mixture was concentrated using the above conditions. The residue was again dissolved in chlorobenzene (200.0 g) and the mixture concentrated.
  • Residual toluene in Compound 28h (50 g, 1 13.3 mmol) was removed by distillation in vacuo from chlorobenzene. The residue from this co-evaporation was dissolved in 1 ,2-dichloroethane (200 mL), and this solution was charged to the solution of silylated nucleoside in chlorobenzene. Tin(IV)chloride (59.0 g, 226.6 mmol, 2 eq.) was added and the mixture was heated to reflux under nitrogen. The reaction mixture was stirred at reflux for 65 h.
  • reaction mixture was cooled to 5 °C, and ethyl acetate (99.8 g, 10 eq.) was added while keeping the temperature at 10-12 ⁇ C.
  • Total weight of mixture 601 .7 g._A quarter of this mixture (150.4 g, in theory 28.3 mmol) was charged to a 250 mL 3 necked round bottom flask, cooled to 5 °C, and
  • dichloromethane 147.5 g, 4 x vol. of EtOAc was added together with Celite (6.25 g).
  • the mixture was stirred for 20 min at 10 °C, then the temperature was adjusted to 25 °C and the mixture was stirred at this temperature for 30 min.
  • the suspension was filtered on a pad of Celite (12.5 g) and the filter cake was washed with dichloromethane (190 mL). The combined filtrate and washings were concentrated to dryness by distillation in vacuo at 60 °C.
  • Dichloromethane (86 mL) was added to the residue then toluene (62 mL). The content of dichloromethane was removed by distillation in vacuo at 50 °C. The resulting suspension was stirred at room temperature for 17 h whereafter the crude title compound was isolated by filtration. The filter cake was washed with toluene (25 mL) and the wet product was dried in an air ventilated dryer at 40 °C, which gave title compound as a solid (5.56 g, 31 .7%).
  • Step k) 1 -((2R,3S,4R,5R)-3-Chloro-3-fluoro-4-hvdroxy-5-(hvdroxymethyl)tetrahvdrofuran-2- yl)pyrimidine-2,4(1 H,3H)-dione (28k)
  • the acidic aqueous phase was extracted with Me-THF (4 x 40 mL), and the combined organic phases were concentrated to dryness by distillation in vacuo at 40 °C.
  • Isopropyl acetate (80 mL) was added to the residue, and the turbid mixture was concentrated in vacuo at 60 °C.
  • Isopropyl acetate (40 mL) was added and the distillation in vacuo was continued.
  • Isopropyl acetate (10 mL) was added to the resulting thick suspension. The suspension was cooled to room temperature and stirred for 30 min. Crude title compound was collected by filtration, and the filter cake was washed with isopropyl acetate (2 x 4 mL).
  • Step I) (S)-lsopropyl 2-(((S)-(((2R.3R.4S.5R)-4-chloro-5-(2.4-dioxo-3.4-dihvdropyrimidin-1 (2H)- yl)-4-fluoro-3-hvdroxytetrahvdrofuran-2-yl)methoxy)(phenoxy)phosphoryl)amino)propanoate (28) THF (0.07% water, 12 mL) was added to Compound 28k (500 mg, 1 .78 mmol) and the solution was cooled to -10 °C under nitrogen.
  • the compounds of formula (I) may be examined for activity in the inhibition of HCV RNA replication in a cellular assay aimed at identifying compounds that inhibit a HCV functional cellular replicating cell line, also known as HCV replicons.
  • a suitable cellular assay is based on a bicistronic expression construct, as described by Lohmann et al. (1999), Science vol. 285 pp. 1 10-1 13 with modifications described by Krieger et al. (2001 ), Journal of Virology 75: 4614- 4624, in a multi-target screening strategy.
  • the assay utilizes the stably transfected cell line Huh-7 luc/neo (hereafter referred to as Huh- Luc).
  • This cell line harbors an RNA encoding a bicistronic expression construct comprising the wild type NS3-NS5B regions of HCV type 1 b translated from an Internal Ribosome Entry Site (IRES) from encephalomyocarditis virus (EMCV), preceded by a reporter portion (FfL- luciferase), and a selectable marker portion (neo R , neomycine phosphotransferase).
  • IRS Internal Ribosome Entry Site
  • EMCV encephalomyocarditis virus
  • FfL- luciferase reporter portion
  • neo R neomycine phosphotransferase
  • the replicon cells are plated in 384 well plates in the presence of the test and control compounds which are added in various concentrations. Following an incubation of three days, HCV replication is measured by assaying luciferase activity (using standard luciferase assay substrates and reagents and a Perkin Elmer ViewLuxTM ultraHTS microplate imager). Replicon cells in the control cultures have high luciferase expression in the absence of any inhibitor. The inhibitory activity of a compound on luciferase activity is monitored on the Huh-Luc cells, enabling a dose-response curve for each test compound. EC 50 values are then calculated, which value represents the amount of the compound required to decrease the level of detected luciferase activity by 50%, or more specifically, the ability of the genetically linked HCV replicon RNA to replicate. Enzyme assay
  • the compounds of the invention are metabolised by cellular kinases in target tissues to the 5'-trisphosphate. It is this triphosphate which is believed to be the antivirally active species.
  • the enzyme assay described here may be used to confirm that compound of the invention are antivirally active as the 5'-triphosphate metabolite.
  • the enzyme assay measures the inhibitory effect of triphosphate compounds in an HCV NS5B- 21 (21 -aminoacid C-terminally truncated version) SPA assay (scintillation proximity assay).
  • the assay is performed by evaluating the amount of radiolabeled ATP incorporated by HCV NS5B- 21 into newly synthesized RNA using an heterogeneous biotinylated RNA template.
  • IC 50 values the compounds are tested at various concentrations in a final volume of 100 ⁇ of reaction mixture. The reaction is stopped by addition of 0.5M EDTA solution.
  • the samples are transferred into flashplates precoated with streptavidin.
  • the incorporated radioactivity is quantified using a scintillation counter (Wallac Microbeta Trilux).
  • Biotinylated RNA template with a sequence of
  • Enzyme HCV NS5B-21 , made up to 500 ⁇ g/ml in water. Replizyme
  • Radiolabeled 3 H-ATP (cat. no TRK747) GE Healthcare
  • Buffer 20 mM tris-HCI, 100 mM ammonium acetate, 20 mM NaCI, 2.5 mM MnCI 2 ,
  • Enzyme NS5B-21 (500 ⁇ g/m ⁇ ) 2 ⁇ g/ml
  • the assay should include enzyme controls (about four, containing 1 ⁇ DMSO instead of inhibitor) and background control containing all ingredients except template. Compounds are serially diluted in DMSO on a separate dilution plate to 1 0Ox the final desired assay concentrations.
  • Sufficient reaction mixture for the number of wells to be used is made up according to the table below and 90 ⁇ /well is added to a 96 well polyproylene plate. 1 ⁇ of compound in DMSO from the dilution plate is added to each well, except the enzyme control wells and background control wells to which 1 ⁇ DMSO is added.
  • AverageEnzymeControlCPM - BackgroundCPM Background Reaction buffer without template.
  • IC 50 is determined using Graphpad Prism. Plot Compound concentration in Log versus percentage inhibition. Fit the curve with nonlinear regression to the Log (Inhibitor) versus Response equation.
  • the inhibition of HCV replication exhibited by the compounds of the invention were tested in the above described replicon assay.
  • the compounds showed sub micromolar activity, with a cell toxicity in the Huh-Luc cell line being in excess of 50 ⁇ .
  • the EC 50 values are presented in Table 1 .
  • compositions of the invention may be examined for activity in the inhibition of HCV RNA replication in a cellular assay aimed at identifying compounds that inhibit a HCV functional cellular replicating cell line, also known as HCV replicons.
  • a suitable cellular assay is based on a bicistronic expression construct, as described by Lohmann et al. (1999), Science vol. 285 pp. 1 10-1 13 with modifications described by Krieger et al. (2001 ), Journal of Virology 75: 4614- 4624, in a multi-target screening strategy.
  • the assay utilizes the stably transfected cell line Huh-7 luc/neo (hereafter referred to as Huh- Luc).
  • This cell line harbors an RNA encoding a bicistronic expression construct comprising the wild type NS3-NS5B regions of HCV type 1 b translated from an Internal Ribosome Entry Site (IRES) from encephalomyocarditis virus (EMCV), preceded by a reporter portion (FfL- luciferase), and a selectable marker portion (neo R , neomycine phosphotransferase).
  • IRS Internal Ribosome Entry Site
  • EMCV encephalomyocarditis virus
  • FfL- luciferase reporter portion
  • neo R neomycine phosphotransferase
  • HCV RNA stably transfected replicon cells that express HCV RNA, which replicates autonomously and to high levels, encoding inter alia luciferase, are used for screening the antiviral compounds.
  • the replicon cells are plated in 384 well plates in the presence of the test and control compounds which are added in various concentrations. Following an incubation of three days, HCV replication is measured by assaying luciferase activity (using standard luciferase assay substrates and reagents and a Perkin Elmer ViewLuxTM ultraHTS microplate imager).
  • Replicon cells in the control cultures have high luciferase expression in the absence of any inhibitor.
  • the inhibitory activity of a compound on luciferase activity is monitored on the Huh-Luc cells, enabling a dose-response curve for each test compound.
  • EC 50 values are then calculated, which value represents the amount of the compound required to decrease the level of detected luciferase activity by 50%, or more specifically, the ability of the genetically linked HCV replicon RNA to replicate.
  • Compound 1A shows an EC 50 value of 0.055 uM (n >10), with a cell toxicity in the Huh-Luc cell line being in excess of 50 ⁇ .
  • the above described replicon assay can further be used to assess synergistic, additive or antagonistic effects of HCV antiviral on each other using the CI isobol method, which method provides a quantitative assessment of synergism between drugs.
  • the combination index (CI) of a combined drug treatment is defined as:
  • D 1 and D 2 are the doses of drug 7 and drug 2, respectively, in the combination; Dx 1 and D 2 are the doses of drug 7 and drug 2, respectively, in the combination; Dx 1 and D 2 are the doses of drug 7 and drug 2, respectively, in the combination; Dx 1 and D 2 are the doses of drug 7 and drug 2, respectively, in the combination; Dx 1 and D 2 are the doses of drug 7 and drug 2, respectively, in the combination; Dx 1 and
  • Dx 2 each is the dose of a treatment with only drug 7 and drug 2 that would give the same effect as that of the combination, respectively.
  • the doses Dx l and Dx 2 need to be estimated from the dose-effect data of single drug treatments.
  • the two drugs under assessment are presented in serial dilution series on the different axes of a well plate - see the attached Figure 1 .
  • Commercially available computer software such as MacSynergy is used to calculate and Bonferroni-adjust the synergy volumes
  • Sofosbuvir is marketed in several countries for the treatment of HCV, predominantly against genotypes 1 and 4.
  • the structure of sofosbuvir is:
  • sofosbuvir differs from the compound of present Example 2, in that it possesses a beta-methyl group at the 2'-position, whereas the compounds of the invention have a beta-chloro substituent at this position.
  • sofosbuvir in the Fission phase III clinical trials reported in Lawitz et al., N. Eng. J. Med., 2013; 368:1878-87, "Response rates in the sofosbuvir -ribavirin group were lower among patients with genotype 3 infection than amongst those with genotype 2 infection (56% vs. 97%)".
  • a threefold better potency for the compound of the invention relative to sofosbuvir is expected to markedly improve viral response rates in the clinic.
  • Example 2 The compound of Example 2 was further evaluated to assess the antiviral activity against genotypes 1 -6 of HCV, both wild type and a number of clinically relevant mutant strains.
  • the result of the evaluation together with the average EC 50 of the genotypes and the corresponding values for sofosbuvir are summarised in Tables 2 and 3.
  • Example 2 has a significant improved potency as compared to sofosbuvir against HCV GT3a both in the wild type strain and in two clinically relevant mutant strains, while keeping the good potency against the other genotypes.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • General Health & Medical Sciences (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • Medicinal Chemistry (AREA)
  • Animal Behavior & Ethology (AREA)
  • Epidemiology (AREA)
  • Engineering & Computer Science (AREA)
  • Immunology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Molecular Biology (AREA)
  • Organic Chemistry (AREA)
  • Virology (AREA)
  • Zoology (AREA)
  • Oncology (AREA)
  • Communicable Diseases (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Biochemistry (AREA)
  • Biotechnology (AREA)
  • Genetics & Genomics (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)

Abstract

L'invention concerne l'utilisation d'un composé représenté par la formule (1A), ou un sel pharmaceutiquement acceptable de ce dernier, dans le traitement contre le VHC chez le mammifère ou l'homme, le composé de formule 1A étant administré en association avec un autre antiviral contre le VHC sélectionné entre : a) l'asunaprevir, le daclatasvir et/ou le beclabuvir ; ou b) le simeprevir et/ou le JNJ56914845 ; ou c) un inhibiteur de protéase du VHC sélectionné entre le paritaprevir ou l'ABT493, et un inhibiteur de NS5A sélectionné entre l'ombitasvir ou l'ABT-530 ; ou d) l'alisporavir ; ou e) l'EDP-239 ; ou f) l'odalasvir et/ou le sovaprevir ; ou g) le grazoprevir et/ou l'elbasvir.
EP16759215.3A 2015-03-02 2016-03-02 Dérivés nucléotidiques qui sont des inhibiteurs du vhc pour une utilisation dans le traitement de l'hépatite c Withdrawn EP3271371A4 (fr)

Applications Claiming Priority (9)

Application Number Priority Date Filing Date Title
SE1550242 2015-03-02
SE1550258 2015-03-04
SE1550257 2015-03-04
SE1550261 2015-03-04
SE1550260 2015-03-04
SE1550256 2015-03-04
SE1550259 2015-03-04
SE1550265 2015-03-04
PCT/SE2016/050166 WO2016140615A1 (fr) 2015-03-02 2016-03-02 Dérivés nucléotidiques qui sont des inhibiteurs du vhc pour une utilisation dans le traitement de l'hépatite c

Publications (2)

Publication Number Publication Date
EP3271371A1 true EP3271371A1 (fr) 2018-01-24
EP3271371A4 EP3271371A4 (fr) 2018-12-05

Family

ID=56851805

Family Applications (1)

Application Number Title Priority Date Filing Date
EP16759215.3A Withdrawn EP3271371A4 (fr) 2015-03-02 2016-03-02 Dérivés nucléotidiques qui sont des inhibiteurs du vhc pour une utilisation dans le traitement de l'hépatite c

Country Status (4)

Country Link
EP (1) EP3271371A4 (fr)
CN (1) CN107531739A (fr)
HK (1) HK1243712A1 (fr)
WO (1) WO2016140615A1 (fr)

Families Citing this family (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN109715161A (zh) * 2016-09-23 2019-05-03 艾伯维公司 剂量调整
CN109762040A (zh) * 2019-01-14 2019-05-17 歌礼生物制药(杭州)有限公司 一种核苷类ns5b聚合酶抑制剂的制备方法

Family Cites Families (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
TWI576352B (zh) * 2009-05-20 2017-04-01 基利法瑪席特有限責任公司 核苷磷醯胺
AU2013329521B2 (en) * 2012-10-08 2018-04-19 Centre National De La Recherche Scientifique 2'-chloro nucleoside analogs for HCV infection
WO2014078436A1 (fr) * 2012-11-14 2014-05-22 Idenix Pharmaceuticals, Inc. Ester de d-alanine d'analogue de sp-nucléoside
UA117375C2 (uk) * 2013-09-04 2018-07-25 Медівір Аб Інгібітори полімерази hcv
US20170198005A1 (en) * 2013-11-27 2017-07-13 Idenix Pharmaceuticals Llc 2'-dichloro and 2'-fluoro-2'-chloro nucleoside analogues for hcv infection

Also Published As

Publication number Publication date
HK1243712A1 (zh) 2018-07-20
EP3271371A4 (fr) 2018-12-05
WO2016140615A1 (fr) 2016-09-09
CN107531739A (zh) 2018-01-02

Similar Documents

Publication Publication Date Title
US10106571B2 (en) HCV polymerase inhibitors
US20130143835A1 (en) HCV Polymerase Inhibitors
WO2013084165A1 (fr) Inhibiteurs de vhc polymérase
RU2534613C2 (ru) Алкил 2-{ [(2r,3s,5r)-5-(4-амино-2-оксо-2н-пиримидин-1-ил)- -гидрокси-тетрагидро-фуран-2-илметокси]-фенокси-фосфориламино} -пропионаты, нуклеозидные ингибиторы рнк-полимеразы hcv ns5b, способы их получения и применения
KR101687084B1 (ko) Hcv rna 복제의 억제제로서 4'-아지도, 3'-플루오로 치환된 뉴클레오시드 유도체
JP5683480B2 (ja) ウラシルシクロプロピルヌクレオチド
JP2019524795A (ja) 置換ヌクレオシド、ヌクレオチド、及びそのアナログ
MX2014007023A (es) Derivados nucleosidos con sustitucion 2',4'-difluoro-2'-metilo como inhibidores de la replicacion de arn del vhc.
US20160271160A1 (en) Hcv polymerase inhibitors
WO2016140615A1 (fr) Dérivés nucléotidiques qui sont des inhibiteurs du vhc pour une utilisation dans le traitement de l'hépatite c
TWI639613B (zh) Hcv聚合酶抑制劑
TWI675664B (zh) Hcv聚合酶抑制劑

Legal Events

Date Code Title Description
STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: THE INTERNATIONAL PUBLICATION HAS BEEN MADE

PUAI Public reference made under article 153(3) epc to a published international application that has entered the european phase

Free format text: ORIGINAL CODE: 0009012

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: REQUEST FOR EXAMINATION WAS MADE

17P Request for examination filed

Effective date: 20170913

AK Designated contracting states

Kind code of ref document: A1

Designated state(s): AL AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HR HU IE IS IT LI LT LU LV MC MK MT NL NO PL PT RO RS SE SI SK SM TR

AX Request for extension of the european patent

Extension state: BA ME

DAV Request for validation of the european patent (deleted)
DAX Request for extension of the european patent (deleted)
RAP1 Party data changed (applicant data changed or rights of an application transferred)

Owner name: MEDIVIR AB

Owner name: MEDIVIR AKTIEBOLAG

RAP1 Party data changed (applicant data changed or rights of an application transferred)

Owner name: MEDIVIR AKTIEBOLAG

A4 Supplementary search report drawn up and despatched

Effective date: 20181029

RIC1 Information provided on ipc code assigned before grant

Ipc: A61K 31/407 20060101ALI20181023BHEP

Ipc: A61K 31/4709 20060101ALI20181023BHEP

Ipc: A61K 38/13 20060101ALI20181023BHEP

Ipc: A61K 38/55 20060101ALI20181023BHEP

Ipc: A61P 31/14 20060101ALI20181023BHEP

Ipc: A61K 31/439 20060101ALI20181023BHEP

Ipc: C07H 19/10 20060101ALI20181023BHEP

Ipc: A61K 31/513 20060101ALI20181023BHEP

Ipc: A61K 38/20 20060101ALI20181023BHEP

Ipc: A61K 38/21 20060101ALI20181023BHEP

Ipc: A61K 31/4725 20060101ALI20181023BHEP

Ipc: A61K 31/498 20060101ALI20181023BHEP

Ipc: A61K 31/4178 20060101ALI20181023BHEP

Ipc: A61K 31/7072 20060101AFI20181023BHEP

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: THE APPLICATION IS DEEMED TO BE WITHDRAWN

18D Application deemed to be withdrawn

Effective date: 20190528