WO2004055016A1 - Pyrrolidine and azetidine compounds as ccr5 antagonists - Google Patents

Pyrrolidine and azetidine compounds as ccr5 antagonists Download PDF

Info

Publication number
WO2004055016A1
WO2004055016A1 PCT/US2003/039618 US0339618W WO2004055016A1 WO 2004055016 A1 WO2004055016 A1 WO 2004055016A1 US 0339618 W US0339618 W US 0339618W WO 2004055016 A1 WO2004055016 A1 WO 2004055016A1
Authority
WO
WIPO (PCT)
Prior art keywords
optionally substituted
compound
dichlorophenyl
triazaspiro
decan
Prior art date
Application number
PCT/US2003/039618
Other languages
French (fr)
Inventor
Hanbiao Yang
Wieslaw Mieczyslaw Kazmierski
Christopher Joseph Aquino
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
Priority to US10/538,134 priority Critical patent/US7271172B2/en
Priority to EP03813415A priority patent/EP1569933B1/en
Priority to JP2004560830A priority patent/JP2006511552A/en
Priority to AU2003296992A priority patent/AU2003296992A1/en
Priority to DE60322423T priority patent/DE60322423D1/en
Publication of WO2004055016A1 publication Critical patent/WO2004055016A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D471/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00
    • C07D471/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00 in which the condensed system contains two hetero rings
    • C07D471/10Spiro-condensed systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/04Drugs for disorders of the alimentary tract or the digestive system for ulcers, gastritis or reflux esophagitis, e.g. antacids, inhibitors of acid secretion, mucosal protectants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • A61P11/06Antiasthmatics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • A61P13/12Drugs for disorders of the urinary system of the kidneys
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/02Drugs for dermatological disorders for treating wounds, ulcers, burns, scars, keloids, or the like
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/08Drugs for skeletal disorders for bone diseases, e.g. rachitism, Paget's disease
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P21/00Drugs for disorders of the muscular or neuromuscular system
    • A61P21/04Drugs for disorders of the muscular or neuromuscular system for myasthenia gravis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/28Drugs for disorders of the nervous system for treating neurodegenerative disorders of the central nervous system, e.g. nootropic agents, cognition enhancers, drugs for treating Alzheimer's disease or other forms of dementia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P27/00Drugs for disorders of the senses
    • A61P27/16Otologicals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/08Drugs for disorders of the metabolism for glucose homeostasis
    • A61P3/10Drugs for disorders of the metabolism for glucose homeostasis for hyperglycaemia, e.g. antidiabetics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/04Antibacterial agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/14Antivirals for RNA viruses
    • A61P31/18Antivirals for RNA viruses for HIV
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/06Immunosuppressants, e.g. drugs for graft rejection
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/08Antiallergic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/10Drugs for disorders of the cardiovascular system for treating ischaemic or atherosclerotic diseases, e.g. antianginal drugs, coronary vasodilators, drugs for myocardial infarction, retinopathy, cerebrovascula insufficiency, renal arteriosclerosis

Definitions

  • HIV human immunodeficiency virus
  • AIDS acquired immunodeficiency syndrome
  • ARC AIDS-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.
  • the present invention features compounds that are useful in the inhibition of HIV replication, the prevention of infection by HIV, 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 antivirals, 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 includes compounds of formula (I)
  • Ring A is a saturated, partially saturated, or aromatic 3-7 monocyclic or 8-10 membered bicyclic ring having one ring nitrogen and 0-4 additional heteroatoms selected from oxygen, phosphorus, sulfur, or nitrogen;
  • Ring B is a saturated 4 or 5 membered ring containing the depicted ring nitrogen;
  • R 1 is alkyl optionally substituted by one or more R 7 , alkenyl optionally substituted by one or more R 7 , alkynyl optionally substituted by one or more R 7 , cycloalkyl optionally substituted by one or more R 8 , heterocyclyl optionally substituted by one or more R 8 , heteroaryl optionally substituted by one or more R 6 , or aryl optionally substituted by one or more R 6 ; or R 1 and X taken together form a saturated, partially saturated or aromatic 5-6 membered ring having 0-3 heteroatoms selected from oxygen, phosphorus, sulfur, or nitrogen that is fused to Ring A; each R 2 is independently selected from -OR 0 , -C(O)-R°, -S(O) 2 -R°, -C(O)-N(R°) 2 , -S(O) 2 -N(R°) 2 , -(CH 2 ) a -N(R°)(-V
  • V is -C(O)-, -C(O)O-, -S(O) 2 -, or -C(O)-N(R°)-;
  • R + is alkyl, cycloalkyl, aralkyl, aryl, heteroaryl, heteroaralkyl, or heterocyclyl, wherein said R + is optionally substituted by one or more R 8 ; m is 0 or 1 ; n is 0-5;
  • R 3 is H, -N(R°) 2 , -N(R°)C(O)R°, -CN, halogen, CF 3 , alkyl optionally substituted by one or more groups selected from R 7 or -S-aryl optionally substituted by -(CH 2 )- ⁇ - 6 - N(R°)SO 2 (R°), alkenyl optionally substituted by one or more groups selected from R 7 or -S-aryl optionally substituted by -(CH 2 ) 1 - 6 -N(R°)SO 2 (R 0 ), alkynyl optionally substituted by one or more groups selected from R 7 or -S-aryl optionally substituted by -(CH 2 ) 1 - 6 -N(R°)SO 2 (R°), cycloalkyl or carbocyclyl optionally substituted by one or more R 8 , aryl optionally substituted by one or more R 6 , heteroaryl optionally substituted by one or more R
  • Y is alkyl, alkenyl, alkynyl, -(CR 4 R 5 ) P -, -C(O)-, -C(O)C(O)-, -C(S)-, -O-(CH 2 ) M -C(O)-, -(CH 2 )o--rC(O)-O-, -N(R°)-C(O)-, -C(O)-N(R 0 )-, -N(R°)-C(S)-,
  • each R 4 is independently H, alkyl optionally substituted by R 7 , alkenyl optionally substituted by R 7 , or alkyny
  • the B ring is pyrrolidine.
  • R 9 is H, while in another embodiment R 9 is oxo.
  • R 1 is optionally substitued aryl.
  • R 1 is phenyl mono- or di- substituted with halogen.
  • R 1 may be phenyl di-substituted with Cl.
  • -(Y) m -R 3 suitably is
  • X is -(CH 2 )-, -(CH 2 -CH 2 )-, or -(CH 2 -CH 2 -CH 2 )-.
  • X is optionally substituted by one or more halogen or oxo.
  • X optionally has 1-3 heteroatoms selected from oxygen, phosphorus, sulfur, or nitrogen.
  • the A ring is selected from the following, where the asterisk (*) indicates the preferred, but not limiting, point(s) of substitution:
  • each R 2 with the asterisk (*) indicating a preferred, but not limiting, point of substitution from Ring A, independently is selected from
  • the ring A with two geminal R 2 s, is selected from:
  • a ring is tropane or piperidine, either optionally substituted with one or more R 2 .
  • A-R 2 is comprised of one of the following:
  • the A ring contains at least one additional nitrogen atom and said A ring optionally is N-substituted.
  • the A ring is N-substituted with - (CH 2 ) a -(V b -R+).
  • Another aspect of the present invention includes a method of treatment
  • the viral infection is an HIV infection.
  • Another aspect of the present invention includes a method of treatment
  • bacterium is Yersinia pestis.
  • Another aspect of the present invention includes a method of treatment
  • Another aspect of the present invention includes a compound of the present invention for use in medical therapy.
  • Another aspect of the present invention includes the use of a compound of the present invention in the manufacture of a medicament for the treatment (including prophylaxis) of a viral infection.
  • a viral infection is a HIV infection.
  • Another aspect of the present invention includes the use of a compound of the present invention in the manufacture of a medicament for the treatment, including prophylaxis, of a bacterial infection.
  • the bacterium is Yersinia pestis.
  • Another aspect of the present invention includes the use of a compound of the present invention in the manufacture of a medicament for the treatment, including prophylaxis, of 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, infectious disorders including bubonic and pneumonic plague, human papilloma virus infection, prostate cancer, wound healing, amyotrophic lateral sclerosis and immune mediated disorders.
  • prophylaxis of multiple sclerosis, rheuma
  • Another aspect of the present invention includes a pharmaceutical composition comprising a pharmaceutically effective amount of a compound of the present invention together with a pharmaceutically acceptable carrier.
  • the pharmaceutical composition is in the form of a tablet, capsule, or liquid.
  • Another aspect of the present invention includes a method of treatment, including prevention, of a viral infection in a mammal comprising administering to said mammal a composition comprising a compound of the present invention and another therapeutic agent.
  • the composition comprises one or more additional therapeutic agent selected from the group consisting of (1 -alpha, 2-beta, 3- alpha)-9-[2,3-bis(hydroxymethyl)cyclobutyl]guanine [(-)BHCG, SQ-34514, lobucavir], 9-[(2R,3R,4S)-3,4-bis(hydroxymethyl)-2-oxetanosyl]adenine (oxetanocin-G), 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-dimethylpropanoi
  • Another aspect of the present invention includes a method of treatment, including prevention, of a viral infection in a mammal comprising administering to said mammal a composition comprising a compound of the present invention and ritonavir.
  • Particular compounds of the present invention include 22
  • the compounds of the present invention include: tert-butyl 3-(3,4-dichlorophenyl)-3-[3-(4-oxo-1-phenyl-1 ,3,8-triazaspiro[4.5]dec-8- yI)propyl]pyrrolidine-1-carboxylate;
  • alkyl refers to a straight-chain or branched-chain saturated aliphatic hydrocarbon radical containing the specified number of carbon atoms.
  • alkyl radicals include, but are not limited to, methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, sec-butyl, tert-butyl, pentyl, isoamyl, n-hexyl and the like.
  • cycloalkyl refers to a monocyclic or polycyclic non-aromatic hydrocarbon ring radical having three to twenty carbon atoms, preferably from three to twelve carbon atoms, and more preferably from three to ten carbon atoms. If polycyclic, each ring in a carbocylyl radical is non-aromatic unless otherwise indicated.
  • a carbocylyl radical is either completely saturated or contains one or more units of unsaturation but is not aromatic. The unsaturation, if present, may occur in any point in the ring that may result in any chemically stable configuration.
  • cycloalkyl also includes hydrocarbon rings that are fused to one or more aromatic rings, such as in tetrahydronaphthyl, where the radical or point of attachment is on the non-aromatic ring.
  • cycloalkyl also includes each possible positional isomer of a non-aromatic hydrocarbon radical, such as in 1-decahydronaphthyl, 2- decahydronaphthyl, 1 -tetrahydronaphthyl and 2-tetrahydronaphthyl.
  • Suitable cycloalkyl groups include, but are not limited to, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cyclohexenyl, decahydronaphthyl, tetrahydronaphthyl and the like.
  • alkenyl refers to a straight-chain or branched-chain alkyl group with at least one carbon-carbon double bond.
  • alkenyl radicals include, but are not limited to, ethenyl, propenyl, isopropenyl, butenyl, isobutenyl, pentenyl, hexenyl, hexadienyl and the like.
  • alkynyl refers to hydrocarbon groups of either a straight or branched configuration with one or more carbon-carbon triple bonds which may occur in any stable point along the chain, such as ethynyl, propynyl, butynyl, pentynyl, and the like.
  • alkoxy refers to an alkyl ether radical, wherein the term “alkyl” is defined above.
  • suitable alkyl ether radicals include, but are not limited to, methoxy, ethoxy, n-propoxy, isopropoxy, n-butoxy, isobutoxy, sec-butoxy, tert- butoxy and the like.
  • aryl refers to an aromatic monocyclic or polycyclic hydrocarbon ring radical containing five to twenty carbon atoms, preferably from six to fourteen carbon atoms, and more preferably from six to ten carbon atoms. Also included within the scope of the term “aryl”, as it is used herein, is a group in which an aromatic hydrocarbon ring is fused to one or more non-aromatic carbocyclic or heteroatom-containing rings, such as in an indanyl, phenanthridinyl or tetrahydronaphthyl, where the radical or point of attachment is on the aromatic hydrocarbon ring.
  • aryl also includes each possible positional isomer of an aromatic hydrocarbon radical, such as in 1-naphthyl, 2- naphthyl, 5-tetrahydronaphthyl, 6-tetrahydronaphthyl, 1 -phenanthridinyl, 2- phenanthridinyl, 3-phenanthridinyl, 4-phenanthridinyl, 7-phenanthridinyl, 8- phenanthridinyl, 9-phenanthridinyl and 10-phenanthridinyl.
  • aryl radicals include, but are not limited to, phenyl, naphthyl, indenyl, azulenyl, fluorenyl, anthracenyl, phenanthrenyl, tetrahydronaphthyl, indanyl, phenanthridinyl and the like.
  • aralkyl further refers to groups of -R a R b , where R a is an alkylene as defined herein and Rb is an aryl as defined herein.
  • heterocycle refers to a non-aromatic monocyclic or polycyclic ring radical containing three to twenty carbon atoms, preferably three to seven carbon atoms if monocyclic and eight to eleven carbon atoms if bicyclic, and in which one or more ring carbons, preferably one to four, are each replaced by a heteroatom such as N, O, and S. If polycyclic, each ring in a heterocyclyl radical is non-aromatic unless otherwise indicated.
  • a heterocyclic ring may be fully saturated or may contain one or more units of unsaturation but is not aromatic.
  • the unsaturation may occur in any point in the ring that may result in any chemically stable configuration.
  • the heterocyclic ring may be attached at a carbon or heteroatom that results in the creation of a stable structure.
  • Preferred heterocycles include 5-7 membered monocyclic heterocycles and 8-10 membered bicyclic heterocycles.
  • heterocycle is a group in which a non-aromatic heteroatom-containing ring is fused to one or more aromatic rings, such as in an indolinyl, chromanyl, phenanthridinyl or tetrahydroquinolinyl, where the radical or point of attachment is on the non-aromatic heteroatom-containing ring.
  • heterocycle also includes each possible positional isomer of a heterocyclic radical, such as in 1-decahydroquinoline, 2-decahydroquinoline, 3- decahydroquinoline, 4-decahydroquinoline, 5-decahydroquinoline, 6- decahydroquinoline, 7-decahydroquinoline, 7-decahydroquinoline, 8- decahydroquinoline, 4a-decahydroquinoline, 8a-decahydroquinoline, 1 -indolinyl, 2- indolinyl, 3-indolinyl, 1-tetrahydroquinoline, 2-tetrahydroquinoline, 3- tetrahydroquinoline and 4-tetrahydroquinoline.
  • heterocyclylalkyl refers to an alkyl group substituted by a heterocyclyl.
  • heterocyclic groups include, but are not limited to, imidazolinyl, 2,3-dihydro-1 H-imidazolyl, imidazolidinyl, indazolinolyl, perhydropyridazyl, pyrrolinyl, pyrrolidinyl, 4H-pyrazolyl, piperidinyl, pyranyl, pyrazolinyl, piperazinyl, morpholinyl, thiamorpholinyl, thiazolidinyl, thiamorpholinyl, oxopiperidinyl, oxopyrrolidinyl, azepinyl, tetrahydrofuranyl, oxoazepinyl, tetrahydropyranyl, thiazolyl, dioxolyl, dioxinyl, oxathiolyl, benzodioxolyl, dithiolyl, dithiolanyl, tetra
  • heteroaryl refers to an aromatic monocyclic or polycyclic ring radical containing five to twenty carbon atoms, preferably five to ten carbon atoms, in which one or more ring carbons, preferably one to four, are each replaced by a heteroatom such as N, O, and S.
  • Preferred heteroaryl groups include 5-6 membered monocyclic heteroaryls and 8-10 membered bicyclic heteroaryls.
  • heteroaryl is a group in which a heteroaromatic ring is fused to one or more aromatic or non-aromatic rings where the radical or point of attachment is on the heteroaromatic ring.
  • heteroaromatic rings include, but are not limited to, pyrido[3,4-d]pyrimidinyl, 7,8-dihydro-pyrido[3,4-d]pyrimidine and 5,6,7,8-tetrahydro-pyrido[3,4-d]pyrimidine.
  • heteroaryl also includes each possible positional isomer of a heteroaryl radical, such as in 2-pyrido[3,4-d]pyrimidinyl and 4-pyrido[3,4-d]pyrimidinyl.
  • heteroaryl groups include, but are not limited to, imidazolyl, quinolyl, isoquinolyl, indolyl, indazolyl, pyridazyl, pyridyl, pyrrolyl, pyrazolyl, pyrazinyl, quinoxalyl, pyrimidinyl, pyridazinyl, furyl, thienyl, triazolyl, thiazolyl, carbazolyl, carbolinyl, tetrazolyl, benzofuranyl, oxazolyl, benzoxazolyl, isoxozolyl, isothiazolyl, thiadiazolyl, furazanyl, oxadiazolyl, benzimidazolyl, benzothienyl, quinolinyl, benzotriazolyl, benzothiazolyl, isoquinolinyl, isoindolyl, acridinyl and benzoisox
  • heteroaryl further refers to groups of -R a R b , where R a is an alkylene as defined herein and R is a heteroaryl as defined herein.
  • heteroatom means nitrogen, oxygen, phosphorus, or sulfur and includes any oxidized forms thereof, including as non-limiting examples oxidized forms of nitrogen such as N(O) ⁇ N + -O " ⁇ , oxidized forms of sulfur such as S(O) and S(O) 2 , and the quaternized form of any basic nitrogen.
  • halogen refers to fluorine, chlorine, bromine or iodine.
  • 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 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.
  • prophylaxis refers to preventing a disease or condition or preventing the occurrence of symptoms of such a disease or condition, in a patient.
  • 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 derivative means any pharmaceutically acceptable salt, ester, salt of an ester, or other derivative of a compound of this invention which, upon administration to a recipient, is capable of providing (directly or indirectly) a compound of this invention or an inhibitorily active metabolite or residue thereof.
  • Particularly favored derivatives and prodrugs are those that increase the bioavailability of the compounds of this invention when such compounds are administered to a mammal (e.g., by allowing an orally administered compound to be more readily absorbed into the blood) or which enhance delivery of the parent compound to a biological compartment (e.g., the brain or lymphatic system) relative to the parent species.
  • 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, maleic, phosphoric, glycollic, lactic, salicyclic, succinic, toluene-p-sulfonic, tartaric, acetic, citric, 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 ⁇ 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 + (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.
  • Esters of the compounds of the present invention are independently selected from the following groups: (1 ) carboxylic acid esters obtained by esterification of the hydroxy groups, in which the non-carbonyl moiety of the carboxylic acid portion of the ester grouping is selected from straight or branched chain alkyl (for example, acetyl, n-propyl, t-butyl, or n-butyl), alkoxyalkyl (for example, methoxymethyl), aralkyl (for example, benzyl), aryloxyalkyl (for example, phenoxymethyl), aryl (for example, phenyl optionally substituted by, for example, halogen, or amino); (2) sulfonate esters, such as alkyl- or aralkylsulfonyl (for example, methanesulfonyl); (3) amino acid esters (for example, L-valyl or L-isoleucyl); (4) phosphonate esters and (5) mono
  • any alkyl moiety present advantageously contains from 1 to 18 carbon atoms, particularly from 1 to 6 carbon atoms, more particularly from 1 to 4 carbon atoms, Any cycloalkyl moiety present in such esters advantageously contains from 3 to 6 carbon atoms. Any aryl moiety present in such esters advantageously comprises a phenyl group.
  • the compounds according to the invention contain one or more asymmetric carbon atoms and thus occur as racemates and racemic mixtures, single enantiomers, diastereomeric mixtures and individual diastereoisomers. All such isomeric forms of these compounds are expressly included in the present invention.
  • Each stereogenic carbon may be of the R or S configuration.
  • structures depicted herein are also meant to include compounds which differ only in the presence of one or more isotopically enriched atoms.
  • compounds having the present structures except for the replacement of a hydrogen by a deuterium or tritium, or the replacement of a carbon by a 13 C- or 1 C-enriched carbon are also within the scope of this invention.
  • the present invention features compounds according to the invention for use in medical therapy, for example for the treatment including 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.
  • ARC AIDS related complex
  • Kaposi's sarcoma Kaposi's sarcoma
  • AIDS dementia AIDS related complex
  • the present invention features use of the compounds of the present invention in the manufacture of a medicament for the treatment including 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 including 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 including 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 treatment including prevention of other CCR5-related diseases and conditions, including 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, infectious disorders including bubonic and pneumonic plague, human papilloma virus infection, prostate cancer, wound healing, amyotrophic lateral sclerosis, immune mediated disorders.
  • CCR5-related diseases and conditions including 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 including 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 including 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 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.
  • agents that are effective for the treatment of viral infections or associated conditions include agents that are effective for the treatment of viral infections or associated conditions.
  • these agents are (1- alpha, 2-beta, 3-alpha)-9-[2,3-bis(hydroxymethyl)cyclobutyl]guanine [(-)BHCG, SQ- 34514, lobucavir], 9-[(2R,3R,4S)-3,4-bis(hydroxymethyl)-2-oxetanosyl]adenine
  • 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-dimethylpropanoic acid (bis- POM PMEA, adefovir dipivoxil), [[(1 R)-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 (tenofovir), and (R
  • 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.
  • 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, suitably in the range of 0.1 to 100 mg per kilogram body weight per day and most suitably 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, suitably 20 to 500 mg, and most suitably 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, 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.
  • 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%, suitably 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.
  • 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.
  • compositions suitable for parenteral administration include aqueous and nonaqueous isotonic sterile injection solutions which may contain anti- oxidants, 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 microparticulate 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.
  • NMO N-methylmorpholine N-oxide
  • the synthesis of the C2-pyrrolidine scaffold was carried out by two procedures. Alkylation of 1 with methyl bromoacetate resulted in a separable mixture of monoalkylated 2 and bis-alkylated 8, each of which was subjected to the desired 10 by a separate route.
  • Compound 2 was converted to 10 in five steps and in 28% overall yield, while the conversion of 8 to 10 could be accomplished in 4 steps and in a higher overall yield of 38%.
  • Example 3 8- ⁇ 3-[3-(3,4-dichlorophenyl)-1 -(isoxazol-5-ylcarbonyl)pyrrolidin-3-yl]propyl ⁇ -1 -phenyl- 1 ,3,8-triazaspiro[4.5]decan-4-one
  • Compound 13 was synthesized by reacting amine 20 with chlorobenzoxazole.
  • Additional compounds of the present invention include oxygen linked compounds as illustrated hereinbelow.
  • Intermediate Compound 5 may be protected with an appropriate protecting group, as will be appreciated by those skilled in the art: 1 ,1- dimethylethyl 3-(3,4-dichlorophenyl)-3-(2-propen-1-yloxy)-1-pyrrolidinecarboxylate
  • Example 23 8- ⁇ 3-[3-(3,4-dichlorophenyl)-1 -(2-furanylcarbonyl)-3- azetidinyl]propyl ⁇ -1-phenyl-1,3,8-triazaspiro[4.5]decan-4-one.
  • Example 24 8- ⁇ 3-[1 -(cyclopentylcarbonyl)-3-(3,4-dichlorophenyl)-3- azetidinyl]propyl ⁇ -1-phenyl-1,3,8-triazaspiro[4.5]decan-4-one.
  • CCR5 SPA CC-Chemokine Receptor-5 Binding by Scintillation Proximity Assay
  • This protocol describes a high-throughput screen using SPA binding to identify compounds that inhibit binding of 125 l-MIP1 ⁇ to the human CCR5 chemokine receptor.
  • CCR5 is a G protein-coupled receptor that binds the natural chemokine ligands
  • CCR5 acts as a co-receptor with CD4 for entry of HIV- 1 into human T-cells and monocytes.
  • Chemokines also play a role in acute and chronic inflammatory processes. Chemokines are soluble proteins produced and released by a wide variety of cell types during the initial phase of a host response to a forgein substance entering the body.
  • Human CCR5 receptors were expressed in Chinese Hamster Ovary (CHO) cells, registration # 12025. Cells were grown in suspension and a 50 to 80 ml CCR5 cell pellet was prepared.
  • Membrane preparation 1) Weigh pellet; 2)Prepare an ice-cold 50mM HEPES buffer, containing 0.0025mg/ml Pefabloc, 0.0001 mg/ml Pepstatin A, 0.0001 mg/ml Leupeptin, 0.0001 mg/ml Aprotinin (protease inhibitor cocktail), pH 7.4; 3) Homogenize pellet in 5 volumes of HEPES buffer; 4) Homogenize again with a glass homogenizer 10 to 20 strokes; 5) Centrifuge homogenate at 18,000 rpm in a F28/36 rotor using a Sorvall RC26 PIUS refrigerated Centrifuge for 30 minutes; 6) Discard supernatant and resuspend pellet in 3 volumes of HEPES buffer; 7) Homogenize and centr
  • the saturation curve binding experiment is performed by adding varying amounts of [ 125 l]MIP1 (0-8.5nM to membranes and beads in concentrations chosen from the optimal protein/bead ratio.
  • the data is analyzed using a non-linear curve-fitting program.
  • the K d and Bmax are derived from the curve.
  • Bacitracin 50mg/ml is dissolved in deionized water, brought to a boil for 5 minutes (to destroy protease activity) and cooled. Prepared 1ml aliquots and store at -80°C.
  • Protease inhibitor cocktail is prepared by dissolving 25 mg/ml of Pefabloc, 1 mg/ml of Leupeptin, 1 mg/ml of Aprotinin and 1 mg/ml of Pepstatin A in 100% DMSO. The cocktail can be aliquoted and stored frozen at -20°C until needed.
  • Sigmacote Any reagent bottles and reservoirs that come in contact with the radioligand are treated with Sigmacote to reduce sticking. Rinse containers with undiluted Sigmacote; rinse with deionized water several times, and allow to air dry before using.
  • Compounds Preparation Compounds for a single concentration determination (One Shots) are delivered in 96 well Packard Optiplates containing 1 ⁇ l of compound in 100% DMSO in columns A1- H10 (80 compounds/plate). Column A11 to H11 is used for total binding (Bo) (vehicle-5 ⁇ l of the appropriate DMSO concentration) and column A12 to D12 is used for determination of nonspecific binding. No further preparation is required.
  • Assay Buffer Preparation 50 mM HEPES buffer pH 7.4, 1mM CaCl 2, 5mM MgCI2(this can be made ahead as a 10OX stock ), 1 % BSA, 0.5 mg/ml Bacitracin, Protease inhibitor Cocktail (see preparation above) 100 uL/100 ml, DMSO is added to equal a final concentration of 2% per well (includes compound %DMSO) if needed.
  • Membrane and SPA Bead preparation The final assay concentration for the membrane is 15 ⁇ g per well.
  • SPA beads are prepared by adding 5 ml of assay buffer to a 500 mg vial. The final concentration of SPA beads in the assay is 0.25 mg/well.
  • Membranes and beads are premixed as a 1 :1 (membrane:bead) mixture and maintained at mixture at 4°C with constant stirring. 50 ⁇ l of the mixture is added to each assay well. After all reagents have been added to the plates (total assay volume 100 ⁇ l), shake plates for 4 hours at room temperature. After 4 hours, place the plates on the TopCount in a count the plates on the TopCount for 30 sec per well using an appropriate program (i.e., one with a quench curve established for the conditions of the assay. Data Reduction:
  • the result of each test well is expressed as % inhibition using the following formula: 100*(1-(U1-C2)/(C1-C2)).
  • U1 is the unknown sample in cpm observed in a particular well
  • C1 is the average of column 12 cpm observed in the absence of any added inhibitor
  • C2 is the average of column 11 cpm observed in the presence of 1 ⁇ M of MIP1 .
  • %B/Bo % total specific binding
  • Each plate contains 12 wells of total binding (column A11-H11). The cpm/well are averaged and are used in data reduction as value d .Each plate also contains 4 wells of non-specific binding (wells A12-D12). The counts of these wells are averaged and used in data reduction as value C2.
  • a standards plate is included in each experiment. This plate contains a 14-point concentration-response curve (in triplicate) for the standard compound MIP1 ⁇ at a starting concentration of 1 ⁇ M. The average historical pKj obtained with MIP1 ⁇ is 7.6.
  • the relevant biological response field for a single concentration is % inhibition. Inhibition values of >40 or >50% were considered positive responses.
  • HOS-LTR-Luciferase Assay The relevant biological response field for a concentration-response experiment is pK
  • Trpsin-EDTA (GibcoBRL #25300-054)
  • FBS Fetal Bovine Serum
  • PBS Phosphate Buffered Saline
  • DMSO Dimethyl Sulfoxide
  • Luclite Luciferase Reporter assay Packard #6016911
  • HOS-CD4.CCR5-LTR-Luciferase Human Osteosarcoma cell line engineered to overexpress human CD4 and human CCR5 (AIDS Repository cat# 3318) stabily transfected with HIV-1 -LTR-Luciferase reporter.
  • the cells were propagated in DMEM containing 2% FBS. Cells were split by standard trypsinization when confluency reached 80% (roughly every 2 to 3 days).
  • HIV-1 virus stocks were titered in the assay system in order to obtain an estimate of the number of infectious particles per unit volume (described as RLU/ml). Virus stocks were diluted into DMEM containing 2% FBS and assayed as described in the "procedure" section below.
  • Black-walled 96-well tissue culture plates were seeded with HOS-CD4.CCR5-LTR- Luciferase @ 0.6 to 1.2 x 10 3 cells per well in 50ul DMEM containing 2% FBS and placed in a humidified incubator @ 37°C, 5% CO 2 overnight. The following day, test compounds were titrated 4-fold at 2X the final concentration in DMEM + 2% FBS + 0.2% DMSO. 50 ⁇ l of titrated compound was transferred to the HOS cells and the plates were placed in a humidified incubator at 37°C, 5% CO 2 for 1hr.
  • RLU Relative Light Units
  • K is IC 5 o
  • Y2 is baseline
  • N Hill Coefficient
  • Each of the compounds of the present invention provides a plC 50 value of at least 5 when tested in each of the above-described assays.
  • Test compounds are employed in free or salt form. All research complied with the principles of laboratory animal care (NIH publication No. 85-23, revised 1985) and standard pharmaceutical industry policy on animal use.

Abstract

The present invention relates to compounds of formula (I), or pharmaceutically acceptable derivatives thereof, 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

PYRROLIDINE AND AZETIDINE COMPOUNDS AS CCR5 ANTAGONISTS
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 AIDS-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.
We have now discovered a series of small molecule nonpeptide 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 HIV, 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 antivirals, 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.
SUMMARY OF THE INVENTION
The present invention includes compounds of formula (I)
Figure imgf000003_0001
and pharmaceutically acceptable derivatives thereof, wherein:
X is a C1-5 alkylene chain, wherein said X is optionally substituted by one or more =O, =S, alkyl, or halogen and wherein said Cι.5 alkylene chain may optionally have 0-3 heteroatoms selected from oxygen, phosphorus, sulfur, or nitrogen;
Ring A is a saturated, partially saturated, or aromatic 3-7 monocyclic or 8-10 membered bicyclic ring having one ring nitrogen and 0-4 additional heteroatoms selected from oxygen, phosphorus, sulfur, or nitrogen;
Ring B is a saturated 4 or 5 membered ring containing the depicted ring nitrogen;
R1 is alkyl optionally substituted by one or more R7, alkenyl optionally substituted by one or more R7, alkynyl optionally substituted by one or more R7, cycloalkyl optionally substituted by one or more R8, heterocyclyl optionally substituted by one or more R8, heteroaryl optionally substituted by one or more R6, or aryl optionally substituted by one or more R6; or R1 and X taken together form a saturated, partially saturated or aromatic 5-6 membered ring having 0-3 heteroatoms selected from oxygen, phosphorus, sulfur, or nitrogen that is fused to Ring A; each R2 is independently selected from -OR0, -C(O)-R°, -S(O)2-R°, -C(O)-N(R°)2, -S(O)2-N(R°)2, -(CH2)a-N(R°)(-Vb-R+), -(CH2)a-(-Vb-R+), halogen, alkyl optionally substituted by one or more R7, alkenyl optionally substituted by one or more R7, alkynyl optionally substituted by one or more R7, aryl optionally substituted by one or more R6, heteroaryl optionally substituted by one or more R6, cycloalkyl optionally substituted by one or more R8, or heterocyclyl optionally substituted by one or more R8; and two adjacent R2s on Ring A are optionally taken together to form a fused, saturated, partially saturated or aromatic 5-6 membered ring having 0-3 heteroatoms selected from oxygen, phosphorus, sulfur, or nitrogen; or two geminal R2s are optionally taken together to form a spiro, saturated, partially saturated or aromatic 5-6 membered ring having 0-3 heteroatoms selected from oxygen, phosphorus, sulfur, or nitrogen, said fused or spiro ring being optionally substituted by one or more R8; each a independently is 0-3; each b independently is 0 or 1 ;
V is -C(O)-, -C(O)O-, -S(O)2-, or -C(O)-N(R°)-;
R+ is alkyl, cycloalkyl, aralkyl, aryl, heteroaryl, heteroaralkyl, or heterocyclyl, wherein said R+ is optionally substituted by one or more R8; m is 0 or 1 ; n is 0-5;
R3 is H, -N(R°)2, -N(R°)C(O)R°, -CN, halogen, CF3, alkyl optionally substituted by one or more groups selected from R7 or -S-aryl optionally substituted by -(CH2)-ι-6- N(R°)SO2(R°), alkenyl optionally substituted by one or more groups selected from R7 or -S-aryl optionally substituted by -(CH2)1-6-N(R°)SO2(R0), alkynyl optionally substituted by one or more groups selected from R7 or -S-aryl optionally substituted by -(CH2)1-6-N(R°)SO2(R°), cycloalkyl or carbocyclyl optionally substituted by one or more R8, aryl optionally substituted by one or more R6, heteroaryl optionally substituted by one or more R6, or heterocyclyl optionally substituted by one or more R8;
Y is alkyl, alkenyl, alkynyl, -(CR4R5)P-, -C(O)-, -C(O)C(O)-, -C(S)-, -O-(CH2)M-C(O)-, -(CH2)o--rC(O)-O-, -N(R°)-C(O)-, -C(O)-N(R0)-, -N(R°)-C(S)-,
-S(O)r, -O-C(=N-CN)-, -O-C(=N-R0)-, -C(=N-CN)-O-, -C(=N-R°)-O-, -C(=N-CN)-S-, -S-C(=N-CN)-, -N(R°)-C(=N-CN)-, -C(=N-CN)-, -N(R°)-C[=N-C(O)-R°], -N(R°)-C[=N-S(O)t-R0], -N(R°)-C(=N-OR0)-, -N(R°)-C(=N-R0)-, or -C(=N-R0)-; each R4 is independently H, alkyl optionally substituted by R7, alkenyl optionally substituted by R7, or alkynyl optionally substituted by R7; each R5 is independently selected from H, -C(O)-OR6, -C(O)-N(R°)2, -S(O)2-N(R°)2, -S(O)2-R°, aryl optionally substituted by R6, or heteroaryl optionally substituted by R6; p is 1-5; t is 1 or 2; each R6 is independently selected from halogen, -CF3, -OCF3, -OR0, -(CH2).-6-OR°, -SR°, -(CH^LB-SR0, -SCF3, -R°, methylenedioxy, ethylenedioxy, -NO2, -CN, -(CH2)1.6-CN, -N(R°)2, -(CH2)1.6-N(R°)2, -NR°C(O)R°, -NR°(CN), -NR°C(O)N(R°)2> -NR°C(S)N(R°)2) -NR°CO2R°, -NR°NR0C(O)R°, -NR0NR°C(O)N(R°)2, -NR°NR°CO2R°, -C(O)C(O)R°, -C(O)CH2C(O)R°, -(CH2)o-6CO2R°, -O-C(O)R°, -C(O)R°, -C(O)N(R0)N(R°)2, -C(O)N(R°)2, -C(O)N(R°)OH, -C(O)N(R°)SO2R0, -OC(O)N(R°)2, -S(O)tR°, -S(O)t-OR°, -S(O)tN(R0)C(O)R°, -S(O)tN(R°)OR°, -NR°SO2N(R°)2, -NR°SO2R°, -C(=S)N(R°)2, -C(=NH)-N(R°)2, -(CH2)1-6-C(O)R°, -C(=N-OR°)-N(R°)2, -O-(CH2)o-6-SO2N(R°)2, -(CH2)1-6NHC(O)R°, or -SO2N(R°)2 wherein the two R°s on the same nitrogen are optionally taken together to form a 5-8 membered saturated, partially saturated, or aromatic ring having additional 0-4 heteroatoms selected from oxygen, phosphorus, nitrogen, or sulfur; each R7 is independently selected from halogen, -CF3, -R°, -OR0, -OCF3,
-(CH2)ι-6-OR°, -SR°, -SCF3, -(CH^-e-SR0, aryl optionally substituted by R6, methylenedioxy, ethylenedioxy, -NO2, -CN, -(CHzKe-CN, -N(R°)2, -(CH2)ι.6-N(R0)2, -NR°C(O)R°, -NR°(CN), -NR0C(O)N(R°)2, -N(R°)C(S)N(R°)2, -NR°CO2R°, -NR°NR°C(O)R°, -NR°NR0C(O)N(R°)2, -NR°NR0CO2R°, -C(O)C(O)R°, -C(O)CH2C(O)R°, -(CH2)o-6-CO2R°, -C(O)R°, -C(O)N(R°)N(R°)2, -C(O)N(R°)2>
-C(O)N(R°)OH, -OC(O)R°, -C(O)N(R°)SO2R°, -OC(O)N(R°)2, -S(O)tR°, -S(O)rOR°, -S(O)tN(R°)C(O)R°, -S(O)tN(R0)OR0, -NR°SO2N(R°)2l -NR°SO2R0, -C(=S)N(R°)2, -C(=NH)-N(R°)2, -(CH2)1-6-C(O)R°, -C(=N-OR°)-N(R°)2, -O-(CH2)0-6-SO2N(R°)2, -(CH2)ι-6-NHC(O)R°, or -SO2N(R°)2 wherein the two R°s on the same nitrogen are optionally taken together to form a 5-8 membered saturated, partially saturated, or aromatic ring having additional 0-4 heteroatoms selected from oxygen, phosphorus, nitrogen, or sulfur; each R8 is independently selected from R7, =O, =S, =N(R°), =N(CN); R9 is H or oxo; each R° is independently selected from hydrogen, alkyl, alkenyl, alkynyl, cycloalkyl, carbocyclylalkyl, aryl, heteroaryl, aralkyl, heteroaralkyl, heterocyclyl, or heterocyclylalkyl, wherein each member of R° except H is optionally substituted by one or more R*, OR*, N(R*)2> =O, =S, halo, CF3, NO2, CN, -C(O)R*, -CO2R*, -C(O)-aryl, -C(O)-heteroaryl, -C(O)-aralkyl, -S(O)raryl, -S(O)t-heteroaryl, -NR*SO2R*, -NR*C(O)R*, -NR*C(O)N(R*)2, -N(R*)C(S)N(R*)2, -NR*CO2R*, -NR*NR*C(O)R*, -NR*NR*C(O)N(R*)2, -NR*NR*CO2R*, -C(O)C(O)R*, -C(O)CH2C(O)R*, -C(O)N(R*)N(R*)2, -C(O)N(R*)2, -C(O)NR*SO2R*, -OC(O)N(R*)2, -S(O)tR*, -NR*SO2N(R*)2) -SO2N(R*)2 wherein the two R*s on the same nitrogen are optionally taken together to form a 5-8 membered saturated, partially saturated or aromatic ring having additional 0-4 heteroatoms selected from oxygen, phosphorus, nitrogen or sulfur; and each R* is independently H, alkyl, alkenyl, alkynyl, cycloalkyl, aryl, or heteroaryl.
In one embodiment of the present invention the B ring is pyrrolidine.
In one embodiment R9 is H, while in another embodiment R9 is oxo.
In one embodiment R1 is optionally substitued aryl. For example, R1 is phenyl mono- or di- substituted with halogen. Suitably R1 may be phenyl di-substituted with Cl.
In one embodiment -(Y)m-R3 suitably is
Figure imgf000006_0001
More suitably -(Y)m-Rό is
Figure imgf000007_0001
Figure imgf000008_0001
Figure imgf000009_0001
CN CN N.C N
A- Λ A^ η 0 T
Figure imgf000010_0001
In one embodiment X is -(CH2)-, -(CH2-CH2)-, or -(CH2-CH2-CH2)-. Suitably X is optionally substituted by one or more halogen or oxo. Further X optionally has 1-3 heteroatoms selected from oxygen, phosphorus, sulfur, or nitrogen.
In one embodiment the A ring is selected from the following, where the asterisk (*) indicates the preferred, but not limiting, point(s) of substitution:
Figure imgf000011_0001
Figure imgf000011_0002
Figure imgf000011_0003
Suitably each R2, with the asterisk (*) indicating a preferred, but not limiting, point of substitution from Ring A, independently is selected from
Figure imgf000011_0004
Figure imgf000012_0001
Figure imgf000013_0001
Figure imgf000014_0001
In one embodiment the ring A, with two geminal R2s, is selected from:
Figure imgf000015_0001
Suitably the A ring is tropane or piperidine, either optionally substituted with one or more R2. Preferrably, A-R2 is comprised of one of the following:
Figure imgf000015_0002
Figure imgf000016_0001
Figure imgf000016_0002
Figure imgf000016_0004
Figure imgf000016_0003
Figure imgf000016_0005
Figure imgf000017_0001
Figure imgf000018_0001
Figure imgf000019_0001
In one embodiment the A ring contains at least one additional nitrogen atom and said A ring optionally is N-substituted. Suitably the A ring is N-substituted with - (CH2)a-(Vb-R+).
Another aspect of the present invention includes a method of treatment
(including prevention) of a viral infection in a mammal comprising administering to said mammal an antiviral effective amount of a compound of the present invention! Preferably the viral infection is an HIV infection.
Another aspect of the present invention includes a method of treatment
(including prevention) of a bacterial infection in a mammal comprising administering to said mammal an effective amount of a compound of the present invention. Preferably the bacterium is Yersinia pestis.
Another aspect of the present invention includes a method of treatment
(including prevention) of 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, infectious disorders including bubonic and pneumonic plague, human papilloma virus infection, prostate cancer, wound healing, amyotrophic lateral sclerosis and immune mediated disorders in a mammal comprising administering to said mammal a pharmceutically effective amount of a compound of the present invention.
Another aspect of the present invention includes a compound of the present invention for use in medical therapy.
Another aspect of the present invention includes the use of a compound of the present invention in the manufacture of a medicament for the treatment (including prophylaxis) of a viral infection. Preferably the viral infection is a HIV infection.
Another aspect of the present invention includes the use of a compound of the present invention in the manufacture of a medicament for the treatment, including prophylaxis, of a bacterial infection. Preferably the bacterium is Yersinia pestis.
Another aspect of the present invention includes the use of a compound of the present invention in the manufacture of a medicament for the treatment, including prophylaxis, of 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, infectious disorders including bubonic and pneumonic plague, human papilloma virus infection, prostate cancer, wound healing, amyotrophic lateral sclerosis and immune mediated disorders.
Another aspect of the present invention includes a pharmaceutical composition comprising a pharmaceutically effective amount of a compound of the present invention together with a pharmaceutically acceptable carrier. Preferably the pharmaceutical composition is in the form of a tablet, capsule, or liquid. Another aspect of the present invention includes a method of treatment, including prevention, of a viral infection in a mammal comprising administering to said mammal a composition comprising a compound of the present invention and another therapeutic agent. Preferably the composition comprises one or more additional therapeutic agent selected from the group consisting of (1 -alpha, 2-beta, 3- alpha)-9-[2,3-bis(hydroxymethyl)cyclobutyl]guanine [(-)BHCG, SQ-34514, lobucavir], 9-[(2R,3R,4S)-3,4-bis(hydroxymethyl)-2-oxetanosyl]adenine (oxetanocin-G), 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), ribonucleotide reductase inhibitors, 2-acetylpyridine 5-[(2-chloroanilino) thiocarbonyl) thiocarbonohydrazone and hydroxyurea, 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), (-)-beta-D-2,6-diaminopurine dioxolane (DAPD), 3'-azido-2',3'-dideoxythymidine-5'-H-phosphophonate (phosphonovir), 2'-deoxy-5-iodo-uridine (idoxuridine), (-)-cis-1-(2-hydroxymethyl)-1 ,3- oxathiolane 5-yl)-cytosine (lamivudine), cis-1 -(2-(hvdroxvmethyl)-1.3-oxathiolan-5- yl)-5-fluorocytosine (FTC), 3'-deoxy-3'-fluorothymidine, 5-chloro-2',3'-dideoxy-3'- fluorouridine, (-)-cis-4-[2-amino-6-(cyclopropylamino)-9jH-purin-9-yl]-2-cyclopentene- 1-methanol (abacavir), 9-[4-hydroxy-2-(hydroxymethyl)but-1-yl]-guanine (H2G), ABT-606 (2HM-H2G) ribavirin, protease inhibitors, indinavir, ritonavir, nelfinavir, amprenavir, saquinavir, fosamprenavir, (R)-N-tert-butyl-3-[(2S,3S)-2-hydroxy-3-N- [(R)-2-N-(isoquinolin-5-yloxyacetyl)amino-3-methylthiopropanoyl]amino-4- phenylbutanoyl]-5,5- dimethyl-1 ,3-thiazolidine-4-carboxamide (KNI-272), 4R-
(4alpha,5alpha,6beta)]-1,3-bis[(3-aminophenyl)methyl] hexahydro-5,6-dihydroxy-4,7- bis(phenylmethyl)-2H-1 ,3-diazepin-2-one dimethanesulfonate (mozenavir), 3-[1-[3-[2- (5-trifluoromethylpyridinyl)-sulfonylamino] phenyl]propyl]-4- hydroxy-6alpha- phenethyl-6beta-propyl-5,6-dihydro-2-pyranone (tipranavir), N'-[2(S)-Hydroxy-3(S)- [N-(methoxycarbonyl)-l-tert-leucylamino]-4- phenylbutyl-N alpha-(methoxycarbonyl)- N'-[4-(2-pyridyl)benzyl]-L- tert-leucylhydrazide (BMS-232632), 3-(2(S)-Hydroxy-3(S)- (3-hydroxy-2-methylbenzamido)-4-phenylbutanoyl)-5,5-dimethyl-N-(2- methylbenzyl)thiazolidine-4(R)-carboxamide (AG-1776), N-(2(R)-hydroxy-1 (S)- indanyl)-2(R)-phenyl-methyl-4(S)-hydroxy-5-(1-(1-(4-benzo[b]furanylmethyl)-2(S)-N'- (tert-butylcarboxamido)piperazinyl)pentanamide (MK-944A), interferons, α-interferon, renal excretion inhibitors, probenecid, nucleoside transport inhibitors, dipyridamole, pentoxifylline, N-acetylcysteine (NAC), Procysteine, α -trichosanthin, phosphonoformic acid, immunomodulators, interleukin II, thymosin, granulocyte macrophage colony stimulating factors, erythropoetin, soluble CD and genetically engineered derivatives thereof, non-nucleoside reverse transcriptase inhibitors (NNRTIs), nevirapine (BI-RG-587), alpha-((2-acetyl-5-methylphenyl)amino)-2,6- dichloro-benzeneacetamide (loviride), 1 -[3-(isopropylamino)-2-pyridyl]-4-[5- (methanesulfonamido)-1H-indol-2-ylcarbonyl]piperazine monomethanesulfonate (delavirdine), (10R, 11S, 12S)-12-hydroxy-6, 6, 10, 11-tetramethyl-4-propyl-11 ,12- dihydro-2H, 6H, 10H-benzo(1 , 2-b:3, 4-b':5, 6-b")tripyran-2-one ((+) calanolide A), (4S)-6-Chloro-4-[1 E)-cyclopropylethenyl)-3,4- dihydro-4-(trifluoromethyl)-2(1 H)- quinazolinone (DPC-083), (S)-6-chloro-4-(cyclopropylethynyl)-1 ,4-dihydro-4- (trifluoromethyl)-2H-3,1-benzoxazin-2-one (efavirenz, DM 266), 1-(ethoxymethyl)-5- (1-methylethyl)-6-(phenylmethyl)-2,4(1H,3H)-pyrimidinedione (MKC-442), and 5- (3,5-dichlorophenyl)thio-4-isopropyl-1 -(4-pyridyl)methyl-1 H-imidazol-2-ylmethyl carbamate (capravirine), glycoprotein 120 antagonists, PRO-2000, PRO-542, 1,4- bis[3-[(2, 4- dichlorophenyl)carbonylamino]-2-oxo-5,8-disodiumsulfanyl]naphthalyl-2, 5-dimethoxyphenyl-1 , 4-dihydrazone (FP-21399), cytokine antagonists, reticulose (Product-R), 1 ,1'-azobis-formamide (ADA), 1,11-(1 ,4-pheny.enebis(methylene))bis- 1 ,4,8, 11 -tetraazacyclotetradecane octahydrochloride (AMD-3100), integrase inhibitors, and fusion inhibitors.
Another aspect of the present invention includes a method of treatment, including prevention, of a viral infection in a mammal comprising administering to said mammal a composition comprising a compound of the present invention and ritonavir.
Particular compounds of the present invention include 22
Figure imgf000023_0001
23
Figure imgf000024_0001
24
Figure imgf000025_0001
Figure imgf000026_0001
W OH
Figure imgf000026_0002
F OH
Figure imgf000026_0003
26
Figure imgf000027_0001
27
Figure imgf000028_0001
28
Figure imgf000029_0001
29
Figure imgf000030_0001
30
Figure imgf000031_0001
Figure imgf000032_0001
Figure imgf000032_0002
Figure imgf000032_0003
Figure imgf000032_0004
32
Figure imgf000033_0001
33
Figure imgf000034_0001
Figure imgf000035_0001
Figure imgf000035_0002
Figure imgf000035_0003
Figure imgf000035_0004
H OH
Figure imgf000036_0001
Figure imgf000036_0002
Figure imgf000036_0003
Figure imgf000036_0004
Figure imgf000037_0001
Figure imgf000038_0001
More particularly, the compounds of the present invention include: tert-butyl 3-(3,4-dichlorophenyl)-3-[3-(4-oxo-1-phenyl-1 ,3,8-triazaspiro[4.5]dec-8- yI)propyl]pyrrolidine-1-carboxylate;
8-{3-[3-(3,4-dichlorophenyl)-1-(2-furoyl)pyrrolidin-3-yl]propyl}-1-phenyl-1 ,3,8- triazaspiro[4.5]decan-4-one; 8-{3-[3-(3,4-dichlorophenyl)-1-(isoxazol-5-ylcarbonyl)pyrrolidin-3-yl]propyl}-1-phenyl-
1,3,8-triazaspiro[4.5]decan-4-one;
8-{3-[3-(3,4-dichlorophenyl)-1-(1H-pyrrol-2-ylcarbonyl)pyrrolidin-3-yl]propyl}-1-phenyl-
1,3,8-triazaspiro[4.5]decan-4-one; 8-{3-[3-(3,4-dichlorophenyl)-1-pentanoylpyrrolidin-3-yl]propyl}-1-[3-
(trifluoromethyl)phenyl]-1 ,3,8-triazaspiro[4.5]decan-4-one;
8-{3-[3-(3,4-dichlorophenyl)-1-(2-furoyl)pyrrolidin-3-yl]propyl}-1-[3-
(trifluoromethyl)phenyl]-1,3,8-triazaspiro[4.5]decan-4-one;
8-{3-[1-(cyclobutylcarbonyl)-3-(3,4-dichlorophenyl)pyrrolidin-3-yl]propyl}-1-[3- (trifluoromethyl)phenyl]-1 ,3,8-triazaspiro[4.5]decan-4-one;
8-{3-[3-(3,4-dichlorophenyl)-1 -pentanoylpyrrolidin-3-yl]propyl}-1 -phenyl-1 ,3,8- triazaspiro[4.5]decan-4-one;
8-{3-[1-(cyclopentylcarbonyl)-3-(3,4-dichlorophenyl)pyrrolidin-3-yl]propyl}-1-phenyl-
1,3,8-triazaspiro[4.5]decan-4-one; 8-{3-[1 -(cyclobutylcarbonyl)-3-(3,4-dichlorophenyl)pyrrolidin-3-yl]propyl}-1 -phenyl-
1 ,3,8-triazaspiro[4.5]decan-4-one;
8-{3-[1-(cyclobutylcarbonyl)-3-(3,4-dichlorophenyl)pyrrolidin-3-yl]propyl}-1-(3- methylphenyl)-1 ,3,8-triazaspiro[4.5]decan-4-one;
3-acetyl-8-{3-[1-acetyl-3-(3,4-dichlorophenyl)pyrrolidin-3-yl]propyl}-1-(3- methylphenyl)-1 ,3,8-triazaspiro[4.5]decan-4-one;
8-{3-[1-(1,3-benzoxazol-2-yl)-3-(3,4-dichlorophenyl)pyrrolidin-3-yl]propyl}-1-phenyl-
1 ,3,8-triazaspiro[4.5]decan-4-one;
8-(2-{[3-(3,4-dichlorophenyl)-1-(2-furoyl)pyrrolidin-3-yl]oxy}ethyl)-1 -phenyl-1 ,3,8- triazaspiro[4.5]decan-4-one; 8-(2-{[1 -(cyclopentylcarbonyl)-3-(3,4-dichlorophenyl)pyrrolidin-3-yl]oxy}ethyl)-1 - phenyl-1 ,3,8-triazaspiro[4.5]decan-4-one;
8-(2-{[1-acetyl-3-(3,4-dichlorophenyl)pyrrolidin-3-yl]oxy}ethyl)-1 -phenyl-1 ,3,8- triazaspiro[4.5]decan-4-one;
8-(2-{[3-(3,4-dichlorophenyl)-1-(phenylsulfonyl)pyrrolidin-3-yl]oxy}ethyl)-1-phenyl- 1 ,3,8-triazaspiro[4.5]decan-4-one;
8-(2-{[3-(3,4-dichlorophenyl)-1-(2-furoyl)pyrrolidin-3-yl]oxy}ethyl)-1-(3- methoxyphenyl)-1,3,8-triazaspiro[4.5]decan-4-one;
8-(2-{[1-(cyclopentylcarbonyl)-3-(3,4-dichlorophenyl)pyrrolidin-3-yl]oxy}ethyl)-1-(3- methoxyphenyl)-1,3,8-triazaspiro[4.5]decan-4-one; 8-(2-{[1-acetyl-3-(3,4-dichlorophenyl)pyrrolidin-3-yl]oxy}ethyl)-1-(3-methoxyphenyl)- 1 ,3,8-triazaspiro[4.5]decan-4-one; and
8-(2-{[3-(3,4-dichlorophenyl)-1-(phenylsulfonyl)pyrrolidin-3-yl]oxy}ethyl)-1-(3- methoxyphenyl)-1 ,3,8-triazaspiro[4.5]decan-4-one.
DETAILED DESCRIPTION OF THE INVENTION The phrase "optionally substituted" is used interchangeably with the phrase "substituted or unsubstituted" or with the term "(un)substituted." Unless otherwise indicated, an optionally substituted group may have a substituent at each substitutable position of the group, and each substitution is independent of the other.
The term "alkyl", alone or in combination with any other term, refers to a straight-chain or branched-chain saturated aliphatic hydrocarbon radical containing the specified number of carbon atoms. Examples of alkyl radicals include, but are not limited to, methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, sec-butyl, tert-butyl, pentyl, isoamyl, n-hexyl and the like.
The term "cycloalkyl", "carbocylyl", "carbocyclic", or "carbocycle", or "carbocyclo", alone or in combination with any other term, refers to a monocyclic or polycyclic non-aromatic hydrocarbon ring radical having three to twenty carbon atoms, preferably from three to twelve carbon atoms, and more preferably from three to ten carbon atoms. If polycyclic, each ring in a carbocylyl radical is non-aromatic unless otherwise indicated. A carbocylyl radical is either completely saturated or contains one or more units of unsaturation but is not aromatic. The unsaturation, if present, may occur in any point in the ring that may result in any chemically stable configuration. The term "cycloalkyl", "carbocylyl", "carbocyclic", or "carbocycle", or "carbocyclo" also includes hydrocarbon rings that are fused to one or more aromatic rings, such as in tetrahydronaphthyl, where the radical or point of attachment is on the non-aromatic ring.
Unless otherwise indicated, the term "cycloalkyl", "carbocylyl", "carbocyclic", or "carbocycle", or "carbocyclo" also includes each possible positional isomer of a non-aromatic hydrocarbon radical, such as in 1-decahydronaphthyl, 2- decahydronaphthyl, 1 -tetrahydronaphthyl and 2-tetrahydronaphthyl. Examples of suitable cycloalkyl groups include, but are not limited to, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cyclohexenyl, decahydronaphthyl, tetrahydronaphthyl and the like.
The term "alkenyl," alone or in combination with any other term, refers to a straight-chain or branched-chain alkyl group with at least one carbon-carbon double bond. Examples of alkenyl radicals include, but are not limited to, ethenyl, propenyl, isopropenyl, butenyl, isobutenyl, pentenyl, hexenyl, hexadienyl and the like.
The term "alkynyl" refers to hydrocarbon groups of either a straight or branched configuration with one or more carbon-carbon triple bonds which may occur in any stable point along the chain, such as ethynyl, propynyl, butynyl, pentynyl, and the like.
The term "alkoxy" refers to an alkyl ether radical, wherein the term "alkyl" is defined above. Examples of suitable alkyl ether radicals include, but are not limited to, methoxy, ethoxy, n-propoxy, isopropoxy, n-butoxy, isobutoxy, sec-butoxy, tert- butoxy and the like.
The term "aryl", alone or in combination with any other term, refers to an aromatic monocyclic or polycyclic hydrocarbon ring radical containing five to twenty carbon atoms, preferably from six to fourteen carbon atoms, and more preferably from six to ten carbon atoms. Also included within the scope of the term "aryl", as it is used herein, is a group in which an aromatic hydrocarbon ring is fused to one or more non-aromatic carbocyclic or heteroatom-containing rings, such as in an indanyl, phenanthridinyl or tetrahydronaphthyl, where the radical or point of attachment is on the aromatic hydrocarbon ring.
Unless otherwise indicated, the term "aryl" also includes each possible positional isomer of an aromatic hydrocarbon radical, such as in 1-naphthyl, 2- naphthyl, 5-tetrahydronaphthyl, 6-tetrahydronaphthyl, 1 -phenanthridinyl, 2- phenanthridinyl, 3-phenanthridinyl, 4-phenanthridinyl, 7-phenanthridinyl, 8- phenanthridinyl, 9-phenanthridinyl and 10-phenanthridinyl. Examples of aryl radicals include, but are not limited to, phenyl, naphthyl, indenyl, azulenyl, fluorenyl, anthracenyl, phenanthrenyl, tetrahydronaphthyl, indanyl, phenanthridinyl and the like. The term "aralkyl" further refers to groups of -RaRb, where Ra is an alkylene as defined herein and Rb is an aryl as defined herein.
The term "heterocycle", "heterocyclic", and "heterocyclyl", alone or in combination with any other term, refers to a non-aromatic monocyclic or polycyclic ring radical containing three to twenty carbon atoms, preferably three to seven carbon atoms if monocyclic and eight to eleven carbon atoms if bicyclic, and in which one or more ring carbons, preferably one to four, are each replaced by a heteroatom such as N, O, and S. If polycyclic, each ring in a heterocyclyl radical is non-aromatic unless otherwise indicated. A heterocyclic ring may be fully saturated or may contain one or more units of unsaturation but is not aromatic. The unsaturation, if present, may occur in any point in the ring that may result in any chemically stable configuration. The heterocyclic ring may be attached at a carbon or heteroatom that results in the creation of a stable structure. Preferred heterocycles include 5-7 membered monocyclic heterocycles and 8-10 membered bicyclic heterocycles.
Also included within the scope of the term "heterocycle", "heterocyclic", or "heterocyclyl" is a group in which a non-aromatic heteroatom-containing ring is fused to one or more aromatic rings, such as in an indolinyl, chromanyl, phenanthridinyl or tetrahydroquinolinyl, where the radical or point of attachment is on the non-aromatic heteroatom-containing ring. Unless otherwise indicated, the term "heterocycle", "heterocyclic", or "heterocyclyl" also includes each possible positional isomer of a heterocyclic radical, such as in 1-decahydroquinoline, 2-decahydroquinoline, 3- decahydroquinoline, 4-decahydroquinoline, 5-decahydroquinoline, 6- decahydroquinoline, 7-decahydroquinoline, 7-decahydroquinoline, 8- decahydroquinoline, 4a-decahydroquinoline, 8a-decahydroquinoline, 1 -indolinyl, 2- indolinyl, 3-indolinyl, 1-tetrahydroquinoline, 2-tetrahydroquinoline, 3- tetrahydroquinoline and 4-tetrahydroquinoline. The term "heterocyclylalkyl" refers to an alkyl group substituted by a heterocyclyl.
Examples of heterocyclic groups include, but are not limited to, imidazolinyl, 2,3-dihydro-1 H-imidazolyl, imidazolidinyl, indazolinolyl, perhydropyridazyl, pyrrolinyl, pyrrolidinyl, 4H-pyrazolyl, piperidinyl, pyranyl, pyrazolinyl, piperazinyl, morpholinyl, thiamorpholinyl, thiazolidinyl, thiamorpholinyl, oxopiperidinyl, oxopyrrolidinyl, azepinyl, tetrahydrofuranyl, oxoazepinyl, tetrahydropyranyl, thiazolyl, dioxolyl, dioxinyl, oxathiolyl, benzodioxolyl, dithiolyl, dithiolanyl, tetrahydrothiophenyl, sulfolanyl, dioxanyl, dioxolanyl, tetahydrofurodihydrofuranyl, dihydropyranyl, tetrahydropyranodihydrofuranyl, tetradyrofurofuranyl, tetrahydropyranofuranyl, diazolonyl, phthalimidinyl, benzoxanyl, benzopyrrolidinyl, benzopiperidinyl, benzoxolanyl, benzothiolanyl and benzothianyl.
The term "heteroaryl", alone or in combination with any other term, refers to an aromatic monocyclic or polycyclic ring radical containing five to twenty carbon atoms, preferably five to ten carbon atoms, in which one or more ring carbons, preferably one to four, are each replaced by a heteroatom such as N, O, and S.
Preferred heteroaryl groups include 5-6 membered monocyclic heteroaryls and 8-10 membered bicyclic heteroaryls.
Also included within the scope of the term "heteroaryl" is a group in which a heteroaromatic ring is fused to one or more aromatic or non-aromatic rings where the radical or point of attachment is on the heteroaromatic ring. Examples include, but are not limited to, pyrido[3,4-d]pyrimidinyl, 7,8-dihydro-pyrido[3,4-d]pyrimidine and 5,6,7,8-tetrahydro-pyrido[3,4-d]pyrimidine. Unless otherwise indicated, the term "heteroaryl" also includes each possible positional isomer of a heteroaryl radical, such as in 2-pyrido[3,4-d]pyrimidinyl and 4-pyrido[3,4-d]pyrimidinyl.
Examples of heteroaryl groups include, but are not limited to, imidazolyl, quinolyl, isoquinolyl, indolyl, indazolyl, pyridazyl, pyridyl, pyrrolyl, pyrazolyl, pyrazinyl, quinoxalyl, pyrimidinyl, pyridazinyl, furyl, thienyl, triazolyl, thiazolyl, carbazolyl, carbolinyl, tetrazolyl, benzofuranyl, oxazolyl, benzoxazolyl, isoxozolyl, isothiazolyl, thiadiazolyl, furazanyl, oxadiazolyl, benzimidazolyl, benzothienyl, quinolinyl, benzotriazolyl, benzothiazolyl, isoquinolinyl, isoindolyl, acridinyl and benzoisoxazolyl.
The term "heteroaralkyl" further refers to groups of -RaRb, where Ra is an alkylene as defined herein and R is a heteroaryl as defined herein.
The term "heteroatom" means nitrogen, oxygen, phosphorus, or sulfur and includes any oxidized forms thereof, including as non-limiting examples oxidized forms of nitrogen such as N(O) {N+-O"}, oxidized forms of sulfur such as S(O) and S(O)2, and the quaternized form of any basic nitrogen. The term "halogen" refers to fluorine, chlorine, bromine or iodine.
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.
The term "pharmaceutically acceptable derivative" means any pharmaceutically acceptable salt, ester, salt of an ester, or other derivative of a compound of this invention which, upon administration to a recipient, is capable of providing (directly or indirectly) a compound of this invention or an inhibitorily active metabolite or residue thereof. Particularly favored derivatives and prodrugs are those that increase the bioavailability of the compounds of this invention when such compounds are administered to a mammal (e.g., by allowing an orally administered compound to be more readily absorbed into the blood) or which enhance delivery of the parent compound to a biological compartment (e.g., the brain or lymphatic system) relative to the parent species.
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, maleic, phosphoric, glycollic, lactic, salicyclic, succinic, toluene-p-sulfonic, tartaric, acetic, citric, 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 C^ 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 NW + (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.
Esters of the compounds of the present invention are independently selected from the following groups: (1 ) carboxylic acid esters obtained by esterification of the hydroxy groups, in which the non-carbonyl moiety of the carboxylic acid portion of the ester grouping is selected from straight or branched chain alkyl (for example, acetyl, n-propyl, t-butyl, or n-butyl), alkoxyalkyl (for example, methoxymethyl), aralkyl (for example, benzyl), aryloxyalkyl (for example, phenoxymethyl), aryl (for example, phenyl optionally substituted by, for example, halogen,
Figure imgf000045_0001
or amino); (2) sulfonate esters, such as alkyl- or aralkylsulfonyl (for example, methanesulfonyl); (3) amino acid esters (for example, L-valyl or L-isoleucyl); (4) phosphonate esters and (5) mono-, di- or triphosphate esters. The phosphate esters may be further esterified by, for example, a Cι-20 alcohol or reactive derivative thereof, or by a 2,3-di (C6-24)acyl glycerol.
In such esters, unless otherwise specified, any alkyl moiety present advantageously contains from 1 to 18 carbon atoms, particularly from 1 to 6 carbon atoms, more particularly from 1 to 4 carbon atoms, Any cycloalkyl moiety present in such esters advantageously contains from 3 to 6 carbon atoms. Any aryl moiety present in such esters advantageously comprises a phenyl group.
The compounds according to the invention contain one or more asymmetric carbon atoms and thus occur as racemates and racemic mixtures, single enantiomers, diastereomeric mixtures and individual diastereoisomers. All such isomeric forms of these compounds are expressly included in the present invention. Each stereogenic carbon may be of the R or S configuration. Although the specific compounds exemplified in this application may be depicted in a particular stereochemical configuration, compounds having either the opposite stereochemistry at any given chiral center or mixtures thereof are also envisioned.
Unless otherwise stated, structures depicted herein are also meant to include compounds which differ only in the presence of one or more isotopically enriched atoms. For example, compounds having the present structures except for the replacement of a hydrogen by a deuterium or tritium, or the replacement of a carbon by a 13C- or 1 C-enriched carbon are also within the scope of this invention.
Certain compounds of this invention may exist in alternative tautomeric forms. All such tautomeric forms of the present compounds are within the scope of the invention. Unless otherwise indicated, the representation of either tautomer is meant to include the other.
The present invention features compounds according to the invention for use in medical therapy, for example for the treatment including 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 including 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 including 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 including 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 treatment including prevention of other CCR5-related diseases and conditions, including 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, infectious disorders including bubonic and pneumonic plague, 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 including 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 including 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 (1- alpha, 2-beta, 3-alpha)-9-[2,3-bis(hydroxymethyl)cyclobutyl]guanine [(-)BHCG, SQ- 34514, lobucavir], 9-[(2R,3R,4S)-3,4-bis(hydroxymethyl)-2-oxetanosyl]adenine
(oxetanocin-G), 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-dimethylpropanoic acid (bis- POM PMEA, adefovir dipivoxil), [[(1 R)-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), ribonucleotide reductase inhibitors, for example 2-acetylpyridine 5- [(2-chloroanilino)thiocarbonyl) thiocarbonohydrazone and hydroxyurea, 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), (-)-beta- D-2,6-diaminopurine dioxolane (DAPD), 3'-azido-2',3'-dideoxythymidine-5'-H- phosphophonate (phosphonovir), 2'-deoxy-5-iodo-uridine (idoxuridine), (-)-cjs-1-(2- hydroxymethyl)-1 ,3-oxathiolane 5-yl)-cytosine (lamivudine), cjs-1-(2-
(hydroxymethyl)-l ,3-oxathiolan-5-yl)-5-fluorocytosine (FTC), 3'-deoxy-3'- , fluorothymidine, 5-chloro-2',3'-dideoxy-3'-fluorouridine, (-)-cis-4-[2-amino-6- (cyclopropylamino)-9H-purin-9-yl]-2-cyclopentene-1-methanol (abacavir), 9-[4- hydroxy-2-(hydroxymethyl)but-1-yl]-guanine (H2G), ABT-606 (2HM-H2G) and ribavirin, protease inhibitors, for example indinavir, ritonavir, nelfinavir, amprenavir, saquinavir, fosamprenavir, (R)-N-tert-butyl-3-[(2S,3S)-2-hydroxy-3-N-[(R)-2-N- (isoquinolin-5-yloxyacetyl)amino-3-methylthiopropanoyl]amino-4-phenylbutanoyl]-5,5- dimethyl-1 ,3-thiazolidine-4-carboxamide (KNI-272), 4R-(4alpha,5alpha,6beta)]-1 ,3- bis[(3-aminophenyl)methyl]hexahydro-5,6-dihydroxy-4,7-bis(phenylmethyl)-2H-1 ,3- diazepin-2-one dimethanesulfonate (mozenavir), 3-[1-[3-[2-(5- trifluoromethylpyridinyl)-sulfonylamino]phenyl]propyl]-4- hydroxy-6alpha-phenethyl- 6beta-propyl-5,6-dihydro-2-pyranone (tipranavir), N'-[2(S)-Hydroxy-3(S)-[N- (methoxycarbonyl)-l-tert-leucylamino]-4- phenylbutyl-N alpha-(methoxycarbonyl)-N'-[4- (2-pyridyl)benzyl]-L- tert-leucylhydrazide (BMS-232632), 3-(2(S)-Hydroxy-3(S)-(3- hydroxy-2-methylbenzamido)-4-phenylbutanoyl)-5,5-dimethyl-N-(2- methylbenzyl)thiazolidine-4(R)-carboxamide (AG-1776), N-(2(R)-hydroxy-1 (S)- indanyl)-2(R)-phenyl-methyl-4(S)-hydroxy-5-(1-(1-(4-benzo[b]furanylmethyl)-2(S)-N'- (tert-butylcarboxamido)piperazinyl)pentanamide (MK-944A), interferons such as α- interferon, renal excretion inhibitors such as probenecid, nucleoside transport inhibitors such as dipyridamole; pentoxifylline, N-acetylcysteine (NAC), Procysteine, α -trichosanthin, phosphonoformic acid, as well as immunomodulators such as interleukin II or thymosin, granulocyte macrophage colony stimulating factors, erythropoetin, soluble CD 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- ylcarbonyljpiperazine monomethanesulfonate (delavirdine), (10R, 11S, 12S)-12- Hydroxy-6, 6, 10, 11-tetramethyl-4-propyl-11,12-dihydro-2H, 6H, 10H-benzo(1, 2-b:3, 4-b':5, 6-b")tripyran-2-one ((+) calanolide A), (4S)-6-Chloro-4-[1 E)- cyclopropylethenyl)-3,4- dihydro-4-(trifluoromethyl)-2(1 H)-quinazolinone (DPC-083), (S)-6-chloro-4-(cyclopropylethynyl)-1 ,4-dihydro-4-(trif luoromethyl)-2H-3, 1 - benzoxazin-2-one (efavirenz, DMP 266), 1-(ethoxymethyl)-5-(1-methylethyl)-6- (phenylmethyl)-2,4(1 H,3H)-pyrimidinedione (MKC-442), and 5-(3,5- dichlorophenyl)thio-4-isopropyl-1 -(4-pyridyl)methyl-1 H-imidazol-2-ylmethyl carbamate (capravirine), glycoprotein 120 antagonists, for example PRO-2000, PRO-542 and 1 ,4-bis[3-[(2, 4- dichlorophenyl)carbonylamino]-2-oxo-5,8- disodiumsulfanyl]naphthalyl-2, 5-dimethoxyphenyl-1 , 4-dihydrazone (FP-21399), cytokine antagonists, for example reticulose (Product-R), 1 ,1'-azobis-formamide (ADA), 1 ,11-(1 ,4-phenylenebis(methylene))bis-1 ,4,8,11 -tetraazacyclotetradecane octahydrochloride (AMD-3100), 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, suitably in the range of 0.1 to 100 mg per kilogram body weight per day and most suitably 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, suitably 20 to 500 mg, and most suitably 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%, suitably 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 anti- oxidants, 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 microparticulate 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.
EXAMPLES
The following examples are intended for illustration only and are not intended to limit the scope of the invention in any way. Low resolution, open-access LC-MS data were acquired in either ESI pos/neg or APCI pos/neg mode with scanning from 100-1100 amu @ 0.5 sec/scan. LC conditions: flowrate 0.8 mL/min. 85% H2O (0.1% formic acid) to 100% MeOH (0.075% formic acid) in 6 minutes. Phenomenex Max-RP column, 2.0x50 mm.
High Resolution Mass Spectra were acquired using Micromass LCT mass spectrometer (time-of-flight) with flow injection (FIA-MS) at 0.3 mL/min with 100% MeOH (0.1% formic acid), run time of 2 minutes, in ESI+ mode, scanning from 100- 1100 amu @ 0.5 sec/scan. Reserpine was used as the lock mass (m/z 609.2812) and to adjust mass scale.
As will be appreciated by those skilled in the art, the following schemes may be followed in preparing the compounds of the present invention. Any variability depicted within the scheme(s) illustrated herein should be limited to the particular scheme and not necessarily extended throughout the rest of the present specification.
Abbreviations used: LHMDS = lithium hexamethyl disilazide THP = tetrahydropyran
NMO = N-methylmorpholine N-oxide
I. B-rinq is pyrrolidine
C3-linker pyrrolidine
Figure imgf000055_0001
1.1. Intermediate Compound 1 (3,4-dichlorophenyl)acetonitrile (50.85 g, 273.38 mmol) was dissolved in THF (200 mL). The solution was cooled down to -78 °C, LHMDS (1 M in THF, 287 mL) was added via cannula. The content was stirred at - 78 °C for 30 min, then it was transferred to a solution of methyl bromoacetate (40.77 mL, 430.58 mmol) at r.t. The reaction was completed in less than four hours. The solvent was then removed under reduced pressure. The residue was dissolved in ethyl acetate. The organic layer was washed with 1 :1 brine:H20, dried, filtrated and concentrated. FCC with hexane:ethyl acetate (5:1) afford 20 g desired intermediate product 2.
I.2. Intermediate Compound 2 (8.2 g, 31.9 mmol) was dissolved in THF (200 mL) and cooled to -78°C. LHMDS (1 M in THF, 33.5 mmol) was added and the content stirred at -78°C for 10 min before being transferred to a solution of 2-(3- bromopropoxy)-tetrahydro-2H-pyran (6.48 mL, 38.29 mmol) at r.t via cannula. The reaction was completed in 4 hrs and solvents were removed under reduced pressure, residue redissolved in ethyl acetate and washed with 1 :1 brine:H20, dried, filtrated and concentrated. FCC with hexane:EtOAc (5:1 to 4:1) afford 7.4 g of the intermediate product 3 as light yellow oil.
1.3. Intermediate Compound 3 was dissolved in MeOH (60 mL), followed by concentrated ammonium hydroxide (4 mL) and Raney-Nickel. The contents were hydrogenated (55 Psi) in the Parr apparatus overnight. The desired intermediate product 4 was secured after filtration and solvent removal.
1.4. Intermediate Compound 4 (3.35 g, 13.48 mmol) was dissolved in THF (50 mL), followed by LAH (13.5 mL, 1 M in ether). The mixture was refluxed under nitrogen for
1 h, and then cooled to 0 °C with ice bath followed by a carefully quench with water. The suspension was filtered through a plug of celite and concentrated to give colorless oil intermediate product 5.
1.5. Intermediate Crude product 5 was dissolved in methanol (15 mL), followed by HCI in dioxane (30 mL, 4M), and the mixture stirred at rt for 1 hr, during which it turned pink. The mizture was then concentrated under reduced pressure, redissolved in THF (40 mL), and added triethylamine (10 mL) and (Boc)2O in THF (14 mL, 1 M). After stirring at rt for 1.5hrs, saturated sodium bicarbonate solution was added and the product extracted with ethyl acetate. The organic layer was then dried over sodium sulfate, solvents removed and the product purified on flash column chromatography with hexane:ethyl acetate (1 :1), affording 2.64 g of oily intermediate product 6.
I.6. Synthesis of intermediate compound 7: Dissolve Dess-Martin periodinane (4.77 g, 11.25 mmo) in dichloromethane (60 mL), followed by t-BuOH (1.07 mL), stir at r.t for 5 min and dropwise add product compound 6 (2.63 g, 7.03 mmol) in dichloromethane (10 mL). After stirring at rt for 30 min, the mixture was diluted with ethyl ether and washed with 1.3 N NaOH. The organic layer was then dried over sodium sulfate, the solvent was removed after filtration and crude product purified on flash column chromatography with hexane: ethyl acetate (3:1) affording 1.88 g product 7 as colorless oil.
The following are representative analytical data for compounds of the present invention: I. Intermediate Compound 2 methyl 3-cyano-3-(3,4-dichlorophenyl)propanoate
1HNMR (400 MHz, CDCI3) δ[ppm]: 7.22 (1H, d, J = 8.2 Hz), 4.25 (1 H, t, J = 7.3 Hz), 3.71 (3H, s), 3.01 (1 H, dd, J = 16.8 Hz, 7.5 Hz), 2.82 (1 H, dd, J = 16.8 Hz, 7.1 Hz). 13CNMR (400 MHz, CDCI3) δ[ppm]: 169.35, 134.61 , 133.39, 131.47, 129.71, 126.97, 119.16, 52.75, 39.60, 32.49. Elemental Analysis calculated for
Figure imgf000057_0001
C 51.19%, H 3.51%, N 5.43% found C 51.02%, H 3.93%, N 4.25%.
I. Intermediate Compound 3 methyl 3-cyano-3-(3,4-dichlorophenyl)-6-(tetrahydro-2H-pyran-2-yloxy)hexanoate 1HNMR (400 MHz, CDCI3) δ[ppm]: 7.47 (1H, d, J = 2.3 Hz), 7.38 (1 H, d, J = 8.4 Hz), 7.24 (1H, dd, J = 8.6 Hz, 2.4 Hz), 4.40 (1 H, dt, J = 17.6 Hz, 3.7 Hz), 3.75-3.58 (2H, m), 3.52 (3H, s), 3.41-3.34 (1H, m), 3.30-3.24 (1H, m), 2.98 (1H, d, J = 16.5 Hz), 2.88 (1H, d, J = 16.4 Hz), 2.15-2.04 (1 H, m), 2.00-1.90 (1H, m), 1.73-1.54 (3H, m), 1.47- 1.28 (5H, m). 13CNMR (400 MHz, CDCI3) δ[ppm]: 168.67, 138.08, 133.43, 132.65, 131.06, 128.30, 125.74, 120.70, 99.18, 99.09, 66.44, 66.32, 62.74, 62.64, 52.26, 44.35, 44.29, 37.52, 30.86, 25.68, 25.60, 19.90, 19.85. HRMS calculated for C19H23CI2NO4 Na (M+Na)+ 422.0922 found 422.0890.
I. Intermediate Compound 7
1 , 1 -dimethylethyl 3-(3,4-dichlorophenyl)-3-(3-oxopropyl)-1 -pyrrolidinecarboxylate 1HNMR (400 MHz, CDCI3) δ[ppm]: 9.49 (1 H, s), 7.29 (1 H, d, J = 8.3 Hz), 7.17 (1 H, s), 6.94 (1 H, d, J = 8.4 Hz), 3.53-3.42 (2H, m), 3.36-3.18 (2H, m), 2.12-1.92 (5H, m), 1.88-1.78 (1H, m). 13CNMR (400 MHz, CDCI3) δ[ppm]: 200.84, 154.70, 154.56,
144.49, 132.91 , 131.04, 130.77, 128.79, 126.20, 79.88, 79.73, 55.71 , 54.86, 48.89, 48.05, 44.47, 44.17, 39.97, 36.61 , 35.23, 31.36, 28.67. II. C2-linker pyrrolidine:
Figure imgf000058_0001
The synthesis of the C2-pyrrolidine scaffold was carried out by two procedures. Alkylation of 1 with methyl bromoacetate resulted in a separable mixture of monoalkylated 2 and bis-alkylated 8, each of which was subjected to the desired 10 by a separate route.
Compound 2 was converted to 10 in five steps and in 28% overall yield, while the conversion of 8 to 10 could be accomplished in 4 steps and in a higher overall yield of 38%.
The following are representative analytical data for compounds of the present invention:
II. Intermediate Compound 8 dimethyl 3-cyano-3-(3,4-dichlorophenyl)pentanedioate 1HNMR (400 MHz, CDCI3) δ[ppm]: 7.54 (1H, d, J = 2.4 Hz), 7.43 (1H, d, J = 8.6 Hz), 7.31 (1H, dd, J = 8.4 Hz, 2.4 Hz), 3.60 (6H, s), 3.22 (2H, d, J = 16.6 Hz), 3.04 (2H, d, J = 16.7 Hz). 13CNMR(400MHz, CDCI3) δ[ppm]: 168.56, 137.38, 133.24, 131.12, 128.15, 125.48, 120.08. HRMS calculated for C14H13CI2NO4Na (M+Na)+ 352.019 found 352.0123.
II. Intermediate Compound 10
1 ,1-dimethylethyl 3-(3,4-dichlorophenyl)-3-(3-oxopropyl)-1-pyrrolidinecarboxylate 1HNMR (400 MHz, CDCI3) δ[ppm]: 9.39 (1H, s), 7.30-7.25 (2H, m), 7.02 (1H, d, J = 8.5 Hz), 3.62-3.51 (2H, m), 3.38-3.23 (2H, m), 2.82-2.62 (2H, m), 2.20-2.01 (2H, m), 1.36 (9H, d, J = 9.2 Hz). 13CNMR(400MHz, CDCI3) δ[ppm]: 199.95, 154.56, 143.96, 132.95, 131.28, 130.80, 128.72, 126.14, 80.06, 79.91, 55.77, 55.66, 52.07, 47.16, 46.38, 44.22, 43.87, 36.35, 35.98, 28.67.
The following representative examples, Examples 1-13 of the invention were synthesized according to the following representative synthetic scheme III:
Figure imgf000059_0001
ples
Figure imgf000059_0002
Example 1 tert-butyl 3-(3,4-dichlorophenyl)-3-[3-(4-oxo-1 -phenyl-1 ,3,8-triazaspiro[4.5]dec-8- yl)propyl]pyrrolidine-1-carboxylate
Figure imgf000060_0001
1H NMR (300MHz, CDCI3) δ 7.42-7.40(m,1 H), 7.31 (m,2H), 7.24-7.07(m,2H), 6.94- 6.87(m,2H), 6.30(m,1 H), 3.57-3.32(m,3H), 2.71-2.57(m,2H), 2.32(m,1 H), 2.13- 2.07(m,2H), 1.83-1.70(m,4H), 1.52-1.49(m,7H), 1.29(s,9H), 0.91-0.86(m,4H). mass spectrometry ES+ =587/589 (M+1 , 2 Cl isotope pattern)
Example 2
8-{3-[3-(3,4-dichlorophenyl)-1-(2-furoyl)pyrrolidin-3-yl]propyl}-1 -phenyl-1 ,3,8- triazaspiro[4.5]decan-4-one
Figure imgf000060_0002
1H NMR (500MHz, DMSO-D6) δ 8.99(s,1 H), 7.89(m,1H), 7.63(m,2H), 7.45- 7.01 (m,4H), 6.89(m,2H), 6.80(m,1H), 6.70-6.60(m1 H), 4.60(s,2H), 4.09(m,1 H), 3.97- 3.68(m,4H), 3.65-3.44(m,4H), 3.02(m,2H), 2.70(m,2H), 2.36-2.00(m,2H), 1.90- 1.62(m,3H), 1.44-1.29(m,2H). mass spectrometry ES+ =581/583 (M+1 , 2 Cl isotope pattern)
Example 3 8-{3-[3-(3,4-dichlorophenyl)-1 -(isoxazol-5-ylcarbonyl)pyrrolidin-3-yl]propyl}-1 -phenyl- 1 ,3,8-triazaspiro[4.5]decan-4-one
Figure imgf000061_0001
1H NMR (500MHz, DMSO-D6) δ 8.97(s,1 H), 8.75(m,1 H), 7.63(m,2H), 7.35-
7.20(m,2H), 7.12-7.06(m,2H), 6.89(m,2H), 6.80(m,1 H), 4.59(s,2H), 4.05(m,1 H), 3.97-
3.45(m,8H), 3.01 (m,2H), 2.67(m,2H), 2.36-2.18(m,2H), 1.86-1.68(m,3H), 1.42-
1.24(m,2H). mass spectrometry ES+ =582/584 (M+1 , 2 Cl isotope pattern)
Example 4
8-{3-[3-(3,4-dichlorophenyl)-1 -(1 H-pyrrol-2-ylcarbonyl)pyrrolidin-3-yl]propyl}-1 -phenyl-
1,3,8-triazaspiro[4.5]decan-4-one
Figure imgf000061_0002
1H NMR (400MHz, DMSO-D6) δ 11.45(m,1H), 8.96(s,1H), 7.59(m,2H), 7.32- 7.11(m,3H), 6.85-6.56(m,5H), 6.10(m,1H), 4.56(s,2H), 4.01-3.40(m,9H), 2.96(m,2H), 2.66(m,2H), 2.31-2.12(m,2H), 1.81-1.56(m,3H), 1.50-1.21 (m,2H). mass spectrometry ES+ =580/582 (M+1, 2 Cl isotope pattern) Example 5
8-{3-[3-(3,4-dichlorophenyl)-1-pentanoylpyrrolidin-3-yl]propyl}-1-[3-
(trifluoromethyl)phenyl]-1 ,3,8-triazaspiro[4.5]decan-4-one
Figure imgf000062_0001
1H NMR (400MHz, CDCI3) δ 7.79(m,1 H), 7.36-7.01 (m,6H), 6.95-6.88(m,1 H), 4.74(s,2H), 3.87-3.78(m,1H), 3.70-3.48(m,3H), 3.41-3.34(m,1H), 2.76(m,3H), 2.36- 2.05(m,6H), 1.81(m,1H), 1.68-1.53(m,5H), 1.42-1.11(m,5H), 0.94-0.83(m,4H). mass spectrometry ES+ =638/640 (M+1 , 2 Cl isotope pattern)
Example 6
8-{3-[3-(3,4-dichlorophenyl)-1-(2-furoyl)pyrrolidin-3-yl]propyl}-1-[3-
(trifluoromethyl)phenyl]-1 ,3,8-triazaspiro[4.5]decan-4-one
Figure imgf000062_0002
1H NMR (400MHz, CDCI3) δ 7.54(m,1 H), 7.47(s,1H), 7.41-7.25(m,4H), 7.18- 7.02(m,4H), 6.46(m,1H), 4.73(s,2H), 4.10-3.74(m,4H), 2.79-2.52(m,3H) 2.38- 2.12(m,3H), 1.81-1.57(m,3H), 1.32-1.10(m,3H), 0.85-0.68(m,4H). mass spectrometry ES+ =649/651 (M+1, 2 Cl isotope pattern) Example 7
8-{3-[1-(cyclobutylcarbonyl)-3-(3,4-dichlorophenyl)pyrrolidin-3-yl]propyl}-1-[3- (trifluoromethyl)phenyl]-1 ,3,8-triazaspiro[4.5]decan-4-one
Figure imgf000063_0001
1H NMR (500MHz, DMSO-D6) δ 9.63-9.51 (m,1 H), 7.62-7.37(m,4H), 7.31-7.22(m,2H), 7.07(s,1H), 5.04(s,2H), 3.94-3.83(m,2H), 3.65(m,1H), 3.60(s,1H), 3.48-3.20(m,3H), 3.01 (m,2H), 2.31-2.02(m,10H), 1.93-1.50(m,6H), 1.41-1.18(m,2H). mass spectrometry ES+ =637/639 (M+1 , 2 Cl isotope pattern)
Example 8
8-{3-[3-(3,4-dichlorophenyl)-1-pentanoylpyrrolidin-3-yl]propyl}-1 -phenyl-1 ,3,8- triazaspiro[4.5]decan-4-one
Figure imgf000063_0002
1H NMR (300MHz, CDCI3) δ 7.50-7.27(m,4H), 7.14-7.04(m,1 H), 6.97-6.86(m,2H), 6.81-6.67(m,1H), 4J5(s,2H), 3.83-3.55(m,3H), 3.44(m,1 H), 3.10-2.65(m,7H), 2.56- 2.42(m,2H), 2.38-2.06(m,4H), 1.88-1.57(m,4H), 1.48-1.27(m,4H), 1.04-1.87(m,3H). mass spectrometry ES+ =571/573 (M+1 , 2 Cl isotope pattern) Example 9
8-{3-[1-(cyclopentylcarbonyl)-3-(3,4-dichlorophenyl)pyrrolidin-3-yl]propyl}-1-phenyl- 1 ,3,8-triazaspiro[4.5]decan-4-one
Figure imgf000064_0001
1H NMR (300MHz, CDCI3) δ 7.69-7.67(m1 H), 7.51-7.49(m,1 H), 7.34(m,1 H), 7.31-
7.18(m,3H), 7.05-6.79(m,2H), 4.69(s,1 H), 4.19(m,3H), 3.76-3.64(m,1 H), 3.51 (m,1 H),
3.28-3.12(m,1 H), 2.65(m,1 H), 2.32-2.09(m,4H), 1.72-1.61 (m,5H), 1.45-1.19(m,7H0,
0.94-0.78(m,8H). mass spectrometry ES+ =583/585 (M+1 , 2 Cl isotope pattern)
Example 10
8-{3-[1-(cyclobutylcarbonyl)-3-(3,4-dichlorophenyl)pyrrolidin-3-yl]propyl}-1-phenyl-
1 ,3,8-triazaspiro[4.5]decan-4-one
Figure imgf000064_0002
1H NMR (300MHz, CDCI3) δ 7.38-7.18(m,4H), 7.04(m,1 H), 6.90-6.85(m,2H), 6.35(m,1H), 4.69(s,1H), 3.78-3.44(m,4H), 2.78-2.63(m,5H), 2.40-2.30(m,1H), 2.17- 2.06(m,2H), 1.91-1.50(m,14H), 1.27-1.21 (m,2H). mass spectrometry ES+ =569/571 (M+1 , 2 Cl isotope pattern)
Example 11
8-{3-[1-(cyclobutylcarbonyl)-3-(3,4-dichlorophenyl)pyrrolidin-3-yl]propyl}-1-(3- methylphenyl)-1 ,3,8-triazaspiro[4.5]decan-4-one
Figure imgf000065_0001
1H NMR (500MHz, DMSO-D6) δ 8.96(s,1H), 7.67-7.49(m,2H), 7.31-7.26(m,1H), 7.08(m,1 H), 6.77(m,1 H), 6.62(m,2H), 4.58(s,2H), 3.67-3.22(m,10H), 2.98(m,2H), 2.66(m,2H), 2.25(s,3H), 2.16-2.04(m,5H), 1.94-1.79(m,3H), 1.74-1.52(m,3H), 1.42- 1.22(m,2H). mass spectrometry ES+ =583/585 (M+1 , 2 Cl isotope pattern)
Example 12 3-acetyl-8-{3-[1-acetyl-3-(3,4-dichlorophenyl)pyrrolidin-3-yl]propyl}-1-(3- methylphenyl)-1,3,8-triazaspiro[4.5]decan-4-one
Figure imgf000065_0002
F 0
1H NMR (500MHz, DMSO-D6) δ 7.61-7.49(m,2H), 7.30(m,1 H), 7.15(m,1 H), 6.89(m,1 H), 6.77(m,2H), 4.93(s,3H), 3.79(m,1H), 3.61-3.34(m,7H), 2.99(m,2H), 2.46(s,4H), 23.1-1.86(m,10H), 1.75-1.59(m,2H), 1.41-1.30(m,2H). mass spectrometry ES+ =543/545 (M+1 , 2 Cl isotope pattern) Example 13
Compound 13 was synthesized by reacting amine 20 with chlorobenzoxazole.
8-{3-[1 -(1 ,3-benzoxazol-2-yl)-3-(3,4-dichlorophenyl)pyrrolidin-3-yl]propyl}-1 -phenyl-
1 ,3,8-triazaspiro[4.5]decan-4-one
Figure imgf000066_0001
1H NMR (400MHz, DMSO-D6) δ 8.96(s,1 H), 7.58(m,2H), 7.37-7.29(m,2H), 7.26- 7.07(m,4H), 6.96(m,1H), 6.85(m,2H), 6.75(m,1H), 4.56(s,2H), 3.94-3.88(m,1H), 3.80(m,1H), 3.67(m,1H), 3.55-3.42(m,6H), 2.96(m,2H), 2.66(m,2H), 2.35-2.24(m,2H), 1.82-1.67(m,3H), 1.48-1.34(m,2H). mass spectrometry ES+ =604/606 (M+1 , 2 Cl isotope pattern)
Additional compounds of the present invention include oxygen linked compounds as illustrated hereinbelow.
Figure imgf000067_0001
The following are representative analytical data for compounds of the present invention:
IV. Intermediate Compound 3 3-(3,4-dichlorophenyl)-1 -(phenylmethyl)-3-pyrrolidinol
1HNMR (400 MHz, CDCI3) δ[ppm]: 7.63 (1 H, d, J= 2.0 Hz), 7.40-7.26 (7H, m), 3.72 (2H, s), 3.42-3.30 (1 H, m), 3.15-3.08 (1 H, m), 2.64 (1 H, d, J= 10.2 Hz), 2.60-2.53 (2H, m), 2.32-2.25 (1 H, m), 2.22-2.14 (1 H, m). 13CNMR (400 MHz, CDCI3) δ[ppm]: 145.19, 138.65, 132.53, 131.12, 130.34, 129.04, 128.68, 127.85, 127.54, 125.02, 80.21 , 68.85, 60.01 , 52.85, 42.44.
IV. Intermediate Compound 4
3-(3,4-dichlorophenyl)-1-(phenylmethyl)-3-(2-propen-1-yloxy)pyrrolidine 1HNMR (400 MHz, CDCI3) δ[ppm]: 7.54 (1 H, d, J= 2.0 Hz), 7.40-7.24 (7H, m), 5.92- 5.82 (1 H, m), 5.28 (1 H, dd, J= 17.2Hz, 1.6 Hz), 5.14 (1 H, dd, J= 10.5 Hz, 1.1 Hz), 3.71-3.67 (4H, m), 3.01-2.77 (4H, m), 2.36-2.30 (1 H, m), 2.18-2.13 (1 H, m). 13CNMR (400 MHz, CDCI3) δ[ppm]: 135.05, 132.71 , 131.20, 130.55, 128.92, 128.63, 128.52, 127.41, 125.76, 116.55, 86.06, 65.63, 64.88, 60.78, 53.50, 39.05.
IV. Intermediate Compound 5
3-(3,4-dichlorophenyl)-3-(2-propen-1-yloxy)pyrrolidine
Figure imgf000068_0001
1HNMR (400 MHz, CDCI3) δ[ppm]: 7.47 (1H, d, J = 2.0 Hz), 7.42 (1 H, d, J = 8.4 Hz), 7.23 (1 H, dd, J = 8.4 Hz, 2.0 Hz), 5.88-5.78 (1 H, m), 5.24 (1 H, dd, J = 9.3 Hz, 1.5 Hz), 3.68-3.60 (3H, m), 3.40 (1H, d, J = 12.0 Hz), 3.337-3.27 (1 H, m), 3.20-3.13 (1 H, m), 2.95 (1 H, d, J = 12.1 Hz), 2.36-2.29 (1 H, m), 2.15-2.06 (1H, m). 13CNMR (400 MHz, CDCI3) δ[ppm]: 141.61 , 134.85, 133.03, 132.09, 130.80, 128.95, 126.28, 116.77, 87.39, 65.16, 57.59, 45.87, 36.21.
Additionally Intermediate Compound 5 may be protected with an appropriate protecting group, as will be appreciated by those skilled in the art: 1 ,1- dimethylethyl 3-(3,4-dichlorophenyl)-3-(2-propen-1-yloxy)-1-pyrrolidinecarboxylate
Figure imgf000068_0002
1HNMR (400 MHz, CDCI3) δ[ppm]: 7.47-7.7.42 (2H, m), 7.24-7.21 (1 H, m), 5.85-5.75 (1 H, m), 5.23 (1 H, d, J = 17.2 Hz), 5.12 (1 H, dd, J = 10.3 Hz, 3.7 Hz), 3.88-3.42 (6H, m), 2.40-2.34 91 H, m), 2.17-2.08 (1 H, m), 1.46 (9H, s). IV. Intermediate Compound 6
1 ,1-dimethylethyl 3-(3,4-dichlorophenyl)-3-[(2-oxoethyl)oxy]-1-pyrrolidinecarboxylate 1HNMR (400 MHz, CDCI3) δ[ppm]: 9.59 (1H, d, J = 7.7 Hz), 7.47-7.45 (2H, m), 7.23- 7.21 (1 H, m), 3.94-3.72 (3H, m), 3.62-3.48 (3H, m), 2.41-2.38 (1 H, m), 2.23-2.18 (1 H, m), 1.47 (9H, s).
Figure imgf000069_0001
Acid Chlorides:
Figure imgf000069_0002
IV. Intermediate Compound 8a
1 ,1-dimethylethyl 3-(3,4-dichlorophenyl)-3-{[2-(4-oxo-1 -phenyl-1 ,3,8- triazaspiro[4.5]dec-8-yl)ethyl]oxy}-1-pyrrolidinecarboxylate
Figure imgf000069_0003
1HNMR (400 MHz, CDCI3) δ[ppm]: 7.54 (1 H, d, J = 7.9 Hz), 7.47 (1 H, d, J = 1.8 Hz), 7.42 (1H, d, J = 8.5 Hz), 7.28 (2H, d, J = 7.6 Hz), 6.90-6.83 (3H, m), 4.72 (2H, s), 3.82 (1 H, dd, J = 40.4 Hz, 11.6 Hz), 3.56 (2H, d, J = 8.5 Hz), 3.50-3.42 (1 H, m), 3.30- 3.20 (2H, m), 2.81-2.58 (8H, m), 2.40-2.37 (1H, m), 2.16-2.05 (1 H, m), 1.68 (2H, d, J = 13.1 Hz), 1.46 (9H, s). HRMS calculated for C3oH39CI2N4O (M+H)+ 589.2348 found 589.2360. IV. Intermediate Compound 8b
1 ,1-dimethylethyl 3-(3,4-dichlorophenyl)-3-[(2-{1-[3-(methyloxy)phenyl]-4-oxo-1 ,3,8- triazaspiro[4.5]dec-8-yl}ethyl)oxy]-1-pyrrolidinecarboxylate
Figure imgf000070_0001
1HNMR (400 MHz, CDCI3) δ[ppm]: 7.97 (1H, s), 7.45-7.41 (2H, m), 7.29-7.25 (1 H, m), 7.16 (1H, t, J = 8.1 Hz), 6.55 (1 H, d, J = 7.8 Hz), 6.40-6.36 (2H, m), 4.69 (2H, s), 3.87-3.76 (4H, m), 3.56-3.41 (3H, m), 3.32-3.19 (2H, m), 2.80-2.57 (8H, m), 2.40- 2.37 (1H, m), 2.16-2.05 (1H, m), 1.64 (2H, d, J = 12.8 Hz), 1.45 (9H, s). HRMS calculated for C3ιH41CI2N4O5 (M+H)+ 619.2454 found 619.2437.
IV. Intermediate Compound 8c
1 ,1-dimethylethyl 3-(3,4-dichlorophenyl)-3-({2-[2-oxo-1-(phenylmethyl)-1 ,3,8- triazaspiro[4.5]dec-8-yl]ethyl}oxy)-1-pyrrolidinecarboxylate
Figure imgf000070_0002
1HNMR (400 MHz, CDCI3) δ[ppm]: 7.41-7.15 (8H, m), 4.31 (2H, s), 3.75 (1H, dd, J = 42.5 Hz, 11.5 Hz), 3.50-3.35 (3H, m), 3.25-3.04 (4H, m), 2.67 (2H, broad s), 2.39- 2.28 (3H, m), 2.08-2.00 (1 H, m), 1.93-1.73 (4H, m), 1.47 (9H, s). LRMS calculated for C3ιH41CI2N4O4 (M+H)+603 found 603.
Additionally, as will be appreciated, the synthesis of O-piperidine analogues may be provided through a parallel synthesis. Example 14
Figure imgf000071_0001
Molecular Weight =583.51
Exact Mass =582
Molecular Formula =C30H32CI2N4O4
8-(2-{[3-(3,4-dichlorophenyl)-1 -(2-furoyl)pyrrolidin-3-yl]oxy}ethyl)-1 -phenyl-1 ,3,8- triazaspiro[4.5]decan-4-one
1HNMR (400 MHz, CDCI3) δ[ppm]: 7.71-7.64 (2H, m), 7.56 (1H, d, J = 8.4 Hz), 7.48- 7.44 (1 H, m), 7.23 (2H, t, J = 7.7 Hz), 7.16 (1 H, d, J = 3.5 Hz), 6.98 (2H, d, J = 8.9 Hz), 6.84 (1 H, t, J = 7.3 Hz), 4.65 (2H, d, J = 3.6 Hz), 4.36 (1 H, dd, J = 67.1 Hz, 11.9 Hz), 4.14-4.10 (1H, m), 3.85-3.79 (1H, m), 3.83 (1H, dd, J = 119.5 Hz, 11.9 Hz), 3.43- 3.37 (1 H, m), 3.28-3.18 (1 H, m), 2.92-2.65 (4H, m), 2.63-2.50 (4H, m), 2.45-2.38 (1 H, m), 1.67-1.57 (2H, m). HRMS calculated for C30H33CI2N-A. (M+H)+ 583.1879 found 583.1865.
Example 15
8-(2-{[1-(cyclopentylcarbonyl)-3-(3,4-dichlorophenyl)pyrrolidin-3-yl]oxy}ethyl)-1- phenyl-1 ,3,8-triazaspiro[4.5]decan-4-one
Figure imgf000071_0002
Molecular Weight =585.57
Exact Mass =584
Molecular Formula =C31H38CI2N403 1HNMR (400 MHz, CDCI3) δ[ppm]: 7.65 (1 H, dd, J = 16.8 Hz, 2.0 Hz), 7.55 (1H, dd, J = 8.3 Hz, 2.0 Hz), 7.45-7.40 (1 H, m), 7.24 (2H, t, J = 7.7 Hz), 7.00 (2H, d, J = 8.5 Hz), 6.84 (1 H, t, J = 7.3 Hz), 4.66 (2H, s), 4.08 (1H, dd, J = 12.6 Hz, 1.7 Hz), 3.82- 3.78 (1H, m), 3.70-3.66 (1H, m), 3.60-3.36 (3H, m), 3.27-3.18 (1H, m), 2.96-2.87 (3H, m), 2.80-2.72 (2H, m), 2.62-2.52>(5H, m), 2.38-2.22 (1 H, m), 1.92-1.82 (2H, m), 1.77- 1.56 (8H, m). HRMS calculated for C3iH39CI2N4O3 (M+H)+ 585.2399 found 585.2384.
Example 16
8-(2-{[1-acetyl-3-(3,4-dichlorophenyl)pyrrolidin-3-yl]oxy}ethyl)-1 -phenyl-1 ,3,8- triazaspiro[4.5]decan-4-one
Figure imgf000072_0001
Molecular Weight =531.48
Exact Mass =530
Molecular Formula =C27H32CI2N403
1HNMR (400 MHz, CDCI3) δ[ppm]: 7.65 (1 H, dd, J = 12.8 Hz, 2.2 Hz), 7.55 (1 H, dd, J
= 8.4 Hz, 2.7 Hz), 7.42 (1 H, td, J = 8.9 Hz, 2.0 Hz), 7.24 (2H, td, J = 9.3 Hz, 2.3 Hz), 6.99 (2H, d, J = 8.2 Hz), 6.84 (1 H, t, J = 7.3 Hz), 4.66 (2H, s), 4.07 (1H, t, J = 11.2
Hz), 3.79-3.66 (2H, m), 3.58-3.35 (2H, m), 3.26-3.19 (1H, m), 2.95-2.83 (2H, m),
2.79-2.70 (2H, m), 2.63-2.52 (5H, m), 2.38-2.22 (1 H, m), 2.07 (2H, d, J = 3.6 Hz),
1.66 (2H, d, J = 13.7 Hz). HRMS calculated for C27H33CI2N4O3 (M+H)+ 531.1929 found 531.1920.
Example 17
8-(2-{[3-(3,4-dichlorophenyl)-1-(phenylsulfonyl)pyrrolidin-3-yl]oxy}ethyl)-1-phenyl- 1 ,3,8-triazaspiro[4.5]decan-4-one
Figure imgf000073_0001
Molecular Weight =629.61
Exact Mass =628
Molecular Formula =C31H34CI2N404S
1HNMR (400 MHz, CDCI3) δ[ppm]: 7.87 (2H, d, J = 7.3 Hz), 7.66 (1 H, t, J = 7.2 Hz),
7.58 (2H, 7.8 Hz), 7.51 (1 H, d, J = 1.8 Hz), 7.47 (1 H, d, J = 8.4 Hz), 7.29 (1 H, dd, J =
8.4 Hz, 2.0 Hz), 7.23 (2H, t, J = 8.2 Hz), 6.96 (2H, d, J = 8.2 Hz), 6.83 (1H, t, J = 7.3
Hz), 4.66 (2H, s), 3.79 (1H, dd, J = 11.3 Hz, 1.1 Hz), 3.57 (1H, t, J = 7.5 Hz), 3.46
(1 H, d, J = 11.4 Hz), 3.42-3.38 (1 H, m), 3.12-3.09 (1H, m), 2.94-2.90 (1H, m), 2.80-
2.70 (2H, m), 2.61-2.40 (5H, m), 2.22-2.13 (3H, m), 1.61 (2H, d, J = 11.4 Hz).
Example 18
8-(2-{[3-(3,4-dichlorophenyl)-1-(2-furoyl)pyrrolidin-3-yl]oxy}ethyl)-1-(3- methoxyphenyl)-1 ,3,8-triazaspiro[4.5]decan-4-one
Figure imgf000073_0002
Molecular Weight =613.54
Exact Mass =612
Molecular Formula =C31H34CI2N405
1HNMR (400 MHz, CDCI3) δ[ppm]: 7.70-7.63 (2H, m), 7.56 (1H, d, J = 8.3 Hz), 7.49- 7.45 (1 H, m), 7.16-7.12 (2H, m), 6.65-6.61 (1 H, m), 6.59-6.55 (1 H, m), 6.44 (2H, d, J = 6.2 Hz), 4.63 (2H, d, J = 3.3 Hz), 4.35 (1 H, dd, J = 67.0 Hz, 11.9 Hz), 4.25-4.20 (1 H, m), 3.85-3.79 (1 H, m), 3.82 (1 H, dd, J = 109.8 Hz, 12.0 Hz), 3.75 (3H, s), 3.42- 3.39 (1 H, m), 3.30-3.18 (1H, m), 2.90-2.65 (5H, m), 2.64-2.52 (4H, m), 2.45-2.39 (1 H, m), 1.65-1.55 (2H, m). HRMS calculated for C3ιH35CI2N4O5 (M+H)+ 613.1985 found 613.1987. Example 19
8-(2-{[1-(cyclopentylcarbonyl)-3-(3,4-dichlorophenyl)pyrrolidin-3-yl]oxy}ethyl)-1-(3- methoxyphenyl)-1 ,3,8-triazaspiro[4.5]decan-4-one
Figure imgf000074_0001
Molecular Weight =615.60 Exact Mass =614
Molecular Formula =C32H40CI2N4O4
1HNMR (400 MHz, CDCI3) δ[ppm]: 7.64 (1 H, dd, J = 17.0 Hz, 2.0 Hz), 7.55 (1 H, dd, J = 8.4 Hz, 5.3 Hz), 7.43 (1H, ddd, J = 8.4 Hz, 7.2 Hz, 2.0 Hz), 7.15 (1H, t, J = 8.4 Hz), 6.65-6.62 (1H, m), 6.45-6.43 (2H, m), 4.64 (2H, s), 4.11 (1 H, q, J = 11.3 Hz), 3.83- 3.79 (1H, m), 3.75 (3H, s), 3.70-3.65 (1 H, m), 3.58-3.52 (1 H, m), 3.47 (1 H, d, J = 12.6 Hz), 3.41-3.35 (1H, m), 3.27-3.19 (1H, m), 2.98-2.84 (3H, m), 2.78-2.70 (2H, m), 2.64-2.52 (4H, m), 2.38-2.21 (1 H, m), 1.92-1.80 (2H, m), 1.75-1.55 (8H, m). HRMS calculated for C32H41CI2N4O4 (M+H)+ 615.2505 found 615.2496.
Example 20
8-(2-{[1-acetyl-3-(3,4-dichlorophenyl)pyrrolidin-3-yl]oxy}ethyl)-1-(3-methoxyphenyl)-
1 ,3,8-triazaspiro[4.5]decan-4-one
Figure imgf000074_0002
Molecular Weight =561.51
Exsct IVlsss —560
Molecular Formula =C28H34CI2N404
1HNMR (400 MHz, CDCI3) δ[ppm]: 7.65 (1 H, dd, J = 12.6 Hz, 2.0 Hz), 7.55 (1 H, dd, J = 8.4 Hz, 3.1 Hz), 7.43 (1H, td, J = 9.3 Hz, 2.0 Hz), 7.15 (1H, td, J = 8.6 Hz, 3.1 Hz), 6.66-6.63 (1 H, m), 6.45-6.43 (2H, m), 4.64 (2H, s), 4.06 (1H, t, J = 12.7 Hz), 3.75 (3H, s), 3.72 (2H, t, J = 4.2 Hz), 3.57-3.50 (1H, m), 3.45 (1H, d, J = 12.6 Hz), 3.41- 3.35 (1H, m), 3.26-3.19 (1 H, m), 2.93-2.83 (2H, m), 2.78-2.69 (2H, m), 2.64-2.52 (4H, m), 2.39-2.22 (1H, m), 2.07 (3H, d, J = 2.7 Hz), 1.64 (2H, d, J = 13.5 Hz). HRMS calculated for C28H35CI2N4O4 (M+H)+ 561.2035 found 561.2051.
Example 21
8-(2-{[3-(3,4-dichlorophenyl)-1-(phenylsulfonyl)pyrrolidin-3-yl]oxy}ethyl)-1-(3- methoxyphenyl)-1 ,3,8-triazaspiro[4.5]decan-4-one
Figure imgf000075_0001
Molecular Weight =659.63
Exact Mass =658
Molecular Formula =C32H36CI2N405S
1HNMR (400 MHz, CDCI3) δ[ppm]: 7.88 (2H, d, J = 7.5 Hz), 7.68-7.64 (1H, m), 7.61- 7.57 (2H, m), 7.52 (1 H, d, J = 2.0 Hz), 7.48 (1 H, d, J = 8.2 Hz), 7.31 (1 H, dd, J = 8.3
Hz, 2.0 Hz), 7.15 (1 H, t, J = 8.8 Hz), 6.62 (1 H, dd, J = 7.4 Hz, 2.0 Hz), 6.45-6.44 (2H, m), 4.65 (2H, s), 3.81-3.76 (4H, m), 3.57 (1H, broad t, J = 3.7 Hz), 3.48-3.38 (2H, m),
3.13-3.07 (1H, m), 2.98-2.85 (2H, m), 2.78-2.71 (2H, m), 2.60-2.49 (4H, m), 2.45-
2.39 (1H, m), 2.24-2.14 (3H, m), 1.63-1.58 (2H, m). HRMS calculated for C32H37CI2N4O5S (M+H)+ 659.1862 found 659.1845.
V. B-rinq is azetidine
Figure imgf000076_0001
ux oride
Figure imgf000076_0002
Synthesis of Intermediate 1 ethyl cyano(3,4-dichlorophenyl)acetate '
To a solution of diethyl carbonate (4.1 mL, 33.88 mmol) in toluene (150 mL) at r.t, sodium hydride (1.43 g, 60% in mineral oil, 35.75 mmol) and 3,4-dichlorophenyl acetonitrile (6.34 g, 34.09 mmol) were added. The content was stirred at 80 °C overnight. After cooling to r.t, HCI was added (1N in H20, 100 mL). The content was extracted with ethyl acetate (3x). The combined organic layer was washed with water, dried (Na2SO4), filtered and concentrated. The residue was purified by flash column chromatography with hexane/ethyl acetate (6/1) to give 7.42 g product (85% yield) as brown oil. 1H NMR (400 MHz, CDCI3) δ[ppm]: 7.60 (1H, broad s), 7.54 (1H, d, J = 8.3 Hz), 7.35 (1H, broad d, J = 8.3 Hz), 4.72 (1H, s), 4.29 (2H, q, J = 14.2 Hz, 7.0 Hz), 1.28 (3H, t, J = 6.7 Hz). LRMS calculated for CnHgCfeNOa M+ 257 found 257. Synthesis of Intermediate 3 ethyl 2-cyano-2-(3,4-dichlorophenyl)-4-(1,3-dioxolan-2-yl)butanoate
To a solution of 1 (7.02 g, 27.32 mmol) and 2-(2-bromoethyl)-1 ,3-dioxolane compound 2 (3.85 mL, 32.78 mmol) in anhydrous DMF (100 mL), potassium t- butoxide (32.78 mL, 1 M in t-BuOH) was added. The content was heated to 80 °C and stirred overnight. H2O (400 mL) and brine (200 mL) were added to the content and the mixture was extracted with ethyl acetate. The combined organic layer was dried over sodium sulfate, filtered and concentrated. Flash column chromatography with hexane/ethyl acetate (6/1) afforded 9.5 g brown oil (-85% product+~15%SM). 1H NMR (400 MHz, CDCI3) δ[ppm]: 7.66 (1H, d, J = 2.3 Hz), 7.48 (1 H, d, J = 8.4 Hz), 7.40 (1 H, dd, J = 8.4 Hz, 2.2 Hz), 4.91 (1 H, t, J = 4.4 Hz), 4.30-4.20 (2H, m), 3.98- 3.94 (2H, m), 3.86-3.83 (2H, m), 2.48 (1H, td, J = 12.2 Hz, 4.5 Hz), 2.24 (1H, td, J = 12.2 Hz, 4.3 Hz), 1.88-1.81 (1 H, m), 1.75-1.68 (1H, m), 1.24 (3H, t, J = 7.1 Hz). 13C NMR (400 MHz, CDCI3) δ[ppm]: 166.90, 134.68, 133.70, 131.28, 128.60, 125.88, 117.58, 103.04, 65.26, 63.91, 53.22, 32.24, 29.88, 14.06.
Synthesis of Intermediate 5 3-(3,4-dichlorophenyl)-3-[2-(1,3-dioxolan-2-yl)ethyll-2-azetidinone
To a solution of 3 (1.50 g, 4.20 mmol) in EtOH (100 mL), Raney-Nickel (1g) was added. The content was stirred at 60 psi H2 overnight. It was then filtered through celite and concentrated. The residue was dissolved in Et2O (20 mL), CH3Mgl (5.6 mL, 3M in Et20, 16.8 mmol) was added. The content was stirred at r.t ovenight. The reaction was quenched with saturated sodium bicarbonate solution, filtered through celite, extracted with EtOAc, dried and then concentrated. The residue was purified with flash column chromatography with hexane/EtOAc (1/2) to give 0.78 g product (59% yield over two steps) as light yellow solid. 1H NMR (400 MHz, CDCI3) δ[ppm]: 7.49 (1 H, d, J = 2.1 Hz), 7.40 (1 H, d, J = 8.3 Hz), 7.25 (1 H, dd, J = 8.2 Hz, 1.9 Hz), 6.23 (1H, s), 4.82 (1H, t, = 4.5 Hz), 3.93-3.88 (2H, m), 3.83- 3.78 (2H, m), 3.50 (2H, dd, J = 15.9 Hz, 5.5 Hz), 2.10-2.02 (2H, m), 1.81-1.72 (1H, m), 1.61-1.52 (1 H, m). 13C NMR (400 MHz, CDCI3) δ[ppm]: 171.44, 140.05, 132.82, 131.48, 130.74, 128.92, 126.42, 103.83, 65.17, 62.98, 48.63, 31.37, 29.36. LRMS calculated for C146CI2NO3 (M+H)+ 316 found 316.
Synthesis of Intermediate 7 (Example 22) 8-{3-[3-(3,4-dichlorophenyl)-1-(phenylsulfonyl)-3-azetidinyl]propyl}-1-phenyl- 1,3,8-triazaspiro[4.5]decan-4-one
To a solution of 5 (0.778 g, 2.47 mmol) in THF (15 mL), LAH (4.94 mL, 1M in Et2OH, 4.94 mmol) was added at r.t. The content was heated to reflux for 5 hrs and then continued stirring ar r.t overnight. The reaction was cooled to 0 °C and quenched with H2O (0.23 mL), sodium hydroxide (15%, 0.23 mL) and H2O (0.69 mL) sequencially. Then it was filtered and concentrated to give a colorless oil.
Half of the oil was dissolved in dichloromethane. Triethylamine (0.518 mL, 3.72 mmol) was added, followed with benzenesulfonyl chloride (0.237 mL. 0.86 mmol). The content was shaked at r.t for 30 mins. The organic layer was washed with 1 N sodium hydroxide, dried over sodium sulfate. Prep. TLC separation with hexane/EtOAc (1/1) afforded 100 mg product as colorless oil.
To a solution of the oil in THF (9 mL), concentrated HCI (1 mL) was added. The content was stirred at r.t overnight. It was quenched with saturated sodium bicarbonate solution, extracted with ethyl acetate. The organic layer was dried over sodium sulfate, filtered and concentrated to give a colorless oil.
To a solution of the oil in THF (5 mL), amine 6 (0.15 g, 0.65 mmol)was added. The content was stirred at r.t for 10 mins before NaBH(OAc)3 (0.27 g, 1.30 mmol) was added. The content was shaken at r.t overnight. The reaction was quenched with saturated sodium bicarbonate and extracted with ethyl acetate. The organic layer was dried over sodium sulfate, filtered and concentrated resulting in the 5:1 mixture of 7 and 8. The residue was purified by PHPLC to remove the monochloro by-product and resulted in 31 mg of product 7 (Example 22) as white solid (formic acid salt). 1H NMR (400 MHz, CDCI3) δ[ppm]: 8.41 (1 H, s), 7.81 (2H, d, J = 7.4 Hz), 7.61 (1 H, t, J = 7.3 Hz), 7.54 (2H, t, J = 7.5 Hz), 7.33 (1 H, d, J = 8.3 Hz), 7.27 (2H, t, J = 7.7 Hz), 6.98 (1 H, d, J = 2.2 Hz), 6.89-6.85 (3H, m), 6.78 (1 H, dd, J = 8.3 Hz, 2.2 Hz), 6.46 (1 H, s), 4.73 (2H, s), 3.91 (2H, d, J = 7.9 Hz), 3.84 (2H, d, J = 7.9 Hz), 3.37- 3.21 (4H, m), 2.99 (2H, td, J = 14.3 Hz, 4.9 Hz), 2.73 (2H, t, J = 7.7 Hz), 1.85-1.73 (4H, m), 1.48-1.40 (2H, m). LRMS calculated for C3ιH35CI2N4O3S (M+H)+ 613.2 found 613.3.
The following two compounds were made in a similar way. Except substituting 2-furancarbonyl chloride (23) , cyclopentanecarbonyl chloride (24) for benzenesulfonyl chloride in the synthesis describe above for example 22.
Figure imgf000079_0001
Molecular Weight =567.51 Molecular Weight =569.57
Exact Mass =566 Exact Mass =568
Molecular Formula =C30H32CI2N4O3 Molecular Formula =C31 H38CI2N4O2
Example 23: 8-{3-[3-(3,4-dichlorophenyl)-1 -(2-furanylcarbonyl)-3- azetidinyl]propyl}-1-phenyl-1,3,8-triazaspiro[4.5]decan-4-one. 1H NMR (400 MHz, CDCI3) δ[ppm]: 7.49 (1 H, s), 7.43 (1 H, d, J = 8.2 Hz), 7.30-7.26 (4H, m), 7.08 (1 H, d, J = 3.3 Hz), 7.00 (1 H, dd, J = 8.2 Hz, 2.0 Hz), 6.94-6.85 (3H, m), 6.50-6.47 (1 H, m), 4.76-4.68 (3H, m), 4.61-4.56 (1H, m), 4.36 (1 H, d, J = 9.9 Hz), 4.22 (1 H, d, J = 9.7 Hz), 2.82-2.52 (6H, broad), 2.46-2.30 (2H, broad s), 2.04-1.96 (2H, m), 1.70 (2H, d, J = 12.8 Hz), 1.37 (2H, broad s). LRMS calculated for C30H33CI2N4O3 (M+H)+ 567 found 567.
Example 24: 8-{3-[1 -(cyclopentylcarbonyl)-3-(3,4-dichlorophenyl)-3- azetidinyl]propyl}-1-phenyl-1,3,8-triazaspiro[4.5]decan-4-one.1H NMR (400 MHz, CDCI3) δ[ppm]: 7.44 (1H, d, J = 8.2 Hz), 7.30-7.25 (3H, m), 7.20 (1 H, d, J - 2.2 Hz), 6.97 (1H, dd, J = 8.4 Hz, 2.0 Hz), 6.92-6.85 (2H, m), 6.45 (1 H, broad s), 4.71 (2H, s), 4.30 (1H, d, J = 8.1 Hz), 4.24 (2H, d, J = 11.4 Hz), 4.18 (1 H, d, J = 9.5 Hz), 2.70 (4H, broad s), 2.63-2.54 (2H, m), 1.95-1.90 (2H, broad), 1.80-1.67 (10H, m), 1.57-1.50 (2H, m), 1.37-1.20 (2H, broad). LRMS calculated for C3iH39CI2N4O3 (M+H)+ 569 found 569. As will be appreciated by those skilled in the art, additional compounds of the present invention may be similarly prepared according to the schemes provided herein.
BIOLOGICAL DATA
The following definitions apply:
Figure imgf000080_0001
CC-Chemokine Receptor-5 Binding by Scintillation Proximity Assay (CCR5 SPA)
Scintillation Proximity Assay for the Human CC-Chemokine Receptor, CCR-5
This protocol describes a high-throughput screen using SPA binding to identify compounds that inhibit binding of 125l-MIP1α to the human CCR5 chemokine receptor.
CCR5 is a G protein-coupled receptor that binds the natural chemokine ligands,
MIP1α, MIP1β and RANTES. CCR5 acts as a co-receptor with CD4 for entry of HIV- 1 into human T-cells and monocytes. Chemokines also play a role in acute and chronic inflammatory processes. Chemokines are soluble proteins produced and released by a wide variety of cell types during the initial phase of a host response to a forgein substance entering the body.
Human CCR5 receptors were expressed in Chinese Hamster Ovary (CHO) cells, registration # 12025. Cells were grown in suspension and a 50 to 80 ml CCR5 cell pellet was prepared. Membrane preparation: 1) Weigh pellet; 2)Prepare an ice-cold 50mM HEPES buffer, containing 0.0025mg/ml Pefabloc, 0.0001 mg/ml Pepstatin A, 0.0001 mg/ml Leupeptin, 0.0001 mg/ml Aprotinin (protease inhibitor cocktail), pH 7.4; 3) Homogenize pellet in 5 volumes of HEPES buffer; 4) Homogenize again with a glass homogenizer 10 to 20 strokes; 5) Centrifuge homogenate at 18,000 rpm in a F28/36 rotor using a Sorvall RC26 PIUS refrigerated Centrifuge for 30 minutes; 6) Discard supernatant and resuspend pellet in 3 volumes of HEPES buffer; 7) Homogenize and centrifuge again using steps 4-6, 2 more times; 8) Reweigh pellet and homogenize in 3X weight-to-volume of HEPES buffer; 9) Aliquot 0.5 to 1.5ml of the membrane preparation into small vials and store at -80 degrees Centigrade; 10) Determine the protein concentration of the membrane preparation using the Bio-Rad or BCA method; 11) The membrane homogenate will need to be characterized for the assay conditions a.) Protein concentration; b.) Optimal protein-to-bead ratio in SPA; and c.) Saturation curve to determine Kd and Bmax in SPA
The saturation curve binding experiment is performed by adding varying amounts of [125l]MIP1 (0-8.5nM to membranes and beads in concentrations chosen from the optimal protein/bead ratio. The data is analyzed using a non-linear curve-fitting program. The Kd and Bmax are derived from the curve.
Bacitracin 50mg/ml is dissolved in deionized water, brought to a boil for 5 minutes (to destroy protease activity) and cooled. Prepared 1ml aliquots and store at -80°C.
Protease inhibitor cocktail is prepared by dissolving 25 mg/ml of Pefabloc, 1 mg/ml of Leupeptin, 1 mg/ml of Aprotinin and 1 mg/ml of Pepstatin A in 100% DMSO. The cocktail can be aliquoted and stored frozen at -20°C until needed.
Sigmacote: Any reagent bottles and reservoirs that come in contact with the radioligand are treated with Sigmacote to reduce sticking. Rinse containers with undiluted Sigmacote; rinse with deionized water several times, and allow to air dry before using.
Color Quench Assay- [125l] SPA PVT color quench kit, Cat. No. RPAQ 4030, Amersham Ltd. A color quench curve is generated for each Packard TopCount and is stored in each counting protocol specific for the assay. This is done to prevent colored compounds from quenching the scintillation counts.
Compounds Preparation: Compounds for a single concentration determination (One Shots) are delivered in 96 well Packard Optiplates containing 1 μl of compound in 100% DMSO in columns A1- H10 (80 compounds/plate). Column A11 to H11 is used for total binding (Bo) (vehicle-5 μl of the appropriate DMSO concentration) and column A12 to D12 is used for determination of nonspecific binding. No further preparation is required.
Compounds for concentration-response curves (10 points) are delivered in 96- Packard Optiplates containing 1 μl of compound in 100% DMSO in columns A1-H10. A 10-point concentration-response curve is desired for each compound with a starting high concentration of 30 μM (in the assauy final). Column A11 to H11 is used for total binding (Bo) (vehicle-5 μl of the appropriate DMSO concentration) and column A12 to D12 is used for determination of nonspecific binding. No further preparation is required
Materials:
1 M HEPES, pH 7.4, Gibco, Cat. No. 15360-080
Bacitracin, Sigma Catalog. Number. B-0125
Bovine Serum Albumin, Sigma, Cat. No. A-7888
MgCI2, J.T. Baker 2444-01 CaCI2, Sigma, Cat. No. C5080
MIP1α, Peprotech, Cat. No. 300-08
Sigmacote, Sigma, Cat. No. SL2
Scintillation Proximity Beads, Wheat Germ Agglutinin, Amersham, Cat No. RPNQ
0001 [125l]MIP1α, NEN (#NEX298)
Packard 96 well flat-bottom Optiplate, Cat. No. 6005190
Falcon 96 well round-bottom plate, Cat. No. 3077
TOPSEAL-S, Packard, Cat. No. 6005161
Dimethyl Sulfoxide, EM Science, Cat. No. MX1458-6 Siliconized Pipette tips, Accutip, volume 200-1300uL, Cat. No. P5048-85 l
Siliconized Pipette tips, Bio Plas, Inc., volume 1-200uL, Cat. No. 60828-908 Reagent Reservoir, Elkay, Cat. No. 175-RBAS-000
Assay Buffer Preparation: 50 mM HEPES buffer pH 7.4, 1mM CaCl2, 5mM MgCI2(this can be made ahead as a 10OX stock), 1 % BSA, 0.5 mg/ml Bacitracin, Protease inhibitor Cocktail (see preparation above) 100 uL/100 ml, DMSO is added to equal a final concentration of 2% per well (includes compound %DMSO) if needed.
Experimental Details: r125l1MIP1α preparation:
Prepared radioligand dilutions in container treated with Sigmacote Reconstitute each 50 μCi vial with 0.5 ml of deionized water and store at 4°C. Specific Activity = 2,000 Ci/mmol
Add -60,000 cpm (0.17 nM) to each assay well in 50uL
Make a 20% DMSO solution and add 5uls of this to each well in col A1 1-H11. This gives a final 2% DMSO concentration for the well when added to the 1% in the assay buffer.
NSB:
Make a stock dilution of MIP1α at 100uM using deionized water; aliquot and freeze. Dilute the MIP-1α stock solution to a concentration of 2 μM in the same 20% DMSO solution used above and add 5 μl to the wells in column A12 to D12 to give a final assay concentration of 100nM. Prepare this in a Sigmacote-treated container
Membrane and SPA Bead preparation- The final assay concentration for the membrane is 15 μg per well. SPA beads are prepared by adding 5 ml of assay buffer to a 500 mg vial. The final concentration of SPA beads in the assay is 0.25 mg/well. Membranes and beads are premixed as a 1 :1 (membrane:bead) mixture and maintained at mixture at 4°C with constant stirring. 50 μl of the mixture is added to each assay well. After all reagents have been added to the plates (total assay volume 100 μl), shake plates for 4 hours at room temperature. After 4 hours, place the plates on the TopCount in a count the plates on the TopCount for 30 sec per well using an appropriate program (i.e., one with a quench curve established for the conditions of the assay. Data Reduction:
Data reduction is performed using the Microsoft Excel Addins Robofit or Robosage.
For single concentration assays (One Shots), the result of each test well is expressed as % inhibition using the following formula: 100*(1-(U1-C2)/(C1-C2)). Where U1 is the unknown sample in cpm observed in a particular well, C1 is the average of column 12 cpm observed in the absence of any added inhibitor, and C2 is the average of column 11 cpm observed in the presence of 1μM of MIP1 .
For concentration-response assays, the result of each test well is expressed as %B/Bo (% total specific binding) using the following formula: 100* (U1-C2)/C1-C2). Curves were generated by plotting the %B/Bo versus the concentration and the IC50 is derived using the equation y=Vmax*(1-(xΛn/(kΛn+xΛn))).
Controls and Standards:
Each plate contains 12 wells of total binding (column A11-H11). The cpm/well are averaged and are used in data reduction as value d .Each plate also contains 4 wells of non-specific binding (wells A12-D12). The counts of these wells are averaged and used in data reduction as value C2.
A standards plate is included in each experiment. This plate contains a 14-point concentration-response curve (in triplicate) for the standard compound MIP1α at a starting concentration of 1 μM. The average historical pKj obtained with MIP1α is 7.6.
The relevant biological response field for a single concentration (One Shots) is % inhibition. Inhibition values of >40 or >50% were considered positive responses.
The relevant biological response field for a concentration-response experiment is pK| HOS Assay (Also referred to as HOS-LTR-Luciferase Assay)
Materials
DMEM (GibcoBRL # 10564-011) Trpsin-EDTA (GibcoBRL #25300-054)
Heat inactivated Fetal Bovine Serum (FBS) (Hyclone # SH30070.03) 96-well, black-walled, clear-bottom, tissue culture-treated plates (Costar # 3904) 96-well, clear-walled, clear-bottom tissue culture-treated plates (Costar # 3598) Phosphate Buffered Saline (PBS) (GibcoBRL #14190-144) Dimethyl Sulfoxide (DMSO) (Sigma # D2650) Luclite Luciferase Reporter assay (Packard #6016911) HOS-CD4.CCR5-LTR-Luciferase (Bioresource Registration # 21164): Human Osteosarcoma cell line engineered to overexpress human CD4 and human CCR5 (AIDS Repository cat# 3318) stabily transfected with HIV-1 -LTR-Luciferase reporter.
Advanced Preparation Growth and Maintenance of the HOS-CD4.CCR5-LTR-Luciferase cell line:
The cells were propagated in DMEM containing 2% FBS. Cells were split by standard trypsinization when confluency reached 80% (roughly every 2 to 3 days).
Titering of virus stocks: HIV-1 virus stocks were titered in the assay system in order to obtain an estimate of the number of infectious particles per unit volume (described as RLU/ml). Virus stocks were diluted into DMEM containing 2% FBS and assayed as described in the "procedure" section below.
Procedure
Black-walled 96-well tissue culture plates were seeded with HOS-CD4.CCR5-LTR- Luciferase @ 0.6 to 1.2 x 103 cells per well in 50ul DMEM containing 2% FBS and placed in a humidified incubator @ 37°C, 5% CO2 overnight. The following day, test compounds were titrated 4-fold at 2X the final concentration in DMEM + 2% FBS + 0.2% DMSO. 50μl of titrated compound was transferred to the HOS cells and the plates were placed in a humidified incubator at 37°C, 5% CO2for 1hr. An additional 60ul of 2X titrated compound was transferred to a clear-walled 96-well tissue culture plate and 60ul of HIV (diluted to appropriate m.o.i.) was added to each well and thoroughly mixed. 100ul of the HIV/compound mixture was transferred to the black- walled plates containing 100ul of cells/compound. The plates were placed in a humidified incubator at 37°C, 5% CO2for 72hr Following the 72 hour incubation, 150ul of supernatant was removed and 50ul of reconstituted LUCLITE (kit reagent) was added to each well. Each plate was sealed and read in a Topcount (Packard) luminometer at 1s/well. Data Reduction
Relative Light Units (RLU) were expressed as % control (RLU at drug [ ] / RLU no drug)*100 = % Control
IC50 values were determined by any one of the following four nonlinear regression models: y=Vmax*(1 -(xΛn/(KΛn+xΛn)))+Y2 y=Vmax*(1 -(xΛn/(KΛn+xΛn))) y=Vmax*(1 -(x/(K+x)))+Y2 y=Vmax*(1-(x/(K+x))) Where: K is IC5o, Y2 is baseline, and N is Hill Coefficient
Each of the compounds of the present invention provides a plC50 value of at least 5 when tested in each of the above-described assays.
Test compounds are employed in free or salt form. All research complied with the principles of laboratory animal care (NIH publication No. 85-23, revised 1985) and standard pharmaceutical industry policy on animal use.
Although specific embodiments of the present invention have been illustrated and described in detail, the invention is not limited thereto. The above detailed description of preferred embodiments is provided for example only and should not be construed as constituting any limitation of the invention. Modifications will be obvious to those skilled in the art, and all modifications that do not depart from the spirit of the invention are intended to be included within the scope of the appended claims.

Claims

What is claimed is:
A compound of formula (I)
Figure imgf000087_0001
and pharmaceutically acceptable derivatives thereof, wherein X is a C-ι-5 alkylene chain, wherein said X is optionally substituted by one or more =O, =S, -S(O)r, alkyl, or halogen and wherein said C^ alkylene chain may optionally have 0-3 heteroatoms selected from oxygen, phosphorus, sulfur, or nitrogen;
Ring A is a saturated, partially saturated, or aromatic 3-7 monocyclic or 8-10 membered bicyclic ring having one ring nitrogen and 0-4 additional heteroatoms selected from oxygen, phosphorus, sulfur, or nitrogen;
Ring B is a saturated 4 or 5 membered ring containing the depicted ring nitrogen;
R1 is alkyl optionally substituted by one or more R7, alkenyl optionally substituted by one or more Rr, alkynyl optionally substituted by one or more R7, cycloalkyl optionally substituted by one or more R8, heterocyclyl optionally substituted by one or more R8, heteroaryl optionally substituted by one or more R6, or aryl optionally substituted by one or more R6; or R1 and X taken together form a saturated, partially saturated or aromatic 5-6 membered ring having 0-3 heteroatoms selected from oxygen, phosphorus, sulfur, or nitrogen that is fused to Ring A; each R2 is independently selected from the group consisting of -OR0, -C(O)-R°, -S(O)2-R°, -C(O)-N(R°)2, -S(O)2-N(R°)2, -(CH2)a-N(R°)(-Vb-R+), -(CH2)a-(-V -R+), halogen, alkyl optionally substituted by one or more R7, alkenyl optionally substituted by one or more R7, alkynyl optionally substituted by one or more R7, aryl optionally substituted by one or more R6, heteroaryl optionally substituted by one or more R6, cycloalkyl optionally substituted by one or more R8, and heterocyclyl optionally substituted by one or more R8; and two adjacent R2s on Ring A are optionally taken together to form a fused, saturated, partially saturated or aromatic 5-6 membered ring having 0-3 heteroatoms selected from oxygen, phosphorus, sulfur, or nitrogen; or two geminal R2s are optionally taken together to form a spiro, saturated, partially saturated or aromatic 5-6 membered ring having 0-3 heteroatoms selected from oxygen, phosphorus, sulfur, or nitrogen, said fused or spiro ring being optionally substituted by one or more R8; each a independently is 0-3; each b independently is 0 or 1 ;
V is -C(O)-, -C(O)O-, -S(O)2-, or -C(O)-N(R0)-;
R+ is alkyl, cycloalkyl, aralkyl, aryl, heteroaryl, heteroaralkyl, or heterocyclyl, wherein said R+ is optionally substituted by one or more R8; m is 0 or 1 ; n is 0-5;
R3 is H, -N(R°)2, -N(R°)C(O)R°, -CN, halogen, CF3, alkyl optionally substituted by one or more groups selected from R7 or -S-aryl optionally substituted by -(CH^-6- N(R0)SO2(R0), alkenyl optionally substituted by one or more groups selected from R7 or -S-aryl optionally substituted by -(CH2)ι-6-N(R0)SO2(R0), alkynyl optionally substituted by one or more groups selected from R7 or -S-aryl optionally substituted by -(CH2)1-6-N(R°)SO2(R°), cycloalkyl or carbocyclyl optionally substituted by one or more R8, aryl optionally substituted by one or more R6, heteroaryl optionally substituted by one or more R6, or heterocyclyl optionally substituted by one or more
R8;
Y is alkyl, alkenyl, alkynyl, -(CR4R5)P-, -C(O)-, -C(O)C(O)-, -C(S)-, -O-(CH2 C(O)-, -(CHs C O-O-, -N(R°)-C(O)-, -C(O)-N(R0)-, -N(R°)-C(S)-, -S(O)r, -O-C(=N-CN)-, -O-C(=N-R0)-, -C(=N-CN)-O-, -C(=N-R°)-O-, -C(=N-CN)-S-, -S-C(=N-CN)-, -N(R°)-C(=N-CN)-, -C(=N-CN)-, -N(R°)-C[=N-C(O)-R°], -N(R0)-C[=N-S(O)rR°], -N(R°)-C(=N-OR0)-, -N(R°)-C(=N-R0)-, or -C(=N-R0)-; each R4 is independently H, alkyl optionally substituted by R7, alkenyl optionally substituted by R7, or alkynyl optionally substituted by R7; each R5 is independently selected from H, -C(O)-OR6, -C(O)-N(R°)2, -S(O)2- N(R°)2, -S(O)2-R°, aryl optionally substituted by R6, or heteroaryl optionally substituted by R6; p is 1-5; each t independently is 1 or 2; each R6 is independently selected from the group consisting of halogen, -CF3, -OCF3, -OR0, -(CH^-e-OR0, -SR°, -(CH2)ι-6-SR°, -SCF3, -R°, methylenedioxy, ethylenedioxy, -NO2, -CN, -(CH^-CN, -N(R°)2, -(CH2)1-6-N(R°)2, -NR°C(O)R°, -NR°(CN), -NR°C(O)N(R°)2, -NR°C(S)N(R°)2, -NR°CO2R°, -NR°NR°C(O)R°, -NR0NR°C(O)N(R°)2, -NR0NR0CO2R°, -C(O)C(O)R0, -C(0)CH2C(0)R°, -(CH2)o-6C02R0, -O-C(O)R0, -C(0)R°, -C(O)N(R0)N(R°)2, -C(0)N(R°)2, -C(O)N(R0)OH, -C(O)N(R°)SO2R0, -OC(O)N(R0)2, -S(0)tR°, -S(0)t-OR°, -S(O),N(R0)C(O)R°, -S(0),N(R°)OR°, -NR0SO2N(R0)2, -NR°S02R°, -C(=S)N(R°)2, -C(=NH)-N(R°)2, -(CH2)1.6-C(O)R0, -C(=N-OR°)-N(R°)2, -0-(CH2)o-6-S02N(R0)2, -(CH2)1.6-NHC(O)R0, and -SO2N(R0)2 wherein the two R°s on the same nitrogen are optionally taken together to form a 5-8 membered saturated, partially saturated, or aromatic ring having additional 0-4 heteroatoms selected from oxygen, phosphorus, nitrogen, or sulfur; each R7 is independently selected from the group consisting of halogen, -CF3, -R°, -OR0, -OCF3, -(CH2)ι-6-OR°, -SR°, -SCF3, -(CH^-SR0, aryl optionally substituted by R6, methylenedioxy, ethylenedioxy, -N02, -CN, -(CH^-CN, -N(R°)2, -(CH2)1.6-N(R°)2, -NR0C(O)R°, -NR°(CN), -NR°C(0)N(R°)2, -N(R°)C(S)N(R°)2, -NR0CO2R°, -NR0NR°C(O)R°, -NR°NR°C(0)N(R°)2, -NR0NR°CO2R°, -C(O)C(O)R0, -C(O)CH2C(O)R0, -(CH2)o-6-C02R°, -C(O)R0, -C(O)N(R°)N(R0)2, -C(O)N(R0)2, -C(O)N(R0)OH, -OC(O)R0, -C(O)N(R0)SO2R°, -OC(O)N(R0)2, -S(O),R0, -S(O),-OR0, -S(O),N(R0)C(O)R°, -S(0),N(R°)OR°, -NR0SO2N(R°)2, -NR0SO2R°, -C(=S)N(R°)2, -C(=NH)-N(R°)2, -(CH^-CO^R0, -C(=N-OR°)-N(R°)2, -O-(CH2)0-6-SO2N(R°)2, -(CH2)1.6-NHC(0)R°, and -SO2N(R0)2 wherein the two R°s on the same nitrogen are optionally taken together to form a 5-8 membered saturated, partially saturated, or aromatic ring having additional 0-4 heteroatoms selected from oxygen, phosphorus, nitrogen, or sulfur; each R8 is independently selected from R7, =0, =S, =N(R°), and =N(CN);
R9 is H or oxo; each R° is independently selected from the group consisting of hydrogen, alkyl, alkenyl, alkynyl, cycloalkyl, carbocyclylalkyl, aryl, heteroaryl, aralkyl, heteroaralkyl, heterocyclyl, and heterooyclylalkyl, wherein each member of R° except H is optionally substituted by one or more R*, OR*, N(R*)2, =0, =S, halo, CF3, N02, CN, -C(0)R*, -C02R*, -C(0)-aryl, -C(0)-heteroaryl, -C(0)-aralkyl, -S(0)raryl, -S(0)t-heteroaryl, -NR*S02R*, -NR*C(0)R*, -NR*C(0)N(R*)2, -N(R*)C(S)N(R*)2, -NR*C02R*, -NR*NR*C(0)R*, -NR*NR*C(0)N(R*)2, -NR*NR*C02R*, -C(0)C(0)R*, -C(0)CH2C(0)R*, -C(0)N(R*)N(R*)2, -C(0)N(R*)2, -C(0)NR*S02R*, -OC(0)N(R*)2, -S(0)tR*, -NR*S02N(R*)2, -S02N(R*)2 wherein the two R*s on the same nitrogen are optionally taken together to form a 5-8 membered saturated, partially saturated or aromatic ring having additional 0-4 heteroatoms selected from oxygen, phosphorus, nitrogen or sulfur; and each R* is independently H, alkyl, alkenyl, alkynyl, cycloalkyl, aryl, or heteroaryl.
2. A compound or salt thereof selected from the group consisting of
Figure imgf000090_0001
90
Figure imgf000091_0001
91
Figure imgf000092_0001
Figure imgf000093_0001
Figure imgf000094_0001
Figure imgf000095_0001
Figure imgf000096_0001
Figure imgf000097_0001
Figure imgf000098_0001
Figure imgf000099_0001
99
Figure imgf000100_0001
Figure imgf000101_0001
Figure imgf000102_0001
Figure imgf000103_0001
Figure imgf000104_0001
3. A compound selected from the group consisting of tert-butyl 3-(3,4-dichlorophenyl)-3-[3-(4-oxo-1 -phenyl-1 ,3,8-triazaspiro[4.5]dec-8- yl)propyl]pyrrolidine-1-carboxylate;
8-{3-[3-(3,4-dichlorophenyl)-1 -(2-furoyl)pyrrolidin-3-yl]propyl}-1 -phenyl-1 ,3,8- triazaspiro[4.5]decan-4-one;
8-{3-[3-(3,4-dichlorophenyl)-1-(isoxazol-5-ylcarbonyl)pyrrolidin-3-yl]propyl}-1-phenyl-
1 ,3,8-triazaspiro[4.5]decan-4-one;
8-{3-[3-(3,4-dichlorophenyI)-1-(1 H-pyrrol-2-ylcarbonyl)pyrrolidin-3-yl]propyl}-1-phenyl-
1 ,3,8-triazaspiro[4.5]decan-4-one;
8-{3-[3-(3,4-dichlorophenyl)-1-pentanoylpyrroIidin-3-yl]propyl}-1-[3-
(trifluoromethyl)phenyl]-1 ,3,8-triazaspiro[4.5]decan-4-one;
8-{3-[3-(3,4-dichlorophenyl)-1 -(2-furoyl)pyrroIidin-3-yl]propyl}-1 -[3-
(trifluoromethyl)phenyl]-1 ,3,8-triazaspiro[4.5]decan-4-one; 8-{3-[1-(cyclobutylcarbonyl)-3-(3,4-dichlorophenyl)pyrrolidin-3-yl]propyl}-1-[3-
(trifluoromethyl)phenyl]-1 ,3,8-triazaspiro[4.5]decan-4-one;
8-{3-[3-(3,4-dichlorophenyl)-1 -pentanoylpyrrolidin-3-yl]propyl}-1 -phenyl-1 ,3,8- triazaspiro[4.5]decan-4-one;
8-{3-[1-(cyclopentylcarbonyl)-3-(3,4-dichlorophenyI)pyrrolidin-3-yl]propyl}-1-phenyl-
1 ,3,8-triazaspiro[4.5]decan-4-one;
8-{3-[1-(cyclobutylcarbonyl)-3-(3,4-dichlorophenyl)pyrrolidin-3-yl]propyl}-1-phenyl-
1,3,8-triazaspiro[4.5]decan-4-one;
8-{3-[1-(cyclobutylcarbonyl)-3-(3,4-dichlorophenyl)pyrrolidin-3-yl]propyl}-1-(3- methylphenyl)-1,3,8-triazaspiro[4.5]decan-4-one;
3-acetyl-8-{3-[1-acetyl-3-(3,4-dichlorophenyl)pyrrolidin-3-yl]propyl}-1-(3- methylphenyl)-1 ,3,8-triazaspiro[4.5]decan-4-one;
8-{3-[1 -(1 ,3-benzoxazol-2-yl)-3-(3,4-dichlorophenyl)pyrrolidin-3-yl]propyl}-1 -phenyl-
1 ,3,8-triazaspiro[4.5]decan-4-one;
8-(2-{[3-(3,4-dichlorophenyl)-1 -(2-furoyl)pyrrolidin-3-yl]oxy}ethyl)-1 -phenyl-1 ,3,8- triazaspiro[4.5]decan-4-one;
8-(2-{[1-(cyclopentylcarbonyl)-3-(3,4-dichlorophenyl)pyrrolidin-3-yl]oxy}ethyl)-1- phenyl-1 ,3,8-triazaspiro[4.5]decan-4-one;
8-(2-{[1 -acetyl-3-(3,4-dichiorophenyl)pyrrolidin-3-yl]oxy}ethyl)-1 -phenyl-1 ,3,8- triazaspiro[4.5]decan-4-one;
8-(2-{[3-(3,4-dichlorophenyl)-1-(phenyIsulfonyl)pyrrolidin-3-yl]oxy}ethyl)-1-phenyl-
1 ,3,8-triazaspiro[4.5]decan-4-one;
8-(2-{[3-(3,4-dichlorophenyl)-1-(2-furoyl)pyrrolidin-3-yl]oxy}ethyl)-1-(3- methoxyphenyl)-1,3,8-triazaspiro[4.5]decan-4-one;
8-(2-{[1-(cyclopentylcarbonyl)-3-(3,4-dichlorophenyl)pyrrolidin-3-yl]oxy}ethyl)-1-(3- methoxyphenyl)-1 ,3,8-triazaspiro[4.5]decan-4-one;
8-(2-{[1-acetyl-3-(3,4-dichlorophenyl)pyrrolidin-3-yl]oxy}ethyl)-1-(3-methoxyphenyl)-
1 ,3,8-triazaspiro[4.5]decan-4-one; and
8-(2-{[3-(3,4-dichlorophenyl)-1-(phenylsulfonyl)pyrrolidin-3-yl]oxy}ethyl)-1-(3- methoxyphenyl)-1 ,3,8-triazaspiro[4.5]decan-4-one.
4. The compound of claim 1 wherein the B ring is pyrrolidine.
5. The compound of claim 4 wherein R9 is H.
6. The compound of claim 4 wherein R9 is oxo.
7. The compound of claim 1 wherein R1 is optionally substitued aryl. The compound of claim 7 wherein R1 is phenyl mono- or di- substituted with halogen.
9. The compound of claim 8 wherein R1 is phenyl di-substituted with Cl. 10. The compound of claim 1 wherein -(Y)m-R3 is selected from the group consisting of
Figure imgf000106_0001
1. The compound of claim 1 wherein -(Y)m-R3 is selected from the group consisting of
Figure imgf000107_0001
Figure imgf000108_0001
Figure imgf000109_0001
Figure imgf000110_0001
Figure imgf000110_0002
Figure imgf000111_0001
12. The compound of claim 1 wherein m is 1 , Y is -C(O)-, and R3 is either aryl or heteroaryl, wherein said aryl or heteroaryl is optionally substituted, with an optionally substituted alkyl, or an optionally substituted cycloalkyl.
13. The compound of claim 1 wherein m is 1 , Y is -(C=N-CN)-O-, and R3 is optionally substituted aryl.
14. The compound of claim 1 wherein m is 1 , Y is ~(CH2)-, and R3 is optionally substituted aryl.
15. The compound of claim 1 wherein m is 1 , Y is -C(O)O-, and R3 is optionally substituted alkyl or optionally substituted aryl.
16. The compound of claim 1 wherein m is 0 and R3 is optionally substituted heteroaryl or optionally substituted heterocyclyl.
17. The compound of claim 1 where X is -(CH2)-, -(CH2-CH2)-, or
18. -(CH2-CH2-CH2)-.
19. The compound of claim 17 wherein X is optionally substituted by one or more halogen or oxo.
20. The compound of claim 18 wherein X is disubstituted with halogen.
21. The compound of claim 19 wherein X is disubstituted with fluoro.
22. The compound of claim 20 wherein X is -CF2-.
23. The compound of claim 17 wherein X optionally has 1-3 heteroatoms selected from oxygen, phosphorus, sulfur, or nitrogen.
24. The compound of claim 22 wherein X is -O- or -C(O)-.
5. The compound of claim 1 wherein the A ring, with an asterisk indicating the point of optional substitution, is selected from the group consisting of
Figure imgf000113_0001
Figure imgf000113_0002
6. The compound of claim 24 wherein each R2, with an asterisk indicating the point of substitution from the A ring, independently is selected from:
Figure imgf000114_0001
Figure imgf000115_0001
Figure imgf000116_0001
7. The compound of claim 1 wherein the A ring, with two geminal R2s, is selected from:
Figure imgf000117_0001
28. The compound of claim 1 wherein the A ring is tropane or piperidine, either optionally substituted with one or more R2.
8. The compound of claim 27 wherein the A ring ring in combination with R is.
Figure imgf000118_0001
Figure imgf000119_0001
Figure imgf000120_0001
Figure imgf000121_0001
29. The compound of claim 1 wherein the A ring contains at least one additional nitrogen atom and said A ring optionally is N-substituted.
30. The compound of claim 29 wherein the A ring is N-substituted with -(CH2)a-(Vb-R+).
31. A method of treatment of a viral infection in a mammal comprising administering to said mammal an antiviral effective amount of a compound according to claims 1-30.
32. A method according to claim 31 wherein the viral infection is an HIV infection.
33. A method of treatment of a bacterial infection in a mammal comprising administering to said mammal an effective amount of a compound according to claims 1-30.
34. A method according to claim 33 wherein the bacterium is Yersinia pestis.
35. A method of treatment of 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, infectious disorders including bubonic and pneumonic plague, human papilloma virus infection, prostate cancer, wound healing, amyotrophic lateral sclerosis and immune mediated disorders in a mammal comprising administering to said mammal a pharmceutically effective amount of a compound according to claims 1-30.
36. A compound according to claims 1-30 for use in medical therapy.
37. Use of a compound according to claims 1-30 in the manufacture of a medicament for the treatment of a viral infection.
39. The use according to claim 37 wherein the viral infection is a HIV infection.
39. Use of a compound according to claims 1-30 in the manufacture of a medicament for the treatment of a bacterial infection.
40. The use according to claim 39 wherein the bacterium is Yersinia pestis.
41. Use of a compound according to claims 1-30 in the manufacture of a medicament for the treatment of 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, infectious disorders including bubonic and pneumonic plague, human papilloma virus infection, prostate cancer, wound healing, amyotrophic lateral sclerosis and immune mediated disorders.
42. A pharmaceutical composition comprising a pharmaceutically effective amount of a compound according to claims 1-30 together with a pharmaceutically acceptable carrier.
43. The pharmaceutical composition according to claim 42 in the form of a tablet or capsule.
44. The pharmaceutical composition according to claim 42 in the form of a liquid.
45. A method of treatment of a viral infection in a mammal comprising administering to said mammal a composition comprising a compound according to claims 1-30 and another therapeutic agent.
46. A method according to claim 45, wherein said composition comprises another therapeutic agent selected from the group consisting of (1 -alpha, 2-beta, 3-alpha)-9- [2,3-bis(hydroxymethyl)cyclobutyl]guanine [(-)BHCG, SQ-34514, lobucavir], 9- [(2R,3R,4S)-3,4-bis(hydroxymethyl)-2-oxetanosyl]adenine (oxetanocin-G), 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), [[(1R)-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), ribonucleotide reductase inhibitors, 2-acetylpyridine 5-[(2- chloroanilino)thiocarbonyl) thiocarbonohydrazone and hydroxyurea, 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), (-)-beta-D-2,6-diaminopurine dioxolane (DAPD), 3'-azido-2\3'-dideoxythymidine-5'-H-phosphophonate (phosphonovir), 2'-deoxy-5-iodo-uridine (idoxuridine), (-)-cjs-1-(2-hydroxymethyl)-1,3- oxathiolane 5-yl)-cytosine (lamivudine), cis-1 -(2-(hydroxymethyl)-1 ,3-oxathiolan-5- yl)-5-fluorocytosine (FTC), 3'-deoxy-3'-fluorothymidine, 5-chloro-2',3'-dideoxy-3'- fluorouridine, (-)-cis-4-[2-amino-6-(cyclopropylamino)-9H-purin-9-yl]-2-cyclopentene- 1-methanol (abacavir), 9-[4-hydroxy-2-(hydroxymethyl)but-1-yl]-guanine (H2G), ABT-606 (2HM-H2G) ribavirin, protease inhibitors, indinavir, ritonavir, nelfinavir, amprenavir, saquinavir, fosamprenavir, (R)-N-tert-butyl-3-[(2S,3S)-2-hydroxy-3-N- [(R)-2-N-(isoquinolin-5-yloxyacetyl)amino-3-methylthiopropanoyl]amino-4- phenylbutanoyl]-5,5- dimethyl-1,3-thiazolidine-4-carboxamide (KNI-272), 4R- (4alpha,5alpha,6beta)]-1,3-bis[(3-aminophenyl)methyl]hexahydro-5,6-dihydroxy-4,7- bis(phenylmethyl)-2H-1,3-diazepin-2-one dimethanesulfonate (mozenavir), 3-[1-[3-[2- (5-trifluoromethylpyridinyl)-sulfonylamino]phenyl]propyl]-4- hydroxy-6alpha- phenethyl-6beta-propyl-5,6-dihydro-2-pyranone (tipranavir), N'-[2(S)-Hydroxy-3(S)- [N-(methoxycarbonyl)-l-tert-leucylamino]-4- phenylbutyl-N alpha-(methoxycarbonyl)- N'-[4-(2-pyridyl)benzyl]-L- tert-leucylhydrazide (BMS-232632), 3-(2(S)-Hydroxy-3(S)- (3-hydroxy-2-methylbenzamido)-4-phenylbutanoyl)-5,5-dimethyl-N-(2- methylbenzyl)thiazolidine-4(R)-carboxamide (AG-1776), N-(2(R)-hydroxy-1 (S)- indanyl)-2(R)-phenyl-methyl-4(S)-hydroxy-5-(1-(1-(4-benzo[b]furanylmethyl)-2(S)-N'- (tert-butylcarboxamido)piperazinyl)pentanamide (MK-944A), interferons, α-interferon, renal excretion inhibitors, probenecid, nucleoside transport inhibitors, dipyridamole, pentoxifylline, N-acetylcysteine (NAC), Procysteine, α -trichosanthin, phosphonoformic acid, immunomodulators, interleukin II, 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- dichloro-benzeneacetamide (loviride), 1 -[3-(isopropylamino)-2-pyridyl]-4-[5- (methanesulfonamido)-1H-indol-2-ylcarbonyl]piperazine monomethanesulfonate (delavirdine), (10R, 11S, 12S)-12-hydroxy-6, 6, 10, 11-tetramethyl-4-propyl-11,12- dihydro-2H, 6H, 10H-benzo(1, 2-b:3, 4-b':5, 6-b")tripyran-2-one '((+) calanolide A), (4S)-6-Chloro-4-[1 E)-cyclopropylethenyl)-3,4- dihydro-4-(trifluoromethyl)-2(1 H)- quinazolinone (DPC-083), (S)-6-chloro-4-(cyclopropylethynyl)-1 ,4-dihydro-4- (trifluoromethyl)-2H-3,1-benzoxazin-2-one (efavirenz, DMP 266), 1-(ethoxymethyl)-5- (1-methylethyl)-6-(phenylmethyl)-2,4(1H,3H)-pyrimidinedione (MKC-442), and 5- (3,5-dichlorophenyl)thio-4-isopropyl-1-(4-pyridyl)methyl-1H-imidazol-2-ylmethyl carbamate (capravirine), glycoprotein 120 antagonists, PRO-2000, PRO-542, 1,4- bis[3-[(2, 4- dichlorophenyl)carbonylamino]-2-oxo-5,8-disodiumsulfanyl]naphthalyl-2, 5-dimethoxyphenyl-1 , 4-dihydrazone (FP-21399), cytokine antagonists, reticulose (Product-R), 1,1'-azobis-formamide (ADA), 1,11-(1,4-phenylenebis(methylene))bis- 1,4,8,11-tetraazacyclotetradecane octahydrochloride (AMD-3100), integrase inhibitors, and fusion inhibitors.
47. A method of treatment of a viral infection in a mammal comprising administering to said mammal a composition comprising a compound according to claims 1-30 and ritonavir.
PCT/US2003/039618 2002-12-13 2003-12-12 Pyrrolidine and azetidine compounds as ccr5 antagonists WO2004055016A1 (en)

Priority Applications (5)

Application Number Priority Date Filing Date Title
US10/538,134 US7271172B2 (en) 2002-12-13 2003-12-12 Pyrrolidine and azetidine compounds as CCR5 antagonists
EP03813415A EP1569933B1 (en) 2002-12-13 2003-12-12 Pyrrolidine and azetidine compounds as ccr5 antagonists
JP2004560830A JP2006511552A (en) 2002-12-13 2003-12-12 Pyrrolidine and azetidine compounds as CCR5 antagonists
AU2003296992A AU2003296992A1 (en) 2002-12-13 2003-12-12 Pyrrolidine and azetidine compounds as ccr5 antagonists
DE60322423T DE60322423D1 (en) 2002-12-13 2003-12-12 PYRROLIDIN AND AZETIDIN COMPOUNDS ALSCCR5 ANTAGONISTS

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US43337202P 2002-12-13 2002-12-13
US60/433,372 2002-12-13

Publications (1)

Publication Number Publication Date
WO2004055016A1 true WO2004055016A1 (en) 2004-07-01

Family

ID=32595168

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2003/039618 WO2004055016A1 (en) 2002-12-13 2003-12-12 Pyrrolidine and azetidine compounds as ccr5 antagonists

Country Status (8)

Country Link
US (1) US7271172B2 (en)
EP (1) EP1569933B1 (en)
JP (1) JP2006511552A (en)
AT (1) ATE402176T1 (en)
AU (1) AU2003296992A1 (en)
DE (1) DE60322423D1 (en)
ES (1) ES2309400T3 (en)
WO (1) WO2004055016A1 (en)

Cited By (24)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP1804919A2 (en) * 2004-10-22 2007-07-11 Merck & Co., Inc. Cgrp receptor antagonists
JP2008509185A (en) * 2004-08-10 2008-03-27 ノバルティス アクチエンゲゼルシャフト Azetidine derivatives as CCR-3 antagonists
WO2008034731A1 (en) * 2006-09-18 2008-03-27 F. Hoffmann-La Roche Ag Octahydropyrrolo [3, 4-c] pyrrole derivatives an their use as antiviral agents
JP2009502786A (en) * 2005-07-18 2009-01-29 メルク エンド カムパニー インコーポレーテッド Spiropiperidine β-secretase inhibitors for treating Alzheimer's disease
WO2009145286A1 (en) 2008-05-30 2009-12-03 武田薬品工業株式会社 Heterocyclic compound
WO2010018131A1 (en) 2008-08-11 2010-02-18 Smithkline Beecham Corporation Purine derivatives for use in the treatment of allergic, inflammatory and infectious diseases
WO2011098451A1 (en) 2010-02-10 2011-08-18 Glaxosmithkline Llc Purine derivatives and their pharmaceutical uses
WO2011098452A1 (en) 2010-02-10 2011-08-18 Glaxosmithkline Llc 6-amino-2-{ [ (1s)-1-methylbutyl] oxy}-9-[5-(1-piperidinyl)-7,9-dihydro-8h-purin-8-one maleate
WO2013090664A1 (en) 2011-12-16 2013-06-20 Glaxosmithkline Llc Derivatives of betulin
WO2015124591A1 (en) 2014-02-20 2015-08-27 Glaxosmithkline Intellectual Property (No.2) Limited Pyrrolo[3,2] pyrimidine derivatives as inducers of human interferon
EP3000813A1 (en) 2008-08-11 2016-03-30 GlaxoSmithKline LLC Purine derivatives for use in the treatment of allergic, inflammatory and infectious diseases
WO2016075661A1 (en) 2014-11-13 2016-05-19 Glaxosmithkline Biologicals Sa Adenine derivatives which are useful in the treatment of allergic diseases or other inflammatory conditions
WO2016205032A1 (en) * 2015-06-16 2016-12-22 Eli Lilly And Company 2-oxo-1,3,8-triazaspiro[4.5]decan-3-yl] carboxylic acid derivatives
WO2017093933A1 (en) 2015-12-03 2017-06-08 Glaxosmithkline Intellectual Property Development Limited Cyclic purine dinucleotides as modulators of sting
WO2017175156A1 (en) 2016-04-07 2017-10-12 Glaxosmithkline Intellectual Property Development Limited Heterocyclic amides useful as protein modulators
WO2017175147A1 (en) 2016-04-07 2017-10-12 Glaxosmithkline Intellectual Property Development Limited Heterocyclic amides useful as protein modulators
EP3246030A1 (en) 2008-08-11 2017-11-22 GlaxoSmithKline LLC Novel adenine derivatives
WO2019069270A1 (en) 2017-10-05 2019-04-11 Glaxosmithkline Intellectual Property Development Limited Modulators of stimulator of interferon genes (sting)
WO2019069269A1 (en) 2017-10-05 2019-04-11 Glaxosmithkline Intellectual Property Development Limited Modulators of stimulator of interferon genes (sting) useful in treating hiv
WO2019219820A1 (en) 2018-05-16 2019-11-21 Ctxt Pty Limited Substituted condensed thiophenes as modulators of sting
WO2020232378A1 (en) 2019-05-16 2020-11-19 Silicon Swat, Inc. Benzo[b][1,8]naphthyridine acetic acid derivatives and methods of use
WO2020232375A1 (en) 2019-05-16 2020-11-19 Silicon Swat, Inc. Oxoacridinyl acetic acid derivatives and methods of use
WO2021009365A1 (en) 2019-07-18 2021-01-21 Ctxt Pty Limited Benzothiophene, thienopyridine and thienopyrimidine derivatives for the modulation of sting
WO2021009362A1 (en) 2019-07-18 2021-01-21 Ctxt Pty Limited Benzothiophene, thienopyridine and thienopyrimidine derivatives for the modulation of sting

Families Citing this family (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7589207B2 (en) * 2002-12-13 2009-09-15 Smithkline Beecham Corporation Cyclohexyl compounds as CCR5 antagonists
WO2008030853A2 (en) * 2006-09-06 2008-03-13 Incyte Corporation Combination therapy for human immunodeficiency virus infection
AU2007338953A1 (en) * 2006-12-22 2008-07-03 Albert Einstein College Of Medicine Of Yeshiva University Azetidine analogues of nucleosidase and phosphorylase inhibitors
CA3121202A1 (en) 2018-11-30 2020-06-04 Nuvation Bio Inc. Pyrrole and pyrazole compounds and methods of use thereof
US11116737B1 (en) 2020-04-10 2021-09-14 University Of Georgia Research Foundation, Inc. Methods of using probenecid for treatment of coronavirus infections

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1994026735A1 (en) * 1993-05-06 1994-11-24 Merrell Dow Pharmaceuticals Inc. Substituted pyrrolidin-3-yl-alkyl-piperidines useful as tachykinin antagonists
WO2002074770A1 (en) * 2001-03-19 2002-09-26 Ono Pharmaceutical Co., Ltd. Triazaspiro[5.5]undecane derivatives and drugs containing the same as the active ingredient

Family Cites Families (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5635510A (en) 1993-05-06 1997-06-03 Merrell Pharmaceuticals Inc. Substituted pyrrolidin-3-yl-alkyl-piperidines
WO1995028389A1 (en) * 1994-04-15 1995-10-26 Yamanouchi Pharmaceutical Co., Ltd. Spiro compound and medicinal composition thereof
US5861417A (en) 1996-12-19 1999-01-19 Hoechst Marion Roussel, Inc. Heterocyclic substituted pyrrolidine amide derivatives

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1994026735A1 (en) * 1993-05-06 1994-11-24 Merrell Dow Pharmaceuticals Inc. Substituted pyrrolidin-3-yl-alkyl-piperidines useful as tachykinin antagonists
WO2002074770A1 (en) * 2001-03-19 2002-09-26 Ono Pharmaceutical Co., Ltd. Triazaspiro[5.5]undecane derivatives and drugs containing the same as the active ingredient
EP1378510A1 (en) * 2001-03-19 2004-01-07 Ono Pharmaceutical Co., Ltd. Triazaspiro 5.5]undecane derivatives and drugs containing the same as the active ingredient

Cited By (33)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP2008509185A (en) * 2004-08-10 2008-03-27 ノバルティス アクチエンゲゼルシャフト Azetidine derivatives as CCR-3 antagonists
AU2005299852B2 (en) * 2004-10-22 2011-08-04 Merck Sharp & Dohme Corp. CGRP receptor antagonists
JP2008517916A (en) * 2004-10-22 2008-05-29 メルク エンド カムパニー インコーポレーテッド CGRP receptor antagonist
EP1804919A4 (en) * 2004-10-22 2009-11-25 Merck & Co Inc Cgrp receptor antagonists
EP1804919A2 (en) * 2004-10-22 2007-07-11 Merck & Co., Inc. Cgrp receptor antagonists
US7745427B2 (en) 2004-10-22 2010-06-29 Merck Sharp & Dohme Corp. CGRP receptor antagonists
JP2009502786A (en) * 2005-07-18 2009-01-29 メルク エンド カムパニー インコーポレーテッド Spiropiperidine β-secretase inhibitors for treating Alzheimer's disease
WO2008034731A1 (en) * 2006-09-18 2008-03-27 F. Hoffmann-La Roche Ag Octahydropyrrolo [3, 4-c] pyrrole derivatives an their use as antiviral agents
US7625905B2 (en) 2006-09-18 2009-12-01 Roche Palo Alto Llc Octahydro-pyrrolo[3,4-c]pyrrole CCR5 receptor antagonists
JP2010503714A (en) * 2006-09-18 2010-02-04 エフ.ホフマン−ラ ロシュ アーゲー Octahydropyrrolo [3,4-C] pyrrole derivatives, their use as antiviral agents
WO2009145286A1 (en) 2008-05-30 2009-12-03 武田薬品工業株式会社 Heterocyclic compound
EP3000813A1 (en) 2008-08-11 2016-03-30 GlaxoSmithKline LLC Purine derivatives for use in the treatment of allergic, inflammatory and infectious diseases
EP3246030A1 (en) 2008-08-11 2017-11-22 GlaxoSmithKline LLC Novel adenine derivatives
WO2010018131A1 (en) 2008-08-11 2010-02-18 Smithkline Beecham Corporation Purine derivatives for use in the treatment of allergic, inflammatory and infectious diseases
WO2011098452A1 (en) 2010-02-10 2011-08-18 Glaxosmithkline Llc 6-amino-2-{ [ (1s)-1-methylbutyl] oxy}-9-[5-(1-piperidinyl)-7,9-dihydro-8h-purin-8-one maleate
WO2011098451A1 (en) 2010-02-10 2011-08-18 Glaxosmithkline Llc Purine derivatives and their pharmaceutical uses
WO2013090664A1 (en) 2011-12-16 2013-06-20 Glaxosmithkline Llc Derivatives of betulin
WO2015124591A1 (en) 2014-02-20 2015-08-27 Glaxosmithkline Intellectual Property (No.2) Limited Pyrrolo[3,2] pyrimidine derivatives as inducers of human interferon
WO2016075661A1 (en) 2014-11-13 2016-05-19 Glaxosmithkline Biologicals Sa Adenine derivatives which are useful in the treatment of allergic diseases or other inflammatory conditions
WO2016205032A1 (en) * 2015-06-16 2016-12-22 Eli Lilly And Company 2-oxo-1,3,8-triazaspiro[4.5]decan-3-yl] carboxylic acid derivatives
WO2017093933A1 (en) 2015-12-03 2017-06-08 Glaxosmithkline Intellectual Property Development Limited Cyclic purine dinucleotides as modulators of sting
EP3366691A1 (en) 2015-12-03 2018-08-29 GlaxoSmithKline Intellectual Property Development Limited Cyclic purine dinucleotides as modulators of sting
WO2017175156A1 (en) 2016-04-07 2017-10-12 Glaxosmithkline Intellectual Property Development Limited Heterocyclic amides useful as protein modulators
WO2017175147A1 (en) 2016-04-07 2017-10-12 Glaxosmithkline Intellectual Property Development Limited Heterocyclic amides useful as protein modulators
EP4032885A1 (en) 2016-04-07 2022-07-27 GlaxoSmithKline Intellectual Property Development Limited Heterocyclic amides useful as protein modulators
WO2019069269A1 (en) 2017-10-05 2019-04-11 Glaxosmithkline Intellectual Property Development Limited Modulators of stimulator of interferon genes (sting) useful in treating hiv
WO2019069270A1 (en) 2017-10-05 2019-04-11 Glaxosmithkline Intellectual Property Development Limited Modulators of stimulator of interferon genes (sting)
WO2019219820A1 (en) 2018-05-16 2019-11-21 Ctxt Pty Limited Substituted condensed thiophenes as modulators of sting
US11613525B2 (en) 2018-05-16 2023-03-28 Ctxt Pty Limited Substituted condensed thiophenes as modulators of sting
WO2020232378A1 (en) 2019-05-16 2020-11-19 Silicon Swat, Inc. Benzo[b][1,8]naphthyridine acetic acid derivatives and methods of use
WO2020232375A1 (en) 2019-05-16 2020-11-19 Silicon Swat, Inc. Oxoacridinyl acetic acid derivatives and methods of use
WO2021009365A1 (en) 2019-07-18 2021-01-21 Ctxt Pty Limited Benzothiophene, thienopyridine and thienopyrimidine derivatives for the modulation of sting
WO2021009362A1 (en) 2019-07-18 2021-01-21 Ctxt Pty Limited Benzothiophene, thienopyridine and thienopyrimidine derivatives for the modulation of sting

Also Published As

Publication number Publication date
ATE402176T1 (en) 2008-08-15
US20060058284A1 (en) 2006-03-16
ES2309400T3 (en) 2008-12-16
AU2003296992A1 (en) 2004-07-09
DE60322423D1 (en) 2008-09-04
EP1569933A1 (en) 2005-09-07
JP2006511552A (en) 2006-04-06
US7271172B2 (en) 2007-09-18
EP1569933B1 (en) 2008-07-23

Similar Documents

Publication Publication Date Title
US7271172B2 (en) Pyrrolidine and azetidine compounds as CCR5 antagonists
US7569579B2 (en) Cyclopropyl compounds as ccr5 antagonists
US7589204B2 (en) Indane compounds as CCR5 antagonists
US7589207B2 (en) Cyclohexyl compounds as CCR5 antagonists
US8691991B2 (en) 2-oxonaphthyridine-3-carboxamides HIV integrase inhibitors
US20090053172A1 (en) Heterocyclic compounds as ccr5 antagonists
US7645771B2 (en) CCR5 antagonists as therapeutic agents
US20080214527A1 (en) Hiv Integrase Inhibitors
US20110003794A1 (en) Naphthyridine Integrase Inhibitors

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A1

Designated state(s): AE AG AL AM AT AU AZ BA BB BG BR BW BY BZ CA CH CN CO CR CU CZ DE DK DM DZ EC EE EG ES FI GB GD GE GH GM HR HU ID IL IN IS JP KE KG KP KR KZ LC LK LR LS LT LU LV MA MD MG MK MN MW MX MZ NI NO NZ OM PG PH PL PT RO RU SC SD SE SG SK SL SY TJ TM TN TR TT TZ UA UG US UZ VC VN YU ZA ZM ZW

AL Designated countries for regional patents

Kind code of ref document: A1

Designated state(s): BW GH GM KE LS MW MZ SD SL SZ TZ UG ZM ZW AM AZ BY KG KZ MD RU TJ TM AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HU IE IT LU MC NL PT RO SE SI SK TR BF BJ CF CG CI CM GA GN GQ GW ML MR NE SN TD TG

DFPE Request for preliminary examination filed prior to expiration of 19th month from priority date (pct application filed before 20040101)
121 Ep: the epo has been informed by wipo that ep was designated in this application
ENP Entry into the national phase

Ref document number: 2006058284

Country of ref document: US

Kind code of ref document: A1

WWE Wipo information: entry into national phase

Ref document number: 2004560830

Country of ref document: JP

Ref document number: 10538134

Country of ref document: US

WWE Wipo information: entry into national phase

Ref document number: 2003813415

Country of ref document: EP

WWP Wipo information: published in national office

Ref document number: 2003813415

Country of ref document: EP

WWP Wipo information: published in national office

Ref document number: 10538134

Country of ref document: US