WO2009058919A1 - Ccr5 antagonists as therapeutic agents - Google Patents

Ccr5 antagonists as therapeutic agents Download PDF

Info

Publication number
WO2009058919A1
WO2009058919A1 PCT/US2008/081662 US2008081662W WO2009058919A1 WO 2009058919 A1 WO2009058919 A1 WO 2009058919A1 US 2008081662 W US2008081662 W US 2008081662W WO 2009058919 A1 WO2009058919 A1 WO 2009058919A1
Authority
WO
WIPO (PCT)
Prior art keywords
amino
methyl
phenyl
oxy
carbonyl
Prior art date
Application number
PCT/US2008/081662
Other languages
French (fr)
Inventor
Wieslaw Mieczyslaw Kazmierski
Pek Yoke Chong
Maosheng Duan
Original Assignee
Smithkline Beecham Corporation
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 Smithkline Beecham Corporation filed Critical Smithkline Beecham Corporation
Publication of WO2009058919A1 publication Critical patent/WO2009058919A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings
    • C07D401/06Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings linked by a carbon chain containing only aliphatic carbon atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D409/00Heterocyclic compounds containing two or more hetero rings, at least one ring having sulfur atoms as the only ring hetero atoms
    • C07D409/14Heterocyclic compounds containing two or more hetero rings, at least one ring having sulfur atoms as the only ring hetero atoms containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D413/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms
    • C07D413/14Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing three or more hetero rings

Definitions

  • the human immunodeficiency virus is the causative agent for acquired immunodeficiency syndrome ("AIDS”), a disease characterized by the destruction of the immune system, particularly of CD4 + T-cells, with attendant susceptibility to opportunistic infections, and its precursor Al DS-related complex (“ARC”), a syndrome characterized by symptoms such as persistent generalized lymphadenopathy, fever and weight loss.
  • AIDS acquired immunodeficiency syndrome
  • ARC Al DS-related complex
  • HIV requires a co-receptor for entry into target cells.
  • the chemokine receptors function together with CD4 as co-receptors for HIV.
  • the chemokine receptors CXCR4 and CCR5 have been identified as the main co- receptors for HIV-1.
  • CCR5 acts as a major co-receptor for fusion and entry of macrophage-tropic HIV into host cells. These chemokine receptors are thought to play an essential role in the establishment and dissemination of an HIV infection. Therefore, CCR5 antagonists are thought to be useful as therapeutic agents active against HIV.
  • Nitrogen-containing heterocyclic compounds that are CCR5 a ⁇ tangonists are disclosed in WO2006030925, WO2004080966, and WO2001087839. However, such compounds can demonstrate limited oral bioavailability or unfavorable cardiovascular profile.
  • the present invention features compounds that are useful in the inhibition of HIV replication, the prevention of infection by HlV, the treatment of infection by HIV and in the treatment of AIDS and/or ARC, either as pharmaceutically acceptable salts or pharmaceutical composition ingredients.
  • the present invention further features methods of treating AIDS, methods of preventing infection by HIV, and methods of treating infection by HIV as monotherapy or in combination with other a ⁇ tivirals, anti- infectives, immunomodulators, antibiotics or vaccines.
  • the present invention also features pharmaceutical compositions, comprising the above-mentioned compounds that are suitable for the prevention or treatment of CCR5-related diseases and conditions.
  • the present invention further features processes for making the above- mentioned compounds.
  • the present invention features a compound selected from the group consisting of 2,4-Difluoro-5-( ⁇ [(1- ⁇ [2-methyl-6-( ⁇ 4-[(methylsulfonyl)amino]phenyl ⁇ oxy)-3- pyridinyl]methyl ⁇ -4-piperidinyl)(phe ⁇ yl)amino]carbonyl ⁇ amino)benzamide; 2,4-Difluoro-5-( ⁇ [(3-fluorophenyl)(1- ⁇ [2-methyl-6-( ⁇ 4- [(rnethylsulfonyl)ami ⁇ o]phenyl ⁇ oxy)-3-pyridinyl]methyl ⁇ -4- piperidinyl)amino]carbonyl ⁇ amino)benzamide;
  • pharmaceutically effective amount refers to an amount of a compound of the invention that is effective in treating a CCR5-related disease, for example a virus infection, for example an HIV infection, in a patient either as monotherapy or in combination with other agents.
  • treatment refers to the
  • asymptomatic patient such as a patient in whom a viral infection has become latent.
  • prophylaxis refers to preventing a disease or condition or preventing the occurrence of symptoms of such a disease or condition, in a patient.
  • the term “patient” refers to a mammal, including a human.
  • pharmaceutically acceptable carrier refers to a carrier that may be administered to a patient, together with a compound of this invention, and which does not destroy the pharmacological activity thereof and is nontoxic when administered in doses sufficient to deliver a therapeutic amount of the therapeutic agent.
  • Pharmaceutically acceptable salts of the compounds according to the invention include those derived from pharmaceutically acceptable inorganic and organic acids and bases.
  • suitable acids include hydrochloric, hydrobromic, sulfuric, nitric, perchloric, fumaric, glutamic, maleic, mandelic, phosphoric, glycollic, lactic, salicyclic, succinic, toluene-p-sulfonic, tartaric, acetic, citric, orotic, toluenesulfonic, methanesulfonic, ethanesulfonic, formic, benzoic, malonic, naphthalene-2-sulfonic and benzenesulfonic acids.
  • Other acids such as oxalic, while not in themselves pharmaceutically acceptable, may be employed in the preparation of salts useful as intermediates in obtaining the compounds of the invention and their pharmaceutically acceptable acid addition salts.
  • Salts derived from appropriate bases include alkali metal (e.g. sodium), alkaline earth metal (e.g., magnesium), ammonium, NW 4 + (wherein W is C 1 J, alkyl) and other amine salts.
  • Physiologically acceptable salts of a hydrogen atom or an amino group include salts of organic carboxylic acids such as acetic, lactic, tartaric, malic, isethionic, lactobionic and succinic acids; organic sulfonic acids such as methanesulfonic, ethanesulfonic, benzenesulfonic and p-toluenesulfonic acids and inorganic acids such as hydrochloric, sulfuric, phosphoric and sulfamic acids.
  • Physiologically acceptable salts of a compound with a hydroxy group include the anion of said compound in combination with a suitable cation such as Na + , NH 4 + , and NW 4 + (wherein W is a C ⁇ alkyl group)
  • Any reference to any of the above compounds also includes a reference to a pharmaceutically acceptable salt thereof
  • Salts of the compounds of the present invention may be made by methods known to a person skilled in the art For example, treatment of a compound of the present invention with an appropriate base or acid in an appropriate solvent will yield the corresponding salt
  • the present invention features compounds according to the invention for use in medical therapy, for example for the treatment or prophylaxis of viral infections such as an HIV infections and associated conditions
  • viral infections such as an HIV infections and associated conditions
  • Reference herein to treatment extends to prophylaxis as well as the treatment of established infections, symptoms, and associated clinical conditions such as AIDS related complex (ARC), Kaposi's sarcoma, and AIDS dementia
  • the present invention features use of the compounds of the present invention in the manufacture of a medicament for the treatment or prophylaxis of a CCR5- related disease or condition, for example, a viral infection, for example, an HIV infection
  • the present invention provides a method for the treatment or prevention of the symptoms or effects of a viral infection in an infected animal, for example, a mammal including a human, which comprises treating said animal with a pharmaceutically effective amount of a compound according to the invention
  • the viral infection is a retroviral infection, in particular an HIV infection
  • a further aspect of the invention includes a method for the treatment or prevention of the symptoms or effects of an HBV infection
  • the compounds according to the invention may also be used in adjuvant therapy in the treatment of HIV infections or HIV-associated symptoms or effects, for example Kaposi's sarcoma.
  • the compounds of the present invention may also be used in the prevention or treatment of other CCR5-related diseases and conditions, including neuropathic pain, multiple sclerosis, rheumatoid arthritis, autoimmune diabetes, chronic implant rejection, asthma, rheumatoid arthritis, Crohns Disease, inflammatory bowel disease, chronic inflammatory disease, glomerular disease, nephrotoxic serum nephritis, kidney disease, Alzheimer's Disease , autoimmune encephalomyelitis, arterial thrombosis, allergic rhinitis, arteriosclerosis, Sjogren's syndrome (dermatomyositis), systemic lupus erythematosus, graft rejection, cancers with leukocyte infiltration of the skin or organs, human papilloma virus infection, prostate cancer, wound healing, amyotrophic lateral sclerosis, immune mediated disorders.
  • CCR5-related diseases and conditions including neuropathic pain, multiple sclerosis, rheumatoid arthritis,
  • the present invention further provides a method for the treatment of a clinical condition in an animal, for example, a mammal including a human which clinical condition includes those which have been discussed hereinbefore, which comprises treating said animal with a pharmaceutically effective amount of a compound according to the invention.
  • the present invention also includes a method for the treatment or prophylaxis of any of the aforementioned diseases or conditions.
  • the present invention provides the use of a compound according to the invention in the manufacture of a medicament for the treatment or prophylaxis of any of the above mentioned viral infections or conditions.
  • Combination therapies according to the present invention comprise the administration of a compound of the present invention or a pharmaceutically acceptable derivative
  • the active ingredient(s) and pharmaceutically active agents may be administered simultaneously in either the same or different pharmaceutical compositions or sequentially in any order.
  • the amounts of the active ingredient(s) and pharmaceutically active agent(s) and the relative timings of administration will be selected in order to achieve the desired combined therapeutic effect.
  • Such therapeutic agents include agents that are effective for the treatment of viral infections or associated conditions.
  • acyclic nucleosides for example acyclovir, valaciclovir, famciclovir, ganciclovir, and penciclovir
  • acyclic nucleoside phosphonates for example (S)-1-(3-hydroxy-2- phosphonyl-methoxypropyl)cytosine (HPMPC), [[[2-(6-amino-9H-purin-9- yl)ethoxy]methyl]phosphinylidene]bis(oxymethylene)-2,2-dirnethyl propanoic acid (bis-POM PMEA, adefovir dipivoxil), [[(1R)-2-(6-amino-9H-purin-9-yl)-1- methylethoxy]methyl]phosphonic acid (tenofovir), and (R)-[[2-(6-Amino-9H-purin-9
  • efavirenz efavirenz, DMP 266
  • rilpivirine efavirine
  • integrase inhibitors efavirine
  • fusion inhibitors for example T-20 and T-1249.
  • the present invention further includes the use of a compound according to the invention in the manufacture of a medicament for simultaneous or sequential administration with at least another therapeutic agent, such as those defined hereinbefore.
  • Compounds of the present invention may be administered with an agent known to inhibit or reduce the metabolism of compounds, for example ritonavir Accordingly, the present invention features a method for the treatment or prophylaxis of a disease as hereinbefore described by administration of a compound of the present invention in combination with a metabolic inhibitor. Such combination may be administered simultaneously or sequentially.
  • a suitable dose for each of the above-mentioned conditions will be in the range of 0.01 to 250 mg per kilogram body weight of the recipient (e.g. a human) per day, preferably in the range of 0.1 to 100 mg per kilogram body weight per day and most preferably in the range 0.5 to 30 mg per kilogram body weight per day and particularly in the range 1.0 to 20 mg per kilogram body weight per day.
  • all weights of active ingredient are calculated as the parent compound of formula (I); for salts or esters thereof, the weights would be increased proportionally.
  • the desired dose may be presented as one, two, three, four, five, six or more sub-doses administered at appropriate intervals throughout the day. In some cases the desired dose may be given on alternative days. These sub-doses may be administered in unit dosage forms, for example, containing 10 to 1000 mg or 50 to 500 mg, preferably 20 to 500 mg, and most preferably 50 to 400 mg of active ingredient per unit dosage form.
  • compositions of the present invention comprise at least one active ingredient, as defined above,
  • Each carrier must be acceptable in the sense of being compatible with the other ingredients of the composition and not injurious to the patient.
  • phrases include those suitable for oral, rectal, nasal, topical (including transdermal, buccal and sublingual), vaginal or parenteral (including subcutaneous, intramuscular, intravenous, intradermal, and intravitreal) administration.
  • the compositions may conveniently be presented in unit dosage form and may be prepared by any methods well known in the art of pharmacy. Such methods represent a further feature of the present invention and include the step of bringing into association the active ingredients with the carrier, which constitutes one or more accessory ingredients.
  • the compositions are prepared by uniformly and intimately bringing into association the active ingredients with liquid carriers or finely divided solid carriers or both, and then if necessary shaping the product.
  • the present invention further includes a pharmaceutical composition as hereinbefore defined wherein a compound of the present invention or a pharmaceutically acceptable derivative thereof and another therapeutic agent are presented separately from one another as a kit of parts.
  • compositions suitable for transdermal administration may be presented as discrete patches adapted to remain in intimate contact with the epidermis of the recipient for a prolonged period of time.
  • patches suitably contain the active compound 1) in an optionally buffered, aqueous solution or 2) dissolved and/or dispersed in an adhesive or 3) dispersed in a polymer.
  • a suitable concentration of the active compound is about 1% to 25%, preferably about 3% to 15%.
  • the active compound may be delivered from the patch by electrotransport or iontophoresis as generally described in Pharmaceutical Research 3 (6), 318 (1986).
  • compositions of the present invention suitable for oral administration may be presented as discrete units such as capsules, caplets, cachets or tablets each containing a predetermined amount of the active ingredients; as a powder or granules; as a solution or a suspension in an aqueous or non-aqueous liquid; or as an oil-in-water liquid emulsion or a water-in-oil liquid emulsion.
  • the active ingredient may also be presented as a bolus, electuary or paste.
  • a tablet may be made by compression or molding, optionally with one or more accessory ingredients.
  • Compressed tablets may be prepared by compressing in a suitable machine the active ingredients in a free-flowing form such as a powder or granules, optionally mixed with a binder (e.g. povidone, gelatin, hydroxypropylmethyl cellulose), lubricant, inert diluent, preservative, disintegrant (e.g. sodium starch glycollate, cross-linked povidone, cross-linked sodium carboxymethyl cellulose) surface-active or dispersing agent.
  • Molded tablets may be made by molding a mixture of the powdered compound moistened with an inert liquid diluent in a suitable machine.
  • the tablets may optionally be coated or scored and may be formulated so as to provide slow or controlled release of the active ingredients therein using, for example, hydroxypropylmethyl cellulose in varying proportions to provide the desired release profile. Tablets may optionally be provided with an enteric coating, to provide release in parts of the gut other than the stomach.
  • compositions suitable for topical administration in the mouth include lozenges comprising the active ingredients in a flavored base, usually sucrose and acacia or tragacanth; pastilles comprising the active ingredient in an inert basis such as gelatin and glycerin, or sucrose and acacia; and mouthwashes comprising the active ingredient in a suitable liquid carrier.
  • compositions suitable for vaginal administration may be presented as pessaries, tampons, creams, gels, pastes, foams or spray Pharmaceutical compositions containing in addition to the active ingredient such carriers as are known in the art to be appropriate.
  • compositions for rectal administration may be presented as a suppository with a suitable carrier comprising, for example, cocoa butter or a salicylate or other materials commonly used in the art.
  • a suitable carrier comprising, for example, cocoa butter or a salicylate or other materials commonly used in the art.
  • the suppositories may be conveniently formed by admixture of the active combination with the softened or melted carrier(s) followed by chilling and shaping in molds.
  • compositions suitable for parenteral administration include aqueous and nonaqueous isotonic sterile injection solutions which may contain antioxidants, buffers, bacteriostats and solutes which render the pharmaceutical composition isotonic with the blood of the intended recipient; and aqueous and nonaqueous sterile suspensions which may include suspending agents and thickening agents; and liposomes or other rnicroparticulate systems which are designed to target the compound to blood components or one or more organs.
  • the pharmaceutical compositions may be presented in unit-dose or multi-dose sealed containers, for example, ampoules and vials, and may be stored in a freeze-dried (lyophilized) condition requiring only the addition of the sterile liquid carrier, for example water for injection, immediately prior to use.
  • Extemporaneous injection solutions and suspensions may be prepared from sterile powders, granules and tablets of the kind previously described.
  • Unit dosage pharmaceutical compositions include those containing a daily dose or daily subdose of the active ingredients, as hereinbefore recited, or an appropriate fraction thereof.
  • compositions of this invention may include other agents conventional in the art having regard to the type of pharmaceutical composition in question, for example, those suitable for oral administration may include such further agents as sweeteners, thickeners and flavoring agents.
  • the compounds of the present invention demonstrate good oral bioavailability and favorable cardiovascular profile.
  • Nitric Acid 120 ml. was added slowly to a cold solution of sulfuric acid (200 mL). 2,4-difluorobenzoic acid (65 g, 0.41 mol, 1 equiv) was added portio ⁇ wise over 1.5 h and the reaction allowed to warm to RT. The reaction mixture-was heated to 50 0 C for 2h and then poured slowly onto ice. The product was extracted into ethyl acetate, dried over Na 2 SO 4 , filtered and concentrated to give intermediate 16 (82 g, 98% yield) as a yellow solid.
  • Intermediate 29 was prepared using chemistry described for the intermediate 12 from intermediates 5 and 28.
  • Intermediate 281 1-dimethylethyl 4-(3-thienylamino)-1- piperidinecarboxylate was synthesized using chemistry described for the intermediate 11.
  • reaction mixture was allowed to warm to RT over 2 h and then slowly poured into cold 3.3N NaOH (100 mL) with stirring. The reaction mixture was then separated and the organics dried over Na 2 SO 4 , filtered and concentrated to give a tan residue. The residue was purified by silica gel flash column chromatography (0 — s- 5% 2N NH 3 in MeOH : CH 2 CI 2 ) to give the intermediate 33 (1.2 g; 67% yield) as a tan solid.
  • Intermediate 39 was prepared from intermediate 38 using chemistry described for intermediate15 except that the product of deprotection was not free-based, but isolated as HCI salt.
  • Example 8 was prepared from intermediates 44 and 3 using chemistry described for example 2.
  • Intermediate 42 was prepared from intermediate 41 using chemistry described for intermediate 13.
  • Intermediate 43 was prepared from intermediate 42 using chemistry described for intermediate 14.
  • Intermediate 47 was prepared from intermediate 46 using chemistry described for intermediate 50.
  • Example 10 was prepared from intermediates 15 and 50 as described for example 2
  • N-(4-hydroxyphenyl)cyclopropanesulfonamide intermediate 48 (1.5 g, 7.2 mmol, 1 equiv), 6-chloro-2-methyl-3-pyridinecarbonitrile intermediate 1 (1.1 g, 7.2 mmol, 1 equiv) and cesium carbonate (4.7 g, 14.4, 2 equiv) in N 1 N-
  • Example 16 5-( ⁇ tbutyl(1- ⁇ [2-cyclopropyl-6-( ⁇ 4-[(methylsulfonyl)am ⁇ no]phenyl ⁇ oxy)- 3-pyr ⁇ d ⁇ nyl]methyl ⁇ -4-PiP er
  • Example 17 5-( ⁇ [(1 - ⁇ [2-cyclopropyl-6-( ⁇ 4-[(methylsulfonyl)am ⁇ no]phenyl ⁇ oxy)-3- pyndi ⁇ yllmethylH-P ⁇ e ⁇ diny ⁇ phenylJaminolcarbonylJamino ⁇ -difiuorobenzamide
  • ⁇ -Chloro-S-pyridinecarbonitrile (2.29g, 16.5 mmol) was dissolved in dichloromethane (60 mL). To this solution was added urea hydrogen peroxide (4.7 g, 49.6 mmol). After the resulting mixture was cooled down to 0 0 C on an ice bath, trifluoroacetic acid anhydride (6.3 mL, 45 mmol) was slowly introduced by syringe. The reaction was stirred for an additional 3 hours at room temperature before quenched with satd. aq. Na 2 S 2 O 3 and extracted with dichloromethane. The combined organic layers were then washed with satd. aq.
  • intermediate 73 (780 mg, 2.6 mmol) was treated with POCI 3 (10 mL) under reflux condition for 1 hour. After evaporation of the solvents, the residue was dissolved in dichloromethane and washed with satd. aq. NaHCO 3 . The organic phase was then dried over anhydrous sodium sulfate. Evaporation of the solvents gave a light gray solid, which was triturated with ether to afford intermediate 74 as an off white solid (720 mg, 86%).
  • PBLs Peripheral Blood Lymphocytes
  • LSM Lymphocyte Separation Medium
  • Whole blood was diluted 1 :1 with sterile phosphate buffered saline (PBS) and overlaid onto LSM in 50 mL conical centrifuge tubes. Samples were centrifuged at 500 x g at room temperature for 30 minutes. PBLs were then gently removed from the interface between LSM and aqueous phases and washed twice in sterile PBS.
  • PBS sterile phosphate buffered saline
  • the cells were resuspended in 150 mL of RPIvII containing 20% fetal bovine serum, 10% T-cell growth factor (IL- 2), 50ug/mL gentamicin (RPMI 20/10/50) and 5ug/mL Phytohaemagluttinin and incubated at 37 0 C, 5% CO 2 for 2 to 3 days. On the second or third day, the cells were washed once with PBS and counted for assay.
  • IL-2 T-cell growth factor
  • RPMI 20/10/50 50ug/mL gentamicin
  • Phytohaemagluttinin Phytohaemagluttinin
  • PHA stimulated PBLs were centrifuged at 260 x g for 15 min, washed once with PBS, centrifuged as before, resuspended to 8x10 6 cells/mL in RPMI 20/10/50, and 50 ⁇ L was distributed to 96-well tissue culture plates.
  • Test compounds were serially diluted in 4-fold increments at 2Xthe final concentration in RPMI 20/10/50 + 0.2% DMSO. Fifty (50) uL of diluted compound was transferred to the PBLs and placed in
  • RT assay buffer 500 mM KCI, 50 mM DTT and 0.5% NP40 in distilled water
  • 40 ⁇ L of RT assay buffer (1.25 mM EGTA, 125 mM Tric/HCI, 12.5 mM MgCI 2 , 68 Ci/mmole methyl- 3 H deoxythymidine-5'-triphosphate, and 0.62 O.D.
  • the first wash consisted of approximately 300 ⁇ L of 5% Na 2 HPO 4 , followed by one wash of approximately 300 ⁇ L of distilled water, and a final wash consisting of 100 ⁇ L of 95% ethanol.
  • the plates were allowed to dry completely at room temperature, bottom-sealed with an adhesive backing, and 50 ⁇ L of scintillation cocktail was added. The plates were topsealed and read in a Topcount (Packard) luminometer at 10s/well.
  • C logiotlC ⁇ o)
  • Compounds of the invention have IC 50 values less than 1OnM.
  • HOS-Luc cells were created by introducing an HIV-1 LTR-luciferase reporter into HOS-CD4.CCR5 (NIH AIDS Research and Reference Reagents Program, cat. #3318). The cells were propagated in DMEM complete medium. Cells were split 1 :20 by standard trypsinization when confluency reached 80% (roughly every 2 to 3 days).
  • Black-walled 96-weIt tissue culture plates were seeded with HOS-Luc at 6x10 3 cells per well in 50 ⁇ L DMEM containing 2% FBS and placed in a humidified incubator at 37 0 C, 5% CO 2 overnight. The following day, test compounds were serially diluted in 4-fold increments at 2X the final concentration in DMEM + 2% FBS + 0.2% DMSO. Diluted compound (50 ⁇ L) was transferred to the HOS-Luc cells and the plates were placed in a humidified incubator at 37 0 C, 5% CO 2 for 1 hr.
  • the HIV/compound mixture (100 ⁇ l_) was transferred to the black-walled plates containing 100 ⁇ L of cells/compound. The plates were placed in a humidified incubator at 37 0 C, 5% CO 2 for four days.
  • RLU Relative Light Unit
  • V 1TCK upper bound of response
  • Compounds of the invention have IC 50 values less than 1OnM.
  • Non-fasted male CD-1 mice (Charles River Laboratories, Raleigh NC), ranging in weight from 28 g to 30 g, were used for each study.
  • a single dose (10 mL/kg of 0.1 mg free base/mL) was administered via tail vein.
  • animals received a single dose (10 mL/kg of 0.1 or 0.3 mg free base/mL) by an oral gavage needle.
  • Blood (approximately 0.8 mL) was taken via cardiac puncture from three mice/dose group at each timepoint of 0.083 (IV only), 0.167 (IV only), 0.25, 0.5, 1, 2, 4, 6, 8, and 24 h and placed on ice.
  • Syringes used for sample collection contained approximately 10 ⁇ L EDTA. Plasma was harvested by centrifugation for 3 min at 4000 x g and 4°C and stored at -80 0 C until analysis.
  • mice were orally administered a single dose (10 mL/kg of 0.1 or 0.3 mg free base/mL) by an oral gavage needle.
  • Plasma samples were thawed to room temperature and vortex-mixed. Aliquots (50 ⁇ L) of standards and study samples were pipetted into 96-well plates. Liver and spleen tissues were thawed to room temperature and weighed. Each pooled liver and spleen sample was diluted with 4 and 9 volumes of water (mL/g), respectively. Following homogenization (Tomtec Autogizer), homogenates were diluted 5-fold in pooled mouse plasma and analyzed with plasma samples. Liver and spleen QC samples were prepared in homogenized blank liver or spleen homogenate.
  • Acetonitrile (200 ⁇ L) containing internal standard was added to each well, samples vortex-mixed, and plates centrifuged for 10 min at 2055 x g and 4 0 C.
  • the acetonitrile supernatant (100 ⁇ L) was transferred to clean 96 well plates containing 100 ⁇ L or 200 ⁇ L of 0.1 % formic acid in water and assayed by LC/MS/MS.
  • the analytical system consisted of a CTC HTS PAL autosampler, Hewlett Packard (HP) 1100 binary pump and Applied Biosystems API 365 mass spectrometer. Samples (10 ⁇ L) were injected onto a 30 x 2 mm, .3micron, Luna CN column
  • Non-compartmental pharmacokinetic parameters [terminal plasma half-life (U 12 ), maximum plasma concentration (C max ), time of maximum plasma concentration (Tmax) plasma clearance (CL), steady-state volume of distribution (V ss ), and area under the plasma concentration-time curve extrapolated to infinite time (AUC D -.)] were calculated based on the composite plasma concentration time data using Wi ⁇ Nonlin Professional 4.1 (Pharsight, Mountain View CA) .
  • Dose-normalized AUC a-*. (DNAUC 0 ⁇ ») was obtained by dividing the AUC 0 ⁇ «with its respective dose and expressed as unit dose AUC 0 ⁇ « (ng*h/mL/mg/kg).
  • Oral bioavailability (F) was calculated using the following equation:
  • test compound For intravenous administration, a single dose of test compound was administered into the jugular cannula followed by a 1 mL flush with saline
  • animals received a single dose by an oral gavage needle
  • Additional animals received capsule doses (shown in the following table) which were administered by a small capsule-dosing syringe (Harvard Apparatus)
  • Plasma samples were taken from the jugular cannula at 0 083 (IV only), 0 167 (IV only), 0 25, 0 5, 1 , 2, 4, 6, 8, and 24 h postdose and placed on ice Syringes used for sample collection contained approximately 10 ⁇ L EDTA Plasma was harvested by centrifugation for 3 mm at 4000 x g and 4°C and stored at -80°C until analysis
  • Plasma samples were thawed to room temperature and vortex-mixed.
  • Standards were prepared in pooled rat plasma containing EDTA.
  • Quality control (QC) samples were prepared from separately weighed sample stocks. Aliquots (50 ⁇ L) of standards, QCs and study samples were pipetted into 96-well plates.
  • Acetonitrile (200 ⁇ L) containing internal standard was added to each well, samples vortex- mixed, and plates centrifuged for 20 min at 2055 x g and 4°C. The acetonitrile supernatant (150 ⁇ L) was transferred to clean 96 well plates containing 100 ⁇ L of 0.1% formic acid in water and assayed by LC/MS/MS.
  • the analytical system consisted of a CTC HTS PAL autosampler, Hewlett Packard (HP) 1100 binary pump and Applied Biosystems API 4000 mass spectrometer. Samples (10 ⁇ L) were injected onto a 30 x 2 mm, 3micron, Atlantis C18 column (Waters) using a gradient eluting system at a flow rate of 0.75 mL/min. Mobile phases consisted of A) water containing 0.5% formic acid and B) acetonitrile containing 0.5% formic acid. The initial mobile phase composition of 90% A:10% B was followed by a linear gradient to 5% A:95% B at 1 min. The 5% A:95% B composition was held for 0.5 min followed by a linear gradient to 90% A:10% B at 2.0 min. Data were acquired and analyzed using Applied BioSystems Analyst version 1.4.1 software.
  • Plasma samples were thawed to room temperature and vortex-mixed.
  • Standards were prepared in pooled rat plasma containing EDTA.
  • Quality control (QC) samples in plasma were prepared from separately weighed sample stocks. Aliquots (50 ⁇ L) of standards, QCs and plasma samples were pipetted into 96-well plates. Liver and spleen tissues were weighed, thawed to room temperature and water (9 volumes, mL/g) added to each sample. Following homogenization (Tomtec Autogizer), homogenate (10 ⁇ L) was added to pooled rat plasma (40 ⁇ L) containing EDTA in 96-well plates.
  • Tomtec Autogizer homogenate (10 ⁇ L) was added to pooled rat plasma (40 ⁇ L) containing EDTA in 96-well plates.
  • Liver and spleen QC samples were prepared in homogenized blank liver or spleen homogenate and aliquots (10 ⁇ L) added to pooled rat plasma (40 ⁇ L) containing EDTA in 96-well plates. Acetonit ⁇ le (200 ⁇ L) containing internal standard was added to each well, samples vortex-mixed, and plates centrifuged for
  • the analytical system consisted of a CTC HTS PAL autosampler, Hewlett Packard (HP) 1100 binary pump and Applied Biosystems API 365 mass spectrometer. Samples (10 ⁇ L) were injected onto a 30 x 2 mm, 2.5micron, Atlantis C18 column (Waters) using an isocratic eluting system at a flow rate of 0.8 mL/min.
  • Mobile phases consisted of A) water containing 0.1% acetic acid, pH adjusted to 6.2 with ammonium hydroxide:acetonitrile (95:5) and B) acetonitrile containing 0.1% acetic acid. The mobile phase composition was 50% A:50% B and total run time was 1.1 min. Data were acquired and analyzed using Applied BioSystems Analyst version 1.4.1 software.
  • Liver and spleen samples were converted to ng/g following quantitation against the plasma calibration curves accounting for the appropriate dilution factor (50-fold).
  • Non-compartmental pharmacokinetic parameters [terminal plasma half-life (tie), maximum plasma concentration (C max ), time of maximum plasma concentration (Tmax) plasma clearance (CL), steady-state volume of distribution (V ss ), and area under the plasma concentration-time curve extrapolated to infinite time (AUC D -4] were calculated based on the individual plasma concentration time data using WinNonlin Professional 4.1 (Pharsight, Mountain View CA) .
  • Dose-normalized AUC 0- + «, (DNAUC o ⁇ ») was obtained by dividing the AUC o ⁇ »by its respective dose and expressed as unit dose AUC D ⁇ » (ng*h/mL/mg/kg).
  • Oral bioavailability (F) was calculated using the following equation:
  • the cloned hERG channel was heterologously expressed in Chinese hamster ovary
  • the membrane-bead suspension was mixed with tritiated dofetilide (final concentration of 7 nM/well) and dispensed into a white Greiner polypropylene 384-well plate (10 ml/well), containing 0 1 ml of test compound in DMSO
  • the assay plates were centrifuged at 10,000 rpm for 1 minute and left to equilibrate for 2-3 hrs at room temperature before reading on a ViewLuxTM
  • Concentration response curve data were generated from an 11 -point inhibition curve (top assay concentration of 60 mM and a 1 :4 step-dilution), a four parameter curve-fit being applied.
  • the reference compounds used were: astemizole, dofetilide, quinidine and cisapride.
  • % inhibition 100 x (1- (sample - NSB) / (TB - NSB), where sample was the signal observed in a particular unknown sample well.
  • the concentration-response curve was analyzed using a non linear equation, 4- parameter logistics, giving a determination of plC50.
  • Curve QC, plate QC (z' > 0.2) and assay QC (plC50 for reference compounds ⁇ 0.5 log units of the rolling mean) were applied when analyzing all experiments.

Landscapes

  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Plural Heterocyclic Compounds (AREA)

Abstract

The present invention relates to compounds useful in the treatment of CCR5-related diseases and disorders, for example, useful in the inhibition of HIV replication, the prevention or treatment of an HIV infection, and in the treatment of the resulting acquired immune deficiency syndrome (AIDS).

Description

CCR5 ANTAGONISTS AS THERAPEUTIC AGENTS
BACKGROUND OF THE INVENTION
The human immunodeficiency virus ("HIV") is the causative agent for acquired immunodeficiency syndrome ("AIDS"), a disease characterized by the destruction of the immune system, particularly of CD4+ T-cells, with attendant susceptibility to opportunistic infections, and its precursor Al DS-related complex ("ARC"), a syndrome characterized by symptoms such as persistent generalized lymphadenopathy, fever and weight loss.
In addition to CD4, HIV requires a co-receptor for entry into target cells. The chemokine receptors function together with CD4 as co-receptors for HIV. The chemokine receptors CXCR4 and CCR5 have been identified as the main co- receptors for HIV-1. CCR5 acts as a major co-receptor for fusion and entry of macrophage-tropic HIV into host cells. These chemokine receptors are thought to play an essential role in the establishment and dissemination of an HIV infection. Therefore, CCR5 antagonists are thought to be useful as therapeutic agents active against HIV.
Nitrogen-containing heterocyclic compounds that are CCR5 aπtangonists are disclosed in WO2006030925, WO2004080966, and WO2001087839. However, such compounds can demonstrate limited oral bioavailability or unfavorable cardiovascular profile.
We have now discovered a series of small molecule compounds that are useful as inhibitors of HIV replication.
BRIEF DESCRIPTION OF THE INVENTION
The present invention features compounds that are useful in the inhibition of HIV replication, the prevention of infection by HlV, the treatment of infection by HIV and in the treatment of AIDS and/or ARC, either as pharmaceutically acceptable salts or pharmaceutical composition ingredients. The present invention further features methods of treating AIDS, methods of preventing infection by HIV, and methods of treating infection by HIV as monotherapy or in combination with other aπtivirals, anti- infectives, immunomodulators, antibiotics or vaccines. The present invention also features pharmaceutical compositions, comprising the above-mentioned compounds that are suitable for the prevention or treatment of CCR5-related diseases and conditions. The present invention further features processes for making the above- mentioned compounds.
DETAILED DESCRIPTION OF THE INVENTION
The present invention features a compound selected from the group consisting of 2,4-Difluoro-5-({[(1-{[2-methyl-6-({4-[(methylsulfonyl)amino]phenyl}oxy)-3- pyridinyl]methyl}-4-piperidinyl)(pheπyl)amino]carbonyl}amino)benzamide; 2,4-Difluoro-5-({[(3-fluorophenyl)(1-{[2-methyl-6-({4- [(rnethylsulfonyl)amiπo]phenyl}oxy)-3-pyridinyl]methyl}-4- piperidinyl)amino]carbonyl}amino)benzamide;
5-{[(Butyl{1-[(2-methyl-6-{[4-(methylsulfonyl)phenyl]oxy}-3-pyridinyl)methyl]-4- piperidinyl}amino)carbonyl]amino}-2,4-difluorobenzamide; 2,4-Difluoro-5-({[(3-fluorophenyl)(1-{[6-({4-[(2-hydroxyethyl)sulfonyl]phenyl}oxy)-2- methyl-3-pyridinyl]methyl}-4-piperidinyl)amino]carbonyl}amino)benzarnide; 2,4-Difluoro-5-({[(1-{[2-methyl-6-({4-[(methylsuIfonyl)amino]phenyl}oxy)-3- pyridinyl]methyl}-4-piperidinyl)(3-thienyl)amino]carbonyl}arnino)benzamide; 2,4-Difluoro-5-({[(3-fluorophenyl)(1-{[2-methyl-6-({4-
[(methylsulfonyl)amino]phenyI}oxy)-3-pyridinyl]rπethyl}-4- piperidinyl)amino]carbonyl}amino)-/V-(1-methylethyl)benzamide; 5-({[(3-Fluorophenyl){H[2-methyl-6-({4-[(methylsulfonyl)amino]phenyl}oxy)-3- pyridinyl]methyl}-4-piperidinyl)amino]carbonyl}amino)-2-(trifluoromethyl)benzamide;
5-({[(2-Cyclopropylethyl)(1-{[2-methyl-6-({4-[(methylsulfonyi)amino]pheπyl}oxy)-3- pyridinyl]methylH-piperidinyl)amino]carbonyl}amino)-2,4-difluorobenzarnide;
2,4-Difluoro-5-[({(3-fluorophenyl)[1-({2-methyl-6-[(4-{[(1- methylethyl)sulfonyl]amino}phenyl)oxy]-3-pyridiπyl}methyI)-4- piperidinyl]amino}carbonyl)amino]benzamide;
5-({[(1-{[6-({4-[(Cyclopropylsulfonyl)amino]phenyI}oxy)-2-methyl-3-pyridinyl]methyl}-
4-piperidiπyl)(3-fluorophenyl)amino]carbonyI}amino)-2,4-difluorobeπzamide;
Λ/-(4-{[5-({4-[(3-Fluorophenyl)({[4-methyl-3-(1,3-oxazoI-2- yI)phenyl]amino}carbonyl)amino]-1-piperidinyi}methyl)-6-methyl-2- pyridinyl]oxy}phenyl)methanesulfonamide;
Λ/-(4-{[5-({4-[({[2,4-Difluoro-5-(1,3,4-oxadiazol-2- yl)phenyl]amino}carbonyl)(pheπyl)amiπo]-1-piperidinyl}methyl)-6-methyl-2- pyridiπyl]oxy}pheπyl)methaπesulfonannide;
Λ/-(4-{[5-({4-t({[2,4-Difluoro-5-(1,3,4-oxadiazol-2-yI)phenyl]amino}carbonyl)(3- fluorophenyl)amino]-1-piperidinyl}methyl)-6-methyl-2- pyridinyl]oxy}phenyl)methanesulfonamide;
Λ/-(4-{[5-({4-[({[2,4-Difluoro-5-(5-methyl-1 ,3,4-oxadiazol-2- yl)phenyl]amino}carbonyl)(phenyl)amino]-1-piperidinyl}methyl)-6-rnethyl-2- pyridinyl]oxy}phenyl)methanesulfonamide;
W-(4-{[5-({4-[({[4-Fluoro-3-(1 ,3-oxazol-2-yl)phenyl]amino}carbonyl)(3- fluoropheπyl)amino]-1-piperidinyl}methyl)-6-methyl-2- pyridinyl]oxy}phenyl)methanesulfonamide;
5-({[butyl(1-{[2-cyclopropyl-6-({4-[(methylsulfonyl)amino]phenyl}oxy)-3- pyridlnyl]methyl}-4-piperidinyl)amiπo]carbonyl}amino)-2,4-difluorobenzamide;
5-({[(1-{[2-cyclopropyl-6-({4-[(methylsulfonyl)amino]phenyl}oxy)-3-pyridinyl]methyl}-
4-piperidinyl)(pheπyl)amino]carboπyl}amino)-2,4-difluorobenzamide; and pharmaceutically acceptable salts thereof.
The term "pharmaceutically effective amount" refers to an amount of a compound of the invention that is effective in treating a CCR5-related disease, for example a virus infection, for example an HIV infection, in a patient either as monotherapy or in combination with other agents. The term "treatment" as used herein refers to the
alleviation of symptoms of a particular disorder in a patient, or the improvement of an ascertainable measurement associated with a particular disorder, and may include the suppression of symptom recurrence in an asymptomatic patient such as a patient in whom a viral infection has become latent. The term "prophylaxis" refers to preventing a disease or condition or preventing the occurrence of symptoms of such a disease or condition, in a patient. As used herein, the term "patient" refers to a mammal, including a human.
The term "pharmaceutically acceptable carrier" refers to a carrier that may be administered to a patient, together with a compound of this invention, and which does not destroy the pharmacological activity thereof and is nontoxic when administered in doses sufficient to deliver a therapeutic amount of the therapeutic agent.
Pharmaceutically acceptable salts of the compounds according to the invention include those derived from pharmaceutically acceptable inorganic and organic acids and bases. Examples of suitable acids include hydrochloric, hydrobromic, sulfuric, nitric, perchloric, fumaric, glutamic, maleic, mandelic, phosphoric, glycollic, lactic, salicyclic, succinic, toluene-p-sulfonic, tartaric, acetic, citric, orotic, toluenesulfonic, methanesulfonic, ethanesulfonic, formic, benzoic, malonic, naphthalene-2-sulfonic and benzenesulfonic acids. Other acids, such as oxalic, while not in themselves pharmaceutically acceptable, may be employed in the preparation of salts useful as intermediates in obtaining the compounds of the invention and their pharmaceutically acceptable acid addition salts.
Salts derived from appropriate bases include alkali metal (e.g. sodium), alkaline earth metal (e.g., magnesium), ammonium, NW4 + (wherein W is C1J, alkyl) and other amine salts. Physiologically acceptable salts of a hydrogen atom or an amino group include salts of organic carboxylic acids such as acetic, lactic, tartaric, malic, isethionic, lactobionic and succinic acids; organic sulfonic acids such as methanesulfonic, ethanesulfonic, benzenesulfonic and p-toluenesulfonic acids and inorganic acids such as hydrochloric, sulfuric, phosphoric and sulfamic acids.
Physiologically acceptable salts of a compound with a hydroxy group include the anion of said compound in combination with a suitable cation such as Na+, NH4 +, and NW4 + (wherein W is a C^alkyl group)
Any reference to any of the above compounds also includes a reference to a pharmaceutically acceptable salt thereof
Salts of the compounds of the present invention may be made by methods known to a person skilled in the art For example, treatment of a compound of the present invention with an appropriate base or acid in an appropriate solvent will yield the corresponding salt
The present invention features compounds according to the invention for use in medical therapy, for example for the treatment or prophylaxis of viral infections such as an HIV infections and associated conditions Reference herein to treatment extends to prophylaxis as well as the treatment of established infections, symptoms, and associated clinical conditions such as AIDS related complex (ARC), Kaposi's sarcoma, and AIDS dementia
The present invention features use of the compounds of the present invention in the manufacture of a medicament for the treatment or prophylaxis of a CCR5- related disease or condition, for example, a viral infection, for example, an HIV infection
According to another aspect, the present invention provides a method for the treatment or prevention of the symptoms or effects of a viral infection in an infected animal, for example, a mammal including a human, which comprises treating said animal with a pharmaceutically effective amount of a compound according to the invention According to one aspect of the invention, the viral infection is a retroviral infection, in particular an HIV infection A further aspect of the invention includes a method for the treatment or prevention of the symptoms or effects of an HBV infection
The compounds according to the invention may also be used in adjuvant therapy in the treatment of HIV infections or HIV-associated symptoms or effects, for example Kaposi's sarcoma.
The compounds of the present invention may also be used in the prevention or treatment of other CCR5-related diseases and conditions, including neuropathic pain, multiple sclerosis, rheumatoid arthritis, autoimmune diabetes, chronic implant rejection, asthma, rheumatoid arthritis, Crohns Disease, inflammatory bowel disease, chronic inflammatory disease, glomerular disease, nephrotoxic serum nephritis, kidney disease, Alzheimer's Disease , autoimmune encephalomyelitis, arterial thrombosis, allergic rhinitis, arteriosclerosis, Sjogren's syndrome (dermatomyositis), systemic lupus erythematosus, graft rejection, cancers with leukocyte infiltration of the skin or organs, human papilloma virus infection, prostate cancer, wound healing, amyotrophic lateral sclerosis, immune mediated disorders.
The present invention further provides a method for the treatment of a clinical condition in an animal, for example, a mammal including a human which clinical condition includes those which have been discussed hereinbefore, which comprises treating said animal with a pharmaceutically effective amount of a compound according to the invention. The present invention also includes a method for the treatment or prophylaxis of any of the aforementioned diseases or conditions.
In yet a further aspect, the present invention provides the use of a compound according to the invention in the manufacture of a medicament for the treatment or prophylaxis of any of the above mentioned viral infections or conditions.
The above compounds according to the invention and their pharmaceutically acceptable derivatives may be employed in combination with other therapeutic agents for the treatment of the above infections or conditions. Combination therapies according to the present invention comprise the administration of a compound of the present invention or a pharmaceutically acceptable derivative
thereof and another pharmaceutically active agent. The active ingredient(s) and pharmaceutically active agents may be administered simultaneously in either the same or different pharmaceutical compositions or sequentially in any order. The amounts of the active ingredient(s) and pharmaceutically active agent(s) and the relative timings of administration will be selected in order to achieve the desired combined therapeutic effect.
Examples of such therapeutic agents include agents that are effective for the treatment of viral infections or associated conditions. Among these agents are acyclic nucleosides, for example acyclovir, valaciclovir, famciclovir, ganciclovir, and penciclovir, acyclic nucleoside phosphonates, for example (S)-1-(3-hydroxy-2- phosphonyl-methoxypropyl)cytosine (HPMPC), [[[2-(6-amino-9H-purin-9- yl)ethoxy]methyl]phosphinylidene]bis(oxymethylene)-2,2-dirnethyl propanoic acid (bis-POM PMEA, adefovir dipivoxil), [[(1R)-2-(6-amino-9H-purin-9-yl)-1- methylethoxy]methyl]phosphonic acid (tenofovir), and (R)-[[2-(6-Amino-9H-purin-9- yl)-1-methylethoxy]methyl]phosphonic acid bis-(isopropoxycarbonyloxymethyl)ester (bis-POC-PMPA), nucleoside reverse transcriptase inhibitors, for example 3'-azido- 3'-deoxythymidine (AZT, zidovudine), 2',3'-dideoxycytidine (ddC, zalcitabine), 2',3'- dideoxyadenosine, 2',3'-dideoxyinosine (ddl, didanosine), 2',3'-didehydrothymidine (d4T, stavudine), (-)-cjs-1-(2-hydroxymethyl)-1,3-oxathiolane 5-yl)-cytosine (lamivudine), cjs-1-(2-(hydroxymethyl)-1,3-oxathiolan-5-yl)-5-fluorocytosine (FTC), (-)-cis-4-t2-amino-6-(cyclopropylamino)-9H-purin-9-yl]-2-cyclopentene-1-methanol (abacavir), and ribavirin, protease inhibitors, for example indinavir, ritonavir, nelfinavir, amprenavir, saquinavir, fosamprenavir, lopinavir, tipranavir, interferons such as oc-interferon, immuπomodulators such as interleukin Il or thymosin, granulocyte macrophage colony stimulating factors, erythropoetin, soluble CD4 and genetically engineered derivatives thereof, non-nucleoside reverse transcriptase inhibitors (NNRTIs), for example nevirapine (BI-RG-587), alpha-((2-acetyl-5- methylphenyl)amino)-2,6-dichloro-benzeneacetamide (loviride), 1 -[3- (isopropylamino)-2-pyridyl]-4-[5-(methanesulfonamido)-1 H-indol-2- ylcarbonyl]piperazine monomethanesulfonate (delavirdine), (S)-6-chloro-4- (cyclopropylethynyl)-i ,4-dihydro-4-(trifluoromethyl)-2H-3, 1 -benzoxazin-2-one
(efavirenz, DMP 266), rilpivirine, integrase inhibitors, or fusion inhibitors, for example T-20 and T-1249.
The present invention further includes the use of a compound according to the invention in the manufacture of a medicament for simultaneous or sequential administration with at least another therapeutic agent, such as those defined hereinbefore.
Compounds of the present invention may be administered with an agent known to inhibit or reduce the metabolism of compounds, for example ritonavir Accordingly, the present invention features a method for the treatment or prophylaxis of a disease as hereinbefore described by administration of a compound of the present invention in combination with a metabolic inhibitor. Such combination may be administered simultaneously or sequentially.
In general a suitable dose for each of the above-mentioned conditions will be in the range of 0.01 to 250 mg per kilogram body weight of the recipient (e.g. a human) per day, preferably in the range of 0.1 to 100 mg per kilogram body weight per day and most preferably in the range 0.5 to 30 mg per kilogram body weight per day and particularly in the range 1.0 to 20 mg per kilogram body weight per day. Unless otherwise indicated, all weights of active ingredient are calculated as the parent compound of formula (I); for salts or esters thereof, the weights would be increased proportionally. The desired dose may be presented as one, two, three, four, five, six or more sub-doses administered at appropriate intervals throughout the day. In some cases the desired dose may be given on alternative days. These sub-doses may be administered in unit dosage forms, for example, containing 10 to 1000 mg or 50 to 500 mg, preferably 20 to 500 mg, and most preferably 50 to 400 mg of active ingredient per unit dosage form.
While it is possible for the active ingredient to be administered alone it is preferable to present it as a pharmaceutical composition. The compositions of the present invention comprise at least one active ingredient, as defined above,
together with one or more acceptable carriers thereof and optionally other therapeutic agents. Each carrier must be acceptable in the sense of being compatible with the other ingredients of the composition and not injurious to the patient.
Phamaceutical compositions include those suitable for oral, rectal, nasal, topical (including transdermal, buccal and sublingual), vaginal or parenteral (including subcutaneous, intramuscular, intravenous, intradermal, and intravitreal) administration. The compositions may conveniently be presented in unit dosage form and may be prepared by any methods well known in the art of pharmacy. Such methods represent a further feature of the present invention and include the step of bringing into association the active ingredients with the carrier, which constitutes one or more accessory ingredients. In general, the compositions are prepared by uniformly and intimately bringing into association the active ingredients with liquid carriers or finely divided solid carriers or both, and then if necessary shaping the product.
The present invention further includes a pharmaceutical composition as hereinbefore defined wherein a compound of the present invention or a pharmaceutically acceptable derivative thereof and another therapeutic agent are presented separately from one another as a kit of parts.
Compositions suitable for transdermal administration may be presented as discrete patches adapted to remain in intimate contact with the epidermis of the recipient for a prolonged period of time. Such patches suitably contain the active compound 1) in an optionally buffered, aqueous solution or 2) dissolved and/or dispersed in an adhesive or 3) dispersed in a polymer. A suitable concentration of the active compound is about 1% to 25%, preferably about 3% to 15%. As one particular possibility, the active compound may be delivered from the patch by electrotransport or iontophoresis as generally described in Pharmaceutical Research 3 (6), 318 (1986).
Pharmaceutical compositions of the present invention suitable for oral administration may be presented as discrete units such as capsules, caplets, cachets or tablets each containing a predetermined amount of the active ingredients; as a powder or granules; as a solution or a suspension in an aqueous or non-aqueous liquid; or as an oil-in-water liquid emulsion or a water-in-oil liquid emulsion. The active ingredient may also be presented as a bolus, electuary or paste.
A tablet may be made by compression or molding, optionally with one or more accessory ingredients. Compressed tablets may be prepared by compressing in a suitable machine the active ingredients in a free-flowing form such as a powder or granules, optionally mixed with a binder (e.g. povidone, gelatin, hydroxypropylmethyl cellulose), lubricant, inert diluent, preservative, disintegrant (e.g. sodium starch glycollate, cross-linked povidone, cross-linked sodium carboxymethyl cellulose) surface-active or dispersing agent. Molded tablets may be made by molding a mixture of the powdered compound moistened with an inert liquid diluent in a suitable machine. The tablets may optionally be coated or scored and may be formulated so as to provide slow or controlled release of the active ingredients therein using, for example, hydroxypropylmethyl cellulose in varying proportions to provide the desired release profile. Tablets may optionally be provided with an enteric coating, to provide release in parts of the gut other than the stomach.
Pharmaceutical compositions suitable for topical administration in the mouth include lozenges comprising the active ingredients in a flavored base, usually sucrose and acacia or tragacanth; pastilles comprising the active ingredient in an inert basis such as gelatin and glycerin, or sucrose and acacia; and mouthwashes comprising the active ingredient in a suitable liquid carrier.
Pharmaceutical compositions suitable for vaginal administration may be presented as pessaries, tampons, creams, gels, pastes, foams or spray Pharmaceutical compositions containing in addition to the active ingredient such carriers as are known in the art to be appropriate.
Pharmaceutical compositions for rectal administration may be presented as a suppository with a suitable carrier comprising, for example, cocoa butter or a salicylate or other materials commonly used in the art. The suppositories may be conveniently formed by admixture of the active combination with the softened or melted carrier(s) followed by chilling and shaping in molds.
Pharmaceutical compositions suitable for parenteral administration include aqueous and nonaqueous isotonic sterile injection solutions which may contain antioxidants, buffers, bacteriostats and solutes which render the pharmaceutical composition isotonic with the blood of the intended recipient; and aqueous and nonaqueous sterile suspensions which may include suspending agents and thickening agents; and liposomes or other rnicroparticulate systems which are designed to target the compound to blood components or one or more organs. The pharmaceutical compositions may be presented in unit-dose or multi-dose sealed containers, for example, ampoules and vials, and may be stored in a freeze-dried (lyophilized) condition requiring only the addition of the sterile liquid carrier, for example water for injection, immediately prior to use. Extemporaneous injection solutions and suspensions may be prepared from sterile powders, granules and tablets of the kind previously described.
Unit dosage pharmaceutical compositions include those containing a daily dose or daily subdose of the active ingredients, as hereinbefore recited, or an appropriate fraction thereof.
It should be understood that in addition to the ingredients particularly mentioned above the pharmaceutical compositions of this invention may include other agents conventional in the art having regard to the type of pharmaceutical composition in question, for example, those suitable for oral administration may include such further agents as sweeteners, thickeners and flavoring agents.
Compounds of the present invention demonstrate advantageous properties compared with CCR5 antangonists disclosed in WO2006030925, WO2004080966, and VVO2001087839. As shown in Examples 20, 21 , and 22, at Tables 1 , 2, and 3,
the compounds of the present invention demonstrate good oral bioavailability and favorable cardiovascular profile.
The following examples are intended for illustration only and are not intended to limit the scope of the invention in any way.
Example 1
2.4-Difluoro-5-(ff(1-([2-methyl-6-α4-ffmethylsulfonvnaminolphenylk)XvV3- pyridinvπmethylM-piperidinvπfphenyl)amino1carbonyl}arnino)benzamide
Figure imgf000013_0001
I ntermediate 3 Λ/-{4-[(5-formyl-6-methyl-2-pyridinyl)oxy]phenyl}methanesulfonamide (46 mg, 0.15 mmol, 1.0 eq) and intermediate1! 0 2,4-difluoro-5-({[phenyl(4- piperidinyl)amino]carbonyl}amino)benzamide hydrochloride (62 mg; 0.15 mmol, 1.0 eq) in 1 ,2-dichloroethane (5.0 mL) was treated with triethylamine (42 uL, 0.3 mmol, 2.0 eq) at RT. Mp-BH(OAc)3 (205 mg, 0.45 mmol, 3.0 eq) was added and the entire reaction mixture was allowed to stir at RT overnight, filtered off the resin, washed with DCM and concentrated down. Residue was purified by ISCO FC with 0-4 % MeOH(W/ 2M NH3) in CH2CI2 to give 2,4-difluoro-5-({[(1-{[2-methyl-6-({4- [(methylsulfonyl)amino]phenyl}oxy)-3-pyridinyl]methyl}-4- piperidinyl)(phenyl)amino]carbonyl}amino)beπzamide as a white solid (37 mg, 37 % yield). 1H NMR (CDCI3, 400 MHz) δ 8.66 (t, J=8.70 Hz, 1 H), 7.41 - 7.54 (m, 4 H), 7.18 - 7.28 (m, 4 H), 7.04 - 7.13 (m, 2 H), 6.79 (t, J=10.44 Hz, 2 H), 6.54 (d, J=8.24 Hz, 1 H), 6.49 (d, J=12.64 Hz, 1 H), 5.90 (d, J=2.01 Hz, 1 H), 5.86 (br. s., 1 H), 4.46 - 4.59 (m, 1 H), 3.34 (s, 2 H), 2.99 (s, 3 H), 2.83 (d, J=10.99 Hz, 2 H), 2.38 (s, 3 H), 2.13 (t, J=11.35 Hz, 2 H), 1.84 (d, .7=10.62 Hz, 2 H), 1.31 - 1.45 (m, 2 H). HRMS: (M+H)* calcd for C33H34F2N6O5S + H, 665.2352; found, 665.2354.
The synthesis of intermediate aldehyde 3:
Figure imgf000014_0001
s
The synthesis of intermediate 1 was accomplished as described in the literature [Singh et al., (1991) Synthesis pp894-896].
Preparation of phenol 4 was accomplished as in J. Med. Chem. 1999, 42, 1041 To a stirred 0 °C suspension of 4-aminophenol (30Og, 2.75 mol) in 3L MeOH was slowly added methanesulfonyl chloride (106 mL, 1.37 mol). The reaction mixture was then stirred at room temperature for 2h and evaporated to give a brown solid residue. The residue was stirred vigorously in 3.5 L 1 N HCI for 1h. The solution was filtered to obtain the phenol 4 as an off-white solid, which was washed with ice-cold water and dried in vacuo over NaOH pellets. Yield obtained was 65% (166.2g). 1H NMR of intermediate 4 MHz, DMSO-cfe) d 9.36 (s, 1 H), 9.17 (s, 1 H), 6.99 - 7.06 (m, 2 H), 6.70 - 6.75 (m, 2 H), 2.84 (s, 3 H). LRMS: (M-H)" calcd for C7H9NO3S - H, 186; found, 186
Solution of chloropyridine 1 (25g, 0.16 mol), phenol 4 (33.7g, 0.18 mol), and Cs2CO3 (106.8g, 0.33 mol) in dry DMF (350 mL) was stirred at 80 °C overnight. The reaction mixture was then filtered and rinsed with CH2CI2. The filtrate was concentrated to a residue by rotary evaporation, and then diluted with water. The pH was adjusted to 7 by the addition of 2M aqueous HCI and then extracted with CH2CI2. The organic extracts were pooled, dried (Na2SO,)), filtered and evaporated to provide a dark brown solid. To this was added 200 mL Et2O, and the solution was stirred overnight and then filtered. The light brown solid was washed with a small amount of Et2O, and dried to provide intermediate 2 as a light brown solid (46.3g, 93%). 1H NMR of 2
(400 MHz, DMSO-d6) δ 9.77 (br. s., 1 H), 8.20 (d, J=8.43 Hz, 1 H), 7.20 - 7.26 (m, 2 H), 7.12 - 7.17 (m, 2 H), 6.95 (d, J=8.07 Hz, 1 H), 2.98 (s, 3 H), 2.48 (s, 3 H). LRMS: (M+H)+ calcd for C14Hi3N3O3S + H, 304; found, 304.
Next, to a stirred suspension of intermediate 2 (46.3g, 152.6 mmol) in 1 L CH2CI2 at 0 0C was added DIBAL (300 mL of a 1M solution in CH2CI2, 300 mmol) dropwise via an addition funnel, over 2h. After addition was complete, the reaction mixture was stirred for 2h at 0 °C. When TLC showed that the reaction was complete, 700 mL sat'd aqueous solution of Rochelle's salt was added carefully and the biphasic mixture was stirred overnight, then filtered. The aqueous layer was extracted with CH2CI2 and the combined organic layers were dried (Na2SO4), filtered, and evaporated to give a brown oil, which was purified by chromatography (0-50% EtOAc/hexanes) to provide intermediate 3 as an off-white solid (29.2g, 62%). 1H NMR of intermediate 3 (400 MHz, DMSO-Gf6) δ 10.18 (s, 1 H), 9.78 (s, 1 H), 8.19 (d, J=8.43 Hz, 1 H), 7.24 - 7.29 (m, 2 H), 7.16 - 7.21 (m, 2 H), 6.95 (d, J=8.43 Hz, 1 H), 3.02 (s, 3 H), 2.63 (s, 3 H). LRMS: (M+H)+ calcd for C14H14N2O4S + H, 307; found, 307.
The synthesis of intermediate 10
Figure imgf000015_0001
Intermediate 7
terf-butyl 4-[({[2,4-difluoro-5- (methoxycarbonylJphenyllaminofcarbonylXphenyOaminoJpiperidine-i-carboxylate
Figure imgf000016_0001
Intermediate 7 was prepared from intermediates 5 and 6 as described for intermediate 12.
1H NMR (400 MHz, DMSO-d6) δ ppm 8.06 (d, J=8.06 Hz, 1 H) 7.40 - 7.53 (m, 3 H) 7.35 (t, J=10.71 Hz, 1 H) 7.26 (dd, J=8.15, 1.37 Hz, 2 H) 7.02 (s, 1 H) 4.34 - 4.50 (m, 1 H) 3.98 (d, ,7=6.04 Hz, 2 H) 3.81 (s, 3 H) 2.74 (br. s., 2 H) 1.76 (d, J=1.47 Hz, 2 H) 1.29 (s, 9 H) 1.07 (qd, 2 H). ES-LCMS: m/z 490.4 (M+H).
Intermediate 8
5-({[[1-(fert-butoxycarbonyl)piperidin-4-yl](phenyl)amino]carbonyl}amino)-2,4- difluorobenzoic acid
Figure imgf000016_0002
Intermediate 8 was prepared using chemistry identical to that described for intermediate 13.
1H NMR (400 MHz, DWISO-dβ) δ ppm 8.03 (t, J=8.52 Hz, 1 H) 7.39 - 7.52 (m, 3 H) 7.23 - 7.34 (m, 3 H) 6.99 (s, 1 H) 4.39 (tt, J=12.00, 3.66 Hz, 1 H) 3.80 - 4.00 (m, 2 H) 2.64 (d, J=2.01 Hz, 2 H) 1.78 (d, J=10.62 Hz, 2 H) 1.29 (s, 9 H) 0.98 - 1.14 (m, J=12.50, 12.38, 12.38, 4.49 Hz, 2 H). ES-LCMS: m/z 476.6 (M+H).
Intermediate 9 tert-butyl 4-[({[5-(aminocarbonyl)-2,4- difluorophenyl]amino}carbonyl)(phenyl)amino]piperidine-1-carboxylate
Figure imgf000017_0001
Intermediate 9 was prepared in a manner identical to that described for intermediate
14.
1H NMR (400 MHz, DMSO-cfe) δ ppm 7.75 - 7.86 (m, 1 H) 7.55 - 7.64 (m, 2 H) 7.47
(d, J=7.51 Hz, 3 H) 7.25 (dd, J=6.04, 2.01 Hz, 3 H) 6.94 - 7.01 (m, 1 H) 4.30 - 4.47
(m, 1 H) 3.83 - 4.01 (m, 2 H) 2.64 - 2.85 (m, 2 H) 1.69 - 1.87 (m, 2 H) 1.29 (s, 9 H)
1.07 (m, 2 H). ES-LCMS: m/z 497.3 (M+Na).
Intermediate 10 enyl(piperidin-4-yl)amino]carbonyl}amino)benzamide
Figure imgf000017_0002
Intermediate 10 was prepared identically to chemistry described towards the intermediate 15.
1H NMR (400 MHz, DMSO-Cf6) δ ppm 7.84 (td, J=5.68, 2.75 Hz, 1 H) 7.60 (br. s., 2 H) 7.38 - 7.54 (m, 3 H) 7.17 - 7.32 (m, J=7.65, 5.06, 5.06, 2.56 Hz, 3 H) 6.86 (br. s., 1 H) 4.30 (d, J=3.11 Hz, 1 H) 3.54 (spt, J=2.59 Hz, 2 H) 2.86 (d, J=11.72 Hz, 2 H) 1.86 (br. s., 1 H) 1.67 (br. s., 2 H) 1.07 (dt, J=6.00, 2.95 Hz, 2 H)ES-LCMS: m/z 375.4 (M+H).
Example 2
2.4-Difluoro-5-(ff(3-fluorophenyl)(1-ff2-methyl-6-((4- f(methylsulfonvDaminolphenyltoxyV3-pyridinyllrnethylH~ piperidinyl)aminolcarbonyl}amino)benzamide
Figure imgf000018_0001
A solution of 2,4-difluoro-5-({[(3-fluorophenyl)(4- piperidinyl)amino]carbonyl}amino)benzamide intermediate 15 (1.0Og, 2.55 mmol, 1 equiv) and Λ/-{4-[(5-formyl-6-methyl-2-pyridinyI)oxy]phenyl}methanesulfonamide intermediate 3 (0.70 g, 2.29 mmol, 0.9 equiv) in dichloromethane (40 mL) was treated with sodium triacetoxyborohydride (1.08g, 5.10 mmol, 2 equiv) at RT. The reaction was stirred at RT for 18 h and then diluted with excess dichloromethane, washed with satd. aq. NaHCO3, dried over Na2SO4, filtered and concentrated. The residue was purified by silica gel flash column chromatography (0 — > 5% 2N NH3 in MeOH : CH2CI2) to give example 2 (945 mg; 55% yield) as a white solid. 1H NMR (400 MHz, DMSO-d6) δ ppm 9.65 (s, 1 H), 7.70 - 7.74 (m, 1 H), 7.63 (s, 2 H), 7.49 - 7.56 (m, 2 H), 7.26 - 7.32 (m, 3 H), 7.15 - 7.24 (m, 3 H)1 7.10 - 7.13 (m, 1 H), 7.04 - 7.08 (m, 1 H), 6.67 - 6.68 (m, 1 H), 4.21 - 4.29 (m, 1 H), 3.32 (s, 3 H), 2.98 (S, 3 H), 2.78 (d, J=11.17 Hz, 2 H), 2.27 (s, 3 H), 2.05 (t, J=11.08 Hz, 2 H), 1.76 - 1.80 (m, 2 H), 1.21 - 1.29 (m, 2 H). ES-LCMS: m/z 683.3 (M+H). HRMS: (M + H)+ calcd for C33H33N6O5F3S + H, 683.2264; found, 683.2251.
Synthesis of Intermediate 15
Figure imgf000018_0002
Intermediate 12
1 , 1 -dimethylethyl 4-[[({2,4-difluoro-5-[(methyloxy)carboπyl]phenyl}amino)carbonyl](3- fluorophenyl)amino]-1-piperidinecarboxylate
Figure imgf000019_0001
A solution of methyl 5-amino-2,4-difluorobenzoate intermediate 5 (5,0 g, 26.5 mmol, 1 equiv) in dichloromethane (150 mL) at -400C was treated with pyridine (6.4 mL, 79.7 mmol, 3.0 equiv) and 1.9 M phosgene in toluene (20.1 mL, 39.8 mmol, 1.5 equiv). After 1.5 h, the reaction was concentrated down and dried under vac. The residue was taken up in dichloromethane (150 mL) and 1 ,1 -dimethylethyl 4-[(3- fluorophenyl)amino]-1-piperidiπecarboxylate intermediate 11 (8.6 g, 29.2 mmol, 1.1 equiv) was added and the reaction was heated to 400C 18 h. The reaction was diluted with dichloromethane, washed with satd. aq. NaHCO3, dried over Na2SO4, filtered and concentrated. The residue was purified by silica gel flash column chromatography (0 → 30% ethyl acetate : hexanes) to give intermediate 12 (12.6 g, 94% yield) a white solid.
1H NMR (400 WIHz, CHLOROFORM-d) δ ppm 8.65 (dd, J=8.88, 7.78 Hz, 1 H), 7.44 - 7.55 (m, 1 H), 7.17 - 7.24 (m, 1 H), 7.04 (d, J=7.87 Hz, 1 H), 6.92 - 6.99 (m, 1 H), 6.80 (t, J=10.26 Hz, 1 H), 5.96 (d, J=3.11 Hz, 1 H), 4.56 - 4.70 (m, 1 H), 4.03 - 4.26 (m, 2 H), 3.80 - 3.92 (m, 3 H), 2.79 (s, 2 H), 1.85 (d, J=13.37 Hz, 2 H), 1.39 (s, 9 H), 1.19 - 1.35 (m, 2 H). ES-LCMS: m/z 530.1 (M+Na).
Intermediate 13
5-({[(HK1 , 1 -dimethylethyl)oxy]carbonylH-piperidinyl)(3- fluorophenyl)amino]carbonyl}amino)-2,4-difluorobenzoic acid
Figure imgf000019_0002
A solution of 1,1-dimethylethyl 4-[[({2,4-difluoro-5- [(methyioxy)carbonyl]phenyl}amino)carbonyl](3-fluorophenyl)amino]-1- piperidinecarboxylate intermediate 12 (12.6 g, 24.9 mmol, 1 equiv) and 1N lithium hydroxide (50 mL) in tetrahydrofuran (200 mL) was stirred at RT for 72 h. The reaction was concentrated down and then acidified to pH 3 by addition of 1 N hydrochloric acid. The aqueous layer was extracted with ethyl acetate and the organics dried over Na2SO4, filtered and concentrated to give intermediate 13 (10.6 g, 86% yield) as a white solid.
1H NMR (400 MHz, DMSO-Cf6) δ ppm 7.89 (t, J=8.42 Hz, 1 H), 7.43 - 7.52 (m, 1 H), 7.22 - 7.33 (m, 3 H), 7.13 - 7.20 (m, 1 H), 7.05 - 7.12 (m, 1 H), 4.29 - 4.40 (m, 1 H), 3.86 - 3.97 (m, 2 H), 2.72 (s, 2 H), 1.76 (s, 2 H), 1.24 - 1.32 (m, 9 H), 1.00 - 1.16 (m, 2 H). ES-LCMS: m/z 492.1 (M-H).
Intermediate 14
1 , 1 -dimethylethyl 4-[({[5-(aminocarbonyl)-2,4-difluorophenyl]amino}carbonyl)(3- peridinecarboxylate
Figure imgf000020_0001
A solution of 5-({[(1-{[(1 ,1-dimethylethyl)oxy]carbonyI}-4-piperidinyl)(3- fluorophenyl)amino]carboπyl}amino)-2,4-difluorobenzoic acid intermediate 13 (6.6, 13.3 mmol, 1 equiv) in Λ/,Λ/-dimethylformamide (100 mL) was treated with HATU (5.0 g, 13.3 mmol, 1 equiv) followed by /V,Λ/-diisopropylethylamine (4.6 mL, 36.7 mmol, 2 equiv). The reaction was stirred at RT for 15 min before the addition of ammonia hydroxide (1.3 mL, 20.0 mmol, 1.5 equiv). The reaction was stirred at RT for 4 h and then partitioned between ethyl acetate and water. The organics were washed with satd. aq. NaHCO3, dried over Na2SO4, filtered and concentrated. The residue was purified by silica gel flash column chromatography (0 → 10% 2N NH3 in MeOH : CH2CI2) to give intermediate 14 (5.8 g; 88% yield) as a tan solid. 1H NMR (400 MHz, DMSO-cfe) δ ppm 7.68 (t, ./=8.42 Hz, 1 H), 7.60 (s, 2 H), 7.44 - 7.54 (m, 1 H), 7.23 - 7.32 (m, 3 H), 7.13 - 7.19 (m, 1 H), 7.09 (dd, J=7.87, 0.92 Hz, 1
H), 4.29 - 4.42 (m, 1 H), 3.86 - 3.99 (m, 2 H), 2.79 (s, 2 H), 1.78 (d, .7=11.35 Hz, 2 H), 1.30 (s, 9 H), 1.02 - 1.16 (m, 2 H). ES-LCMS: mfz 515.3 (M+Na).
Intermediate 15 2,4-difluoro-5-({[(3-fluorophenyl)(4-piperidinyl)amino]carbonyI}amino)benzamide
Figure imgf000021_0001
A solution of 1,1-dimethylethyl 4-[({[5-(aminocarbonyl)-2,4- difluorophenyl]amino}carbonyl)(3-fluorophenyl)amino]-1-piperidiπecarboxylate intermediate 14 (5.8 g, 11.7 mmol, 1 equiv) in 4N HCI in dioxane (29.0 mL, 117.7 mmol, 10 equiv) was stirred at RT for 2 h and concentrated to a yellow solid. The solid was dissolved in water and washed with ethyl acetate. The aqueous layer was made basic (pH 8) by addition of 1 N NaOH and then extracted with dichloromethane. The organics were dried over Na2SO4, filtered and concentrated to give intermediate 15 (4.1g, 89 % yield) as a yellow solid
1H NMR (400 WIHz, DMSO-cfe) δ ppm 7.67 - 7.74 (m, 1 H), 7.60 (s, 2 H), 7.45 - 7.54 (m, 1 H), 7.24 - 7.31 (m, 2 H), 7.21 (s, 1 H), 7.05 - 7.16 (m, 2 H), 4.20 - 4.33 (m, 1 H), 3.29 (s, 1 H), 2.82 - 2.92 (m, 2 H), 2.38 - 2.50 (m, 2 H), 1.69 (dd, J=11.54, 1.65 Hz, 2 H), 1.00 - 1.14 (m, 2 H). ES-LCMS: m/z 393.3 (M+H).
Example 3
5-{r(Butyl{1-|'(2-methyl-6-ff4-fmethylsulfonyl')phenyl1oxv}-3-pyridinyl)methylH- piperidinyllamino)carbonvnamino)-2.4-difluorobenzamide
Figure imgf000021_0002
To a solution of 5-({[butyI(4-piperidinyl)arnino]carbonyl}arnino)-2,4- difluorobenzamide intermediate 22 (49 mg, 0.14 mmol) in 1 ,2-dichloroethane was
added 2-methyl-6-{[4-(methylsulfoπyl)phenyl]oxy}-3-pyridinecarbaldehyde intermediate 24 (40 mg, 0.14 mmol). Acetic acid (46 μl_, 0.80 mmol) was added, upon which a gum momentarily precipitated, but returned to solution after sonication. The reaction mixture was stirred at room temperature for 2 hours. Sodium triacetoxyborohydride was added and stirred 16 hours. The reaction mixture was diluted with dichloromethaπe and quenched with saturated aqueous sodium bicarbonate solution. The organic layer was dried over sodium sulfate. Filtration and concentration, followed by semi-preparative HPLC provided 5- {[(butyl{1-[(2-methyl-6-{[4-(methylsulfonyl)phenyl]oxy}-3-pyridinyI)methyl]-4- piperidinyl}amino)carbonyl]amino}-2,4-difluorobenzamide, example 3 (39 mg, 45%) as a white solid. 1H NMR (400 MHz, CD3OD) δ 7.96 (d, J = 8.7 Hz, 2 H), 7.86 (app t, J = 8.3 Hz, 1 H), 7.77 (d, J = 8.3 Hz, 1 H), 7.28 (d, J = 8.8 Hz, 2 H), 7.12 (app t, J = 10.3 Hz, 1 H), 6.86 (d, J = 8.2 Hz, 1 H), 4.04 (m, 1 H), 3.51 (s, 2 H), 3.26 (m, 2 H), 3.13 (s, 3 H), 2.97 (br d, J = 11.2 Hz, 2 H), 2.47 (s, 3 H), 2.18 (app td, J = 11.6, 2.0 Hz, 2 H), 1.83 (app qd, J = 12.0, 3.7 Hz, 2 H), 1.73 (m, 2 H), 1.61 (m, 2 H), 1.37 (m, 2 H), 0.96 (t, J = 7.3 Hz, 3 H); MS m/z 630 (M+H)+.
The synthesis of intermediate 22
Figure imgf000023_0001
Intermediate 16 2,4-difluoro-5-nitrobenzoic acid
F O
Nitric Acid (120 ml.) was added slowly to a cold solution of sulfuric acid (200 mL). 2,4-difluorobenzoic acid (65 g, 0.41 mol, 1 equiv) was added portioπwise over 1.5 h and the reaction allowed to warm to RT. The reaction mixture-was heated to 50 0C for 2h and then poured slowly onto ice. The product was extracted into ethyl acetate, dried over Na2SO4, filtered and concentrated to give intermediate 16 (82 g, 98% yield) as a yellow solid.
1H NMR (400 MHz, DMSO-CZ6) δ ppm 8.56 (t, J=I, .87 Hz, 1 H), 7.82 (t, ./=10.71 Hz, 1 H).
Intermediate 17 methyl 2,4-difluoro-5-nitrobenzoate
Figure imgf000024_0001
Sulfuric acid (3 mL) was added to a solution of 2,4-difluoro-5-nitrobeπzoic acid intermediate 16 (100 g, 0.49 mol, 1 equiv) in methanol (700 mL) and the reaction heated to reflux for 18 h. The reaction mixture was concentrated and the residue taken up in water and brought to pH 8 by addition of 1 N NaOH. The product was extracted into ethyl acetate, dried over Na2SO4, filtered and concentrated. The residue was purified by silica gel flash column chromatography (0 → 25% EtOAc: Hexanes) to give intermediate! 7 (90 g, 85% yield) as a tan solid. 1H NMR (400 MHz, DMSO-cfe) δ ppm 8.56 - 8.64 (m, 1 H), 7.83 - 7.94 (m, 1 H), 3.85 - 3.92 (m, 3 H).
Intermediate 5 methyl 5-amino-2,4-difluorobenzoate
Figure imgf000024_0002
Iron powder (12.3 g, 0.22 mol, 4 equiv) was added to a solution of methyl 2,4- difluoro-5-nitrobeπzoate intermediate 17 (12.0 g, 0.06 mol, 1 equiv) in acetic acid (150 mL) and water (150 mL) and the reaction was heated to 400C for 1.5 h. The reaction mixture was filtered thru a pad of celite and the filtered solution extracted with ethyl acetate. The organics were washed with satd. aq. NaCI, dried over Na- 2SO4, filtered and concentrated to give intermediate 5 (10.0g, 96% yield) as a brown solid.
1H NMR (400 MHz, DMSO-d6) δ ppm 7.27 (dd, J=9.98, 7.42 Hz, 1 H), 7.14 (t, J=10.99 Hz, 1 H), 5.28 (s, 2 H), 3.77 (s, 3 H). ES-LCMS: m/z 189.0 (M+H).
Intermediate 18
1 ,1-dimethylethyl 4-(butylamiπo)-1-piperidinecarboxylate
Figure imgf000025_0001
Sodium triacetoxyborohydride (88.5 g, 0.42 mol, 1.6 equiv) was added portionwise over 1 h to a solution of 4-BOC-piperidone (52.0 g, 0.26 mol, 1 equiv), n-butyl amine (30.9 mL, 0.31 moi, 1.2 equiv) and acetic acid (44.7 mL, 0.78 mol, 3 equiv) in dichloromethane (700 mL) at 0 0C. The reaction mixture was allowed to warm to RT over 2 h and then slowly poured into cold 3.3N NaOH (1L) with stirring. The reaction mixture was then separated and the organics dried over Na2SO4, filtered and concentrated to give intermediate 18 (66 g, 98% yield) as an oil. 1H NMR (400 MHz, CHLOROFORM-d) δ ppm 4.03 (s, 2 H), 2.77 (t, J=11.99 Hz, 2 H), 2.52 - 2.66 (m, 3 H), 1.82 (d, J=11.90 Hz, 2 H), 1.39 - 1.52 (m, 11 H), 1.29 - 1.39 (m, 2 H), 1.16 - 1.29 (m, 2 H), 1.06 (s, 1 H), 0.84 - 0.96 (m, 3 H).
Intermediate 19
1 ,1-dimethylethyl 4-{butyl[({2,4-difluoro-5-
[(methyloxy)carbonyl]phenyl}amino)carbonyl]amino}-1-piperidinecarboxylate
Figure imgf000025_0002
A solution of methyl 5-amino-2,4-difluorobenzoate intermediate 5 (5.5 g, 29.5 mmol, 1 equiv) in dichloromethane (100 mL) at -400C was treated with pyridine (7.16 mL, 88.6 mmol, 3.0 equiv) and 1.9 M phosgene in toluene (23.3 mL, 44.3 mmol, 1.5 equiv). After 1.5 h, a solution of 1 ,1-dimethylethyl 4-(butylamino)-1- piperidinecarboxylate intermediate 18 (9.1 g, 35.4 mmol, 1.2 equiv) in dichloromethane (50 mL) was added and the reaction was allowed to warm to RT for 18 h. The reaction was diluted with dichloromethane, washed with satd. aq. NaHCO3, dried over Na24, filtered and concentrated to give intermediate 19 (13.1 g, 95% yield) a white solid.
1H NMR (400 MHz, DMSO-Cf6) δ ppm 8.09 (s, 1 H), 7.89 - 7.99 (m, 1 H), 7.42 (t, J=10.62 Hz, 1 H), 3.88 - 3.99 (m, 1 H), 3.85 (s, 3 H), 3.11 - 3.22 (m, 2 H), 2.96 (d,
J=12.27 Hz, 2 H), 2.41 - 2.53 (m, 3 H), 1.43 - 1.59 (m, 5 H), 1.21 - 1.35 (m, 2 H), 1.20 (s, 9 H), 0.90 (t, J=7.33 Hz, 3 H). ES-LCMS: m/z 468.1 (M-H).
Intermediate 20 δ-KIbutyKI-IKI .I-dimethylethyOoxylcarbonylH-piperidinyOaminolcarbonyiyamino)- 2,4-difluorobenzoic acid
Figure imgf000026_0001
A solution of 1,1-dimethylethyl 4-{butyl[({2,4-difluoro-5-
[(methyloxyJcarbonyllphenylJaminoJcarbonyllaminoM-piperidinecarboxylate intermediate 19 (13.Og, 27.7 mmol, 1 equiv) and 1N lithium hydroxide (15 mL) in tetrahydrofuran (30 mL) was stirred at RT for 2 h. The reaction was concentrated down and then acidified to pH 2 by addition of 1N hydrochloric acid. The aqueous layer was extracted with ethyl acetate and the organics dried over Na2SO4, filtered and concentrated to give intermediate 20 (12.0 g, 95% yield) as a white solid. 1H NMR (400 MHz, DMSOd6) δ ppm 8.09 (s, 1 H), 7.81 - 7.91 (m, 1 H), 7.29 - 7.38 (m, 1 H), 3.93 - 4.07 (m, 1 H), 3.24 - 3.38 (m, 2 H), 3.09 - 3.20 (m, 2 H), 2.72 (d, J=6.97 Hz, 2 H), 1.52 - 1.65 (m, 4 H), 1.42 - 1.51 (m, 2 H), 1.34 - 1.41 (m, 9 H), 1.19 - 1.33 (m, 2 H), 0.87 (t, J=7.33 Hz, 3 H). ES-LCMS: m/z 454.43 (M-H).
Intermediate 21
1 ,1-dimethylethyl 4-[({[5-(aminocarbonyl)-2,4- difluorophenyl]amino}carbonyl)(butyl)amino]-1-piperidinecarboxylate
Figure imgf000026_0002
A solution of 5-({[butyl(1-{[(1 ,1-dimethylethyl)oxy]carbonyl}-4- piperidinyl)amino]carbonyl}amino)-2,4-difluorobenzoic acid intermediate 20
(12.0, 26.3 mmol, 1 equiv) in Λ/,W-dimethylformamide (100 mL) was treated with HATU (10.0 g, 26.3 mmol, 1 equiv) followed by W,Λ/-diisopropylethylamine (9.2mL, 52.6 mmol, 2 equiv). The reaction was stirred at RT for 15 min before the addition of ammonia hydroxide (2.6 mL, 39.4 mmol, 1.5 equiv). The reaction was stirred at RT for 2 h and then partitioned between ethyl acetate and water. The organics were washed with satd. aq. NaHCO3, dried over Na2SO4, filtered and concentrated. The residue was purified by silica gel flash column chromatography (0 -+ 5% 2N NH3 in MeOH : CH2CI2) to give intermediate 21 (10.1 g; 80% yield) as a white solid. 1H NMR (400 MHz, DMSO-cfe) δ ppm 8.04 (s, 1 H), 7.55 - 7.70 (m, 3 H), 7.29 (t, J=10.35 Hz, 1 H), 3.98 (s, 3 H), 3.24 - 3.29 (m, 1 H), 3.03 - 3.19 (m, 2 H), 2.57 - 2.79 (m, 2 H), 1.50 - 1.65 (m, 3 H), 1.45 (s, 2 H), 1.37 (s, 9 H), 1.16 - 1.30 (m, 2 H), 0.86 (t, J=7.23 Hz, 3 H). ES-LCMS: m/z 453.4 (M-H).
Intermediate 22 5-({[butyl(4-piperidinyl)amino]carbonyl}amino)-2,4-difluorobeπzamide
Figure imgf000027_0001
A solution of 1 ,1-dimethylethyl 4-[({[5-(aminocarbonyl)-2,4- difluorophenyl]amino}carbonyl)(butyl)amino]-1-piperidinecarboxylate intermediate 21 (3.0 g, 6.60 mmol, 1 equiv) in 4N HCI in dioxane (16.5 mL, 66.0 mol, 10 equiv) was stirred at RT for 2 h and concentrated to a yellow solid. The solid was dissolved In water and washed with ethyl acetate. The aqueous layer was made basic (pH 8) by addition of 1N NaOH and then extracted with dichloromethane. The organics were dried over Na2SO4, filtered and concentrated to give intermediate 22 (1.9g, 81% yield) as a yellow solid.
1H NMR (400 MHz, DMSO-cfe) S ppm 8.00 (s, 1 H), 7.58 - 7.72 (m, 3 H), 7.30 (t, J=10.45 Hz, 1 H), 3.86 - 3.97 (m, 1 H), 3.57 - 3.62 (m, 1 H), 3.30 (s, 2 H), 3.10 - 3.19 (m, 2 H), 2.90 - 3.00 (m, 2 H), 2.39 - 2.47 (m, 1 H), 1.69 - 1.79 (m, 1 H), 1.41 - 1.58 (m, 4 H), 1.20 - 1.34 (m, 2 H), 0.88 (t, J=7.33 Hz, 3 H). ES-LCMS: m/z 355.1 (M+H).
Synthesis of intermediate 24
Figure imgf000028_0001
24
Intermediate 23: 2-methyl-6-{[4-(methylsulfonyl)phenyl]oxy}-3-pyrιdιπecarbonιtrιle
Figure imgf000028_0002
To a stirred solution of δ-chloro-Z-methyl-S-pyπdinecarbonitπle intermediate 1 (1 00 g, 6 56 mmol) in /V,Λ/-dιmethylformamιde (10 mL) was added 4- (methylsulfonyl)phenol (1 24 g, 721 mmol) and cesium carbonate (427 g, 13 1 mmol) The reaction mixture was stirred at room temperature for 16 hours The reaction mixture was filtered and the filtrate was diluted with toluene The organic layer was washed with water and dried over sodium sulfate The organic layer was filtered and concentrated in vacuo to remove the majority of the toluene Ether was added and the resulting solution was placed in a freezer for 3 days The precipitated solids were collected on a filter to provide 2-methyl-6-{[4- (methylsulfonyl)phenyl]oxy}-3-pyrιdιnecarbonιtrιle intermediate 23 (1 47 g, 78 %) as a light brown solid 1H NMR (400 MHz, DMSO-Cf6) δ 829 (d, J = 86 Hz, 1 H), 7 98 (d, J = 8 6 Hz, 2 H), 745 (d, J = 8 5 Hz, 2 H), 7 14 (d, J = 8 5 Hz, 1 H), 3 25 (s, 3 H), 249 (s, 3 H), MS m/z 289 (M+H)+ lntemediate 24: 2-methyl-6-{[4-(methylsulfonyl)phenyl]oxy)-3-pyrιdinecarbaldehyde
Figure imgf000028_0003
To a cold (-35 0C) solution of 2-methyl-6-([4-(methylsulfonyl)phenyl]oxy}-3- pyπdinecarboπitπle intermediate 23 (900 mg, 3 12 mmol) in dichloromethane (20
mL) was added diisobutylaluminum hydride (9.88 mL, 1.0 Wl solution in dichloromethane, 9.88 mmol) dropwise, maintaining the cooling bath temperature between -40 °C and -35 °C. The reaction mixture was stirred at -35 °C for an additional 20 minutes. The reaction mixture was quenched with methanol (700 μL), added dropwise, followed by saturated aqueous citric acid solution. The reaction mixture was stirred at room temperature for 6 hours, then diluted with dichloromethane. The organic layer was separated, washed with water, washed with saturated aqueous sodium bicarbonate solution, then dried over sodium sulfate. Filtration and concentration provided 2-methyl-6-{[4-(methylsulfonyl)phenyl]oxy}-3- pyridinecarbaldehyde intermediate 24 (769 mg, 85 %) as a tan solid. 1H NMR (400 MHz, CD3OD) δ 10.24 (s, 1 H)1 8.25 (d, J = 8.4 Hz, 1 H), 8.02 (d, J = 8.6 Hz, 2 H), 7.42 (d, J = 8.7 Hz, 2 H), 7.04 (d, J = 8.3 Hz, 1 H), 3.16 (s, 3 H), 2.67 (s, 3 H); MS m/z 292 (M+H)*.
Example 4
2.4-Difluoro-5-(ff(3-fluorophenyl)(1-ff6-({4-r(2-hvdroxyethyl)sulfonyllphenylloxyV2- methyl-3-pyridinvnmethyl)-4-piperidinyl)aminolcarbonyl)amino)beπzamide
Figure imgf000029_0001
A mixture of 6-({4-[(2-hydroxyethyl)sulfoπyl]pheπyl}oxy)-2-methyl-3- pyridinecarbaldehyde 27 (32.1 mg, 0.10 mmol, 1.0 eq) and intermediate 15 2,4- difluoro-5-({t(3-fluorophenyl)(4-piperidinyl)amino]carbonyl}amiπo)benzamide (39.2 mg; 0.10 mmol, 1.0 eq) and NaBH(OAc)3 (42.4 mg; 0.30 mmol, 2.0 eq) in 1 ,2- dichloroethane (6.0 mL) was stirred for 24 h at RT. The mixture was diluted with excess DCM and washed with saturated aq. NaHCO3. Organics were separated and aqueous layer extracted with 10 % MeOH in DCM. Organics were then combined, dried over Na2SO4, filtered and concentrated down. Residue was purified by preparative TLC (5 % MeOH(w/ 2M NH3) in CH2CI2) to give 2,4-difluoro-5-({[(3- fluoropheπyl)(1-{[6-({4-[(2-hydroxyethyl)sulfonyl]phenyl}oxy)-2-methyl-3-
pyridinyl]methyl3-4-pipβridiπyl)amino]carboπyl}amino)beπzamide, example 4 (52 mg, 75 % yield). 1H NMR (CDCI3, 400 MHz) δ 8.65 (t, J=8.62 Hz, 1 H), 7.89 (d, J=8.43 Hz, 2 H), 7.60 (d, J=8.07 Hz, 1 H), 7.46 - 7.53 (m, 1 H), 7.24 - 7.28 (m, 3 H), 7.18 - 7.24 (m, 1 H), 7.06 (d, J=8.80 Hz, 1 H), 6.96 - 7.01 (m, 1 H), 6.83 (t, J=10.45 Hz, 1 H), 6.74 (d, J=8.43 Hz, 1 H), 6.51 (br. s., 1 H), 5.89 (d, J=2.57 Hz, 1 H), 5.77 (s, 1 H), 4.49 - 4.60 (m, 1 H), 3.98 - 4.05 (m, 2 H), 3.40 (s, 2 H), 3.33 - 3.38 (m, 2 H), 2.87 (d, .7=11.73 Hz, 2 H), 2.41 (s, 3 H), 2.12 - 2.23 (m, 2 H), 1.85 (s, 2 H), 1.35 - 1.49 (m, 2 H). HRMS: (M+H)+ calcd for C34H34F3N5O6S + H, 698.2260; found, 698.2255.
Synthesis of intermediate 27
Figure imgf000030_0001
42 % intermediate 26 intermediate 1 intermediate 25
Figure imgf000030_0002
22 % intermediate 27
4-[(2-hydroxyethyl)sulfonyl]phenol 25
Figure imgf000030_0003
The synthesis of 4-[(2-hydroxyethyl)sulfonyl]phenol 25 was accomplished as described in [Waldmeier et al., (2003) Organic Process Research & Development. pp418-425].
1H NMR (DMSO-D6, 400 MHz) δ 3 3 (m, 2 H) 36 (m, 2 H) 4 8 (t, J=A 7 Hz, 1 H) 6 9 (dd, J=87, 2 3 Hz, 2 H) 7 6 (dd, J=8 7, 2 3 Hz, 2 H) 10 5 (s, 1 H) LCMS (M+Hf calcd for C8H10O4S+H, 203, found, 203
6-({4-[(2-hydroxyethyI)sulfonyl]phenyl}oxy)-2-methyl-3-pyπdinecarbonιtπle 26
Figure imgf000031_0001
Synthesized from 4-[(2-hydroxyethyl)sulfonyl]phenoI using chemistry described for intermediate 2 1H NMR (CDCI3, 400 MHz) δ 2 6 (s, 3 H) 34 (m, 2 H) 4 1 (m, 2 H) 6 9 (d, J=B 4 Hz, 1 H) 74 (d, J=84 Hz 2 H) 7 9 (d, J=84 Hz, 1 H) 8 0 (d, J=8 8 Hz, 2 H) LCMS (M+Hf calcd for C15H14N2O4SH-H, 319, found, 319
6-({4-[(2-hydroxyethyl)sulfonyi]phenyl}oxy)-2-methyl-3-pyπdιnecarbaldehyde 27
Figure imgf000031_0002
Synthesized from 6-({4-[(2-hydroxyethyl)sulfonyl]phenyI}oxy)-2-methyl-3- pyπdinecarbonitrile 26 using chemistry described for intermediate 3 1H NMR (CDCI3, 400 MHz) δ 2 7 (s, 3 H) 34 (m, 2 H) 4 1 (m, 2 H) 6 9 (d, J=84 Hz, 1 H) 74 (m, 2 H) 8 0 (d, J=B 4 Hz, 2 H) 82 (d, J=B 4 Hz, 1 H) 10 3 (s, 1 H) LCMS (M+H)+ calcd for C15H15NO5S+H, 319, found, 319
Example 5
2,4-Dιfluoro-5-f(rπ-(r2-methyl-6-(f4-r(methylsulfonyl)amιno1phenyl>oxy)-3- pyπdιnvπmethylM-piperιdιnvh(3-thιenyDamιno1carbonyltømιno)beπzamιde
Figure imgf000032_0001
Example 5 was prepared using the chemistry described for example 2 1H NMR (400 MHz, DMSO-cfe) δ ppm 7 86 - 7 92 (m, 1 H), 7 80 (t, J=I 56 Hz, 1 H), 7 66 (t, J=I 69 Hz, 2 H), 7 56 - 763 (m, 2 H), 741 (s, 1 H), 7 07 (t, J=10 16 Hz, 1 H), 4 33 - 445 (m, 1 H), 3 90 - 3 98 (m, 2 H), 3 50 (s, 1 H), 276 (s, 2 H), 1 99 (d, J=2 01 Hz, 1 H), 1 83 (d, J=10 80 Hz, 2 H), 1 47 - 1 58 (m, 1 H), 1 36 - 1 42 (m, 1 H), 1 28 - 1 35 (m, 9 H), 0 99 - 1 14 (m, 3 H), 0 84 - 0 94 (m, 1 H) ES-LCMS m/z 671 5 (M+H) HRMS (Wl + H)* calcd for C31H32N6O5F2 S2 + H, 671 1922, found, 671 1911
Synthesis of intermediate 31
Figure imgf000032_0002
Intermediate 29
1 , 1 -dimethylethyl 4-[[({2,4-dιfluoro-5-[(methyloxy)carbonyl]phenyl}amιno)carbonyl](3- thιenyl)amιno]-1-pιperιdιnecarboxylate
Figure imgf000033_0001
Intermediate 29 was prepared using chemistry described for the intermediate 12 from intermediates 5 and 28. Intermediate 281 ,1-dimethylethyl 4-(3-thienylamino)-1- piperidinecarboxylate was synthesized using chemistry described for the intermediate 11. 1H NMR (400 MHz, DMSO-Cf6) δ ppm 8.03 - 8.18 (m, 1 H), 7.66 (dd, J=5.13, 3.11 Hz, 1 H), 7.59 (dd, J=3.11 , 1.47 Hz, 1 H), 7.37 (t, J=10.71 Hz, 1 H), 7.17 (s, 1 H), 7.00 (dd, J=5.04, 1.37 Hz, 1 H), 4.30 - 4.43 (m, 1 H), 3.87 - 4.00 (m, 1 H), 3.78 - 3.84 (m, 3 H), 2.74 (s, 2 H), 1.65 - 1.79 (m, 2 H), 1.34 - 1.39 (m, 1 H), 1.24 - 1.33 (m, 9 H), 0.99 - 1.15 (m, 2 H). ES-LCWIS: m/z 518.3 (M+Na)
Intermediate 30
5-({[(1-{[(1,1-dimethylethyl)oxy]carbonyl}-4-pipβridinyl)(3- thienyOaminolcarbonylJamino^^-difluorobenzoic acid
Figure imgf000033_0002
Intermediate 30 was prepared from intermediate 29 using chemistry described for intermediate 13. 1H NMR (400 MHz, DMSO-cfe) δ ppm 7.96 - 8.13 (m, 1 H), 7.65 (dd, J=5.13, 3.11 Hz, 1 H), 7.58 (dd, J=3.11 , 1.46 Hz, 1 H), 7.31 (t, J=10.62 Hz, 1 H), 7.14 (S, 1 H), 7.00 (dd, J=5.04, 1.37 Hz, 1 H), 4.25 - 4.43 (m, 1 H), 3.96 (d, J=5.68 Hz, 1 H), 2.74 (s, 2 H), 1.74 (dd, J=11.90, 1.83 Hz, 2 H), 1.37 (s, 1 H), 1.25 - 1.34 (m, 9 H), 1.00 - 1.15 (m, 2 H). ES-LCMS: m/z 504.3 (M+Na).
Intermediate 31
2,4-difluoro-5-({[4-pipericIinyl(3-thienyl)amino]carbonyl}amino)benzamide
Figure imgf000034_0001
Intermediate 31 was prepared from intermediate 30 using chemistry described for intermediates 14 and 15. 1H NMR (400 MHz, DMSO-αfe) δ ppm 7.89 (dd, J=8.88, 7.97 Hz, 1 H), 7.65 (dd, J=5.13, 3.11 Hz, 1 H), 7.60 (s, 1 H), 7.54 (dd, J=3.11 , 1.47 Hz, 1 H), 7.27 (t, J=10.44 Hz, 1 H), 7.02 (s, 1 H), 6.98 (dd, J=5.04, 1.37 Hz, 1 H), 4.17 - 4.31 (m, 1 H), 3.54 (S1 1 H), 2.81 - 2.93 (m, 2 H), 2.39 - 2.47 (m, 2 H), 1.83 (s, 1 H), 1.64 (dd, J=11.54, 1.83 Hz, 2 H), 1.01 - 1.19 (m, 2 H). ES-LCMS: m/z 381.9 (M+H).
Example 6
2.4-Difluoro-5-»f(3-fluorophenyl)(14r2-methyl-6-α4- r(methylsulfonyl)aminolphenyltoxy)-3-pyridinyl1rnethyl}-4- piperidinylteminolcarbonyl}amino)-N-n-rnethylethvDbenzarriide
Figure imgf000034_0002
A solution of 2,4-difluoro-5-({[(3-fluorophenyl)(1-{[2-methyl-6-({4- [(methylsulfoπyl)amino]phenyl}oxy)-3-pyridinyl]methyl}-4- piperidiπyl)amino]carbonyl}amino)benzoic acid intermediate 35 (100 mg, 0.15 mmol, 1 equiv) in N,Λ/-dimethylformamide (3 mL) was treated with HATU (55.6 mg, 0.15 mmol, 1 equiv) followed by Λ/,W-diisopropylethylamine (50.9 mL, 0.29 mmol, 2 equiv). The reaction was stirred at RT for 5 min before the addition of isopropyl amine (18.6 mL, 0.22 mmol, 1.5 equiv). The reaction was stirred at RT for 4 h and
then partitioned between ethyl acetate and water The organics were washed with satd aq NaHCO3, dπed over Na2SO4, filtered and concentrated The residue was purified by silica gel flash column chromatography (O → 4% 2N NH3 in MeOH CH2CI2) to give example 6 (50 mg, 45% yield) as a white solid 1H NMR (400 MHz, CHLOROFORM-d) δ ppm 847 - 8 51 (m, 1 H), 746 - 7 53 (m, 2 H), 7 18 - 7 27 (m, 3 H), 709 - 7 13 (m, 2 H), 7 07 (s, 1 H), 6 97 - 7 01 (m, 1 H), 6 80 (t, J=10 44 Hz, 1 H), 6 58 (d, J=B 24 Hz, 1 H), 6 29 - 629 (m, 1 H), 5 86 (s, 1 H), 452 - 454 (m, 1 H), 423 - 4 32 (m, 1 H), 3 38 (s, 1 H), 3 02 (s, 3 H), 2 87 - 2 87 (m, 2 H), 241 (s, 3 H), 2 17 (d, J=8 61 Hz, 2 H), 1 83 - 1 88 (m, 2 H), 1 60 - 1 62 (m, 2 H), 1 38 - 1 46 (m, 2 H), 1 26 (s, 3 H), 1 24 (s, 3 H) ES-LCMS m/z 7252 (M+H) HRMS (M + H)+ calcd for C36H39N6O5F3S + H, 7252733, found, 725 2737 Synthesis of intermediate 35
Figure imgf000035_0001
Intermediate 11
1 , 1 -climethylethyl 4-[(3-fluorophenyl)amino]-1 -pipeπdmecarboxylate
Figure imgf000035_0002
Sodium tπacetoxyborohydπde (79 7 g, 0 37 mol, 1 5 equiv) was added portionwise over 1 h to a solution of 4-BOC-pιpendone (50 0 g, 0 25 mol, 1 equiv), 3- fluoroaniline (28 9 mL, 0 30 mol, 1 2 equiv) and acetic acid (86 2 mL, 1 51 mol, 6 equiv) in dichloroethane ( 700 mL) at 0 0C The reaction mixture was allowed to
warm to RT over 2 h and then slowly poured into cold 3 3N NaOH (1L) with stirπng The reaction mixture was then separated and the organics dned over Na2SO4, filtered and concentrated to give a tan solid The solid was suspended in 3 1 hexanes ethyl acetate, stirred overnight and filtered to give 11 (67 g, 91% yield) as a white solid
1 H NMR (400 MHz, DMSO-cfe) δ ppm 6 96 - 7 05 (m, 1 H), 6 29 - 640 (m, 2 H), 6 18 - 6 25 (m, 1 H), 5 80 (d, J=824 Hz, 1 H), 3 81 (s, 2 H), 3 30 - 342 (m, 1 H), 287 (s, 2 H), 1 81 (s, 2 H), 1 32 - 1 42 (m, 9 H), 1 09 - 1 24 (m, 2 H)
Intermediate 32 W-(3-fluorophenyl)-4-pιpeπdιnamιne
Figure imgf000036_0001
4N HCI in dioxane (12 7 mL, 50 9 mmol, 10 equiv) was added to a solution of 1 ,1- dimethylethyl 4-[(3-fluorophenyl)amino]-1-pιperιdιnecarboxylate intermediate 11 (1 5 g, 5 1 mmol, 1 equiv) in dichloromethane (10 mL) and stirred at RT for 3 h The reaction mixture was concentrated and then dissolved in water and brought to pH 8 by addition of 1N NaOH The aqueous layer was extracted with dichloromethane and the organics were dried over Na2SO4, filtered and concentrated to give intermediate 32 (740 mg, 75% yield) as a white solid
1H NMR (400 MHz, CHLOROFORM-cQ δ ppm 6 97 - 7 16 (m, 1 H), 6 17 - 6 45 (m, 3 H), 3 53 - 3 78 (m, 1 H), 3 21 - 340 (m, 1 H), 3 01 - 3 19 (m, 2 H), 2 58 - 2 81 (m, 2 H), 1 92 - 2 14 (m, 2 H), 1 52 (s, 1 H), 1 17 - 1 41 (m, 2 H) ES-LCMS m/z 195 0 (M+H)
Intermediate 33
N-(4-{[5-({4-[(3-fluorophenyl)amιno]-1-pιperιdιnyl}methyl)-6-rnethyl-2- pyπdιnyl]oxy}phenyl)methanesulfonamιde
Figure imgf000036_0002
Sodium triacetoxyborohydride (1.2 g, 5.7 mmol, 1.5 equiv) was added portionwise over 30 min to a solution of Λ/-(3-fluorophenyI)-4-piperidiπamine 32 (740 mg, 3.8 mmol, 1 equiv), Λ/-{4-[(5-formyl-6-methyl-2- pyridinyl)oxy]pheπyl}methanesulfonamide 3 (1.1 g, 3.81 mmol, 1 equiv) and acetic acid (0.4 mL, 7.6 mmol, 2 equiv) in dichloroethane (20 mL) at 0 0C. The reaction mixture was allowed to warm to RT over 2 h and then slowly poured into cold 3.3N NaOH (100 mL) with stirring. The reaction mixture was then separated and the organics dried over Na2SO4, filtered and concentrated to give a tan residue. The residue was purified by silica gel flash column chromatography (0 — s- 5% 2N NH3 in MeOH : CH2CI2) to give the intermediate 33 (1.2 g; 67% yield) as a tan solid. 1H NMR (400 WIHz, DMSO-cfe) δ ppm 9.63 (s, 1 H), 7.59 (d, J=8.24 Hz, 1 H), 7.14 - 7.24 (m, 1 H), 7.03 - 7.10 (m, 2 H), 6.95 - 7.03 (m, 1 H), 6.68 (d, J=8.24 Hz, 1 H), 6.35 (d, J=8.06 Hz, 2 H), 6.22 (s, 1 H), 5.66 - 5.82 (m, 1 H), 3.29 (d, J=1.10 Hz, 2 H), 3.18 (s, 1 H), 2.95 (d, J=0.92 Hz, 3 H), 2.70 (s, 2 H), 2.32 (s, 3 H), 2.08 (s, 2 H), 1.82 (S1 2 H), 1.32 (s, 2 H). ES-LCMS: m/z 485.1 (M+H).
Intermediate 34 methyl 2,4-difluoro-5-({[(3-fluorophenyl)(1 -{[2-ππethyl-6-({4-
[(methylsulfonyl)amino]phenyl}oxy)-3-pyridinyl]methyl}-4- piperidinyl)amino]carbonyl}amino)benzoate
Figure imgf000037_0001
A solution of methyl 5-amino-2,4-difluorobenzoate intermediate 5 (400 mg g, 2.1 mmol, 1 equiv) in dichloromethane (30 mL) at -400C was treated with N,N- diisopropylethylamine (0.89 mL, 5.1 mmol, 2.4 equiv) and 1.9 M phosgene in toluene (1.3 mL, 2.5 mmol, 1.2 equiv). After 1.5 h, the reaction was concentrated down and dried under vac. The residue was taken up in dichloromethane (30 mL) and Λ/-(4-{[5-({4-[(3-fluorophenyl)amino]-1-piperidinyl}methyl)-6-methyl-2- pyridinyl]oxy}phenyl)methanesulfonamide intermediate 33 (1.0 g, 2.13 mmol, 1 equiv) was added and the reaction was stirred at RT for 18 h. The reaction was
diluted with dichloromethane, washed with satd. aq. NaHCO3, dried over Na2SO4, filtered and concentrated. The residue was purified by silica gel flash column chromatography (O → 5% 2N NH3 in MeOH : CH2CI2) to give intermediate 34 (675 mg, 46% yield) as a white solid.
1H NWIR (400 MHz, DMSO-cfe) δ ppm 9.61 (s, 1 H), 7.95 (t, J=8.15 Hz, 1 H), 7.45 - 7.56 (m, 2 H), 7.32 - 7.40 (m, 1 H), 7 24 - 7.31 (m, 2 H), 7.17 (t, J=8.88 Hz, 3 H), 7.09 (d, J=7.87 Hz, 1 H), 6.99 - 7.06 (m, 2 H), 6.64 (d, J=8.24 Hz, 1 H), 4.22 (s, 1 H), 3.80 (s, 3 H), 3.31 (s, 2 H), 2.94 (d, J=0.73 Hz, 3 H), 2.74 (s, 2 H), 2.24 (s, 3 H), 1.92 - 2.09 (m, 2 H), 1.73 (s, 2 H), 1.11 - 1.29 (m, 2 H). ES-LCMS: m/z 698.2 (M+H).
Intermediate 35
2,4-difluoro-5-({[(3-fluorophenyl)(1-{[2-methyI-6-({4- [(methylsulfonyl)amino]phenyl}oxy)-3-pyridinyl]methyl}-4- piperidinyl)amino]carbonyl}amino)benzoic acid
Figure imgf000038_0001
Intermediate 35 was prepared from intermediate 34 using chemistry described for the intermediate 13
1H NMR (400 MHz, DMSO-dβ) δ ppm 9.70 (s, 1 H), 7.75 - 7.92 (m, 1 H), 7.47 - 7.58 (m, 1 H), 7.40 - 7.44 (m, 1 H), 7.27 - 7.36 (m, 2 H), 7.18 - 7.26 (m, 3 H), 7.13 (d, J=7.69 Hz, 1 H), 7.03 - 7.10 (m, 2 H), 6.80 (s, 1 H), 4.38 - 4.62 (m, 1 H), 4.20 (s, 2 H), 3.03 - 3.43 (m, 6 H), 2.91 - 3.01 (m, 3 H), 2.23 - 2.41 (m, 3 H), 1.86 - 2.07 (m, 2 H), 1.58 (s, 2 H). ES-LCMS: m/z 684.2 (M+H).
Example 7
5-αr(3-Fluorophenyl)f1-{r2-methyl-6-(f4-[(methylsulfonyl)aminolphenyl>oxy)-3- ρyridinvnmethylV4-piperidinvπamino1carbonyltamino)-2-(trifluoromethyl)benzamide
Figure imgf000039_0001
A solution of 5-({[(3-fluorophenyl)(4-piperidinyl)amino]carbonyl}amino)-2- (trifluoromethyl)benzamide hydrochloride 39 (368 mg, 0.8 mmoi, 1 equiv) and Λ/-{4- [(5-formyl-6-methyl-2-pyridinyl)oxy]phenyl}methanesulfonamide intermediate 3 (232 mg, 0.76 mmol, 0.95 equiv) in dichloromethane (10 mL) was treated with triethylamine (122 mL, 0.88 mmol, 1.1 equiv) followed by sodium triacetoxyborohydride (1.08g, 5.10 mmol, 2 equiv) at RT. The reaction was stirred at RT for 18 h and then diluted with excess dichloromethane, washed with satd. aq. NaHCO3, dried over Na2SO4, filtered and concentrated. The residue was purified by silica gel flash column chromatography (0 → 5% 2N NH3 in MeOH : CH2CI2) to give example 7 (306 mg; 54% yield) as a white solid.
1H NMR (400 MHz, DMS0-dβ) δ ppm 9.65 (s, 1 H), 7.97 (s, 1 H), 7.80 (s, 1 H), 7.72 (d, J=8.42 Hz, 1 H), 7.47 - 7.58 (m, 4 H), 7.30 (t, J=7.87 Hz, 1 H)1 7.21 (d, J=8.79 Hz, 2 H), 7.16 (s, 1 H), 7.06 (d, J=8.06 Hz, 3 H), 6.68 (d, J=8.06 Hz, 1 H), 4.26 - 4.32 (m, 1 H), 3.29 - 3.35 (m, 3 H), 2.98 (s, 3 H), 2.78 (d, J=6.59 Hz, 2 H), 2.27 (s, 3 H), 2.07 (t, J=11.17 Hz, 2 H), 1.78 (d, J=14.47 Hz, 2 H), 1.17 - 1.31 (m, 2 H). ES- LCMS: m/z 715.3 (M+H). HRMS: (M + Hf calcd for C34H34N6O5F4S + H, 715.2326; found, 715.2317.
Preparation of intermediate 39
Figure imgf000040_0001
Intermediate 36 5-amino-2-(trifluoromethyl)benzonitrile
Figure imgf000040_0002
A solution of 5-fluoro-2-(trifluoromethyI)benzonitrile (7.Og, 37.0 mmol, 1 equiv) and 1 ,4-dioxane (10 mL) in ammonia hydroxide (100 mL) 1 ,4-dioxane (10 mL) was heated to 120 0C in Parr bomb under 200 psi for 18 h. The reaction mixture was concentrated down and the residue partitioned between dichloromethane and water. The organics were dried over Na2SO4, filtered and concentrated and the residue purified by silica gel flash column chromatography (0 → 40% ethyl acetate : hexanes) to give intermediate 36 (3.5 g, 52% yield) as a white solid. 1H NMR (400 MHz, DMSOd6) δ ppm 7.49 (d, J=8.79 Hz, 1 H), 7.02 (d, J=2.20 Hz, 1 H), 6.77 - 6.91 (m, 1 H), 6.32 (s, 2 H). ES-LCMS: m/z 187.2 (M+H).
Intermediate 37
1 , 1 -dimethylethyl 4-[({[3-cyano-4-(trifluoromethyl)phenyl]amino}carbonyl)(3- fluorophenyl)amino]-1-piperidinecarboxylate
Figure imgf000041_0001
Preparation of intermediate 37 from intermediates 36 and 11 was performed as descπbed for intermediate 12.
1H NMR (400 MHz, DMSOd6) δ ppm 8 18 - 8 23 (m, 1 H), 8 12 (d, J=Z 01 Hz, 1 H), 7 89 - 7 97 (m, 1 H), 7 75 - 7 84 (m, 1 H), 741 - 7 53 (m, 1 H), 7 23 - 7 32 (m, 1 H), 7 13 - 7 21 (m, 1 H), 7 03 - 7 10 (m, 1 H), 4 30 - 446 (m, 1 H), 3 96 (d, J=330 Hz, 2 H), 273 (s, 2 H), 1 76 (s, 2 H), 1 23 - 1 33 (m, 9 H), 1 00 - 1 18 (m, 2 H) ES-LCMS m/z 5054 (M-H)
Intermediate 38
1 ,1-dιmethylethyl 4-[({[3-(amιnocarbonyl)-4-
(trιfluoromethyl)phenyl]amιno}carbonyl)(3-fluorophenyl)amιno]-1- pipeπdinecarboxylate
Figure imgf000041_0002
Urea hydrogen peroxide (2 7 g, 28 9 mmol, 24 equiv) and potassium carbonate (60 mg, 04 mmol, 0 36 equiv) were added to a solution of 1 ,1-dιmethylethyl 4-[({[3- cyano^trifiuoromethylJphenylJaminoJcarbonylHS-fluorophenyOarninol-i- pipeπdinecarboxylate intermediate 37 (610 mg, 1 21 mmo, 1 equiv) in acetone (10 mL) and water (5 mL) and the reaction was stirred at RT for 72 h The reaction mixture was concentrated and the residue dissolved in dichloromethane and washed with water The orgaπics were dried over Na2SO4, filtered and concentrated and the residue purified by silica gel flash column chromatography (0 → 70% ethyl acetate hexaπes) to give intermediate 28(421 mg, 67% yield) as a white solid 1H NMR (400 MHz, DMSO-Cf6) δ ppm 8 82 (s, 1 H), 8 19 (s, 1 H), 8 05 (s, 1 H), 7 76 (s, 1 H), 765 - 772 (m, 1 H), 741 - 7 58 (m, 2 H), 7 24 - 7 34 (m, 1 H), 7 19 (d,
J=9.89 Hz, 1 H), 7.08 (s, 1 H), 4.51 (s, 1 H), 3.21 (s, 2 H), 2.97 (s, 2 H), 1.78 - 2.03 (m, 2 H), 1.23 (s, 9 H), 1.29 - 1.59 (m, 2 H). ES-LCMS: m/z 523.4 (M-H).
Intermediate 39
5-({[(3-fluorophenyl)(4-piperidinyl)amino]carbonyl}amino)-2- mide hydrochloride
Figure imgf000042_0001
Intermediate 39 was prepared from intermediate 38 using chemistry described for intermediate15 except that the product of deprotection was not free-based, but isolated as HCI salt.
1H NMR (400 MHz, DMSO-cfe) δ ppm 8.82 (s, 1 H), 8.19 (s, 1 H), 8.05 (s, 1 H), 7 76 (s, 1 H), 7.65 - 7.72 (m, 1 H), 7.41 - 7.58 (m, 3 H), 7.24 - 7.34 (m, 1 H), 7.19 (d, J=9.89 Hz, 1 H), 7.08 (s, 1 H), 4.51 (s, 1 H), 3.21 (s, 2 H), 2.97 (s, 2 H), 1.78 - 2.03 (m, 2 H), 1.29 - 1.59 (m, 2 H). ES-LCMS: m/z 425.22 (M+H).
Example 8
5-((f(2-Cvclopropylethvπ(1-(f2-metrιyl-6-((4-f(methylsulfonyl)amino1phenyl>oxy')-3- pyridinvnmethylV4-piperidinyl)amino1carbonyl)aminoV2,4-difluorobenzamide
Figure imgf000042_0002
Example 8 was prepared from intermediates 44 and 3 using chemistry described for example 2.
1H NMR (400 MHz, DMSO-cfe) δ ppm 9.66 (s, 1 H), 8.04 (s, 1 H), 7.57 - 7.74 (m, 3 H), 7.32 (t, J=10.26 Hz, 1 H), 7.23 (d, J=8.42 Hz, 2 H), 7.09 (d, J=8.61 Hz, 2 H), 6.72 (d, J=8.06 Hz, 1 H), 3.82 - 3.96 (m, 1 H), 3.32 (s, 3 H), 3.22 - 3.28 (m, 2 H),
2 99 (s, 3 H), 2 84 (d, J=10 62 Hz, 2 H), 2 35 (s, 3 H), 2 04 (t, J=10 62 Hz, 2 H), 1 56 - 1 71 (m, 4 H), 1 42 (s, 2 H), 0 68 (s, 1 H), 0 39 (d, J=I 33 Hz, 2 H), 0 06 (d, J=A 58 Hz, 2 H) ES-LCMS m/z 657 4 (M+H) HRMS (M + H)+ calcd for C32H38N6O5F2S + H, 657 2671, found, 657 2673
Preparation of intermediate 44
Figure imgf000043_0001
Intermediate 40
Figure imgf000043_0002
1 N DIBAL (71 0 mL, 71 0 mmol, 1 1 equiv) was added dropwise via an addition funnel to a solution of cyclopropylacetonitπle (524 g, 64 6 mmol, 1 equiv) in dichloromethane (75 mL) at 0 0C and the reaction was stirred at this temperature for 3 h MeOH and 1 N NaHSO4 were added and the reaction was stirred at RT for 4 h
The organics were separated, dried over Na2SO,t, filtered and concentrated to a ~100 mL solution of the aldehyde in dichloromethaπe This solution was cooled to 0 0C before the addition of 1 , 1 -dimethylethyl 4-amino-1 -piperidinecarboxylate (11 6 g, 58 14 mmol, 0 9 equiv) followed by sodium tπacetoxyborohydπde (27 3 g, 129 2 mmol, 2 equiv) The reaction mixture was allowed to warm to RT over 3 h and then poured into cold 3 3 N NaOH (1L) and then stirred for an additional 2 h The aqueous layer was extracted with dichloromethane The organics were washed with satd aq NaCI, dπed over Na2SO4, filtered and concentrated to give intermediate 40 (12 g, 70% yield) as an oil
1H NMR (400 MHz, CHLOROFORM-cQ δ ppm 3 99 (s, 2 H), 261 - 2 84 (m, 4 H), 249 - 2 62 (m, 1 H), 1 80 (d, J=12 09 Hz, 2 H), 1 37 - 1 45 (m, 9 H), 1 34 (q, J=7 14 Hz, 2 H), 1 03 - 1 28 (m, 3 H), 0 51 - 068 (m, 1 H), 0 28 - 043 (m, 2 H), -0 07 - 0 04 (m, 2 H)
Intermediate 41
1 ,1 -dimethylethyl 4-{(2-cycIopropylethyl)[({2,4-dιfluoro-5-
[(methyloxy)carbonyl]phenyl}amιno)carbonyl]arnιno}-1-pιperιdιnecarboxylate
Figure imgf000044_0001
Intermediate 41 was prepared from intermediate 40 using chemistry described for intermediate 12
1H NMR (400 MHz, DMSO-Cf6) δ ppm 8 08 (s, 1 H), 7 91 (t, J=8 33 Hz, 1 H), 7 38 (t, J=W 62 Hz, 1 H), 3 87 - 4 09 (m, 3 H), 375 - 3 83 (m, 3 H), 3 14 - 3 25 (m, 2 H), 2 69 (s, 2 H), 1 56 (s, 4 H)1 1 36 (s, 11 H), 0 64 (s, 1 H), 027 - 0 39 (m, 2 H), -0 05 - 0 07 (m, 2 H) ES-LCMS m/z 50445 (M+Na)
Intermediate 42
5-({[(2-cyclopropylethyl)(1 -{[(1 , 1 -dιmethylethyI)oxy]carbonyl}-4- pipeπdinylJaminolcarbonylJaminoJ-Z^-difluorobenzoic acid
Figure imgf000045_0001
Intermediate 42 was prepared from intermediate 41 using chemistry described for intermediate 13.
1H NMR (400 MHz, DMSOd6) δ ppm 8 09 (s, 1 H), 7 88 (t, J=8 42 Hz, 1 H), 7 33 (t, J=1053 Hz, 1 H), 3 85 - 4 11 (m, 3 H), 3 14 - 3 29 (m, 2 H), 256 - 283 (m, 2 H), 1 45 - 1 67 (m, 4 H), 1 27 - 1 46 (m, 11 H), 0 53 - 073 (m, 1 H), 026 - 044 (m, 2 H), -0 05 - 0 14 (m, 2 H) ES-LCMS m/z 4902 (M+Na)
Intermediate 43
1 , 1 -dimethylethyl 4-[({[5-(amιnocarbonyl)-2,4-dιfluorophenyl]amino}carbonyl)(2- cyclopropylethyl)amino]-1-piperidinecarboxylate
Figure imgf000045_0002
Intermediate 43 was prepared from intermediate 42 using chemistry described for intermediate 14.
1H NMR (400 MHz, DMSOd6) δ ppm 7 97 - 8 09 (m, 1 H), 7 55 - 7 70 (m, 3 H), 7 25 - 735 (m, 1 H), 384 - 4 05 (m, 2 H), 3 14 - 327 (m, 2 H), 2 57 - 280 (m, 2 H), 1 43 - 1 65 (m, 4 H), 1 30 - 1 44 (m, 11 H), 1 13 - 1 26 (m, 1 H), 0 55 - 072 (m, 1 H), 029 - 042 (m, 2 H), -0 04 - 0 08 (m, 2 H) ES-LCMS m/z 465 5 (M-H)
Intermediate 44
5-({[(2-cyclopropylethyl)(4-pιperιdιnyl)amιno]carbonyl}amιno)-2,4-dιfluorobenzamιde
Figure imgf000045_0003
Intermediate 44 was prepared from intermediate 43 using chemistry described for intermediate 15.
1H NMR (400 MHz, DMSO-d6) 5 ppm 7 99 (s, 1 H), 7 55 - 7 71 (m, 3 H), 7 23 - 7 35 (m, 1 H), 3 97 (s, 1 H), 3 17 - 336 (m, 3 H), 2 86 - 3 02 (m, 2 H), 2 38 - 2 52 (m, 2 H), 1 45 - 1 59 (m, 4 H), 1 31 - 1 45 (m, 2 H), 0 56 - 0 72 (m, 1 H), 026 - 045 (m, 2 H), -0 03 - 0 12 (m, 2 H) ES-LCMS rn/z 3674 (M+H)
Example 9
2.4-Dιfluoro-5-rffl3-fluorophen yl)f 1 -({ 2-methyl-6-[ (4-fl f 1 - methylethyl)sulfonyllamιno>phenyl)oxyl-3-pyrιdιnyl>methyl)-4- pipeπdinyllaminotoarbonvDaminoibenzamide
Figure imgf000046_0001
Example 9 was prepared from intermediates 15 and 47 as described for example 2 1H NMR (400 MHz, DMSOd6) δ ppm 9 68 (s, 1 H), 7 66 - 7 72 (m, 1 H), 760 (s, 2 H), 745 - 754 (m, 2 H), 7 23 - 7 31 (m, 3 H), 7 18 - 722 (m, 2 H), 7 12 - 7 16 (m, 1 H), 7 07 - 7 10 (m, 1 H), 6 98 - 7 04 (m, 2 H), 663 (d, J=8 24 Hz, 1 H), 4 17 - 428 (m, 1 H), 3 30 (s, 4 H), 3 12 - 3 22 (m, 1 H), 2 75 (d, J=11 17 Hz, 2 H), 2 20 - 2 28 (m, 3 H), 1 99 - 2 08 (m, 2 H), 1 75 (d, J= 11 35 Hz, 2 H), 1 23 (s, 3 H), 1 21 (s, 3 H) ES-LCMS m/z 711 4 (M+H) HRMS (M + H)* calcd for C35H37N6O5F3S + H, 711 2577, found, 711 2566
Preparation of intermediate 47
800C
Figure imgf000047_0001
intermediate 1
Figure imgf000047_0002
intermediate 46 intermediate 47
Intermediate 45 Λ/-(4-hydroxyphenyl)-2-propanesulfonamide
Figure imgf000047_0003
Intermediate 45 was preparation from 4-aminophenol using chemistry described for intermediate 48.
1H NMR (400 MHz, DMSO-dB) δ ppm 9.26 (d, J=I lOO Hz, 1 H), 6.90 - 7.10 (m, 2 H), 6.56 - 6.80 (m, 2 H), 3.31 (s, 1 H), 3.04 (t, J=6.78 Hz, 1 H), 1.20 (s, 3 H), 1.17 - 1.19 (m, 3 H). ES-LCMS: m/z 238.2 (M+Na).
Intermediate 46 Λ/-{4-[(5-cyano-6-methyl-2-pyridinyl)oxy]phenyl}-2-propanesulfonamide
Λr
KA0XJ o' Ao
Intermediate 46 was prepared from 45 using chemistry described for intermediate
49.
1H NMR (400 MHz, DMSOd6) δ ppm 9.79 (s, 1 H), 8.18 (d, J=8.61 Hz, 1 H), 7.17 -
7.31 (m, 2 H), 7.07 - 7.17 (m, 2 H), 6.92 (d, J=8.61 Hz, 1 H), 3.15 - 3.28 (m, 1 H),
2.46 (s, 3 H), 1.24 (s, 3 H), 1.22 (s, 3 H). ES-LCMS: m/z 332.3 (M+H).
Intermediate 47 pyrιdιnyl)oxy]phenyl}-2-propanesulfonamιde
Figure imgf000048_0001
Intermediate 47 was prepared from intermediate 46 using chemistry described for intermediate 50.
1H NMR (400 MHz, DMSO-c/6) δ ppm 1015 (s, 1 H), 978 (s, 1 H), 816 (d, J=S 61
Hz, 1 H), 718 - 730 (m, 2 H), 706 - 719 (m, 2 H), 691 (d, J=B 61 Hz, 1 H), 314 -
3 29 (m, 1 H), 259 (s, 3 H), 1 24 (s, 3 H), 1 23 (s, 3 H) ES-LCMS m/z 335 3
(M+H)
Example 10
5-(<T(1-(r6-(f4-f(Cvclopropylsulfonyl)amιno1phenyltoxy)-2-methyl-3-pyπdιnvnmetrιyll- 4-piperιdιnyl)(3-fluorophenyl)amιno1carbonyl)amιno)-2.4-dιfluorobenzamιde
Figure imgf000048_0002
Example 10 was prepared from intermediates 15 and 50 as described for example 2
1H NMR (400 MHz, DMSO-dβ) S ppm 964 (s, 1 H), 7 68 - 7 75 (m, 1 H), 7 63 (s, 2 H), 748 - 7 58 (m, 2 H), 721 - 7 34 (m, 5 H), 7 14 - 720 (m, 1 H), 7 11 (d, J=787 Hz, 1 H), 7 01 - 7 08 (m, 2 H), 6 67 (d, J=8 24 Hz, 1 H), 4 25 (t, J= 11 99 Hz, 1 H), 3 32 (s, 3 H), 2 78 (d J= 11 17 Hz, 2 H), 2 55 - 263 (m, 1 H), 2 26 (s, 2 H), 2 05 (t, J=10 99 Hz, 2 H), 1 78 (d, J=10 99 Hz, 2 H), 1 19 - 1 31 (m, 2 H), 0 87 - 0 97 (m, 4 H) ES-LCMS m/z 709 5 (M+H) HRMS (M + H)+ calcd for C35H35N6O5F3S + H, 709 2420, found, 709 2426
Preparation of intermediate 50
1
Figure imgf000049_0001
intermediate 49 intermediate 50
Intermediate 48 N-(4-hydroxyphenyl)oyclopropanesulfonamide
ΛβrA ό b
Cyclopropaπesulfoπyl chloride (3.2 g, 22.9 mmol, 0.5 equiv) was added to a solution of 4-aminophenol (5.0 g, 45.8 mmol, 1 equiv) in methanol (40 ml_) at 0 0C. The reaction was allowed to warm to RT over 3h and then concentrated down. The solid residue was suspended in 1N hydrochloric acid, stirred for 3 h and filtered to give intermediate 48 (4.1g, 84% yield) as a tan solid.
1H NMR (400 MHz, DMSO-Cf6) δ ppm 9.15 (s, 1 H), 6.89 - 7.16 (m, 2 H), 6.48 - 6.84 (m, 2 H), 3.65 (s, 1 H), 2.36 - 2.46 (m, 1 H), 0.67 - 0.98 (m, 4 H). ES-LCMS: m/z 236.2 (M+Na).
Intermediate 49 Λ/-{4-t(5-cyano-6-methyl-2-pyridinyl)oxy]phenyl}cyclopropanesulfonamide
Figure imgf000049_0002
A solution of N-(4-hydroxyphenyl)cyclopropanesulfonamide intermediate 48 (1.5 g, 7.2 mmol, 1 equiv), 6-chloro-2-methyl-3-pyridinecarbonitrile intermediate 1 (1.1 g, 7.2 mmol, 1 equiv) and cesium carbonate (4.7 g, 14.4, 2 equiv) in N1N-
dimethylformamide (40 ml_) was heated to 80 0C for 3 h. The reaction was diluted with ethyl acetate and washed with 1:1 satd. aq. NaCI : water. The organics were dried over Na2SO4, filtered and concentrated and residue purified by silica gel flash column chromatography (0 → 60% ethyl acetate : hexanes) to give intermediate 49 (1.6 g, 67% yield) as a yellow solid.
1H NMR (400 MHz, DMSOd6) S ppm 9.73 (s, 1 H), 8.18 (d, J=8.61 Hz, 1 H), 7.20 - 7.31 (m, 2 H), 7.06 - 7.19 (m, 2 H), 6.94 (d, J=8.61 Hz, 1 H), 2.53 - 2.67 (m, 1 H), 2.43 - 2.51 (m, 3 H), 0.84 - 0.96 (m, 4 H). ES-LCMS: m/z 330.3 (M+H).
Intermediate 50
Figure imgf000050_0001
1N DIBAL (9.1 mL, 9.13 mmol, 1.8 equiv) was added dropwise via an addition funnel to a solution of W-{4-[(5-cyano-6-methyl-2- pyridinyl)oxy]phenyl}cyclopropanesulfonamide intermediate 49 (1.6 g, 4.8 mmol, 1 equiv) in dichloromethane (20 mL) at 00C and the reaction was stirred at this temperature for 3 h. MeOH and 1N NaHSO4 were added and the reaction was stirred at RT for 4 h. The organics were separated, dried over Na2SO4, filtered and concentrated. The residue was purified by silica gel flash column chromatography (0 → 70% ethyl acetate : hexanes) to give intermediate 50 (1.0 g, 65% yield) as a white solid.
1H NWIR (400 MHz, DMSO-cfe) δ ppm 10.15 (s, 1 H), 9.72 (s, 1 H), 8.16 (d, J=8.42 Hz, 1 H), 7.20 - 7.32 (m, 2 H), 7.06 - 7.19 (m, 2 H), 6.92 (d, J=8.61 Hz, 1 H), 2.54 - 2.64 (m, 4 H), 0.83 - 0.97 (m, 4 H). ES-LCMS: m/z 333.3 (M+H).
Example 11
N-(44r5-(f4-r(3-Fluorophenvh«T4-methyl-3-(1,3-oxazol-2- yl)phenyllamino>carbonyl)aminol-1-piperidinyl>methylV6-methyl-2- pyridinyπoxy>phenyOmethanesulfonamide
Figure imgf000051_0001
The title compound was obtained by reacting intermediates 56 and 3, as described in example 1
1H NMR (CDCI3, 400 MHz) δ 7.69 (d, J=2.57 Hz, 1 H), 7.68 (d, J=0.73 Hz, 1 H), 7.42 - 7.51 (m, 3 H), 7.37 (br. s., 1 H), 7.13 - 7.25 (m, 5 H), 7.02 - 7.08 (m, 3 H), 6.97 (dt, J=9.16, 2.20 Hz, 1 H), 6.53 (d, J=8.43 Hz, 1 H), 5.91 (s, 1 H), 4.46 - 4.63 (m, 1 H), 3.35 (S, 2 H), 2.95 (s, 3 H), 2.84 (d, J=9.90 Hz, 2 H), 2.57 (s, 3 H), 2.39 (s, 3 H), 2.14 (t, J=11.18 Hz, 2 H), 1.84 (d, J=10.26 Hz, 2 H), 1.28 - 1.45 (m, 2 H). HRMS: (M+H)* calcd for C36H37FN6O5S + H, 685.2603; found, 685.2605.
Preparation of intermediate 56
Figure imgf000051_0002
Intermediate 51 Λ/-(2-hydroxyethyl)-2-methyl-5-nitrobenzamide
Figure imgf000052_0001
Thioπyl chloride (2.4 ml_, 33.0 mmol) and 1 drop of pyridine was added to 2-methyI- 5-nitrobenzoic acid (1 g, 5.5 mmol). The mixture was heated at 50 °C for 2 h, and evaporated under reduced pressure to provide the acid chloride as a white solid. To a solution of the acid chloride in 5 mL CH2CI2 was slowly added 2-aminoethanol (0.6 g, 9.8 mmol). The reaction mixture was stirred at RT for 24 h, filtered, and washed with CH2CI2. The filtrate was washed with satd. aq. NaHCO3, dried (Na2SO4), and concentrated. The residue was purified by silica gel flash column chromatography (5→100% EtOAc : hexanes) to give 51 (670.1 mg, 54% yield) as a white solid. 1H NMR (400 MHz, MeOH-Cf4) δ 8.25 (d, J=2.56 Hz, 1 H), 8.18 (dd, J=8.42, 2.56 Hz, 1 H), 7.49 (d, J=8.42 Hz, 1 H), 3.71 (t, J=5.77 Hz, 2 H), 3.49 (t, J=5.68 Hz, 2 H), 2.50 (s, 3 H). ES-LCMS: m/z 225.2 (M+H).
Intermediate 52 2-(2-methyl-5-nitrophenyl)-4,5-dihydro-1 ,3-oxazole
Figure imgf000052_0002
To a solution of Λ/-(2-hydroxyethyl)-2-methyl-5-nitrobeπzamide 51 (650 mg, 2.9 mmol) in 10 mL CH2CI2 was added thionyl chloride (634 μL, 8.7 mmol) and the solution was stirred at RT for 2 h. The mixture was cooled to 0 °C and diluted with water (10 mL). The organic layer was isolated and the aqueous layer was basified with NaOH pellets and then extracted with CHCI3. The combined organic layers was dried (Na2SO4) and concentrated to afford Λ/-(2-chloroethyl)-2-methyl-5- nitrobenzamide (705.0 mg, quant.) as a white solid. 1H NMR (400 MHz, CDCI3) δ 8.26 (d, J=2.56 Hz, 1 H), 8.18 (dd, J=BAZ, 2.56 Hz, 1 H), 7.42 (d, J=8.43 Hz, 1 H), 6.28 (br. s., 1 H), 3.74 - 3.86 (m, 4 H), 2.57 (s, 3 H). ES-LCMS: m/z 243.2 (M+H).
The above N-(2-chloroethyl)-2-methyl-5-nitrobeπzamide (705.0 mg, 2.9 mmol) was dissolved in EtOH (6 mL) and 1M aq. NaOH (6 mL) and the solution was heated at
80 °C for 2 h The mixture was then extracted with CH2CI2, dried (Na2SO4) and concentrated to give 2-(2-methyl-5-nιtrophenyl)-4,5-dιhydro-1,3-oxazole, 52 (350 9 mg, 59% yield) as an orange solid which was used without further purification 1H NMR (400 MHz, CDCI3) δ ppm 8 72 (d, J=257 Hz, 1 H), 8 19 (dd, J=843, 2 57 Hz,
1 H), 743 (d, J=B 43 Hz, 1 H), 4 50 (t, J=9 71 Hz, 2 H), 4 19 (t, J=Q 53 Hz, 2 H),
2 73 (s, 3 H) ES-LCMS m/z 207 1 (M+H) ■
Intermediate 53 2-(2-methyl-5-nιtrophenyl)-1 ,3-oxazole
Figure imgf000053_0001
To a solution of 2-(2-methyl-5-nιtrophenyl)-4,5-dιhydro-1,3-oxazole, 52 (306 9 mg, 1 5 mmol) in benzene (12 rnL) was added nickel (II) peroxide (3 5 g, 32 mmol) The reaction mixture was refluxed for 24 h, concentrated and purified by silica gel flash column chromatography (0→50% EtOAc hexanes) to give 2-(2-methyl-5- nιtrophenyl)-1 ,3-oxazole, 53 (73 7 mg, 24% yield) as a white solid 1H NWIR (400 MHz, CDCI3) δ 885 (d, J=2 20 Hz, 1 H), 8 16 (dd, J=8 62, 238 Hz, 1 H), 7 80 (s, 1 H), 7 47 (d, J=843 Hz, 1 H), 7 33 (s, 1 H), 2 77 - 2 85 (m, 3 H) ES-LCMS m/z 205 1 (M+H)
Intermediate 55
1,1-dimethylethyl 4-[(3-fluorophenyl)({[4-methyl-3-(1 ,3-oxazol-2- yl)phenyl]amino}carbonyl)ammo]-1-piperidinecarboxylate
Figure imgf000053_0002
A solution of 2-(2-methyl-5-nιtropheπyl)-1,3-oxazole, 53 (73 7 mg, 0 36 mmol) in MeOH (10 mL) with catalytic 5% Pd/C was stirred vigorously under an atmosphere of H2 gas for 25 h The mixture was filtered through celite and concentrated to provide
4-methyl-3-(1 ,3-oxazol-2-yl)anιlιne 54 in quantitative yield as a white solid
A solution of the above aniline 54 (204 mg, 0 12 mmol) in CH2CI2 (1 3 mL) at 0 0C was treated with pyπdine (28 μL, 0 35 mmol) and 1 9 M phosgene in toluene (93 μL, 0 18 mmol) After 24 h, the reaction mixture was evaporated, dπed in vacuo and redissolved in CH2CI2 (2 mL) To this, intermediate 11, 1,1-dιmethylethyl 4-[(3- f!uorophenyl)amιno]-1-pιperιdιnecarboxylate (50 mg, 0 17 mmol) was added and the solution was stirred at RT for 24h The mixture was concentrated and purified by silica gel flash column chromatography (0→50% EtOAc hexanes) to give 1 ,1- dimethylethyl 4-[(3-fluorophenyl)({[4-methyl-3-(1 ,3-oxazol-2- yl)phenyl]amιno}carbonyl)amino]-1-pιpeπdιnecarboxylate, 55 (47 7 mg, 80% yield) as a white solid 1H NMR (400 MHz, CDCI3) δ 7 69 (s, 1 H), 7 67 (d, J=2 20 Hz, 1 H), 744 - 7 52 (m, 2 H), 726 (s, 1 H), 7 19 - 7 23 (m, 1 H), 7 17 (d, J=843 Hz, 1 H), 7 01 - 7 05 (m, 1 H), 6 96 (dt, J=9 16, 220 Hz, 1 H), 5 90 (s, 1 H), 4 62 - 472 (m, 1 H), 4 07 - 4 22 (m, 2 H), 2 73 - 2 86 (m, 2 H), 2 58 (s, 3 H), 1 80 - 1 89 (m, 2 H), 1 39 (s, 9 H), 1 18 - 1 32 (m, 2 H) ES-LCMS m/z 495 2 (M+H)
Intermediate 56 Λ/-(3-fluorophenyl)-W-[4-methyl-3-(1 ,3-oxazol-2-yl)phenyl]-Λ/-4-pιperιdιnylurea
Figure imgf000054_0001
A solution of 1 ,1-dιmethylethyl 4-[(3-fluorophenyl)({[4-methyI-3-(1 ,3-oxazol-2- y!)phenyl]amιno}carbonyl)amιno]-1-pιperιdιnecarboxylate 55 (47 7 mg, 0 096 mmol) in 4N HCI in dioxane (0 5 mL, 2 mmol) and CH2CI2 (1 5 mL) was stirred at RT for 2h and concentrated to a white solid The solid was stirred in a mixture Of CH2CI2 (5 mL) and sat'd aq NaHCC>3 for 15 mins The aqueous layer was extracted with CH2CI2, and the organic layers were pooled, dried (Na2SC\4), and concentrated to provide Λ/-(3-fluorophenyl)-W-[4-methyl-3-(1 ,3-oxazol-2-yl)phenyl]- Λ/-4-pιpeπdιnylurea, 56 in quantitative yield as a white solid, which was used without further purification
Example 12
Λ/-(4-{[5-({4-ff{r2,4-Dιfluoro-5-(1,3,4-oxadιazol-2- yl)phenyllamιno)carbonyl)(phenyl)amιnol-1-pιperιdιnyl}methyl)-6-methyl-2-
Figure imgf000055_0001
Λ/-(4-{[5-({4-[({[2,4-dιfluoro-5-(1,3,4-oxadιazoI-2- yl)phenyl]amιno}carbonyl)(phenyl)amino]-1-pιperιdinyl}methyl)-6-methyl-2- pyπdιnyl]oxy}phenyl)methanesulfonamιde example 12 was prepared from intermediates 60 and 3 using chemistry described in example 1. 1H NMR (CDCI3, 400 MHz) δ 8 83 - 8 90 (m, 1 H), 849 (s, 1 H), 744 - 7 56 (m, 4 H), 7 21 - 7 28 (m, 4 H), 7 04 - 7 11 (m, 3 H), 6 92 (t, J=10 08 Hz, 1 H), 6 55 (d, J=S 43 Hz, 1 H), 6 09 (d, J=Z 93 Hz, 1 H), 447 - 460 (m, 1 H), 336 (s, 2 H), 2 99 (s, 3 H), 2 85 (d, J=9 17 Hz, 2 H), 2 39 (s, 3 H), 2 15 (t, J=IO 63 Hz, 2 H), 1 86 (d, J=10 63 Hz, 2 H), 1 33 - 1 48 (m, 2 H) HRMS (M+H)+ calcd for C34H33F2N7O5S + H, 690 2310, found, 690 2297
Preparartion of intermediate 60
Figure imgf000056_0001
Intermediate 57
1 ,1-dimethylethyl 4-[[({5-[(bis{[(1 ,1-dimethylethyl)oxy]carbonyI}amino)carbonyl]-2,4- difluorophenyIK[(1 , 1 -dimethylethyl)oxy]carbonyl}amino)carbonyl]{phenyl)amino]-1 - piperidinecarboxylate
Figure imgf000056_0002
To a solution of intermediate 9; 1 ,1-dimethylethyl 4-[({[5-(aminocarbonyl)-2,4- difluorophenyl]amino}carbonyl)(3-fluorophenyi)amino]-1 -piperidinecarboxylate (300 mg, 0.73 mmol) and 4-(dimethylamino)pyridine (40 mg, 0.33 mmol) in CH3CN (2.5 mL) and CH2CI2 (2.5 mL) was added di-ferf-butyl dicarbonate (1.6 g, 7.3 mmol). The solution was stirred at RT for 3 h, then evaporated, redissolved in CH2CL2 (5 mL) and another portion of di-terf-butyl dicarbonate (800 mg, 3.7 mmol) was added. After 24 h, the reaction mixture was concentrated and purified by silica gel flash column chromatography (0→40% EtOAc : hexanes) to give 1 ,1-dimethylethyl 4-[[({5- [(bis{[(1 , 1 -dimethylethyl)oxy]carbonyl}amino)carbonyl]-2,4-difluorophenyl}{[(1 , 1 - dimethylethyl)oxy]carbonyl}amino)carbonyl](phenyl)amino]-1 -piperidinecarboxylate
57 (509.2 mg, 90 % yield) as a white solid. 1H NMR (400 MHz, CDCI3) δ 7.47 (t, J=7.60 Hz, 1 H), 7.21 - 7.32 (m, 3 H), 6.84 - 6.91 (m, 2 H), 6.61 (t, J=9.71 Hz, 1 H), 4.38 - 4.49 (m, 1 H), 4.03 - 4.17 (m, 2 H), 2.68 - 2.81 (m, 2 H), 1.79 - 1.88 (m, 2 H), 1.47 (s, 18 H), 1.37 (s, 9 H), 1.36 (s, 9 H), 1.26 - 1.33 (m, 2 H). ES-LCMS: m/z 797.5 (M+Na).
Intermediate 58
1,1-dimethylethyl 4-[[([2,4-difluoro-5-(hydraziπocarbonyl)phenyl]{[(1 ,1- dimethylethyl)oxy]carbonyl}amino)carbonyl](pheπyl)amino]-1-piperidiπecarboxylate
Figure imgf000057_0001
A solution of 1 ,1-dimethylethyl 4-[[({5-[(bis{[(1 ,1- dimethylethyl)oxy]carbonyl}amino)carbonyl]-2,4-difluorophenyiχ[(1 , 1 - dimethylethylJoxylcarbonylJaminoJcarbonylKphenylJaminol-i-piperidinecarboxylate,
57 (250 mg, 0.32 mmol) and hydrazine hydrate (20 μL, 0.64 mmol) in CH2CI2 (0.6 mL) was stirred at RT for 24 h, then concentrated and purfied by silica gel flash column chromatography (0→100% EtOAc : hexanes) to give 1,1-dimethylethyl 4- [[([2,4-difluoro-5-(hydrazinocarbonyl)phenyl]{[(1 , 1 - dimethylethyQoxyJcarbonytyaminoJcarbonylKphenyOaminol-i-piperidinecarboxylate,
58 (166.2 mg, 87 % yield) as a white solid. 1H NMR (400 MHz, CDCI3) δ 7.92 (br. S., 1 H), 7.80 (t, J=8.06 Hz, 1 H), 7.16 - 7.27 (m, 3 H), 6.84 (d, J=6.78 Hz, 2 H), 6.66 (dd, J=10.62, 9.71 Hz, 1 H), 4.34 - 4.46 (m, 1 H), 3.93 - 4.12 (m, 4 H), 2.63 - 2.76 (m, 2 H), 1.73 - 1.85 (m, 2 H), 1.32 (s, 9 H), 1.31 (s, 9 H), 1.19 - 1.29 (m, 2 H). ES- LCMS: m/z 588.4 (M-H).
Intermediate 59
1 , 1 -dimethylethyl 4-[[([2,4-difluoro-5-(1 ,3,4-oxadiazol-2-yl)phenyl]{[(1 ,1 - dimethylethyl)oxy]carbonyl}amino)carbonyl](phenyl)amlno]-1-piperidinecarboxylate
Figure imgf000058_0001
A solution of 1,1-dimethylethyl 4-[[([2,4-difluoro-5-(hydrazinocarbonyl)phenyl]{[(1 ,1- dimethylethyl)oxy]carbonyl}amino)carbonyl](phenyl)amino]-1-piperidinecarboxylate,
58 (166.2 mg, 0.28 mmol) in triethyl orthoformate (2 mL, 12 mmoi) was heated at
110°C for 24 h, concentrated and purified by silica gel flash column chromatography (0→50% EtOAc : hexanes) to give 1 ,1-dimethylethyl 4-[[([2,4-difluoro-5-(1 ,3,4- oxadiazol-2-yl)phenyI]{[(1 , 1 - dimethylethyl)oxy]carbonyl}amino)carbonyl](phenyl)amino]-1-piperidinecarboxylate,
59 (127.1 mg, 75 % yield) as a white solid. 1H NMR (400 MHz, CDCI3) δ 8.52 (s, 1 H), 7.81 (t, .7=7.51 Hz, 1 H), 7.20 - 7.29 (m, 3 H), 6.86 - 6.90 (m, 2 H), 6.82 (t, J=9.71 Hz, 1 H), 4.37 - 4.50 (m, 1 H), 3.99 - 4.17 (m, 2 H), 2.65 - 2.81 (m, 2 H), 1.76 - 1.88 (m, 2 H), 1.35 (s, 9 H), 1.34 (s, 9 H), 1.23 - 1.32 (m, 2 H). ES-LCMS: m/z 600.4 (M+H).
Intermediate 60 1 ,3,4-oxadiazol-2-yi)phenyl]-/V-phenyl-Λ/-4-piperidinylurea
Figure imgf000058_0002
A solution of 1 ,1-dimethylethyl 4-[[([2,4-difluoro-5-(1 ,3,4-oxadiazol-2-yl)phenyl]{[(1,1- dimethylethyl)oxy]carbonyl}amino)carbonyl](phenyl)amino]-1-piperidinecarboxylate, 59 (60.0 mg, 0.10 mmol) was stirred in a solution of trifluoroacetic acid (0.5 mL, 6.5 mmol) and CH2CI2 (1 mL) at RT for 3 h. The mixture was neutralized with sat'd aq. NaHCO3 and the aqueous phase was then extracted with CH2CI2. The organic layers were combined and evaporated to provide W-[2,4-difluoro-5-(1,3,4-oxadiazol- 2-yl)pheπyl]-Λ/-phenyl-Λ/-4-piperidinylurea, 60 (38.8 mg, 97 % yield) as a white solid which was used without further purification. ES-LCMS: m/z 400.3 (M+H).
Example 13
Λ/-(4-(r5-((4-rffr2.4-Difluoro-5-(1.3,4-oxadiazol-2-yl)pheπyllamino>carbonvnf3- fluorophenyl)amino1-1-piperidinyl)methvπ-6-mβthyl-2- pyridinylloxyk>henyl)methanesulfoπamide
Figure imgf000059_0001
Λ/-(4-{[5-({4-[({[2,4-difluoro-5-(1 ,3,4-oxadiazol-2-yl)phenyI]amino}carbonyl)(3- fluorophenyl)amino]-1-piperidinyl}rnethyl)-6-methyl-2- pyridiny[]oxy}phenyl)methanesulfonamide was prepared from intermediates 64 and 3 using chemistry described in example 1
1H NMR (CDCI3, 400 MHz) δ 8.80 - 8.87 (m, 1 H), 8.48 - 8.50 (m, 1 H), 7.45 - 7.54 (m, 2 H), 7.19 - 7.28 (m, 3 H), 7.15 (br. s., 1 H), 7.05 - 7.10 (m, 3 H), 6.99 (dt, ,7=9.16, 2.20 Hz, 1 H), 6.94 (t, J=10.08 Hz, 1 H), 6.55 (d, J=B.07 Hz, 1 H), 6.08 (d, .7=3.30 Hz, 1 H), 4.46 - 4.58 (m, 1 H), 3.36 (s, 2 H), 2.99 (s, 3 H), 2.86 (d, J=11.36 Hz, 2 H), 2.39 (s, 3 H), 2.15 (t, J=11.18 Hz, 2 H), 1.85 (d, .7=11.36 Hz, 2 H), 1.34 - 1.49 (m, 2 H). HRMS: (M+H)+ calcd for C34H32F3N7O5S + H, 708.2216; found, 708.2207.
The synthesis of intermediate 64
Figure imgf000060_0001
CH2CI2
Figure imgf000060_0002
Figure imgf000060_0003
Intermediate 61
1 ,1-dimethylethyl 4-[[({5-[(bis{[(1 ,1-dimethylethyl)oxy]carbonyl}amino)carbonyl]-2,4- difluorophenyIK[(1,1-dimethylethyI)oxy]carbonyl}amino)carbonyl](3- fluorophenyl)amino]-1-piperidinecarboxylate
Figure imgf000060_0004
Prepared by a procedure identical to that used for Intermediate 57 1H NMR (400 MHz, CDCI3) 67.48 - 7.54 (m, 1 H), 7.19 - 7.26 (m, 1 H), 7.00 - 7.07 (m, 1 H), 6.62 - 6.72 (m, 3 H)1 4.35 - 4.48 (m, 1 H), 4.05 - 4.18 (m, 2 H), 2.67 - 2.81 (m, 2 H), 1.75 - 1.89 (m, 2 H), 1.47 (s, 18 H), 1.38 (s, 9 H), 1.37 (s, 9 H), 1.23 - 1.33 (m, 2 H). ES-LCMS: m/z 815.5 (M+Na).
Intermediate 62
1 ,1-dimethylethyl 4-[[([2,4-difluoro-5-(hydrazinocarbonyl)phenyl]{[(1 ,1- dimethylethyl)oxy]carbonyl}amino)carbonyl](3-fluorophenyl)amino]-1- piperidinecarboxylate
Figure imgf000061_0001
Prepared by a procedure identical to that used for intermediate 58 1H NMR (400 MHz, CDCI3) δ 7.98 (br. s., 1 H), 7.86 (t, J=8.06 Hz, 1 H), 7.18 - 7.26 (m, 1 H), 7.01 (td, J=8.29, 1.92 Hz, 1 H), 6.60 - 6.76 (m, 3 H), 4.34 - 4.47 (m, 1 H), 3.83 - 4.18 (m, 4 H), 2.62 - 2.79 (m, 2 H), 1.75 - 1.86 (m, 2 H), 1.34 (s, 9 H), 1.33 (s, 9 H), 1.25 - 1.30 (m, 2 H). ES-LCMS: m/z 606.4 (M-H).
Intermediate 63
1 ,1-dimethylethyl 4-[[([2,4-difluoro-5-(1 ,3,4-oxadiazol-2-yl)phenyl]{[(1,1- dimethylethyl)oxy]carbonyl}amino)carbonyl](3-fluorophenyl)amino]-1- piperidinecarboxylate
Figure imgf000061_0002
Prepared by a procedure identical to that used for intermediate 59 1H NMR (400 MHz, CDCI3) δ 8.52 (s, 1 H), 7.89 (t, J=7.60 Hz, 1 H), 7.21 - 7.30 (m, 1 H), 7.00 - 7.07 (m, 1 H), 6.88 (t, J=9.71 Hz, 1 H), 6.66 - 6.74 (m, 2 H), 4.36 - 4.49 (m, 1 H), 4.05 - 4.17 (m, 2 H), 2.67 - 2.80 (m, 2 H), 1.78 - 1.88 (m, 2 H), 1.36 (s, 18 H), 1.27 - 1.34 (m, 2 H). ES-LCMS: m/z 618.3 (M+H).
Intermediate 64
W-t2,4-difluoro-5-(1,3,4-oxadiazol-2-yl)phenyl]-N-(3-fluorophenyl)-/V-4-
Figure imgf000061_0003
Prepared by a procedure identical to that used for Intermediate 60 ES-LCMS: m/z 418.2 (M+H).
Example 14
Λf-(4-fr5-α4-rαr2.4-Difluoro-5-(5-methyl-1.3.4-oxadiazol-2- vπphenvπamino)carbonvh(Dhenvπaminol-1-piperidinyl>methyl)-6-methyl-2- pyridinvHoxytohenvDmethanesulfonamide
Figure imgf000062_0001
Λ/-(4-{[5-({4-[({[2,4-difluoro-5-(5-methyl-1 ,3,4-oxadiazol-2- yl)phenyl]amino}carboπyl)(phenyl)amino]-1-piperidinyl}methyl)-6-methyl-2- pyridinyi]oxy}phenyi)methanesulfonamide was prepared from intermediates 66 and 3 using chemistry described in example 1
1H NMR (CDCI3, 400 MHz) δ 8.73 - 8.84 (m, 1 H), 7.43 - 7.56 (m, 4 H), 7.20 - 7.29 (m, 4 H), 7.02 - 7.11 (m, 3 H), 6.89 (t, J=10.08 Hz, 1 H), 6.54 (d, J=8.07 Hz, 1 H), 6.07 (d, J=3.30 Hz, 1 H), 4.48 - 4.60 (m, 1 H), 3.35 (s, 2 H), 2.99 (s, 3 H), 2.85 (d, J=HOO Hz, 2 H), 2.61 (s, 3 H), 2.39 (s, 3 H), 2.15 (t, J=11.18 Hz, 2 H), 1.86 (d, J=11.00 Hz, 2 H), 1.32 - 1.49 (m, 2 H). HRMS: (M+H)* calcd for C35H35F2N7O5S + H, 704.2467; found, 704.2449.
Preparartion of intermediate 66
Figure imgf000063_0001
Intermediate 65
1 ,1-dimethylethyl 4-[[([2,4-difluoro-5-(5-methyl-1 ,3,4-oxadiazol-2-yl)phenyl]{[(1 ,1- dimethylethylJoxyfcarbonylJarriinoJcarbonylKphenyOarninol-i-piperidinecarboxylate
Figure imgf000063_0002
A solution of 1 ,1-dimethylethyl 4-[[([2,4-difluoro-5-(hydrazinocarbonyl)phenyl]{[(1 ,1- dimethylethyl)oxy]carbonyl}amino)carbonyl](phenyl)amino]-1-piperidinecarboxylate, 58 (544.2 mg, 0.92 mmol) in tπethyl orthoacetate (4 mL, 21.8 mmol) was heated at 1100C for 24 h, concentrated and purified by silica gel flash column chromatography (0→70% EtOAc : hexanes) to give 1 ,1-dimethylethyl 4-[[([2,4-difluoro-5-(5-methyl- 1 ,3,4-oxadiazol-2-yl)pheny!]{[(1 , 1 - dimethylethyOoxylcarbonylJaminoJcarbonylKphenytyaminol-i-piperidinecarboxylate, 65 (521.2 mg, 92 % yield) as a white solid. 1H NMR (400 MHz, CDCI3) δ 7.80 (t, J=7.70 Hz, 1 H), 7.24 - 7.34 (m, 3 H), 6.87 - 6.93 (m, 2 H), 6.81 (t, J=9.71 Hz, 1 H), 4.39 - 4.54 (m, 1 H), 4.03 - 4.19 (m, 2 H), 2.67 - 2.83 (m, 2 H), 2.63 (s, 3 H), 1.80 - 1.88 (m, 2 H), 1.38 (s, 9 H)1 1.37 (s, 9 H), 1.26 - 1.34 (m, 2 H). ES-LCMS: m/z 614.4 (M+H).
Intermediate 66
W-[2,4-difluoro-5-(5-methyl-1 ,3,4-oxadiazol-2-yl)phenyl]-Λ/-phenyl-Λ/-4- piperidinylurea
Figure imgf000064_0001
Prepared by a procedure identical to that used for intermediate 22. ES-LCMS: m/z 414.3 (M+H).
Example 15
/V-(4-ff5-((4-r((r4-Fluoro-3-(1,3-oxazol-2-yl)phenvπamino>carbonyl)(3- fluorophenyl)aminol-1-piperidinylTmethvπ-6-methyl-2- pyridinylloxyl-phenvDmetrianesulfonamide
Figure imgf000064_0002
W-(4-{[5-({4-[({[4-fluoro-3-(1 ,3-oxazol-2-yl)phenyl]amino}carbonyl)(3- fluorophenyl)amino]-1-piperidinyl}methyl)-6-methyl-2- pyridinyl]oxy}phenyl)methanesulfonamide was prepared from intermediates 71 and 3 using chemistry described in example 1
1H NMR (CDCi3, 400 MHz) δ 7.73 (s, 1 H), 7.67 (dd, J=6.05, 2.75 Hz, 1 H), 7.59 - 7.64 (m, 1 H), 7.46 - 7.53 (m, 2 H), 7.19 - 7.27 (m, 4 H), 7.07 - 7.14 (m, 3 H), 7.03 - 7.07 (m, J=8.43 Hz, 1 H), 6.98 (dt, J=9.16, 2.20 Hz, 1 H), 6.54 - 6.59 (m, J=8.07 Hz, 2 H), 5.91 (s, 1 H), 4.48 - 4.61 (m, 1 H), 3.37 (s, 2 H), 3.00 (s, 3 H), 2.86 (d, J=11.00 Hz, 2 H), 2.40 (s, 3 H), 2.16 (t, J=HOO Hz, 2 H), 1.79 - 1.89 (m, 2 H), 1.32 - 1.45 (m, 2 H). HRMS: (M+H)+ calcd for C35H34F2N6O5S + H, 689.2358; found, 689.2353.
Preparartion of intermediate 71
Figure imgf000065_0001
Intermediate 68 2-(2-fluorophenyl)-1 ,3-oxazole
Figure imgf000065_0002
To a -78 °C solution of oxazole (750 μL, 11.4 mmol) in THF (11 mL) was slowly added π-BuLi (7.0 mL of a 1.93 IVI solution in hexanes). The reaction mixture was stirred at -78 °C for 20 miπ and then anhydrous ZnCI2 was added portionwise. After an additional 15 min, the suspension was allowed to slowly warm to RT1 and stirred another 20 minutes at RT resulting in 67. To this solution 1-fluoro-2-iodobenzene (2.53 g, 11.4 mmol) and Pd(PPh3)* (660 mg, 0.57 mmol) were added and the reaction mixture was heated at 60 °C for 3 h. The mixture was concentrated and stirred in a mixture of sat'd aq. NH4CI (20 mL) and EtOAc (20 mL). The organic layer was dried (Na2SO4), concentrated and purified by silica gel flash column chromatography (5→40% EtOAc : hexanes) to give 2-(2-fluorophenyl)-1,3-oxazole, 68 in quantitative yield as an oily brown solid. 1H NMR (400 MHz, CDCI3) δ 8.02 (td,
J=7.60, 1.83 Hz, 1 H), 7.75 (s, 1 H), 7.38 - 7.45 (m, 1 H), 7.28 (s, 1 H), 7.15 - 7.25 (m, 2 H). ES-LCMS: m/z 164.1 (M+H).
Intermediate 69 2-(2-fluoro-5-nitrophenyl)-1 ,3-oxazole
Figure imgf000066_0001
To a 0 °C solution of 2-(2-fluorophenyl)-1 ,3-oxazole, 68 (1.86 g, 11.4 mmol) in cone. H2SO4 (6 mL) was added solid KNO3 (1.38 g, 13.6 mmol) portionwise. The mixture was allowed to warm to RT and stirred for 6 h. The solution was then poured into ice and extracted with EtOAc. The organic layers were combined, dried (Na2SO4), concentrated and purified by silica gel flash column chromatography (0→40% EtOAc : hexanes) to give 2-(2-fluoro-5-nitrophenyl)-1 ,3-oxazole, 69 (575.1 mg, 24% yield) as a light yellow solid. 1H NMR (400 MHz, CDCI3) δ 8.89 (dd, J=6.23, 2.93 Hz, 1 H), 8.25 - 8.31 (m, 1 H), 7.83 (s, 1 H), 7.32 - 7.38 (m, 2 H). ES-LCMS: m/z 209.3 (M+H).
Intermediate 70
1 ,1-dimethylethyl 4-[({[4-fluoro-3-(1 ,3-oxazol-2-yl)phenyl]amino}carbonyl)(3- fluorophenyl)amino]-1-piperidinecarboxylate
Figure imgf000066_0002
Prepared by a procedure identical to that used for intermediate 55 1H NWIR (400 MHz, CDCI3) δ 7.69 (s, 1 H), 7.65 (dd, J=6.23, 2.93 Hz, 1 H), 7.52 - 7.58 (m, 1 H), 7.40 - 7.48 (m, 1 H), 7.14 - 7.20 (m, 2 H), 7.02 - 7.09 (m, 1 H), 7.00 (d, J=8.07 Hz, 1 H), 6.90 - 6.95 (m, 1 H), 5.97 (s, 1 H), 4.55 - 4.67 (m, 1 H), 4.03 - 4.19 (m, 2 H), 2.68 - 2.83 (m, 2 H), 1.77 - 1.85 (m, 2 H), 1.36 (s, 9 H), 1.15 - 1.28 (m, 2 H). ES-LCMS: m/z 499.3 (M+H).
Intermediate 71 W-[4-fluoro-3-(1 ,3-oxazol-2-yl)phenyl]-/V-(3-fluorophenyl)-/V-4-piperidinylurea
Figure imgf000067_0001
Prepared by a procedure identical to that used for intermediate 56
Example 16: 5-({tbutyl(1-{[2-cyclopropyl-6-({4-[(methylsulfonyl)amιno]phenyl}oxy)- 3-pyrιdιnyl]methyl}-4-PiPer|dιnyl)amιπo]carbonyl}amιno)-2,4-difluorobenzarπιde
Figure imgf000067_0002
5-({[Butyl(1-{[2-cyclopropyl-6-({4-[(methylsulfonyl)amino]phenyl}oxy)-3- ρyrιdιnyl]methyl}-4-pιperιdιnyl)amιno]carbonyl}amιno)-2,4-difluorobenzamιde was prepared from intermediates 76 and 22 using the chemistry described in Example 3 1H NMR (400 MHz, METHANOL-Cf4) δ ppm 7 8 (t, J=B 2 Hz, 1 H), 7 7 (d, J=S 2 Hz, 1 H), 7 3 (m, 2 H), 7 1 (t, J=10 3 Hz, 1 H), 7 0 (m, 3 H), 6 6 (d, J=82 Hz, 1 H), 4 1 (d, J=I 1 Hz, 1 H), 3 9 (s, 2 H), 3 2 (s, 6 H), 2 9 (s, 3 H), 2 5 (s, 2 H), 2 2 (d, J=A 6 Hz, 1 H), 2 0 (d, J=16 8 Hz, 2 H), 1 8 (s, 2 H), 1 6 (s, 2 H), 1 3 (m, 2 H), 0 9 (m, 4 H), 0 8 (m, 4 H) HRMS (M+H)+ calcd for C33H40F2N6O5S + H, 671 2822, found, 671 2822
Example 17: 5-({[(1 -{[2-cyclopropyl-6-({4-[(methylsulfonyl)amιno]phenyl}oxy)-3- pyndiπyllmethylH-P^eπdiny^phenylJaminolcarbonylJamino^^-difiuorobenzamide
Figure imgf000067_0003
5-({[(1-{[2-Cyclopropyl-6-({4-[(methylsulfonyl)amino]phenyl}oxy)-3-pyriciinyl]methyl}- 4-piperidinyl)(phenyl)amino]carbonyl}amino)-2,4-difluorobenzamide was prepared from intermediate 76 and 10 as described in Example 1. 1H NMR (400 MHz, CHLOROFORM-d) δ ppm 8.7 (t, J=8.8 Hz, 1 H), 7.5 (m, 4 H), 7.2 (m, 4 H), 7.1 (m, 2 H), 6.8 (t, J=10.4 Hz, 2 H), 6.5 (d, J=8.4 Hz, 1 H), 5.9 (d, J=2.6 Hz, 1 H), 4.5 (m, 1 H), 3.5 (m, 2 H), 3.0 (m, 2 H), 2.9 (s, 2 H), 2.1 (m, 3 H), 1.8 (s, 2 H), 1.8 (s, 2 H), 1.6 (s, 1 H), 1.4 (d, J=13.9 Hz, 2 H), 0.8 (m, 4 H). HRMS: (M+H)+ calod for C35H36F2N6O5S + H, 691.2509; found, 691.2512.
Synthesis of intermediate 76:
Figure imgf000068_0001
δ-Chloro-S-pyridinecarbonitrile (2.29g, 16.5 mmol) was dissolved in dichloromethane (60 mL). To this solution was added urea hydrogen peroxide (4.7 g, 49.6 mmol). After the resulting mixture was cooled down to 00C on an ice bath, trifluoroacetic acid anhydride (6.3 mL, 45 mmol) was slowly introduced by syringe. The reaction was stirred for an additional 3 hours at room temperature before quenched with satd. aq. Na2S2O3 and extracted with dichloromethane. The combined organic layers were then washed with satd. aq. NaHCO3, dried over anhydrous sodium sulfate and concentrated. The residue was purified by flash column chromatography (silical gel, 0-100% EtOAc/ hexaπe ) to provide intermediate 72 as a white solid (2.1 g, 82%). 1H NMR (400 MHz, CHLOROFORM- φ δ ppm 8.5 (d, J=I.6 Hz, 1 H), 7.6 (d, J=8.4 Hz, 1 H), 7.4 (dd, J=8.4, 1.6 Hz, 1 H). MS m/z 155 (M+H)*.
To a solution of intermediate 72 (616 mg, 4 mmol) in DMF (25mL) were added intermediate 4 (748 mg, 4 mmol) and Cs2CO3 (1 ,3 g, 4 mmol). The mixture was stirred for 4 hours at 45 0C before cooled down to room temperature and diluted with water. Intermediate 73 was precipitated and collected as white solid by filtration (1.1 g, 90%). 1H NMR (400 MHz, DMSO-d6) δ ppm 9.8 (s, 1 H), 9.0 (s, 1 H), 7.7 (s, 1 H), 7.2 (m, 2 H), 7.1 (d, J=8.8 Hz, 2 H), 3.0 (s, 3 H). MS m/z 306 (M+H)*.
At N2 atmosphere, intermediate 73 (780 mg, 2.6 mmol) was treated with POCI3 (10 mL) under reflux condition for 1 hour. After evaporation of the solvents, the residue was dissolved in dichloromethane and washed with satd. aq. NaHCO3. The organic phase was then dried over anhydrous sodium sulfate. Evaporation of the solvents gave a light gray solid, which was triturated with ether to afford intermediate 74 as an off white solid (720 mg, 86%). 1H NMR (400 MHz, CHLOROFORM-d) δ ppm 7.9 (d, J=8.4 Hz, 1 H), 7.3 (d, .7=4.4 Hz, 2 H), 7.1 (m, 2 H), 6.9 (d, J=8.6 Hz, 1 H), 3.0 (s, 3 H).
Intermediate 74 (533 mg, 1.6 mmol), cyclopropyl boronic acid (424 mg, 4.9 mmol), K3PO4 (1040 mg, 4.9 mmol) and PEPPSI™-IPr (87 mg) were mixed in schlenk flask charging with 20 mL of toluene. The mixture was purged with N2 (3X) before heated
to 100 0C for 20 hours. After cooled down to room temperature, the solid was filtered off and washed with EtOAc. The filtrate was evaporated and the residue was purified by chromatography (silical gel, 0-50% EtOAc/hexane) to provide intermediate 75 as a sloid (373 mg, 71%). 1H NMR (400 WIHz, CHLOROFORM-c/) δ ppm 7.8 (d, J=BA Hz, 1 H), 7.3 (s, 2 H), 7.1 (d, J=8.8 Hz, 2 H), 6.7 (d, J=BA Hz, 1 H), 6.5 (s, 1 H), 3.0 (s, 3 H), 2.4 (dt, J=SA1 3.9 Hz, 1 H), 1.0 (dd, J=7.7, 2.9 Hz, 2 H), 0.9 (dd, J=4.4, 2.9 Hz, 2 H). MS rn/z 330 (M+H)+.
Intermediate 76 (440 mg, 94%) was synthesized from intermediate 75 (450 mg, 1.5 mmol) in a similar procedure as described in the preparation of intermediate 3. 1 H NMR (400 MHz, DMSO-d6) δ ppm 10.3 (s, 1 H), 8.1 (d, J=8.6 Hz, 1 H), 7.2 (d, J=8.8 Hz, 2 H), 7.1 (m, 2 H), 6.8 (d, J=8.4 Hz, 1 H), 2.9 (m, 4 H), 0.9 (d, J=7.7 Hz, 2 H), 0.7 (br. s., 2 H).
Example 18: Biological Activity
PBL Assay
Whole blood samples were obtained from normal human donors. Peripheral Blood Lymphocytes (PBLs) were separated from whole blood by density gradient centrifugation with Lymphocyte Separation Medium (LSM). Whole blood was diluted 1 :1 with sterile phosphate buffered saline (PBS) and overlaid onto LSM in 50 mL conical centrifuge tubes. Samples were centrifuged at 500 x g at room temperature for 30 minutes. PBLs were then gently removed from the interface between LSM and aqueous phases and washed twice in sterile PBS. The cells were resuspended in 150 mL of RPIvII containing 20% fetal bovine serum, 10% T-cell growth factor (IL- 2), 50ug/mL gentamicin (RPMI 20/10/50) and 5ug/mL Phytohaemagluttinin and incubated at 370C, 5% CO2 for 2 to 3 days. On the second or third day, the cells were washed once with PBS and counted for assay.
PHA stimulated PBLs were centrifuged at 260 x g for 15 min, washed once with PBS, centrifuged as before, resuspended to 8x106 cells/mL in RPMI 20/10/50, and 50 μL was distributed to 96-well tissue culture plates. Test compounds were serially diluted in 4-fold increments at 2Xthe final concentration in RPMI 20/10/50 + 0.2% DMSO. Fifty (50) uL of diluted compound was transferred to the PBLs and placed in
a humidified incubator at 370C, 5% CO2 for 1hr. An additional 60 μL of diluted compound was transferred to a separate 96-well plate containing 60 μL of HIV-1 Ba- L (diluted 1:50 into RPMI 20/10/50) and thoroughly mixed. The input of Ba-L was previously determined by titrating the virus under assay conditions without compound and selecting a dilution that gives a response in the linear range of the curve and a signal to background of at least 30. One hundred (100) μL of this mixture was transferred onto the PBL/compound mixture and placed in a humidified incubator at 370C, 5% CO2 for seven days.
On day seven of the assay, 50 μL of culture supernatant was transferred to a new 96-well plate. The plates were frozen at -80°C and then thawed to room temperature for 30 min. Ten (10) μL of reverse transcriptase (RT) extraction buffer (500 mM KCI, 50 mM DTT and 0.5% NP40 in distilled water) was added to the supernatant followed by the addition of 40 μL of RT assay buffer (1.25 mM EGTA, 125 mM Tric/HCI, 12.5 mM MgCI2, 68 Ci/mmole methyl-3H deoxythymidine-5'-triphosphate, and 0.62 O.D. units/mL of poly(rA) p(dT)12_ in distilled water). The plates were thoroughly mixed on a platform shaker and placed in a humidified incubator at 370C, 5% CO2 for 2h. Whatman Unifilter DE-81 96-well plates were placed on a vacuum manifold and 100 μL of the RT reaction was added. Full vacuum from an in-house line was applied until all of the reaction volume had seeped through the plate. Each plate was then washed a total of three times with all washes performed under full, in-house vacuum. All wash buffers were delivered with a 12 channel Nunc plate washer connected to a carboy containing at least 2 liters of buffer. The first wash consisted of approximately 300 μL of 5% Na2HPO4, followed by one wash of approximately 300 μL of distilled water, and a final wash consisting of 100 μL of 95% ethanol. The plates were allowed to dry completely at room temperature, bottom-sealed with an adhesive backing, and 50 μL of scintillation cocktail was added. The plates were topsealed and read in a Topcount (Packard) luminometer at 10s/well.
Raw data from the PBL assay were expressed as Counts Per Minute (CPM) and normalized according to the following formula:
Log10(CPM) - Log10 (Geometric mean PC) + Log-m (Geometric mean NC) - Log™ (Geometric mean PC)
Where:
PC = Positive Control = mock infected wells in column 12 on each assay plate
NC = Negative Control = untreated but infected wells in column 11 on each assay plate
The potency of test compounds are reported as IC50 values derived from the unconstrained four parameter logistic equation, defined as: y = A + ((B-C))/(1+(10Λx)/10ΛC)ΛD) Where:
A = minimum y D = slope factor B = maximum y x = Iog10(molar compound concentration) C = logiotlCδo)
Compounds of the invention have IC50 values less than 1OnM.
Example 19: Biological Activity
HOS Assay
HOS-Luc cells were created by introducing an HIV-1 LTR-luciferase reporter into HOS-CD4.CCR5 (NIH AIDS Research and Reference Reagents Program, cat. #3318). The cells were propagated in DMEM complete medium. Cells were split 1 :20 by standard trypsinization when confluency reached 80% (roughly every 2 to 3 days).
Black-walled 96-weIt tissue culture plates were seeded with HOS-Luc at 6x103 cells per well in 50 μL DMEM containing 2% FBS and placed in a humidified incubator at 370C, 5% CO2 overnight. The following day, test compounds were serially diluted in 4-fold increments at 2X the final concentration in DMEM + 2% FBS + 0.2% DMSO. Diluted compound (50 μL) was transferred to the HOS-Luc cells and the plates were placed in a humidified incubator at 370C, 5% CO2 for 1 hr. An additional 60 μL of 2X compound was transferred to a clear-walled 96-well tissue culture plate and 60 μL of virus (diluted 1:50 in tissue culture media) was added to each well and thoroughly mixed. The virus input was previously determined by titrating the virus under assay
conditions without compound and selecting a dilution that gives a response in the linear range of the curve and a signal to background of at least 30. The HIV/compound mixture (100 μl_) was transferred to the black-walled plates containing 100 μL of cells/compound. The plates were placed in a humidified incubator at 370C, 5% CO2 for four days.
Following the four-day incubation, 150 μL of supernatant was removed and 50 μL of reconstituted Luciite (Promega) was added to each well. For cytotoxicity assessment, 50 μL of reconstituted CellTiter-GLo (Promega) was added to each well. Each plate was topsealed and read in a Topcount (Packard) luminometer at 1s/well.
Raw data from the HOS-Luc assay were expressed as Relative Light Units (RLU) and normalized according to the following formula:
(RLU at drug [ ] / RLU no drug)*100 = % control
The potency of test compounds are reported as IC50 values derived from the four parameter Hill equation, defined as: y=Vrnax*(1 -(xΛn/(KΛn+xΛn)))+Y2 Where: x = Logio[compound] y = normalized response data
V1TCK= upper bound of response
K = IC50
Y2 = lower bound or baseline of response n = hill coefficient
Compounds of the invention have IC50 values less than 1OnM.
Example 20: Pharmacokinetics in mouse
Non-fasted male CD-1 mice (Charles River Laboratories, Raleigh NC), ranging in weight from 28 g to 30 g, were used for each study.
Test Article Administration and Sample Collection
For intravenous administration, a single dose (10 mL/kg of 0.1 mg free base/mL) was administered via tail vein. For oral solution administration, animals received a single dose (10 mL/kg of 0.1 or 0.3 mg free base/mL) by an oral gavage needle. Blood (approximately 0.8 mL) was taken via cardiac puncture from three mice/dose group at each timepoint of 0.083 (IV only), 0.167 (IV only), 0.25, 0.5, 1, 2, 4, 6, 8, and 24 h and placed on ice. Syringes used for sample collection contained approximately 10 μL EDTA. Plasma was harvested by centrifugation for 3 min at 4000 x g and 4°C and stored at -800C until analysis.
Mice were orally administered a single dose (10 mL/kg of 0.1 or 0.3 mg free base/mL) by an oral gavage needle.
Blood (approximately 0.8 mL) was taken via cardiac puncture from three mice per dose group at each timepoint (0.5, 1 , 2, 4, 8, 12, and 24 h) and placed on ice. Syringes used for sample collection contained approximately 10 μL EDTA. Livers and spleens were also harvested immediately after blood collection, combined (n = 3 at each timepoint/dose group), and placed on ice. Blood and tissue samples were stored at -80°C until analysis.
Sample Analysis
Plasma samples were thawed to room temperature and vortex-mixed. Aliquots (50 μL) of standards and study samples were pipetted into 96-well plates. Liver and spleen tissues were thawed to room temperature and weighed. Each pooled liver and spleen sample was diluted with 4 and 9 volumes of water (mL/g), respectively. Following homogenization (Tomtec Autogizer), homogenates were diluted 5-fold in pooled mouse plasma and analyzed with plasma samples. Liver and spleen QC samples were prepared in homogenized blank liver or spleen homogenate. Acetonitrile (200 μL) containing internal standard was added to each well, samples vortex-mixed, and plates centrifuged for 10 min at 2055 x g and 40C. The acetonitrile supernatant (100 μL) was transferred to clean 96 well plates containing 100 μL or 200 μL of 0.1 % formic acid in water and assayed by LC/MS/MS. The analytical system consisted of a CTC HTS PAL autosampler, Hewlett Packard (HP) 1100 binary pump and Applied Biosystems API 365 mass spectrometer. Samples (10 μL) were injected onto a 30 x 2 mm, .3micron, Luna CN column
(Phenomenex) using an isocratic eluting system at a flow rate of 0.8 mL/min. Mobile phases consisted of A) water containing 0.1% acetic acid, pH adjusted to 6.2 with ammonium hydroxide:acetonitrile (95:5) and B) acetonitrile containing 0.1% acetic acid. The mobile phase composition was 50% A:50% B and total run time was 1.1 miπ. Test compounds and internal standard were detected in the positive turboionspray mode by multiple reaction monitoring (MRM) of the transitions m/z 645 to m/z 291 , and m/z 665 to m/z 467, respectively. Data were acquired and analyzed using Applied Biosystems Analyst version 1.4.1 software. Liver and spleen samples were converted to ng/g following quantitation against the plasma calibration curves accounting for the appropriate dilution factor (25- and 50- fold, respectively).
Data Analysis
Non-compartmental pharmacokinetic parameters [terminal plasma half-life (U12), maximum plasma concentration (Cmax), time of maximum plasma concentration (Tmax) plasma clearance (CL), steady-state volume of distribution (Vss), and area under the plasma concentration-time curve extrapolated to infinite time (AUCD-.)] were calculated based on the composite plasma concentration time data using WiπNonlin Professional 4.1 (Pharsight, Mountain View CA) . Dose-normalized AUC a-*. (DNAUC 0→») was obtained by dividing the AUC 0→«with its respective dose and expressed as unit dose AUC 0→« (ng*h/mL/mg/kg). Oral bioavailability (F) was calculated using the following equation:
F (%) = [(DNAUCa-., oral)/(DNAUCo-», IV] * 100%
Table 1 Plasma Pharmacokinetic Parameter Estimates for Compounds in Non-fasted Male CD-1 Mice Following Intravenous (1 mg free base/kg) and Oral (1 and 3 mg free base/kg) Administration
Figure imgf000076_0001
* WO200S/030925
Example 21: Pharmacokinetics in rat
Male Sprague-Dawley CD rats (Charles River Laboratories, Raleigh NC), were either surgically implanted with a jugular cannula and housed in individual cages or no surgery was required Food and water were offered freely
Test Article Administration and Sample Collection
For intravenous administration, a single dose of test compound was administered into the jugular cannula followed by a 1 mL flush with saline For oral solution administration, animals received a single dose by an oral gavage needle Additional animals received capsule doses (shown in the following table) which were administered by a small capsule-dosing syringe (Harvard Apparatus)
Blood samples (approximately 0 5 mL each) were taken from the jugular cannula at 0 083 (IV only), 0 167 (IV only), 0 25, 0 5, 1 , 2, 4, 6, 8, and 24 h postdose and placed on ice Syringes used for sample collection contained approximately 10 μL EDTA Plasma was harvested by centrifugation for 3 mm at 4000 x g and 4°C and stored at -80°C until analysis
For oral solution administration, animals received a single dose by an oral gavage needle Animals (n=2) were euthanized at 1 , 2, 4, and 24 h postdose, and blood samples (approximately 1 mL with EDTA anticoagulant) taken by cardiac puncture and placed on ice Plasma was harvested by centrifugation for 3 mm at 4000 x g
and 4°C and stored at -800C until analysis. Spleen and liver were also dissected, immediately frozen and stored at -80°C until analysis.
Sample Analysis
Plasma samples were thawed to room temperature and vortex-mixed. Standards were prepared in pooled rat plasma containing EDTA. Quality control (QC) samples were prepared from separately weighed sample stocks. Aliquots (50 μL) of standards, QCs and study samples were pipetted into 96-well plates. Acetonitrile (200 μL) containing internal standard was added to each well, samples vortex- mixed, and plates centrifuged for 20 min at 2055 x g and 4°C. The acetonitrile supernatant (150 μL) was transferred to clean 96 well plates containing 100 μL of 0.1% formic acid in water and assayed by LC/MS/MS.
The analytical system consisted of a CTC HTS PAL autosampler, Hewlett Packard (HP) 1100 binary pump and Applied Biosystems API 4000 mass spectrometer. Samples (10 μL) were injected onto a 30 x 2 mm, 3micron, Atlantis C18 column (Waters) using a gradient eluting system at a flow rate of 0.75 mL/min. Mobile phases consisted of A) water containing 0.5% formic acid and B) acetonitrile containing 0.5% formic acid. The initial mobile phase composition of 90% A:10% B was followed by a linear gradient to 5% A:95% B at 1 min. The 5% A:95% B composition was held for 0.5 min followed by a linear gradient to 90% A:10% B at 2.0 min. Data were acquired and analyzed using Applied BioSystems Analyst version 1.4.1 software.
Plasma samples were thawed to room temperature and vortex-mixed. Standards were prepared in pooled rat plasma containing EDTA. Quality control (QC) samples in plasma were prepared from separately weighed sample stocks. Aliquots (50 μL) of standards, QCs and plasma samples were pipetted into 96-well plates. Liver and spleen tissues were weighed, thawed to room temperature and water (9 volumes, mL/g) added to each sample. Following homogenization (Tomtec Autogizer), homogenate (10 μL) was added to pooled rat plasma (40 μL) containing EDTA in 96-well plates. Liver and spleen QC samples were prepared in homogenized blank liver or spleen homogenate and aliquots (10 μL) added to pooled rat plasma (40 μL) containing EDTA in 96-well plates. Acetonitπle (200 μL) containing internal standard was added to each well, samples vortex-mixed, and plates centrifuged for
20 min at 2055 x g and 4°C. The acetonitrile supernatant (100 μL) was transferred to clean 96 well plates containing 400 μL of 0.1 % formic acid in water and assayed by LC/MS/MS.
The analytical system consisted of a CTC HTS PAL autosampler, Hewlett Packard (HP) 1100 binary pump and Applied Biosystems API 365 mass spectrometer. Samples (10 μL) were injected onto a 30 x 2 mm, 2.5micron, Atlantis C18 column (Waters) using an isocratic eluting system at a flow rate of 0.8 mL/min. Mobile phases consisted of A) water containing 0.1% acetic acid, pH adjusted to 6.2 with ammonium hydroxide:acetonitrile (95:5) and B) acetonitrile containing 0.1% acetic acid. The mobile phase composition was 50% A:50% B and total run time was 1.1 min. Data were acquired and analyzed using Applied BioSystems Analyst version 1.4.1 software.
Liver and spleen samples were converted to ng/g following quantitation against the plasma calibration curves accounting for the appropriate dilution factor (50-fold).
Data Analysis
Non-compartmental pharmacokinetic parameters [terminal plasma half-life (tie), maximum plasma concentration (Cmax), time of maximum plasma concentration (Tmax) plasma clearance (CL), steady-state volume of distribution (Vss), and area under the plasma concentration-time curve extrapolated to infinite time (AUCD-4] were calculated based on the individual plasma concentration time data using WinNonlin Professional 4.1 (Pharsight, Mountain View CA) . Dose-normalized AUC 0-+«, (DNAUC o→») was obtained by dividing the AUC o→»by its respective dose and expressed as unit dose AUCD→» (ng*h/mL/mg/kg). Oral bioavailability (F) was calculated using the following equation:
F (%) = [(DNAUCo-., oral)/(DNAUC0., IV] * 100% where DNAUC0-», IV was the mean DNAUC0.« following IV dose administration.
Table 2 Plasma Pharmacokinetic Parameter Estimates for Compounds in Non-Fasted Male Sprague-Dawley CD Rat Following Intravenous, Oral Solution and Oral Capsule
Figure imgf000079_0001
* WO2006/03O925
Example 22: hERG dofetilide binding assay
1. hERG Membrane Preparation
The cloned hERG channel was heterologously expressed in Chinese hamster ovary
(CHO)-KI cells. Cells were grown in medium comprising DMEM / Ham's F12 (with
15 mM HEPES), 10% FBS, 2 mM Glutamax and 500 mg/ml G418 (no selection pressure in final passage) at 310C in a humidified CO2 atmosphere Membranes were prepared from cells cultured in 1800 cm2 roller bottles All buffers and equipment were cooled before use and kept in ice throughout the process The cells were harvested with HBSS containing NaHCO3 (84 mM) and EDTA (0 6 mM) and spun down at 250 g for 5 mins at 40C This spin was repeated after resuspending the pellets in 200 mis of the same solution All subsequent steps were performed at 40C The cells were homogenized within a glass Waring blender for 2 x 15 sees in 200 mis of a solution containing HEPES (50 mM), leupeptin (0 1 mM), bacitracin (25mg/ml), EDTA (1 mM), PMSF (1 mM) and Pepstatin A (2mM) Pepstatin A and PMSF were prepared as concentrated stocks in absolute ethanol (pepstatin 50Ox, PMSF 10Ox) and were added just before use The blender was plunged into ice for 5 mins after the first burst and 10-40 mins after the final burst to allow foam to dissipate The material was then spun at 50Og for 20 mins and the supernatant spun for 36 mins at 48,00Og The pellet was resuspended in the same buffer as above but without PMSF and Pepstatin A The material was then forced through a 0 6 mm needle, made up to the required volume, aliquoted and stored frozen at -80 0C
2 Experimental Protocol
Compound potencies were determined by a radioligand (3H-dofetιlιde) competition assay In this assay, hERG-expressing CHO-K1 membranes (1 mg membranes/well ) were adhered to wheat germ agglutinin-coated SPA PEI imaging beads from Amersham (60 mg/well) in assay buffer, containing HEPES (25 mM), MgCI2 (1 2 mM) and pH adjusted to 7 4 using KOH (5 M) The final potassium concentration in the assay was 13 mM On the day of the assay, pluronic acid was added to the buffer to get a 0 2% final concentration
After one hour on ice, the membrane-bead suspension was mixed with tritiated dofetilide (final concentration of 7 nM/well) and dispensed into a white Greiner polypropylene 384-well plate (10 ml/well), containing 0 1 ml of test compound in DMSO The assay plates were centrifuged at 10,000 rpm for 1 minute and left to equilibrate for 2-3 hrs at room temperature before reading on a ViewLux™
luminescence imager. Concentration response curve data were generated from an 11 -point inhibition curve (top assay concentration of 60 mM and a 1 :4 step-dilution), a four parameter curve-fit being applied.
3. Drugs and Materials
Compounds were dissolved in DMSO at a concentration of 10 mM. For concentration-response assays, dilutions were prepared in 100% DMSO using a 1 in 4 dilution step. The top assay concentration was 60 mM and the final DWISO assay concentration was 1%. On each test compound plate 0.1 ml of DMSO was included in one column to assess total binding (TB) and dofetilide or astemizole (10 mM final concentration) added to one column to determine non specific binding (NSB).
Four reference compounds were included in each assay for quality control (QC) purposes. The reference compounds used were: astemizole, dofetilide, quinidine and cisapride.
4. Data Analysis
For a single concentration assay, the results of each well are expressed as % inhibition:
% inhibition = 100 x (1- (sample - NSB) / (TB - NSB), where sample was the signal observed in a particular unknown sample well.
For concentration-response assays, the results of each test well were expressed as
% of controls. The normalization equation used was:
% control: 100 x (sample - NSB) / (TB - NSB)
The concentration-response curve was analyzed using a non linear equation, 4- parameter logistics, giving a determination of plC50.
Curve QC, plate QC (z' > 0.2) and assay QC (plC50 for reference compounds ± 0.5 log units of the rolling mean) were applied when analyzing all experiments.
Table 3
Figure imgf000082_0001
* WO2006/030925

Claims

Claims
1. A compound selected from the group consisting of
2,4-Difluoro-5-({[(1-{[2-methyl-6-({4-[(methyIsulfonyl)amino]phenyl}oxy)-3- pyridinyl]methylJ-4-piPeric'inyl)(phenyl)amino]carbonyi}amino)benzamide;
2,4-Difluoro-5-({[(3-fluorophenyl)(1-{[2-methyl-6-({4-
[(methylsulfonyl)amino]phenyl}oxy)-3-pyridiπyl]methyl}-4- piperidinyl)amino]carboπyl}amino)beπzamide;
5-{[(Butyl{1-[(2-methyl-6-{[4-(methylsu[foπyl)phenyl]oxy}-3-pyridinyl)methyl]-4- piperidinyl}amino)carboπyl]amino}-2,4-difluorobenzamide;
2,4-Difluoro-5-({t(3-fluorophenyl)(1-{[6-({4-[(2-hydroxyethyl)sulfonyI]phenyl}oxy)-2- . methyi-3-pyridinyl]methyl}-4-piperidinyl)amino]carbonyI}amino)benzamide;
2,4-Difluoro-5-({[(1-{[2-methyl-6-({4-[(methylsulfonyl)amino]phenyl}oxy)-3- pyridinyl]methyl}-4-piperidinyl)(3-thieriyl)amino]carbonyl}amino)benzamide;
2,4-Difluoro-5-({[(3-fluorophenyi)(1-{[2-methyl-6-({4-
[(methylsulfonyl)amino]phenyl}oxy)-3-pyridiπyl]methyl}-4- piperidiπyl)amino]carbonyl}amino)-Λ/-(1-methylethyl)benzamide;
5-({[(3-Fluorophenyl)(1-{[2-methyl-6-({4-[(methy!sulfonyl)amino]phenyl}oxy)-3- pyridinyl]methyl}-4-piperidinyl)amino]carbonyI}amino)-2-(trifluoromethyl)benzamide;
5-({[(2-Cyclopropylethyl)(1-{[2-methyl-6-({4-[(methylsulfonyl)amino]phenyl}oxy)-3- pyridinyl]methyl}-4-piperidinyl)amiπo]carbonyl}amino)-2,4-difluorobenzamide;
2,4-Difluoro-5-[({(3-fluorophenyl)[1 -({2-methyl-6-[(4-{[(1 - methylethyl)sulfoπyI]amino}phenyI)oxy]-3-pyridinyl}methyl)-4- piperidinyl]amino}carbonyl)amino]benzamide;
5-{{[(H[6-({4-[(Cyclopropylsulfonyl)amino]phenyI}oxy)-2-methyl-3-pyridinyl]methyl}-
4-piperidinyl)(3-fluorophenyi)amino]carbonyl}amino)-2,4-difluorobenzamide;
Λ/-(4-{[5-({4-[(3-Fluoropheπyl)({[4-methyl-3-(1,3-oxazol-2- yl)phenyl]amino}carbonyl)amino]-1-piperidinyl}methyl)-6-methyl-2- pyridinyl]oxy}phenyl)methaπesulfonamide;
Λ/-(4-{[5-({4-[({t2,4-Difluoro-5-(1 ,3,4-oxadiazol-2- yl)pheπyl]amino}carbonyl)(prιenyl)amiπo]-1-piperidinyl}methyl)-6-metrιyl-2- pyridinyl]oxy}phenyl)methanesulfonamide;
Λ/-(4-{[5-({4-[({[2,4-Difluoro-5-(1 ,3,4-oxadiazol-2-yl)phenyl]amino}carboπyI)(3- fluorophenyI)amino]-1-piperidiπyl}methyl)-6-rnethyl-2- pyridinyl]oxy}phenyl)methanesulfonamide;
Λ/-(4-{[5-({4-[({[2,4-Difluoro-5-(5-methyl-1 ,3,4-oxadiazol-2- yl)pheny[]amino}carbonyl)(phenyl)amino]-1-piperidinyl}methyl)-6-methyl-2- pyridinyl]oxy}phenyl)methanesulfonamide;
Λ/-(4-{[5-({4-[({[4-Fluoro-3-(1,3-oxazol-2-y[)pheπyl]amiπo}carbonyl)(3- fluorophenyl)amino]-1-piperidinyl}rnethyl)-6-methyl-2- pyridiny[]oxy}phenyl)methanesulfonamide;
5-({[butyl(1-{[2-cyclopropyl-6-({4-[(methyIsulfonyl)amino]phenyl}oxy)-3- pyridinyl]methyl}-4-piperidinyl)amino]carbonyl}amino)-2,4-difluorobenzamide;
5-({[(1-{[2-cyclopropyl-6-({4-[(methylsulfonyl)amino]phenyl}oxy)-3-pyridinyl]methyl}-
4-piperidinyl)(phenyl)amino]carbonyl}amino)-2,4-difluorobenzamide and pharmaceutically acceptable salts thereof.
2. A method of treatment or prevention of a HIV infection in a human comprising administering to said human an effective amount of a compound according to claim 1.
3. A method of treatment or prevention of neuropathic pain, multiple sclerosis, rheumatoid arthritis, autoimmune diabetes, chronic implant rejection, asthma, rheumatoid arthritis, Crohns Disease, inflammatory bowel disease, chronic inflammatory disease, glomerular disease, nephrotoxic serum nephritis, kidney disease, Alzheimer's Disease , autoimmune encephalomyelitis, arterial thrombosis, allergic rhinitis, arteriosclerosis, Sjogren's syndrome (dermatomyositis), systemic lupus erythematosus, graft rejection, cancers with leukocyte infiltration of the skin or organs, human papilloma virus infection, prostate cancer, wound healing, amyotrophic lateral sclerosis and immune mediated disorders in a human comprising administering to said human a pharmceutically effective amount of a compound according to claim 1.
4. A compound according to claim 1 for use in medical therapy.
5. Use of a compound according to claim 1 in the manufacture of a medicament for the treatment or prophylaxis of an HIV infection.
6. The use of a compound according to claim 1 in the manufacture of a medicament for the treatment or prophylaxis of neuropathic pain, multiple sclerosis, rheumatoid arthritis, autoimmune diabetes, chronic implant rejection, asthma, rheumatoid arthritis, Crohns Disease, inflammatory bowel disease, chronic inflammatory disease, glomerular disease, nephrotoxic serum nephritis, kidney disease, Alzheimer's Disease , autoimmune encephalomyelitis, arterial thrombosis, allergic rhinitis, arteriosclerosis, Sjogren's syndrome (dermatomyositis), systemic lupus erythematosus, graft rejection, cancers with leukocyte infiltration of the skin or organs, human papilloma virus infection, prostate cancer, wound healing, amyotrophic lateral sclerosis and immune mediated disorders.
7. A pharmaceutical composition comprising a pharmaceuticaily effective amount of a compound according to claim 1 together with a pharmaceutically acceptable carrier.
8. A pharmaceutical composition according to claim 7 in the form of a tablet or capsule.
9. A pharmaceutical composition according to claim 7 in the form of a liquid.
10. A method of treatment or prevention of an HIV infection in a human comprising administering to said human a composition comprising a compound according to claim 1 and another therapeutic agent.
11. A method according to claim 10, wherein said composition comprises another therapeutic agent selected from the group consisting of acyclic nucleosides, acyclovir, valaciclovir, famciclovir, ganciclovir, penciclovir, acyclic nucleoside phosphonates, (S)-1 -(3-hydroxy-2-phosphonyl-methoxypropyl)cytosine (HPMPC),
[[[2-(6-amino-9H-purin-9- yl)ethoxy]methyl]phosphinylidene]bis(oxymethylene)-2,2- dimethylpropanoic acid (bis-POM PMEA, adefovir dipivoxil), [[(1 R)-2-(6-amino-9H- purin-9-yl)-1-methylethoxy]methyl]phosphonic acid (tenofovir), (R)-[[2-(6-Amino-9H- purin-9-yl)-1-methylethoxy)methyl]phosphonic acid bis- (isopropoxycarbonyloxymethyl)ester (bis-POC-PMPA), nucleoside reverse transcriptase inhibitors, 3'-azido-3'-deoxythymidine (AZT, zidovudine), 2', 3'- dideoxycytidine (ddC, zalcitabine), 2',3'-dideoxyadenosine, 2',3'-dideoxyinosine (ddl, didanosine), 2',3'-didehydrothymidine (d4T, stavudine), (-)-cis-1-(2-hydroxymethyl)- 1 ,3-oxathioIane 5-yl)-cytosine (lamivudine), cjs-1-(2-(hydroxymethyl)-1 ,3- oxathiolan-5-yl)-5-fluorocytosine (FTC), (-J-cis-Λ-p-amino-δ-fcyclopropylamiπoJ-θhH- purin-9-yl]-2-cyclopentene-1 -methanol (abacavir), ribavirin, protease inhibitors, indinavir, ritonavir, nelfinavir, amprenavir, saquinavir, fosamprenavir, lopinavir, tipranavir, interferons, α-interferon, immunomodulators, interleukin Il or thymosin, granulocyte macrophage colony stimulating factors, erythropoetin, soluble CD4 and genetically engineered derivatives thereof, non-nucleoside reverse transcriptase inhibitors (NNRTIs), nevirapine (BI-RG-587), alpha-((2-acetyl-5- methylphenyl)amino)-2,6-dichIoro-benzeneacetamide (loviride), 1 -[3- (isopropylamino)-2-pyridylH-[5-(methanesulfonarnido)-1 H-indol-2- ylcarbonyl]piperazine monomethanesulfonate (delavirdine), (S)-6-chloro-4- (cyclopropylethynyl)-i ,4-dihydro-4-(trifluoromethyl)-2H-3, 1 -benzoxazin-2-one (efavirenz, DMP 266), rilpivirine, integrase inhibitors, fusion inhibitors, T-20 and T- 1249.
PCT/US2008/081662 2007-10-31 2008-10-30 Ccr5 antagonists as therapeutic agents WO2009058919A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US98408407P 2007-10-31 2007-10-31
US60/984,084 2007-10-31

Publications (1)

Publication Number Publication Date
WO2009058919A1 true WO2009058919A1 (en) 2009-05-07

Family

ID=40591446

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2008/081662 WO2009058919A1 (en) 2007-10-31 2008-10-30 Ccr5 antagonists as therapeutic agents

Country Status (1)

Country Link
WO (1) WO2009058919A1 (en)

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2010101299A1 (en) * 2009-03-05 2010-09-10 住友化学株式会社 Method for producing halogen-substituted benzene dimethanol
CN106928099A (en) * 2017-04-27 2017-07-07 重庆丽澄环保科技有限公司 A kind of synthetic method of benzene sulfonyl hydrazide

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2006030925A1 (en) * 2004-09-13 2006-03-23 Ono Pharmaceutical Co., Ltd. Nitrogenous heterocyclic derivative and medicine containing the same as an active ingredient

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2006030925A1 (en) * 2004-09-13 2006-03-23 Ono Pharmaceutical Co., Ltd. Nitrogenous heterocyclic derivative and medicine containing the same as an active ingredient
CA2579501A1 (en) * 2004-09-13 2006-03-23 Ono Pharmaceutical Co., Ltd. Nitrogenous heterocyclic derivative and medicine containing the same as an active ingredient

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2010101299A1 (en) * 2009-03-05 2010-09-10 住友化学株式会社 Method for producing halogen-substituted benzene dimethanol
CN106928099A (en) * 2017-04-27 2017-07-07 重庆丽澄环保科技有限公司 A kind of synthetic method of benzene sulfonyl hydrazide

Similar Documents

Publication Publication Date Title
CA2753434C (en) Soluble guanylate cyclase activators
TWI344955B (en) Heterocyclic rinf having nitrogen atom derivatives and medicament containing the derivatives as active ingredient
US7071213B2 (en) Cannabinoid receptor ligands
KR101456721B1 (en) New compounds
US8153658B2 (en) Piperidine derivative or salt thereof
CA3104131A1 (en) Inhibitors of cyclin-dependent kinases
JP2009542640A (en) New pyridine analogues
CA2938855C (en) 1,2-substituted cyclopentanes as orexin receptor antagonists
EA015835B1 (en) 4-oxa(thia)methylpiperidine derivatives, use thereof as antidiabetic compounds
SK8762001A3 (en) Piperidines as CCR5 modulators
JP2007522197A (en) Pyrimidine derivatives as orexin receptor antagonists
TW200806610A (en) Compounds for the treatment of inflammatory disorders and microbial diseases
JP2012529535A (en) Nicotinamide compounds useful as kinase modulators
US8367698B2 (en) 3-substituted sulfonyl piperidine derivative
NO326349B1 (en) CCR5 antagonists for the treatment of AIDS, pharmaceuticals containing them, and their use in the manufacture of medicine.
TW200932727A (en) New acetyl coenzyme a carboxylase (ACC) inhibitors and uses in treatments of obesity and diabetes mellitus
MXPA05006381A (en) Novel piperidine derivatives as modulators of chemokine receptor ccr5.
JPWO2002022588A1 (en) New pyrimidine and pyridine derivatives
WO2009058919A1 (en) Ccr5 antagonists as therapeutic agents
WO2009058921A1 (en) Ccr5 antagonists as therapeutic agents
EP1219605A1 (en) Urea compounds, process for producing the same and use thereof
WO2009058923A1 (en) Ccr5 antagonists as therapeutic agents
RU2321586C2 (en) Derivatives of thiazole as npy antagonists
WO2009058924A1 (en) Ccr5 antagonists as therapeutic agents
WO2009075960A1 (en) Ccr5 antagonists as therapeutic agents

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: 08843446

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 08843446

Country of ref document: EP

Kind code of ref document: A1