WO2007143521A1 - Cyclopropyl fused indolobenzazepine hcv inhibitors - Google Patents

Cyclopropyl fused indolobenzazepine hcv inhibitors Download PDF

Info

Publication number
WO2007143521A1
WO2007143521A1 PCT/US2007/070173 US2007070173W WO2007143521A1 WO 2007143521 A1 WO2007143521 A1 WO 2007143521A1 US 2007070173 W US2007070173 W US 2007070173W WO 2007143521 A1 WO2007143521 A1 WO 2007143521A1
Authority
WO
WIPO (PCT)
Prior art keywords
hcv
alkyl
compound
inhibitors
hydrogen
Prior art date
Application number
PCT/US2007/070173
Other languages
French (fr)
Inventor
Kap-Sun Yeung
Katharine A. Grant-Young
Min Ding
Robert G. Gentles
John F. Kadow
Original Assignee
Bristol-Myers Squibb Company
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Bristol-Myers Squibb Company filed Critical Bristol-Myers Squibb Company
Publication of WO2007143521A1 publication Critical patent/WO2007143521A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D487/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00
    • C07D487/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00 in which the condensed system contains two hetero rings
    • C07D487/04Ortho-condensed systems
    • 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

Definitions

  • HCV Hepatitis C virus
  • HCV is a positive-stranded R ⁇ A virus. Based on a comparison of the deduced amino acid sequence and the extensive similarity in the 5 '-untranslated region, HCV has been classified as a separate genus in the Flaviviridae family. All members of the Flaviviridae family have enveloped virions that contain a positive stranded R ⁇ A genome encoding all known virus-specific proteins via translation of a single, uninterrupted, open reading frame.
  • the single strand HCV R ⁇ A genome is approximately 9500 nucleotides in length and has a single open reading frame (ORF) encoding a single large polyprotein of about 3000 amino acids. In infected cells, this polyprotein is cleaved at multiple sites by cellular and viral proteases to produce the structural and non-structural (NS) proteins. In the case of HCV, the generation of mature non-structural proteins (NS2, NS3, NS4A, NS4B, NS5A, and NS5B) is effected by two viral proteases.
  • the first one is believed to be a metalloprotease and cleaves at the NS2-NS3 junction; the second one is a serine protease contained within the N-terminal region of NS3 (also referred to as NS3 protease) and mediates all the subsequent cleavages downstream of NS3, both in cis, at the NS3-NS4A cleavage site, and in trans, for the remaining NS4A-NS4B, NS4B-NS5A, NS5A-NS5B sites.
  • the NS4A protein appears to serve multiple functions, acting as a cofactor for the NS3 protease and possibly assisting in the membrane localization of NS3 and other viral replicase components.
  • NS5B (also referred to as HCV polymerase) is a RNA-dependent RNA polymerase that is involved in the replication of HCV.
  • the HCV NS5B protein is described in "Structural Analysis of the Hepatitis C Virus RNA Polymerase in Complex with Ribonucleotides (Bressanelli; S. et al, Journal of Virology 2002, 3482-3492; and Defrancesco and Rice, Clinics in Liver Disease 2003, 7, 211-242.
  • One aspect of the invention is a compound of formula I
  • R 1 is CO 2 R 5 or CONR 6 R 7 ;
  • R 2 is N(R 1O )(R U );
  • R 3 is hydrogen, halo, alkyl, alkenyl, hydroxy, benzyloxy, alkoxy, or haloalkoxy;
  • R 4 is cycloalkyl
  • R 5 is hydrogen or alkyl
  • R 6 is hydrogen, alkyl, alkylSO 2 , cycloalkylSO 2 , haloalkylSO 2 , (R 7 )(R 8 )NSO 2 , or
  • R 7 is hydrogen or alkyl
  • R 8 is hydrogen or alkyl
  • R 9 is azetidinyl, pyrrolidinyl, piperidinyl, piperazinyl, morpholinyl, thiomorpholinyl, homopiperidinyl, or homomorpholinyl, and is substituted with 0-3 substituents selected from alkyl and alkoxy;
  • R 10 is hydrogen, alkyl, alkoxyCO, or (R 12 )(R 13 )NCO;
  • R » 11 is hydrogen or alkyl
  • R 12 is hydrogen, alkyl, alkoxyalkyl, aminoalkyl, (alkylamino)alkyl,
  • R 13 is hydrogen alkyl, alkoxyalkyl, aminoalkyl, (alkylamino)alkyl, or (dialkylamino)alkyl;
  • R 12 )(R 13 )N taken together is azetidinyl, pyrrolidinyl, piperidinyl, piperazinyl, morpholinyl, thiomorpholinyl, homopiperidinyl, or homomorpholinyl and is substituted with 0-3 substituents selected from alkyl and alkoxy;
  • R , 14 is hydrogen or alkyl
  • R » 15 is hydrogen or alkyl
  • R , 16 is hydrogen or alkyl
  • N(R 15 )(R 16 ) taken together is azetidinyl, pyrrolidinyl, piperidinyl, piperazinyl, morpholinyl, thiomorpholinyl, homopiperidinyl, or homomorpholinyl and is substituted with 0-3 substituents selected from alkyl and alkoxy;
  • R 1 is CONHR 6 and R 6 is alkylSO 2 , cycloalkylSO 2 , haloalkylSO 2 , (R 7 )(R 8 )NSO 2 , or (R 9 )SO 2 .
  • Another aspect of the invention is a compound of formula I where R is hydrogen.
  • Another aspect of the invention is a compound of formula I where R 3 is methoxy.
  • Another aspect of the invention is a compound of formula I where R 4 is cyclohexyl.
  • Another aspect of the invention is a compound of formula I where R 6 is (R 9 ⁇ NSO 2 or (R 10 )SO 2 .
  • Another aspect of the invention is a compound of formula I where R 10 is (R 12 )(R 13 )NCO.
  • R 12 is alkoxyalkyl, (dialkylamino)alkyl, (CON(R 15 )(R 16 ))alkyl, or (tetrahydropyranyl)alkyl.
  • Another aspect of the invention is a compound of formula I where (R 12 )(R 13 )N taken together is azetidinyl, pyrrolidinyl, piperidinyl, piperazinyl, morpholinyl, thiomorpholinyl, homopiperidinyl, or homomorpholinyl and is substituted with 0-3 substituents selected from alkyl and alkoxy.
  • Another aspect of the invention is a compound of formula I with the following stereochemistry.
  • Another aspect of the invention is a compound of formula I with the following stereochemistry.
  • any scope of any variable including R 1 , R 2 , R 3 , R 4 , R 5 , R 6 , R 7 , R 8 , R 9 , R 10 , R 11 , R 12 , R 13 , R 14 , R 15 , and R 16 , can be used independently with the scope of any other instance of a variable. Unless specified otherwise, these terms have the following meanings.
  • Alkyl means a straight or branched alkyl group composed of 1 to 6 carbons.
  • Alkenyl means a straight or branched alkyl group composed of 2 to 6 carbons with at least one double bond.
  • Cycloalkyl means a monocyclic ring system composed of 3 to 7 carbons.
  • Haldroxy alkyl means an alkyl substituent further substituted with the substituent R.
  • the invention includes all pharmaceutically acceptable salt forms of the compounds.
  • Pharmaceutically acceptable salts are those in which the counter ions do not contribute significantly to the physiological activity or toxicity of the compounds and as such function as pharmacological equivalents. These salts can be made according to common organic techniques employing commercially available reagents. Some anionic salt forms include acetate, acistrate, besylate, bromide, chloride, citrate, fumarate, glucouronate, hydrobromide, hydrochloride, hydroiodide, iodide, lactate, maleate, mesylate, nitrate, pamoate, phosphate, succinate, sulfate, tartrate, tosylate, and xinofoate.
  • Some cationic salt forms include ammonium, aluminum, benzathine, bismuth, calcium, choline, diethylamine, diethanolamine, lithium, magnesium, meglumine, 4-phenylcyclohexylamine, piperazine, potassium, sodium, tromethamine, and zinc.
  • the invention includes all stereoisomeric forms, including enantiomers and diastereomers as well as mixtures of stereoisomers such as racemates. Some stereoisomers can be made using methods known in the art.
  • Stereoisomeric mixtures of the compounds and related intermediates can be separated into individual isomers according to methods known in the art.
  • the compounds may be made by methods known in the art including those described below. Some reagents and intermediates are known in the art. Other reagents and intermediates can be made by methods known in the art using readily available materials.
  • the variables (e.g. numbered "R" substituents) used to describe the synthesis of the compounds are intended only to illustrate how to make the compounds and are not to be confused with variables used in the claims or in other sections of the specification.
  • the compounds can be made by methods known in the art and described in the application.
  • 2-bromo-3-cyclohexyl-lH-indole-6-carboxylic acid can be condensed with a variety of sulfonyl ureas, using for example l,l '-carbonyldiimidazole in combination with l,8-diazabicyclo[5.4.0]undec-7-ene in anhydrous THF.
  • the resultant acyl sulfamides can be subjected to known coupling reactions, for example Suzuki coupling conditions with a diversity of 2-formyl boronic acids or esters to provide cyclic hemiaminal intermediates of the type depicted.
  • the resultant acids can be converted to ureas by methods known in the art, for example treatment with diphenylphosphoryl azide and triethylamine in toluene, and the intermediate acyl azide can be isolated, and then treated in a separate step with a diversity of amines to afford the urea examples as shown in Scheme 2.
  • the intermediate acid ester shown in Scheme 3 can be subjected to a Curtius reaction using a diversity of amines, and the resultant esters can be hydro lyzed to provide examples of carboxamide acids.
  • a coupling with a sulfamide or sulfonamide as shown in the last step of Scheme 3 can provide an alternate preparation of compounds of the invention.
  • HCVNS5B RdRp cloning, expression, and purification The cDNA encoding the NS5B protein of HCV, genotype Ib, was cloned into the pET21a expression vector. The protein was expressed with an 18 amino acid C-terminal truncation to enhance the solubility.
  • the E. coli competent cell line BL21(DE3) was used for expression of the protein. Cultures were grown at 37 0 C for ⁇ 4 hours until the cultures reached an optical density of 2.0 at 600 nm. The cultures were cooled to 20 0 C and induced with 1 mM IPTG. Fresh ampicillin was added to a final concentration of 50 ⁇ g/ml and the cells were grown overnight at 20 0 C.
  • Cell pellets (3L) were lysed for purification to yield 15-24 mgs of purified NS5B.
  • the lysis buffer consisted of 20 mM Tris-HCl, pH 7.4, 500 mM NaCl, 0.5% triton X-100, 1 mM DTT, ImM EDTA, 20% glycerol, 0.5 mg/ml lysozyme, 10 mM MgC12, 15 ug/ml deoxyribonuclease I, and Complete TM protease inhibitor tablets (Roche). After addition of the lysis buffer, frozen cell pellets were resuspended using a tissue homogenizer.
  • aliquots of the lysate were sonicated on ice using a microtip attached to a Branson sonicator.
  • the sonicated lysate was centrifuged at 100,000 x g for lhr at 4 0 C and filtered through a 0.2 ⁇ m filter unit (Corning).
  • the protein was purified using three sequential chromatography steps: Heparin sepharose CL-6B, polyU sepharose 4B, and Hitrap SP sepharose (Pharmacia).
  • the chromatography buffers were identical to the lysis buffer but contained no lysozyme, deoxyribonuclease I, MgC12 or protease inhibitor and the NaCl concentration of the buffer was adjusted according to the requirements for charging the protein onto the column.
  • Each column was eluted with a NaCl gradient which varied in length from 5-50 column volumes depending on the column type.
  • the resulting purity of the enzyme is >90% based on SDS-PAGE analysis.
  • the enzyme was aliquoted and stored at -80 0 C.
  • HCV RdRp genotype Ib assays were run in a final volume of 60 ⁇ l in 96 well plates (Costar 3912).
  • the assay buffer is composed of 20 mM Hepes, pH 7.5, 2.5 mM KCl, 2.5 mM MgC12, 1 mM DTT, 1.6 U RNAse inhibitor (Promega N2515), 0.1 mg/ml BSA (Promega R3961), and 2 % glycerol. All compounds were serially diluted (3 -fold) in DMSO and diluted further in water such that the final concentration of DMSO in the assay was 2%.
  • HCV RdRp genotype Ib enzyme was used at a final concentration of 28 nM.
  • a polyA template was used at 6 nM, and a biotinylated oligo-dT12 primer was used at 180 nM final concentration. Template was obtained commercially (Amersham 27- 4110).
  • Biotinylated primer was prepared by Sigma Genosys. 3H-UTP was used at 0.6 ⁇ Ci (0.29 ⁇ M total UTP). Reactions were initiated by the addition of enzyme, incubated at 30 0 C for 60 min, and stopped by adding 25 ⁇ l of 50 mM EDTA containing SPA beads (4 ⁇ g/ ⁇ l, Amersham RPNQ 0007). Plates were read on a Packard Top Count NXT after >lhr incubation at room temperature.
  • Modified HCVNS5B RdRp enzyme assay A modified enzyme assay was performed essentially as described for the standard enzyme assay except for the following: The biotinylated oligo dT12 primer was precaptured on streptavidin- coated SPA beads by mixing primer and beads in assay buffer and incubating at room temperature for one hour. Unbound primer was removed after centrifugation. The primer-bound beads were resuspended in 20 mM Hepes buffer, pH 7.5 and used in the assay at final concentrations of 20 nM primer and 0.67 ⁇ g/ ⁇ l beads.
  • enzyme 14 nM was added to diluted compound followed by the addition of a mixture of template (0.2 nM) , 3H-UTP (0.6 ⁇ Ci, 0.29 ⁇ M), and primer-bound beads, to initiate the reaction; concentrations given are final. Reactions were allowed to proceed for 4 hours at 30° C.
  • FRET Assay Preparation. To perform the HCV FRET screening assay, 96- well cell culture plates were used.
  • the FRET peptide (Anaspec, Inc.) (Taliani et al, Anal. Biochem. 1996, 240, 60-67) contains a fluorescence donor, EDANS, near one end of the peptide and an acceptor, DABCYL, near the other end.
  • the fluorescence of the peptide is quenched by intermolecular resonance energy transfer (RET) between the donor and the acceptor, but as the NS3 protease cleaves the peptide the products are released from RET quenching and the fluorescence of the donor becomes apparent.
  • RET intermolecular resonance energy transfer
  • the assay reagent was made as follows: 5X cell Luciferase cell culture lysis reagent from Promega (#E153A) diluted to IX with dH 2 O, NaCl added to 150 mM final, the FRET peptide diluted to 20 uM final from a 2 mM stock.
  • HCV replicon cells with or without a Renilla luciferase reporter gene, were trypsinized and placed into each well of a 96-well plate with titrated test compounds added in columns 3 through 12; columns 1 and 2 contained a control compound (HCV protease inhibitor), and the bottom row contained cells without compound.
  • the plates were then placed in a CO 2 incubator at 37 0 C.
  • FRET Assay Preparation at various times the plate was removed and Alamar blue solution (Trek Diagnostics, #00-100) was added per well as a measure of cellular toxicity. After reading in a Cytoflour 4000 instrument (PE Biosystems), plates were rinsed with PBS and then used for FRET assay by the addition of 30 ul of the FRET peptide assay reagent described above (FRET Assay Preparation) per well. The plate was then placed into the Cytoflour 4000 instrument which had been set to 340 excite/490 emission, automatic mode for 20 cycles and the plate read in a kinetic mode. Typically, the signal to noise using an endpoint analysis after the reads was at least three- fold.
  • Compound analysis was determined by quantification of the relative HCV replicon inhibition and the relative cytotoxicity values.
  • cytoxicity values the average Alamar Blue fluorescence signals from the control wells were set as 100% non-toxic. The individual signals in each of the compound test wells were then divided by the average control signal and multiplied by 100% to determine percent cytotoxicity.
  • HCV replicon inhibition values an average background value was obtained from the two wells containing the highest amount of HCV protease inhibitor at the end of the assay period. These numbers were similar to those obtained from na ⁇ ve Huh-7 cells.
  • the background numbers were then subtracted from the average signal obtained from the control wells and this number was used as 100% activity.
  • the individual signals in each of the compound test wells were then divided by the averaged control values after background subtraction and multiplied by 100% to determine percent activity.
  • EC50 values for a protease inhibitor titration were calculated as the concentration which caused a 50% reduction in FRET or luciferase activity.
  • the two numbers generated for the compound plate, percent cytoxicity and percent activity were used to determine compounds of interest for further analysis.
  • compositions comprising a compound, or a pharmaceutically acceptable salt or solvate thereof, and a pharmaceutically acceptable carrier.
  • a composition further comprising a compound having anti-HCV activity.
  • Another aspect of the invention is a composition where the compound having anti-HCV activity is an interferon.
  • the interferon is selected from interferon alpha 2B, pegylated interferon alpha, consensus interferon, interferon alpha 2A, and lymphoblastoid interferon tau.
  • compositions where the compound having anti-HCV activity is a cyclosporin.
  • cyclosporin is cyclosporin A.
  • compositions where the compound having anti-HCV activity is selected from the group consisting of interleukin 2, interleukin 6, interleukin 12, a compound that enhances the development of a type 1 helper T cell response, interfering RNA, anti-sense RNA, Imiqimod, ribavirin, an inosine 5'- monophospate dehydrogenase inhibitor, amantadine, and rimantadine.
  • Another aspect of the invention is a composition where the compound having anti-HCV activity is effective to inhibit the function of a target selected from HCV metalloprotease, HCV serine protease, HCV polymerase, HCV helicase, HCV NS4B protein, HCV entry, HCV assembly, HCV egress, HCV NS5A protein, IMPDH, and a nucleoside analog for the treatment of an HCV infection.
  • a target selected from HCV metalloprotease, HCV serine protease, HCV polymerase, HCV helicase, HCV NS4B protein, HCV entry, HCV assembly, HCV egress, HCV NS5A protein, IMPDH, and a nucleoside analog for the treatment of an HCV infection.
  • compositions comprising a compound, or a pharmaceutically acceptable salt thereof, a pharmaceutically acceptable carrier, an interferon and ribavirin.
  • Another aspect of the invention is a method of inhibiting the function of the HCV replicon comprising contacting the HCV replicon with a compound or a pharmaceutically acceptable salt thereof.
  • Another aspect of the invention is a method of inhibiting the function of the HCV NS5B protein comprising contacting the HCV NS5B protein with a compound or a pharmaceutically acceptable salt thereof.
  • Another aspect of the invention is a method of treating an HCV infection in a patient comprising administering to the patient a therapeutically effective amount of a compound or a pharmaceutically acceptable solvate or salt thereof.
  • the compound is effective to inhibit the function of the HCV replicon.
  • the compound is effective to inhibit the function of the HCV NS5B protein.
  • Another aspect of the invention is a method of treating an HCV infection in a patient comprising administering to the patient a therapeutically effective amount of a compound, or a pharmaceutically acceptable solvate or salt thereof, in conjunction with (prior to, after, or concurrently) another compound having anti-HCV activity.
  • Another aspect of the invention is the method where the other compound having anti-HCV activity is an interferon.
  • interferon is selected from interferon alpha 2B, pegylated interferon alpha, consensus interferon, interferon alpha 2A, and lymphoblastoid interferon tau.
  • Another aspect of the invention is the method where the other compound having anti-HCV activity is a cyclosporin.
  • Another aspect of the invention is the method where the cyclosporin is cyclosporin A.
  • Another aspect of the invention is the method where the other compound having anti-HCV activity is selected from interleukin 2, interleukin 6, interleukin 12, a compound that enhances the development of a type 1 helper T cell response, interfering RNA, anti-sense RNA, Imiqimod, ribavirin, an inosine 5'-monophospate dehydrogenase inhibitor, amantadine, and rimantadine.
  • Another aspect of the invention is the method where the other compound having anti-HCV activity is effective to inhibit the function of a target selected from the group consisting of HCV metalloprotease, HCV serine protease, HCV polymerase, HCV helicase, HCV NS4B protein, HCV entry, HCV assembly, HCV egress, HCV NS5A protein, IMPDH, and a nucleoside analog for the treatment of an HCV infection.
  • a target selected from the group consisting of HCV metalloprotease, HCV serine protease, HCV polymerase, HCV helicase, HCV NS4B protein, HCV entry, HCV assembly, HCV egress, HCV NS5A protein, IMPDH, and a nucleoside analog for the treatment of an HCV infection.
  • Another aspect of the invention is the method where the other compound having anti-HCV activity is effective to inhibit the function of target in the HCV life cycle other than the HCV NS5B protein.
  • “Therapeutically effective” means the amount of agent required to provide a meaningful patient benefit as understood by practitioners in the field of hepatitis and HCV infection.
  • Patient means a person infected with the HCV virus and suitable for therapy as understood by practitioners in the field of hepatitis and HCV infection.
  • the compounds of this invention are generally given as pharmaceutical compositions comprised of a therapeutically effective amount of a compound or its pharmaceutically acceptable salt and a pharmaceutically acceptable carrier and may contain conventional excipients.
  • a therapeutically effective amount is that which is needed to provide a meaningful patient benefit.
  • Pharmaceutically acceptable carriers are those conventionally known carriers having acceptable safety profiles.
  • compositions encompass all common solid and liquid forms including capsules, tablets, losenges, and powders as well as liquid suspensions, syrups, elixers, and solutions. Compositions are made using common formulation techniques, and conventional excipients (such as binding and wetting agents) and vehicles (such as water and alcohols) are generally used for compositions.
  • compositions are normally formulated in dosage units and compositions providing from about 1 to 1000 mg of the active ingredient per dose are preferred. Some examples of dosages are 1 mg, 10 mg, 100 mg, 250 mg, 500 mg, and 1000 mg. Generally, other agents will be present in a unit range similar to agents of that class used clinically. Typically, this is 0.25-1000 mg/unit.
  • Liquid compositions are usually in dosage unit ranges. Generally, the liquid composition will be in a unit dosage range of 1-100 mg/mL. Some examples of dosages are 1 mg/mL, 10 mg/mL, 25 mg/mL, 50 mg/mL, and 100 mg/mL. Generally, other agents will be present in a unit range similar to agents of that class used clinically. Typically, this is 1-100 mg/mL.
  • the invention encompasses all conventional modes of administration; oral and parenteral methods are preferred.
  • the dosing regimen will be similar to other agents used clinically.
  • the daily dose will be 1-100 mg/kg body weight daily.
  • more compound is required orally and less parenterally.
  • the specific dosing regime will be determined by a physician using sound medical judgement.
  • the invention also encompasses methods where the compound is given in combination therapy. That is, the compound can be used in conjunction with, but separately from, other agents useful in treating hepatitis and HCV infection.
  • the compound will generally be given in a daily dose of 1-100 mg/kg body weight daily in conjunction with other agents.
  • the other agents generally will be given in the amounts used therapeutically.
  • the specific dosing regime will be determined by a physician using sound medical judgement.
  • N 5 N- Dimethylsulfamide (0.94 g, 7.56 mmol) was added followed by the dropwise addition of a solution of DBU (1.34 g ,8.8 mmol) in THF (4 mL). Stirring was continued for 24 hr. The mixture was partitioned between ethyl acetate and dilute HCl. The ethyl acetate layer was washed with water followed by brine and dried over Na 2 SO 4 . The extract was concentrated to dryness to leave the title product as a pale yellow friable foam, (2.0 g, 74 %, >90 % purity , estimated from NMR).
  • the reaction mixture was then diluted with ethyl acetate and water, and then acidified with an excess of dilute HCl.
  • the ethyl acetate layer was then collected and washed with dilute HCl, water and brine.
  • the organic solution was then dried (magnesium sulfate), filtered and concentrated to give a gum.
  • the gum was diluted with hexanes (250 ml) and ethyl acetate (25 mL), and the mixture was stirred for 20 hr at 22° C during which time the product was transformed into a bright yellow granular solid (4.8 g) which was used directly without further purification.
  • DPPA Diphenylphosphorylazide
  • 3 equivalents of triethylamine The solution was stirred overnight at room temperature.
  • the crude mixture was passed through a 24g SiliCycle-IscoTM silica gel cartridge with DCE to give acyl azide after solvent evacuation (50-65% yield).
  • the acyl azide was found to be stable at room temperature in a vacuum desiccator for up to three months.
  • To a 50 mL RBF was added 0.2mmol of acyl azide in 5.0 mL of dry toluene. The mixture was heated in an oil bath at 120 0 C for 15 minutes then quickly cooled to room temperature.
  • Examples 12 - 17 use the general methods below. Analytical HPLC and LC/MS were performed by using Shimadzu-VP instrument with UV detection at 220 nm and Waters Micromass. Biotage Horizon was used for flash chromatography as indicated.
  • hydrochloride salt of (l ⁇ R,12W ⁇ )-l ⁇ -amino-8-cyclohexyl-N- (dimethylsulfamoyl)- 11 -methoxy- 1 , 1 a,2, 12&-tetrahydrocyclopropa[ ⁇ i]indolo[2, 1 - ⁇ ][2]benzazepine-5-carboxamide was prepared from (BMS-807598) tert-butyl (( 1 ciR, 12&S)-8-cyclohexyl-5 -((dimethylsulfamoyl)carbamoyl)- 11 -methoxy- 1 , 12b- dihydrocyclopropa[ ⁇ i]indolo[2,l- ⁇ ][2]benzazepin-l ⁇ (2H)-yl)carbamate in a similar manner as described above.
  • hydrochloride salt of (l ⁇ «S,12/?R)-la-Amino-8-cyclohexyl-N- (dimethylsulfamoyl)- 11 -methoxy- 1 , 1 a,2, 12&-tetrahydrocyclopropa[ ⁇ i]indolo[2, 1 - ⁇ ][2]benzazepine-5-carboxamide was prepared from tert-butyl ((laS,l2bR)-8- cyclohexyl-5 -((dimethylsulfamoyl)carbamoy I)- 11 -methoxy- 1,12b- dihydrocyclopropa[ ⁇ i]indolo[2,l- ⁇ ][2]benzazepin-l ⁇ (2H)-yl)carbamate in a similar manner as described above.

Landscapes

  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Oncology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Communicable Diseases (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Virology (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

The invention encompasses compounds (I) as well as compositions and methods of using the compounds. The compounds have activity against hepatitis C virus (HCV) and are useful in treating those infected with HCV.

Description

CYCLOPROPYL FUSED INDOLOBENZAZEPINE HCV INHIBITORS
CROSS-REFERENCE TO RELATED APPLICATIONS
This application claims the benefit of U.S. Provisional Application Serial
Numbers 60/894,764 filed March 14, 2007 and 60/804,203 filed June 8, 2006.
BACKGROUND OF THE INVENTION
Hepatitis C virus (HCV) is a major human pathogen, infecting an estimated
170 million persons worldwide - roughly five times the number infected by human immunodeficiency virus type 1. A substantial fraction of these HCV infected individuals develop serious progressive liver disease, including cirrhosis and hepatocellular carcinoma (Lauer, G. M.; Walker, B. D. N. Engl. J. Med. 2001, 345, 41-52).
HCV is a positive-stranded RΝA virus. Based on a comparison of the deduced amino acid sequence and the extensive similarity in the 5 '-untranslated region, HCV has been classified as a separate genus in the Flaviviridae family. All members of the Flaviviridae family have enveloped virions that contain a positive stranded RΝA genome encoding all known virus-specific proteins via translation of a single, uninterrupted, open reading frame.
Considerable heterogeneity is found within the nucleotide and encoded amino acid sequence throughout the HCV genome. At least six major genotypes have been characterized, and more than 50 subtypes have been described. The major genotypes of HCV differ in their distribution worldwide, and the clinical significance of the genetic heterogeneity of HCV remains elusive despite numerous studies of the possible effect of genotypes on pathogenesis and therapy.
The single strand HCV RΝA genome is approximately 9500 nucleotides in length and has a single open reading frame (ORF) encoding a single large polyprotein of about 3000 amino acids. In infected cells, this polyprotein is cleaved at multiple sites by cellular and viral proteases to produce the structural and non-structural (NS) proteins. In the case of HCV, the generation of mature non-structural proteins (NS2, NS3, NS4A, NS4B, NS5A, and NS5B) is effected by two viral proteases. The first one is believed to be a metalloprotease and cleaves at the NS2-NS3 junction; the second one is a serine protease contained within the N-terminal region of NS3 (also referred to as NS3 protease) and mediates all the subsequent cleavages downstream of NS3, both in cis, at the NS3-NS4A cleavage site, and in trans, for the remaining NS4A-NS4B, NS4B-NS5A, NS5A-NS5B sites. The NS4A protein appears to serve multiple functions, acting as a cofactor for the NS3 protease and possibly assisting in the membrane localization of NS3 and other viral replicase components. The complex formation of the NS3 protein with NS4A seems necessary to the processing events, enhancing the proteolytic efficiency at all of the sites. The NS3 protein also exhibits nucleoside triphosphatase and RNA helicase activities. NS5B (also referred to as HCV polymerase) is a RNA-dependent RNA polymerase that is involved in the replication of HCV. The HCV NS5B protein is described in "Structural Analysis of the Hepatitis C Virus RNA Polymerase in Complex with Ribonucleotides (Bressanelli; S. et al, Journal of Virology 2002, 3482-3492; and Defrancesco and Rice, Clinics in Liver Disease 2003, 7, 211-242.
Currently, the most effective HCV therapy employs a combination of alpha- interferon and ribavirin, leading to sustained efficacy in 40% of patients (Poynard, T. et al. Lancet 1998, 352, 1426-1432). Recent clinical results demonstrate that pegylated alpha-interferon is superior to unmodified alpha-interferon as monotherapy (Zeuzem, S. et al. N. Engl. J. Med. 2000, 343, 1666-1672). However, even with experimental therapeutic regimens involving combinations of pegylated alpha- interferon and ribavirin, a substantial fraction of patients do not have a sustained reduction in viral load. Thus, there is a clear and important need to develop effective therapeutics for treatment of HCV infection.
DESCRIPTION OF THE INVENTION
One aspect of the invention is a compound of formula I
Figure imgf000004_0001
where:
R1 is CO2R5 or CONR6R7;
R2 is N(R1O)(RU);
R3 is hydrogen, halo, alkyl, alkenyl, hydroxy, benzyloxy, alkoxy, or haloalkoxy;
R4 is cycloalkyl;
R5 is hydrogen or alkyl;
R6 is hydrogen, alkyl, alkylSO2, cycloalkylSO2, haloalkylSO2, (R7)(R8)NSO2, or
(R9)SO2;
R7 is hydrogen or alkyl;
R8 is hydrogen or alkyl;
R9 is azetidinyl, pyrrolidinyl, piperidinyl, piperazinyl, morpholinyl, thiomorpholinyl, homopiperidinyl, or homomorpholinyl, and is substituted with 0-3 substituents selected from alkyl and alkoxy;
R10 is hydrogen, alkyl, alkoxyCO, or (R12)(R13)NCO;
R » 11 is hydrogen or alkyl; R12 is hydrogen, alkyl, alkoxyalkyl, aminoalkyl, (alkylamino)alkyl,
( (ddiiaallkkyyllaammiinnoo))aallkkyyll,, ((CCOOj2R14)alkyl, (CON(R15)(R16))alkyl, or
(tetrahydropyranyl)alkyl;
R13 is hydrogen alkyl, alkoxyalkyl, aminoalkyl, (alkylamino)alkyl, or (dialkylamino)alkyl;
or (R12)(R13)N taken together is azetidinyl, pyrrolidinyl, piperidinyl, piperazinyl, morpholinyl, thiomorpholinyl, homopiperidinyl, or homomorpholinyl and is substituted with 0-3 substituents selected from alkyl and alkoxy;
R , 14 is hydrogen or alkyl;
R » 15 is hydrogen or alkyl; and
R , 16 is hydrogen or alkyl; or
N(R15)(R16) taken together is azetidinyl, pyrrolidinyl, piperidinyl, piperazinyl, morpholinyl, thiomorpholinyl, homopiperidinyl, or homomorpholinyl and is substituted with 0-3 substituents selected from alkyl and alkoxy;
or a pharmaceutically acceptable salt thereof.
Another aspect of the invention is a compound of formula I where R1 is CONHR6 and R6 is alkylSO2, cycloalkylSO2, haloalkylSO2, (R7)(R8)NSO2, or (R9)SO2.
Another aspect of the invention is a compound of formula I where R is hydrogen.
Another aspect of the invention is a compound of formula I where R3 is methoxy.
Another aspect of the invention is a compound of formula I where R4 is cyclohexyl. Another aspect of the invention is a compound of formula I where R6 is (R9^NSO2 or (R10)SO2.
Another aspect of the invention is a compound of formula I where R10 is (R12)(R13)NCO.
Another aspect of the invention is a compound of formula I where R12 is alkoxyalkyl, (dialkylamino)alkyl, (CON(R15)(R16))alkyl, or (tetrahydropyranyl)alkyl.
Another aspect of the invention is a compound of formula I where (R12)(R13)N taken together is azetidinyl, pyrrolidinyl, piperidinyl, piperazinyl, morpholinyl, thiomorpholinyl, homopiperidinyl, or homomorpholinyl and is substituted with 0-3 substituents selected from alkyl and alkoxy.
Another aspect of the invention is a compound of formula I with the following stereochemistry.
Figure imgf000006_0001
Another aspect of the invention is a compound of formula I with the following stereochemistry.
Figure imgf000006_0002
Any scope of any variable, including R1, R2, R3, R4, R5, R6, R7, R8, R9, R10, R11, R12, R13, R14, R15, and R16, can be used independently with the scope of any other instance of a variable. Unless specified otherwise, these terms have the following meanings. "Alkyl" means a straight or branched alkyl group composed of 1 to 6 carbons. "Alkenyl" means a straight or branched alkyl group composed of 2 to 6 carbons with at least one double bond. "Cycloalkyl" means a monocyclic ring system composed of 3 to 7 carbons. "Hydroxy alkyl," "alkoxy" and other terms with a substituted alkyl moiety include straight and branched isomers composed of 1 to 6 carbon atoms for the alkyl moiety. "Haloalkyl" and "haloalkoxy" include all halogenated isomers from monohalo substituted alkyl to perhalo substituted alkyl. "Aryl" includes carbocyclic and heterocyclic aromatic substituents. Parenthetic and multiparenthetic terms are intended to clarify bonding relationships to those skilled in the art. For example, a term such as ((R)alkyl) means an alkyl substituent further substituted with the substituent R.
The invention includes all pharmaceutically acceptable salt forms of the compounds. Pharmaceutically acceptable salts are those in which the counter ions do not contribute significantly to the physiological activity or toxicity of the compounds and as such function as pharmacological equivalents. These salts can be made according to common organic techniques employing commercially available reagents. Some anionic salt forms include acetate, acistrate, besylate, bromide, chloride, citrate, fumarate, glucouronate, hydrobromide, hydrochloride, hydroiodide, iodide, lactate, maleate, mesylate, nitrate, pamoate, phosphate, succinate, sulfate, tartrate, tosylate, and xinofoate. Some cationic salt forms include ammonium, aluminum, benzathine, bismuth, calcium, choline, diethylamine, diethanolamine, lithium, magnesium, meglumine, 4-phenylcyclohexylamine, piperazine, potassium, sodium, tromethamine, and zinc.
Some of the compounds of the invention possess asymmetric carbon atoms (for example, the structures below). The invention includes all stereoisomeric forms, including enantiomers and diastereomers as well as mixtures of stereoisomers such as racemates. Some stereoisomers can be made using methods known in the art.
Stereoisomeric mixtures of the compounds and related intermediates can be separated into individual isomers according to methods known in the art.
Figure imgf000008_0001
Synthetic Methods
The compounds may be made by methods known in the art including those described below. Some reagents and intermediates are known in the art. Other reagents and intermediates can be made by methods known in the art using readily available materials. The variables (e.g. numbered "R" substituents) used to describe the synthesis of the compounds are intended only to illustrate how to make the compounds and are not to be confused with variables used in the claims or in other sections of the specification.
Abbreviations used in the schemes generally follow conventions used in the art.
The compounds can be made by methods known in the art and described in the application.
2-bromo-3-cyclohexyl-lH-indole-6-carboxylic acid can be condensed with a variety of sulfonyl ureas, using for example l,l '-carbonyldiimidazole in combination with l,8-diazabicyclo[5.4.0]undec-7-ene in anhydrous THF. The resultant acyl sulfamides can be subjected to known coupling reactions, for example Suzuki coupling conditions with a diversity of 2-formyl boronic acids or esters to provide cyclic hemiaminal intermediates of the type depicted. These compounds can be converted to indolobenzazepines derivatives, using as one example, the methodology shown in Scheme 1. Related fused cyclopropyl ester derivatives can be generated by methods known in the art, including treatment of the indolobenzazepine esters with trimethyl sulfoxonium iodide under strongly basic conditions in DMSO. The residual aliphatic ester moiety in the resultant fused cyclopropanes can be hydrolyzed using sodium hydroxide in an aqueous methanolic THF mixture. Scheme 1.
Figure imgf000009_0001
Figure imgf000009_0002
Scheme Ia.
Na2CO3 80°C
Figure imgf000010_0002
Figure imgf000010_0003
Figure imgf000010_0001
The resultant acids can be converted to ureas by methods known in the art, for example treatment with diphenylphosphoryl azide and triethylamine in toluene, and the intermediate acyl azide can be isolated, and then treated in a separate step with a diversity of amines to afford the urea examples as shown in Scheme 2.
Scheme 2.
Figure imgf000011_0001
As an alternative, the intermediate acid ester shown in Scheme 3 can be subjected to a Curtius reaction using a diversity of amines, and the resultant esters can be hydro lyzed to provide examples of carboxamide acids. A coupling with a sulfamide or sulfonamide as shown in the last step of Scheme 3 can provide an alternate preparation of compounds of the invention.
Scheme 3.
DCE
Figure imgf000012_0002
Figure imgf000012_0001
HNR6R7
TMSOK
THF C
Figure imgf000012_0003
Figure imgf000012_0004
Biological Methods
The compounds demonstrated activity against HCV NS5B as determined in the following HCV RdRp assays.
HCVNS5B RdRp cloning, expression, and purification. The cDNA encoding the NS5B protein of HCV, genotype Ib, was cloned into the pET21a expression vector. The protein was expressed with an 18 amino acid C-terminal truncation to enhance the solubility. The E. coli competent cell line BL21(DE3) was used for expression of the protein. Cultures were grown at 37 0C for ~ 4 hours until the cultures reached an optical density of 2.0 at 600 nm. The cultures were cooled to 20 0C and induced with 1 mM IPTG. Fresh ampicillin was added to a final concentration of 50 μg/ml and the cells were grown overnight at 20 0C. Cell pellets (3L) were lysed for purification to yield 15-24 mgs of purified NS5B. The lysis buffer consisted of 20 mM Tris-HCl, pH 7.4, 500 mM NaCl, 0.5% triton X-100, 1 mM DTT, ImM EDTA, 20% glycerol, 0.5 mg/ml lysozyme, 10 mM MgC12, 15 ug/ml deoxyribonuclease I, and Complete TM protease inhibitor tablets (Roche). After addition of the lysis buffer, frozen cell pellets were resuspended using a tissue homogenizer. To reduce the viscosity of the sample, aliquots of the lysate were sonicated on ice using a microtip attached to a Branson sonicator. The sonicated lysate was centrifuged at 100,000 x g for lhr at 4 0C and filtered through a 0.2 μm filter unit (Corning).
The protein was purified using three sequential chromatography steps: Heparin sepharose CL-6B, polyU sepharose 4B, and Hitrap SP sepharose (Pharmacia). The chromatography buffers were identical to the lysis buffer but contained no lysozyme, deoxyribonuclease I, MgC12 or protease inhibitor and the NaCl concentration of the buffer was adjusted according to the requirements for charging the protein onto the column. Each column was eluted with a NaCl gradient which varied in length from 5-50 column volumes depending on the column type. After the final chromatography step, the resulting purity of the enzyme is >90% based on SDS-PAGE analysis. The enzyme was aliquoted and stored at -80 0C.
Standard HCVNS5B RdRp enzyme assay. HCV RdRp genotype Ib assays were run in a final volume of 60 μl in 96 well plates (Costar 3912). The assay buffer is composed of 20 mM Hepes, pH 7.5, 2.5 mM KCl, 2.5 mM MgC12, 1 mM DTT, 1.6 U RNAse inhibitor (Promega N2515), 0.1 mg/ml BSA (Promega R3961), and 2 % glycerol. All compounds were serially diluted (3 -fold) in DMSO and diluted further in water such that the final concentration of DMSO in the assay was 2%. HCV RdRp genotype Ib enzyme was used at a final concentration of 28 nM. A polyA template was used at 6 nM, and a biotinylated oligo-dT12 primer was used at 180 nM final concentration. Template was obtained commercially (Amersham 27- 4110). Biotinylated primer was prepared by Sigma Genosys. 3H-UTP was used at 0.6 μCi (0.29 μM total UTP). Reactions were initiated by the addition of enzyme, incubated at 30 0C for 60 min, and stopped by adding 25 μl of 50 mM EDTA containing SPA beads (4 μg/μl, Amersham RPNQ 0007). Plates were read on a Packard Top Count NXT after >lhr incubation at room temperature.
Modified HCVNS5B RdRp enzyme assay. A modified enzyme assay was performed essentially as described for the standard enzyme assay except for the following: The biotinylated oligo dT12 primer was precaptured on streptavidin- coated SPA beads by mixing primer and beads in assay buffer and incubating at room temperature for one hour. Unbound primer was removed after centrifugation. The primer-bound beads were resuspended in 20 mM Hepes buffer, pH 7.5 and used in the assay at final concentrations of 20 nM primer and 0.67 μg/μl beads. Order of addition in the assay: enzyme (14 nM) was added to diluted compound followed by the addition of a mixture of template (0.2 nM) , 3H-UTP (0.6 μCi, 0.29 μM), and primer-bound beads, to initiate the reaction; concentrations given are final. Reactions were allowed to proceed for 4 hours at 30° C.
IC50 values for compounds were determined using seven different [I]. IC50 values were calculated from the inhibition using the formula y = A+((B- A)/(l+((C/x)ΛD))).
FRET Assay Preparation. To perform the HCV FRET screening assay, 96- well cell culture plates were used. The FRET peptide (Anaspec, Inc.) (Taliani et al, Anal. Biochem. 1996, 240, 60-67) contains a fluorescence donor, EDANS, near one end of the peptide and an acceptor, DABCYL, near the other end. The fluorescence of the peptide is quenched by intermolecular resonance energy transfer (RET) between the donor and the acceptor, but as the NS3 protease cleaves the peptide the products are released from RET quenching and the fluorescence of the donor becomes apparent. The assay reagent was made as follows: 5X cell Luciferase cell culture lysis reagent from Promega (#E153A) diluted to IX with dH2O, NaCl added to 150 mM final, the FRET peptide diluted to 20 uM final from a 2 mM stock.
To prepare plates, HCV replicon cells, with or without a Renilla luciferase reporter gene, were trypsinized and placed into each well of a 96-well plate with titrated test compounds added in columns 3 through 12; columns 1 and 2 contained a control compound (HCV protease inhibitor), and the bottom row contained cells without compound. The plates were then placed in a CO2 incubator at 37 0C.
Assays. Subsequent to addition of the test compounds described above
(FRET Assay Preparation), at various times the plate was removed and Alamar blue solution (Trek Diagnostics, #00-100) was added per well as a measure of cellular toxicity. After reading in a Cytoflour 4000 instrument (PE Biosystems), plates were rinsed with PBS and then used for FRET assay by the addition of 30 ul of the FRET peptide assay reagent described above (FRET Assay Preparation) per well. The plate was then placed into the Cytoflour 4000 instrument which had been set to 340 excite/490 emission, automatic mode for 20 cycles and the plate read in a kinetic mode. Typically, the signal to noise using an endpoint analysis after the reads was at least three- fold. Alternatively, after Alamar blue reading, plates were rinsed with PBS, 50 ul of DMEM (high glucose) without phenol red was added and plates were then used for luciferase assay using the Promega Dual-Glo Luciferase Assay System.
Compound analysis was determined by quantification of the relative HCV replicon inhibition and the relative cytotoxicity values. To calculate cytoxicity values, the average Alamar Blue fluorescence signals from the control wells were set as 100% non-toxic. The individual signals in each of the compound test wells were then divided by the average control signal and multiplied by 100% to determine percent cytotoxicity. To calculate the HCV replicon inhibition values, an average background value was obtained from the two wells containing the highest amount of HCV protease inhibitor at the end of the assay period. These numbers were similar to those obtained from naϊve Huh-7 cells.
The background numbers were then subtracted from the average signal obtained from the control wells and this number was used as 100% activity. The individual signals in each of the compound test wells were then divided by the averaged control values after background subtraction and multiplied by 100% to determine percent activity. EC50 values for a protease inhibitor titration were calculated as the concentration which caused a 50% reduction in FRET or luciferase activity. The two numbers generated for the compound plate, percent cytoxicity and percent activity were used to determine compounds of interest for further analysis.
Representative data for compounds are reported in Table 1.
Figure imgf000016_0001
Figure imgf000017_0001
Figure imgf000018_0001
Figure imgf000019_0001
A>1 μM, B 0 02 μM - 1 μM, EC50 C >10 μM, D 1 μM - 10 μM, E 1 0 μM - 0 07 μM,F <0 02μM,G >0 37μM IC50 values were determined using the preincubation protocol EC50 values were determined using the FRET assay
Pharmaceutical Compositions and Methods of Treatment
The compounds demonstrate activity against HCV NS5B and can be useful in treating HCV and HCV infection. Therefore, another aspect of the invention is a composition comprising a compound, or a pharmaceutically acceptable salt or solvate thereof, and a pharmaceutically acceptable carrier. Another aspect of the invention is a composition further comprising a compound having anti-HCV activity.
Another aspect of the invention is a composition where the compound having anti-HCV activity is an interferon. Another aspect of the invention is where the interferon is selected from interferon alpha 2B, pegylated interferon alpha, consensus interferon, interferon alpha 2A, and lymphoblastoid interferon tau.
Another aspect of the invention is a composition where the compound having anti-HCV activity is a cyclosporin. Another aspect of the invention is where the cyclosporin is cyclosporin A.
Another aspect of the invention is a composition where the compound having anti-HCV activity is selected from the group consisting of interleukin 2, interleukin 6, interleukin 12, a compound that enhances the development of a type 1 helper T cell response, interfering RNA, anti-sense RNA, Imiqimod, ribavirin, an inosine 5'- monophospate dehydrogenase inhibitor, amantadine, and rimantadine.
Another aspect of the invention is a composition where the compound having anti-HCV activity is effective to inhibit the function of a target selected from HCV metalloprotease, HCV serine protease, HCV polymerase, HCV helicase, HCV NS4B protein, HCV entry, HCV assembly, HCV egress, HCV NS5A protein, IMPDH, and a nucleoside analog for the treatment of an HCV infection.
Another aspect of the invention is a composition comprising a compound, or a pharmaceutically acceptable salt thereof, a pharmaceutically acceptable carrier, an interferon and ribavirin.
Another aspect of the invention is a method of inhibiting the function of the HCV replicon comprising contacting the HCV replicon with a compound or a pharmaceutically acceptable salt thereof. Another aspect of the invention is a method of inhibiting the function of the HCV NS5B protein comprising contacting the HCV NS5B protein with a compound or a pharmaceutically acceptable salt thereof.
Another aspect of the invention is a method of treating an HCV infection in a patient comprising administering to the patient a therapeutically effective amount of a compound or a pharmaceutically acceptable solvate or salt thereof. In another embodiment the compound is effective to inhibit the function of the HCV replicon. In another embodiment the compound is effective to inhibit the function of the HCV NS5B protein.
Another aspect of the invention is a method of treating an HCV infection in a patient comprising administering to the patient a therapeutically effective amount of a compound, or a pharmaceutically acceptable solvate or salt thereof, in conjunction with (prior to, after, or concurrently) another compound having anti-HCV activity.
Another aspect of the invention is the method where the other compound having anti-HCV activity is an interferon.
Another aspect of the invention is the method where the interferon is selected from interferon alpha 2B, pegylated interferon alpha, consensus interferon, interferon alpha 2A, and lymphoblastoid interferon tau.
Another aspect of the invention is the method where the other compound having anti-HCV activity is a cyclosporin.
Another aspect of the invention is the method where the cyclosporin is cyclosporin A.
Another aspect of the invention is the method where the other compound having anti-HCV activity is selected from interleukin 2, interleukin 6, interleukin 12, a compound that enhances the development of a type 1 helper T cell response, interfering RNA, anti-sense RNA, Imiqimod, ribavirin, an inosine 5'-monophospate dehydrogenase inhibitor, amantadine, and rimantadine.
Another aspect of the invention is the method where the other compound having anti-HCV activity is effective to inhibit the function of a target selected from the group consisting of HCV metalloprotease, HCV serine protease, HCV polymerase, HCV helicase, HCV NS4B protein, HCV entry, HCV assembly, HCV egress, HCV NS5A protein, IMPDH, and a nucleoside analog for the treatment of an HCV infection.
Another aspect of the invention is the method where the other compound having anti-HCV activity is effective to inhibit the function of target in the HCV life cycle other than the HCV NS5B protein.
"Therapeutically effective" means the amount of agent required to provide a meaningful patient benefit as understood by practitioners in the field of hepatitis and HCV infection.
"Patient" means a person infected with the HCV virus and suitable for therapy as understood by practitioners in the field of hepatitis and HCV infection.
"Treatment," "therapy," "regimen," "HCV infection," and related terms are used as understood by practitioners in the field of hepatitis and HCV infection.
The compounds of this invention are generally given as pharmaceutical compositions comprised of a therapeutically effective amount of a compound or its pharmaceutically acceptable salt and a pharmaceutically acceptable carrier and may contain conventional excipients. A therapeutically effective amount is that which is needed to provide a meaningful patient benefit. Pharmaceutically acceptable carriers are those conventionally known carriers having acceptable safety profiles.
Compositions encompass all common solid and liquid forms including capsules, tablets, losenges, and powders as well as liquid suspensions, syrups, elixers, and solutions. Compositions are made using common formulation techniques, and conventional excipients (such as binding and wetting agents) and vehicles (such as water and alcohols) are generally used for compositions.
Solid compositions are normally formulated in dosage units and compositions providing from about 1 to 1000 mg of the active ingredient per dose are preferred. Some examples of dosages are 1 mg, 10 mg, 100 mg, 250 mg, 500 mg, and 1000 mg. Generally, other agents will be present in a unit range similar to agents of that class used clinically. Typically, this is 0.25-1000 mg/unit.
Liquid compositions are usually in dosage unit ranges. Generally, the liquid composition will be in a unit dosage range of 1-100 mg/mL. Some examples of dosages are 1 mg/mL, 10 mg/mL, 25 mg/mL, 50 mg/mL, and 100 mg/mL. Generally, other agents will be present in a unit range similar to agents of that class used clinically. Typically, this is 1-100 mg/mL.
The invention encompasses all conventional modes of administration; oral and parenteral methods are preferred. Generally, the dosing regimen will be similar to other agents used clinically. Typically, the daily dose will be 1-100 mg/kg body weight daily. Generally, more compound is required orally and less parenterally. The specific dosing regime, however, will be determined by a physician using sound medical judgement.
The invention also encompasses methods where the compound is given in combination therapy. That is, the compound can be used in conjunction with, but separately from, other agents useful in treating hepatitis and HCV infection. In these combination methods, the compound will generally be given in a daily dose of 1-100 mg/kg body weight daily in conjunction with other agents. The other agents generally will be given in the amounts used therapeutically. The specific dosing regime, however, will be determined by a physician using sound medical judgement.
Some examples of compounds suitable for compositions and methods are listed in Table 2. Table 2.
Figure imgf000024_0001
Figure imgf000025_0001
Figure imgf000026_0002
DESCRIPTION OF SPECIFIC EMBODIMENTS
Analytical HPLC and LC/MS were performed by using Shimadzu-VP instrument with UV detection at 220 nM and Waters Micromass. Biotage Horizon was used for flash chromatography as indicated.
Intermediate 1
Figure imgf000026_0001
2-Bromo-3-cyclohexyl- N-[(dimethylamino)sulfonyl]-lH-indole-6- carboxamide. 1,1 '-Carbonyldiimidazole (1.17 g, 7.2 mmol) was added to a stirred solution of 2-bromo-3-cyclohexyl-lH-indole-6-carboxylic acid (2.03 g, 6.3 mmol) in THF (6 mL) at 220C. The evolution of CO2 was instantaneous and when it slowed the solution was heated at 500C for 1 hr and then cooled to 220C. N5N- Dimethylsulfamide (0.94 g, 7.56 mmol) was added followed by the dropwise addition of a solution of DBU (1.34 g ,8.8 mmol) in THF (4 mL). Stirring was continued for 24 hr. The mixture was partitioned between ethyl acetate and dilute HCl. The ethyl acetate layer was washed with water followed by brine and dried over Na2SO4. The extract was concentrated to dryness to leave the title product as a pale yellow friable foam, (2.0 g, 74 %, >90 % purity , estimated from NMR). 1H NMR (300 MHz, DMSO-D6) δ ppm 1.28 - 1.49 (m, 3 H) 1.59 - 2.04 (m, 7 H) 2.74 - 2.82 (m, 1 H) 2.88 (s, 6 H) 7.57 (dd, J=8.42, 1.46 Hz, 1 H) 7.74 (d, J=8.78 Hz, 1 H) 7.91 (s, 1 H) 11.71 (s, 1 H) 12.08 (s, 1 H).
Intermediate 2
Figure imgf000027_0001
3-cyclohexyl-N-(N,N-dimethylsulfamoyl)-2-(2-formyl-4-methoxyphenyl)-lH- indole-6-carboxamide. A mixture of the 2-Bromo-3-cyclohexyl- N- [(dimethylamino)sulfonyl]-lH-indole-6-carboxamide (4.28g, 0.01 mol), boronic acid (2.7g, 0.015 mol), 2-dicyclohexylphosphino-2',6'-dimethoxy-biphenyl (41 mg, 0.0001 mol), palladium acetate (11.2 mg), and finely ground potassium carbonate (4.24g, 0.02 mol) in toluene (30 mL) was stirred under reflux and under nitrogen for 30 min, at which time LC/MS analysis showed the reaction to be complete. The reaction mixture was then diluted with ethyl acetate and water, and then acidified with an excess of dilute HCl. The ethyl acetate layer was then collected and washed with dilute HCl, water and brine. The organic solution was then dried (magnesium sulfate), filtered and concentrated to give a gum. The gum was diluted with hexanes (250 ml) and ethyl acetate (25 mL), and the mixture was stirred for 20 hr at 22° C during which time the product was transformed into a bright yellow granular solid (4.8 g) which was used directly without further purification.
Intermediate 3
Figure imgf000028_0001
6-Carbomethoxy-13-cyclohexyl-N-[(dimethylamino)sulfonyl]-5H-indolo[2,l- a] [2] benzazepine-10-carboxamide. A mixture of the 3-cyclohexyl-N-(N,N- dimethylsulfamoyl)-2-(2-formyl-4-methoxyphenyl)-lH-indole-6-carboxamide (4.8g, 0.01 mol), and cesium carbonate (7.1g, 0.02 mol) in DMF (28mL) was stirred for 20 hr at an oil bath temperature of 55 ° C. The mixture was poured into ice-water and acidified with dilute HCl to precipitate the crude product. The solid was collected, dried and flash chromatographed on Siθ2 (11Og) using an ethyl acetate and methylene chloride (1: 10) solution containing 2% acetic acid. Homogeneous fractions were combined and evaporated to afford the title compound as a pale yellow solid (3.9g, 71 % yield). MS: 552 (M=H+).
Intermediate 4
Figure imgf000028_0002
(+/~) Cycloprop[d]indolo[2, 1-a] [2]benzazepine-5-carboxylic acid, 8- cyclohexyl- 1 , Ia, 2, 12b-tetrahydro-l l-methoxy-la-[(4-morphoHnylcarbonyl)amino]- methyl ester. DMSO (5 mL) was added to a mixture of trimethylsulfoxonium iodide (199 mg, 0.906 mmol) and NaH (38 mg in 60% oil dispersion, 0.953 mmol) in a round-bottomed flask. The reaction mixture was stirred at rt for 0.5 hr. 7H- Indolo[2, 1 -a] [2]benzazepine-6-carboxylic acid, 13-cyclohexyl- 10- [[[(dimethylamino)sulfonyl]amino]carbonyl]-3-(methoxy)-, methyl ester (125 mg, 0.227 mmol) was then added and the reaction mixture was stirred at rt. for 3 hr., and then at 500C for a further 3 hr. The reaction was then quenched with water and acidified with IN HCl solution. The crude product then precipitated as a light yellow solid which was collected by filtration and air dried, (106 mg, 83% yield). 6 mg of this material was then purified by Prep. HPLC to afford the title compound as a light yellow solid (1.8 mg). MS m/z 566(MH+), Retention time: 3.850 min.lH NMR (500 MHz, MeOD) δ ppm 0.28 (m, 0.36 H) 1.19 - 2.20 (m, 11.64 H) 2.70 - 3.02 (m, 2 H) 3.03 (s, 2.16 H) 3.05 (s, 3.84 H) 3.49 (d, J=15.26 Hz, 0.64 H) 3.54 (s, 1.92 H) 3.83 (s, 1.08 H) 3.91 (s, 3 H) 4.08 (d, J=15.26 Hz, 0.36 H) 5.29 (d, J=15.26 Hz, 0.36 H) 5.50 (d, J=14.95 Hz, 0.64 H) 6.98 - 7.06 (m, 1 H) 7.16 (d, J=2.44 Hz, 0.36 H) 7.23 (d, J=2.44 Hz, 0.64 H) 7.30 (d, J=8.55 Hz, 0.64 H) 7.34 (d, J=8.55 Hz, 0.36 H) 7.56 (dd, J=8.55, 1.53 Hz, 0.64 H) 7.63 (dd, J=8.55, 1.53 Hz, 0.36 H) 7.88 (d, J=8.55 Hz, 0.64 H) 7.91 (d, J=8.55 Hz, 0.36 H) 8.12 (s, 0.36 H) 8.33 (d, J=I.53 Hz, 0.64 H).
Intermediate 5
Figure imgf000029_0001
(+/-) Cycloprop[d]indolo[2,l-a] ' [2] benzazepine-1 a(2H)-carboxylic acid, 8- cyclohexyl-5-[[[(dimethylamino)sulfonyl] amino] carbonyl] -1 ,12b-dihydro-l 1- methoxy-. To a solution of (+/-) Cycloprop[d]indolo[2,l-a][2]benzazepine-5- carboxylic acid, 8-cyclohexyl-l,la,2,12b-tetrahydro-l 1 -methoxy- la-[(4- morpholinylcarbonyl)amino]-methyl ester (100 mg, 0.177 mmol) in THF/Methanol mixture (2.0 mL/2.0 mL), 2N NaOH solution (1.0 mL) was added. The reaction mixture was heated at 900C under microwave conditions for 5 min. It was then concentrated, acidified with IN HCl solution and extracted with ethyl acetate (2X20 mL). The organic layers were combined, dried (MgSO4), filtered and concentrated. The residue was purified by Prep. HPLC to afford the desired product as a light yellow solid, (59 mg, 60% yield). MS m/z 552(MH+), Retention time: 3.850 min. lH NMR (300 MHz, MeOD) δ ppm 0.25 (m, 0.38 H) 1.14 - 2.22 (m, 11.62 H) 2.69 - 2.98 (m, 2 H) 3.02 (s, 2.28 H) 3.02 (s, 3.72 H) 3.41 (d, J=15.00 Hz, 0.62 H) 3.88 (s, 3 H) 4.01 (d, J=15.00 Hz, 0.38 H) 5.26 (d, J=15.00 Hz, 0.38 H) 5.45 (d, J=14.64 Hz, 0.62 H) 6.94 - 7.02 (m, 1 H) 7.13 (d, J=2.56 Hz, 0.38 H) 7.21 (d, J=2.20 Hz, 0.62 H) 7.26 (d, J=8.42 Hz, 0.62 H) 7.30 (d, J=8.78 Hz, 0.38 H) 7.53 (dd, J=8.42, 1.46 Hz, 0.62 H) 7.61 (dd, J=8.60, 1.65 Hz, 0.38 H) 7.85 (d, J=8.42 Hz, 0.62 H) 7.89 (d, J=8.42 Hz, 0.38 H) 8.10 (s, 0.38 H) 8.28 (d, ./=1.46 Hz, 0.62 H).
Intermediate 6
Figure imgf000030_0001
(+/-) Cycloprop[d]indolo[2,l-a][2]benzazepine-la(2H)-carbonyl azide, 8- cyclohexyl-5-[[[(dimethylamino)sulfonyl] amino] carbonyl] -1 ,12b-dihydro-l 1- methoxy-To a mixture of the acid (+/-) Cycloprop[d]indolo[2,l-a][2]benzazepine- la(2H)-carboxylic acid, 8-cyclohexyl-5- [[[(dimethylamino)sulfonyl]amino]carbonyl]-l,12b-dihydro-l l-methoxy (1 g, 1.81 mmol) in PhMe (18 ml) at r.t. under N2 was added triethylamine (0.38 ml, 2.73 mmol), followed by diphenylphosphoryl azide (DPPA) (0.59 ml, 2.73 mmol). The mixture was stirred at r.t. for 2.5 hr. The volatiles were then evaporated and the residue purified by Biotage flash chromatography (gradient elution, 0 to 60% EtOAc/Hexane) to give the product acyl azide (725.4 mg). Analytical HPLC method: Solvent A = 10% MeOH-90% H2O-0.1% TFA, Solvent B = 90% MeOH- 10%H2O-0.1% TFA, Start %B = 0, Final %B = 100, Gradient time = 2 min, Flow Rate = 5 ml/min, Column: Xterra MS C18 S7 3.0 x 50mm; LC/MS: (ES+) m/z (M+H)+ = 577.29, HPLC Rt = 2.015 min. Analytical HPLC method: Solvent A = 5% MeCN-95% H2O-IO mM NH4OAc, Solvent B = 95% MeCN-5%H2O-10 mM NH4OAc, Start %B = 0, Final %B = 100, Gradient time = 2 min, Flow Rate = 5 ml/min, Column: Phenomenex Lina C18 5um 3.0 x 50mm; LC/MS: (ES+) m/z (M+H)+ = 577.18, HPLC Rt = 1.633 min.
An alternative procedure: To 1.0 g of carboxylic acid in a 100 mL round bottom flask (RBF) equipped with a septa under nitrogen, was added 20 mL of dry dichloroethane (DCE). To this solution was then added 1.2 equivalents of
Diphenylphosphorylazide (DPPA) in one portion followed by 3 equivalents of triethylamine. The solution was stirred overnight at room temperature. The reaction progress was followed by an analytical Shimadzu LC/MS. The crude mixture was passed through a 24g SiliCycle-Isco™ silica gel cartridge with DCE to give acyl azide after solvent evacuation (50-65% yield). The acyl azide was found to be stable at room temperature in a vacuum desiccator for up to three months. To a 50 mL RBF was added 0.2mmol of acyl azide in 5.0 mL of dry toluene. The mixture was heated in an oil bath at 1200C for 15 minutes then quickly cooled to room temperature. To this mixture was then added 3.0 equivalents of amine, and the flask was returned to the oil bath and heated at 1200C for 60 minutes. The crude reaction mixture was then evacuated to near dryness, taken up in 1.2 mL of methanol and purified using a Shimadzu preparative HPLC employing methanol/water and 0.1% trifluoroacetic acid buffer with a Phenomenex Luna, C 18, 21 mm x 100 mm, 10 μm column at a gradient of 40-100% B (where A = 10% HPLC grade methanol/ 0.1% trifluoroacetic acid/ 90% HPLC grade water and B = 90% HPLC grade methanol/ 0.1% trifluoroacetic acid/ 10% HPLC grade water ) and a flow rate of 25 mL/min. over 10 minutes with a 5-10 minute hold, to give dimethylamino sulfamide ureas 3 as yellow amorphous solids (35-50% yield). Post-purification LC/MS data was obtained on a Shimadzu analytical LC /Micromass Platform LC (ESI+) at 220nm using the following set of conditions: Column I (Phenomenex lOμm C 18, 4.6 x 30mm),
Solvent system I (gradient of 0-100%B where A = 10% HPLC grade methanol/ 0.1% trifluoroacetic acid/ 90% HPLC grade water and B = 90% HPLC grade methanol/ 0.1% trifluoroacetic acid/ 10% HPLC grade water ), in 2 minutes with a 1 minute hold at a flow rate of 5 mL/minute.
Example 1
Figure imgf000032_0001
(+/-) Cycloprop [d] indolo [2, l-a][2]benzazepine-5-carboxamide, 8- cyclohexyl-N-[(dimethylamino)sulfonyl]-la-[[[(2R,6S)-2,6-dimethyl-4- morpholinyl] carbonyl] amino] - 1 , Ia, 2, 12b-tetrahydro-l 1-methoxy-. A mixture of the acyl azide (+/-) cycloprop[d]indolo[2,l-a] ' [2] benzazepine-la(2H) -carbonyl azide, 8- cyclohexyl-5-[[[(dimethylamino)sulfonyl] amino] carbonyl] -1 ,12b-dihydro-l 1- methoxy- (42.7 mg, 74 μmol) in MeCN (2 ml) was stirred at 50°C under N2 for 1 hr. The mixture was added a solution of cis-2,6-dimethylmorpholine (30 mg, 0.26 mmol) in MeCN (0.5 ml). The reaction mixture was stirred at 50°C for 1 hr. 15 min and then evaporated. The product urea C was isolated from the residue by preparative TLC (5% MeOH/CH2Cl2) Analytical HPLC method: Solvent A = 10% MeOH-90% H2O-0.1% TFA, Solvent B = 90% MeOH-10%H2O-0.1% TFA, Start %B = 0, Final %B = 100, Gradient time = 2 min, Flow Rate = 5 ml/min, Column: Xterra MS C18 S7 3.0 x 50mm; LC/MS: (ES+) m/z (M+H)+ = 664.45, HPLC Rt = 1.900 min. Analytical HPLC method: Solvent A = 5% MeCN-95% H2O-IO mM NH4OAc,
Solvent B = 95% MeCN-5%H2O-10 mM NH4OAc, Start %B = 0, Final %B = 100, Gradient time = 2 min, Flow Rate = 5 ml/min, Column: Phenomenex Lina Cl 8 5um 3.0 x 50mm; LC/MS: (ES+) m/z (M+H)+ = 664.38, HPLC Rt = 1.440 min. Example 2
Figure imgf000033_0001
(+/-) Cycloprop[d]indolo[2, 1-a] [2] benzazepine-5-carboxamide, 8- cyclohexyl-N-[(dimethylamino)sulfonyl]-l, Ia, 2, 12b-tetrahydro-l l-methoxy-la-[[(3- methoxy-l-azetidinyl)carbonyl] amino] -. A mixture of the acyl azide (+/-) cycloprop[d]indolo[2, l-a][2]benzazepine-la(2H)-carbonyl azide, 8-cyclohexyl-5- [[[(dimethylamino)sulfonyl] amino] carbonyl]-l,12b-dihydro-ll-methoxy- (98.3 mg, 0.17 mmol) in PhMe (2 ml) was stirred at 120° C under N2 for 1 hr 20 min. The mixture was added 3-methoxyazetidine hydrochloride (63.2 mg, 0.51 mmol) followed by EtsN (85 μl, 0.61 ml). The reaction mixture was stirred at 120°C for 30 min and then evaporated. The residue was purified by preparative TLC (5% MeOH/CH2Cl2) to give the product. Analytical HPLC method: Solvent A = 10% MeOH-90% H2O-0.1% TFA, Solvent B = 90% MeOH-10%H2O-0.1% TFA, Start %B = 0, Final %B = 100, Gradient time = 2 min, Flow Rate = 5 ml/min, Column: Xterra MS C18 S7 3.0 x 50mm; LC/MS: (ES+) m/z (M+H)+ = 636.46, HPLC Rt = 1.878 min. Analytical HPLC method: Solvent A = 5% MeCN-95% H2O-IO mM NH4OAc, Solvent B = 95% MeCN-5%H2O-10 mM NH4OAc, Start %B = 0, Final %B = 100, Gradient time = 2 min, Flow Rate = 5 ml/min, Column: Phenomenex Lina C18 5um 3.0 x 50mm; LC/MS: (ES+) m/z (M+H)+ = 636.31, HPLC Rt = 1.363 min. Example 3
Figure imgf000034_0001
(+/~) Cycloprop[d]indolo[2, 1-a] [2]benzazepine-5-carboxylic acid, 8- cyclohexyl- 1 , Ia, 2, 12b-tetrahydro -la-[(4-morphoHnylcarbonyl)amino]-, methyl ester. To a solution of cycloprop[<i]indolo[2,l-α][2]benzazepine-la,5(2H)- dicarboxylic acid, 8-cyclohexyl-l,12b-dihydro, 5-methyl ester (112 mg, 0.261 mmol) in CH2Cl2 (5 mL), triethyl amine (0.109 mL, 0.783 mmol) and DPPA (0.067 mL, 0.313 mmol) were added. The reaction mixture was stirred at rt. for overnight. Then the reaction mixture went through a Biotage column and washed with CH2Cl2. The fractions were collected and concentrated. The residue was then dissolved in toluene (4 mL) and morpholine (0.034 mL, 0.392 mmol) was added. The reaction mixture was heated at 1000C under microwave condition for 30 min. Toluene was evaporated and methanol was added. A white solid was collected as pure compound. (98 mg, 73% yield). MS m/z 514(MH+), Retention time: 3.875 min. IH NMR (500 MHz, DMSO-D6) δ ppm -0.06 (m, 0.62 H) 0.84 (m, 0.62 H) 1.07 - 2.15 (m, 10.76 H) 2.29 - 2.35 (m, 1 H) 2.66 - 2.78 (m, 0.38 H) 2.90 - 2.99 (m, 2 H) 3.10 - 3.19 (m, 1.62 H) 3.24 - 3.44 (m, 4 H) 3.53 - 3.60 (m, 1.62 H) 3.87 (s, 1.86 H) 3.89 (s, 1.14 H) 4.18 (d, J=14.65 Hz, 0.38 H) 4.77 (d, J=14.34 Hz, 0.38 H) 5.22 (d, J=15.26 Hz, 0.62 H) 6.59 (s, 0.62 H) 7.30 - 7.49 (m, 3.38 H) 7.52 - 7.58 (m, 1 H) 7.61 (dd, J=8.55, 1.53 Hz, 0.62 H) 7.66 (dd, J=8.39, 1.37 Hz, 0.38 H) 7.87 - 7.94 (m, 1.62 H) 8.21 (s, 0.38 H). Example 4
Figure imgf000035_0001
(+/~) Cycloprop[d]indolo[2, 1-a] [2]benzazepine-5-carboxylic acid, 8- cyclohexyl- 1 , Ia, 2, 12b-tetrahydro-la-[(4-morphoHnylcarbonyl)amino]-. To a mixture of (+/-) Cycloprop[d]indolo[2,l-a][2]benzazepine-5-carboxylic acid, 8- cyclohexyl-l,la,2,12b-tetrahydro -la-[(4-morpholinylcarbonyl)amino]-, methyl ester (40 mg, 0.078 mmol) and potassium trimethylsilanolate (50 mg, 0.389 mmol) in a round-bottomed flask, THF (5 mL) was added. The reaction mixture was stirred t rt. for 8 hr. Then it was acidified with IN HCl solution and extracted with ethyl acetate. The organic layer was separated, dried (MgSO4) and concentrated. The residue was then purified by Prep. HPLC column to give a light yellow solid as final compound. (15 mg, 44% yield). MS m/z 500(MH+), Retention time: 3.721 min. IH NMR (500 MHz, DMSO-D6) δ ppm -0.03 (m, 0.4 H) 0.84 (m, 0.4 H) 1.05 - 2.16 (m, 11 H) 2.24
- 2.34 (m, 1 H) 2.67 - 2.79 (m, 0.6 H) 2.84 - 2.98 (m, 2 H) 3.08 - 3.18 (m, 1.4 H) 3.21
- 3.64 (m, 5.6 H) 4.17 (d, J=14.34 Hz, 0.4 H) 4.75 (d, J=14.34 Hz, 0.4 H) 5.20 (d, J=14.95 Hz, 0.6 H) 6.57 (s, 0.6 H) 7.29 - 7.49 (m, 3.4 H) 7.51 - 7.58 (m, 1 H) 7.60 (d, J=8.55 Hz, 0.6 H) 7.65 (d, J=8.55 Hz, 0.4 H) 7.87 (m, 1 H) 7.91 (s, 0.6 H) 8.18 (s, 0.4 H). Example 5
Figure imgf000036_0001
(+/~) Cycloprop[d]indolo[2, 1-a] [2]benzazepine-5-carboxylic acid, 8- cyclohexyl- 1 , Ia, 2, 12b-tetrahydro-l l-methoxy-la-[(4-morphoHnylcarbonyl)amino]-, methyl ester. To a solution of cycloprop[<i]indolo[2,l-α][2]benzazepine-la,5(2H)- dicarboxylic acid, 8-cyclohexyl-l,12b-dihydro-l l-methoxy-, 5-methyl ester (105 mg, 0.228 mmol) in CH2Cl2 (5 mL), triethyl amine (0.095 mL, 0.684 mmol) and DPPA (0.059 mL, 0.274 mmol) were added. The reaction mixture was stirred at rt. for 2hr. Then the reaction mixture went through a Biotage column and washed with CH2Cl2. The fractions were collected and concentrated. The residue was then dissolved in toluene (4 mL) and morpholine (0.030 mL, 0.342 mmol) was added. The reaction mixture was heated at 1000C under microwave condition for 30 min. Toluene was evaporated and methanol was added. A white solid was collected as pure compound. (48 mg, 39% yield). MS m/z 544(MH+), Retention time: 3.891 min. IH NMR (500 MHz, DMSO-D6) δ ppm -0.03 (m, 0.38 H) 0.73 - 0.94 (m, 0.62 H) 1.05 - 2.16 (m, 11 H) 2.25 - 2.35 (m, 1 H) 2.67 - 2.80 (m, 0.38 H) 2.82 - 3.01 (m, 2 H) 3.09 - 3.20 (m, 1.38 H) 3.23 - 3.49 (m, 4.24 H) 3.51 - 3.67 (m, 1.62 H) 3.83 - 3.91 (m, 6 H) 4.19 (d, J=14.04 Hz, 0.38 H) 4.76 (d, J=14.65 Hz, 0.38 H) 5.22 (d, J=14.95 Hz, 0.62 H) 6.60 (s, 0.62 H) 6.99 (dd, J=8.55, 2.44 Hz, 0.38 H) 7.05 (dd, J=8.39, 2.59 Hz, 0.62 H) 7.11 (d, J=2.44 Hz, 0.62 H) 7.15 (d, J=2.44 Hz, 0.38 H) 7.24 - 7.34 (m, 1 H) 7.44 (s, 0.38 H) 7.61 (d, J=8.54 Hz, 0.62 H) 7.66 (d, J=8.55 Hz, 0.38 H) 7.88 (d, J=8.55 Hz, 1 H) 7.90 (s, 0.62 H) 8.20 (s, 0.38 H). Example 6
Figure imgf000037_0001
(+/~) Cycloprop[d]indolo[2, 1-a] [2]benzazepine-5-carboxylic acid, 8- cyclohexyl- 1 , Ia, 2, 12b-tetrahydro-l l-methoxy-la-[(4-morphoHnylcarbonyl)amino]-. To a mixture of (+/-) cycloprop[d]indolo[2,l-a][2]benzazepine-5-carboxylic acid, 8- cyclohexyl- 1 , 1 a,2, 12b-tetrahydro- 11 -methoxy- 1 a- [(4-morpholinylcarbonyl)amino] -, methyl ester (43 mg, 0.079 mmol) and potassium trimethylsilanolate (51 mg, 0.395 mmol) in a round-bottomed flask, THF (5 mL) was added. The reaction mixture was stirred at rt. overnight. Then it was acidified with IN HCl solution and extracted with ethyl acetate. The organic layer was separated, dried (MgSO4) and concentrated. The residue was then purified by Prep.HPLC column to give the product as a light yellow solid, (10 mg, 24% yield). MS m/z 530(MH+), Retention time: 3.733 min. IH NMR (300 MHz, DMSO-D6) δ ppm -0.04 (m, 0.38 H) 0.81 (m, 0.62 H) 0.96 - 2.15 (m, 11 H) 2.19 - 2.34 (m, 1 H) 2.66 - 2.78 (m, 0.38 H) 2.80 - 2.99 (m, 2 H) 3.01 - 3.61 (m, 7.24 H) 3.83 (s, 1.14 H) 3.85 (s, 1.86 H) 4.15 (d, J=14.27 Hz, 0.38 H) 4.71(d, J=13.90Hz, 0.38 H) 5.17 (d, J=15.37 Hz, 0.62 H) 6.58 (s, 0.62 H) 6.91 - 7.15 (m, 2 H) 7.20 - 7.32 (m, 1 H) 7.42 (s, 0.38 H) 7.57 (d, ./=8.41 Hz, 0.62 H) 7.62 (d, J=8.79 Hz, 0.38 H) 7.83 (d, J=8.42 Hz, 1 H) 7.87 (s, 0.62 H) 8.14 (s, 0.38 H) 12.52 (s, 1 H). Example 7
Figure imgf000038_0001
8-Cyclohexyl-N-(dimethylsulfamoyl)-ll-methoxy-la-((methyl(tetrahydro-2H- pyran-4-ylmethyl)carbamoyl)amino)-l, Ia, 2, 12b-tetrahydrocyclopropa[d]indolo[2, 1- a][2]benzazepine-5-carboxamide. 1H NMR (500 MHz, CD3OD) δ ppm: 0.17 (m, 0.25 H), 0.83 - 0.95 (m, 0.75 H), 1.22 (m, 4 H), 1.44 (m, 3 H), 1.68 (m, 1 H), 1.79 (m, 2 H), 1.93 (m, 1 H), 1.99 (m, 1 H), 2.09 (m, 2 H), 2.35 (m, 1 H), 2.62 (m, 2 H), 2.79 (m, 1 H), 2.85 (m, 1 H), 2.92 (m, 1 H), 2.99 (s, 6 H), 3.00 (s, 3 H), 3.17 (m, 2 H), 3.40 (m, 1 H), 3.74 (m, 1 H), 3.86 (s, 3 H), 3.88 (m, IH), 5.23 (d, J=14.65 Hz, 1 H), 6.96 (m, 1 H), 7.10 - 7.17 (m, 1 H), 7.26 - 7.30 (m, 1 H), 7.55 - 7.58 (m, 1 H), 7.84 - 7.90 (m, 1 H), 7.94 - 8.09 (m, 1 H). LC/MS: m/z 678.25 (MH+), Rf 2.07 min., 100% purity.
Example 8
Figure imgf000039_0001
8-Cyclohexyl-la-(((2-(dimethylamino)ethyl)(methyl)carbamoyl)amino)-N- (dimethylsulfamoyl)-l 1-methoxy-l, Ia, 2, 12b-tetrahydrocyclopropa[d] indolo[2, 1- a][2]benzazepine-5-carboxamide. 1H NMR (500 MHz, CD3OD) δ ppm: 0.08 (m, 0.20 H), 0.78 (m, 0.20 H), 1.20 (m, 0.8 H), 1.27 (m, 0.8 H), 1.40 (m, 2 H), 1.66 (m, 1 H), 1.76 (m, 1 H), 1.88 - 1.96 (m, 2 H), 2.05 (m, 2 H), 2.26 (m, 1 H), 2.37 (s, 3 H), 2.81 (s, 1 H), 2.92 (s, 5 H), 2.95 (s, 6 H), 3.30 (m, 3 H), 3.15 (m, 1 H), 3.33 (m, 1 H), 3.38 - 3.41 (m, 1 H), 3.46 (m, 1 H), 3.62 (m, 1 H), 3.82 (s, 3H), 5.18 (d, J=14.65 Hz, 1 H), 6.95 (dd, J= 8.55, 1.83 Hz, 1 H), 7.08 (d, J=I.83 Hz, 1 H), 7.27 (m, 1 H), 7.50 (dd, J=8.54, 1.53 Hz, 1 H), 7.81 - 7.86 (m, 2 H). LC/MS: m/z 651.19 (MH+), Rf 1.83 min., 98.6% purity.
Example 9
Figure imgf000040_0001
8-Cyclohexyl-N-(dimethylsulfamoyl)-ll-methoxy-la-(((2-((2- methoxyethyl)amino)-2-oxoethyl) (methyl)carbamoyl)amino)-l, Ia, 2, 12b- tetrahydrocyclopropa [djindolo [2,l-a][2]benzazepine-5-carboxamide. 1H NMR (500 MHz, CD3OD) δ ppm: 0.17 (t, J= 6.41 Hz, 0.25 H), 0.86 (m, 0.25 H), 1.22 (m, 0.75 H), 1.28 (m, 1 H), 1.35 (m, 0.75 H), 1.44 (m, 2 H), 1.70 (m, 1 H), 1.80 (m, 2 H), 1.93 (m, 1 H), 2.01 (m, 1 H), 2.10 (m, 2 H), 2.29 (m, 0.75 H), 2.35 (m, 0.25 H), 2.93 (m, 2 H), 2.99 (m, 1 H), 3.01 (s, 6 H), 3.33 (m, 2 H), 3.37 (m, 2 H), 3.44 (m, 3 H), 3.58 (d, J= 16.79 Hz, 1 H), 3.85 (s, 2.25 H), 3.88 (s, 0.75 H), 3.95 (m, 2 H), 4.03 (m, IH), 5.13 (d, J=14.65 Hz, 1 H), 6.92 (dd, J= 8.55, 1.83 Hz, 0.25 H), 6.98 (dd, J= 8.55, 1.83 Hz, 0.75 H), 7.13 (m, 1 H), 7.30 (m, 1 H), 7.56 (m, 1 H), 7.85 - 7.89 (m, 1 H), 7.97 (s, 0.75 H), 8.11 (s, 0.25 H). LC/MS: m/z 695.27 (MH+), Rf 2.05 min., 95.0% purity. Example 10
Figure imgf000041_0001
la-(((2-Amino-2-oxoethyl)(methyl)carbamoyl)amino)-8-cyclohexyl-N-
(dimethylsulfamoyl)-l 1-methoxy-l, Ia, 2, 12b-tetrahydrocyclopropa[d] indolo[2, 1- a] [2]benzazepine-5-carboxamide. 1H NMR (500 MHz, CD3OD) δ ppm: 0.16 (t, J= 6.40 Hz, 0.20 H), 0.88 (m, 0.20 H), 1.21 (t, J=6.40 Hz, 0.8 H), 1.28 (m, 1 H), 1.30 (m, 0.8 H), 1.45 (m, 2 H), 1.81 (m, 3 H), 1.97 (m, 2 H), 2.13 (m, 2 H), 2.36 (m, 1 H), 2.59 (s, 2 H), 2.86 - 2.95 (m, 1 H), 3.30 (s, 6 H), 3.42 (d, J= 14.95 Hz, 1 H), 3.61 (d, J= 17.09 Hz, 1 H), 3.86 (s, 2.40 H), 3.88 (s, 0.60 H), 3.97 (m, 1 H), 4.22 (m, IH), 5.10 (d, J=14.95 Hz, 1 H), 6.91 (dd, J= 8.55, 2.75 Hz, 0.20 H), 6.95 (dd, J= 8.55, 2.75 Hz, 0.80 H), 7.13 (m, 1 H), 7.28 (m, 1 H), 7.58 (m, 1 H), 7.85 - 7.90 (m, 1 H), 7.96 (m, 0.80 H), 8.11 (s, 0.20 H). LC/MS: m/z 637.16 (MH+), Rf 2.04 min., 95.0% purity.
Example 11
Figure imgf000042_0001
8-Cyclohexyl-N-(dimethylsulfamoyl)-ll-methoxy-la-((methyl(2-(4-methyl-l- piperazinyl)-2-oxoethyl)carbamoyl)amino)-l, Ia, 2, 12b-tetrahydrocyclopropa[d] indolo[2,l-a][2]benzazepine-5-carboxamide. 1H NMR (500 MHz, CD3OD) δ ppm: 0.15 (m, 0.20 H), 0.84 (m, 0.20 H), 1.22 (t, J=5.90 Hz, 0.8 H), 1.28 (m, 1 H), 1.34 (m, 0.8 H), 1.46 (m, 2 H), 1.70 (m, 1 H), 1.80 (m, 2 H), 1.92 - 1.99 (m, 2 H), 2.10 (m, 2 H), 2.29 (m, 0.8 H), 2.36 (m, 0.2 H), 2.48 (s, 2 H), 2.85 (m, 1 H), 2.92 - 3.03 (m, 12 H), 3.07 (m, 2 H), 3.44 (d, J= 14.95 Hz, 1 H), 3.54 (m, 2 H), 3.86 (m, 2.40 H), 3.88 (m, 0.60 H), 3.92 (m, 1 H), 4.07 (m, IH), 4.19 (m, IH), 4.29 (m, 1 H), 5.13 (d, J=14.95 Hz, 1 H), 6.93 (m, 0.20 H), 6.97 (m, 0.80 H), 7.12 (m, 1 H), 7.29 (m, 1 H), 7.55 - 7.61 (m, 1 H), 7.84 - 7.89 (m, 1 H), 8.02 (s, 0.80 H), 8.10 (s, 0.20 H). LC/MS: m/z 720.18 (MH+), R/1.87 min., 97.8% purity.
Examples 12 - 17 use the general methods below. Analytical HPLC and LC/MS were performed by using Shimadzu-VP instrument with UV detection at 220 nm and Waters Micromass. Biotage Horizon was used for flash chromatography as indicated. Example 12
Figure imgf000043_0001
1, 1-Dimethylethyl (8-cyclohexyl-5-
((((dimethylamino)sulfonyl)amino)carbonyl)-ll-(methyloxy)-l,12b- dihydrocyclopropa[d]indolo[2, 1-aJ ' [2] benzazepin-1 a(2H)-yl)carbamate. A mixture of the azide, 8-cyclohexy 1-5 -(((dimethylamino)sulfonyl)carbamoy I)-11-methoxy- 1 , 12&-dihydrocyclopropa[<i] indolo[2, 1 -a] [2]benzazepine- 1 α(2H)-carbonyl azide, (102 mg) in PhMe (2 ml) under N2 was stirred at 120°C for 1 hr. 35 min., cooled to r.t. and then concentrated. The residue was added tert-butanol (2 ml) and stirred at 120° C for 1 hr. 45 min., and then evaporated. The crude product was purified by flash chromatography (gradient elution 0-60% EtOAc/Ηexane) to give (BMS- 792039) 1,1-dimethylethyl (8-cyclohexyl-5- ((((dimethylamino)sulfonyl)amino)carbonyl)- 11 -(methyloxy)- 1,126- dihydrocyclopropa[<i]indolo[2,l-α][2]benzazepin-lα(2H)-yl)carbamate as a pale yellow solid. Analytical ΗPLC method: Solvent A = 10% MeOΗ-90% H2O-0.1% TFA, Solvent B = 90% MeOH- 10%H2O-0.1% TFA, Start %B = 0, Final %B = 100, Gradient time = 2 min, Flow Rate = 5 ml/min, Column: Xterra MS Cl 8 S7 3.0 x 50mm; LC/MS: (ES+) m/z (M+H)+ = 623.45, HPLC Rt = 1.957 min. Analytical HPLC method: Solvent A = 5% MeCN-95% H2O-IO mM NH4OAc, Solvent B = 95% MeCN-5%H2O-10 mM NH4OAc, Start %B = 0, Final %B = 100, Gradient time = 2 min, Flow Rate = 5 ml/min, Column: Phenomenex Lina Cl 8 5um 3.O x 50mm; LC/MS: (ES+) m/z (M+H)+ = 623.37, HPLC Rt = 1.628 min. Example 13
Figure imgf000044_0001
la-Amino-8-cyclohexyl-N-((dimethylamino)sulfonyl)-ll-(methyloxy)-
1, Ia, 2, 12b-tetrahydrocyclopropa[d]indolo[2, 1-a] [2] benzazepine-5-carboxamide. To 1, 1-dimethylethyl (8-cyclohexyl-5-((((dimethylamino)sulfonyl)amino)carbonyl)-l 1- (methyloxy)- 1 , 12&-dihydrocyclopropa[<i] indolo [2, 1 -a] [2]benzazepin- 1 α(2H)- yl)carbamate (75.3 mg) at r.t. under N2 was added a solution of HCl in 1,4-dioxane (0.5 ml, 4M). The mixture was stirred for 3 hr. 35 min., evaporated to give the hydrochloride salt of lα-amino-8-cyclohexyl-N-((dimethylamino)sulfonyl)-l l- (methyloxy)-l,lα,2,12έ-tetrahydrocyclopropa[(i]indolo[2,l-β][2]benzazepine-5- carboxamide, which was used without further purification. Analytical ΗPLC method: Solvent A = 10% MeOΗ-90% H2O-0.1% TFA, Solvent B = 90% MeOH- 10%H2O-0.1% TFA, Start %B = 0, Final %B = 100, Gradient time = 2 min, Flow Rate = 5 ml/min, Column: Xterra MS C18 S7 3.0 x 50mm; LC/MS: (ES+) m/z (M+H)+ = 523.39, HPLC Rt = 1.632 min.
Example 14
Figure imgf000044_0002
tert-Butyl ((laR,12bS)-8-cyclohexyl-5-((dimethylsulfamoyl)carbamoyl)-ll- methoxy-l,12b-dihydrocyclopropa[d]indolo[2,l-a][2]benzazepin-la(2H)- yl) carbamate. tert-Butyl ((lαR,12W>)-8-cyclohexyl-5- ((dimethylsulfamoyl)carbamoyl)- 11 -methoxy- 1,12b- dihydrocyclopropa[<i]indolo[2,l-β][2]benzazepin-lα(2H)-yl)carbamate was prepared from the corresponding chiral acid,
Figure imgf000045_0001
((dimethylsulfamoyl)carbamoyl)- 11 -methoxy- 1,12b- dihydrocyclopropa[<i]indolo[2,l-α][2]benzazepine-lα(2H)-carboxylic acid, in a similar manner as described above. Analytical HP LC method: Solvent A = 10% MeOH-90% H2O-0.1% TFA, Solvent B = 90% MeOH-10%H2O-0.1% TFA, Start %B = 0, Final %B = 100, Gradient time = 2 min, Flow Rate = 5 ml/min, Column: Xterra MS C18 S7 3.0 x 50mm; LC/MS: (ES+) m/z (M+H)+ = 623.16, HPLC Rt = 1.980 min. Analytical HPLC method: Solvent A = 5% MeCN-95% H2O-IO mM NH4OAc, Solvent B = 95% MeCN-5%H2O-10 mM NH4OAc, Start %B = 0, Final %B = 100, Gradient time = 2 min, Flow Rate = 5 ml/min, Column: Phenomenex Lina C18 5um 3.0 x 50mm; LC/MS: (ES+) m/z (M+H)+ = 623.48, HPLC Rt = 1.600 min. Average specific rotation = -56.55° (1.29 mg/ml in MeOH; Wavelength 589 nm; 100 mm cell).
Example 15
Figure imgf000045_0002
(laR,12bS)-la-Amino-8-cyclohexyl-N-(dimethylsulfamoyl)-ll-methoxy- 1,1 a, 2, 12b-tetrahydrocyclopropa[d]indolo[2, 1-a] [2] benzazepine-5-carboxamide. The hydrochloride salt of (lαR,12WΪ)-lα-amino-8-cyclohexyl-N- (dimethylsulfamoyl)- 11 -methoxy- 1 , 1 a,2, 12&-tetrahydrocyclopropa[<i]indolo[2, 1 - α][2]benzazepine-5-carboxamide was prepared from (BMS-807598) tert-butyl (( 1 ciR, 12&S)-8-cyclohexyl-5 -((dimethylsulfamoyl)carbamoyl)- 11 -methoxy- 1 , 12b- dihydrocyclopropa[<i]indolo[2,l-β][2]benzazepin-lα(2H)-yl)carbamate in a similar manner as described above. Analytical ΗPLC method: Solvent A = 10% MeOΗ-90% H2O-0.1% TFA, Solvent B = 90% MeOH-10%H2O-0.1% TFA, Start %B = 0, Final %B = 100, Gradient time = 2 min, Flow Rate = 5 ml/min, Column: Xterra MS C18 S7 3.0 x 50mm; LC/MS: (ES+) m/z (M+H)+ = 523.26, HPLC Rt = 1.640 min. Average specific rotation = -36.95° (1.19 mg/ml in MeOH; Wavelength 589 nm; 50 mm cell).
Example 16
Figure imgf000046_0001
tert-Butyl ((laS,12bR)-8-cyclohexyl-5-((dimethylsulfamoyl)carbamoyl)-ll- methoxy- 1, 12b-dihydrocyclopropa[d]indolo[2, 1-a] [2]benzazepin-la(2H)- yl) carbamate. tert-Butyl ((lαS,12fø?)-8-cyclohexyl-5- ((dimethylsulfamoyl)carbamoyl)- 11 -methoxy- 1,12b- dihydrocyclopropa[<i]indolo[2,l-α][2]benzazepin-lα(2H)-yl)carbamate was prepared from the corresponding chiral acid, (lα,S,12M)-8-cyclohexyl-5- ((dimethylsulfamoyl)carbamoyl)- 11 -methoxy- 1,12b- dihydrocyclopropa[<i]indolo[2,l-α][2]benzazepine-lα(2H)-carboxylic acid, in a similar manner as described above. Analytical ΗPLC method: Solvent A = 10% MeOΗ-90% H2O-0.1% TFA, Solvent B = 90% MeOH-10%H2O-0.1% TFA, Start %B = 0, Final %B = 100, Gradient time = 2 min, Flow Rate = 5 ml/min, Column: Xterra MS C18 S7 3.0 x 50mm; LC/MS: (ES+) m/z (M+H)+ = 623.10, HPLC Rt = 1.935 min. Analytical HPLC method: Solvent A = 5% MeCN-95% H2O-IO mM NH4OAc, Solvent B = 95% MeCN-5%H2O-10 mM NH4OAc, Start %B = 0, Final %B = 100, Gradient time = 2 min, Flow Rate = 5 ml/min, Column: Phenomenex Lina C18 5um 3.0 x 50mm; LC/MS: (ES-) m/z (M-H)+ = 621.26, HPLC Rt = 1.653 min. Average specific rotation = +56.07° (2.57 mg/ml in MeOH; Wavelength 589 nm; 50 mm cell).
Example 17
Figure imgf000047_0001
(laS,12bR)-la-Amino-8-cyclohexyl-N-(dimethylsulfamoyl)-ll-methoxy- 1, Ia, 2, 12b-tetrahydrocyclopropa[d]indolo[2, 1-a] [2] benzazepine-5-carboxamide. The hydrochloride salt of (lα«S,12/?R)-la-Amino-8-cyclohexyl-N- (dimethylsulfamoyl)- 11 -methoxy- 1 , 1 a,2, 12&-tetrahydrocyclopropa[<i]indolo[2, 1 - α][2]benzazepine-5-carboxamide was prepared from tert-butyl ((laS,l2bR)-8- cyclohexyl-5 -((dimethylsulfamoyl)carbamoy I)- 11 -methoxy- 1,12b- dihydrocyclopropa[<i]indolo[2,l-α][2]benzazepin-lα(2H)-yl)carbamate in a similar manner as described above. Analytical ΗPLC method: Solvent A = 10% MeOΗ-90% H2O-0.1% TFA, Solvent B = 90% MeOH-10%H2O-0.1% TFA, Start %B = 0, Final %B = 100, Gradient time = 2 min, Flow Rate = 5 ml/min, Column: Xterra MS C18 S7 3.0 x 50mm; LC/MS: (ES+) m/z (M+H)+ = 523.32, HPLC Rt = 1.603 min. Example 18
Figure imgf000048_0001
8-Cyclohexyl-N-(dimethylsulfamoyl)-ll-methoxy-la-(methylamino)-
1, Ia, 2, 12b-tetrahydrocyclopropa[d]indolo[2, 1-a] [2] benzazepine-5-carboxamide. The hydrochloride salt of 8-cyclohexyl-N-(dimethylsulfamoyl)-l 1-methoxy-lα- (methylamino)- \,\a,2,\ 2&-tetrahydrocyclopropa[<i]indolo[2, 1 -a] [2]benzazepine-5- carboxamide was prepared from 1,1-dimethylethyl (8-cyclohexyl-5- ((((dimethylamino)sulfonyl)amino)carbonyl)-l l-(methyloxy)-l,12έ- dihydrocyclopropa[<i]indolo[2,l-β][2]benzazepin-lα(2H)-yl)carbamate in two steps (MeI, ΝaΗ, DMF, r.t. followed by HCl, 1,4-dioxane, r.t.) Analytical ΗPLC method: Solvent A = 10% MeOΗ-90% H2O-0.1% TFA, Solvent B = 90% MeOH- 10%H2O-0.1% TFA, Start %B = 0, Final %B = 100, Gradient time = 2 min, Flow Rate = 5 ml/min, Column: Xterra MS C18 S7 3.0 x 50mm; LC/MS: (ES+) m/z (M+H)+ = 537.12, HPLC Rt = 1.627 min. Analytical HPLC method: Solvent A = 5% MeCΝ-95% H2O-IO mM NH4OAc, Solvent B = 95% MeCN-5%H2O-10 mM NH4OAc, Start %B = 0, Final %B = 100, Gradient time = 2 min, Flow Rate = 5 ml/min, Column: Phenomenex Luna Cl 8 5um 3.O x 50mm; LC/MS: (ES+) m/z (M+H)+ = 537.55, HPLC Rt = 1.392 min.
Example 19
Figure imgf000049_0001
8-Cyclohexyl-N-((dimethylamino)sulfonyl)-ll-methoxy-la-((((3R, 5S)-3, 4, 5- trimethyl-l-piperazinyl)carbonyl)amino)-l, Ia, 2, 12b- tetrahydrocyclopropa[d]indolo[2, 1-a] [2] benzazepine-5-carboxamide. A mixture of the acyl azide, 8-cyclohexyl-5-(((dimethylamino)sulfonyl)carbamoyl)-l l-methoxy- l,12/?-dihydrocyclopropa[(i]indolo[2,l-«][2]benzazepine-la(2H)-carbonyl azide, (98 mg, 0.17 mmol) in PhMe (2 ml) was stirred at 120°C under N2 for 1 hr 30 min. The mixture was added 1,2,6-trimethylpiperazine bistrifluoroacetic acid salt (182 mg, 0.51 mmol) followed by EtsN (170 μl, 1.22 mmol). The reaction mixture was stirred at 120°C for 30 min and then evaporated. The residue was purified by Shimadzu-VP preparative reverse phase HP LC with separation method: Solvent A = 10% MeOH-90% H2O-0.1% TFA, Solvent B = 90% MeOH-10%H2O-0.1% TFA, Start %B = 0, Final %B = 100, Gradient time = 6min, Flow Rate = 30mL/min, Column: Xterra Prep MS C18 5u 30x50mm, Fraction Collection: 6.31 - 6.70 min. (UV detection at 220 nm) to give the trifluoroacetic acid salt of the compound of Example 19, 8-cyclohexyl-N-((dimethylamino)sulfonyl)-l l-methoxy-lα-((((3R,55)-3,4,5- trimethyl-l-piperazinyl)carbonyl)amino)-l,lα,2,12ό- tetrahydrocyclopropa[<i]indolo[2,l-α][2]benzazepine-5-carboxamide, (56.3 mg) as a light yellow solid;. Analytical HPLC method: Solvent A = 10% MeOH-90% H2O-0.1% TFA, Solvent B = 90% MeOH-10%H2O-0.1% TFA, Start %B = 0, Final %B = 100, Gradient time = 2 min, Flow Rate = 5 ml/min, Column: Xterra MS C18 S7 3.0 x 50mm; LC/MS: (ES+) m/z (M+H)+ = 677.56, HPLC Rt = 1.703 min. Analytical HPLC method: Solvent A = 5% MeCN-95% H2O-IO mM NH4OAc, Solvent B = 95% MeCN-5%H2O-10 mM NH4OAc, Start %B = 0, Final %B = 100, Gradient time = 2 min, Flow Rate = 5 ml/min, Column: Phenomenex Luna C18 5um 3.0 x 50mm; LC/MS: (ES+) m/z (M+H)+ = 677.37, HPLC Rt = 1.247 min.

Claims

CLAIMS We claim:
1. A compound of formula I
Figure imgf000051_0001
where:
R1 is CO2R5 or CONR6R7;
R2 is N(R1O)(RU);
R3 is hydrogen, halo, alkyl, alkenyl, hydroxy, benzyloxy, alkoxy, or haloalkoxy;
R4 is cycloalkyl;
R5 is hydrogen or alkyl;
R6 is hydrogen, alkyl, alkylSO2, cycloalkylSO2, haloalkylSO2, (R7)(R8)NSO2, or (R9)SO2;
R7 is hydrogen or alkyl;
R8 is hydrogen or alkyl; R9 is azetidinyl, pyrrolidinyl, piperidinyl, piperazinyl, morpholinyl, thiomorpholinyl, homopiperidinyl, or homomorpholinyl, and is substituted with 0-3 substituents selected from alkyl and alkoxy;
R10 is hydrogen, alkyl, alkoxyCO, or (R12)(R13)NCO;
R » 11 is hydrogen or alkyl;
R12 is hydrogen, alkyl, alkoxyalkyl, aminoalkyl, (alkylamino)alkyl, (dialkylamino)alkyl, (CO2R14)alkyl, (CON(R15)(R16))alkyl, or (tetrahydropyranyl)alkyl;
R13 is hydrogen alkyl, alkoxyalkyl, aminoalkyl, (alkylamino)alkyl, or (dialkylamino)alkyl;
or (R12)(R13)N taken together is azetidinyl, pyrrolidinyl, piperidinyl, piperazinyl, morpholinyl, thiomorpholinyl, homopiperidinyl, or homomorpholinyl and is substituted with 0-3 substituents selected from alkyl and alkoxy;
R , 14 is hydrogen or alkyl;
R is hydrogen or alkyl; and
R , 16 is hydrogen or alkyl; or
N(R15)(R16) taken together is azetidinyl, pyrrolidinyl, piperidinyl, piperazinyl, morpholinyl, thiomorpholinyl, homopiperidinyl, or homomorpholinyl and is substituted with 0-3 substituents selected from alkyl and alkoxy;
or a pharmaceutically acceptable salt thereof.
2. A compound of claim 1 where R1 is CONHR6 and R6 is alkylSO2, cycloalkylSO2, haloalkylSO2, (R7)(R8)NSO2, or (R9)SO2.
3. A compound of claim 1 where R3 is hydrogen.
4. A compound of claim 1 where R3 methoxy.
5. A compound of claim 1 where R4 is cyclohexyl.
6. A compound of claim 1 where R6 is (R7)(R8)NSO2 or (R9)SO2.
7. A compound of claim 1 where R10 is (R12)(R13)NCO.
8. A compound of claim 1 where R12 is alkoxyalkyl, (dialkylamino)alkyl, (CON(R15)(R16))alkyl, or (tetrahydropyranyl)alkyl.
9. A compound of claim 1 where (R12)(R13)N taken together is azetidinyl, pyrrolidinyl, piperidinyl, piperazinyl, morpholinyl, thiomorpholinyl, homopiperidinyl, or homomorpholinyl and is substituted with 0-3 substituents selected from alkyl and alkoxy.
10. A compound of claim 1 according to the following stereochemistry.
Figure imgf000053_0001
11. A compound of claim 1 according to the following stereochemistry.
Figure imgf000053_0002
2. A compound of claim 1 selected from the group consisting of
Figure imgf000054_0001
Figure imgf000055_0001
or a pharmaceutically acceptable salt thereof.
13. A composition comprising a compound of claim 1, or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable carrier.
14. The composition of claim 13 further comprising at least one additional compound having therapeutic benefits for HCV wherein the compound is selected from the group consisting of interferons, cyclosporins, interleukins, HCV metalloprotease inhibitors, HCV serine protease inhibitors, HCV polymerase inhibitors, HCV helicase inhibitors, HCV NS4B protein inhibitors, HCV entry inhibitors, HCV assembly inhibitors, HCV egress inhibitors, HCV NS5A protein inhibitors, HCV NS5B protein inhibitors, and HCV replicon inhibitors.
15. A method of treating hepatitis C infection comprising administering a therapeutically effective amount of a compound of claim 1, or a pharmaceutically acceptable salt thereof, to a patient.
16. The method of claim 15 further comprising administering at least one additional compound having therapeutic benefits for HCV wherein the compound is selected from the group consisting of interferons, cyclosporins, interleukins, HCV metalloprotease inhibitors, HCV serine protease inhibitors, HCV polymerase inhibitors, HCV helicase inhibitors, HCV NS4B protein inhibitors, HCV entry inhibitors, HCV assembly inhibitors, HCV egress inhibitors, HCV NS5A protein inhibitors, HCV NS5B protein inhibitors, and HCV replicon inhibitors.
PCT/US2007/070173 2006-06-08 2007-06-01 Cyclopropyl fused indolobenzazepine hcv inhibitors WO2007143521A1 (en)

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US80420306P 2006-06-08 2006-06-08
US60/804,203 2006-06-08
US89476407P 2007-03-14 2007-03-14
US60/894,764 2007-03-14

Publications (1)

Publication Number Publication Date
WO2007143521A1 true WO2007143521A1 (en) 2007-12-13

Family

ID=38577257

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2007/070173 WO2007143521A1 (en) 2006-06-08 2007-06-01 Cyclopropyl fused indolobenzazepine hcv inhibitors

Country Status (2)

Country Link
US (1) US7452876B2 (en)
WO (1) WO2007143521A1 (en)

Cited By (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2008097796A1 (en) * 2007-02-02 2008-08-14 Bristol-Myers Squibb Company Indolobenzazepine derivatives for the treatment of hepatitis c
WO2008112863A1 (en) * 2007-03-15 2008-09-18 Bristol-Myers Squibb Company Compounds for the treatment of hepatitis c
WO2008112841A1 (en) * 2007-03-14 2008-09-18 Bristol-Myers Squibb Company Compounds for the treatment of hepatitis c
WO2008112851A1 (en) * 2007-03-14 2008-09-18 Bristol-Myers Squibb Company Cyclopropyl fused indolobenzazepine hcv inhibitors
WO2009076747A1 (en) 2007-12-19 2009-06-25 Boehringer Ingelheim International Gmbh Viral polymerase inhibitors
US7659263B2 (en) 2004-11-12 2010-02-09 Japan Tobacco Inc. Thienopyrrole compound and use thereof as HCV polymerase inhibitor
WO2010080874A1 (en) 2009-01-07 2010-07-15 Scynexis, Inc. Cyclosporine derivative for use in the treatment of hcv and hiv infection
WO2010099159A1 (en) 2009-02-26 2010-09-02 Bristol-Myers Squibb Company Cyclopropyl fused indolobenzazepine hcv ns5b inhibitors
US7977331B1 (en) 2004-02-24 2011-07-12 Japan Tobacco Inc. Tetracyclic fused heterocyclic compound and use thereof as HCV polymerase inhibitor
EP2494991A1 (en) 2007-05-04 2012-09-05 Vertex Pharmaceuticals Incorporated Combination therapy for the treatment of HCV infection

Families Citing this family (23)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2005049622A1 (en) * 2003-11-19 2005-06-02 Japan Tobacco Inc. 5-5-membered fused heterocyclic compound and use thereof as hcv polymerase inhibitor
US7521443B2 (en) * 2006-05-17 2009-04-21 Bristol-Myers Squibb Company Cyclopropyl fused indolobenzazepine HCV NS5B inhibitors
US7541351B2 (en) * 2007-01-11 2009-06-02 Bristol-Myers Squibb Company Compounds for the treatment of hepatitis C
US7517872B2 (en) * 2007-02-22 2009-04-14 Bristol-Myers Squibb Company Compounds for the treatment of hepatitis C
US7998951B2 (en) * 2007-03-05 2011-08-16 Bristol-Myers Squibb Company HCV NS5B inhibitors
US7541353B2 (en) * 2007-03-14 2009-06-02 Bristol-Myers Squibb Company Compounds for the treatment of hepatitis C
US7547690B2 (en) * 2007-03-14 2009-06-16 Bristol-Myers Squibb Company Compounds for the treatment of Hepatitis C
US7642251B2 (en) * 2007-08-09 2010-01-05 Bristol-Myers Squibb Company Compounds for the treatment of hepatitis C
US7652004B2 (en) * 2007-08-09 2010-01-26 Bristol-Myers Squibb Company Compounds for the treatment of hepatitis C
US8143243B2 (en) * 2007-08-09 2012-03-27 Bristol-Myers Squibb Company Compounds for the treatment of hepatitis C
US8129367B2 (en) * 2007-11-21 2012-03-06 Bristol-Myers Squibb Company Compounds for the treatment of Hepatitis C
US8124601B2 (en) 2007-11-21 2012-02-28 Bristol-Myers Squibb Company Compounds for the treatment of Hepatitis C
JP2011515486A (en) * 2008-03-27 2011-05-19 ブリストル−マイヤーズ スクイブ カンパニー Compound for the treatment of hepatitis C
JP2011515484A (en) * 2008-03-27 2011-05-19 ブリストル−マイヤーズ スクイブ カンパニー Pyrrolidine-fused indolobenzazepine HCVNS5B inhibitor
EP2268643B1 (en) * 2008-03-27 2014-08-06 Bristol-Myers Squibb Company Dioxolane and dioxolanone fused indolobenzadiazepine hcv ns5b inhibitors
ATE531373T1 (en) 2008-03-27 2011-11-15 Bristol Myers Squibb Co AROMATIC HETEROCYCLIC CONDENSED INDOLOBENZADIAZEPINE HCV NS5B INHIBITORS
US8133884B2 (en) * 2008-05-06 2012-03-13 Bristol-Myers Squibb Company Compounds for the treatment of hepatitis C
UY32099A (en) 2008-09-11 2010-04-30 Enanta Pharm Inc HEPATITIS C SERINA PROTEASAS MACROCYCLIC INHIBITORS
CN106986862A (en) 2010-12-22 2017-07-28 Abbvie 公司 Hepatitis c inhibitor and application thereof
WO2012092411A2 (en) 2010-12-30 2012-07-05 Enanta Pharmaceuticals, Inc. Phenanthridine macrocyclic hepatitis c serine protease inhibitors
MX2013007677A (en) 2010-12-30 2013-07-30 Abbvie Inc Macrocyclic hepatitis c serine protease inhibitors.
US10201584B1 (en) 2011-05-17 2019-02-12 Abbvie Inc. Compositions and methods for treating HCV
EP3089757A1 (en) 2014-01-03 2016-11-09 AbbVie Inc. Solid antiviral dosage forms

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2006020082A1 (en) * 2004-08-09 2006-02-23 Bristol-Myers Squibb Company Inhibitors of hcv replication
WO2007033032A1 (en) * 2005-09-12 2007-03-22 Bristol-Myers Squibb Company Cyclopropyl fused indolobenzazepine hcv ns5b inhibitors
WO2007033175A1 (en) * 2005-09-13 2007-03-22 Bristol-Myers Squibb Company Indolobenzazepine hcv ns5b inhibitors

Family Cites Families (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
ATE428714T1 (en) 2004-02-24 2009-05-15 Japan Tobacco Inc CONDENSED HETEROTETRACYCLIC COMPOUNDS AND THEIR USE AS HCV POLYMERASE INHIBITORS
US7348425B2 (en) 2004-08-09 2008-03-25 Bristol-Myers Squibb Company Inhibitors of HCV replication
CA2584179C (en) 2004-10-13 2013-01-29 Wolfgang Staehle N,n'-diphenylurea derivatives which are suitable as kinase inhibitors
AU2005298412B2 (en) 2004-10-26 2011-06-09 Istituto Di Ricerche Di Biologia Molecolare P Angeletti Spa Tetracyclic indole derivatives as antiviral agents
GB0518390D0 (en) 2005-09-09 2005-10-19 Angeletti P Ist Richerche Bio Therapeutic compounds
US7456165B2 (en) 2006-02-08 2008-11-25 Bristol-Myers Squibb Company HCV NS5B inhibitors

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2006020082A1 (en) * 2004-08-09 2006-02-23 Bristol-Myers Squibb Company Inhibitors of hcv replication
WO2007033032A1 (en) * 2005-09-12 2007-03-22 Bristol-Myers Squibb Company Cyclopropyl fused indolobenzazepine hcv ns5b inhibitors
WO2007033175A1 (en) * 2005-09-13 2007-03-22 Bristol-Myers Squibb Company Indolobenzazepine hcv ns5b inhibitors

Cited By (12)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7977331B1 (en) 2004-02-24 2011-07-12 Japan Tobacco Inc. Tetracyclic fused heterocyclic compound and use thereof as HCV polymerase inhibitor
US7659263B2 (en) 2004-11-12 2010-02-09 Japan Tobacco Inc. Thienopyrrole compound and use thereof as HCV polymerase inhibitor
WO2008097796A1 (en) * 2007-02-02 2008-08-14 Bristol-Myers Squibb Company Indolobenzazepine derivatives for the treatment of hepatitis c
CN101646674B (en) * 2007-02-02 2013-05-29 百时美施贵宝公司 Indolobenzazepine derivatives for the treatment of hepatitis c
WO2008112841A1 (en) * 2007-03-14 2008-09-18 Bristol-Myers Squibb Company Compounds for the treatment of hepatitis c
WO2008112851A1 (en) * 2007-03-14 2008-09-18 Bristol-Myers Squibb Company Cyclopropyl fused indolobenzazepine hcv inhibitors
CN101679442B (en) * 2007-03-14 2013-02-20 百时美施贵宝公司 Compounds for the treatment of hepatitis c
WO2008112863A1 (en) * 2007-03-15 2008-09-18 Bristol-Myers Squibb Company Compounds for the treatment of hepatitis c
EP2494991A1 (en) 2007-05-04 2012-09-05 Vertex Pharmaceuticals Incorporated Combination therapy for the treatment of HCV infection
WO2009076747A1 (en) 2007-12-19 2009-06-25 Boehringer Ingelheim International Gmbh Viral polymerase inhibitors
WO2010080874A1 (en) 2009-01-07 2010-07-15 Scynexis, Inc. Cyclosporine derivative for use in the treatment of hcv and hiv infection
WO2010099159A1 (en) 2009-02-26 2010-09-02 Bristol-Myers Squibb Company Cyclopropyl fused indolobenzazepine hcv ns5b inhibitors

Also Published As

Publication number Publication date
US7452876B2 (en) 2008-11-18
US20070287694A1 (en) 2007-12-13

Similar Documents

Publication Publication Date Title
WO2007143521A1 (en) Cyclopropyl fused indolobenzazepine hcv inhibitors
US7521441B2 (en) Cyclopropyl fused indolobenzazepine HCV NS5B inhibitors
EP2029606B1 (en) Cyclopropyl fused indolobenzazepine hcv ns5b inhibitors
JP5306230B2 (en) Compound for the treatment of hepatitis C
JP5343011B2 (en) Compounds for the treatment of hepatitis C
EP2209789B1 (en) Cyclopropyl fused indolobenzazepine hcv ns5b inhibitors
WO2007140200A2 (en) Cyclopropyl fused indolobenzazepine hcv ns5b inhibitors
JP5301473B2 (en) Compounds for the treatment of hepatitis C
WO2008112851A1 (en) Cyclopropyl fused indolobenzazepine hcv inhibitors
WO2008103637A1 (en) Compounds for the treatment of hepatitis c
WO2009067481A1 (en) Compounds for the treatment of hepatitis c
WO2008109584A1 (en) Hcv ns5b inhibitors
WO2009067392A1 (en) Cyclopropyl fused indolobenzazepine derivatives for the treatment of hepatitis c
EP2114947B1 (en) Indolobenzazepine derivatives for the treatment of hepatitis c
EP2280978A1 (en) Pyrrolidine fused indolobenzadiazepine hcv ns5b inhibitors
WO2009120650A1 (en) Aromatic heterocyclic fused indolobenzadiazepine hcv ns5b inhibitors
EP2268643B1 (en) Dioxolane and dioxolanone fused indolobenzadiazepine hcv ns5b inhibitors
AU2009228214B2 (en) Dioxolane and dioxolanone fused indolobenzadiazepine HCV NS5B inhibitors
AU2009228325B2 (en) Pyrrolidine fused indolobenzadiazepine HCV NS5B inhibitors

Legal Events

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

Ref document number: 07815077

Country of ref document: EP

Kind code of ref document: A1

122 Ep: pct application non-entry in european phase

Ref document number: 07815077

Country of ref document: EP

Kind code of ref document: A1