WO2010011566A1 - Macrocyclic quinoxaline compounds as hcv ns3 protease inhibitors - Google Patents

Macrocyclic quinoxaline compounds as hcv ns3 protease inhibitors Download PDF

Info

Publication number
WO2010011566A1
WO2010011566A1 PCT/US2009/050915 US2009050915W WO2010011566A1 WO 2010011566 A1 WO2010011566 A1 WO 2010011566A1 US 2009050915 W US2009050915 W US 2009050915W WO 2010011566 A1 WO2010011566 A1 WO 2010011566A1
Authority
WO
WIPO (PCT)
Prior art keywords
compound
hcv
methyl
compounds
solution
Prior art date
Application number
PCT/US2009/050915
Other languages
French (fr)
Inventor
Steven Harper
Vincenzo Summa
Nigel J. Liverton
Johh A. Mccauley
Original Assignee
Merck & Co., Inc.
Istituto Di Ricerche Di Biologia Molecolare P. Angeletti S.P.A.
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
Family has litigation
First worldwide family litigation filed litigation Critical https://patents.darts-ip.com/?family=41130248&utm_source=google_patent&utm_medium=platform_link&utm_campaign=public_patent_search&patent=WO2010011566(A1) "Global patent litigation dataset” by Darts-ip is licensed under a Creative Commons Attribution 4.0 International License.
Priority to BRPI0916235A priority Critical patent/BRPI0916235B8/en
Priority to MEP-2012-463A priority patent/ME02024B/en
Priority to EP09790553A priority patent/EP2310095B1/en
Priority to EA201170241A priority patent/EA019327B1/en
Priority to KR1020117003982A priority patent/KR101313675B1/en
Priority to SI200930359T priority patent/SI2310095T1/en
Priority to PL09790553T priority patent/PL2310095T3/en
Priority to AU2009274190A priority patent/AU2009274190B2/en
Priority to CA2731177A priority patent/CA2731177C/en
Priority to ES09790553T priority patent/ES2392611T3/en
Priority to RS20120463A priority patent/RS52534B/en
Application filed by Merck & Co., Inc., Istituto Di Ricerche Di Biologia Molecolare P. Angeletti S.P.A. filed Critical Merck & Co., Inc.
Priority to NZ590638A priority patent/NZ590638A/en
Priority to MX2011000826A priority patent/MX2011000826A/en
Priority to JP2011520110A priority patent/JP4920797B2/en
Priority to CN200980137118.6A priority patent/CN102159285B/en
Priority to DK09790553.3T priority patent/DK2310095T3/en
Publication of WO2010011566A1 publication Critical patent/WO2010011566A1/en
Priority to IL210580A priority patent/IL210580A/en
Priority to TN2011000014A priority patent/TN2011000014A1/en
Priority to MA33556A priority patent/MA32502B1/en
Priority to HRP20120866AT priority patent/HRP20120866T1/en
Priority to FR16C1027C priority patent/FR16C1027I2/en
Priority to LTPA2016049C priority patent/LTC2310095I2/en
Priority to NL300857C priority patent/NL300857I2/nl
Priority to LU00002C priority patent/LUC00002I2/fr
Priority to NO2017004C priority patent/NO2017004I1/en
Priority to CY2017005C priority patent/CY2017005I2/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D241/00Heterocyclic compounds containing 1,4-diazine or hydrogenated 1,4-diazine rings
    • C07D241/36Heterocyclic compounds containing 1,4-diazine or hydrogenated 1,4-diazine rings condensed with carbocyclic rings or ring systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K5/00Peptides containing up to four amino acids in a fully defined sequence; Derivatives thereof
    • C07K5/04Peptides containing up to four amino acids in a fully defined sequence; Derivatives thereof containing only normal peptide links
    • C07K5/08Tripeptides
    • C07K5/0802Tripeptides with the first amino acid being neutral
    • C07K5/0804Tripeptides with the first amino acid being neutral and aliphatic
    • C07K5/0808Tripeptides with the first amino acid being neutral and aliphatic the side chain containing 2 to 4 carbon atoms, e.g. Val, Ile, Leu
    • 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
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/16Drugs for disorders of the alimentary tract or the digestive system for liver or gallbladder disorders, e.g. hepatoprotective agents, cholagogues, litholytics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • 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
    • 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
    • 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
    • A61P31/20Antivirals for DNA viruses
    • A61P31/22Antivirals for DNA viruses for herpes viruses
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D403/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00
    • C07D403/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings
    • C07D403/12Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings linked by a chain containing hetero atoms as chain links

Definitions

  • the present invention relates to macrocyclic compounds that are useful as inhibitors of the hepatitis C virus (HCV) NS3 protease, the synthesis of such compounds, and the use of such compounds for treating HCV infection and/or reducing the likelihood or severity of HCV infection.
  • HCV hepatitis C virus
  • HCV infection is a major health problem that leads to chronic liver disease, such as cirrhosis and hepatocellular carcinoma, in a substantial number of infected individuals.
  • Current treatments for HCV infection include immunotherapy with recombinant interferon- ⁇ alone or in combination with the nucleoside analog ribavirin.
  • NS3 metal loprotease
  • NS3 serine protease
  • NS3 a helicase
  • NS5B RNA- dependent RNA polymerase
  • the NS3 protease is located in the N-terminal domain of the NS3 protein.
  • NS4A provide a cofactor for NS3 activity.
  • the present invention relates to a macrocyclic compound of formula (I) and pharmaceutically acceptable salts thereof.
  • the compound and its salts are HCV NS3 protease inhibitors.
  • the compound and its salts have therapeutic and research applications.
  • a first aspect of the present invention describes a compound of formula (I), or a pharmaceutical acceptable salt thereof:
  • the present invention also includes pharmaceutical compositions containing a compound of the present invention and methods of preparing such pharmaceutical compositions.
  • the present invention further includes methods of treating or reducing the likelihood or severity of HCV infection.
  • the present invention includes a compound of formula (I), and pharmaceutically acceptable salts thereof.
  • the compound and its pharmaceutically acceptable salts are useful in the inhibition of HCV NS3 protease, the treatment of HCV infection and/or the reduction of the likelihood or severity of an HCV infection.
  • Prophylactic applications include, for example, treatment after suspected past exposure to HCV by such means as blood transfusion, exchange of body fluids, bites, accidental needle stick, or exposure to patient blood during surgery.
  • the compounds and salts may be the primary active therapeutic agent.
  • the compound may be combined with other therapeutic agents including but not limited to other HCV antivirals, anti-infectives, immunomodulators, antibiotics or vaccines.
  • NS3 inhibitors are also useful in the preparation and execution of screening assays for antiviral compounds.
  • such compounds can be used to isolate enzyme mutants, which are excellent screening tools for more powerful antiviral compounds.
  • the compounds may be used to establish or determine the binding site of other antivirals to HCV protease, e.g., by competitive inhibition.
  • Example 2 the formula (I) compound was compared to the compound of Examples 110 and 118 of WO 2008/057209, and has several advantages. WO 2008/057209 is not admitted to be prior art to the claimed invention.
  • composition comprising an effective amount of a compound of formula (I) and a pharmaceutically acceptable carrier.
  • composition of (a) further comprising a second therapeutic agent selected from the group consisting of HCV antiviral agents, immunomodulators, and anti-infective agents.
  • HCV antiviral agent is an antiviral selected from the group consisting of HCV protease inhibitors and HCV NS5B polymerase inhibitors.
  • a pharmaceutical combination which is (i) a compound of formula (I) and (ii) a second therapeutic agent selected from the group consisting of HCV antiviral agents, immunomodulators, and anti-infective agents; wherein the compound of formula (I) and the second therapeutic agent are each employed in an amount that renders the combination effective for inhibiting HCV NS3 protease, or for treating HCV infection and/or reducing the likelihood or severity of HCV infection.
  • HCV antiviral agent is an antiviral selected from the group consisting of HCV protease inhibitors and HCV NS5B polymerase inhibitors.
  • HCV antiviral agent is an antiviral selected from the group consisting of HCV protease inhibitors and HCV NS5B polymerase inhibitors.
  • (j) A method of inhibiting HCV NS3 protease in a subject in need thereof which comprises administering to the subject the pharmaceutical composition of (a), (b), or (c) or the combination of (d) or (e).
  • (k) A method of treating HCV infection and/or reducing the likelihood or severity of HCV infection in a subject in need thereof which comprises administering to the subject the pharmaceutical composition of (a), (b), or (c) or the combination of (d) or (e).
  • Reference to a compound also includes stable complexes of the compound such as a stable hydrate.
  • a “stable” compound is a compound which can be prepared and isolated and whose structure and properties remain or can be caused to remain essentially unchanged for a period of time sufficient to allow use of the compound for the purposes described herein (e.g., therapeutic or prophylactic administration to a subject).
  • administration means providing the compound or a prodrug of the compound to the individual in need of treatment
  • administration means providing the compound or a prodrug of the compound to the individual in need of treatment
  • active agents e.g., antiviral agents useful for treating HCV infection
  • administration and its variants are each understood to include concurrent and sequential provision of the compound or salt and other agents.
  • the compounds of the present invention may be administered in the form of pharmaceutically acceptable salts.
  • pharmaceutically acceptable salt refers to a salt of the parent compound which has activity and which is not biologically or otherwise undesirable (e.g., is neither toxic nor otherwise deleterious to the recipient thereof).
  • Suitable salts include acid addition salts which may, for example, be formed by mixing a solution of a compound with a solution of a pharmaceutically acceptable acid such as hydrochloric acid, sulfuric acid, acetic acid, trifluoroacetic acid, or benzoic acid.
  • Compounds carrying an acidic moiety can be mixed with suitable pharmaceutically acceptable salts to provide, for example, alkali metal salts (e.g., sodium or potassium salts), alkaline earth metal salts (e.g., calcium or magnesium salts), and salts formed with suitable organic ligands such as quaternary ammonium salts.
  • suitable pharmaceutically acceptable salts for example, alkali metal salts (e.g., sodium or potassium salts), alkaline earth metal salts (e.g., calcium or magnesium salts), and salts formed with suitable organic ligands such as quaternary ammonium salts.
  • suitable pharmaceutically acceptable esters can be employed to modify the solubility or hydrolysis characteristics of the compound.
  • prodrug is intended to encompass an inactive drug form or compound that is converted into an active drug form or compound by the action of enzymes, chemicals or metabolic processes in the body of an individual to whom it is administered.
  • composition is intended to encompass a product comprising the specified ingredients, as well as any product which results, directly or indirectly, from combining the specified ingredients.
  • pharmaceutically acceptable is meant that the ingredients of the pharmaceutical composition must be compatible with each other and not deleterious to the recipient thereof.
  • subject refers to an animal, preferably a mammal, most preferably a human, who has been the object of treatment, observation or experiment.
  • an effective amount indicates a sufficient amount to exert a therapeutic or prophylactic effect.
  • an effective amount is sufficient to achieve one or more of the following effects: reduce the ability of HCV to replicate, reduce HCV load, and increase viral clearance.
  • an effective amount is sufficient to achieve one or more of the following: a reduced susceptibility to HCV infection, and a reduced ability of the infecting virus to establish persistent infection for chronic disease.
  • the compounds of the present invention can be administered by means that produces contact of the active agent with the agent's site of action. They can be administered by conventional means available for use in conjunction with pharmaceuticals, either as individual therapeutic agents or in a combination of therapeutic agents. They can be administered alone, but typically are administered with a pharmaceutical carrier selected on the basis of the chosen route of administration and standard pharmaceutical practice.
  • Compounds can, for example, be administered by one or more of the following routes: orally, parenterally (including subcutaneous injections, intravenous, intramuscular, intrastemal injection or infusion techniques), by inhalation (such as in a spray form), or rectally, in the form of a unit dosage of a pharmaceutical composition containing an effective amount of the compound and conventional non-toxic pharmaceutically-acceptable carriers, adjuvants and vehicles.
  • Liquid preparations suitable for oral administration e.g., suspensions, syrups, elixirs and the like
  • Solid preparations suitable for oral administration e.g.
  • powders, pills, capsules and tablets can be prepared according to techniques known in the art and can employ such solid excipients as starches, sugars, kaolin, lubricants, binders, disintegrating agents and the like.
  • Parenteral compositions can be prepared according to techniques known in the art and typically employ sterile water as a carrier and optionally other ingredients, such as solubility aids.
  • injectable solutions can be prepared according to methods known in the art wherein the carrier comprises a saline solution, a glucose solution or a solution containing a mixture of saline and glucose. Further guidance for methods suitable for use in preparing pharmaceutical compositions of the present invention and of ingredients suitable for use in said compositions is provided in Remington's Pharmaceutical Sciences, 20 th edition (ed. A. R. Gennaro, Mack Publishing Co., 2000).
  • the compounds of this invention can be administered orally in a dosage range of 0.001 to 1000 mg/kg of mammal (e.g., human) body weight per day in a single dose or in divided doses.
  • mammal e.g., human
  • One dosage range is 0.01 to 500 mg/kg body weight per day orally in a single dose or in divided doses.
  • Another dosage range is 0.1 to 100 mg/kg body weight per day orally in single or divided doses.
  • the compositions can be provided in the form of tablets or capsules containing 1.0 to 500 mg of the active ingredient, particularly 1, 5, 10, 15, 20, 25, 50, 75, 100, 150, 200, 250, 300, 400, 500, and 750 mg of the active ingredient for the symptomatic adjustment of the dosage to the patient to be treated.
  • the specific dose level and frequency of dosage for any particular patient may be varied and will depend upon a variety of factors including the activity of the specific compound employed, the metabolic stability and length of action of that compound, the age, body weight, general health, sex, diet, mode and time of administration, rate of excretion, drug combination, the severity of the particular condition, and the host undergoing therapy.
  • the quinoxaline macrocyclic compounds described herein can be used in a combination treatment involving one or more additional therapeutic agents.
  • Additional therapeutic agents include those also targeting HCV, targeting a different disease causing agent, or those enhancing the immune system.
  • Agents enhancing the immune system include those generally enhancing an immune system function and those producing a specific immune response against HCV.
  • Additional therapeutic agents targeting HCV include agents targeting NS3 and agents targeting other HCV activities such as NS5A and NS5B, and agents targeting host cell activities involved in HCV replication.
  • HCV inhibitors are described in different publications. Macrocyclic compounds useful as inhibitors the HCV protease inhibitors are described in WO 06/119061, WO 7/015785, WO 7/016441, WO 07/148135, WO 08/051475, WO 08/051477, WO 08/051514, WO 08/057209.
  • HCV NS3 protease inhibitors are disclosed in International Patent Application Publications WO 98/22496, WO 98/46630, WO 99/07733, WO 99/07734, WO 99/38888, WO 99/50230, WO 99/64442, WO 00/09543, WO 00/59929, WO 02/48116, WO 02/48172, British Patent No. GB 2 337 262, and U.S. Patent No. 6,323,180.
  • therapeutic agents that may be present in a combination include ribavirin, levovirin, viramidine, thymosin alpha- 1, interferon- ⁇ , interferon- ⁇ , pegylated interferon- ⁇ (peginterferon- ⁇ ), a combination of interferon- ⁇ and ribavirin, a combination of peginterferon- ⁇ and ribavirin, a combination of interferon- ⁇ and levovirin, and a combination of peginterferon- ⁇ and levovirin.
  • Interferon- ⁇ includes recombinant interferon- ⁇ 2a (such as ROFERON interferon available from Hoffmann-LaRoche, Nutley, NJ), pegylated interferon- ⁇ 2a (PEGASYS), interferon- ⁇ 2b (such as INTRON-A interferon available from Schering Corp., Kenilworth, NJ), pegylated interferon- ⁇ z2b (PEGINTRON), a recombinant consensus interferon (such as interferon al ⁇ hacon-1), and a purified interferon- ⁇ product.
  • Amgen's recombinant consensus interferon has the brand name INFERGEN.
  • Levovirin is the L-enantiomer of ribavirin which has shown immunomodulatory activity similar to ribavirin.
  • Viramidine represents an analog of ribavirin disclosed in WO 01/60379.
  • the individual components of the combination can be administered separately at different times during the course of therapy or concurrently in divided or single combination forms.
  • Ribavirin, levovirin, and viramidine may exert their anti-HCV effects by modulating intracellular pools of guanine nucleotides via inhibition of the intracellular enzyme inosine monophosphate dehydrogenase (IMPDH).
  • IMPDH inosine monophosphate dehydrogenase
  • Ribavirin is readily phosphorylated intracellularly and the monophosphate derivative is an inhibitor of IMPDH.
  • inhibition of IMPDH represents another useful target for the discovery of inhibitors of HCV replication.
  • the compounds of the present invention may also be administered in combination with an inhibitor of IMPDH, such as VX-497, which is disclosed in International Patent Application Publications WO 97/4121 1 and WO 01/00622; another IMPDH inhibitor, such as that disclosed in WO 00/25780; or mycophenolate mofetil. See A.C. Allison and E.M. Eugui, 44 (Suppl.) Agents Action 165 (1993).
  • an inhibitor of IMPDH such as VX-497, which is disclosed in International Patent Application Publications WO 97/4121 1 and WO 01/00622
  • another IMPDH inhibitor such as that disclosed in WO 00/25780
  • mycophenolate mofetil See A.C. Allison and E.M. Eugui, 44 (Suppl.) Agents Action 165 (1993).
  • the compounds of the present invention may also be administered in combination with the antiviral agent amantadine (1-aminoadamantane).
  • amantadine 1-aminoadamantane
  • the compounds of the present invention may also be administered in combination with the antiviral agent polymerase inhibitor R7128 (Roche).
  • the compounds of the present invention may also be combined for the treatment of HCV infection with antiviral 2'-C-branched ribonucleosides disclosed in R. E. Harry-O'Kuru et al., 62 J. Org. Chem. 1754-59 (1997); M. S. Wolfe et al., 36 Tet. Lett. 761 1-14 (1995); U.S. Patent No. 3,480,613; and International Patent Application Publications WO 01/90121, WO 01/92282, WO 02/32920, WO 04/002999, WO 04/003000 and WO 04/002422; the contents of each of which are incorporated by reference in their entirety.
  • Such 2'-C-branched ribonucleosides include, but are not limited to, 2'-C-methyl-cytidine, 2'-C-methyl-uridine, 2'-C- methyl-adenosine, 2'-C-methyl-guanosine, and 9-(2-C-methyl- ⁇ -D-ribofuranosyl)-2,6- diaminopurine, and the corresponding amino acid ester of the ribose C-2', C-3', and C-5' hydroxyls and the corresponding optionally substituted cyclic 1,3 -propanediol esters of the 5'- phosphate derivatives.
  • the compounds of the present invention may also be combined for the treatment of HCV infection with other nucleosides having anti-HCV properties, such as those disclosed in International Patent Application Publications WO 02/51425, WO 01/79246, WO 02/32920, WO 02/48165 and WO2005/003147 (including Rl 656, (2'/?)-2'-deoxy-2'-fluoro-2'-C- methylcytidine, shown as compounds M on page 77); WO 01/68663; WO 99/43691; WO 02/18404 and WO2006/021341, and U.S.
  • nucleosides having anti-HCV properties such as those disclosed in International Patent Application Publications WO 02/51425, WO 01/79246, WO 02/32920, WO 02/48165 and WO2005/003147 (including Rl 656, (2'/?)-2'-deoxy-2'-fluoro-2'-C- methylcytidine, shown as compounds M on page 77); WO 01/6
  • Patent Application Publication US 2005/0038240 including 4'-azido nucleosides such as Rl 626, 4'-azidocytidine; U.S. Patent Application Publications US 2002/0019363, US 2003/0236216, US 2004/0006007 and US 2004/0063658; and International Patent Application Publications WO 02/100415, WO 03/026589, WO 03/026675, WO 03/093290, WO 04/011478, WO 04/013300 and WO 04/028481 ; the content of each is incorporated herein by reference in its entirety.
  • the compounds of the present invention may also be administered in combination with an agent that is an inhibitor of HCV NS5B polymerase.
  • HCV NS5B polymerase inhibitors that may be used as combination therapy include, but are not limited to, those disclosed in International Patent Application Publications WO 02/057287, WO 02/057425, WO 03/068244, WO 2004/000858, WO 04/003138 and WO 2004/007512; U.S. Patent No. 6,777,392 and U.S. Patent Application Publication US2004/0067901; the content of each is incorporated herein by reference in its entirety.
  • Other such HCV polymerase inhibitors include, but are not limited to, valopicitabine (NM-283; Idenix) and 2 > -F-2'-beta-methylcytidine (see also WO 2005/003147).
  • nucleoside HCV NS5B polymerase inhibitors that are used in combination with the present HCV NS3 protease inhibitors are selected from the following compounds: 4-amino-7- ⁇ 2-C-methyl- ⁇ -D-arabinofuranosyl)-7H-pyrrolo[2,3-cf)pyrimidine; 4- amino-7-(2-C-memyI-p ⁇ D-ribofuranosyl)-7H ⁇ vrmlot2,3- ⁇ pyrimidine; 4-methylamino-7-(2-C- methyl- ⁇ -D-ribofuranosyl)-7H-pyrrolo[2,3- ⁇ i]pyrimidine; 4-dimethylamino-7- ⁇ 2-C-methyl- ⁇ -D- rirx)furanosyl)-7H-pyrrolo[2,3-cf]pyiimidine; 4-cyclopropylarnino-7-(2-C-methyl- ⁇ -D- rirx)furanosyl)-7H-pyrrolo[2,3
  • the compounds of the present invention may also be combined for the treatment of ⁇ CV infection with non-nucleoside inhibitors of ⁇ CV polymerase such as those disclosed in International Patent Application Publications WO 01/77091; WO 01/47883; WO 02/04425; WO 02/06246; WO 02/20497; WO 2005/016927 (in particular JTK003); the content of each is incorporated herein by reference in its entirety; and ⁇ CV-796 (Viropharma Inc.).
  • non-nucleoside inhibitors of ⁇ CV polymerase such as those disclosed in International Patent Application Publications WO 01/77091; WO 01/47883; WO 02/04425; WO 02/06246; WO 02/20497; WO 2005/016927 (in particular JTK003); the content of each is incorporated herein by reference in its entirety; and ⁇ CV-796 (Viropharma Inc.).
  • non-nucleoside HCV NS5B polymerase inhibitors that are used in combination with the present HCV NS 3 protease inhibitors are selected from the following compounds: 14-cyclohexyl-6-[2- ⁇ dimethylamino)ethyl]-7-oxo-5 > 6,7,8- tetrahydroindolo[2,l- ⁇ ][2,5]benzodiazocine-l 1-carboxylic acid; 14-cyclohexyl-6- ⁇ 2-morpholin- 4-ylethyl)-5,6,7,8-tetrahydroindolo[2,l- ⁇ ][2,5]benzodiazocine-l 1-carboxylic acid; 14- cyclohexyl-6-[2-(dimemylamino)ethyl]-3-methoxy-5,6,7,8-tetrahydroindolo[2,l- ⁇ ] [2,5]benzodiazocine-l 1-carboxylic acid;
  • a NS3 protease time-resolved fluorescence (TRF) assay is ⁇ CV NS3 protease time-resolved fluorescence (TRF) assay as described below and in Mao et al,, Anal. Biochem. 373: 1-8, 2008 and International Patent Application Publication WO 2006/102087.
  • a NS3 protease assay can be performed, for example, in a final volume of 100 ⁇ l assay buffer containing 50 mM ⁇ EPES, p ⁇ 7.5, 150 mM NaCl, 15 % glycerol, 0.15 % TRITON X-100, 10 mM DTT, and 0.1 % PEG 8000.
  • NS3 and NS4A protease is pre-incubated with various concentrations of inhibitors in DMSO for 30 minutes. The reaction is initiated by adding the TRF peptide substrate (final concentration 100 nM). NS3 mediated hydrolysis of the substrate is quenched after 1 hour at room temperature with 100 ⁇ l of 500 mM MES, pH 5.5. Product fluorescence is detected using either a VICTOR V2 or FUSION fluorophotometer (Perkin Elmer Life and Analytical Sciences) with excitation at 340 nm and emission at 615 nm with a 400 ⁇ s delay. Testing concentrations of different enzyme forms are selected to result in a signal to background ratio (S/B) of 10-30. IC 50 values are derived using a standard four-parameter fit to the data. K 1 values are derived from IC 50 values using the following formula,
  • ICs 0 Ki (1 + [S] / K M ), Eqn (l), where [S] is the concentration of substrate peptide in the reaction and K M is the Michaelis constant.
  • the present invention also includes processes for making compounds of formula (I).
  • the compounds of the present invention can be readily prepared according to the following reaction schemes and examples, or modifications thereof, using readily available starting materials, reagents and conventional synthesis procedures. In these reactions, it is also possible to make use of variants which are themselves known to those of ordinary skill in this art. Other methods for preparing compounds of the invention will be readily apparent to the person of ordinary skill in the art in light of the following reaction schemes and examples. Unless otherwise indicated, all variables are as defined above. The following reaction schemes and examples serve only to illustrate the invention and its practice,
  • Olefin metathesis catalysts include the following Ruthenium based species: F. Miller et al., 118 J. AM. CHEM. SOC. 9606 (1996); G. Kingsbury et al., 121 J. Am. Chem. Soc. 791 (1999); H. Scholl et al., 1 ORG. LETT. 953 (1999); U.S. Patent Application Publication US20O2/01O7138; K. Furstner et al., 64 J. ORG. CHEM. 8275 (1999).
  • the utility of these catalysts in ring closing metathesis is well known in the literature (e.g. Trnka and Grubbs, 34 Ace. CHEM. RES. 18 (2001).
  • Step 1 [(lEVhq ⁇ ta-L ⁇ -dien-l-yloxyJCtrimethvDsilane
  • Step 2 /ro/ty-2-pent-4-en-l-ylcyclopropanol
  • Step 3 methyl 3 -methyl -/'/-(oxomethyleneVL-valinate
  • Step 4 methyl 3 -methyl -N-(If(I j? ⁇ 2J?)-2-pent-4-en-l-ylcvclopropyl]oxy)carbonyl)-L-valinate and methyl 3-methyl-N-( If(15 l ,2SV2-pent-4-en- 1 -ylcyclopropyll oxy ⁇ carbonylVL- valinate
  • Step 3 1-ferr-butyl 2-methyl (2S,4R)-4-r(3-chloro-7-methoxyquinoxalin-2-yl)oxy1pyrrolidine- 1 ,2-dicarboxylate
  • Step 4 methyl (4RM-[(3-chloro-7-methoxyqumoxdin-2- ⁇ l)oxy[-L-prolinate hydrochloride
  • Step 1 methyl 3-methyl-N-( ⁇ [(lR,2R)-2-pent-4-en-l-ylcvclopropyl
  • Step 3 methyl riaR.5S.8S.10R.18E.22aRV5-tert-butyl-14-methoxy-3.6-dioxo- l,la.3 ⁇ 5,6.9.10.20 ⁇ 1.22.22a-dodecahvdro-8H-7,10- methanocyclopropa[18J9in.l0.3,6]dioxadiazacvclononadecino[l lJ2-b1quinoxaline-8- carboxylate
  • Step 4 methyl ⁇ aR.5S.8S.10R.22aRV5-tert-butyl-14-methoxy-3.6-dioxo- l.la.3.4.5.6.9.10.18.19.20.21.22 ⁇ 2a-tetradecahvdro-8H-7.10- methanocvclopropa[ 18 ⁇ 9) [ IJ 0,3 ⁇ Idioxadiazacvclononadecinor 11.12-b]quinoxaline-8- carbox ⁇ late
  • Step 5 (laR,SS.8S.10It22aRVS-tert-but ⁇ l-14-methoxy-3.6-dioxo- l.la.3.4.5.6.9.10J 8.19 ⁇ 0 ⁇ U2 ⁇ 2a-tetradecahvdro-8H-7.10- methanocyclopropaf 18.191 f 1.10,3. ⁇ ldioxadiazacyclononadecinof 1 1,12-b1quinoxaline-8- carboxylic acid
  • Step 6 (I aR.5S.8S.10R ⁇ 2aRV 5-tert-butyl-N-(f 1 RJ2SV 1 - ( [(cvclopropylsulfonvnaminol carbonvU-2-vinylcvclopropyl>-14-methoxy-3.6-dioxo-l.la.3.4,5.6,9.10.18J9,20 ⁇ 1 ⁇ 2 ⁇ 2a- tetradecahydro-8H-7, 10-methanocvclopropaf 18.191[Ll OJ ⁇ ldioxadiazacvclononadecinof 11.12- biquinoxaline-8-carbo ⁇ amide
  • Example 1 The compound of Example 1 was compared to the compound of Examples 110 and 118 of WO 2008/057209. The results are shown in Tables 1 and 2 below. As illustrated in the tables and the discussion of the results, the compound of formula (I) appears to have several advantageous properties compared to both the WO 2008/057209 Example 1 18 compound and the WO 2008/057209 Example 110 compound.
  • Ki Inhibition constant, reference to ⁇ 0.016 nM indicates that the observed activity is less than 0.016 nM, the exact amount less than 0.016 nM was not determined by the assay; EC50; Effective concentration achieving 50% viral replication suppression; gt: Genotype; AUC: Area Under the plasma concentration/time curve; LOQ: Limit of quantitation (3 pmol/mg); BLQ: Below limit of quantitation.
  • Example 110 compound Advantageous properties of the formula (I) compound versus the WO 2008/057209
  • Example 110 compound are the following: 1 ) Physical properties (no salt disproportionation for the Compound of formula (I));
  • the differences in properties are particularly advantageous for the formulation and administration of the formula (I) compound compared to the WO 2008/057209 Example 1 10 compound.
  • the lack of salt disproportionation for the formula (I) compound enables dissolution of 1.8 mg/ml of the K+ salt form of the Compound of Example 1 in water.
  • the K+ salt form of the WO 2008/057209 Example 110 compound has improved aqueous solubility (9.7 mg/mL), the compound thus dissolved disproportionates to give a crystalline zwitterionic form which has low aqueous solubility ( ⁇ 0.009 mg/ml).
  • Example 118 is its resistance profile against different mutant enzymes.
  • HTV protease inhibitors e.g. HTV protease inhibitors
  • HCV NS3 protease inhibitors e.g., VX -950, telaprevir
  • the compound of Example 1 showed improved enzyme affinity (Ki) against different mutant enzymes that are known to confer resistance to HCV NS3 protease inhibitors.
  • Table 2 summarizes activity against different mutant enzymes.
  • an advantage of compound 1 may be an increased barrier to the development of resistant virus when administered to patients. It also provides the potential advantage to treat patients who have failed other therapies because of the development of resistance, since compound 1 may inhibit this resistant virus.
  • Example 110 compound Further expected advantageous properties of the formula (I) compound versus the WO 2008/057209 Example 110 compound include the following:
  • the formula (I) compound was found to have very good covalent binding in vivo characteristics and pharmacokinetic properties. Based on observed covalent binding in vivo and pharmacokinetic properties of the Example 1 compound and the WO 2008/057209 Example 118 compound, the formula (I) compound has significantly better in vivo covalent binding characteristics and pharmacokinetic properties .
  • Example 1 showed undetectable binding to plasma proteins following oral administration of a single 20 mg/kg dose to rats. (See Table 1.) Under analogous conditions, the WO 2008/057209 Example 118 compound demonstrated detectable binding to rat liver proteins (see Table 1), and therefore may be considered a less advantageous compound for administration to human subjects than the compound of formula (I).
  • Table 3 provides some additional in vivo covalent binding data observed for related compounds from WO 2008/057209 that contain the (R,R)-trans-2-alkylcyclopentanol moiety incorporated in Example 118.
  • Target organ exposure in multiple preclinical species provides a rationale that high target organ exposure is achievable for the compound in patients, and high liver exposure in both rat and dog allows for confident evaluation of preclinical toxicity. High liver exposure is especially advantageous for HCV since this is the target organ for the drug.
  • the compound of Example 1 had a very good rat plasma and liver exposure.
  • the observed rat liver exposure was at a level greater than for WO 2008/057209 compound 118 and compound 1 10.
  • Table 1 Based on these results and on testing several different compounds from WO 2008/057209 by oral administration to both rat (25mpk) and dog (5mpk), the compound of formula (I) is also expected to have dog liver exposures greater than WO2008/057209 compound 1 18 and compound 110.
  • NS 3/4A Inhibitory Activity 1 (Ki): NS 3/4A Inhibitory Activity was determined as described in Section IV. Compound Evaluation supra., and Mao et al, Anal Biochem 373 ⁇ - 8, 2008.
  • Rcplicon Activity 2 ECs ⁇ Replicon Activity was determined using the procedures described in Carroll et al, J. Biol. Chem. 278: ⁇ 1979-1 1984, 2003 and Olsen et al., ArUi Microb. Agents 4S-.3944-3953, 2004.
  • Pharmacokinetic parameters were calculated based on non-compartmental analysis (e.g. using WATSON®, W ⁇ NNOL ⁇ N®). Predose concentrations that were below the limit of quantitation (BLQ) were assigned a value of 0. For oral AUC estimation, the first BLQ value in the terminal phase were given a value equal to 1 /2 Lowest Limit Of Quantitation, while subsequent values in the terminal phase were assigned a value of 0. Standard pharmacokinetic parameters CLp, Vdss, half-life (only for IV), % F, Cm ⁇ , Tm 8x , AUQ>-Ust > AUCo-. ⁇ finity were calculated. AUC values were calculated using linear trapezoidal method for ascending concentrations and the log trapezoidal method for descending concentrations.
  • Counting of the plasma samples Place a 200 ⁇ L aliquot in a 20 mL scintillation vial. Add 500 ⁇ L of SOLVABLETM and incubate at 1 h with shaking at 55°C. Remove, allow to cool prior to the addition of 15 mL scintillation cocktail, and count. Plasma samples (200 ⁇ L aliquot) were then processed as described below for liver proteins.
  • Tissue homogenization Weighed liver samples were diluted with 2 vol 100 mM phosphate buffer (pH 7.4) and homogenized on ice.
  • Counting of the liver homogenate Aliquots were placed in a 20 mL scintillation vial, diluted with 1 mL of SOLVABLETM and incubated for 1 h with shaking at 55°C. After removal from the incubator and cooling 15 mL scintillation cocktail and 30% H 2 O 2 were added and the radioactivity counted Protein precipitation: Take 500 ⁇ L aliquot, add 1 :8 homogenate:acetonitrile (if compound is suspected to have low solubility in acetonitrile, another solvent may be selected), vortex and centrifuge (3500 rcf for 20 min). Discard the supernatant.
  • Protein precipitate resuspension Sonication (minimal intensity, ⁇ 5 sec) and vortexing until the pellet crumbles in 80% MeOH: 20% water.
  • Counting of the final pellet 1 mL of dissolved pellet, 15 mL scintillation cocktail (if another scintillation cocktail other than ULTIMA GOLDTM is used, pellet may require neutralization using 1 N HCl), and count.
  • Protein concentration of the final pellet BCA or BlO-RAD kit using BSA as a standard.
  • Counting of the dosing solution Count a known volume of the dosing solution in triplicate.
  • Counting of the dosing solution count a known volume of the dosing solution in triplicate.

Abstract

The present invention relates to macrocyclic a compound of formula (I) and its use as inhibitors of the hepatitis C virus (HCV) NS3 protease, and in treating or preventing HCV infections. Formula (I)

Description

TITLE OF THE APPLICATION
MACROCYCLIC QUINOXALINE COMPOUNDS AS HCV NS3 PROTEASE INHIBITORS
FIELD OF THE INVENTION
The present invention relates to macrocyclic compounds that are useful as inhibitors of the hepatitis C virus (HCV) NS3 protease, the synthesis of such compounds, and the use of such compounds for treating HCV infection and/or reducing the likelihood or severity of HCV infection.
BACKGROUND OF THE INVENTION
Hepatitis C virus (HCV) infection is a major health problem that leads to chronic liver disease, such as cirrhosis and hepatocellular carcinoma, in a substantial number of infected individuals. Current treatments for HCV infection include immunotherapy with recombinant interferon-α alone or in combination with the nucleoside analog ribavirin.
Several virally-encoded enzymes are putative targets for therapeutic intervention, including a metal loprotease (NS2-3), a serine protease (NS3), a helicase (NS3), and an RNA- dependent RNA polymerase (NS5B). The NS3 protease is located in the N-terminal domain of the NS3 protein. NS4A provide a cofactor for NS3 activity.
Potential treatments for HCV infection have been discussed in the different references including Balsano, Mini Rev. Med. Chem. 5^:307-318, 2008, Rδnn et al., Current Topics in Medicinal Chemistry 5:533-562, 2008, Sheldon et al., Expert Opin. Investig. Drugs 16(8)ύ 171-1181, 2007, and De Francesco et al, Antiviral Research 55:1-16, 2003.
SUMMARY OF THE INVENTION
The present invention relates to a macrocyclic compound of formula (I) and pharmaceutically acceptable salts thereof. The compound and its salts are HCV NS3 protease inhibitors. The compound and its salts have therapeutic and research applications.
Thus, a first aspect of the present invention describes a compound of formula (I), or a pharmaceutical acceptable salt thereof:
Figure imgf000002_0001
The present invention also includes pharmaceutical compositions containing a compound of the present invention and methods of preparing such pharmaceutical compositions. The present invention further includes methods of treating or reducing the likelihood or severity of HCV infection.
Other embodiments, aspects and features of the present invention are either further described in or will be apparent from the ensuing description, examples and appended claims.
DETAILED DESCRIPTION QF THE INVENTION
The present invention includes a compound of formula (I), and pharmaceutically acceptable salts thereof. The compound and its pharmaceutically acceptable salts are useful in the inhibition of HCV NS3 protease, the treatment of HCV infection and/or the reduction of the likelihood or severity of an HCV infection. Prophylactic applications include, for example, treatment after suspected past exposure to HCV by such means as blood transfusion, exchange of body fluids, bites, accidental needle stick, or exposure to patient blood during surgery.
As pharmaceutical composition ingredients, the compounds and salts may be the primary active therapeutic agent. When appropriate, the compound may be combined with other therapeutic agents including but not limited to other HCV antivirals, anti-infectives, immunomodulators, antibiotics or vaccines.
NS3 inhibitors are also useful in the preparation and execution of screening assays for antiviral compounds. For example, such compounds can be used to isolate enzyme mutants, which are excellent screening tools for more powerful antiviral compounds. Furthermore, the compounds may be used to establish or determine the binding site of other antivirals to HCV protease, e.g., by competitive inhibition.
As further described in Example 2 the formula (I) compound was compared to the compound of Examples 110 and 118 of WO 2008/057209, and has several advantages. WO 2008/057209 is not admitted to be prior art to the claimed invention.
I. Compositions and Methods
Different embodiments include the following:
(a) A pharmaceutical composition comprising an effective amount of a compound of formula (I) and a pharmaceutically acceptable carrier.
(b) The pharmaceutical composition of (a), further comprising a second therapeutic agent selected from the group consisting of HCV antiviral agents, immunomodulators, and anti-infective agents. (c) The pharmaceutical composition of (b), wherein the HCV antiviral agent is an antiviral selected from the group consisting of HCV protease inhibitors and HCV NS5B polymerase inhibitors.
(d) A pharmaceutical combination which is (i) a compound of formula (I) and (ii) a second therapeutic agent selected from the group consisting of HCV antiviral agents, immunomodulators, and anti-infective agents; wherein the compound of formula (I) and the second therapeutic agent are each employed in an amount that renders the combination effective for inhibiting HCV NS3 protease, or for treating HCV infection and/or reducing the likelihood or severity of HCV infection.
(e) The combination of (d), wherein the HCV antiviral agent is an antiviral selected from the group consisting of HCV protease inhibitors and HCV NS5B polymerase inhibitors.
(f) A method of inhibiting HCV NS 3 protease in a subject in need thereof which comprises administering to the subject an effective amount of a compound of formula (I).
(g) A method of treating HCV infection and/or reducing the likelihood or severity of HCV infection in a subject in need thereof which comprises administering to the subject an effective amount of a compound of formula (I).
(h) The method of (g), wherein the compound of formula (I) is administered in combination with an effective amount of at least one second therapeutic agent selected from the group consisting of HCV antiviral agents, immunomodulators, and anti-infective agents.
(i) The method of (h), wherein the HCV antiviral agent is an antiviral selected from the group consisting of HCV protease inhibitors and HCV NS5B polymerase inhibitors.
(j) A method of inhibiting HCV NS3 protease in a subject in need thereof which comprises administering to the subject the pharmaceutical composition of (a), (b), or (c) or the combination of (d) or (e).
(k) A method of treating HCV infection and/or reducing the likelihood or severity of HCV infection in a subject in need thereof which comprises administering to the subject the pharmaceutical composition of (a), (b), or (c) or the combination of (d) or (e).
(1) A compound of formula (I) for use in medicine, for use in prevention or treatment of HCV infection, or for use (i) in, (ii) as a medicament for, or (iii) in the preparation of a medicament for: (a) inhibiting HCV NS3 protease, or (b) treating HCV infection and/or reducing the likelihood or severity of HCV infection, hi these uses, the compounds of the present invention can optionally be employed in combination with one or more second therapeutic agents selected from HCV antiviral agents, anti-infective agents, and immunomodulators. In all of these embodiments, the compound may optionally be used in the form of a pharmaceutically acceptable salt.
The term "or," as used herein, denotes alternatives that may, where appropriate, be combined. Thus, the term "or" includes each listed alternative separately as well as their combination if the combination is not mutually exclusive.
Reference to a compound also includes stable complexes of the compound such as a stable hydrate. A "stable" compound is a compound which can be prepared and isolated and whose structure and properties remain or can be caused to remain essentially unchanged for a period of time sufficient to allow use of the compound for the purposes described herein (e.g., therapeutic or prophylactic administration to a subject).
II. Administration and Compositions
The term "administration" and variants thereof (e.g., "administering" a compound) means providing the compound or a prodrug of the compound to the individual in need of treatment When a compound of the invention or a prodrug thereof is provided in combination with one or more other active agents (e.g., antiviral agents useful for treating HCV infection), "administration" and its variants are each understood to include concurrent and sequential provision of the compound or salt and other agents.
The compounds of the present invention may be administered in the form of pharmaceutically acceptable salts. The term "pharmaceutically acceptable salt" refers to a salt of the parent compound which has activity and which is not biologically or otherwise undesirable (e.g., is neither toxic nor otherwise deleterious to the recipient thereof). Suitable salts include acid addition salts which may, for example, be formed by mixing a solution of a compound with a solution of a pharmaceutically acceptable acid such as hydrochloric acid, sulfuric acid, acetic acid, trifluoroacetic acid, or benzoic acid. Compounds carrying an acidic moiety can be mixed with suitable pharmaceutically acceptable salts to provide, for example, alkali metal salts (e.g., sodium or potassium salts), alkaline earth metal salts (e.g., calcium or magnesium salts), and salts formed with suitable organic ligands such as quaternary ammonium salts. Also, in the case of an acid (-COOH) or alcohol group being present, pharmaceutically acceptable esters can be employed to modify the solubility or hydrolysis characteristics of the compound.
As used herein, the term "prodrug" is intended to encompass an inactive drug form or compound that is converted into an active drug form or compound by the action of enzymes, chemicals or metabolic processes in the body of an individual to whom it is administered.
As used herein, the term "composition" is intended to encompass a product comprising the specified ingredients, as well as any product which results, directly or indirectly, from combining the specified ingredients. By "pharmaceutically acceptable" is meant that the ingredients of the pharmaceutical composition must be compatible with each other and not deleterious to the recipient thereof.
The term "subject" (alternatively referred to herein as "patient") as used herein refers to an animal, preferably a mammal, most preferably a human, who has been the object of treatment, observation or experiment.
The term "effective amount" indicates a sufficient amount to exert a therapeutic or prophylactic effect. For a patient infected with HCV, an effective amount is sufficient to achieve one or more of the following effects: reduce the ability of HCV to replicate, reduce HCV load, and increase viral clearance. For a patient not infected with HCV, an effective amount is sufficient to achieve one or more of the following: a reduced susceptibility to HCV infection, and a reduced ability of the infecting virus to establish persistent infection for chronic disease.
For the purpose of inhibiting HCV NS3 protease and treating HCV infection and/or reducing the likelihood or severity of symptoms of HCV infection, the compounds of the present invention, optionally in the form of a salt, can be administered by means that produces contact of the active agent with the agent's site of action. They can be administered by conventional means available for use in conjunction with pharmaceuticals, either as individual therapeutic agents or in a combination of therapeutic agents. They can be administered alone, but typically are administered with a pharmaceutical carrier selected on the basis of the chosen route of administration and standard pharmaceutical practice.
Compounds can, for example, be administered by one or more of the following routes: orally, parenterally (including subcutaneous injections, intravenous, intramuscular, intrastemal injection or infusion techniques), by inhalation (such as in a spray form), or rectally, in the form of a unit dosage of a pharmaceutical composition containing an effective amount of the compound and conventional non-toxic pharmaceutically-acceptable carriers, adjuvants and vehicles. Liquid preparations suitable for oral administration (e.g., suspensions, syrups, elixirs and the like) can be prepared according to techniques known in the art and can employ any of the usual media such as water, glycols, oils, alcohols and the like. Solid preparations suitable for oral administration (e.g. , powders, pills, capsules and tablets) can be prepared according to techniques known in the art and can employ such solid excipients as starches, sugars, kaolin, lubricants, binders, disintegrating agents and the like. Parenteral compositions can be prepared according to techniques known in the art and typically employ sterile water as a carrier and optionally other ingredients, such as solubility aids. Injectable solutions can be prepared according to methods known in the art wherein the carrier comprises a saline solution, a glucose solution or a solution containing a mixture of saline and glucose. Further guidance for methods suitable for use in preparing pharmaceutical compositions of the present invention and of ingredients suitable for use in said compositions is provided in Remington's Pharmaceutical Sciences, 20th edition (ed. A. R. Gennaro, Mack Publishing Co., 2000).
The compounds of this invention can be administered orally in a dosage range of 0.001 to 1000 mg/kg of mammal (e.g., human) body weight per day in a single dose or in divided doses. One dosage range is 0.01 to 500 mg/kg body weight per day orally in a single dose or in divided doses. Another dosage range is 0.1 to 100 mg/kg body weight per day orally in single or divided doses. For oral administration, the compositions can be provided in the form of tablets or capsules containing 1.0 to 500 mg of the active ingredient, particularly 1, 5, 10, 15, 20, 25, 50, 75, 100, 150, 200, 250, 300, 400, 500, and 750 mg of the active ingredient for the symptomatic adjustment of the dosage to the patient to be treated. The specific dose level and frequency of dosage for any particular patient may be varied and will depend upon a variety of factors including the activity of the specific compound employed, the metabolic stability and length of action of that compound, the age, body weight, general health, sex, diet, mode and time of administration, rate of excretion, drug combination, the severity of the particular condition, and the host undergoing therapy.
Ill, Combination Treatment
The quinoxaline macrocyclic compounds described herein can be used in a combination treatment involving one or more additional therapeutic agents. Additional therapeutic agents include those also targeting HCV, targeting a different disease causing agent, or those enhancing the immune system. Agents enhancing the immune system include those generally enhancing an immune system function and those producing a specific immune response against HCV. Additional therapeutic agents targeting HCV include agents targeting NS3 and agents targeting other HCV activities such as NS5A and NS5B, and agents targeting host cell activities involved in HCV replication.
Different HCV inhibitors are described in different publications. Macrocyclic compounds useful as inhibitors the HCV protease inhibitors are described in WO 06/119061, WO 7/015785, WO 7/016441, WO 07/148135, WO 08/051475, WO 08/051477, WO 08/051514, WO 08/057209. Additional HCV NS3 protease inhibitors are disclosed in International Patent Application Publications WO 98/22496, WO 98/46630, WO 99/07733, WO 99/07734, WO 99/38888, WO 99/50230, WO 99/64442, WO 00/09543, WO 00/59929, WO 02/48116, WO 02/48172, British Patent No. GB 2 337 262, and U.S. Patent No. 6,323,180.
Additional examples of therapeutic agents that may be present in a combination include ribavirin, levovirin, viramidine, thymosin alpha- 1, interferon-β, interferon-α, pegylated interferon-α (peginterferon-α), a combination of interferon-α and ribavirin, a combination of peginterferon-α and ribavirin, a combination of interferon-α and levovirin, and a combination of peginterferon-α and levovirin. Interferon-α includes recombinant interferon-α2a (such as ROFERON interferon available from Hoffmann-LaRoche, Nutley, NJ), pegylated interferon-α2a (PEGASYS), interferon-α2b (such as INTRON-A interferon available from Schering Corp., Kenilworth, NJ), pegylated interferon-<z2b (PEGINTRON), a recombinant consensus interferon (such as interferon alρhacon-1), and a purified interferon-α product. Amgen's recombinant consensus interferon has the brand name INFERGEN. Levovirin is the L-enantiomer of ribavirin which has shown immunomodulatory activity similar to ribavirin. Viramidine represents an analog of ribavirin disclosed in WO 01/60379. The individual components of the combination can be administered separately at different times during the course of therapy or concurrently in divided or single combination forms.
Ribavirin, levovirin, and viramidine may exert their anti-HCV effects by modulating intracellular pools of guanine nucleotides via inhibition of the intracellular enzyme inosine monophosphate dehydrogenase (IMPDH). IMPDH is the rate-limiting enzyme on the biosynthetic route in de novo guanine nucleotide biosynthesis. Ribavirin is readily phosphorylated intracellularly and the monophosphate derivative is an inhibitor of IMPDH. Thus, inhibition of IMPDH represents another useful target for the discovery of inhibitors of HCV replication. Therefore, the compounds of the present invention may also be administered in combination with an inhibitor of IMPDH, such as VX-497, which is disclosed in International Patent Application Publications WO 97/4121 1 and WO 01/00622; another IMPDH inhibitor, such as that disclosed in WO 00/25780; or mycophenolate mofetil. See A.C. Allison and E.M. Eugui, 44 (Suppl.) Agents Action 165 (1993).
For the treatment of HCV infection, the compounds of the present invention may also be administered in combination with the antiviral agent amantadine (1-aminoadamantane). For a comprehensive description of this agent, see J. Kirschbaum, 12 Anal. Profiles Drug Subs. 1-36 (1983).
For the treatment of HCV infection, the compounds of the present invention may also be administered in combination with the antiviral agent polymerase inhibitor R7128 (Roche).
The compounds of the present invention may also be combined for the treatment of HCV infection with antiviral 2'-C-branched ribonucleosides disclosed in R. E. Harry-O'Kuru et al., 62 J. Org. Chem. 1754-59 (1997); M. S. Wolfe et al., 36 Tet. Lett. 761 1-14 (1995); U.S. Patent No. 3,480,613; and International Patent Application Publications WO 01/90121, WO 01/92282, WO 02/32920, WO 04/002999, WO 04/003000 and WO 04/002422; the contents of each of which are incorporated by reference in their entirety. Such 2'-C-branched ribonucleosides include, but are not limited to, 2'-C-methyl-cytidine, 2'-C-methyl-uridine, 2'-C- methyl-adenosine, 2'-C-methyl-guanosine, and 9-(2-C-methyl-β-D-ribofuranosyl)-2,6- diaminopurine, and the corresponding amino acid ester of the ribose C-2', C-3', and C-5' hydroxyls and the corresponding optionally substituted cyclic 1,3 -propanediol esters of the 5'- phosphate derivatives.
The compounds of the present invention may also be combined for the treatment of HCV infection with other nucleosides having anti-HCV properties, such as those disclosed in International Patent Application Publications WO 02/51425, WO 01/79246, WO 02/32920, WO 02/48165 and WO2005/003147 (including Rl 656, (2'/?)-2'-deoxy-2'-fluoro-2'-C- methylcytidine, shown as compounds M on page 77); WO 01/68663; WO 99/43691; WO 02/18404 and WO2006/021341, and U.S. Patent Application Publication US 2005/0038240, including 4'-azido nucleosides such as Rl 626, 4'-azidocytidine; U.S. Patent Application Publications US 2002/0019363, US 2003/0236216, US 2004/0006007 and US 2004/0063658; and International Patent Application Publications WO 02/100415, WO 03/026589, WO 03/026675, WO 03/093290, WO 04/011478, WO 04/013300 and WO 04/028481 ; the content of each is incorporated herein by reference in its entirety.
For the treatment of HCV infection, the compounds of the present invention may also be administered in combination with an agent that is an inhibitor of HCV NS5B polymerase. Such HCV NS5B polymerase inhibitors that may be used as combination therapy include, but are not limited to, those disclosed in International Patent Application Publications WO 02/057287, WO 02/057425, WO 03/068244, WO 2004/000858, WO 04/003138 and WO 2004/007512; U.S. Patent No. 6,777,392 and U.S. Patent Application Publication US2004/0067901; the content of each is incorporated herein by reference in its entirety. Other such HCV polymerase inhibitors include, but are not limited to, valopicitabine (NM-283; Idenix) and 2>-F-2'-beta-methylcytidine (see also WO 2005/003147).
In one embodiment, nucleoside HCV NS5B polymerase inhibitors that are used in combination with the present HCV NS3 protease inhibitors are selected from the following compounds: 4-amino-7-{2-C-methyl-β-D-arabinofuranosyl)-7H-pyrrolo[2,3-cf)pyrimidine; 4- amino-7-(2-C-memyI-p^D-ribofuranosyl)-7Hφvrmlot2,3-^pyrimidine; 4-methylamino-7-(2-C- methyl-β-D-ribofuranosyl)-7H-pyrrolo[2,3-<i]pyrimidine; 4-dimethylamino-7-{2-C-methyl-β-D- rirx)furanosyl)-7H-pyrrolo[2,3-cf]pyiimidine; 4-cyclopropylarnino-7-(2-C-methyl-β-D- rirx)furanosyl)-7H-pyrrolo[2,3-<i]pyrimidine; 4-amino-7-{2-C-vάnyl-β-D-ribofuranosyl)-7H- ρvrrolo(2,3-d]pyrimidine; 4-amino-7-(2-C-hydroxymethyl-β-D-ribofuranosyl)-7H- pyrrolo[2,3-<i]pyrimidine; 4-amino-7-{2-C-fluoromethyl-β-D-ribofuranosyl)-7H- pyrrolo[2,3-<i]pyrimidine; 4-amino-5-methyl-7-{2-C-methyl-β-D-ribofuranosyl)-7H- pyrrolo[2,3-<i]pyrimidine; 4-amino-7-(2-C-methyl-β-D-ribofuranosyl)-7H- pyrrolo[2,3-J]pyrimidine-5-carboxylic acid; 4-amino-5-bromo-7-(2-C-methyl-β-D- ribofuranosyl)-7H-pyrrolo[2,3-</]pyrimidine; 4-amino-5-chloro-7-(2-C-methyl-β-D- ribofuranosyl)-7H-pyrrolo[2,3-ci]pyrirnidine; 4-amino-5-fluoro-7-(2-C-methyl-β-D- ribofuranosyl)-7H-pyrrolo[2,3-t(]pyrirnidine; 2,4-diamino-7-(2-C-methyl-β-D-ribofuranosyl)- 7//-pyrrolo[2,3-J]pyrimidine; 2-amino-7-(2-C-methyl-β-D-ribofuranosyl)-7H- pyrrolo[2,3-J]pyrimidine; 2-amincMlΗ;ycIopropylaniino-7-(2-C-methyl-β-D-ribofuranosyl)-7H- pyrrolo[2,3-J]pyrimidine; 2-amino-7-(2-C-methyl-β-D-ribofuranosyl)-7H- pyrrolo[2,3-J]pyrimidin-4(3H)-one; 4-amino-7-(2-C-ethyl-β-D-ribofuranosyl)-7H- pyrrolo[2,3-if]pyrimidine; 4-amino-7-(2-C,2-O-dimethyl-β-D-ribofuτanosyl)-7H- pyrrolo[2,3-t/]pyrimidine; 7-(2-C-methyl-β-D-rilx)furanosyl)-7H-pyττolo[2)3-<i]pyrimidin-4(3H)- one; 2-amino-5-methyl-7-{2-C, 2-0-dimethyl-β-D-ribofuranosyl)-7H-pyτrolo[2,3-<i]pyrimidin- 4(3H)-one; 4-amino-7^3-deoxy-2-C-methyl-β-D-ribofuranosyl)-7H-pyrrolo[2,3-J]pyrimidine; 4-amino-7-(3-deoxy-2^-memyl-β-D-arabinofuranosyl)-7H-pyπOlo[2,3-^ 2-fluoro-7-(2-C-methyl-β-D-ribofuranosyl)-7H-pyrrolo[2,3-<i]pyrimidiiie; 4-amino-7-(3-C- methyl-β-D-ribofuranosyl)-7H-pyiτolo[2,3-</]pyrimidine; 4-amino-7-(3-C-methyl-β-D- xylofuranosyl)-7H-pyrrolo[2,3-^pyrimidine; 4-amino-7-(2,4-di-C-methyl-β-D-ribofuranosyl)- 7H-pyτrolo[2,3-<f]pyrimidine; 4-amino-7-(3-deoxy-3-fluoro-2-C-methyl-β-D-ribofuranosyl)-7H- pyrrolo[2,3-</]pyrimidine; and the corresponding 5'-triphosphates; or a pharmaceutically acceptable salt thereof.
The compounds of the present invention may also be combined for the treatment of ΗCV infection with non-nucleoside inhibitors of ΗCV polymerase such as those disclosed in International Patent Application Publications WO 01/77091; WO 01/47883; WO 02/04425; WO 02/06246; WO 02/20497; WO 2005/016927 (in particular JTK003); the content of each is incorporated herein by reference in its entirety; and ΗCV-796 (Viropharma Inc.).
In one embodiment, non-nucleoside HCV NS5B polymerase inhibitors that are used in combination with the present HCV NS 3 protease inhibitors are selected from the following compounds: 14-cyclohexyl-6-[2-{dimethylamino)ethyl]-7-oxo-5>6,7,8- tetrahydroindolo[2,l-α][2,5]benzodiazocine-l 1-carboxylic acid; 14-cyclohexyl-6-{2-morpholin- 4-ylethyl)-5,6,7,8-tetrahydroindolo[2,l-Λ][2,5]benzodiazocine-l 1-carboxylic acid; 14- cyclohexyl-6-[2-(dimemylamino)ethyl]-3-methoxy-5,6,7,8-tetrahydroindolo[2,l-α] [2,5]benzodiazocine-l 1-carboxylic acid; 14-cyclohexyl-3-methoxy-6-methyl-5,6,7,8- tetrahydroindolo[2,l-tf][2,5]benzodiazocine-l 1-carboxylic acid; methyl ({[(14-cyclohexyl-3- methoxy-ό-methyl-Sjό^^-tetrahydroindolo^l-αJp.SJbenzodiazocin-l 1- yl)carbonyl]amino}sulfonyl)acetate; ({[(M-cyclohexyl-S-methoxy-ό-methyl-S^J.δ- te^rahydroindolo[2,l-α][2,5]benzcκiiazocm-l l-yl)carbκ)nyl]amino}sulfonyl)acetic acid; 14- cyclohexyl-iV-[(dimethylamino)sulfonyl]-3-methoxy-6-methyl-5,6,7,8-tetrahydroindolo[2,l-α] [2,5]benzodiazocine-l 1-carboxamide; 3-chloro-14-cyclohexyl-6-[2-(dimethylamino)ethyl]-7- oxo-S.δJjδ-tetrahydroindolo^l-αJ^SJbenzodiazocine 11-carboxylic acid; .V-(11-carboxy- 14- cyclohexyl-7,8-dihydro-6H-indolo[ 1 ,2-e] [ 1 ,5]benzoxazocin-7-yl)-7vr^V-dimethylethane- 1 ,2- diaminium bis(trifluoroacetate); 14-cyclohexyl-7,8-dihydro-6H-indolo[ 1 ,2-e] [ 1 ,5] benzoxazocine-11-carboxylic acid; 14-cyclohexyl-6-methyl-7-oxo-5,6,7,8-tetrahydroindolo [2,l-α][2,5]benzodiazocine-l 1-carboxylic acid; H-cyclohexyl-S-methoxy-β-methyl-T-oxo- 5,6,7, 8-tetrahydroindolo[2,l-α][2,5]benzodiazocine-l 1-carboxylic acid; 14-cyclohexyl-6-[2- (dimethylamino)ethyl]-3-methoxy-7-oxo-5,6,7,8-tetrahydroindolo[2,l-<i][2,5]benzodiazocine- 11-carboxylic acid; 14-cyclohexyl-6-[3-(dimethylamino)propyl]-7-oxo-5,6,7,8-tetrahydroindolo [2,l-α][2,5]benzodiazocine-l 1-carboxylic acid; 14-cyclohexyl-7-oxo-6-{2-piperidin-l-ylethyl)- 5,6,7,8-tetrahydroindolo[2,l-α][2,5]ben2xκiiazocine-l 1-carboxylic acid; 14-cyclohexyl-6-(2- moφholin-4-ylethyl)-7-oxo-5,6J,8-tetrahydroindolo[2,l-α][2,5]benzodiazocine-l l-carboxyIic acid; 14-cyclohexyl -6- [2-(diethylamino)ethyl] -7-oxo-5 ,6,7, 8-tetrahydroindolo [2, 1 -a] [2,5]benzodiazocine-l 1-carboxylic acid; 14-cyclohexyl-6-(l-rnethylpiperidin-4-yl)-7-oxo- 5,6,7,8-tetrahydroindolo[2,l-α][2,5]benzodiazocine-l 1-carboxylic acid; 14-cyclohexyl-/1/- [(dimethylamino)sulfonyl]-7-oxo-6-(2-piperidin- 1 -ylethyl)-5,6,7,8-tetrahydroindolo[2, 1 -a] [2,5]benzodiazocine-l 1-carboxamide; 14-cyclohexyl-6-[2-(dimethylamino)ethyl]-N- [(dimethylamino)sulfonyl]-7-oxo-5,6,7,8-tetrahydroindolo[2,l-α][2,5]benzodiazocine-l l- carboxamide; 14-cyclopentyl-6-[2-(dimethylamino)ethyl]-7-oxo-5>6,7>8-tetrahydroindolo[2, 1 -a] [2,5]benzodiazocine-l 1-carboxylic acid; 14-cycIohexyl-5,6,7,8-tetrahydroindolo[2>l-iϊ] [2,5]benzodiazocine-l 1-carboxylic acid; 6-allyl-14-cyclohexyl-3-methoxy-5,6,7,8- tetrahydroindo Io [2, 1-α] [2, 5 ]benzodiazocine-l 1-carboxylic acid; 14-cyclopentyl-6-[2- (dimethylamino^thylJ-S.δJ.δ-tetrahydroindoloP,! -α][2,5]benzodiazocine-l 1 -carboxylic acid; 14-cyclohexyl-6-[2-(dimethylamino)ethyl]-5,6,7,8-tetrahydroindolo[2,l-α][2,5]ben2θdiazocine- 11 -carboxylic acid; \3^γc\ohexy\-5-mcϋ^y\A,5,6jΛc\xBhy<kofuw[y,T:βt7][\,4]ώazocmo[\,^- α] indole- 10-carboxylic acid; 15-cyclohexyl-6-[2-{dimethylamino)ethyl]-7-oxo-6, 7,8,9- tetrahydro-5H-indolo[2, 1 -a] [2,6]benzodiazonine- 12-carboxylic acid; 15-cyclohexyl-8-oxo- 6,7,8,9-tetrahydro-5H-indolo[2,l-σ][2,5]ben2θdiazonine-12-carboxylic acid; 13-cyclohexyl-6- oxo-6,7-dihydro-5H-indolo[l,2-<f)[l ,4)benzodiazq)ine-10-carboxylic acid; and pharmaceutically acceptable salts thereof.
IV. Compound Evaluation
Compounds described herein can be evaluated for different activities such as the ability to inhibit ΗCV NS3 activity, ΗCV replicon activity, and ΗCV replication activity using techniques well-known in the art. (See, for example, Carroll et al, J. Biol. Chem. 278: 11979— 11984, 2003.)
One such assay is ΗCV NS3 protease time-resolved fluorescence (TRF) assay as described below and in Mao et al,, Anal. Biochem. 373: 1-8, 2008 and International Patent Application Publication WO 2006/102087. A NS3 protease assay can be performed, for example, in a final volume of 100 μl assay buffer containing 50 mM ΗEPES, pΗ 7.5, 150 mM NaCl, 15 % glycerol, 0.15 % TRITON X-100, 10 mM DTT, and 0.1 % PEG 8000. NS3 and NS4A protease is pre-incubated with various concentrations of inhibitors in DMSO for 30 minutes. The reaction is initiated by adding the TRF peptide substrate (final concentration 100 nM). NS3 mediated hydrolysis of the substrate is quenched after 1 hour at room temperature with 100 μl of 500 mM MES, pH 5.5. Product fluorescence is detected using either a VICTOR V2 or FUSION fluorophotometer (Perkin Elmer Life and Analytical Sciences) with excitation at 340 nm and emission at 615 nm with a 400 μs delay. Testing concentrations of different enzyme forms are selected to result in a signal to background ratio (S/B) of 10-30. IC50 values are derived using a standard four-parameter fit to the data. K1 values are derived from IC50 values using the following formula,
ICs0 = Ki (1 + [S] / KM), Eqn (l), where [S] is the concentration of substrate peptide in the reaction and KM is the Michaelis constant. See P. Gallinari et al., 38 BlOCHEM. 5620-32(1999); P. Gallinari et al., 72 J. ViROL. 6758-69 (1998); M. Taliani et al., 240 ANAL. BIOCHEM. 60-67 (1996); Mao et al, Analytical Biochemistry 373: 1-8, 2008.
V. General Compound Production
The present invention also includes processes for making compounds of formula (I). The compounds of the present invention can be readily prepared according to the following reaction schemes and examples, or modifications thereof, using readily available starting materials, reagents and conventional synthesis procedures. In these reactions, it is also possible to make use of variants which are themselves known to those of ordinary skill in this art. Other methods for preparing compounds of the invention will be readily apparent to the person of ordinary skill in the art in light of the following reaction schemes and examples. Unless otherwise indicated, all variables are as defined above. The following reaction schemes and examples serve only to illustrate the invention and its practice,
Olefin metathesis catalysts include the following Ruthenium based species: F. Miller et al., 118 J. AM. CHEM. SOC. 9606 (1996); G. Kingsbury et al., 121 J. Am. Chem. Soc. 791 (1999); H. Scholl et al., 1 ORG. LETT. 953 (1999); U.S. Patent Application Publication US20O2/01O7138; K. Furstner et al., 64 J. ORG. CHEM. 8275 (1999). The utility of these catalysts in ring closing metathesis is well known in the literature (e.g. Trnka and Grubbs, 34 Ace. CHEM. RES. 18 (2001).
The following examples serve only to illustrate the invention and its practice. The examples are not to be construed as limitations on the scope or spirit of the invention.
List of Abbreviations
List of Abbreviations
DCM / CH2Cl2 dichloromethane
DCE 1,2-dichloroethane DIEA diisopropylethylamine
DMF dimethylformamide
DMSO dimethyl sulfoxide
Dppf diphenylphosphinoferrocene
Et2θ diethyl ether
ElOAc ethyl acetate
HATU <9-(7-azabenzotr iazol- UyI)-N1N1N ',N '-tetramethyluronium hexafluoropho sphate
HCl hydrochloric acid
TMSCl Chlorotήmethylsilane
TBAF Tetra-butyl ammonium fluoride
DMAP Dimethylamino pyridine
MeCN acetonitrile
MeOH methanol
Pd/C palladium on carbon
TBTU O-benzotriazol- 1 -y\-N,N,N ',N '-tetramethyluronium tetrafluoroborate
TFA trifluoroacetatic acid
THF tetrahydofuran
Flash Chromatography Purification using Biotage Horizon using silica gel cartridge and specified mobile phase gradient
HPLC Automated mass or UV triggered high performance liquid chromatography using acidified MeCN and H2O gradients as mobile phase
MHz Megahertz
Synthesis of Intermediates
Intermediates A
Intermediate # Structure Name Lit. Reference
Al (lϋ,25>l-Amino-iV- Wang et al, (cyclopropylsulfonyl)-2- US 6,995,174 vinylcyclopropanecarboxamide
Figure imgf000013_0001
hydrochloride Intermediate Bl : 3-methvI-iV-(|[(li? JifVl-pent-^n-l-ylcvclopropylioxylcarbonvD-L- valine
Figure imgf000014_0002
Step 1 : [(lEVhq^ta-Lό-dien-l-yloxyJCtrimethvDsilane
Figure imgf000014_0001
A solution (0.5 M) of butenyl magnesium bromide in THF (1.4 eq) was treated at -78°C with Cu(I)Br.SMe2 (0.05 eq) and HMPA (2.4 eq). The mixture was stirred for 10 min then a solution (1 M) of acrolein (1 eq) and TMSCl (2 eq) in THF was added over 1 h such that the internal temperature remained below -68 0C. The resulting mixture was stirred at -78 0C for 2 h then treated with excess Et3N and diluted with hexane. After reaching room temperature the mixture was treated with a small portion of H2O and filtered through CELITE. The filtrate was washed 10 times with H2O and then with brine. The organic layer was dried and the volatiles were removed to give a residue that was distilled under reduced pressure (20 mbar). The fraction collected at 80-86 0C contained the title compound (58%) as a colorless liquid. l H NMR (400 MHz, CDCl3) δ 6.19 (d, J = 1 1.6 Hz, IH), 5.85-5.75 (m, IH), 5.02-4.92 (m, 3H), 2.08-2.02 (m, 2H), 1.94-1.88 (m, 2H), 1.46-1.38 (m, 2H), 0.18 (s, 9H).
Step 2: /ro/ty-2-pent-4-en-l-ylcyclopropanol
Figure imgf000014_0003
A solution (0.45 M) of the preceding compound in hexane was treated with a solution (15%) Of Et2Zn (1.2 eq) in toluene and the resulting solution was cooled in an ice bath. Diiodomethane (1.2 eq) was added dropwise then the solution was stirred for 1 h before being warmed to 20 0C. Pyridine (6 eq) was added and the slurry was stirred for 15 min then poured onto petroleum ether. The mixture was filtered repeatedly through CELITE until a transparent solution was obtained. This mixture was concentrated at 100 mbar and the solution which remained (that contained trimethyl{[(/rαrw)-2-pent-4-en-l-ylcyclopropyl]oxy}silane, toluene and pyridine) was further diluted with THF. The mixture was cooled to 0 0C then treated dropwise with a solution (1 M) of TBAF (1.2 eq) in THF. After 10 min the mixture was allowed to warm to 20 0C, and after a further 1 h was poured into H2O. The aqueous phase was extracted with EtOAc and the combined organic extracts were washed with brine then dried. Removal of the volatiles afforded a residue that was purified by flash chromatography (eluent 0-66% Et2O/petroleum ether) to furnish the title compound (71%) as a colorless liquid. 1H NMR (400 MHz, CIX:i3) δ 5.85-5J5 (m, IH), 5.00 Cd^ J= H1I, 1.6 Hz, IH), 4.94 (br d, J= 10.4 Hz, IH), 3.20 (apparent dt, J= 6.4, 2.5 Hz, IH), 2.10-2.04 (m, 2H), 1.52-1.44 (m, 2H), 1.29-1.19 (m, IH), 1.15-1.07 (m, IH), 0.95-0.87 (m, IH), 0.71-0.66 (m,lH), 0.31 (apparent q, J= 6.0 Hz, IH).
Step 3: methyl 3 -methyl -/'/-(oxomethyleneVL-valinate
Figure imgf000015_0002
A solution (0.39 M) of methyl 3-methyI-L-valinate in a 2:1 mixture of saturated aqueous NaHCO3 and CH2Cl2 was cooled in an ice bath and stirred rapidly. The mixture was treated with triphosgene (0.45 eq) in one portion and the resulting mixture was stirred for 0.5 h. The reaction was diluted with CH2Cl2 and the layers were separated. The aqueous phase was extracted with CH2Cl2 then the combined organics were washed with brine and dried. Removal of the solvent gave the title compound as clear oil that was kept for 12 h under vacuum (0.1 mbar) then used directly in the subsequent step. 1H NMR (400 MHz, CDCl3) δ 3.79 (s, 3H), 3.75 (s, IH), 1.00 (s, 9H).
Step 4: methyl 3 -methyl -N-(If(I j?<2J?)-2-pent-4-en-l-ylcvclopropyl]oxy)carbonyl)-L-valinate and methyl 3-methyl-N-( If(15l,2SV2-pent-4-en- 1 -ylcyclopropyll oxy} carbonylVL- valinate
Figure imgf000015_0001
A solution (0.45 M) of /r<ms-2-pent-4-en-l-ylcyclopropanol in toluene was treated with methyl 3-methyl-N-(oxomethylene)-L-valinate (1.1 eq) and then DMAP (1 eq). The resulting mixture was heated under reflux for 12 h then cooled to 20 0C. H2O and EtOAc were added and the organic layer was separated and washed with IN HCl, brine and dried. Removal of the volatiles afforded a residue that was purified twice by flash chromatography (eluent 0-30% Et2O/petroleum ether). The first fractions contained methyl 3-methyl-N-({[(li?,2i?)-2-pent-4-en- l-ylcyclopropyl]oxy}carbonyl>L-valinate (38%) as an oil. MS (ES+) m/z 298 (M+H)+
The later fractions contained methyl 3 -methyl -N-(If(I^, 25)-2-pent-4-en- 1 - ylcyclopropyl]oxy}carbonyl)-L-valinate (28%) as an oil. MS (ES") m/z 298 (M+H)+ Step 5 : 3 -methy\-N-( { [( 1 R2 J?V2-pent-4-en- 1 - ylcvclopropyll oxy ) carbonvD-L-valine
Figure imgf000016_0001
A solution (0.1 M) of methyl 3-methyl-ΛK{[(l/?,2Λ>2-pent-4-en-l- ylcyclopropyl]oxy}carbonyl)-L-valinate in 2:1 mixture of MeOH/H2O was treated with LiOH-H2O (4 eq) and then heated at 60 °C for 4 h. The mixture was cooled and concentrated to half volume then diluted with EtOAc and acidified with aqueous HCl (1 N). The organic layer was separated and washed with brine then dried. Removal of the volatiles afforded the title compound (98%) as an oil. MS (ES+) m/z 284 (M+H)+
Intermediates C
Intermediate Cl: methyl (4R)-4-[(3-chJoro-7-methoiyqnJnoxaLin-2-yl)oxyl-L7pro-iiiate hydrochloride
Figure imgf000016_0002
Step 1 : 6-methoxyquinoxaJine-2,3-diol
Figure imgf000016_0003
A suspension of 4-methoxybenzene-l,2-diamine dihydrochloride in diethyl oxalate (8 eq) was treated with Et3N (2 eq) and then heated at 150 °C for 2 h. The mixture was cooled and filtered, and then the collected solid was washed with H2O and EtOH. The residue was dried to give the title compound (69%). MS (ES+) m/z 193 (M+H)+
Step 2: 3-chloro-6-methoxyquinoxalin-2-ol
Figure imgf000016_0004
A solution (1.53 M) of 6-methoxyquinoxaline-2,3-diol in DMF was treated with SOCl2 (1 eq) and heated at 1 10 °C. After 1.5 h the reaction mixture was cooled and poured into aqueous HCl (1 N). The resulting precipitate was filtered and washed with H2O and Et2O. The dried solid contained predominantly the title compound as a mixture with 6- methoxyquinoxaline-2,3-diol and 2,3-dichloro-6-methoxyquinoxaline. This material was used directly in the subsequent step. MS (ES+) m/z 211 (M+H)+
Step 3: 1-ferr-butyl 2-methyl (2S,4R)-4-r(3-chloro-7-methoxyquinoxalin-2-yl)oxy1pyrrolidine- 1 ,2-dicarboxylate
Figure imgf000017_0001
A solution (0.35 M) of 3-chloro-6-methoxyquinoxalin-2-ol in NMP was treated with Cs2CO3 (1.5 eq) and 1-terr-butyl 2-methyl (2S,4S>4-{[(4- bromophenyl)sulfonyl]oxy} pyrrolidine- 1 ,2-dicarboxylate (1.1 eq). The resulting mixture was stirred at 50 °C for 18 h then a further portion (0.1 eq) of 1 -rer/-butyl 2-methyl (2S,4S)-4-{[(4- bromophenyl)sulfonyl]oxy} pyrrolidine- 1 ,2-dicarboxylate was added. After stirring for 2 h the mixture was cooled and diluted with H2O and EtOAc. The organic phases were washed with aqueous HCl (1 N), saturated aqueous NaHCO3 and brine. The dried organic phase was concentrated to a residue that was purified by flash-chromatography (0-60% EtO Ac/petroleum ether) to give the title compound (35% for two steps) as a solid. MS (ES+) m/z 438 (M+H)+
Step 4: methyl (4RM-[(3-chloro-7-methoxyqumoxdin-2-γl)oxy[-L-prolinate hydrochloride
Figure imgf000017_0002
A solution (0.62 M) of 1-terf-butyl 2-methyl (2S,4R)-4-[(3-chloro-7- methoxyquinoxalin-2-yl)oxy]pyrrolidine-l,2-dicarboxylate in CH2Cl2 was treated with a solution (4 M) of HCl in dioxane (5 eq). The mixture was stirred at 20 °C for 2 h then treated with a solution (4 M) of HCl in dioxane (2 eq). After 5 h the reaction was judged complete and the mixture was concentrated under reduced pressure. The residue was triturated with Et2O to give the title compound (95%) as a solid. MS (ES+) m/z 338 (M+H)+
Example 1; Potassium (I(lIUS>-l-((KlaR^S,8S.10IL22aR>-S-tert-butyl-14-methoxy-3<6- dioiQ-l,la3.4^.6.9.10.18.19v20t21<22<22a-tetradecahvdro-8H-7.10- methaDocvclopropan8.19|[lJ0<3,61dioxadiazacvclononadecipoU1.12-blquinoialiD-8- yllcarbonγl}amino>-2-vinylcγclopropγllcarbonylKcvclopropylsιiIfonγπazapide
X
Figure imgf000018_0001
Step 1 : methyl 3-methyl-N-({[(lR,2R)-2-pent-4-en-l-ylcvclopropyl|oxylcarfaonyl>-L-valyl- (4R)-4-[(3-chloro-7-methoxyquinoxalm-2-yl')oxy1-L-prolinate
Figure imgf000018_0002
A solution (0.2 M) of methyl (4R)-4-[(3-chloro-7-methoxyquinoxalin-2-yl)oxy]- L-prolinate hydrochloride in DMF was treated with 3-methyl-N-({[(lR,2R)-2-pent-4-en-l- ylcyclopropyl]oxy}carbonyl)-L-valine (1.1 eq), DIEA (5 eq) and HATU (1.2 eq). The resulting mixture was stirred at 20 0C for 5 h then diluted with EtOAc. The organic layer was separated and washed with aqueous HCl (1 N), saturated aqueous NaHCO3 and brine. The dried organic phase was concentrated under reduced pressure to give a residue that was purified by flash chromatography (eluent 10-30% EtOAc/petroleum ether) to furnish the title compound (96%) as an oil. MS (ES+) m/z 604 (M+H)+ Step 2: methyl 3-methyl-N-(([(lR,2R>-2-pent-4-en-l-ylcvclopropylloxylcarbonyl)-L-valyl- (4RV4-f(7-methoxy-3-vinylquinoxalin-2-yl')oxy1-L-prolinate
Figure imgf000019_0001
A solution (0.1 M) of methyl 3-methyl-N-({[(lR^2R)-2-pent-4-en-l- ylcyclopropyl]oxy}carbonyl)-L-valyl-{4R)-4-[(3-chloro-7-methoxyquinoxalin-2-yl)oxy]-L- prolinate in EtOH was treated with potassium trifluoro(vinyl)borate (1.5 eq) and triethylamine (1.5 eq). The resulting mixture was degassed, then PdCl2(dppf)-CH2Cl2 adduct (0.1 eq) was added. The mixture was heated under reflux for 1 h then cooled to room temperature and diluted with H2O and EtOAc. The organic phase was separated, washed with H2O and brine then dried. Removal of the volatiles afforded a residue that was purified by flash chromatography (20-30% EtO Ac/petroleum ether) to give the title compound as a yellow foam that was used directly in the subsequent step. MS (ES+) m/z 595 (M+H)+
Step 3: methyl riaR.5S.8S.10R.18E.22aRV5-tert-butyl-14-methoxy-3.6-dioxo- l,la.3Λ5,6.9.10.20^1.22.22a-dodecahvdro-8H-7,10- methanocyclopropa[18J9in.l0.3,6]dioxadiazacvclononadecino[l lJ2-b1quinoxaline-8- carboxylate
Figure imgf000019_0002
A solution (0.02 M) of methyl 3-methyl-N-({[(lR,2R>2-pent-4-en-l- ylcyclopropyl]oxy}carbonyl)-L-valyl-(4R)-4-[(7-methoxy-3-vinylquinoxalin-2-yl)oxy]-L- prolinate in DCE was heated to 80 0C then treated with Zhan 1 catalyst (0.15 eq). The resulting mixture was stirred at 80 °C for 1 h then cooled to room temperature and concentrated under reduced pressure. The residue was purified by flash chromatography (20-50% EtOAc/ petroleum ether) to give the title compound (25% for 2 steps) as a foam. MS (ES+) m/z 567 (M+H)+
Step 4: methyl αaR.5S.8S.10R.22aRV5-tert-butyl-14-methoxy-3.6-dioxo- l.la.3.4.5.6.9.10.18.19.20.21.22^2a-tetradecahvdro-8H-7.10- methanocvclopropa[ 18 Λ 9) [ IJ 0,3 ^Idioxadiazacvclononadecinor 11.12-b]quinoxaline-8- carboxγlate
Figure imgf000020_0001
A solution (0.05 M) of methyl (laR,5S,8S,10R, 18E,22aR)-5-tert-butyl-14- methoxy-3,6-dioxo-l,la,3,4,5,6,9,10,20,21 ,22,22a-dodecahydro-8H-7,10- methanocyclopropa[ 18, 19] [ 1, 10,3 ,6] dioxadiazacyclononadecino [11,12-b]quinoxaline-8- carboxylate in MeOH/dioxane (1: 1 ratio) was treated with Pd/C (8% in weight). The resulting mixture was stirred under atmosphere of hydrogen for 4 h. The catalyst was filtered off and the filtrate was concentrated under reduced pressure to give the title compound (98%) as a solid. MS (ES+) m/z 569 (M+H)+
Step 5: (laR,SS.8S.10It22aRVS-tert-butγl-14-methoxy-3.6-dioxo- l.la.3.4.5.6.9.10J 8.19^0^U2^2a-tetradecahvdro-8H-7.10- methanocyclopropaf 18.191 f 1.10,3.όldioxadiazacyclononadecinof 1 1,12-b1quinoxaline-8- carboxylic acid
Figure imgf000020_0002
A solution (0.1 M) of methyl (laR,5S,8S,10R,22aR)-5-tert-butyl-14-methoxy-3)6- dioxo- 1 , 1 a,3 ,4,5,6,9, 10, 18, 19,20,21 ,22,22a-tetradecahydro-8H-7, 10- methanocyclopropa[ 18, 19] [1 , 10,3,6]dioxadiazacyclononadecino[l 1 , 12-b]quinoxaline-8- carboxylate in a 1 :1 mixture Of H2OzTHF was treated with LiOH-H2O (3 eq). The resulting mixture was stirred at 20 0C for 18 h, acidified with aqueous HCl (0.2 M) and diluted with EtOAc. The organic phase was separated, washed with aqueous HCl (0.2 M) and brine then dried. Removal of the volatiles afforded the title compound (98%) as a solid. MS (ES+) m/z 555 (M+H)+
Step 6: (I aR.5S.8S.10R^2aRV 5-tert-butyl-N-(f 1 RJ2SV 1 - ( [(cvclopropylsulfonvnaminol carbonvU-2-vinylcvclopropyl>-14-methoxy-3.6-dioxo-l.la.3.4,5.6,9.10.18J9,20^1^2^2a- tetradecahydro-8H-7, 10-methanocvclopropaf 18.191[Ll OJ^ldioxadiazacvclononadecinof 11.12- biquinoxaline-8-carboχamide
Figure imgf000021_0001
A solution (0.1 M) of (laR,5S,8S,10Rt22aR)-5-tert-butyl-14-methoxy-3,6-dioxo- 1 , 1 a,3,4,5,6,9, 10, 18, 19,20,21 ,22,22a-tetradecahydro-8H-7, 10- methanocyclopropa[ 18, 19] [ 1, 10,3,6]dioxadiazacyclononadecino[l 1 , 12-b]quinoxaIine-8- carboxylic acid in CH2Cl2 was treated with (lR,2S)-l-{[(cyclopropylsulfonyl)amino]carbonyl}- 2-vinylcyclopropanaminium chloride (1.3 eq), DIEA (3 eq), DMAP (1.5 eq) and TBTU (1.45 eq), The resulting mixture was stirred at 20 0C for 18 h and then diluted with EtOAc. The solution was washed with aqueous HCl (0.2 M), saturated aqueous NaHCO3 and brine. The organic phases were dried and concentrated to give a residue that was purified by flash- chromatography (eluent 2.5% MeOH/CH2Cl2) to give the title compound (89%) as a solid. 13C NMR (100 MHz, DMSO-^) δ 172.32, 170.63, 169.04, 159.86, 156.95, 154.74, 148.10, 140.41, 133.55 (2 signals), 128.94, 118.21, 1 17.58, 105.89, 74.88, 59.75, 58.71, 55.68, 54.13, 54.01, 40.13, 34.49, 34.04, 33.76, 32.68, 30.71, 30.43, 28.55, 27.69, 27.28, 26.38, 21.98, 18.49, 10.67, 5.69, 5.46; MS (ES+) m/z 767 (M+H)+ Step 7: potassium U(lR.2SVHU(laR.5S.8S.10R.22aRV5-tert-butyl-14-mctfaoxy-3.6-dioxo- 1.1 a.3 A5.6.9.10.18.19.20.21.22.22a-tetradecahvdro-8H-7, 10- methanocyclopropa[ 18.19] [ 1 , 10,3.βjdioxadiazacyclononadecinof 1 1.12-b~|quinoχalin-8- vϊi carbonyl ) amino)-2-vinylcyclopropyl] carbonyl } (cyclopropylsulfonyPazanide
Figure imgf000022_0001
The preceding material was taken up in EtOH and the resulting solution (0.025 M) was cooled to 0 0C. A solution (0.02 M) of /erf-BuOK (1.5 eq) in EtOH was added leading to the formation of a precipitate. The mixture was stirred at 20 0C for 18 h then the solid was collected by filtration. This material was washed with EtOH and dried to give the title compound (93%) as a white crystalline solid. MS (ES+) m/z 761 (M+H)+
Example 2: Comparison of Different Compounds
The compound of Example 1 was compared to the compound of Examples 110 and 118 of WO 2008/057209. The results are shown in Tables 1 and 2 below. As illustrated in the tables and the discussion of the results, the compound of formula (I) appears to have several advantageous properties compared to both the WO 2008/057209 Example 1 18 compound and the WO 2008/057209 Example 110 compound.
Figure imgf000023_0001
Ki: Inhibition constant, reference to < 0.016 nM indicates that the observed activity is less than 0.016 nM, the exact amount less than 0.016 nM was not determined by the assay; EC50; Effective concentration achieving 50% viral replication suppression; gt: Genotype; AUC: Area Under the plasma concentration/time curve; LOQ: Limit of quantitation (3 pmol/mg); BLQ: Below limit of quantitation.
Formula (I) Compound Compared to WO 2008/057209 Example UO
Advantageous properties of the formula (I) compound versus the WO 2008/057209 Example 110 compound are the following: 1 ) Physical properties (no salt disproportionation for the Compound of formula (I));
2) Pharmacokinetic profile in rats following administration of the potassium salt; and
3) Liver (target organ) exposure.
The differences in properties are particularly advantageous for the formulation and administration of the formula (I) compound compared to the WO 2008/057209 Example 1 10 compound. The lack of salt disproportionation for the formula (I) compound enables dissolution of 1.8 mg/ml of the K+ salt form of the Compound of Example 1 in water. Although the K+ salt form of the WO 2008/057209 Example 110 compound has improved aqueous solubility (9.7 mg/mL), the compound thus dissolved disproportionates to give a crystalline zwitterionic form which has low aqueous solubility (<0.009 mg/ml). The lack of this behavior for the Compound of Example 1 provides an unexpected advantage in its formulation for pharmaceutical administration and results in improved pharmacokinetic properties as reported in Table 1 (plasma AUC and liver exposure for rat and dog). High plasma and liver exposure in preclinical species is advantageous for the selection of safe and efficacious doses for use in the treatment of patients.
Formula (I) Compound Compared to WO 2008/057209 Example 118
An observed advantage of the compound of formula (I) compared to
WO2008/057209 Example 118 is its resistance profile against different mutant enzymes. In line with data from clinical studies with antiviral agents from related classes (e.g. HTV protease inhibitors), and also from studies with HCV NS3 protease inhibitors (e.g., VX -950, telaprevir), it is expected that viral resistance may develop in response to treatment with the current compounds. The compound of Example 1 showed improved enzyme affinity (Ki) against different mutant enzymes that are known to confer resistance to HCV NS3 protease inhibitors. Table 2 summarizes activity against different mutant enzymes. Thus, an advantage of compound 1 may be an increased barrier to the development of resistant virus when administered to patients. It also provides the potential advantage to treat patients who have failed other therapies because of the development of resistance, since compound 1 may inhibit this resistant virus.
Figure imgf000025_0001
1 Comparative data collected in the same run of the enzyme assays
Further expected advantageous properties of the formula (I) compound versus the WO 2008/057209 Example 110 compound include the following:
1) Low in vi vo co valent binding; and
2) High plasma and liver exposure,
The formula (I) compound was found to have very good covalent binding in vivo characteristics and pharmacokinetic properties. Based on observed covalent binding in vivo and pharmacokinetic properties of the Example 1 compound and the WO 2008/057209 Example 118 compound, the formula (I) compound has significantly better in vivo covalent binding characteristics and pharmacokinetic properties .
Compounds which covalently bind to proteins, or which form metabolites that subsequently become covalently bound to proteins, potentially give rise to adverse events in patients such as immunological toxicities mediated by antibody responses to the drug-protein conjugate, and other idiosyncratic toxicities. (See Chem. Res. Toxicol. 2004, 17, 3-16.)
The compound of Example 1 showed undetectable binding to plasma proteins following oral administration of a single 20 mg/kg dose to rats. (See Table 1.) Under analogous conditions, the WO 2008/057209 Example 118 compound demonstrated detectable binding to rat liver proteins (see Table 1), and therefore may be considered a less advantageous compound for administration to human subjects than the compound of formula (I).
Table 3 provides some additional in vivo covalent binding data observed for related compounds from WO 2008/057209 that contain the (R,R)-trans-2-alkylcyclopentanol moiety incorporated in Example 118.
Figure imgf000026_0001
It is advantageous to have high plasma and liver exposure in preclinical species to effectively demonstrate that the potential drug candidate does not elicit undesired toxicities. It is also more likely that a compound that has high liver and plasma exposure in animals displays the same behavior in man than one that does not. For such a compound, the required efficacious exposure in man can be reached with a lower dose, advantageous both for the cost and ease of manufacturing the drug, but also potentially lowering the likelihood of adverse effects. Target organ exposure in multiple preclinical species provides a rationale that high target organ exposure is achievable for the compound in patients, and high liver exposure in both rat and dog allows for confident evaluation of preclinical toxicity. High liver exposure is especially advantageous for HCV since this is the target organ for the drug.
The compound of Example 1 had a very good rat plasma and liver exposure. The observed rat liver exposure was at a level greater than for WO 2008/057209 compound 118 and compound 1 10. (See Table 1) Based on these results and on testing several different compounds from WO 2008/057209 by oral administration to both rat (25mpk) and dog (5mpk), the compound of formula (I) is also expected to have dog liver exposures greater than WO2008/057209 compound 1 18 and compound 110.
Methods
NS 3/4A Inhibitory Activity1 (Ki): NS 3/4A Inhibitory Activity was determined as described in Section IV. Compound Evaluation supra., and Mao et al, Anal Biochem 373\\- 8, 2008. Rcplicon Activity2 ECs^: Replicon Activity was determined using the procedures described in Carroll et al, J. Biol. Chem. 278:\ 1979-1 1984, 2003 and Olsen et al., ArUi Microb. Agents 4S-.3944-3953, 2004.
Rat Plasma AUC (a). 25mpk per os3: Test compounds were dissolved in a suitable dosing vehicle for iv administration (e.g. 20%:60%:20% DMSO:PEG400:Water) or per os administration (e.g., 10% POLYSORBATE80: 90% Water or 100% PEG400). Animals were administered (n = 3) using a crossover study design for non-rodents. Plasma samples were collected at time points between 2 minutes and 24 h, and compound levels were determined by RP-HPLC. Liver samples were collected post mortem in rat and following anesthesia (0.5 h prior to biopsy) in dog. Liver samples were weighed, homogenized, and diluted using techniques known to those skilled in the art, and compound levels were determined by RP- HPLC.
Pharmacokinetic parameters were calculated based on non-compartmental analysis (e.g. using WATSON®, WΓNNOLΓN®). Predose concentrations that were below the limit of quantitation (BLQ) were assigned a value of 0. For oral AUC estimation, the first BLQ value in the terminal phase were given a value equal to 1 /2 Lowest Limit Of Quantitation, while subsequent values in the terminal phase were assigned a value of 0. Standard pharmacokinetic parameters CLp, Vdss, half-life (only for IV), % F, Cm, Tm8x, AUQ>-Ust> AUCo-.πfinity were calculated. AUC values were calculated using linear trapezoidal method for ascending concentrations and the log trapezoidal method for descending concentrations.
In-Vivo Covalent Binding : Test compounds were suitably radiolabeled (3H) and a 20 mg/kg dose containing 25-75 mCi/rat (purity >98.5%) radioactivity was prepared by combination of the cold compound and evaporated radiotracer stock solution. This mixture was dissolved in a dosing vehicle suitable for per os administration (see above) then administered orally to rat (n = 3 per timepoint, 2h, 6h, 24h). Plasma and liver were collected and flash frozen/stored at -80°C before analysis.
Counting of the plasma samples: Place a 200 μL aliquot in a 20 mL scintillation vial. Add 500 μL of SOLVABLE™ and incubate at 1 h with shaking at 55°C. Remove, allow to cool prior to the addition of 15 mL scintillation cocktail, and count. Plasma samples (200 μL aliquot) were then processed as described below for liver proteins.
Tissue homogenization: Weighed liver samples were diluted with 2 vol 100 mM phosphate buffer (pH 7.4) and homogenized on ice.
Counting of the liver homogenate: Aliquots were placed in a 20 mL scintillation vial, diluted with 1 mL of SOLVABLE™ and incubated for 1 h with shaking at 55°C. After removal from the incubator and cooling 15 mL scintillation cocktail and 30% H2O2 were added and the radioactivity counted Protein precipitation: Take 500 μL aliquot, add 1 :8 homogenate:acetonitrile (if compound is suspected to have low solubility in acetonitrile, another solvent may be selected), vortex and centrifuge (3500 rcf for 20 min). Discard the supernatant.
Protein precipitate resuspension: Sonication (minimal intensity, < 5 sec) and vortexing until the pellet crumbles in 80% MeOH: 20% water.
Wash of the protein pellet: 2-5 mL 80:20 MeOH: water. If needed, remove 1.0 mL of the supernatant, add 15 mL of scintillation cocktail and count. Continue to wash the protein pellet until radioactivity in supernatant is < 200 DPM or DPMs cease to decrease by more than 200 in consecutive washes.
Dissolution of the final pellet: 1 mL of 1 N NaOH or SOLVABLE™, incubated at 500C overnight or until completely dissolved.
Counting of the final pellet: 1 mL of dissolved pellet, 15 mL scintillation cocktail (if another scintillation cocktail other than ULTIMA GOLD™ is used, pellet may require neutralization using 1 N HCl), and count.
Protein concentration of the final pellet: BCA or BlO-RAD kit using BSA as a standard.
Counting blank samples: 15 mL scintillation cocktail, in duplicate.
Counting of the dosing solution: Count a known volume of the dosing solution in triplicate.
Data Analysis: Average the radioactivity counts (DPM) of the dosing solution and calculate the specific activity of the dosing solution in μCi/mol. Average the radioactivity counts of the blank samples. Subtract the counts of the averaged blank sample from the counts obtained from each liver and plasma pellet. Calculate the amount of radioactivity (μCi) per unit volume (L) for each liver and plasma pellet. Calculate the concentration of radioactivity in each plasma and liver pellet by dividing the value obtained above (μCi/L) by the specific activity (μCi/mol). Calculate the amount of covalently bound radioactivity to protein in pmol/mg protein.
Counting blank samples: 15 mL scintillation cocktail, in duplicate.
Counting of the dosing solution: count a known volume of the dosing solution in triplicate.
Physical properties5: The crystalline test compounds (potassium salt, ca. 5 mg) was weighed in a glass vial and water or aqueous buffer was added (100 μL). The slurry obtained was stirred for 24 h at room temperature. After centrifuging, the supernatant was analyzed by reversed-phase HPLC and the equilibrium solubility determined by comparison with a calibration curve. The solid material was in part transferred onto an XRPD plate, dried and then analyzed by X-Ray Powder Diffraction. The XRPD pattern was compared with positive controls for crystalline K+ salt, crystalline zwitterionic (or acidic) and amorphous forms of the test compound. A further determination of the salt form was obtained from a second portion of the solid material which was analyzed by 400 MHz NMR (Bruker) following dissolution in DMSO-^6- IH-NMR spectra were compared to positive controls described above.
None of the references described throughout the present application are admitted to be prior art to the claimed invention.
Other embodiments are within the following claims. While several embodiments have been shown and described, various modifications may be made without departing from the spirit and scope of the present invention.

Claims

WHAT IS CLAIMED IS:
1. A compound of formula (I), or a pharmaceutically acceptable salt thereof:
Figure imgf000030_0001
2. A pharmaceutical composition comprising an effective amount of the compound of claim 1 and a pharmaceutically acceptable carrier.
3. The pharmaceutical composition of claim 2, further comprising a second therapeutic agent selected from the group consisting of a HCV protease inhibitor and a HCV NS5B polymerase inhibitors.
4. The compound of claim 1 for use in medicine.
5. The compound of claim 1 for use in prevention or treatment of HCV infection.
6. Use of the compound or composition according to anyone of claims 1-3 in the preparation of a medicament for inhibiting HCV NS3 protease activity in a subject in need thereof.
7. Use of the compound or composition according to anyone of claims 1-3 in the preparation of a medicament for preventing or treating infection by HCV in a subject in need thereof.
8. A method of treating a patient infected with HCV comprising the step of administering to said patient an effective amount of the compound or composition of anyone of claims 1-3.
PCT/US2009/050915 2008-07-22 2009-07-17 Macrocyclic quinoxaline compounds as hcv ns3 protease inhibitors WO2010011566A1 (en)

Priority Applications (26)

Application Number Priority Date Filing Date Title
NZ590638A NZ590638A (en) 2008-07-22 2009-07-17 MACROCYCLIC QUINOXALINE COMPOUNDS AS Hepatitis C Virus NS3 PROTEASE INHIBITORS
JP2011520110A JP4920797B2 (en) 2008-07-22 2009-07-17 Macrocyclic quinoxaline compounds as HCV NS3 protease inhibitors
EP09790553A EP2310095B1 (en) 2008-07-22 2009-07-17 Macrocyclic quinoxaline compounds as hcv ns3 protease inhibitors
EA201170241A EA019327B1 (en) 2008-07-22 2009-07-17 Macrocyclic quinoxaline compounds as hcv ns3 protease inhibitors
KR1020117003982A KR101313675B1 (en) 2008-07-22 2009-07-17 Macrocyclic quinoxaline compounds as HCV NS3 Protease inhibitors
SI200930359T SI2310095T1 (en) 2008-07-22 2009-07-17 Macrocyclic quinoxaline compounds as hcv ns3 protease inhibitors
PL09790553T PL2310095T3 (en) 2008-07-22 2009-07-17 Macrocyclic quinoxaline compounds as hcv ns3 protease inhibitors
AU2009274190A AU2009274190B2 (en) 2008-07-22 2009-07-17 Macrocyclic quinoxaline compounds as HCV NS3 protease inhibitors
CA2731177A CA2731177C (en) 2008-07-22 2009-07-17 Macrocyclic quinoxaline compounds as hcv ns3 protease inhibitors
ES09790553T ES2392611T3 (en) 2008-07-22 2009-07-17 Macrocyclic quinoxaline compounds as HCV NS3 protease inhibitors
RS20120463A RS52534B (en) 2008-07-22 2009-07-17 Macrocyclic quinoxaline compounds as hcv ns3 protease inhibitors
MEP-2012-463A ME02024B (en) 2008-07-22 2009-07-17 Macrocyclic quinoxaline compounds as hcv ns3 protease inhibitors
MX2011000826A MX2011000826A (en) 2008-07-22 2009-07-17 Macrocyclic quinoxaline compounds as hcv ns3 protease inhibitors.
BRPI0916235A BRPI0916235B8 (en) 2008-07-22 2009-07-17 compound, pharmaceutical composition, and, use of the compound or composition
DK09790553.3T DK2310095T3 (en) 2008-07-22 2009-07-17 MACROCYCLIC QUINOXALIN COMPOUNDS AS HCV-NS3 PROTEASE INHIBITORS
CN200980137118.6A CN102159285B (en) 2008-07-22 2009-07-17 Macrocyclic quinoxaline compounds as HCV NS3 protease inhibitors
IL210580A IL210580A (en) 2008-07-22 2011-01-11 Macrocyclic quinoxaline compound as hcv ns3 protease inhibitor
TN2011000014A TN2011000014A1 (en) 2008-07-22 2011-01-13 Macrocyclic quinoxaline compounds as hcv ns3 protease inhibitors
MA33556A MA32502B1 (en) 2008-07-22 2011-01-25 MACROCYCLIC QUINOXALINE COMPOUNDS AS HEPATITIS C VIRUS (HCV) NS3 PROTEASE INHIBITORS
HRP20120866AT HRP20120866T1 (en) 2008-07-22 2012-10-26 Macrocyclic quinoxaline compounds as hcv ns3 protease inhibitors
FR16C1027C FR16C1027I2 (en) 2008-07-22 2016-12-26 MACROCYCLIC QUINOXALINE COMPOUNDS AS HEPATITIS C VIRUS (HCV) NS3 PROTEASE INHIBITORS
LTPA2016049C LTC2310095I2 (en) 2008-07-22 2016-12-29 Macrocyclic quinoxaline compounds as inhibitors of HCV NS3 protease
NL300857C NL300857I2 (en) 2008-07-22 2016-12-30
LU00002C LUC00002I2 (en) 2008-07-22 2017-01-03
NO2017004C NO2017004I1 (en) 2008-07-22 2017-01-20 Grazoprevir or a pharmaceutically acceptable substance thereof
CY2017005C CY2017005I2 (en) 2008-07-22 2017-01-20 QUINOXALINE MACROCYCLIC COMPOUNDS AS INHIBITORS OF HCV NS3 PROTEINS

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US13555908P 2008-07-22 2008-07-22
US61/135,559 2008-07-22

Publications (1)

Publication Number Publication Date
WO2010011566A1 true WO2010011566A1 (en) 2010-01-28

Family

ID=41130248

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2009/050915 WO2010011566A1 (en) 2008-07-22 2009-07-17 Macrocyclic quinoxaline compounds as hcv ns3 protease inhibitors

Country Status (44)

Country Link
US (2) US7973040B2 (en)
EP (3) EP2310095B1 (en)
JP (1) JP4920797B2 (en)
KR (1) KR101313675B1 (en)
CN (1) CN102159285B (en)
AR (1) AR072588A1 (en)
AU (1) AU2009274190B2 (en)
BR (1) BRPI0916235B8 (en)
CA (1) CA2731177C (en)
CL (1) CL2011000145A1 (en)
CO (1) CO6351757A2 (en)
CR (1) CR20110089A (en)
CY (3) CY1113752T1 (en)
DK (2) DK2540350T3 (en)
DO (1) DOP2011000023A (en)
EA (1) EA019327B1 (en)
EC (1) ECSP11010777A (en)
ES (2) ES2392611T3 (en)
FR (1) FR16C1027I2 (en)
HK (2) HK1173402A1 (en)
HN (1) HN2011000209A (en)
HR (2) HRP20120866T1 (en)
HU (1) HUS1700001I1 (en)
IL (1) IL210580A (en)
LT (1) LTC2310095I2 (en)
LU (1) LUC00002I2 (en)
MA (1) MA32502B1 (en)
ME (2) ME02132B (en)
MX (1) MX2011000826A (en)
MY (1) MY152070A (en)
NI (1) NI201100023A (en)
NL (1) NL300857I2 (en)
NO (1) NO2017004I1 (en)
NZ (1) NZ590638A (en)
PE (1) PE20110212A1 (en)
PL (2) PL2540350T3 (en)
PT (2) PT2540350E (en)
RS (2) RS53420B (en)
SI (2) SI2310095T1 (en)
SV (1) SV2011003813A (en)
TN (1) TN2011000014A1 (en)
TW (1) TWI441638B (en)
UA (1) UA100436C2 (en)
WO (1) WO2010011566A1 (en)

Cited By (23)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7973040B2 (en) 2008-07-22 2011-07-05 Merck Sharp & Dohme Corp. Macrocyclic quinoxaline compounds as HCV NS3 protease inhibitors
WO2012050850A1 (en) * 2010-09-29 2012-04-19 Merck Sharp & Dohme Corp. Polycyclic heterocycle derivatives and methods of use thereof for the treatment of viral diseases
WO2013028470A1 (en) 2011-08-19 2013-02-28 Merck Sharp & Dohme Corp. Process and intermediates for preparing macrolactams
WO2013074386A2 (en) 2011-11-15 2013-05-23 Merck Sharp & Dohme Corp. Hcv ns3 protease inhibitors
KR20130098369A (en) * 2010-09-21 2013-09-04 이난타 파마슈티칼스, 인코포레이티드 Macrocyclic proline derived hcv serine protease inhibitors
WO2014008285A1 (en) 2012-07-03 2014-01-09 Gilead Sciences, Inc. Inhibitors of hepatitis c virus
US8828930B2 (en) 2009-07-30 2014-09-09 Merck Sharp & Dohme Corp. Hepatitis C virus NS3 protease inhibitors
WO2014145095A1 (en) * 2013-03-15 2014-09-18 Gilead Sciences, Inc. Macrocyclic and bicyclic inhibitors of hepatitis c virus
US8957203B2 (en) 2011-05-05 2015-02-17 Bristol-Myers Squibb Company Hepatitis C virus inhibitors
WO2015095430A1 (en) * 2013-12-20 2015-06-25 Merck Sharp & Dohme Corp. Methods and intermediates for the preparation of macrolactams
WO2015100145A1 (en) * 2013-12-23 2015-07-02 Gilead Sciences, Inc. Synthesis of a macrocyclic hcv ns3 inhibiting tripeptide
WO2015100144A1 (en) * 2013-12-23 2015-07-02 Gilead Sciences, Inc. Crystalline forms of a macrocyclic hcv ns3 inhibiting tripeptide
EP2773342A4 (en) * 2011-10-31 2015-08-26 Merck Sharp & Dohme Compositions useful for the treatment of viral diseases
US9120818B2 (en) 2010-12-14 2015-09-01 Merck Sharp & Dohme Corp. Process and intermediates for preparing macrolactams
US9334279B2 (en) 2012-11-02 2016-05-10 Bristol-Myers Squibb Company Hepatitis C virus inhibitors
US9409943B2 (en) 2012-11-05 2016-08-09 Bristol-Myers Squibb Company Hepatitis C virus inhibitors
US9499550B2 (en) 2012-10-19 2016-11-22 Bristol-Myers Squibb Company Hepatitis C virus inhibitors
US9580463B2 (en) 2013-03-07 2017-02-28 Bristol-Myers Squibb Company Hepatitis C virus inhibitors
US9598433B2 (en) 2012-11-02 2017-03-21 Bristol-Myers Squibb Company Hepatitis C virus inhibitors
US9643999B2 (en) 2012-11-02 2017-05-09 Bristol-Myers Squibb Company Hepatitis C virus inhibitors
EP3057926A4 (en) * 2013-10-18 2017-07-12 Merck Sharp & Dohme Corp. Methods and intermediates for preparing macrolactams
WO2017198102A1 (en) * 2016-05-16 2017-11-23 深圳市塔吉瑞生物医药有限公司 Substituted macrocyclic quinoxaline compound and pharmaceutical composition and use thereof
WO2021209563A1 (en) 2020-04-16 2021-10-21 Som Innovation Biotech, S.A. Compounds for use in the treatment of viral infections by respiratory syndrome-related coronavirus

Families Citing this family (34)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
MY140680A (en) 2002-05-20 2010-01-15 Bristol Myers Squibb Co Hepatitis c virus inhibitors
BRPI0718161A2 (en) * 2006-10-27 2013-11-26 Merck & Co Inc COMPOUND, PHARMACEUTICAL COMPOSITION, AND, USE OF THE COMPOUND.
US8461107B2 (en) * 2008-04-28 2013-06-11 Merck Sharp & Dohme Corp. HCV NS3 protease inhibitors
SI2410844T1 (en) 2009-03-27 2016-06-30 Merck Sharp & Dohme Corp. Inhibitors of hepatitis c virus replication
US20190127365A1 (en) 2017-11-01 2019-05-02 Merck Sharp & Dohme Corp. Inhibitors of hepatitis c virus replication
WO2010132163A1 (en) * 2009-05-13 2010-11-18 Enanta Pharmaceuticals, Inc. Macrocyclic compounds as hepatitis c virus inhibitors
RS53856B1 (en) * 2009-06-11 2015-08-31 Abbvie Bahamas Ltd. Heterocyclic compounds as inhibitors of hepatitis c virus (hcv)
US8937150B2 (en) 2009-06-11 2015-01-20 Abbvie Inc. Anti-viral compounds
NZ605440A (en) 2010-06-10 2014-05-30 Abbvie Bahamas Ltd Solid compositions comprising an hcv inhibitor
AU2011314168A1 (en) 2010-09-29 2013-04-04 Merck Sharp & Dohme Corp. Fused tetracycle derivatives and methods of use thereof for the treatment of viral diseases
WO2012122716A1 (en) 2011-03-17 2012-09-20 Merck Sharp & Dohme Corp. Tetracyclic xanthene derivatives and methods of use thereof for treatment of viral diseases
US10201584B1 (en) 2011-05-17 2019-02-12 Abbvie Inc. Compositions and methods for treating HCV
US9249204B2 (en) * 2011-06-01 2016-02-02 Jyant Technologies, Inc. Chemokine-immunoglobulin fusion polypeptides, compositions, method of making and use thereof
US8492386B2 (en) 2011-10-21 2013-07-23 Abbvie Inc. Methods for treating HCV
EA201490837A1 (en) 2011-10-21 2014-11-28 Эббви Инк. METHODS OF TREATING HCV, INCLUDING, AT THE LITERATURE, TWO ANTI-VIRAL AGENTS OF DIRECT ACTION, RIBAVIRIN, BUT NOT INTERFERON
DE112012003457T5 (en) 2011-10-21 2015-03-12 Abbvie Inc. Combination treatment (e.g., with ABT-072 or ABT-333 from DAAs for use in the treatment of HCV)
US8466159B2 (en) 2011-10-21 2013-06-18 Abbvie Inc. Methods for treating HCV
US9034832B2 (en) 2011-12-29 2015-05-19 Abbvie Inc. Solid compositions
WO2014110687A1 (en) 2013-01-16 2014-07-24 Merck Sharp & Dohme Corp. Thiazolyl-substitued tetracyclic compounds and methods of use thereof for treatment of viral diseases
US11484534B2 (en) 2013-03-14 2022-11-01 Abbvie Inc. Methods for treating HCV
WO2015103490A1 (en) 2014-01-03 2015-07-09 Abbvie, Inc. Solid antiviral dosage forms
EP3102210A4 (en) * 2014-02-05 2017-10-11 Merck Sharp & Dohme Corp. Pharmaceutical composition of selective hcv ns3/4a inhibitors
WO2015119924A2 (en) * 2014-02-05 2015-08-13 Merck Sharp & Dohme Corp. Fixed-dose combinations of antiviral compounds
CA2948902C (en) 2014-06-06 2022-11-22 Abbvie Inc. Crystal forms of hcv protease inhibitors and use thereof
US20180228828A1 (en) * 2015-08-04 2018-08-16 Merck Sharp & Dohme Corp. Fixed-dose combinations of antiviral compounds
WO2017023714A1 (en) * 2015-08-04 2017-02-09 Merck Sharp & Dohme Corp. Fixed-dose combinations of antiviral compounds
CN105753806B (en) * 2016-02-02 2018-06-05 厦门市蔚嘉化学科技有限公司 A kind of heterogeneous synthetic method of Ritonavir intermediate and its application
EA201892448A1 (en) 2016-04-28 2019-06-28 Эмори Юниверсити ALKYN-CONTAINING NUCLEOTIDE AND NUCLEOSIDE THERAPEUTIC COMPOSITIONS AND RELATED APPLICATION METHODS
WO2017197036A1 (en) 2016-05-10 2017-11-16 C4 Therapeutics, Inc. Spirocyclic degronimers for target protein degradation
WO2017197055A1 (en) 2016-05-10 2017-11-16 C4 Therapeutics, Inc. Heterocyclic degronimers for target protein degradation
CN109641874A (en) 2016-05-10 2019-04-16 C4医药公司 C for target protein degradation3The glutarimide degron body of carbon connection
RU2650610C1 (en) 2017-02-28 2018-04-16 Васильевич Иващенко Александр Antiviral composition and method of its application
WO2020247736A1 (en) * 2019-06-07 2020-12-10 University Of Massachusetts Hepatitis c virus ns3/4a protease inhibitors
CN111057045A (en) * 2019-12-18 2020-04-24 安徽红杉生物医药科技有限公司 HCV NS3/4A protease inhibitor intermediate, and synthesis method and application thereof

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2006119061A2 (en) * 2005-05-02 2006-11-09 Merck & Co., Inc. Hcv ns3 protease inhibitors
WO2007016441A1 (en) * 2005-08-01 2007-02-08 Merck & Co., Inc. Macrocyclic peptides as hcv ns3 protease inhibitors
WO2008002924A2 (en) * 2006-06-26 2008-01-03 Enanta Pharmaceuticals, Inc. Quinoxalinyl macrocyclic hepatitis c virus serine protease inhibitors
WO2008057209A1 (en) * 2006-10-27 2008-05-15 Merck & Co., Inc. Hcv ns3 protease inhibitors

Family Cites Families (103)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3480613A (en) 1967-07-03 1969-11-25 Merck & Co Inc 2-c or 3-c-alkylribofuranosyl - 1-substituted compounds and the nucleosides thereof
US6128582A (en) 1996-04-30 2000-10-03 Vertex Pharmaceuticals Incorporated Molecules comprising an IMPDH-like binding pocket and encoded data storage medium capable of graphically displaying them
GB9623908D0 (en) 1996-11-18 1997-01-08 Hoffmann La Roche Amino acid derivatives
GB9707659D0 (en) 1997-04-16 1997-06-04 Peptide Therapeutics Ltd Hepatitis C NS3 Protease inhibitors
AU757072B2 (en) 1997-08-11 2003-01-30 Boehringer Ingelheim (Canada) Ltd. Hepatitis C inhibitor peptide analogues
DE69829381T2 (en) 1997-08-11 2006-04-13 Boehringer Ingelheim (Canada) Ltd., Laval HEPATITIS C INHIBITOR PEPTIDE
IT1299134B1 (en) 1998-02-02 2000-02-29 Angeletti P Ist Richerche Bio PROCEDURE FOR THE PRODUCTION OF PEPTIDES WITH PROTEAS INHIBITING THE NS3 PROTEASIS OF THE HCV VIRUS, PEPTIDES SO OBTAINABLE AND PEPTIDES
ES2276515T3 (en) 1998-02-25 2007-06-16 Emory University 2'-FLUORONUCLEOSIDS.
GB9806815D0 (en) 1998-03-30 1998-05-27 Hoffmann La Roche Amino acid derivatives
WO1999050230A1 (en) 1998-03-31 1999-10-07 Vertex Pharmaceuticals Incorporated Inhibitors of serine proteases, particularly hepatitis c virus ns3 protease
GB9812523D0 (en) 1998-06-10 1998-08-05 Angeletti P Ist Richerche Bio Peptide inhibitors of hepatitis c virus ns3 protease
US6602982B1 (en) 1998-08-10 2003-08-05 Hokkaido Electric Power Company, Incorporated Process for the preparation of regular glycopeptides
US6323180B1 (en) 1998-08-10 2001-11-27 Boehringer Ingelheim (Canada) Ltd Hepatitis C inhibitor tri-peptides
WO2000025780A1 (en) 1998-10-29 2000-05-11 Bristol-Myers Squibb Company Compounds derived from an amine nucleus that are inhibitors of impdh enzyme
UA74546C2 (en) 1999-04-06 2006-01-16 Boehringer Ingelheim Ca Ltd Macrocyclic peptides having activity relative to hepatitis c virus, a pharmaceutical composition and use of the pharmaceutical composition
US6608027B1 (en) 1999-04-06 2003-08-19 Boehringer Ingelheim (Canada) Ltd Macrocyclic peptides active against the hepatitis C virus
MXPA02000294A (en) 1999-06-25 2002-06-21 Vertex Pharma Prodrugs of carbamate inhibitors of impdh.
CN1623984A (en) 1999-12-27 2005-06-08 日本烟草产业株式会社 Fused-ring compounds and use thereof as drugs
US6455508B1 (en) 2000-02-15 2002-09-24 Kanda S. Ramasamy Methods for treating diseases with tirazole and pyrrolo-pyrimidine ribofuranosyl nucleosides
US6495677B1 (en) 2000-02-15 2002-12-17 Kanda S. Ramasamy Nucleoside compounds
AU2001235278A1 (en) 2000-02-18 2001-08-27 Shire Biochem Inc Method for the treatment or prevention of flavivirus infections using nucleoside analogues
KR20030036152A (en) 2000-04-05 2003-05-09 쉐링 코포레이션 Macrocyclic NS3-serine protease inhibitors of hepatitis C virus comprising N-cyclic P2 moieties
US6727267B2 (en) 2000-04-05 2004-04-27 Tularik Inc. NS5B HVC polymerase inhibitors
EP1284741B1 (en) 2000-04-13 2008-11-19 Pharmasset, Inc. 3'-or 2'-hydroxymethyl substituted nucleoside derivatives for treatment of viral infections
MY164523A (en) 2000-05-23 2017-12-29 Univ Degli Studi Cagliari Methods and compositions for treating hepatitis c virus
CN101099745A (en) 2000-05-26 2008-01-09 艾登尼科斯(开曼)有限公司 Methods and compositions for treating flaviviruses and pestiviruses
US6448281B1 (en) 2000-07-06 2002-09-10 Boehringer Ingelheim (Canada) Ltd. Viral polymerase inhibitors
GB0017676D0 (en) 2000-07-19 2000-09-06 Angeletti P Ist Richerche Bio Inhibitors of viral polymerase
JP3943015B2 (en) 2000-08-10 2007-07-11 トラスティーズ オブ ボストン カレッジ Recyclable metathesis catalyst
US6955174B2 (en) 2000-08-18 2005-10-18 Uryovascular Systems, Inc. Cryotherapy method for detecting and treating vulnerable plaque
US20030008841A1 (en) 2000-08-30 2003-01-09 Rene Devos Anti-HCV nucleoside derivatives
AU2001282528A1 (en) 2000-09-01 2002-03-22 Shionogi And Co., Ltd. Compounds having anti-hepatitis c virus effect
KR20090089922A (en) 2000-10-18 2009-08-24 파마셋 인코포레이티드 Modified nucleosides for treatment of viral infections and abnormal cellular proliferation
AU2002236591B2 (en) 2000-12-12 2005-07-14 Schering Corporation Diaryl peptides as NS3-serine protease inhibitors of hepatits C virus
WO2002048116A2 (en) 2000-12-13 2002-06-20 Bristol-Myers Squibb Pharma Company Inhibitors of hepatitis c virus ns3 protease
BR0116221A (en) 2000-12-15 2005-09-13 Pharmasset Ltd Antiviral agents for treatment of flaviviridae infections
EP1346724A4 (en) 2000-12-26 2004-11-17 Mitsubishi Pharma Corp Remedies for hepatitis c
JP3914156B2 (en) 2001-01-22 2007-05-16 メルク エンド カムパニー インコーポレーテッド Nucleoside derivatives as RNA-dependent RNA viral polymerase inhibitors
GB0114286D0 (en) 2001-06-12 2001-08-01 Hoffmann La Roche Nucleoside Derivatives
JP4537057B2 (en) 2001-08-14 2010-09-01 テルアビブ・ユニバーシティ・フューチャー・テクノロジー・デベロップメント・エルピー Lipidated glycosaminoglycan particles and their use in drug and gene delivery for diagnosis and treatment
WO2003026675A1 (en) 2001-09-28 2003-04-03 Idenix (Cayman) Limited Methods and compositions for treating flaviviruses and pestiviruses using 4'-modified nucleoside
AU2002330154A1 (en) 2001-09-28 2003-04-07 Centre National De La Recherche Scientifique Methods and compositions for treating hepatitis c virus using 4'-modified nucleosides
EP1465862A1 (en) 2002-01-17 2004-10-13 SmithKline Beecham Corporation Cycloalkyl ketoamides derivatives useful as cathepsin k inhibitors
GB0201179D0 (en) 2002-01-18 2002-03-06 Angeletti P Ist Richerche Bio Therapeutic agents
CA2369711A1 (en) 2002-01-30 2003-07-30 Boehringer Ingelheim (Canada) Ltd. Macrocyclic peptides active against the hepatitis c virus
EP1476169B1 (en) 2002-02-13 2013-03-20 Merck Sharp & Dohme Corp. Inhibiting orthopoxvirus replication with nucleoside compounds
US6777392B2 (en) 2002-03-28 2004-08-17 Council Of Scientific And Industrial Research 8-(C-β-D-glucopyranosyl)-7, 3', 4'-trihydroxyflavone, process of isolation thereof, pharmaceutical composition and method for the treatment of diabetes
US20040063658A1 (en) 2002-05-06 2004-04-01 Roberts Christopher Don Nucleoside derivatives for treating hepatitis C virus infection
MY140680A (en) 2002-05-20 2010-01-15 Bristol Myers Squibb Co Hepatitis c virus inhibitors
WO2004000858A2 (en) 2002-06-21 2003-12-31 Merck & Co., Inc. Nucleoside derivatives as inhibitors of rna-dependent rna viral polymerase
AU2003269892A1 (en) 2002-06-27 2004-01-19 Isis Pharmaceuticals, Inc. Nucleoside derivatives as inhibitors of rna-dependent rna viral polymerase
JP5087211B2 (en) 2002-06-28 2012-12-05 イデニクス(ケイマン)リミテツド 2 'and 3'-nucleoside prodrugs for the treatment of flavivirus infection
RS113904A (en) 2002-06-28 2007-02-05 Idenix (Cayman) Limited 2'-c-methyl-3'-o-l-valine ester ribofuranosyl cytidine for treatment of flaviviridae infections
PL374831A1 (en) 2002-06-28 2005-10-31 Idenix (Cayman) Limited Modified 2' and 3' -nucleoside prodrugs for treating flaviridae infections
AU2003269902A1 (en) 2002-07-16 2004-02-02 Isis Pharmaceuticals, Inc. Nucleoside derivatives as inhibitors of rna-dependent rna viral polymerase
CA2492607A1 (en) 2002-07-25 2004-02-05 Micrologix Biotech Inc. Anti-viral 7-deaza d-nucleosides and uses thereof
EP1545545A4 (en) 2002-08-01 2008-09-03 Pharmasset Inc COMPOUNDS WITH THE BICYCLO 4.2.1 NONANE SYSTEM FOR THE TREATMENT OF i FLAVIVIRIDAE /i INFECTIONS
KR20050059199A (en) 2002-09-30 2005-06-17 제네랩스 테크놀로지스, 인코포레이티드 Nucleoside derivatives for treating hepatitis c virus infection
HN2003000348A (en) 2002-11-01 2008-10-14 Viropharma Inc BENZOFURAN COMPOUNDS, COMPOSITIONS AND METHODS FOR TREATMENT AND VIRAL INFECTION OF HEPATITIS C AND ASSOCIATED ILLNESSES.
US20040254159A1 (en) 2003-02-27 2004-12-16 Hasvold Lisa A. Heterocyclic kinase inhibitors
EP1601685A1 (en) 2003-03-05 2005-12-07 Boehringer Ingelheim International GmbH Hepatitis c inhibiting compounds
CA2520886A1 (en) 2003-04-02 2004-11-04 Boehringer Ingelheim International Gmbh Pharmaceutical compositions for hepatitis c viral protease inhibitors
GB0307891D0 (en) 2003-04-04 2003-05-14 Angeletti P Ist Richerche Bio Chemical compounds,compositions and uses
ATE422895T1 (en) 2003-04-16 2009-03-15 Bristol Myers Squibb Co MACROCYCLIC ISOQUINOLINE PEPTIDE INHIBITORS OF HEPATITIS C VIRUS
US7176208B2 (en) 2003-04-18 2007-02-13 Enanta Pharmaceuticals, Inc. Quinoxalinyl macrocyclic hepatitis C serine protease inhibitors
AU2004240704B9 (en) 2003-05-21 2009-10-22 Boehringer Ingelheim International Gmbh Hepatitis C inhibitor compounds
KR100883703B1 (en) 2003-05-30 2009-02-12 파마셋 인코포레이티드 Modified fluorinated nucleoside analogues
GB0313250D0 (en) 2003-06-09 2003-07-16 Angeletti P Ist Richerche Bio Therapeutic agents
EP1644395B1 (en) 2003-06-19 2006-11-22 F. Hoffmann-La Roche Ag Processes for preparing 4'azido nucleoside derivatives
TW200510425A (en) 2003-08-13 2005-03-16 Japan Tobacco Inc Nitrogen-containing fused ring compound and use thereof as HIV integrase inhibitor
GB0321003D0 (en) 2003-09-09 2003-10-08 Angeletti P Ist Richerche Bio Compounds, compositions and uses
GB0323845D0 (en) 2003-10-10 2003-11-12 Angeletti P Ist Richerche Bio Chemical compounds,compositions and uses
US7132504B2 (en) 2003-11-12 2006-11-07 Bristol-Myers Squibb Company Hepatitis C virus inhibitors
DE602005025855D1 (en) 2004-01-21 2011-02-24 Boehringer Ingelheim Pharma MACROCYCLIC PEPTIDES WITH EFFECT TO THE HEPATITIS C VIRUS
WO2005080399A1 (en) 2004-02-24 2005-09-01 Japan Tobacco Inc. Fused heterotetracyclic compounds and use tehreof as hcv polymerase inhibitor
GB0413087D0 (en) 2004-06-11 2004-07-14 Angeletti P Ist Richerche Bio Therapeutic compounds
GB0416396D0 (en) 2004-07-22 2004-08-25 Angeletti P Ist Richerche Bio Therapeutic agents
US7153848B2 (en) 2004-08-09 2006-12-26 Bristol-Myers Squibb Company Inhibitors of HCV replication
CA2577526A1 (en) 2004-08-23 2006-03-02 Joseph Armstrong Martin Antiviral 4'-azido-nucleosides
GB0419850D0 (en) 2004-09-07 2004-10-13 Angeletti P Ist Richerche Bio Therapeutic agents
JP2008517986A (en) 2004-10-26 2008-05-29 イステイチユート・デイ・リチエルケ・デイ・ビオロジア・モレコラーレ・ピ・アンジエレツテイ・エツセ・ピー・アー Tetracyclic indole derivatives as antiviral agents
WO2006102087A2 (en) 2005-03-22 2006-09-28 Merck & Co., Inc. Hcv protease substrates
GB0509326D0 (en) 2005-05-09 2005-06-15 Angeletti P Ist Richerche Bio Therapeutic compounds
AR057456A1 (en) * 2005-07-20 2007-12-05 Merck & Co Inc HCV PROTEASA NS3 INHIBITORS
US7462035B2 (en) 2005-07-27 2008-12-09 Physical Optics Corporation Electrical connector configured as a fastening element
WO2007028789A1 (en) 2005-09-07 2007-03-15 Istituto Di Ricerche Di Biologia Molecolare P. Angeletti Spa Quinazoline derivatives as antiviral agents
GB0518390D0 (en) 2005-09-09 2005-10-19 Angeletti P Ist Richerche Bio Therapeutic compounds
GB0519797D0 (en) 2005-09-29 2005-11-09 Istituto Di Ricerche D Biolog Therapeutic agents
GB0609492D0 (en) 2006-05-15 2006-06-21 Angeletti P Ist Richerche Bio Therapeutic agents
EP2029741A4 (en) 2006-06-08 2011-03-16 Merck Sharp & Dohme A rapid method to determine inhibitor sensitivity of ns3/4a protease sequences cloned from clinical samples
GB0612423D0 (en) 2006-06-23 2006-08-02 Angeletti P Ist Richerche Bio Therapeutic agents
WO2008051475A2 (en) 2006-10-24 2008-05-02 Merck & Co., Inc. Hcv ns3 protease inhibitors
JP5345941B2 (en) 2006-10-24 2013-11-20 メルク・シャープ・アンド・ドーム・コーポレーション HCV NS3 protease inhibitor
EP2076278B1 (en) * 2006-10-24 2015-05-06 Merck Sharp & Dohme Corp. Macrocyclic HCV NS3 protease inhibitors
US8377874B2 (en) * 2006-10-27 2013-02-19 Merck Sharp & Dohme Corp. HCV NS3 protease inhibitors
CN101557910B (en) 2006-11-09 2011-08-17 双刃技术控股瑞典股份公司 Hub device
EP2481807A3 (en) 2007-03-09 2013-04-03 Merck Sharp & Dohme Corp. In vivo HCV resistance to anti-viral inhibitors
ES2541434T3 (en) 2007-06-29 2015-07-20 Gilead Sciences, Inc. Purine derivatives and their use as modulators of the Toll 7 receptor
CA2699891C (en) 2007-07-19 2013-10-22 Nigel Liverton Macrocyclic compounds as antiviral agents
CL2008003384A1 (en) 2007-11-14 2009-12-11 Enanta Pharm Inc Macrocyclic quinoxaline derived compounds, serine protease inhibitors; pharmaceutical composition comprising them; and its use in the treatment of hepatitis c.
MX2010005261A (en) 2007-11-14 2010-10-15 Enanta Pharm Inc Macrocyclic tetrazolyl hepatitis c serine protease inhibitors.
US8461107B2 (en) * 2008-04-28 2013-06-11 Merck Sharp & Dohme Corp. HCV NS3 protease inhibitors
DK2540350T3 (en) 2008-07-22 2014-08-04 Merck Sharp & Dohme COMBINATIONS OF A MACROCYCLIC QUINOXAL COMPOUND, WHICH IS ANHCV-NS3 PROTEASE INHIBITOR WITH OTHER HCV AGENTS

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2006119061A2 (en) * 2005-05-02 2006-11-09 Merck & Co., Inc. Hcv ns3 protease inhibitors
WO2007016441A1 (en) * 2005-08-01 2007-02-08 Merck & Co., Inc. Macrocyclic peptides as hcv ns3 protease inhibitors
WO2008002924A2 (en) * 2006-06-26 2008-01-03 Enanta Pharmaceuticals, Inc. Quinoxalinyl macrocyclic hepatitis c virus serine protease inhibitors
WO2008057209A1 (en) * 2006-10-27 2008-05-15 Merck & Co., Inc. Hcv ns3 protease inhibitors

Cited By (60)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8080654B2 (en) 2008-07-22 2011-12-20 Insituto di Ricerche di Biologia Molecolare P. Angeletti SpA Macrocyclic quinoxaline compounds as HCV NS3 protease inhibitors
US7973040B2 (en) 2008-07-22 2011-07-05 Merck Sharp & Dohme Corp. Macrocyclic quinoxaline compounds as HCV NS3 protease inhibitors
US8828930B2 (en) 2009-07-30 2014-09-09 Merck Sharp & Dohme Corp. Hepatitis C virus NS3 protease inhibitors
KR102128232B1 (en) 2010-09-21 2020-06-30 이난타 파마슈티칼스, 인코포레이티드 Macrocyclic proline derived hcv serine protease inhibitors
KR20190069623A (en) * 2010-09-21 2019-06-19 이난타 파마슈티칼스, 인코포레이티드 Macrocyclic proline derived hcv serine protease inhibitors
JP2016104756A (en) * 2010-09-21 2016-06-09 エナンタ ファーマシューティカルズ インコーポレイテッド Macrocyclic proline derived hcv serine protease inhibitors
KR20180099929A (en) * 2010-09-21 2018-09-05 이난타 파마슈티칼스, 인코포레이티드 Macrocyclic proline derived hcv serine protease inhibitors
KR20130098369A (en) * 2010-09-21 2013-09-04 이난타 파마슈티칼스, 인코포레이티드 Macrocyclic proline derived hcv serine protease inhibitors
JP2013538235A (en) * 2010-09-21 2013-10-10 エナンタ ファーマシューティカルズ インコーポレイテッド Macrocyclic proline-derived HCV serine protease inhibitor
JP2020015730A (en) * 2010-09-21 2020-01-30 エナンタ ファーマシューティカルズ インコーポレイテッド Macrocyclic proline derived hcv serine protease inhibitors
KR101894704B1 (en) * 2010-09-21 2018-09-05 이난타 파마슈티칼스, 인코포레이티드 Macrocyclic proline derived hcv serine protease inhibitors
KR20190110648A (en) * 2010-09-21 2019-09-30 이난타 파마슈티칼스, 인코포레이티드 Macrocyclic proline derived hcv serine protease inhibitors
KR102163902B1 (en) 2010-09-21 2020-10-12 이난타 파마슈티칼스, 인코포레이티드 Macrocyclic proline derived hcv serine protease inhibitors
JP2021063130A (en) * 2010-09-21 2021-04-22 エナンタ ファーマシューティカルズ インコーポレイテッド Macrocyclic proline derived hcv serine protease inhibitors
KR101990279B1 (en) 2010-09-21 2019-06-19 이난타 파마슈티칼스, 인코포레이티드 Macrocyclic proline derived hcv serine protease inhibitors
JP2018162265A (en) * 2010-09-21 2018-10-18 エナンタ ファーマシューティカルズ インコーポレイテッド Macrocyclic proline derived hcv serine protease inhibitors
WO2012050850A1 (en) * 2010-09-29 2012-04-19 Merck Sharp & Dohme Corp. Polycyclic heterocycle derivatives and methods of use thereof for the treatment of viral diseases
EP2621501A4 (en) * 2010-09-29 2014-04-09 Merck Sharp & Dohme Polycyclic heterocycle derivatives and methods of use thereof for the treatment of viral diseases
EP2621501A1 (en) * 2010-09-29 2013-08-07 Merck Sharp & Dohme Corp. Polycyclic heterocycle derivatives and methods of use thereof for the treatment of viral diseases
US9120818B2 (en) 2010-12-14 2015-09-01 Merck Sharp & Dohme Corp. Process and intermediates for preparing macrolactams
US8957203B2 (en) 2011-05-05 2015-02-17 Bristol-Myers Squibb Company Hepatitis C virus inhibitors
US9527885B2 (en) 2011-05-05 2016-12-27 Bristol-Myers Squibb Company Hepatitis C virus inhibitors
EP2744507A4 (en) * 2011-08-19 2015-01-28 Merck Sharp & Dohme Crystal forms of a hcv protease inhibitor
US9242917B2 (en) 2011-08-19 2016-01-26 Merck Sharp & Dohme Limited Crystal forms of a HCV protease inhibitor
JP2014521750A (en) * 2011-08-19 2014-08-28 メルク・シャープ・アンド・ドーム・コーポレーション Methods and intermediates for preparing macrolactams
EP2744507A1 (en) * 2011-08-19 2014-06-25 Merck Sharp & Dohme Corp. Crystal forms of a hcv protease inhibitor
WO2013028470A1 (en) 2011-08-19 2013-02-28 Merck Sharp & Dohme Corp. Process and intermediates for preparing macrolactams
WO2013028471A1 (en) * 2011-08-19 2013-02-28 Merck Sharp & Dohme Corp. Methods and intermediates for preparing macrolactams
US9238604B2 (en) 2011-08-19 2016-01-19 Merck Sharp & Dohme Corp. Process and intermediates for preparing macrolactams
US9073825B2 (en) 2011-08-19 2015-07-07 Merck Sharp & Dohme Limited Methods and intermediates for preparing macrolactams
EP2773342A4 (en) * 2011-10-31 2015-08-26 Merck Sharp & Dohme Compositions useful for the treatment of viral diseases
WO2013074386A2 (en) 2011-11-15 2013-05-23 Merck Sharp & Dohme Corp. Hcv ns3 protease inhibitors
US9328138B2 (en) 2011-11-15 2016-05-03 Msd Italia S.R.L. HCV NS3 protease inhibitors
EP2780026A4 (en) * 2011-11-15 2016-06-08 Merck Sharp & Dohme Hcv ns3 protease inhibitors
EP3825312A1 (en) 2012-07-03 2021-05-26 Gilead Pharmasset LLC Intermediates for preparing inhibitors of hepatitis c virus
US10603318B2 (en) 2012-07-03 2020-03-31 Gilead Pharmasset Llc Inhibitors of hepatitis C virus
WO2014008285A1 (en) 2012-07-03 2014-01-09 Gilead Sciences, Inc. Inhibitors of hepatitis c virus
EP3159345A1 (en) 2012-07-03 2017-04-26 Gilead Sciences, Inc. Inhibitors of hepatitis c virus
EP3492464A1 (en) 2012-07-03 2019-06-05 Gilead Sciences, Inc. Intermediates for preparing inhibitors of hepatitis c virus
US10335409B2 (en) 2012-07-03 2019-07-02 Gilead Pharmasset Llc Inhibitors of hepatitis C virus
US9296782B2 (en) 2012-07-03 2016-03-29 Gilead Sciences, Inc. Inhibitors of hepatitis C virus
US9499550B2 (en) 2012-10-19 2016-11-22 Bristol-Myers Squibb Company Hepatitis C virus inhibitors
US9598433B2 (en) 2012-11-02 2017-03-21 Bristol-Myers Squibb Company Hepatitis C virus inhibitors
US9334279B2 (en) 2012-11-02 2016-05-10 Bristol-Myers Squibb Company Hepatitis C virus inhibitors
US9643999B2 (en) 2012-11-02 2017-05-09 Bristol-Myers Squibb Company Hepatitis C virus inhibitors
US9409943B2 (en) 2012-11-05 2016-08-09 Bristol-Myers Squibb Company Hepatitis C virus inhibitors
US9580463B2 (en) 2013-03-07 2017-02-28 Bristol-Myers Squibb Company Hepatitis C virus inhibitors
AU2014233390B2 (en) * 2013-03-15 2018-03-01 Gilead Sciences, Inc. Macrocyclic and bicyclic inhibitors of hepatitis C virus
EA029088B1 (en) * 2013-03-15 2018-02-28 Джилид Сайэнс, Инк. Macrocyclic and bicyclic inhibitors of hepatitis c virus
WO2014145095A1 (en) * 2013-03-15 2014-09-18 Gilead Sciences, Inc. Macrocyclic and bicyclic inhibitors of hepatitis c virus
US9617310B2 (en) 2013-03-15 2017-04-11 Gilead Sciences, Inc. Inhibitors of hepatitis C virus
US9873707B2 (en) 2013-10-18 2018-01-23 Merck Sharp & Dohme Corp. Methods and intermediates for preparing macrolactams
EP3057926A4 (en) * 2013-10-18 2017-07-12 Merck Sharp & Dohme Corp. Methods and intermediates for preparing macrolactams
WO2015095430A1 (en) * 2013-12-20 2015-06-25 Merck Sharp & Dohme Corp. Methods and intermediates for the preparation of macrolactams
WO2015100145A1 (en) * 2013-12-23 2015-07-02 Gilead Sciences, Inc. Synthesis of a macrocyclic hcv ns3 inhibiting tripeptide
US9440991B2 (en) 2013-12-23 2016-09-13 Gilead Sciences, Inc. Synthesis of an antiviral compound
WO2015100144A1 (en) * 2013-12-23 2015-07-02 Gilead Sciences, Inc. Crystalline forms of a macrocyclic hcv ns3 inhibiting tripeptide
US10030033B2 (en) 2013-12-23 2018-07-24 Gilead Sciences, Inc. Synthesis of an antiviral compound
WO2017198102A1 (en) * 2016-05-16 2017-11-23 深圳市塔吉瑞生物医药有限公司 Substituted macrocyclic quinoxaline compound and pharmaceutical composition and use thereof
WO2021209563A1 (en) 2020-04-16 2021-10-21 Som Innovation Biotech, S.A. Compounds for use in the treatment of viral infections by respiratory syndrome-related coronavirus

Also Published As

Publication number Publication date
NL300857I2 (en) 2017-04-25
HK1173403A1 (en) 2013-05-16
AU2009274190A1 (en) 2010-01-28
CY2017005I1 (en) 2017-06-28
EP2310095B1 (en) 2012-08-29
PT2540350E (en) 2014-08-27
EP2540350A1 (en) 2013-01-02
HUS1700001I1 (en) 2017-02-28
EA201170241A1 (en) 2011-10-31
US7973040B2 (en) 2011-07-05
LTPA2016049I1 (en) 2017-01-25
ES2491090T3 (en) 2014-09-05
EP2310095A1 (en) 2011-04-20
FR16C1027I1 (en) 2017-02-03
HRP20120866T1 (en) 2013-01-31
EP2540350B1 (en) 2014-05-21
CN102159285B (en) 2014-05-14
SV2011003813A (en) 2011-05-25
US20100029666A1 (en) 2010-02-04
AR072588A1 (en) 2010-09-08
AU2009274190B2 (en) 2012-07-05
DOP2011000023A (en) 2011-03-31
ECSP11010777A (en) 2011-02-28
PL2540350T3 (en) 2014-10-31
EP2540349B1 (en) 2014-02-12
JP4920797B2 (en) 2012-04-18
CO6351757A2 (en) 2011-12-20
KR20110036627A (en) 2011-04-07
BRPI0916235B1 (en) 2020-09-29
CL2011000145A1 (en) 2011-05-06
NI201100023A (en) 2011-09-20
HRP20140693T1 (en) 2014-11-21
RS52534B (en) 2013-04-30
US8080654B2 (en) 2011-12-20
CR20110089A (en) 2011-04-27
PE20110212A1 (en) 2011-04-10
US20110224134A1 (en) 2011-09-15
ME02132B (en) 2014-08-22
ME02024B (en) 2015-05-20
TW201023860A (en) 2010-07-01
LUC00002I1 (en) 2017-01-19
CY1113752T1 (en) 2016-07-27
JP2011528713A (en) 2011-11-24
EP2540349A1 (en) 2013-01-02
CA2731177A1 (en) 2010-01-28
HN2011000209A (en) 2013-04-22
PL2310095T3 (en) 2013-03-29
SI2540350T1 (en) 2015-01-30
NO2017004I2 (en) 2017-01-20
CN102159285A (en) 2011-08-17
HK1173402A1 (en) 2013-05-16
BRPI0916235B8 (en) 2021-05-25
EA019327B1 (en) 2014-02-28
ES2392611T3 (en) 2012-12-12
CY1115503T1 (en) 2017-01-04
NO2017004I1 (en) 2017-01-20
SI2310095T1 (en) 2013-01-31
IL210580A0 (en) 2011-03-31
BRPI0916235A8 (en) 2018-06-12
MA32502B1 (en) 2011-07-03
MY152070A (en) 2014-08-15
DK2310095T3 (en) 2012-12-10
IL210580A (en) 2014-08-31
MX2011000826A (en) 2011-02-23
KR101313675B1 (en) 2013-10-02
UA100436C2 (en) 2012-12-25
TWI441638B (en) 2014-06-21
CY2017005I2 (en) 2017-06-28
NZ590638A (en) 2012-06-29
FR16C1027I2 (en) 2017-12-08
CA2731177C (en) 2013-10-29
DK2540350T3 (en) 2014-08-04
BRPI0916235A2 (en) 2018-03-13
PT2310095E (en) 2012-11-16
RS53420B (en) 2014-12-31
TN2011000014A1 (en) 2012-09-05
LTC2310095I2 (en) 2018-11-12
LUC00002I2 (en) 2017-04-04

Similar Documents

Publication Publication Date Title
EP2540350B1 (en) Combinations of a macrocyclic quinoxaline compound which is an hcv ns3 protease inhibitor with other hcv agents
EP2271345B1 (en) Hcv ns3 protease inhibitors
EP1924593B9 (en) Hcv ns3 protease inhibitors
AU2007318164B2 (en) HCV NS3 protease inhibitors
EP1879607B1 (en) Hcv ns3 protease inhibitors
EP2076278B1 (en) Macrocyclic HCV NS3 protease inhibitors
EP2086982B1 (en) Hcv ns3 protease inhibitors

Legal Events

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

Ref document number: 200980137118.6

Country of ref document: CN

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

Ref document number: 09790553

Country of ref document: EP

Kind code of ref document: A1

WWE Wipo information: entry into national phase

Ref document number: 2009790553

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 328/DELNP/2011

Country of ref document: IN

WWE Wipo information: entry into national phase

Ref document number: 2731177

Country of ref document: CA

ENP Entry into the national phase

Ref document number: 2011520110

Country of ref document: JP

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: 2011010126

Country of ref document: EG

Ref document number: 11005448

Country of ref document: CO

WWE Wipo information: entry into national phase

Ref document number: 590638

Country of ref document: NZ

Ref document number: 2009274190

Country of ref document: AU

Ref document number: DZP2011000055

Country of ref document: DZ

WWE Wipo information: entry into national phase

Ref document number: 000067-2011

Country of ref document: PE

Ref document number: 12011500151

Country of ref document: PH

Ref document number: MX/A/2011/000826

Country of ref document: MX

NENP Non-entry into the national phase

Ref country code: DE

WWE Wipo information: entry into national phase

Ref document number: CR2011-000089

Country of ref document: CR

ENP Entry into the national phase

Ref document number: 2009274190

Country of ref document: AU

Date of ref document: 20090717

Kind code of ref document: A

ENP Entry into the national phase

Ref document number: 20117003982

Country of ref document: KR

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: 201170241

Country of ref document: EA

WWE Wipo information: entry into national phase

Ref document number: A201102068

Country of ref document: UA

WWE Wipo information: entry into national phase

Ref document number: P-2012/0463

Country of ref document: RS

ENP Entry into the national phase

Ref document number: PI0916235

Country of ref document: BR

Kind code of ref document: A2

Effective date: 20110119