EP1866276A1 - Alpha ketoamide compounds as cysteine protease inhibitors - Google Patents

Alpha ketoamide compounds as cysteine protease inhibitors

Info

Publication number
EP1866276A1
EP1866276A1 EP06739282A EP06739282A EP1866276A1 EP 1866276 A1 EP1866276 A1 EP 1866276A1 EP 06739282 A EP06739282 A EP 06739282A EP 06739282 A EP06739282 A EP 06739282A EP 1866276 A1 EP1866276 A1 EP 1866276A1
Authority
EP
European Patent Office
Prior art keywords
ethyl
amino
methylamino
alkyl
ylco
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Withdrawn
Application number
EP06739282A
Other languages
German (de)
French (fr)
Inventor
John O. Link
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
CELERA GENOMICS
Original Assignee
CELERA GENOMICS
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 CELERA GENOMICS filed Critical CELERA GENOMICS
Publication of EP1866276A1 publication Critical patent/EP1866276A1/en
Withdrawn legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C237/00Carboxylic acid amides, the carbon skeleton of the acid part being further substituted by amino groups
    • C07C237/02Carboxylic acid amides, the carbon skeleton of the acid part being further substituted by amino groups having the carbon atoms of the carboxamide groups bound to acyclic carbon atoms of the carbon skeleton
    • C07C237/22Carboxylic acid amides, the carbon skeleton of the acid part being further substituted by amino groups having the carbon atoms of the carboxamide groups bound to acyclic carbon atoms of the carbon skeleton having nitrogen atoms of amino groups bound to the carbon skeleton of the acid part, further acylated
    • 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
    • A61P17/00Drugs for dermatological disorders
    • A61P17/06Antipsoriatics
    • 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
    • A61P25/04Centrally acting analgesics, e.g. opioids
    • 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
    • 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
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • 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

  • the present invention is directed to compounds that are inhibitors of cysteine proteases, in particular, cathepsins B, K, L, F, and S and are therefore useful in treating diseases mediated by these proteases.
  • the present invention is also directed to pharmaceutical compositions comprising these compounds and processes for preparing them.
  • Cysteine proteases represent a class of peptidases characterized by the presence of a cysteine residue in the catalytic site of the enzyme. Cysteine proteases are associated with the normal degradation and processing of proteins. The aberrant activity of cysteine proteases, e.g., as a result of increased expression or enhanced activation, however, may have pathological consequences. In this regard, certain cysteine proteases are associated with a number of disease states, including arthritis, muscular dystrophy, inflammation, tumor invasion, glomerulonephritis, malaria, periodontal disease, metachromatic leukodystrophy and others.
  • cathepsin B levels and redistribution of the enzyme are found in tumors; thus, suggesting a role for the enzyme in tumor invasion and metastasis.
  • aberrant cathepsin B activity is implicated in such disease states as rheumatoid arthritis, osteoarthritis, Pneumocystis carinii, acute pancreatitis, inflammatory airway disease and bone and joint disorders.
  • cathepsin K in osteoclasts and osteoclast-related multinucleated cells and its high collagenolytic activity suggest that the enzyme is involved in osteoclast-mediated bone resorption and, hence, in bone abnormalities such as occurs in osteoporosis.
  • cathepsin K expression in the lung and its elastinolytic activity suggest that the enzyme plays a role in pulmonary disorders as well.
  • Cathepsin L is implicated in normal lysosomal proteolysis as well as in several disease states, including, but not limited to, metastasis of melanomas.
  • Cathepsin S is implicated in Alzheimer's disease and certain autoimmune disorders, including, but not limited to juvenile onset diabetes, multiple sclerosis, pemphigus vulgaris, Graves' disease, myasthenia gravis, systemic lupus erythemotasus, rheumatoid arthritis, neuropathic pain, and Hashimoto's thyroiditis.
  • cathepsin S is implicated in: allergic disorders, including, but not limited to asthma; and allogeneic immune reponses, including, but not limited to, rejection of organ transplants or tissue grafts.
  • this invention is directed to a compound of Formula (I):
  • R 1 is hydrogen or alkyl
  • R 2 is cycloalkyl, cycloalkylalkyl, aralkyl, heteroaryl, or heteroaralkyl optionally substituted with one or two substitutents independently selected from alkyl, alkoxy, or halo;
  • R 3 is hydrogen, alkyl or alkoxyalkyl;
  • R 4 is alkyl; or R 3 and R 4 together with the carbon atom to which they are attached form cycloalkylene optionally substituted with one to four fluoro or heterocycloalkylene optionally substituted with alkyl, alkoxyalkyl, hydroxyalkyl, acyl, cycloalkyl, cycloalkylalkyl, or haloalkyl;
  • R 5 is -alkylene-SO 2 NR ⁇ R 12 where R 11 is hydrogen, alkyl, haloalkyl, alkoxyalkyl, hydroxyalkyl, aminoalkyl, carboxyalkyl, alkoxycarbonylalkyl,
  • R 7 is hydrogen, alkyl, or haloalkyl
  • R 8 is hydrogen, alkyl, haloalkyl, cycloalkyl, aryl, heteroaryl, heterocycloalkyl attached via a carbon atom wherein the aromatic or alicyclic ring in R is optionally substituted with one, two, or three R e independently selected from alkyl, halo, haloalkyl, hydroxy, alkoxy, haloalkoxy, alkoxycarbonyl, carboxy, cyano, alkylcarbonyl, alkylsulfonyl, alkylsulfonylamino, alkylaminocarbonyl, dialkylaminocarbonyl, or aminosulfonyl; or a pharmaceutically acceptable salts thereof.
  • this invention is directed to a pharmaceutical composition comprising a compound of Formula (I) or a pharmaceutically acceptable salt thereof in admixture with one or more suitable excipients.
  • this invention is directed to a method for treating a disease in an animal mediated by cysteine proteases, in particular cathepsin S, which method comprises administering to the animal a pharmaceutical composition comprising a therapeutically effective amount of a compound of Formula (I) or a pharmaceutically acceptable salt thereof in admixture with one or more suitable excipients.
  • this invention is directed to processes for preparing compounds of Formula (I).
  • this invention is directed to a method of treating a patient undergoing a therapy wherein the therapy causes an immune response, preferably a deleterious immune response, in the patient comprising administering to the patient a compound of Formula (I) or a pharmaceutically acceptable salt thereof.
  • the immune response is mediated by MHC class II molecules.
  • the compound of this invention can be administered prior to, simultaneously, or after the therapy.
  • the therapy involves treatment with a biologic.
  • the therapy involves treatment with a small molecule.
  • the biologic is a protein, preferably an antibody, more preferably a monoclonal antibody. More preferrably, the biologic is Remicade ® , Refacto ® , Referon-A ® , Factor VIII, Factor VII, Betaseron ® , Epogen ® , Enbrel ® , Interferon beta, Botox ® , Fabrazyme ® , Elspar ® , Cerezyme ® , Myobloc ® , Aldurazyme ® , Verluma ® , Interferon alpha, Humira ® , Aranesp ® , Zevalin ® or OKT3.
  • the treatment involves use of heparin, low molecular weight heparin, procainamide or hydralazine.
  • this invention is directed to a method of treating immune response in an animal that is caused by administration of a biologic to the animal which method comprises administering to the animal in need of such treatment a therapeutically effective amount of a compound of Formula (I) or a pharmaceutically acceptable salt thereof.
  • this invention is directed to a method of conducting a clinical trial for a biologic comprising administering to an individual participating in the clinical trial a compound of Formula (I) or a pharmaceutically acceptable salt thereof with the biologic.
  • this invention is directed to a method of prophylactically treating a patient undergoing treatment with a biologic with a compound of Formula (I) or a pharmaceutically acceptable salt thereof to treat the immune response caused by the biologic in the patient.
  • this invention is directed to a method of determing the loss in the efficacy of a biologic in an animal due to the immune response caused by the biologic comprising administering the biologic to the animal in the presence and absence of a compound of Formula (I) or a pharmaceutically acceptable salt thereof.
  • this invention is directed to a method of improving efficacy of a biologic in an animal comprising administering the biologic to the animal with a compound of Formula (I) or a pharmaceutically acceptable salt thereof.
  • this invention is directed to the use of a compound of Formula (I) or a pharmaceutically acceptable salt thereof for the manufacture of a medicament.
  • the medicament is for use in the treatment of a disease mediated by Cathepsin S.
  • this invention is directed to the use of a compound of Formula (I) or a pharmaceutically acceptable salt thereof for the manufacture of a medicament for combination therapy with a biologic, wherein the compound of this invention treats the immune response caused by the biologic.
  • the compound(s) of the invention is administered prior to the administration of the biological agent.
  • the compound(s) of the invention is administered concomitantly with the biological agent.
  • the compound(s) of the invention is administered after the administration of the biological agent.
  • Alicyclic means a moiety characterized by arrangement of the carbon atoms in closed non-aromatic ring structures e.g., cycloalkyl and heterocycloalkyl rings as defined herein.
  • Alkyl represented by itself means a straight or branched, saturated aliphatic radical containing one to eight carbon atoms, unless otherwise indicated e.g., alkyl includes methyl, ethyl, propyl, isopropyl, butyl, sec-butyl, isobutyl, tert-butyl, and the like.
  • Alkylene unless indicated otherwise, means a straight or branched, saturated aliphatic, divalent radical having the number of one to six carbon atoms, e.g., methylene (-CH 2 -), ethylene (-CH 2 CH 2 -), trimethylene (-CH 2 CH 2 CH 2 -), tetramethylene (-CH 2 CH 2 CH 2 CH 2 -) 2-methyltetramethylene (-CH 2 CH(CH 3 )CH 2 CH 2 -), pentamethylene (-CH 2 CH 2 CH 2 CH 2 CH 2 -), and the like.
  • Alkylsulfonyl means -SO 2 R radical where R is alkyl as defined herein e.g., methylsulfonyl, ethylsulfonyl, and the like.
  • Alkylsulfonylamino refers to a -NHSO 2 R radical where R is an alkyl group as defined above e.g., methylsulfonylamino, ethylsulfonylamino, and the like.
  • Alkoxy refers to a -OR radical where R is an alkyl group as defined above e.g., methoxy, ethoxy, and the like.
  • Alkoxyalkyl means a linear monovalent hydrocarbon radical of one to six carbon atoms or a branched monovalent hydrocarbon radical of three to six carbons substituted with at least one alkoxy group, preferably one or two alkoxy groups, as defined above, e.g., 2- methoxy-ethyl, 1-, 2-, or 3-methoxypropyl, 2-ethoxyethyl, and the like.
  • Alkoxycarbonyl refers to a -C(O)OR radical where R is an alkyl group as defined above e.g., methoxycarbonyl, ethoxycarbonyl, and the like.
  • Alkoxycarbonylalkyl means a linear monovalent hydrocarbon radical of one to six carbon atoms or a branched monovalent hydrocarbon radical of three to six carbons substituted with at least one, preferably one or two, alkoxycarbonyl group(s) as defined herein e.g., methoxycarboxymethyl, ethoxycarbonylmethyl, methoxycarbonylethyl, and the like.
  • Aminoalkyl means a linear monovalent hydrocarbon radical of one to six carbon atoms or a branched monovalent hydrocarbon radical of three to six carbons substituted with at least one, preferably one or two, -NRR' where R is hydrogen, alkyl, acyl, hydroxyalkyl, alkoxyalkyl, carboxyalkyl, alkoxycarbonylalkyl, aryl, aralkyl, heteroaryl, heteroaralkyl or heterocycloalkylalkyl and R is hydrogen, alkyl, haloalkyl, acyl, hydroxyalkyl, alkoxyalkyl, carboxyalkyl alkoxycarbonylalkyl, aryl, aralkyl, heteroaryl, heteroaralkyl, heterocycloalkyl, heterocycloalkylalkyl, cycloalkyl, cycloalkylalkyl, aminocarbonyl, aminosulfonyl, -C(O)
  • Acyl refers to a— COR radical where R is hydrogen, alkyl, cycloalkyl, cycloalkylalkyl, haloalkyl, aryl, aralkyl, heteroaryl, heteroaralkyl, or heterocycloalkyl as defined herein, e.g., formyl, acetyl, trifluoroacetyl, benzoyl, piperazin-1-ylcarbonyl, and the like.
  • R is alkyl it is referred to in this application as alkylcarbonyl.
  • Acylalkyl means a linear monovalent hydrocarbon radical of one to six carbon atoms or a branched monovalent hydrocarbon radical of three to six carbons substituted with at least one, preferably one or two, acyl group(s) as defined herein e.g., methylcarbonylmethyl, ben ⁇ oylethyl, piperidin-1-ylcarbonylmethyl or ethyl, and the like.
  • Aminocarbonyl means -CONRR' radical where R and R' are independently selected from hydrogen, alkyl, aryl, aralkyl, heteroaryl, heteroaralkyl, or heterocycloalkylalkyl or R and R' together with the nitrogen atom to which they are attached form heterocycloamino as defined herein.
  • R group is H or alkyl and R' is alkyl
  • such groups may be referred to in this Application as alkylaminocarbonyl and dialkylaminocarbonyl respectively and are subset of aminocarbonyl group e.g., methylaminocarbonyl or dimethylaminocarbonyl.
  • Aminosulfonyl means -SO 2 NRR' radical where R and R' are independently selected from hydrogen, alkyl, aryl, aralkyl, heteroaryl, heteroaralkyl, or heterocycloalkylalkyl or R and R' together with the nitrogen atom to which they are attached form heterocycloamino as defined herein.
  • Animal includes humans, non-human mammals (e.g., dogs, cats, rabbits, cattle, horses, sheep, goats, swine, deer, and the like) and non-mammals (e.g., birds, and the like).
  • Aromatic refers to a moiety wherein the constituent atoms make up an unsaturated ring system, all atoms in the ring system are sp 2 hybridized and the total number of pi electrons is equal to 4n+2.
  • Aryl refers to a monocyclic or fused bicyclic ring assembly containing 6 to 10 ring carbon atoms wherein each ring is aromatic e.g., phenyl or naphthyl.
  • Aryloxy refers to a — O-R radical where R is aryl as defined above e.g., phenoxy, napthyloxy, and the like.
  • Aryloxycarbonyl refers to a -C(O)OR radical where R is aryl as defined above e.g., phenyloxycarbonyl, naphthyloxycarbonyl, and the like.
  • Aralkyl refers to a -(alkylene)-R radical where R is aryl as defined above e.g., benzyl, phenethyl, and the like.
  • Aralkyloxy refers to a -O-R radical where R is aralkyl as defined above e.g., benzyloxy, phenethyloxy, and the like.
  • Alkyloxycarbonyl refers to a -C(O)OR radical where R is aralkyl as defined above e.g., benzyloxycarbonyl, phenethyloxycarbonyl, and the like.
  • Biologic means a therapeutic agent originally derived from living organisms for the treatment or management of a disease. Examples include, but are not limited to, proteins (recombinant and plasma derived), monoclonal or polyclonal, humanized or murine antibodies, toxins, hormones, and the like. Biologies are currently available for the treatment of a variety of diseases such as cancer, rheumatoid arthritis, and hemophilia.
  • Bridged azabicyclic ring means a bridged bicylic ring containing 7 or 8 ring atoms wherein one or two ring atoms are nitrogen and the remaining ring atoms being carbon. The ring is attached to the sulfonyl group via the nitrogen atom. Representative examples include, but are not limited to, the following:
  • Carboxy refers to -C(O)OH radical.
  • Carboxyalkyl means a linear monovalent hydrocarbon radical of one to six carbon atoms or a branched monovalent hydrocarbon radical of three to six carbons substituted with at least one, preferably one or two, carboxy group(s) e.g., carboxymethyl, carboxyethyl, and the like.
  • Cycloalkyl refers to a monovalent saturated monocyclic ring containing three to eight ring carbon atoms e.g., cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, and the like.
  • Cycloalkylalkyl refers to a -(alkylene)-R radical where R is cycloalkyl as defined above e.g., cyclopropylmethyl, cyclobutylethyl, cyclobutylmethyl, and the like.
  • Cycloalkyloxycarbonyl refers to a -C(O)OR radical where R is cyccloalkyl as defined above e.g., cyclopropyloxycarbonyl, cyclopentyloxycarbonyl, and the like.
  • Cycloalkylalkyloxycarbonyl refers to a -C(O)OR radical where R is cycloalkylalkyl as defined above e.g., cyclopropylmethyloxycarbonyl, cyclopentylmethyloxycarbonyl, and the like.
  • Cycloalkylene refers to a divalent saturated monocyclic ring containing three to eight ring carbon atoms.
  • R 1 and R 2 together with the carbon atom to which both R 1 and R 2 are attached form cycloalkylene includes, but is not limited to, the following:
  • Disease specifically includes any unhealthy condition of an animal or part thereof and includes an unhealthy condition that may be caused by, or incident to, medical or veterinary therapy applied to that animal, i.e., the "side effects" of such therapy.
  • Deleterious immune response means an immune response that prevents effective treatment of a patient or causes disease in a patient.
  • dosing a patient with a murine antibody either as a therapy or a diagnostic agent causes the production of human antimouse antibodies that prevent or interfere with subsequent treatments.
  • the incidence of antibody formation versus pure murine monoclonals can exceed 70%. ⁇ see Khazaeli, M. B. et al. J. Immunother, 1994, 15, pp 42-52; Dillman R. O. et al. Cancer Biother. 1994, 9, pp 17- 28; and Reinsberg, J. Hybridoma. 1995, 14, pp 205-208).
  • factor VIII blood-clotting factors
  • factor VIII When administered to hemophilia A patients, factor VIII restores the ability of the blood to clot. Although factor VIII is a human protein, it still elicits an immune response in hemophiliacs as endogenous factor VIII is not present in their blood and thus it appears as a foreign antigen to the immune system. Approximately 29-33% of new patients will produce antibodies that bind and neutralize the therapeutically administered factor VIII ⁇ see Lusher J. M. Semin Thromb Hemost. 2002, 28(3), pp 273-276).
  • Retroviral therapy remains experimental and is of limited utility. One reason is that the application of a therapeutic virus generates an immune response capable of blocking any subsequent administration of the same or similar virus ⁇ see Yiping Yang et al. J. of Virology. 1995, 69, pp 2004-2015). This ensures that retroviral therapies must be based on the transient expression of a protein or the direct incorporation of viral sequence into the host genome.
  • a "deleterious immune response” also encompasses diseases caused by therapeutic agents.
  • a specific example of this is the immune response to therapy with recombinant human erythropoietin (EPO).
  • EPO erythropoietin
  • Erythropoietin is used to stimulate the growth or of red cells and restore red blood cell counts in patients who have undergone chemotherapy or dialysis.
  • a small percentage of patients develop antibodies to EPO and subsequently are unresponsive to both therapeutically administered EPO and their own endogenous EPO ⁇ see Casadevall, N. et al, NEJM.
  • Humira ® is a monoclonal antibody directed against TNF and is used to treat rheumatoid arthritis patients. When taken alone ⁇ 12% of patients develop neutralizing antibodies. In addition, a small percentage of patients given the drug also contract a systemic lupus erthematosus-like condition that is an IgG- mediated immune response induced by the therapeutic agent (see Humira package label).
  • Another example of "deleterious immune response” is a host reaction to small molecule drugs. It is known to those skilled in the art that certain chemical structures will conjugate with host proteins to stimulate immune recognition (see Ju. C. et al. 2002. Current Drug Metabolism 3, pp 367-377 and Kimber I. et al. 2002, Toxicologic Pathology 30, pp 54- 58.) A substantial portion of these host reactions are IgG mediated. Specific "deleterious immune responses" that are IgG mediated include: hemolytic anemia, Steven- Johnson syndrome and drug induced Lupus.
  • Halo refers to fluoro, chloro, bromo or iodo.
  • Haloalkyl refers to alkyl as defined above substituted by one or more, preferably one to seven, "halo" atoms, as such terms are defined in this Application.
  • Haloalkyl includes monohaloalkyl, dihaloalkyl, trihaloalkyl, perhaloalkyl and the like e.g. chloromethyl, dichloromethyl, difluoromethyl, trifluoromethyl, 2,2,2-trifluoroethyl, perfluoroethyl, 2,2,2-trifluoro-l,l-dichloroethyl, and the like).
  • Haloalkoxy refers to a -OR radical where R is haloalkyl group as defined above e.g., trifluoromethoxy, 2,2,2-trifluoroethoxy, difluoromethoxy, and the like.
  • Heteroaryl as a group or part of a group denotes an aromatic monocyclic or bicyclic moiety of 5 to 10 ring atoms in which one or more, preferably one, two, or three, of the ring atom(s) is(are) selected from nitrogen, oxygen or sulfur, the remaining ring atoms being carbon.
  • heteroaryl rings include, but are not limited to, pyrrolyl, furanyl, thienyl, oxazolyl, isoxazolyl, thiazolyl, imidazolyl, triazolyl, tetrazolyl, pyridinyl, pyrimidinyl, pyrazinyl, pyridazinyl, indolyl, benzofuranyl, benzothiophenyl, benzimidazolyl, quinolinyl, isoquinolinyl, quinazolinyl, quinoxalinyl, pyrazolyl, and the like.
  • Heteroaryloxy refers to a -OR radical where R is heteroaryl as defined above e.g., furanyloxy, pyridinyloxy, indolyloxy, and the like.
  • Heteroaryl oxycarbonyl refers to a -C(O)O-R radical where R is heteroaryl as defined above e.g., pyridinyloxycarbonyl, pyrimidinyloxycarbonyl, and the like.
  • Heteroaralkyl refers to a -(alkylene)-R radical where R is heteroaryl as defined above e.g., pyridinylmethyl, 1- or 2-furanylethyl, imidazolylmethyl, and the like.
  • ⁇ eteroaralkyloxy refers to a -O-R radical where R is heteroaralkyl as defined above e.g., pyridinylmethyloxy, furanylethyloxy, and the like.
  • Heteroaralkyloxycarbonyl refers to a -C(O)O-R radical where R is heteroaralkyl as defined above e.g., pyridinylmethyloxycarbonyl, pyrimidinylmethyloxycarbonyl, and the like.
  • the heterocycloalkyl ring is optionally fused to cycloalkyl, aryl or heteroaryl ring as defined herein.
  • Representative examples include, but are not limited to, imidazolidinyl, morpholinyl, thiomorpholinyl, thiomorpholino-1 -oxide, thiomorpholino-1,1- dioxide, tetrahydrofuranyl, tetrahydropyranyl, tetrahydrothiopyranyl, 1 -oxo- tetrahydrothiopyranyl, 1,1-dioxotetrathio-pyranyl, indolinyl, piperazinyl, piperidyl, pyrrolidinyl, pyrrolinyl, quinuclidinyl,3,4-dihydroisoquinolinyl, dihydroindolyl, and the like.
  • heterocycloalkyl group contains at least one nitrogen ring atom it is referred to herein as "heterocycloamino" and is a subset of the heterocycloalkyl group as defined above.
  • Heterocyclylalkylene refers to a divalent heterocyclyl group, as defined in this Application, e.g., the instance wherein R and R 4 together with the carbon atom to which both R 3 and R 4 are attached form heterocyclylalkylene” includes, but is not limited to, the following:
  • Heterocycloalkylalkyl refers to a -(alkylene)-R radical where R is heterocycloalkyl as defined above e.g., pyrrolidinylmethyl, tetrahydrofuranylethyl, pyridinylmethylpiperidinylmethyl, and the like.
  • Heterocycloalkyloxycarbonyl refers to a -C(O)OR radical where R is heterocycloalkyl as defined above e.g., pyridinyloxycarbonyl, pyrimidinyloxycarbonyl, and the like.
  • Heterocycloalkylalkyloxycarbonyl refers to a -C(O)OR radical where R is heterocycloalkyla;lyl as defined above e.g., pyridinylmethyloxycarbonyl, pyrimidinylmethyloxycarbonyl, and the like.
  • Heterocycloalkylaminocarbonyl refers to a -CONHR radical where R is heterocycloalkyl as defined above e.g., tetrahydrofuranylaminocarbonyl, tetrahydropyranylaminocarbonyl, and the like.
  • Hydroxy means -OH radical. Unless indicated otherwise, the compounds of the invention containing hydroxy radicals include protected derivatives thereof. Suitable protecting groups for hydroxy moieties include benzyl and the like.
  • Hydroalkyl means a linear monovalent hydrocarbon radical of one to six carbon atoms or a branched monovalent hydrocarbon radical of three to six carbons substituted with one or two hydroxy groups, provided that if two hydroxy groups are present they are not both on the same carbon atom.
  • Representative examples include, but are not limited to, hydroxymethyl, 2-hydroxyethyl, 2-hydroxypropyl, 3-hydroxypro ⁇ yl, l-(hydroxymethyl)-2- methylpropyl, 2-hydroxybutyl, 3-hydroxybutyl, 4-hydroxybutyl, 2,3-dihydroxypropyl, 1- (hydroxymethyl)-2-hydroxyethyl, 2,3-dihydroxybutyl, 3,4-dihydroxybutyl and 2- (hydroxymethyl)-3-hydroxypropyl, preferably 2-hydroxyethyl, 2,3-dihydroxypropyl, and 1- (hydroxymethyl)-2-hydroxyethyl.
  • Isomers mean compounds of Formula (I) having identical molecular formulae but differ in the nature or sequence of bonding of their atoms or in the arrangement of their atoms in space. Isomers that differ in the arrangement of their atoms in space are termed “stereoisomers”. Stereoisomers that are not mirror images of one another are termed “diastereomers” and stereoisomers that are nonsuperimposable mirror images are termed “enantiomers” or sometimes "optical isomers”. A carbon atom bonded to four nonidentical substituents is termed a "chiral center”. A compound with one chiral center that has two enantiomeric forms of opposite chirality is termed a "racemic mixture”.
  • a compound that has more than one chiral center has 2" "1 enantiomeric pairs, where n is the number of chiral centers.
  • Compounds with more than one chiral center may exist as either an individual diastereomer or as a mixture of diastereomers, termed a "diastereomeric mixture".
  • a stereoisomer may be characterized by the absolute configuration of that chiral center. Absolute configuration refers to the arrangement in space of the substituents attached to the chiral center.
  • Enantiomers are characterized by the absolute configuration of their chiral centers and described by the R- and ⁇ -sequencing rules of Cahn, Ingold and Prelog.
  • JV-oxide derivatives of a compound of Formula (I) mean a compound of Formula (I) in which a nitrogen atom is in an oxidized state (i.e., N-»O) e.g., pyridine iV-oxide, and which possess the desired pharmacological activity.
  • "Pathology" of a disease means the essential nature, causes and development of the disease as well as the structural and functional changes that result from the disease processes.
  • “Pharmaceutically acceptable” means that which is useful in preparing a pharmaceutical composition that is generally safe, non-toxic and neither biologically nor otherwise undesirable and includes that which is acceptable for veterinary use as well as human pharmaceutical use.
  • “Pharmaceutically acceptable salts” means salts of compounds of Formula (I) which are pharmaceutically acceptable, as defined above, and which possess the desired pharmacological activity.
  • Such salts include acid addition salts formed with inorganic acids such as hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid, phosphoric acid, and the like; or with organic acids such as acetic acid, propionic acid, hexanoic acid, heptanoic acid, cyclopentanepropionic acid, glycolic acid, pyruvic acid, lactic acid, malonic acid, succinic acid, malic acid, maleic acid, fumaric acid, tartaric acid, citric acid, benzoic acid, o-(4-hydroxybenzoyl)benzoic acid, cinnamic acid, mandelic acid, methylsulfonic acid, ethanesulfonic acid, 1 ,2-ethanedisulfonic acid, 2-hydroxy-ethanesulf
  • Pharmaceutically acceptable salts also include base addition salts which may be formed when acidic protons present are capable of reacting with inorganic or organic bases.
  • Acceptable inorganic bases include sodium hydroxide, sodium carbonate, potassium hydroxide, aluminum hydroxide and calcium hydroxide.
  • Acceptable organic bases include ethanolamine, diethanolamine, triethanolamine, tromethamine, JV-methylglucamine and the like.
  • the present invention also includes prodrugs of a compound of Formula (I).
  • Prodrug means a compound that is convertible in vivo by metabolic means (e.g. by hydrolysis) to a compound of Formula (I).
  • an ester of a compound of Formula (I) containing a hydroxy group may be convertible by hydrolysis in vivo to the parent molecule.
  • an ester of a compound of Formula (I) containing a carboxy group may be convertible by hydrolysis in vivo to the parent molecule.
  • Suitable esters of compounds of Formula (I) containing a hydroxy group are for example acetates, citrates, lactates, tartrates, malonates, oxalates, salicylates, propionates, succinates, fumarates, maleates, methylene-bis- ⁇ b-hydroxynaphthoates, gentisates, isethionates, di-p-toluoyltartrates, methylsulphonates, ethanesulphonates, benzenesulphonates, j9-toluenesulphonates, cyclohexylsulphamates and quinates.
  • esters of compounds of Formula (I) containing a carboxy group are for example those described by Leinweber, FJ. Drug Metab. Res., 1987, 18, page 379.
  • An especially useful class of esters of compounds of Formula (I) containing a hydroxy group may be formed from acid moieties selected from those described by Bundgaard et al., J. Med. Chem., 1989, 32, pp 2503-2507, and include substituted (aminomethyl)-benzoates, for example, dialkylamino-methylbenzoates in which the two alkyl groups may be joined together and/or interrupted by an oxygen atom or by an optionally substituted nitrogen atom, e.g.
  • an alkylated nitrogen atom more especially (morpholino-methyl)benzoates, e.g. 3- or 4-(mo ⁇ holinomethyl)-benzoates, and (4-alkylpiperazin-l-yl)benzoates, e.g. 3- or 4-(4-alkylpiperazin-l-yl)benzoates.
  • (morpholino-methyl)benzoates e.g. 3- or 4-(mo ⁇ holinomethyl)-benzoates
  • (4-alkylpiperazin-l-yl)benzoates e.g. 3- or 4-(4-alkylpiperazin-l-yl)benzoates.
  • Protected derivatives means derivatives of compounds of Formula (I) in which a reactive site or sites are blocked with protecting groups.
  • Protected derivatives of compounds of Formula (I) are useful in the preparation of compounds of Formula (I) or in themselves may be active cathepsin S inhibitors.
  • a comprehensive list of suitable protecting groups can be found in T. W. Greene, Protective Groups in Organic Synthesis, 3rd edition, John Wiley & Sons, Inc. 1999.
  • “Therapeutically effective amount” means that amount which, when administered to an animal for treating a disease, is sufficient to effect such treatment for the disease.
  • Treatment or “treating” means any administration of a compound of the present invention and includes:
  • Treatment or “treating” with respect to combination therapy i.e., use with a biologic means any administration of a compound of the present invention and includes:
  • R 5 is optionally substituted with one, two, or three R a independently selected from alkyl, haloalkyl, alkoxy, hydroxy, haloalkoxy, or halo; or optionally substituted with one or two R b independently selected from hydrogen, alkyl, haloalkyl, alkoxy, hydroxy, haloalkoxy, halo, carboxy, or alkoxycarbonyl and one R c selected from hydroxyalkyl, alkoxyalkyl, aminoalkyl, aryl, heteroaryl, aralkyl, heteroaralkyl, cycloalkyl, cycloalkylalkyl, " in the definition of R 5 in the compound of Formula (I) means that all the aromatic and alicyclic rings within the scope of R 5 whether directly or indirectly attached (e.g., R 5 is cycloalkylalkyl, -alkylene-X-R 9 where
  • R 1 is hydrogen or methyl, preferably hydrogen
  • R 2 is cyclopropyl, 1-phenylethyl, or lH-pyrazol-5-yl; preferably cyclopropyl.
  • R 3 is hydrogen and R 4 is alkyl, preferably methyl, ethyl, propyl or butyl, more preferably R 4 is ethyl or propyl.
  • a more preferred group of compounds is that wherein R 3 is alkyl, preferably methyl or ethyl and R 4 is alkyl, preferably methyl, ethyl, propyl or butyl, more preferably R 4 is methyl.
  • R 3 and R 4 are methyl.
  • a more preferred group of compounds is that wherein R 3 and R 4 together with the carbon atom to which they are attached form cycloalkylene, preferably cyclopropylene, cyclopentylene, or cyclohexylene, more preferably cyclopropylene.
  • a more preferred group of compounds is that wherein R 3 and R 4 together with the carbon atom to which they are attached form piperidin-4-yl substituted at the nitrogen atom with ethyl, 2,2,2-trifluoroethyl or cyclopropyl, tetrahydropyran-4-yl, tetrahydrothiopyran-4- yl, or l,l-dioxotetrahydrothiopyran-4-yl.
  • R 6 is haloalkyl, preferably, difluoromethyl, trifiuoromethyl, 2,2,2-trifluoroethyl, 2,2,3,3,3-pentafluoropropyl, 2,2,3,3,4,4,4-heptafluorobutyl and R 7 and R 8 are hydrogen.
  • a more preferred group of compounds is that wherein R 6 is haloalkyl, preferably, difluoromethyl, trifiuoromethyl, 2,2,2-trifluoroethyl, or 2,2,3,3,3- pentafluoropropyl, R 7 is haloalkyl, preferably, trifiuoromethyl, 2,2,2-trifluoroethyl, or 2,2,3,3,3-pentafluoropropyl, and R 8 is hydrogen.
  • a more preferred group of compounds is that wherein R 6 is haloalkyl, preferably, difluoromethyl, trifluoromethyl, 2,2,2-trifiuoroethyl, or 2,2,3,3,3- pentafluoropropyl, R 7 is alkyl, preferably, methyl, ethyl, or propyl, and R 8 is hydrogen.
  • a more preferred group of compounds is that wherein R 6 is haloalkyl, preferably, difluoromethyl, trifluoromethyl, 2,2,2-trifluoroethyl, or 2,2,3,3,3- pentafluoropropyl, R 7 is haloalkyl, preferably, trifluoromethyl or 2,2,2-trifluoroethyl, and R 8 is aryl optionally substituted with one, two, or three R e .
  • R 8 is phenyl, A- fluorophenyl, 2,3-, 2,4-, 2,5-, 2,6-, 3,4, or 3,5-difluorophenyl. More preferably, R 6 and R 7 are trifluoromethyl and R 8 is phenyl, 4-fluorophenyl, 2,3-, 2,4-, 2,5-, 2,6-, 3,4, or 3,5- difluorophenyl.
  • a more preferred group of compounds is that wherein R 6 is haloalkyl, preferably, difluoromethyl, trifluoromethyl, 2,2,2-trifluoroethyl, or 2,2,3,3,3- pentafluoropropyl, R 7 is alkyl, preferably, methyl or ethyl, and R 8 is aryl optionally substituted with one, two, or three R e .
  • R 8 is phenyl, 4-fluorophenyl, 2,3-, 2,4-, 2,5-, 2,6-, 3,4, or 3,5-difluorophenyl. More preferably, R 6 and R 7 are trifluoromethyl and R 8 is phenyl, 4-fluorophenyl, 2,3-, 2,4-, 2,5-, 2,6-, 3,4, or 3,5-difluorophenyl.
  • a more preferred group of compounds is that wherein R 6 is haloalkyl, preferably, trifluoromethyl, difluoromethyl, 2,2,2-trifluoroethyl, or 2,2,3,3,3- pentafluoropropyl, R 7 is hydrogen, and R 8 is aryl optionally substituted with one, two, or three R e .
  • R 8 is phenyl, 4-fluorophenyl, 2,3-, 2,4-, 2,5-, 2,6-, 3,4, or 3,5- difluorophenyl.
  • R 6 is trifluoromethyl and R 8 is phenyl, 4-fluorophenyl, 2,3- , 2,4-, 2,5-, 2,6-, 3,4, or 3,5-difluorophenyl, preferably 2,4-difluorophenyl.
  • a more preferred group of compounds is that wherein R 6 is haloalkyl, preferably, trifluromethyl, 2,2,2-trifluoroethyl, or 2,2,3,3,3-pentafluoropropyl, R 7 is haloalkyl, preferably, trifluoromethyl or 2,2,2-trifluoroethyl, and R 8 is heteroaryl optionally substituted with one, two, or three R e .
  • R 8 is indol-5-yl, benzoxazol-5-yl, thiophen- 3-yl, thiophen-2-yl, furan-2-yl, pyridin-4-yl, pyridin-3-yl, pyridin-2-yl, imidazol-5-yl, pyrimidin-2-yl, pyrazin-2-yl, pyrimidin-5-yl, pyrimdin-4-yl, pyridazin-4-yl, isoxazol-4-yl, imidazol-2-yl, [1.2.3]thiadiazol-4-yl, imidazol-4-yl, pyrazol-4-yl, thiazol-2-yl, pyrazol-4-yl, pyrrol-2-yl, pyrrol-3-yl, thiazol-4-yl, thiazol-5-yl optionally substituted with one or two methyl.
  • a more preferred group of compounds is that wherein R 6 is haloalkyl, preferably, trifluromethyl, 2,2,2-trifiuoroethyl, or 2,2,3,3,3-pentafluoropropyl, R 7 is alkyl, preferably, methyl or ethyl, and and R 8 is heteroaryl optionally substituted with one, two, or three R e .
  • R 8 is indol-5-yl, benzoxazol-5-yl 5 thiophen-3-yl, thiophen-2-yl, furan-2- yl, pyridine-4-yl, pyridin-3-yl, pyridin-2-yl, imidazol-5-yl, pyrimidin-2-yl, pyrazin-2-yl, pyrimidin-5-yl, pyrimdin-4-yl, pyridazin-4-yl, isoxazol-4-yl, imidazol-2-yl, [1.2.3]thiadiazol- 4-yl, imidazol-4-yl, pyrazol-4-yl, thiazol-2-yl, pyrazol-4-yl, pyrrol-2-yl, pyrrol-3-yl, thiazol- 4-yl, thiazol-5-yl optionally substituted with one or two methyl.
  • a more preferred group of compounds is that wherein R 6 is haloalkyl, preferably, trifluromethyl, 2,2,2-trifiuoroethyl, or 2,2,3,3,3-pentafluoropropyl, R 7 is hydrogen, and and R 8 is heteroaryl optionally substituted with one, two, or three R e .
  • R 8 is indol-5-yl, benzoxazol-5-yl, thiophen-3-yl, thiophen-2-yl, furan-2-yl, pyridin-4-yl, pyridin-3-yl, pyridin-2-yl, imidazol-5-yl, pyrimidin-2-yl, pyrazin-2-yl, pyrimidin-5-yl, pyrimdin-4-yl, pyridazin-4-yl, isoxazol-4-yl, imidazol-2-yl, [1.2.3]thiadiazol- 4-yl, imidazol-4-yl, pyrazol-4-yl, thiazol-2-yl, pyrazol-4-yl, pyrrol-2-yl, pyrrol-3-yl, thiazol- 4-yl, thiazol-5-yl optionally substituted with one or two methyl.
  • R 11 is hydrogen, alkyl, haloalkyl, alkoxyalkyl, hydroxyalkyl, aminoalkyl, carboxyalkyl, alkoxycarbonylalkyl, acyl, cycloalkyl, cycloalkylalkyl, aryl, heteroaryl, aralkyl, heteroaralkyl, heterocycloalkylalkyl, acylalkyl, or heterocycloalkylaminocarbonyl; and R 12 is hydrogen, alkyl, aryl, heteroaryl, aralkyl, heteroaralkyl, cycloalkyl, cycloalkylalkyl, heterocycloalkyl, or heterocycloalkylalkyl; wherein the aromatic or alicyclic ring are optionally substituted with one, two, or three R a independently selected from alkyl, haloalkyl, alkoxy, hydroxy, haloalkoxy, cyano, halo, carboxy or alkoxy
  • R 11 is methyl, ethyl, propyl, butyl, 2,2,2-trifluoroethyl, 2-hydroxethyl, 2- or 3-hydroxypropyl, 2-methoxy or ethoxyethyl, 2- or 3-methoxy or ethoxypropyl, methylaminoethyl, methylaminopropyl, acetylaminoethyl, 2-carboxy ethyl, 3-carboxypropyl, methoxycarbonylethyl, acetyl, cyclopropyl, cyclopropylmethyl, benzyl, phenylethyl, pyridinylethyl, pyridinylmethyl, pyrimidinylmethyl, furanylmethyl, pyrrolylmethyl, indolylmethyl, quinolinylmethyl, isoquinolinylmethyl, or tetrahydroquinolinylmethyl and R
  • R 5 is -alkylene-SO 2 NR n R 12 where:
  • R 11 is alkoxyalkyl, hydroxyalkyl, aminoalkyl, carboxyalkyl, alkoxycarbonylalkyl, cycloalkyl, cycloalkylalkyl, aryl, heteroaryl, aralkyl, heteroaralkyl, heterocycloalkylalkyl and
  • R 12 is hydrogen, alkyl, aryl, heteroaryl, aralkyl, heteroaralkyl, cycloalkyl, cycloalkylalkyl, heterocycloalkyl, or heterocycloalkylalkyl; one, two, or three R a independently selected from alkyl, haloalkyl, alkoxy, hydroxy, haloalkoxy, cyano, halo, carboxy or alkoxycarbonyl; or optionally substituted with one or two R b independently selected from hydrogen, alkyl, haloalkyl, alkoxy, hydroxy, haloalkoxy, halo, carboxy, or alkoxycarbonyl and one R c selected from cyano, hydroxyalkyl, alkoxyalkyl, aminoalkyl, aryl, heteroaryl, aralkyl, heteroaralkyl, cycloalkyl, cycloalkylalkyl, heterocycloalkyl, heterocycloalkyl
  • R 11 is 2-hydroxethyl, 2- or 3-hydroxypropyl, 2-methoxy or ethoxyethyl, 2- or 3-methoxy or ethoxypropyl, methylaminoethyl, methylaminopropyl, acetylaminoethyl, 2-carboxyethyl, 3-carboxypropyl, methoxycarbonylethyl, acetyl, cyclopropyl, cyclopropylmethyl, benzyl, phenylethyl, pyridinylethyl, pyridinylmethyl, pyrimidinylmethyl, furanylmethyl, pyrrolylmethyl, indolylmethyl, quinolinylmethyl, isoquinolinylmethyl, tetrahydroquinolinylmethyl or -(CH 2 ) 2 -NRR (where R is methyl, ethyl, hydroxyethyl,
  • R 5 is — alkylene-SO 2 NR n R 12 where R 11 and R 12 together with the nitrogen atom to which they are attached form heterocycloamino substituted with one, two, or three R a independently selected from alkyl, haloalkyl, alkoxy, hydroxy, haloalkoxy, cyano, halo, carboxy or alkoxycarbonyl; or optionally substituted with one or two R b independently selected from hydrogen, alkyl, haloalkyl, alkoxy, hydroxy, haloalkoxy, halo, carboxy, or alkoxycarbonyl and one R c selected from cyano, hydroxyalkyl, alkoxyalkyl, aminoalkyl, aryl, heteroaryl, aralkyl, heteroaralkyl, cycl
  • R 11 and R 12 together with the nitrogen atom to which they are attached form 2,3-dihydroindol-l-yl, l,3-dihydroisoindol-2-yl, 3,4-dihydroisoquinolin-2-yl, morpholinyl, piperidin- 1 -yl or piperazin- 1 -yl, preferably piperazin- 1 -yl, optionally substituted with one or two R b independently selected from hydrogen, methyl, trifluoromethyl, methoxy, hydroxy, trifluoromethoxy, carboxy, fluoro, or methoxycarbonyl and one R c selected from hydroxymethyl, hydroxyethyl, 2-or 3-hydroxypropyl, 2- dimethylaminoethyl, phenyl, benzyl, pyridinyl, pyrimidinyl, pyrazinyl, pyrrolyl, cyclopropyl, cyclopropyl, cycloprop
  • R 5 is -alkylene-SO 2 NR u R 12 where R 11 and R 12 together with the nitrogen atom to which they are attached form a bridged azabicyclic ring optionally substituted with one, two, or three R a independently selected from alkyl, haloalkyl, alkoxy, hydroxy, haloalkoxy, or halo; or optionally substituted with one or two R b independently selected from hydrogen, alkyl, haloalkyl, alkoxy, hydroxy, haloalkoxy, halo, carboxy, or alkoxycarbonyl and one R 0 selected from hydroxyalkyl, alkoxyalkyl, aminoalkyl, aryl, heteroaryl, aralkyl, heteroaralkyl, cycloalkyl, cycloalkyl, cycloalkyl, cycloalkyl, cycloalkyl, cycloalkyl, cycloalkyl, cycloalkyl, cyclo
  • R 5 is -alkylene-S0 2 NR n R 12 where R 11 and R 12 together with the nitrogen atom to which they are attached form heterocycloamino optionally substituted with one, two, or three R a independently selected from alkyl, haloalkyl, alkoxy, hydroxy, haloalkoxy, or halo.
  • R 11 and R 12 together with the nitrogen atom to which they are attached form 2,3-dihydroindol-l-yl, 3,4-dihydroisoquinolin-2-yl, morpholinyl, piperidin-1-yl or piperazin-1-yl optionally substituted with one, two, or three R a independently selected from methyl, ethyl, trifluoromethyl, methoxy, hydroxyl, ethoxy, trifluoromethoxy, or fluoro.
  • R 5 is -alkylene-SO 2 NR u R 12 where R 11 and R 12 together with the nitrogen atom to which they are attached form heterocycloamino optionally substituted with one or two R b independently selected from hydrogen, alkyl, haloalkyl, alkoxy, hydroxy, haloalkoxy, halo, carboxy, or alkoxycarbonyl and one R c selected from aryl, heteroaryl, aralkyl, heteroaralkyl, cycloalkyl, cycloalkylalkyl, acyl, or alkoxycarbonyl; and further wherein the aromatic or alicyclic ring in R c is optionally substituted with one, two, or three R independently selected from alkyl, haloalkyl, alkoxy, hydroxy, hal
  • R 11 and R 12 together with the nitrogen atom to which they are attached form 2,3- dihydroindol- 1 -yl, 3 ,4-dihydroisoquinolin-2-yl, morpholinyl, piperidin- 1 -yl or piperazin- 1 -yl optionally substituted with with one or two R b independently selected from hydrogen, methyl, trifiuoromethyl, methoxy, hydroxyl, trifluoromethoxy, carboxy, fluoro, or methoxycarbonyl and one R c selected from phenyl, pyridinyl, pyrimidinyl, benzyl, cyclopropyl, cyclopropylmethyl, benzoyl, acetyl, or trifluoroacetyl; and further wherein the aromatic or alicyclic ring in R c is optionally substituted with one, two, or three R d independently selected from methyl, ethyl, trifiuor
  • R 5 is -alkylene-SO 2 NR u R 12 where R 11 is 2- imidazol-4-ylethyl 5 imidazol-4-ylmethyl, l-methylimidazol-4-ylmethyl, 2-(l-methylimidazol- 4-yl)ethyl, 4-CF 3 pyridin-3-yl, 4-CNpyridin-3-yl, 3-CF 3 pyridin-2-yl, 3-CN ⁇ yridin-2-yl, 3- CF 3 pyridin-4-yl, 3-CN ⁇ yridin-4-yl, 2-CF 3 pyridin-3-yl, 2-CNpyridin-3-yl, 2- JV- methylaminoethyl, 2-JV,JV-dimethylaminoethyl, 2-iV-e
  • N(CH 3 )CO)amino]ethyl 2-[iV-(4-CO 2 HphenylmethylN(CH 3 )CO)amino]ethyl, 2-[iV-(2- CONH 2 phenylmethylN(CH 3 )CO)amino]ethyl, 2-[N-(3-CONH 2 phenylmethylN(CH 3 )CO)- amino]ethyl, 2-[N-(4-CONH 2 ⁇ henylmethylN(CH 3 )CO)amino]ethyl 2-[iV-(pyridin-2- ylmethylN(CH 3 )CO)amino]ethyl, 2-[iV-(pyridin-3-ylmethylN(CH 3 )CO)amino]ethyl, 2-[N- (pyridin-4-ylmethylN(CH 3 )CO)amino]ethyl, 2-[N-(pyrimidin-2-ylmethylN(CH 3
  • R 5 is -alkylene-SC ⁇ NR 11 R 12 where here R 11 and R 12 together with the nitrogen atom to which they are attached form piperidin-1-yl, A- methylpiperidin-1-yl, 4-ethylpiperidin-l-yl, 4-(2,2,2-trifluoroethyl)piperidin-l-yl, 4-(2- isopropyl)piperidin- 1 -yl, 4-(2-trifluoromethyl-2,2,2-trifluoroethyl) ⁇ iperidin- 1 -yl A- (cyclopropyl)piperidin-l-yl, 4-(cyclobutyl)piperidin-l-yl, 4-(oxetan-3-yl)piperidin-l-yl A- (azetidin-3 -
  • y l 5 4.(6-O ⁇ -2,3-dihydiO-lH-indol-l-ylCO)piperidin-l-yl, 4-(5-O ⁇ -2,3-dihydro-lH-indol- l-ylCO)piperidin-l-yl, 4-(7-CO 2 ⁇ -2,3-dihydro-lH-indol-l-ylCO)piperidin-l-yl, 4-(6-CO 2 H- 2,3-dihydro-lH-indol-l-ylCO)piperidin-l-yl, 4-(5-CO 2 H-2,3-dihydro-lH-indol-l- ylCO)piperidin- 1 -yl, 4-(2,3 -dihydro- 1 ⁇ -pyrrolo [ 2,3 -b]pyridin- 1 -ylCO)piperidin- 1 -yl, 4-(2,
  • R 5 is -alkylene-SO 2 NR R where here R 11 and R 12 together with the nitrogen atom to which they are attached form piperazin-1-yl, A- methylpiperazin-1-yl, 4-ethylpiperazin-l-yl, 4-(2,2,2-trifluoroethyl)piperazin-l-yl, 4-(2- isopropyl)piperazin- 1 -yl, 4-(2-trifluoromethyl-2,2,2-triiluoroethyl)piperazin- 1 -yl A- (cyclopropyl)piperazin-l-yl, 4-(cyclobutyl)piperazin-l-yl, 4-(oxetan-3-yl)piperazin-l-yl A- (azetidin-3-yl)
  • A6 [(2-OCH 3 phenyl)methylNCO]piperazin-l-yl, 4-[(3-CNphenyl)methylNCO]piperazin-l-yl, 4- [(4-CNphenyl)methylNCO]piperazin- 1 -yl, 4- [(2-CNphenyl)methylNCO]piperazin- 1 -yl, 4- [(3-OHphenyl)methylNCO]piperazin-l-yl, 4-[(4-OHphenyl)methylNCO]piperazin-l-yl, 4- [(2-OHphenyl)methylNCO]piperazin- 1 -yl, 4-[(pyridin-2-yl)methylNCO]piperazin- 1 -yl, 4- [(pyridin-3-yl)methylNCO]piperazin-l-yl, 4-[(pyridin-4-yl)methylNCO]piperazin-l-yl, 4- [(pyrimidin-2-yl)methylNCO]pipe
  • the stereochemistry at the carbon to which R 5 is attached is (R) and to which R 4 and R 6 are attached is (S).
  • the stereochemistry at the carbon to which R 5 and R 6 are attached is (R) and to which R 4 is attached is (S).
  • R 3 is alkyl, preferably methyl or ethyl and R 4 is alkyl, preferably methyl, ethyl, propyl or butyl, more preferably R 4 is methyl. Preferably, R 3 and R 4 are methyl.
  • R 3 and R 4 are methyl.
  • Yet another preferred group of compounds of Formula (I) is that wherein R 3 and R 4 together with the carbon atom to which they are attached form cycloalkylene, preferably cyclopropylene, cyclopentylene, or cyclohexylene, more preferably cyclopropylene.
  • R 6 is haloalkyl, preferably, difluoromethyl, trifluoromethyl, 2,2,2-trifluoroethyl, or 2,2,3,3,3- pentafluoropropyl and R 7 and R 8 are hydrogen.
  • R 6 is haloalkyl, preferably, difluoromethyl, trifluoromethyl, 2,2,2-trifluoroethyl, or 2,2,3,3,3- pentafluoropropyl
  • R 7 is haloalkyl, preferably, trifluoromethyl, 2,2,2-trifluoroethyl, or 2,2,3,3,3-pentafluoropropyl
  • R 8 are hydrogen.
  • R 6 is haloalkyl, preferably, difluoromethyl, trifluoromethyl, 2,2,2-trifluoroethyl, or 2,2,3,3,3- pentafluoropropyl
  • R 7 is alkyl, preferably, methyl, ethyl, or propyl
  • R 8 are hydrogen.
  • more preferred groups of compounds are those wherein R 1 , R 2 , R 5 , R 6 , R 7 and R 8 are as defined for group (A) above.
  • more preferred groups of compounds are those wherein R 1 , R 2 , R 3 , R 4 , and R 5 are as defined for group (A) above.
  • the starting materials and the intermediates of the reaction may be isolated and purified if desired using conventional techniques, including but not limited to filtration, distillation, crystallization, chromatography and the like. Such materials may be characterized using conventional means, including physical constants and spectral data.
  • the reactions described herein take place at atmospheric pressure over a temperature range from about -78 0 C to about 150 0 C, more preferably from about 0 0 C to about 125 0 C and most preferably at about room (or ambient) temperature, e.g., about 20 0 C.
  • Reaction of a ketone of formula 1 where R 6 and R 8 are as defined in the Summary of the Invention with an ⁇ -amino ester of formula 2 where R is a carboxy protecting group, preferably an alkyl group, preferably methyl, and R 5 is as defined in the Summary of the Invention under reductive amination reaction conditions provide a compound of formula 3.
  • the reaction is carried out in the presence of a suitable dehydrating agent such as TiCl 4 , magnesium sulfate, isopropyl trifluoroacetate, in the presence of a base such as diisopropylethylamine, pyridine, and the like and in a suitable organic solvent such as methylene chloride to give an imine.
  • the imine is reduced with a suitable reducing agent such as sodium borohydride, sodium cyanoborohydride, and the like in a suitable organic solvent such as methanol, ethanol, and the like.
  • Hydrolysis of the ester group in compound 3 provides a compound of formula 4.
  • the hydrolysis conditions depend on the nature of the protecting group. For example, when R is alkyl the hydrolysis is carried out under aqueous basic hydrolysis reaction conditions to give the corresponding acid of formula 4.
  • the reaction is typically carried out with cesium carbonate, lithium hydroxide, and the like in an aqueous alcohol such as methanol, ethanol, and the like.
  • Compound 4 is then reacted with an ⁇ -hydroxyketoamide of formula 5 to give a compound of Formula 6.
  • the reaction is typically carried out in the presence of a suitable coupling agent e.g., benzotriazole-1-yloxytrispyrrolidinophosphonium hexafluorophosphate (PyBOP®), O-benzotriazol-l-yl-iV ⁇ N'jiV'-tetramethyl-uronium hexafluorophosphate (HBTU), ⁇ 9-(7-azabenzotriazol- 1 -yl)- 1 , 1 ,3,3 -tetramethyl-uronium hexafluorophosphate (HATU), l-(3-dimethylaminopropyl)-3-ethylcarbodiimide hydrochloride (EDC), or l ⁇ -dicyclohexyl-carbodiimide (DCC), optionally in the presence of 1-hydroxy-benzotri
  • reaction solvents are inert organic solvents such as halogenated organic solvents (e.g., methylene chloride, chloroform, and the like), acetonitrile, iV,7V-dimethylformamide, ethereal solvents such as tetrahydrofuran, dioxane, and the like.
  • halogenated organic solvents e.g., methylene chloride, chloroform, and the like
  • acetonitrile e.g., methylene chloride, chloroform, and the like
  • iV,7V-dimethylformamide ethereal solvents
  • ethereal solvents such as tetrahydrofuran, dioxane, and the like.
  • the above coupling step can be carried out by first converting 4 into an active acid derivative such as succinimide ester and then reacting it with an ⁇ - hydroxyketoamide of formula 5.
  • the reaction typically requires 2 to 3 h to complete.
  • the conditions utilized in this reaction depend on the nature of the active acid derivative. For example, if it is an acid chloride derivative of 4, the reaction is carried out in the presence of a suitable base (e.g. triethylamine, diisopropylethylamine, pyridine, and the like).
  • Suitable reaction solvents are polar organic solvents such as acetonitrile, ⁇ , ⁇ f-dimethylformarnide, dichloromethane, or any suitable mixtures thereof.
  • Compounds of formula 5 can be prepared by methods well known in the art e.g., they can be prepared by the procedures described in PCT application publication No. WO 02/18369, the disclosure of which is incorporated herein by reference in its entirety.
  • Oxidation of the hydroxyl group in compound 6 with a suitable oxidizing agent such as OXONE ® provides a compound of Formula (I).
  • Invention and PG is a suitable oxygen protecting group with a hemiacetal of formula 7 where R 6 is as defined in the Summary of the Invention provides an imine compound of of formula 9.
  • Treatment of 9 with an organolithium compound of formula R Li where R is not hydrogen provides compound 10.
  • Removal of the oxygen protecting group, followed by oxidation of the resulting alcohol 11 provides a compound of formula 4 which is then converted to a compound of Formula (I) as described in Scheme 1 above.
  • Suitable oxygen protecting groups and reaction conditions for putting them on and removing them can be found in Greene, T. W.; and Wuts, P. G. M.; Protecting Groups in Organic Synthesis; John Wiley & Sons, Inc. 1999.
  • Reaction of an amino acid compound of formula 2 where R is alkyl and R 5 is as defined in the Summary of the Invention with a hemiacetal compound of formula 7 provides a 2-(l-hydroxy-2,2,2-trifluoroethylarnino)acetate compound of formula 12.
  • the reaction is carried out in the presence of a catalytic amount of an acid such as ⁇ >-toluenesulfonic acid and in an aromatic hydrocarbon solvent such as toluene, benzene, and the like.
  • Reaction of a compound of formula 13 where R is as defined in Summary of the Invention with a compound of formula 14 where R' is hydrogen or a carboxy protecting group and R z is R 5 or a precursor group (e.g., -alkylene-S-trityl, and the like) to R 5 group provides a compound of formula 15.
  • the reaction is carried out in a suitable organic solvent, including but not limited to, diethyl ether, tetrahydrofuran, acetonitrile, benzene, toluene, xylene, and the like, or mixtures thereof and optionally in the presence of an organic or inorganic base.
  • the organic base is triethylamine, pyridine, JV-methylmorpholine, collidine, diisopropylethylamine, and the like.
  • the inorganic base is cesium carbonate, sodium carbonate, sodium bicarbonate, and the like.
  • the reaction is optionally carried out in the presence of a drying agent such as molecular sieves. Preferably, the reaction is carried out at room temperature.
  • Compounds of formula 13 can be prepared by methods well known in the art.
  • a compound of formula 13 where R 8 is phenyl or 4-fluorophenyl and R 6 is trifluoromethyl can be readily prepared from commercially available 2,2,2- trifluoroacetophenone or 2,2,2,4'-tetrafluoroacetophenone respectively, by reducing the keto group to an alcoholic group by suitable reducing agent such as sodium borohydride, lithium aluminum hydride, and the like.
  • suitable reducing agent such as sodium borohydride, lithium aluminum hydride, and the like.
  • the solvent used depends on the type of reducing agent. For example, when sodium borohydride is used the reaction is carried out in an alcoholic organic solvent such as methanol, ethanol, and the like.
  • Optically enriched compound of formula 15 can be obtained by reduction of the corresponding halogenated acetophenone with a suitable reducing agent such as catecholborane or BH 3 -DMS complex in the presence of a suitable catalyst such as (S) or (i?)-methyl CBS oxazaborolidine catalyst or (S) or (R)-a,a -diphenyl-2-pyrrolidine-methanol in the presence of BBN to provide chiral alcohol which is then converted to compound 13 as described above.
  • a suitable catalyst such as (S) or (i?)-methyl CBS oxazaborolidine catalyst or (S) or (R)-a,a -diphenyl-2-pyrrolidine-methanol in the presence of BBN to provide chiral alcohol which is then converted to compound 13 as described above.
  • Compounds of formula 14 are either commercially available or they can be prepared by methods well known in the art.
  • Removal of the carboxy protecting group from a compound of formula 15 where R' is a protecting group provides a compound of formula 16.
  • the conditions used to remove the carboxy protecting group depend on the nature of the carboxy protecting group. For example, if R' is allcyl, it is removed under basic hydrolysis reaction conditions utilizing aqueous base such as aqueous lithium hydroxide, sodium hydroxide, and the like in an alcoholic solvent such as methanol, ethanol, and the like. Additionally, if the R z group in compound 14 is a precursor group to R 5 , it can be converted to R 5 prior or after the ester hydrolysis step.
  • Compound 15 (where R' is hydrogen) or 16 is then converted to an activated acid derivative 17 (X is a leaving group) and which upon reaction with an aminoacetonitrile compound of formula 5 provides a compound of Formula (I) when R z is R 5 or a precursor compound to (I) when R z is a precursor group to R 5 .
  • the activated acid derivative can be prepared and then reacted with compound 5 in a stepwise manner or the activated acid derivative can be generated in situ in the presence of compound 5.
  • the activated acid is acid halide it is first prepared by reacting 16 with a halogenating agent such as thionyl chloride, oxalyl chloride and the like and then reacted with compound 5.
  • the activated acid derivative is generated in situ by reacting compound 16 and 5 in the presence of a suitable coupling agent e.g., benzotriazole- 1 -yloxytrispyrrolidinophosphonium hexafluorophosphate (PyBOP®), O-benzotriazol- 1 -yl-iV,iV,iV',iV'-tetramethyl-uronium hexafluorophosphate (HBTU), (9-(7-azabenzotriazol- 1 -yl)- 1 , 1 ,3 ,3 -tetramethyl-uronium hexafluorophosphate (HATU), l-(3-dimethylaminopropyl)-3-ethylcarbodiimide hydrochloride (EDC), 1,3-dicyclohexyl- carbodiimide (DCC), an the like, optionally in the presence of 1-hydroxybenzotriazole (HOBT), and in the presence of
  • Suitable reaction solvents are inert organic solvents such as halogenated organic solvents (e.g., methylene chloride, chloroform, and the like), acetonitrile, ⁇ f-dimethylformamide, ethereal solvents such as tetrahydrofuran, dioxane, and the like.
  • the activated acid can be reacted with CR 1 R 2 CNH 2 )CONH 2 where R 1 and R 2 are as described in the Summary of the Invention, followed by conversion of the -CONH 2 group to the cyano group by methods well known in the art.
  • R z is a precursor group to R 5 , it is converted to R 5 group to provide a compound of Formula (I) e.g, conversion of — alkylene-S-NR ⁇ R 12 to -alkylene-SO 2 -NR ⁇ R 12 under oxidation reaction conditions.
  • Amines of formula NHR 11 R 12 are either commercially available or they can be prepared by methods known in the art.
  • 1-cyclopropylpiperazine was prepared according to Gillaspy, M. A.; Lefker, B. A.; Hada, W. A.; Hoover, DJ Tetrahedron Lett. 1995, 36, 7399-7402.
  • Other cyclic amines can be prepared from commercially available starting materials.
  • analogs of piperazine can be prepared from 1-tert- butoxycarbonylpiperazine or 1-benzyloxy-carbonylpiperazine utilizing procedures well known in the art.
  • acylation of the 4-position can be performed by treatment with an acyl chloride (e.g.
  • benzoyl chloride) or sulfonylation can be achieved by treatment with a sulfonyl chloride (e.g. methane sulfonyl chloride) in the presence of triethylamine or diisopropylethylamine in a suitable solvent such as, but not limited to, methylene chloride.
  • Urea formation was achieved by treatment with an isocyanate (e.g. isopropylisocyanate) in a suitable solvent such as methylene chloride.
  • Alkylation was achieved using alkyl electrophiles bearing a suitable leaving group such as halide, tosylate, or triflate (e.g.
  • 2,2,2- trifluoroethyl trifluoromethanesulfonate prepared by the treatment of 2,2,2-trifuoroethanol with triflic anhydride in the presence of diisopropylethylamine in methylene chloride) in a suitable solvent such as methylene chloride or diethyl ether in the presence of triethylamine or diisopropylamine if necessary.
  • Alkylation can also be achieved via reductive animation using a suitable aldehyde in the presence of an acid catalyst and sodium cyanoborohydride in an acceptable solvent such as methanol.
  • Removal of the tert-butyloxycarbonyl protection group can be achieved using trifluoroacetic acid in methylene chloride to produce the trifluoroacetate salt or 4 M hydrochloric acid in dioxane (Aldrich) to produce the HCl salt after solvent removal.
  • the benzyloxycarbonyl group can be removed using 30% hydrobromic acid in acetic acid (Aldrich) in methylene chloride or by hydrogenation utilizing 10% Pd/C under an atmosphere of hydrogen gas in a suitable solvent such as ethanol.
  • a compound of Formula (I) can be converted to other compounds of Formula (I). For example:
  • a compound of Formula (I) containing a hydroxy group may be prepared by de- alkylation/benzylation of an alkoxy/benzyloxy substituent; those containing an acid group, by hydrolysis of an ester group; and those containing a cyano, by displacement of a bromine atom on the corresponding compounds of Formula (I).
  • a compound of Formula (I) containing a cyano group can be converted to a corresponding carboxy containing compound by hydrolysis of the cyano group.
  • the carboxy group in turn, can be converted to an ester group.
  • a compound of Formula (I) can be prepared as a pharmaceutically acceptable acid addition salt by reacting the free base form of the compound with a pharmaceutically acceptable inorganic or organic acid.
  • a pharmaceutically acceptable base addition salt of a compound of Formula (I) can be prepared by reacting the free acid form of the compound with a pharmaceutically acceptable inorganic or organic base.
  • inorganic and organic acids and bases suitable for the preparation of the pharmaceutically acceptable salts of compounds of Formula (I) are set forth in the definitions section of this Application.
  • the salt forms of the compounds of Formula (I) can be prepared using salts of the starting materials or intermediates.
  • the free acid or free base forms of the compounds of Formula (I) can be prepared from the corresponding base addition salt or acid addition salt form.
  • a compound of Formula (I) in an acid addition salt form can be converted to the corresponding free base by treating with a suitable base (e.g., ammonium hydroxide solution, sodium hydroxide, and the like).
  • a suitable base e.g., ammonium hydroxide solution, sodium hydroxide, and the like.
  • a compound of Formula (I) in a base addition salt form can be converted to the corresponding free acid by treating with a suitable acid (e.g., hydrochloric acid, etc).
  • JV-oxides of compounds of Formula (I) can be prepared by methods known to those of ordinary skill in the art.
  • iV-oxides can be prepared by treating an unoxidized form of the compound of Formula (I) with an oxidizing agent (e.g., trifluoroperacetic acid, permaleic acid, perbenzoic acid, peracetic acid,
  • an oxidizing agent e.g., trifluoroperacetic acid, permaleic acid, perbenzoic acid, peracetic acid,
  • the JV ⁇ oxides of the compounds of Formula (I) can be prepared from the iV-oxide of an appropriate starting material.
  • Compounds of Formula (I) in unoxidized form can be prepared from iV-oxides of compounds of Formula (I) by treating with a reducing agent (e.g., sulfur, sulfur dioxide, triphenyl phosphine, lithium borohydride, sodium borohydride, phosphorus trichloride, tribromide, or the like) in a suitable inert organic solvent (e.g., acetonitrile, ethanol, aqueous dioxane, or the like) at 0 to 8O 0 C.
  • a reducing agent e.g., sulfur, sulfur dioxide, triphenyl phosphine, lithium borohydride, sodium borohydride, phosphorus trichloride, tribromide, or the like
  • a suitable inert organic solvent e.g., acetonitrile, ethanol, aqueous dioxane, or the like
  • Prodrug derivatives of the compounds of Formula (I) can be prepared by methods known to those of ordinary skill in the art (e.g., for further details see Saulnier et ⁇ /.(1994), Bioorganic and Medicinal Chemistry Letters, Vol. 4, p. 1985).
  • appropriate prodrugs can be prepared by reacting a non-derivatized compound of Formula (I) with a suitable carbamylating agent (e.g., l j l-acyloxyalkylcarbonochloridatejp ⁇ ra-nitrophenyl carbonate, or the like).
  • Hydrates of compounds of the present invention may be conveniently prepared or formed during the process of the invention, as solvates (e.g. hydrates). Hydrates of compounds of the present invention may be conveniently prepared by recrystallisation from an aqueous/organic solvent mixture, using organic solvents such as dioxin, tetrahydrofuran or methanol.
  • Compounds of Formula (I) can be prepared as their individual stereoisomers by reacting a racemic mixture of the compound with an optically active resolving agent to form a pair of diastereoisomeric compounds, separating the diastereomers and recovering the optically pure enantiomer. While resolution of enantiomers can be carried out using covalent diasteromeric derivatives of compounds of Formula (I), dissociable complexes are preferred (e.g., crystalline diastereoisomeric salts). Diastereomers have distinct physical properties (e.g., melting points, boiling points, solubilities, reactivity, etc.) and can be readily separated by taking advantage of these dissimilarities.
  • the diastereomers can be separated by chromatography or, preferably, by separation/resolution techniques based upon differences in solubility.
  • the optically pure enantiomer is then recovered, along with the resolving agent, by any practical means that would not result in racemization.
  • a more detailed description of the techniques applicable to the resolution of stereoisomers of compounds from their racemic mixture can be found in Jean Jacques Andre Collet, Samuel H. Wilen, Enantiomers, Racemates and Resolutions, John Wiley & Sons, Inc. (1981).
  • Monoclonal antibodies can be prepared using standard techniques well known in the art such as by the method of Kohler and Milstein, Nature 1975, 256:495, or a modification thereof, such as described by Buck et al. 1982, In Vitro 18:377.
  • a mouse or rat is immunized with the MenB PS derivative conjugated to a protein carrier, boosted and the spleen (and optionally several large lymph nodes) removed and dissociated into single cells.
  • the spleen cells may be screened (after removal of non- specifically adherent cells) by applying a cell suspension to a plate or well coated with the antigen.
  • B-cells expressing membrane-bound immunoglobulin specific for the antigen, will bind to the plate, and will not be rinsed away with the rest of the suspension. Resulting B- cells, or all dissociated spleen cells, are then induced to fuse with myeloma cells to form hybridomas.
  • Representative murine myeloma lines for use in the hybridizations include those available from the American Type Culture Collection (ATCC).
  • Chimeric antibodies composed of human and non-human amino acid sequences may be formed from the mouse monoclonal antibody molecules to reduce their immunogenicity in humans (Winter et al. Nature 1991 349:293; Lobuglio et al. Proc. Nat. Acad. ScL USA 1989 86:4220; Shaw et al. J Immunol. 1987 138:4534; and Brown et al. Cancer Res. 1987 47:3577; Riechmann et al. Nature 1988 332:323; Verhoeyen et al. Science 1988 239:1534; and Jones et al. Nature 1986 321:522; EP Publication No.519, 596, published Dec. 23, 1992; and U.K. Patent Publication No. GB 2,276,169, published Sep. 21, 1994).
  • Antibody molecule fragments e.g., F(ab') 2 , FV, and sFv molecules, that are capable of exhibiting immunological binding properties of the parent monoclonal antibody molecule can be produced using known techniques. Inbar et al. Proc. Nat. Acad. Sd. USA 1972 69:2659; Hochman et al. Biochem. 1976 15:2706; Ehrlich et al. Biochem. 1980 19:4091; Huston et al. Proc. Nat. Acad. ScI USA 1988 85(16):5879; and U.S. Pat. Nos. 5,091,513 and 5,132,405, and U.S. Pat. No. 4,946,778.
  • F(ab') 2 , FV, and sFv molecules that are capable of exhibiting immunological binding properties of the parent monoclonal antibody molecule can be produced using known techniques. Inbar et al. Proc. Nat. Acad. Sd. USA 1972 69:2659
  • a phage-display system can be used to expand the monoclonal antibody molecule populations in vitro. Saiki, et al. Nature 1986 324:163; Scharf et al. Science 1986 233:1076; U.S. Pat. Nos. 4,683,195 and 4,683,202; Yang et al. J. MoI. Biol. 1995 254:392; Barbas, III et al Methods: Comp. Meth Enzymol. 1995 8:94; Barbas, III et al. Proc. Natl. Acad. Sci. USA 1991 88:7978.
  • the coding sequences for the heavy and light chain portions of the Fab molecules selected from the phage display library can be isolated or synthesized, and cloned into any suitable vector or replicon for expression.
  • Any suitable expression system can be used, including, for example, bacterial, yeast, insect, amphibian and mammalian systems. Expression systems in bacteria include those described in Chang et al. Nature 1978 275:615, Goeddel et al Nature 1979 281:544, Goeddel et al. Nucleic Acids Res. 1980 8:4057, European Application No. EP 36,776, U.S. Pat. No. 4,551,433, deBoer et al Proc. Natl. Acad. Sci. USA 1983 80:21-25, and Siebenlist et al Cell 1980 20:269.
  • Expression systems in yeast include those described in Hinnen et al Proc. Natl.
  • Mammalian expression can be accomplished as described in Dijkema et al EMBO J. 1985 4:761, Gorman et al Proc. Natl. Acad. Sci. USA 1982 79:6777, Boshart et al Cell 1985 41:521, and U.S. Pat. No. 4,399,216. Other features of mammalian expression can be facilitated as described in Ham et al. Meth. Enz. 1979 58:44, Barnes et al. Anal. Biochem. 1980 102:255, U.S. Pat. Nos. 4,767,704, 4,657,866, 4,927,762, 4,560,655 and Reissued U.S. Pat. No. RE 30,985, and in International Publication Nos. WO 90/103430, WO 87/00195. [0178] The production of recombinant adenoviral vectors are described in U.S. Pat. No. 6,485,958.
  • Botulinum toxin type A can be obtained by establishing and growing cultures of Clostridium botulinum in a fermenter and then harvesting and purifying the fermented mixture in accordance with known procedures.
  • the compounds of the invention are selective inhibitors of cysteine proteases such as cathepsin S, K, B, and/or F, and in particular cathepsin S, and accordingly are useful for treating diseases in which cysteine protease activity contributes to the pathology and/or symptomatology of the disease.
  • the compounds of the invention are useful in treating autoimmune disorders, including, but not limited to, juvenile onset diabetes, psoriasis, multiple sclerosis, pemphigus vulgaris, Graves' disease, myasthenia gravis, systemic lupus erythemotasus, rheumatoid arthritis and Hashimoto's thyroiditis, allergic disorders, including, but not limited to, asthma, allogeneic immune responses, including, but not limited to, organ transplants or tissue grafts and endometriosis.
  • autoimmune disorders including, but not limited to, juvenile onset diabetes, psoriasis, multiple sclerosis, pemphigus vulgaris, Graves' disease, myasthenia gravis, systemic lupus erythemotasus, rheumatoid arthritis and Hashimoto's thyroiditis
  • allergic disorders including, but not limited to, asthma, allogeneic immune responses, including, but not limited to, organ transplants or tissue graf
  • Cathepsin S is also implicated in disorders involving excessive elastolysis, such as chronic obstructive pulmonary disease (e.g., emphysema), bronchiolitis, excessive airway elastolysis in asthma and bronchitis, pneumonities and cardiovascular disease such as plaque rupture and atheroma.
  • Cathepsin S is implicated in fibril formation and, therefore, inhibitors of cathepsins S are of use in treatment of systemic amyloidosis.
  • cysteine protease inhibitory activities of the compounds of Formula (I) can be determined by methods known to those of ordinary skill in the art. Suitable in vitro assays for measuring protease activity and the inhibition thereof by test compounds are known. Typically, the assay measures protease-induced hydrolysis of a peptide-based substrate. Details of assays for measuring protease inhibitory activity are set forth in Biological Examples 1-5, infra. ADMINISTRATION AND PHARMACEUTICAL COMPOSITIONS [0184] In general, compounds of Formula (I) will be administered in therapeutically effective amounts via any of the usual and acceptable modes known in the art, either singly or in combination with one or more therapeutic agents.
  • a therapeutically effective amount may vary widely depending on the severity of the disease, the age and relative health of the subject, the potency of the compound used and other factors.
  • therapeutically effective amounts of a compound of Formula (I) may range from about 10 micrograms per kilogram body weight ( ⁇ g/kg) per day to about 100 milligram per kilogram body weight (mg/kg) per day, typically from about 100 ⁇ g/kg/day to about 10 mg/kg/day. Therefore, a therapeutically effective amount for an 80 kg human patient may range from about 1 mg/day to about 8 g/day, typically from about 1 mg/day to about 800 mg/day.
  • a therapeutically effective amount of a compound of Formula (I) for treating a given disease may range from about 10 micrograms per kilogram body weight ( ⁇ g/kg) per day to about 100 milligram per kilogram body weight (mg/kg) per day, typically from about 100 ⁇ g/kg/day to about 10 mg/kg/day. Therefore, a therapeutically effective amount for an 80 kg human patient may range from about
  • compositions can take the form of tablets, pills, capsules, semisolids, powders, sustained release formulations, solutions, suspensions, elixirs, aerosols, or any other appropriate composition and are comprised of, in general, a compound of Formula (I) in combination with at least one pharmaceutically acceptable excipient.
  • Acceptable excipients are non-toxic, aid administration, and do not adversely affect the therapeutic benefit of the active ingredient.
  • excipient may be any solid, liquid, semisolid or, in the case of an aerosol composition, gaseous excipient that is generally available to one of skill in the art.
  • Solid pharmaceutical excipients include starch, cellulose, talc, glucose, lactose, sucrose, gelatin, malt, rice, flour, chalk, silica gel, magnesium stearate, sodium stearate, glycerol monostearate, sodium chloride, dried skim milk, and the like.
  • Liquid and semisolid excipients may be selected from water, ethanol, glycerol, propylene glycol and various oils, including those of petroleum, animal, vegetable or synthetic origin (e.g., peanut oil, soybean oil, mineral oil, sesame oil, and the like).
  • Preferred liquid carriers, particularly for injectable solutions include water, saline, aqueous dextrose and glycols.
  • a composition of a compound of Formula (I) for treating a given disease will comprise from 0.01 %w to 90%w, preferably 5%w to 50%w, of active ingredient with the remainder being the excipient or excipients.
  • the pharmaceutical composition is administered in a single unit dosage form for continuous treatment or in a single unit dosage form ad libitum when relief of symptoms is specifically required.
  • Representative pharmaceutical formulations containing a compound of Formula (I) are described below.
  • the present invention is further exemplified, but not limited by, the following examples that illustrate the preparation of compounds of Formula (I) (Examples) and intermediates (References) according to the invention.
  • the reaction mixture was diluted with ethyl acetate and washed with a saturated brine solution.
  • the organic layer was dried over magnesium sulfate, filtered and concentrated to provide a solid.
  • the solid was suspended in hexanes and filtered off.
  • the hexanes filtrate containing the desired product was concentrated and the residue subjected to flash chromatography (10 hexanes: 1 ethylacetate) to provide the title compound as colorless oil (2.2g, 87% yield).
  • the ratio of enantiomers was determined to be 95:5 by chiral HPLC (Chiralcel OD column, 95 hexanes: 5 isopropanol mobile phase. Ret. time major product 6.757 min. Ret. time minor isomer 8.274 min.).
  • Step 2 A solution of 1-aminocyclopropanecarboxylic acid chlorohydrate (3.6 g, 26.2 mmol) in MeOH (100 mL), containing potassium carbonate (4.0 g, 28.94 mmol) was stirred at room temperature for 48 h. After filtration, MeOH was removed under reduced pressure to yield 1-aminocyclopropanecarboxylic acid (2.64 g) which was used in the next step without further purification.
  • Step l To a solution of (2-2ydroxy-l,l-dimethylethyl)-carbamic acid tert-hutyl ester (284 mg, 1.5 mmol) in CH 2 Cl 2 (5 mL) was added at 0 0 C Dess-Martin periodane (763 mg, 1.8 mmol). After 1.5 h, a solution of 0.26M Na 2 S 2 O 3 in saturated NaHCO 3 (6 mL) was added and the resulting mixture was stirred for 15 min. The layers were separated and the aqueous layer was extracted with CH 2 Cl 2 .
  • Step 3 To a solution of acetic acid 2-fert-butoxycarbonylamino- 1 -cyclopropylcarbamoyl-2- methylpropyl ester in MeOH (10 mL) was added 10% NaOH (1.5 mL). The reaction mixture was stirred at room temperature for 3 h and then acidified with IN Hydrochloric acid to pH 7. The reaction mixture was extracted with EtOAc. The organic layer was dried (Na 2 SO 4 ) concentrated to yield (2-cyclopropylcarbamoyl-2-hydroxy-l,l-dimethylethyl)-carbamic acid tert-hutyl ester which was used in the next step without further purification.
  • 3-Amino-iV-benzyl-2-hydroxy-3-methylbutyramide was made by the procedure described for 3-amino-N-cyclopropyl-2-hydroxy-3-methylbutyramide by substituting cyclopropyl isocyanide with benzyl isocyanide.
  • Example 1 Cathepsin B Assay Solutions of test compounds in varying concentrations were prepared in 10 ⁇ L of dimethyl sulfoxide (DMSO) and then diluted into assay buffer (40 ⁇ L, comprising: N,N- bis(2-hydroxyethyl)-2-aminoethanesulfonic acid (BES), 50 mM (pH 6); polyoxyethylenesorbitan monolaurate, 0.05%; and dithiothreitol (DTT), 2.5 mM). Human cathepsin B (0.025 pMoles in 25 ⁇ L of assay buffer) was added to the dilutions.
  • DMSO dimethyl sulfoxide
  • assay buffer 40 ⁇ L, comprising: N,N- bis(2-hydroxyethyl)-2-aminoethanesulfonic acid (BES), 50 mM (pH 6); polyoxyethylenesorbitan monolaurate, 0.05%; and dithiothreitol (DTT),
  • the assay solutions were mixed for 5-10 seconds on a shaker plate, covered and incubated for 30 min at room temperature.
  • Z-FR-AMC (20 nMoles in 25 ⁇ L of assay buffer) was added to the assay solutions and hydrolysis was followed spectrophotometrically at ( ⁇ 460 nm) for 5 min.
  • Apparent inhibition constants (K 1 ) were calculated from the enzyme progress curves using standard mathematical models.
  • Example 2 Cathepsin K Assay [0214] Solutions of test compounds in varying concentrations were prepared in 10 ⁇ L of dimethyl sulfoxide (DMSO) and then diluted into assay buffer (40 ⁇ L, comprising: MES 5 50 mM (pH 5.5); EDTA, 2.5 mM; and DTT, 2.5 mM). Human cathepsin K (0.0906 pMoles in 25 ⁇ L of assay buffer) was added to the dilutions. The assay solutions were mixed for 5-10 seconds on a shaker plate, covered and incubated for 30 min at room temperature.
  • DMSO dimethyl sulfoxide
  • test compounds in varying concentrations were prepared in 10 ⁇ L of dimethyl sulfoxide (DMSO) and then diluted into assay buffer (40 ⁇ L, comprising: MES, 50 niM (pH 5.5); EDTA, 2.5 niM; and DTT, 2.5 mM).
  • assay buffer 40 ⁇ L, comprising: MES, 50 niM (pH 5.5); EDTA, 2.5 niM; and DTT, 2.5 mM).
  • Human cathepsin L (0.05 pMoles in 25 ⁇ L of assay buffer) was added to the dilutions.
  • the assay solutions were mixed for 5-10 seconds on a shaker plate, covered and incubated for 30 min at room temperature.
  • test compounds in varying concentrations were prepared in 10 ⁇ L of dimethyl sulfoxide (DMSO) and then diluted into assay buffer (40 ⁇ L, comprising: MES, 50 mM (pH 6.5); EDTA, 2.5 mM; and NaCl, 100 mM); ⁇ -mercaptoethanol, 2.5 mM; and BSA, 0.00%.
  • Assay buffer 40 ⁇ L, comprising: MES, 50 mM (pH 6.5); EDTA, 2.5 mM; and NaCl, 100 mM); ⁇ -mercaptoethanol, 2.5 mM; and BSA, 0.00%.
  • Human cathepsin S (0.05 pMoles in 25 ⁇ L of assay buffer) was added to the dilutions.
  • the assay solutions were mixed for 5-10 seconds on a shaker plate, covered and incubated for 30 min at room temperature.
  • test compounds in varying concentrations were prepared in 10 ⁇ L of dimethyl sulfoxide (DMSO) and then diluted into assay buffer (40 ⁇ L, comprising: MES, 50 mM (pH 6.5); EDTA, 2.5 mM; and NaCl, 100 raM); DTT, 2.5 mM; and BSA, 0.01%.
  • Assay buffer 40 ⁇ L, comprising: MES, 50 mM (pH 6.5); EDTA, 2.5 mM; and NaCl, 100 raM); DTT, 2.5 mM; and BSA, 0.01%.
  • Human cathepsin F 0.1 pMoles in 25 ⁇ L of assay buffer was added to the dilutions.
  • the assay solutions were mixed for 5-10 seconds on a shaker plate, covered and incubated for 30 min at room temperature.
  • Example 1 Representative pharmaceutical formulations Containing a Compound of Formula (I)
  • Citric Acid Monohydrate 1.05 mg Sodium Hydroxide 0.18 mg

Abstract

The present invention is directed to compounds that are inhibitors of cysteine proteases, in particular, cathepsins B, K, L, F, and S and are therefore useful in treating diseases mediated by these proteases. The present invention is directed to pharmaceutical compositions comprising these compounds and processes for preparing them.

Description

ALPHA KETOAMIDE COMPOUNDS AS CYSTEINE PROTEASE
INHIBITORS
[0001] This application claims the benefit of Provisional Patent Application No. 60/664,041 , filed March 21 , 2005 the content of which is incorporated herein by reference.
STATEMENT AS TO RIGHTS TO INVENTIONS MADE UNDER FEDERALLY SPONSORED RESEARCH OR DEVELOPMENT
[0002] NOT APPLICABLE
REFERENCE TO A "SEQUENCE LISTING," A TABLE, OR A COMPUTER PROGRAM LISTING APPENDIX SUBMITTED ON A COMPACT DISK.
[0003] NOT APPLICABLE
FIELD OF THE INVENTION
[0004] The present invention is directed to compounds that are inhibitors of cysteine proteases, in particular, cathepsins B, K, L, F, and S and are therefore useful in treating diseases mediated by these proteases. The present invention is also directed to pharmaceutical compositions comprising these compounds and processes for preparing them.
STATE OF THE ART
[0005] Cysteine proteases represent a class of peptidases characterized by the presence of a cysteine residue in the catalytic site of the enzyme. Cysteine proteases are associated with the normal degradation and processing of proteins. The aberrant activity of cysteine proteases, e.g., as a result of increased expression or enhanced activation, however, may have pathological consequences. In this regard, certain cysteine proteases are associated with a number of disease states, including arthritis, muscular dystrophy, inflammation, tumor invasion, glomerulonephritis, malaria, periodontal disease, metachromatic leukodystrophy and others. For example, increased cathepsin B levels and redistribution of the enzyme are found in tumors; thus, suggesting a role for the enzyme in tumor invasion and metastasis. In addition, aberrant cathepsin B activity is implicated in such disease states as rheumatoid arthritis, osteoarthritis, Pneumocystis carinii, acute pancreatitis, inflammatory airway disease and bone and joint disorders.
[0006] The prominent expression of cathepsin K in osteoclasts and osteoclast-related multinucleated cells and its high collagenolytic activity suggest that the enzyme is involved in osteoclast-mediated bone resorption and, hence, in bone abnormalities such as occurs in osteoporosis. In addition, cathepsin K expression in the lung and its elastinolytic activity suggest that the enzyme plays a role in pulmonary disorders as well.
[0007] Cathepsin L is implicated in normal lysosomal proteolysis as well as in several disease states, including, but not limited to, metastasis of melanomas. Cathepsin S is implicated in Alzheimer's disease and certain autoimmune disorders, including, but not limited to juvenile onset diabetes, multiple sclerosis, pemphigus vulgaris, Graves' disease, myasthenia gravis, systemic lupus erythemotasus, rheumatoid arthritis, neuropathic pain, and Hashimoto's thyroiditis. In addition, cathepsin S is implicated in: allergic disorders, including, but not limited to asthma; and allogeneic immune reponses, including, but not limited to, rejection of organ transplants or tissue grafts.
[0008] In view of the number of diseases wherein it is recognized that an increase in cysteine protease activity contributes to the pathology and/or symptomatology of the disease, molecules which inhibit the activity of this class of enzymes, in particular molecules which inhibit cathepsins B, K, L, F, and/or S, will therefore be useful as therapeutic agents.
SUMMARY OF THE INVENTION
[0009] In one aspect, this invention is directed to a compound of Formula (I):
where: R1 is hydrogen or alkyl;
R2 is cycloalkyl, cycloalkylalkyl, aralkyl, heteroaryl, or heteroaralkyl optionally substituted with one or two substitutents independently selected from alkyl, alkoxy, or halo; R3 is hydrogen, alkyl or alkoxyalkyl; R4 is alkyl; or R3 and R4 together with the carbon atom to which they are attached form cycloalkylene optionally substituted with one to four fluoro or heterocycloalkylene optionally substituted with alkyl, alkoxyalkyl, hydroxyalkyl, acyl, cycloalkyl, cycloalkylalkyl, or haloalkyl; R5 is -alkylene-SO2NRπR12 where R11 is hydrogen, alkyl, haloalkyl, alkoxyalkyl, hydroxyalkyl, aminoalkyl, carboxyalkyl, alkoxycarbonylalkyl, acyl, cycloalkyl, cycloalkylalkyl, aryl, heteroaryl, aralkyl, heteroaralkyl, heterocycloalkylalkyl, acylalkyl, or heterocycloalkylaminocarbonyl and R12 is hydrogen, alkyl, hydroxyalkyl, alkoxyalkyl, aryl, heteroaryl, aralkyl, heteroaralkyl, cycloalkyl, cycloalkylalkyl, heterocycloalkyl, or heterocycloalkylalkyl); or R11 and R12 together with the nitrogen atom to which they are attached form heterocycloamino or bridged azabicyclic ring, wherein the aromatic or alicyclic ring in R5 is optionally substituted with one, two, or three Ra independently selected from alkyl, haloalkyl, alkoxy, hydroxy, haloalkoxy, cyano, halo, carboxy or alkoxycarbonyl; or optionally substituted with one or two Rb independently selected from hydrogen, alkyl, haloalkyl, alkoxy, hydroxy, haloalkoxy, halo, carboxy, or alkoxycarbonyl and one Rc selected from cyano, hydroxyalkyl, alkoxyalkyl, aminoalkyl, aryl, heteroaryl, aralkyl, heteroaralkyl, cycloalkyl, cycloalkylalkyl, heterocycloalkyl, heterocycloalkylalkyl, acyl, acylalkyl, alkoxycarbonyl, aryloxycarbonyl, aralkyloxycarbonyl, heteroaryloxycarbonyl, heteroaralkyloxycarbonyl, heterocycloalkyloxycarbonyl, heterocycloalkylalkyloxycarbonyl, cycloalkyloxycarbonyl, cycloalkylalkyloxycarbonyl, aryloxy, heteroaryloxy, aralkyloxy, heteroaralkyloxy, aminocarbonyl, aminosulfonyl, or -SO2R13 (where R13 is alkyl, cycloalkyl, aryl, heteroaryl, or heterocycloalkyl); and further wherein the aromatic or alicyclic ring in Rc is optionally substituted with one, two, or three Rd independently selected from alkyl, haloalkyl, alkoxy, hydroxy, haloalkoxy, halo, cyano, -CONH2, alkylaminocarbonyl, dialkylaminocarbonyl, alkylcarbonyl, alkylsulfonyl, or alkylsulfonylamino; R6 is haloalkyl;
R7 is hydrogen, alkyl, or haloalkyl; and
R8 is hydrogen, alkyl, haloalkyl, cycloalkyl, aryl, heteroaryl, heterocycloalkyl attached via a carbon atom wherein the aromatic or alicyclic ring in R is optionally substituted with one, two, or three Re independently selected from alkyl, halo, haloalkyl, hydroxy, alkoxy, haloalkoxy, alkoxycarbonyl, carboxy, cyano, alkylcarbonyl, alkylsulfonyl, alkylsulfonylamino, alkylaminocarbonyl, dialkylaminocarbonyl, or aminosulfonyl; or a pharmaceutically acceptable salts thereof. [0010] In a second aspect, this invention is directed to a pharmaceutical composition comprising a compound of Formula (I) or a pharmaceutically acceptable salt thereof in admixture with one or more suitable excipients.
[0011] In a third aspect, this invention is directed to a method for treating a disease in an animal mediated by cysteine proteases, in particular cathepsin S, which method comprises administering to the animal a pharmaceutical composition comprising a therapeutically effective amount of a compound of Formula (I) or a pharmaceutically acceptable salt thereof in admixture with one or more suitable excipients.
[0012] In a fourth aspect, this invention is directed to processes for preparing compounds of Formula (I).
[0013] In a fifth aspect, this invention is directed to a method of treating a patient undergoing a therapy wherein the therapy causes an immune response, preferably a deleterious immune response, in the patient comprising administering to the patient a compound of Formula (I) or a pharmaceutically acceptable salt thereof. Preferably, the immune response is mediated by MHC class II molecules. The compound of this invention can be administered prior to, simultaneously, or after the therapy. Preferably, the therapy involves treatment with a biologic. Preferably, the therapy involves treatment with a small molecule.
[0014] Preferably, the biologic is a protein, preferably an antibody, more preferably a monoclonal antibody. More preferrably, the biologic is Remicade®, Refacto®, Referon-A®, Factor VIII, Factor VII, Betaseron®, Epogen®, Enbrel®, Interferon beta, Botox®, Fabrazyme®, Elspar®, Cerezyme®, Myobloc®, Aldurazyme®, Verluma®, Interferon alpha, Humira®, Aranesp®, Zevalin® or OKT3.
[0015] Preferably, the treatment involves use of heparin, low molecular weight heparin, procainamide or hydralazine.
[0016] In a sixth aspect, this invention is directed to a method of treating immune response in an animal that is caused by administration of a biologic to the animal which method comprises administering to the animal in need of such treatment a therapeutically effective amount of a compound of Formula (I) or a pharmaceutically acceptable salt thereof. [0017] In a seventh aspect, this invention is directed to a method of conducting a clinical trial for a biologic comprising administering to an individual participating in the clinical trial a compound of Formula (I) or a pharmaceutically acceptable salt thereof with the biologic.
[0018] In an eigth aspect, this invention is directed to a method of prophylactically treating a patient undergoing treatment with a biologic with a compound of Formula (I) or a pharmaceutically acceptable salt thereof to treat the immune response caused by the biologic in the patient.
[0019] In a ninth aspect, this invention is directed to a method of determing the loss in the efficacy of a biologic in an animal due to the immune response caused by the biologic comprising administering the biologic to the animal in the presence and absence of a compound of Formula (I) or a pharmaceutically acceptable salt thereof.
[0020] In a tenth aspect, this invention is directed to a method of improving efficacy of a biologic in an animal comprising administering the biologic to the animal with a compound of Formula (I) or a pharmaceutically acceptable salt thereof.
[0021] In an eleventh aspect, this invention is directed to the use of a compound of Formula (I) or a pharmaceutically acceptable salt thereof for the manufacture of a medicament. Preferably, the medicament is for use in the treatment of a disease mediated by Cathepsin S.
[0022] In a twelfth aspect, this invention is directed to the use of a compound of Formula (I) or a pharmaceutically acceptable salt thereof for the manufacture of a medicament for combination therapy with a biologic, wherein the compound of this invention treats the immune response caused by the biologic. Preferably, the compound(s) of the invention is administered prior to the administration of the biological agent. Preferably, the compound(s) of the invention is administered concomitantly with the biological agent. Preferably, the compound(s) of the invention is administered after the administration of the biological agent.
DETAILED DESCRIPTION OF THE INVENTION Definitions:
[0023] Unless otherwise stated, the following terms used in the specification and claims are defined for the purposes of this Application and have the following meanings. [0024] "Alicyclic" means a moiety characterized by arrangement of the carbon atoms in closed non-aromatic ring structures e.g., cycloalkyl and heterocycloalkyl rings as defined herein.
[0025] "Alkyl" represented by itself means a straight or branched, saturated aliphatic radical containing one to eight carbon atoms, unless otherwise indicated e.g., alkyl includes methyl, ethyl, propyl, isopropyl, butyl, sec-butyl, isobutyl, tert-butyl, and the like.
[0026] "Alkylene", unless indicated otherwise, means a straight or branched, saturated aliphatic, divalent radical having the number of one to six carbon atoms, e.g., methylene (-CH2-), ethylene (-CH2CH2-), trimethylene (-CH2CH2CH2-), tetramethylene (-CH2CH2CH2CH2-) 2-methyltetramethylene (-CH2CH(CH3)CH2CH2-), pentamethylene (-CH2CH2CH2CH2CH2-), and the like.
[0027] "Alkylsulfonyl" means -SO2R radical where R is alkyl as defined herein e.g., methylsulfonyl, ethylsulfonyl, and the like.
[0028] "Alkylsulfonylamino" refers to a -NHSO2R radical where R is an alkyl group as defined above e.g., methylsulfonylamino, ethylsulfonylamino, and the like.
[0029] " Alkoxy" refers to a -OR radical where R is an alkyl group as defined above e.g., methoxy, ethoxy, and the like.
[0030] "Alkoxyalkyl" means a linear monovalent hydrocarbon radical of one to six carbon atoms or a branched monovalent hydrocarbon radical of three to six carbons substituted with at least one alkoxy group, preferably one or two alkoxy groups, as defined above, e.g., 2- methoxy-ethyl, 1-, 2-, or 3-methoxypropyl, 2-ethoxyethyl, and the like.
[0031] "Alkoxycarbonyl" refers to a -C(O)OR radical where R is an alkyl group as defined above e.g., methoxycarbonyl, ethoxycarbonyl, and the like.
[0032] " Alkoxycarbonylalkyl" means a linear monovalent hydrocarbon radical of one to six carbon atoms or a branched monovalent hydrocarbon radical of three to six carbons substituted with at least one, preferably one or two, alkoxycarbonyl group(s) as defined herein e.g., methoxycarboxymethyl, ethoxycarbonylmethyl, methoxycarbonylethyl, and the like.
[0033] " Aminoalkyl" means a linear monovalent hydrocarbon radical of one to six carbon atoms or a branched monovalent hydrocarbon radical of three to six carbons substituted with at least one, preferably one or two, -NRR' where R is hydrogen, alkyl, acyl, hydroxyalkyl, alkoxyalkyl, carboxyalkyl, alkoxycarbonylalkyl, aryl, aralkyl, heteroaryl, heteroaralkyl or heterocycloalkylalkyl and R is hydrogen, alkyl, haloalkyl, acyl, hydroxyalkyl, alkoxyalkyl, carboxyalkyl alkoxycarbonylalkyl, aryl, aralkyl, heteroaryl, heteroaralkyl, heterocycloalkyl, heterocycloalkylalkyl, cycloalkyl, cycloalkylalkyl, aminocarbonyl, aminosulfonyl, -C(O)OR where (R is alkyl, cycloalkyl, cycloalkylalkyl, aryl, aralkyl, heteroaryl, heteroaralkyl, or heterocycloalkyl) or -SO2R (where R is alkyl, haloalkyl, cycloalkyl, aryl, heteroaryl, or heterocycloalkyl) as defined herein e.g., aininomethyl, methylaminoethyl, dimethylaniinoethyl, 1,3-diaminopropyl, acetylaminopropyl, and the like.
[0034] "Acyl" refers to a— COR radical where R is hydrogen, alkyl, cycloalkyl, cycloalkylalkyl, haloalkyl, aryl, aralkyl, heteroaryl, heteroaralkyl, or heterocycloalkyl as defined herein, e.g., formyl, acetyl, trifluoroacetyl, benzoyl, piperazin-1-ylcarbonyl, and the like. When R is alkyl it is referred to in this application as alkylcarbonyl.
[0035] " Acylalkyl" means a linear monovalent hydrocarbon radical of one to six carbon atoms or a branched monovalent hydrocarbon radical of three to six carbons substituted with at least one, preferably one or two, acyl group(s) as defined herein e.g., methylcarbonylmethyl, benκoylethyl, piperidin-1-ylcarbonylmethyl or ethyl, and the like.
[0036] "Aminocarbonyl" means -CONRR' radical where R and R' are independently selected from hydrogen, alkyl, aryl, aralkyl, heteroaryl, heteroaralkyl, or heterocycloalkylalkyl or R and R' together with the nitrogen atom to which they are attached form heterocycloamino as defined herein. When the R group is H or alkyl and R' is alkyl, such groups may be referred to in this Application as alkylaminocarbonyl and dialkylaminocarbonyl respectively and are subset of aminocarbonyl group e.g., methylaminocarbonyl or dimethylaminocarbonyl.
[0037] "Aminosulfonyl" means -SO2NRR' radical where R and R' are independently selected from hydrogen, alkyl, aryl, aralkyl, heteroaryl, heteroaralkyl, or heterocycloalkylalkyl or R and R' together with the nitrogen atom to which they are attached form heterocycloamino as defined herein.
[0038] "Animal" includes humans, non-human mammals (e.g., dogs, cats, rabbits, cattle, horses, sheep, goats, swine, deer, and the like) and non-mammals (e.g., birds, and the like). [0039] "Aromatic" refers to a moiety wherein the constituent atoms make up an unsaturated ring system, all atoms in the ring system are sp2 hybridized and the total number of pi electrons is equal to 4n+2.
[0040] "Aryl" refers to a monocyclic or fused bicyclic ring assembly containing 6 to 10 ring carbon atoms wherein each ring is aromatic e.g., phenyl or naphthyl.
[0041] "Aryloxy" refers to a — O-R radical where R is aryl as defined above e.g., phenoxy, napthyloxy, and the like.
[0042] "Aryloxycarbonyl" refers to a -C(O)OR radical where R is aryl as defined above e.g., phenyloxycarbonyl, naphthyloxycarbonyl, and the like.
[0043] "Aralkyl" refers to a -(alkylene)-R radical where R is aryl as defined above e.g., benzyl, phenethyl, and the like.
[0044] "Aralkyloxy" refers to a -O-R radical where R is aralkyl as defined above e.g., benzyloxy, phenethyloxy, and the like.
[0045] "Aralkyloxycarbonyl" refers to a -C(O)OR radical where R is aralkyl as defined above e.g., benzyloxycarbonyl, phenethyloxycarbonyl, and the like.
[0046] "Biologic" means a therapeutic agent originally derived from living organisms for the treatment or management of a disease. Examples include, but are not limited to, proteins (recombinant and plasma derived), monoclonal or polyclonal, humanized or murine antibodies, toxins, hormones, and the like. Biologies are currently available for the treatment of a variety of diseases such as cancer, rheumatoid arthritis, and hemophilia.
[0047] "Bridged azabicyclic ring" means a bridged bicylic ring containing 7 or 8 ring atoms wherein one or two ring atoms are nitrogen and the remaining ring atoms being carbon. The ring is attached to the sulfonyl group via the nitrogen atom. Representative examples include, but are not limited to, the following:
and the like.
[0048] "Carboxy" refers to -C(O)OH radical. [0049] "Carboxyalkyl" means a linear monovalent hydrocarbon radical of one to six carbon atoms or a branched monovalent hydrocarbon radical of three to six carbons substituted with at least one, preferably one or two, carboxy group(s) e.g., carboxymethyl, carboxyethyl, and the like.
[0050] "Cycloalkyl" refers to a monovalent saturated monocyclic ring containing three to eight ring carbon atoms e.g., cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, and the like.
[0051] "Cycloalkylalkyl" refers to a -(alkylene)-R radical where R is cycloalkyl as defined above e.g., cyclopropylmethyl, cyclobutylethyl, cyclobutylmethyl, and the like.
[0052] "Cycloalkyloxycarbonyl" refers to a -C(O)OR radical where R is cyccloalkyl as defined above e.g., cyclopropyloxycarbonyl, cyclopentyloxycarbonyl, and the like.
[0053] "Cycloalkylalkyloxycarbonyl" refers to a -C(O)OR radical where R is cycloalkylalkyl as defined above e.g., cyclopropylmethyloxycarbonyl, cyclopentylmethyloxycarbonyl, and the like.
[0054] "Cycloalkylene" refers to a divalent saturated monocyclic ring containing three to eight ring carbon atoms. For example, the instance wherein "R1 and R2 together with the carbon atom to which both R1 and R2 are attached form cycloalkylene" includes, but is not limited to, the following:
and the like.
[0055] "Disease" specifically includes any unhealthy condition of an animal or part thereof and includes an unhealthy condition that may be caused by, or incident to, medical or veterinary therapy applied to that animal, i.e., the "side effects" of such therapy.
[0056] "Derived" means a similar agent can be traced to.
[0057] "Deleterious immune response" means an immune response that prevents effective treatment of a patient or causes disease in a patient. As an example, dosing a patient with a murine antibody either as a therapy or a diagnostic agent causes the production of human antimouse antibodies that prevent or interfere with subsequent treatments. The incidence of antibody formation versus pure murine monoclonals can exceed 70%. {see Khazaeli, M. B. et al. J. Immunother, 1994, 15, pp 42-52; Dillman R. O. et al. Cancer Biother. 1994, 9, pp 17- 28; and Reinsberg, J. Hybridoma. 1995, 14, pp 205-208). Additional examples of known agents that suffer from deleterious immune responses are blood-clotting factors such as factor VIII. When administered to hemophilia A patients, factor VIII restores the ability of the blood to clot. Although factor VIII is a human protein, it still elicits an immune response in hemophiliacs as endogenous factor VIII is not present in their blood and thus it appears as a foreign antigen to the immune system. Approximately 29-33% of new patients will produce antibodies that bind and neutralize the therapeutically administered factor VIII {see Lusher J. M. Semin Thromb Hemost. 2002, 28(3), pp 273-276). These neutralizing antibodies require the administration of larger amounts of factor VIII in order to maintain normal blood clotting parameters; an expensive regimen of treatment in order to induce immune tolerance {see Briet E et al. Adv. Exp. Med. Bio. 2001, 489, pp 89-97). Another immunogenic example is adenoviral vectors. Retroviral therapy remains experimental and is of limited utility. One reason is that the application of a therapeutic virus generates an immune response capable of blocking any subsequent administration of the same or similar virus {see Yiping Yang et al. J. of Virology. 1995, 69, pp 2004-2015). This ensures that retroviral therapies must be based on the transient expression of a protein or the direct incorporation of viral sequence into the host genome. Directed research has identified multiple viral neutralizing epitopes recognized by host antibodies {see Hanne, Gahery-Segard et al. J. of Virology 1998. 72, pp 2388-2397) suggesting that viral modifications will not be sufficient to overcome this obstacle. This invention will enable a process whereby an adenoviral therapy will have utility for repeated application. Another example of an immunogenic agent that elicits neutralizing antibodies is the well-known cosmetic agent Botox. Botulin toxin protein, is purified from the fermentation of Clostridium botulinum. As a therapeutic agent, it is used for muscle disorders such as cervical dystonia in addition to cosmetic application. After repeated exposure patients generate neutralizing antibodies to the toxin that results in reduced efficacy {see Birklein F. et al. Ann Neurol. 2002, 52, pp 68-73 and Rollnik, J. D. et al. Neurol. CHn. Neurophysiol. 2001, 2001(3), pp 2-4). A "deleterious immune response" also encompasses diseases caused by therapeutic agents. A specific example of this is the immune response to therapy with recombinant human erythropoietin (EPO). Erythropoietin is used to stimulate the growth or of red cells and restore red blood cell counts in patients who have undergone chemotherapy or dialysis. A small percentage of patients develop antibodies to EPO and subsequently are unresponsive to both therapeutically administered EPO and their own endogenous EPO {see Casadevall, N. et al, NEJM. 2002, 346, pp 469-475). They contract a disorder, pure red cell aplasia, in which red blood cell production is severely diminished (see Gershon S. K. et. al. NEJM. 2002, 346, pp 1584-1586). This complication of EPO therapy is lethal if untreated. Another specific example is the murine antibody, OKT3 (a.k.a., Orthoclone) a monoclonal antibody directed towards CD-3 domain of activated T-cells. In clinical trials 20-40% of patients administered OKT3 produce antibodies versus the therapy. These antibodies, besides neutralizing the therapy, also stimulate a strong host immune reaction. The immune reaction is severe enough that patients with high titers of human anti- mouse antibodies are specifically restricted from taking the drug (see Orthoclone package label). A final example is a human antibody therapeutic. Humira® is a monoclonal antibody directed against TNF and is used to treat rheumatoid arthritis patients. When taken alone ~12% of patients develop neutralizing antibodies. In addition, a small percentage of patients given the drug also contract a systemic lupus erthematosus-like condition that is an IgG- mediated immune response induced by the therapeutic agent (see Humira package label).
[0058] Another example of "deleterious immune response" is a host reaction to small molecule drugs. It is known to those skilled in the art that certain chemical structures will conjugate with host proteins to stimulate immune recognition (see Ju. C. et al. 2002. Current Drug Metabolism 3, pp 367-377 and Kimber I. et al. 2002, Toxicologic Pathology 30, pp 54- 58.) A substantial portion of these host reactions are IgG mediated. Specific "deleterious immune responses" that are IgG mediated include: hemolytic anemia, Steven- Johnson syndrome and drug induced Lupus.
[0059] "Halo" refers to fluoro, chloro, bromo or iodo.
[0060] "Haloalkyl" refers to alkyl as defined above substituted by one or more, preferably one to seven, "halo" atoms, as such terms are defined in this Application. Haloalkyl includes monohaloalkyl, dihaloalkyl, trihaloalkyl, perhaloalkyl and the like e.g. chloromethyl, dichloromethyl, difluoromethyl, trifluoromethyl, 2,2,2-trifluoroethyl, perfluoroethyl, 2,2,2-trifluoro-l,l-dichloroethyl, and the like).
[0061] "Haloalkoxy" refers to a -OR radical where R is haloalkyl group as defined above e.g., trifluoromethoxy, 2,2,2-trifluoroethoxy, difluoromethoxy, and the like.
[0062] "Heteroaryl" as a group or part of a group denotes an aromatic monocyclic or bicyclic moiety of 5 to 10 ring atoms in which one or more, preferably one, two, or three, of the ring atom(s) is(are) selected from nitrogen, oxygen or sulfur, the remaining ring atoms being carbon. Representative heteroaryl rings include, but are not limited to, pyrrolyl, furanyl, thienyl, oxazolyl, isoxazolyl, thiazolyl, imidazolyl, triazolyl, tetrazolyl, pyridinyl, pyrimidinyl, pyrazinyl, pyridazinyl, indolyl, benzofuranyl, benzothiophenyl, benzimidazolyl, quinolinyl, isoquinolinyl, quinazolinyl, quinoxalinyl, pyrazolyl, and the like.
[0063] "Heteroaryloxy" refers to a -OR radical where R is heteroaryl as defined above e.g., furanyloxy, pyridinyloxy, indolyloxy, and the like.
[0064] "Heteroaryl oxycarbonyl" refers to a -C(O)O-R radical where R is heteroaryl as defined above e.g., pyridinyloxycarbonyl, pyrimidinyloxycarbonyl, and the like.
[0065] "Heteroaralkyl" refers to a -(alkylene)-R radical where R is heteroaryl as defined above e.g., pyridinylmethyl, 1- or 2-furanylethyl, imidazolylmethyl, and the like.
[0066] Ηeteroaralkyloxy" refers to a -O-R radical where R is heteroaralkyl as defined above e.g., pyridinylmethyloxy, furanylethyloxy, and the like.
[0067] "Heteroaralkyloxycarbonyl" refers to a -C(O)O-R radical where R is heteroaralkyl as defined above e.g., pyridinylmethyloxycarbonyl, pyrimidinylmethyloxycarbonyl, and the like.
[0068] "Heterocycloalkyl" refers to a saturated or partially unsaturated, mono or bicyclic radical of 4, 5 or 6 carbon ring atoms wherein one or more, preferably one, two, or three of the ring carbon atoms are replaced by a heteroatom selected from -N=, -N-, -O-, -S-, -SO-, or -S(O)2- and further wherein one or two ring carbon atoms are optionally replaced by a keto (- CO-) group. The heterocycloalkyl ring is optionally fused to cycloalkyl, aryl or heteroaryl ring as defined herein. Representative examples include, but are not limited to, imidazolidinyl, morpholinyl, thiomorpholinyl, thiomorpholino-1 -oxide, thiomorpholino-1,1- dioxide, tetrahydrofuranyl, tetrahydropyranyl, tetrahydrothiopyranyl, 1 -oxo- tetrahydrothiopyranyl, 1,1-dioxotetrathio-pyranyl, indolinyl, piperazinyl, piperidyl, pyrrolidinyl, pyrrolinyl, quinuclidinyl,3,4-dihydroisoquinolinyl, dihydroindolyl, and the like.
[0069] When the heterocycloalkyl group contains at least one nitrogen ring atom it is referred to herein as "heterocycloamino" and is a subset of the heterocycloalkyl group as defined above.
[0070] "Heterocyclylalkylene" refers to a divalent heterocyclyl group, as defined in this Application, e.g., the instance wherein R and R4 together with the carbon atom to which both R3 and R4 are attached form heterocyclylalkylene" includes, but is not limited to, the following:
in which R is a substituent defined in the Summary of the Invention [0071] "Heterocycloalkylalkyl" refers to a -(alkylene)-R radical where R is heterocycloalkyl as defined above e.g., pyrrolidinylmethyl, tetrahydrofuranylethyl, pyridinylmethylpiperidinylmethyl, and the like.
[0072] "Heterocycloalkyloxycarbonyl" refers to a -C(O)OR radical where R is heterocycloalkyl as defined above e.g., pyridinyloxycarbonyl, pyrimidinyloxycarbonyl, and the like.
[0073] "Heterocycloalkylalkyloxycarbonyl" refers to a -C(O)OR radical where R is heterocycloalkyla;lyl as defined above e.g., pyridinylmethyloxycarbonyl, pyrimidinylmethyloxycarbonyl, and the like.
[0074] "Heterocycloalkylaminocarbonyl" refers to a -CONHR radical where R is heterocycloalkyl as defined above e.g., tetrahydrofuranylaminocarbonyl, tetrahydropyranylaminocarbonyl, and the like.
[0075] "Hydroxy" means -OH radical. Unless indicated otherwise, the compounds of the invention containing hydroxy radicals include protected derivatives thereof. Suitable protecting groups for hydroxy moieties include benzyl and the like.
[0076] "Hydroxyalkyl" means a linear monovalent hydrocarbon radical of one to six carbon atoms or a branched monovalent hydrocarbon radical of three to six carbons substituted with one or two hydroxy groups, provided that if two hydroxy groups are present they are not both on the same carbon atom. Representative examples include, but are not limited to, hydroxymethyl, 2-hydroxyethyl, 2-hydroxypropyl, 3-hydroxyproρyl, l-(hydroxymethyl)-2- methylpropyl, 2-hydroxybutyl, 3-hydroxybutyl, 4-hydroxybutyl, 2,3-dihydroxypropyl, 1- (hydroxymethyl)-2-hydroxyethyl, 2,3-dihydroxybutyl, 3,4-dihydroxybutyl and 2- (hydroxymethyl)-3-hydroxypropyl, preferably 2-hydroxyethyl, 2,3-dihydroxypropyl, and 1- (hydroxymethyl)-2-hydroxyethyl.
[0077] "Isomers" mean compounds of Formula (I) having identical molecular formulae but differ in the nature or sequence of bonding of their atoms or in the arrangement of their atoms in space. Isomers that differ in the arrangement of their atoms in space are termed "stereoisomers". Stereoisomers that are not mirror images of one another are termed "diastereomers" and stereoisomers that are nonsuperimposable mirror images are termed "enantiomers" or sometimes "optical isomers". A carbon atom bonded to four nonidentical substituents is termed a "chiral center". A compound with one chiral center that has two enantiomeric forms of opposite chirality is termed a "racemic mixture". A compound that has more than one chiral center has 2""1 enantiomeric pairs, where n is the number of chiral centers. Compounds with more than one chiral center may exist as either an individual diastereomer or as a mixture of diastereomers, termed a "diastereomeric mixture". When one chiral center is present a stereoisomer may be characterized by the absolute configuration of that chiral center. Absolute configuration refers to the arrangement in space of the substituents attached to the chiral center. Enantiomers are characterized by the absolute configuration of their chiral centers and described by the R- and ^-sequencing rules of Cahn, Ingold and Prelog. Conventions for stereochemical nomenclature, methods for the determination of stereochemistry and the separation of stereoisomers are well known in the art (e.g., see "Advanced Organic Chemistry", 4th edition, March, Jerry, John Wiley & Sons, New York, 1992). It is understood that the names and illustration used in this Application to describe compounds of Formula (I) are meant to be encompassed all possible stereoisomers.
[0078] "Optional" or "optionally" or "may be" means that the subsequently described event or circumstance may or may not occur, and that the description includes instances where the event or circumstance occurs and instances in which it does not. For example, the phrase "wherein the aromatic ring in Ra is optionally substituted with one or two substituents independently selected from alkyl" means that the aromatic ring may or may not be substituted with alkyl in order to fall within the scope of the invention.
[0079] The present invention also includes JV-oxide derivatives of a compound of Formula (I). JV-oxide derivative mean a compound of Formula (I) in which a nitrogen atom is in an oxidized state (i.e., N-»O) e.g., pyridine iV-oxide, and which possess the desired pharmacological activity. [0080] "Pathology" of a disease means the essential nature, causes and development of the disease as well as the structural and functional changes that result from the disease processes.
[0081] "Pharmaceutically acceptable" means that which is useful in preparing a pharmaceutical composition that is generally safe, non-toxic and neither biologically nor otherwise undesirable and includes that which is acceptable for veterinary use as well as human pharmaceutical use.
[0082] "Pharmaceutically acceptable salts" means salts of compounds of Formula (I) which are pharmaceutically acceptable, as defined above, and which possess the desired pharmacological activity. Such salts include acid addition salts formed with inorganic acids such as hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid, phosphoric acid, and the like; or with organic acids such as acetic acid, propionic acid, hexanoic acid, heptanoic acid, cyclopentanepropionic acid, glycolic acid, pyruvic acid, lactic acid, malonic acid, succinic acid, malic acid, maleic acid, fumaric acid, tartaric acid, citric acid, benzoic acid, o-(4-hydroxybenzoyl)benzoic acid, cinnamic acid, mandelic acid, methylsulfonic acid, ethanesulfonic acid, 1 ,2-ethanedisulfonic acid, 2-hydroxy-ethanesulfonic acid, benzenesulfonic acid, jc-chlorobenzenesulfonic acid, 2-naphthalenesulfonic acid, p-toluenesulfonic acid, camphorsulfonic acid, 4-methylbicyclo[2.2.2]oct-2-ene-l-carboxylic acid, glucoheptonic acid, 4,4'-methylenebis(3-hydroxy-2-ene-l-carboxylic acid), 3-phenylpropionic acid, trimethylacetic acid, tertiary butylacetic acid, lauryl sulfuric acid, gluconic acid, glutamic acid, hydroxynaphthoic acid, salicylic acid, stearic acid, muconic acid and the like.
[0083] Pharmaceutically acceptable salts also include base addition salts which may be formed when acidic protons present are capable of reacting with inorganic or organic bases. Acceptable inorganic bases include sodium hydroxide, sodium carbonate, potassium hydroxide, aluminum hydroxide and calcium hydroxide. Acceptable organic bases include ethanolamine, diethanolamine, triethanolamine, tromethamine, JV-methylglucamine and the like.
[0084] The present invention also includes prodrugs of a compound of Formula (I). Prodrug means a compound that is convertible in vivo by metabolic means (e.g. by hydrolysis) to a compound of Formula (I). For example, an ester of a compound of Formula (I) containing a hydroxy group may be convertible by hydrolysis in vivo to the parent molecule. Alternatively an ester of a compound of Formula (I) containing a carboxy group may be convertible by hydrolysis in vivo to the parent molecule. Suitable esters of compounds of Formula (I) containing a hydroxy group, are for example acetates, citrates, lactates, tartrates, malonates, oxalates, salicylates, propionates, succinates, fumarates, maleates, methylene-bis-βb-hydroxynaphthoates, gentisates, isethionates, di-p-toluoyltartrates, methylsulphonates, ethanesulphonates, benzenesulphonates, j9-toluenesulphonates, cyclohexylsulphamates and quinates. Suitable esters of compounds of Formula (I) containing a carboxy group, are for example those described by Leinweber, FJ. Drug Metab. Res., 1987, 18, page 379. An especially useful class of esters of compounds of Formula (I) containing a hydroxy group, may be formed from acid moieties selected from those described by Bundgaard et al., J. Med. Chem., 1989, 32, pp 2503-2507, and include substituted (aminomethyl)-benzoates, for example, dialkylamino-methylbenzoates in which the two alkyl groups may be joined together and/or interrupted by an oxygen atom or by an optionally substituted nitrogen atom, e.g. an alkylated nitrogen atom, more especially (morpholino-methyl)benzoates, e.g. 3- or 4-(moφholinomethyl)-benzoates, and (4-alkylpiperazin-l-yl)benzoates, e.g. 3- or 4-(4-alkylpiperazin-l-yl)benzoates.
[0085] "Protected derivatives" means derivatives of compounds of Formula (I) in which a reactive site or sites are blocked with protecting groups. Protected derivatives of compounds of Formula (I) are useful in the preparation of compounds of Formula (I) or in themselves may be active cathepsin S inhibitors. A comprehensive list of suitable protecting groups can be found in T. W. Greene, Protective Groups in Organic Synthesis, 3rd edition, John Wiley & Sons, Inc. 1999.
[0086] "Therapeutically effective amount" means that amount which, when administered to an animal for treating a disease, is sufficient to effect such treatment for the disease.
[0087] "Treatment" or "treating" means any administration of a compound of the present invention and includes:
[0088] (1) preventing the disease from occurring in an animal which may be predisposed to the disease but does not yet experience or display the pathology or symptomatology of the disease,
[0089] (2) inhibiting the disease in an animal that is experiencing or displaying the pathology or symptomatology of the diseased (i.e., arresting further development of the pathology and/or symptomatology), or [0090] (3) ameliorating the disease in an animal that is experiencing or displaying the pathology or symptomatology of the diseased (i.e., reversing the pathology and/or symptomatology).
[0091] "Treatment" or "treating" with respect to combination therapy i.e., use with a biologic means any administration of a compound of the present invention and includes:
[0092] (1) preventing the immune response from occurring in an animal which may be predisposed to the immune response but does not yet experience or display the pathology or symptomatology of the immune response,
[0093] (2) inhibiting the immune response in an animal that is experiencing or displaying the pathology or symptomatology of the immune response (i.e., arresting further development of the pathology and/or symptomatology), or
[0094] (3) ameliorating the immune response in an animal that is experiencing or displaying the pathology or symptomatology of the immune response (i.e., reducing in degree or severity, or extent or duration, the overt manifestations of the immune response or reversing the pathology and/or symptomatology e.g., reduced binding and presentation of antigenic peptides by MHC class II molecules, reduced activation of T-cells and B-cells, reduced humoral and cell-mediated responses and, as appropriate to the particular immune response, reduced inflammation, congestion, pain, necrosis, reduced loss in the efficacy of a biologic agent, and the like).
[0095] The expression "wherein the aromatic or alicyclic ring in R5 is optionally substituted with one, two, or three Ra independently selected from alkyl, haloalkyl, alkoxy, hydroxy, haloalkoxy, or halo; or optionally substituted with one or two Rb independently selected from hydrogen, alkyl, haloalkyl, alkoxy, hydroxy, haloalkoxy, halo, carboxy, or alkoxycarbonyl and one Rc selected from hydroxyalkyl, alkoxyalkyl, aminoalkyl, aryl, heteroaryl, aralkyl, heteroaralkyl, cycloalkyl, cycloalkylalkyl, " in the definition of R5 in the compound of Formula (I) means that all the aromatic and alicyclic rings within the scope of R5 whether directly or indirectly attached (e.g., R5 is cycloalkylalkyl, -alkylene-X-R9 where X is as defined in the Summary of the Invention and R9 is ary, aralkyl, etc, ..) are optionally substituted with R\ or Rb and Rc, or Re alone. PREFERRED EMBODIMENTS
[0096] I. Certain compounds of Formula (I) within the broadest scope set forth in the Summary of the Invention are preferred. For example:
[0097] (A) A preferred group of compounds is that wherein:
R1 is hydrogen or methyl, preferably hydrogen;
R2 is cyclopropyl, 1-phenylethyl, or lH-pyrazol-5-yl; preferably cyclopropyl. [0098] (1) Within the above preferred group (A) and more preferred group contained therein, a more preferred group of compounds is that wherein R3 is hydrogen and R4 is alkyl, preferably methyl, ethyl, propyl or butyl, more preferably R4 is ethyl or propyl.
[0099] (2) Within the above preferred group (A) and more preferred group contained therein, a more preferred group of compounds is that wherein R3 is alkyl, preferably methyl or ethyl and R4 is alkyl, preferably methyl, ethyl, propyl or butyl, more preferably R4 is methyl. Preferably, R3 and R4 are methyl.
[0100] (3) Within the above preferred group (A) and more preferred groups contained therein, a more preferred group of compounds is that wherein R3 and R4 together with the carbon atom to which they are attached form cycloalkylene, preferably cyclopropylene, cyclopentylene, or cyclohexylene, more preferably cyclopropylene.
[0101] (4) Within the above preferred group (A) and more preferred group contained therein, a more preferred group of compounds is that wherein R3 and R4 together with the carbon atom to which they are attached form piperidin-4-yl substituted at the nitrogen atom with ethyl, 2,2,2-trifluoroethyl or cyclopropyl, tetrahydropyran-4-yl, tetrahydrothiopyran-4- yl, or l,l-dioxotetrahydrothiopyran-4-yl.
[0102] (i) Within the above preferred groups (A) and A(I -4) and more preferred groups contained therein, a more preferred group of compounds is that wherein R6 is haloalkyl, preferably, difluoromethyl, trifiuoromethyl, 2,2,2-trifluoroethyl, 2,2,3,3,3-pentafluoropropyl, 2,2,3,3,4,4,4-heptafluorobutyl and R7 and R8 are hydrogen.
[0103] (ii) Within the above preferred groups (A) and A(I -4) and more preferred groups contained therein, a more preferred group of compounds is that wherein R6 is haloalkyl, preferably, difluoromethyl, trifiuoromethyl, 2,2,2-trifluoroethyl, or 2,2,3,3,3- pentafluoropropyl, R7 is haloalkyl, preferably, trifiuoromethyl, 2,2,2-trifluoroethyl, or 2,2,3,3,3-pentafluoropropyl, and R8 is hydrogen. [0104] (iii) Within the above preferred groups (A) and A(I -4) and more preferred groups contained therein, a more preferred group of compounds is that wherein R6 is haloalkyl, preferably, difluoromethyl, trifluoromethyl, 2,2,2-trifiuoroethyl, or 2,2,3,3,3- pentafluoropropyl, R7 is alkyl, preferably, methyl, ethyl, or propyl, and R8 is hydrogen.
[0105] (iv) Within the above preferred groups (A) and A(I -4) and more preferred groups contained therein, a more preferred group of compounds is that wherein R6 is haloalkyl, preferably, difluoromethyl, trifluoromethyl, 2,2,2-trifluoroethyl, or 2,2,3,3,3- pentafluoropropyl, R7 is haloalkyl, preferably, trifluoromethyl or 2,2,2-trifluoroethyl, and R8 is aryl optionally substituted with one, two, or three Re. Preferably R8 is phenyl, A- fluorophenyl, 2,3-, 2,4-, 2,5-, 2,6-, 3,4, or 3,5-difluorophenyl. More preferably, R6 and R7 are trifluoromethyl and R8 is phenyl, 4-fluorophenyl, 2,3-, 2,4-, 2,5-, 2,6-, 3,4, or 3,5- difluorophenyl.
[0106] (iv) Within the above preferred groups (A) and A(I -4) and more preferred groups contained therein, a more preferred group of compounds is that wherein R6 is haloalkyl, preferably, difluoromethyl, trifluoromethyl, 2,2,2-trifluoroethyl, or 2,2,3,3,3- pentafluoropropyl, R7 is alkyl, preferably, methyl or ethyl, and R8 is aryl optionally substituted with one, two, or three Re. Preferably R8 is phenyl, 4-fluorophenyl, 2,3-, 2,4-, 2,5-, 2,6-, 3,4, or 3,5-difluorophenyl. More preferably, R6 and R7 are trifluoromethyl and R8 is phenyl, 4-fluorophenyl, 2,3-, 2,4-, 2,5-, 2,6-, 3,4, or 3,5-difluorophenyl.
[0107] (v) Within the above preferred groups (A) and A(I -4) and more preferred groups contained therein, a more preferred group of compounds is that wherein R6 is haloalkyl, preferably, trifluoromethyl, difluoromethyl, 2,2,2-trifluoroethyl, or 2,2,3,3,3- pentafluoropropyl, R7 is hydrogen, and R8 is aryl optionally substituted with one, two, or three Re. Preferably R8 is phenyl, 4-fluorophenyl, 2,3-, 2,4-, 2,5-, 2,6-, 3,4, or 3,5- difluorophenyl. More preferably, R6 is trifluoromethyl and R8 is phenyl, 4-fluorophenyl, 2,3- , 2,4-, 2,5-, 2,6-, 3,4, or 3,5-difluorophenyl, preferably 2,4-difluorophenyl.
[0108] (vi) Within the above preferred groups (A) and A(l-4) and more preferred groups contained therein, a more preferred group of compounds is that wherein R6 is haloalkyl, preferably, trifluromethyl, 2,2,2-trifluoroethyl, or 2,2,3,3,3-pentafluoropropyl, R7 is haloalkyl, preferably, trifluoromethyl or 2,2,2-trifluoroethyl, and R8 is heteroaryl optionally substituted with one, two, or three Re. Preferably R8 is indol-5-yl, benzoxazol-5-yl, thiophen- 3-yl, thiophen-2-yl, furan-2-yl, pyridin-4-yl, pyridin-3-yl, pyridin-2-yl, imidazol-5-yl, pyrimidin-2-yl, pyrazin-2-yl, pyrimidin-5-yl, pyrimdin-4-yl, pyridazin-4-yl, isoxazol-4-yl, imidazol-2-yl, [1.2.3]thiadiazol-4-yl, imidazol-4-yl, pyrazol-4-yl, thiazol-2-yl, pyrazol-4-yl, pyrrol-2-yl, pyrrol-3-yl, thiazol-4-yl, thiazol-5-yl optionally substituted with one or two methyl.
[0109] (vii) Within the above preferred groups (A) and A(I -4) and more preferred groups contained therein, a more preferred group of compounds is that wherein R6 is haloalkyl, preferably, trifluromethyl, 2,2,2-trifiuoroethyl, or 2,2,3,3,3-pentafluoropropyl, R7 is alkyl, preferably, methyl or ethyl, and and R8 is heteroaryl optionally substituted with one, two, or three Re. Preferably R8 is indol-5-yl, benzoxazol-5-yl5 thiophen-3-yl, thiophen-2-yl, furan-2- yl, pyridine-4-yl, pyridin-3-yl, pyridin-2-yl, imidazol-5-yl, pyrimidin-2-yl, pyrazin-2-yl, pyrimidin-5-yl, pyrimdin-4-yl, pyridazin-4-yl, isoxazol-4-yl, imidazol-2-yl, [1.2.3]thiadiazol- 4-yl, imidazol-4-yl, pyrazol-4-yl, thiazol-2-yl, pyrazol-4-yl, pyrrol-2-yl, pyrrol-3-yl, thiazol- 4-yl, thiazol-5-yl optionally substituted with one or two methyl.
[0110] (viii) Within the above preferred groups (A) and A(I -4) and more preferred groups contained therein, a more preferred group of compounds is that wherein R6 is haloalkyl, preferably, trifluromethyl, 2,2,2-trifiuoroethyl, or 2,2,3,3,3-pentafluoropropyl, R7 is hydrogen, and and R8 is heteroaryl optionally substituted with one, two, or three Re. Preferably R8 is indol-5-yl, benzoxazol-5-yl, thiophen-3-yl, thiophen-2-yl, furan-2-yl, pyridin-4-yl, pyridin-3-yl, pyridin-2-yl, imidazol-5-yl, pyrimidin-2-yl, pyrazin-2-yl, pyrimidin-5-yl, pyrimdin-4-yl, pyridazin-4-yl, isoxazol-4-yl, imidazol-2-yl, [1.2.3]thiadiazol- 4-yl, imidazol-4-yl, pyrazol-4-yl, thiazol-2-yl, pyrazol-4-yl, pyrrol-2-yl, pyrrol-3-yl, thiazol- 4-yl, thiazol-5-yl optionally substituted with one or two methyl.
[0111] (a) Within the above preferred groups (A), A(l-4), A(l-viii) and A(i-4)(i-viii), and more preferred groups contained therein, an even more preferred group of compounds is that wherein R5 is -alkylene-SO2NRπR12 where:
R11 is hydrogen, alkyl, haloalkyl, alkoxyalkyl, hydroxyalkyl, aminoalkyl, carboxyalkyl, alkoxycarbonylalkyl, acyl, cycloalkyl, cycloalkylalkyl, aryl, heteroaryl, aralkyl, heteroaralkyl, heterocycloalkylalkyl, acylalkyl, or heterocycloalkylaminocarbonyl; and R12 is hydrogen, alkyl, aryl, heteroaryl, aralkyl, heteroaralkyl, cycloalkyl, cycloalkylalkyl, heterocycloalkyl, or heterocycloalkylalkyl; wherein the aromatic or alicyclic ring are optionally substituted with one, two, or three Ra independently selected from alkyl, haloalkyl, alkoxy, hydroxy, haloalkoxy, cyano, halo, carboxy or alkoxycarbonyl; or optionally substituted with one or two Rb independently selected from hydrogen, alkyl, haloalkyl, alkoxy, hydroxy, haloalkoxy, halo, carboxy, or alkoxycarbonyl and one Rc selected from cyano, hydroxyalkyl, alkoxyalkyl, aminoalkyl, aryl, heteroaryl, aralkyl, heteroaralkyl, cycloalkyl, cycloalkylalkyl, heterocycloalkyl, heterocycloalkylalkyl, acyl, acylalkyl, alkoxycarbonyl, aryloxycarbonyl, aralkyloxycarbonyl, heteroaryloxycarbonyl, heteroaralkyloxycarbonyl, heterocycloalkyloxycarbonyl, heterocycloalkylalkyloxycarbonyl, cycloalkyloxycarbonyl, cycloalkylalkyloxycarbonyl, aryloxy, heteroaryloxy, aralkyloxy, heteroaralkyloxy, aminocarbonyl, aminosulfonyl, or - SO2R13 (where R13 is alkyl, cycloalkyl, aryl, heteroaryl, or heterocycloalkyl); and further wherein the aromatic or alicyclic ring in Rc is optionally substituted with one, two, or three R independently selected from alkyl, haloalkyl, alkoxy, hydroxy, haloalkoxy, halo, cyano, - CONH2, alkylaminocarbonyl, dialkylaminocarbonyl, alkylcarbonyl, alkylsulfonyl, or alkylsulfonylamino . [0112] Preferably, R11 is methyl, ethyl, propyl, butyl, 2,2,2-trifluoroethyl, 2-hydroxethyl, 2- or 3-hydroxypropyl, 2-methoxy or ethoxyethyl, 2- or 3-methoxy or ethoxypropyl, methylaminoethyl, methylaminopropyl, acetylaminoethyl, 2-carboxy ethyl, 3-carboxypropyl, methoxycarbonylethyl, acetyl, cyclopropyl, cyclopropylmethyl, benzyl, phenylethyl, pyridinylethyl, pyridinylmethyl, pyrimidinylmethyl, furanylmethyl, pyrrolylmethyl, indolylmethyl, quinolinylmethyl, isoquinolinylmethyl, or tetrahydroquinolinylmethyl and R12 is hydrogen, methyl, ethyl, phenyl, benzyl, pyridinylmethyl or ethyl, pyrimidinylmethyl or ethyl, indolylmethyl or ethyl, quinolinylmethyl or ethyl, dihydroindolylmethyl or ethyl, piperidinylmethyl or ethyl, piperazinylmethyl or ethyl, pyrrolidinylmethyl or ethyl, or morpholinylmethyl or ethyl wherein the aromatic rings or alicyclic rings in R11 and R12 are optionally substituted with one, two, or three Ra independently selected from methyl, ethyl, trifluoromethyl, trifluoromethoxy, methoxy, hydroxy, or fluoro; or optionally substituted with one or two Rb independently selected from hydrogen, methyl, ethyl, trifluoromethyl, methoxy, hydroxy, trifluoromethoxy, or fluoro and one Rc selected from hydroxymethyl, hydroxyethyl, 2- or 3-hydroxypropyl, cyclopropylmethyl, phenyl, pyridinyl, benzyl, cyclopropyl, benzoyl, methoxycarbonyl, ethoxycarbonyl, acetyl, trifluoroacetyl, benzyloxycarbonyl, dimethylaminocarbonyl, or methylaminocarbonyl; and further wherein the aromatic or alicyclic ring in Rc is optionally substituted with one, two, or three Rd independently selected from methyl, ethyl, trifluoromethyl, methoxy, hydroxyl, trifluormethoxy or fluoro. [0113] (b) Within the above preferred groups (A), A( 1 -4), A(I -viii) and A(i-4)(i-viii), and more preferred groups contained therein, R5 is -alkylene-SO2NRnR12 where:
R11 is alkoxyalkyl, hydroxyalkyl, aminoalkyl, carboxyalkyl, alkoxycarbonylalkyl, cycloalkyl, cycloalkylalkyl, aryl, heteroaryl, aralkyl, heteroaralkyl, heterocycloalkylalkyl and
R12 is hydrogen, alkyl, aryl, heteroaryl, aralkyl, heteroaralkyl, cycloalkyl, cycloalkylalkyl, heterocycloalkyl, or heterocycloalkylalkyl; one, two, or three Ra independently selected from alkyl, haloalkyl, alkoxy, hydroxy, haloalkoxy, cyano, halo, carboxy or alkoxycarbonyl; or optionally substituted with one or two Rb independently selected from hydrogen, alkyl, haloalkyl, alkoxy, hydroxy, haloalkoxy, halo, carboxy, or alkoxycarbonyl and one Rc selected from cyano, hydroxyalkyl, alkoxyalkyl, aminoalkyl, aryl, heteroaryl, aralkyl, heteroaralkyl, cycloalkyl, cycloalkylalkyl, heterocycloalkyl, heterocycloalkylalkyl, acyl, acylalkyl, alkoxycarbonyl, aryloxycarbonyl, aralkyloxycarbonyl, heteroaryloxycarbonyl, heteroaralkyloxycarbonyl, heterocycloalkyloxycarbonyl, heterocycloalkylalkyloxycarbonyl, cycloalkyloxycarbonyl, cycloalkylalkyloxycarbonyl, aryloxy, heteroaryloxy, aralkyloxy, heteroaralkyloxy, aminocarbonyl, aminosulfonyl, or -SO2R13 (where R13 is alkyl, cycloalkyl, aryl, heteroaryl, or heterocycloalkyl); and further wherein the aromatic or alicyclic ring in Rc is optionally substituted with one, two, or three R independently selected from alkyl, haloalkyl, alkoxy, hydroxy, haloalkoxy, halo, cyano, -CONH2, alkylaminocarbonyl, dialkylaminocarbonyl, alkylcarbonyl, alkylsulfonyl, or alkylsulfonylamino;
[0114] Preferably, R11 is 2-hydroxethyl, 2- or 3-hydroxypropyl, 2-methoxy or ethoxyethyl, 2- or 3-methoxy or ethoxypropyl, methylaminoethyl, methylaminopropyl, acetylaminoethyl, 2-carboxyethyl, 3-carboxypropyl, methoxycarbonylethyl, acetyl, cyclopropyl, cyclopropylmethyl, benzyl, phenylethyl, pyridinylethyl, pyridinylmethyl, pyrimidinylmethyl, furanylmethyl, pyrrolylmethyl, indolylmethyl, quinolinylmethyl, isoquinolinylmethyl, tetrahydroquinolinylmethyl or -(CH2)2-NRR (where R is methyl, ethyl, hydroxyethyl, hydroxypropyl, or methoxyethyl and R' is hydroxyethyl, hydroxypropyl, methoxyethyl, methoxypropyl, phenyl, benzyl, cyclopropyl, or cyclopropylmethyl) and R is hydrogen, methyl, ethyl, phenyl, benzyl, pyridinylmethyl or ethyl, pyrimidinylmethyl or ethyl, indolylmethyl or ethyl, quinolinylmethyl or ethyl, dihydroindolylmethyl or ethyl, piperidinylmethyl or ethyl, piperazinylmethyl or ethyl, pyrrolidinylmethyl or ethyl, or morpholinylmethyl or ethyl wherein the aromatic rings or alicyclic rings are optionally substituted with one, two, or three Ra independently selected from methyl, ethyl, trifluoromethyl, trifluoromethoxy, methoxy, hydroxy, or fluoro; or optionally substituted with one or two Rb independently selected from hydrogen, methyl, ethyl, trifluoromethyl, methoxy, hydroxy, trifluoromethoxy, or fluoro and one Rc selected from hydroxymethyl, hydroxy ethyl, 2- or 3-hydroxypropyl, cyclopropylmethyl, phenyl, pyridinyl, benzyl, cyclopropyl, benzoyl, methoxycarbonyl, ethoxycarbonyl, acetyl, trifluoroacetyl, benzyloxycarbonyl, dimethylaminocarbonyl, or methylaminocarbonyl; and further wherein the aromatic or alicyclic ring in Rc is optionally substituted with one, two, or three Rd independently selected from methyl, ethyl, trifluoromethyl, methoxy, hydroxyl, trifluormethoxy or fluoro.
[0115] (c) Within the above preferred groups (A), A(I -4), A(I -viii) and A(i-4)(i-viii), and more preferred groups contained therein, R5 is — alkylene-SO2NRnR12 where R11 and R12 together with the nitrogen atom to which they are attached form heterocycloamino substituted with one, two, or three Ra independently selected from alkyl, haloalkyl, alkoxy, hydroxy, haloalkoxy, cyano, halo, carboxy or alkoxycarbonyl; or optionally substituted with one or two Rb independently selected from hydrogen, alkyl, haloalkyl, alkoxy, hydroxy, haloalkoxy, halo, carboxy, or alkoxycarbonyl and one Rc selected from cyano, hydroxyalkyl, alkoxyalkyl, aminoalkyl, aryl, heteroaryl, aralkyl, heteroaralkyl, cycloalkyl, cycloalkylalkyl, heterocycloalkyl, heterocycloalkylalkyl, acyl, acylalkyl, alkoxycarbonyl, aryloxycarbonyl, aralkyloxycarbonyl, heteroaryloxycarbonyl, heteroaralkyloxycarbonyl, heterocycloalkyloxycarbonyl, heterocycloalkylalkyloxycarbonyl, cycloalkyloxycarbonyl, cycloalkylalkyloxycarbonyl, aryloxy, heteroaryloxy, aralkyloxy, heteroaralkyloxy, aminocarbonyl, aminosulfonyl, or -SO2R13 (where R13 is alkyl, cycloalkyl, aryl, heteroaryl, or heterocycloalkyl); and further wherein the aromatic or alicyclic ring in Rc is optionally substituted with one, two, or three Rd independently selected from alkyl, haloalkyl, alkoxy, hydroxy, haloalkoxy, halo, cyano, -CONH2, alkylaminocarbonyl, dialkylaminocarbonyl, alkylcarbonyl, alkylsulfonyl, or alkylsulfonylamino.
[0116] Preferably, R11 and R12 together with the nitrogen atom to which they are attached form 2,3-dihydroindol-l-yl, l,3-dihydroisoindol-2-yl, 3,4-dihydroisoquinolin-2-yl, morpholinyl, piperidin- 1 -yl or piperazin- 1 -yl, preferably piperazin- 1 -yl, optionally substituted with one or two Rb independently selected from hydrogen, methyl, trifluoromethyl, methoxy, hydroxy, trifluoromethoxy, carboxy, fluoro, or methoxycarbonyl and one Rc selected from hydroxymethyl, hydroxyethyl, 2-or 3-hydroxypropyl, 2- dimethylaminoethyl, phenyl, benzyl, pyridinyl, pyrimidinyl, pyrazinyl, pyrrolyl, cyclopropyl, cyclopropylmethyl, benzoyl, pyridinylcarbonyl, benzyloxycarbonyl, cyclopropyloxycarbonyl, tetrahydropyran-4-yloxycarbonyl, methylaminocarbonyl, or dimethylaminocarbonyl; and further wherein the aromatic or alicyclic ring in Rc is optionally substituted with one, two, or three Rd independently selected from methyl, ethyl, trifluoromethyl, methoxy, hydroxyl, ethoxy, trifluoromethoxy, or fluoro. More preferably, R11 and R12 together with the nitrogen atom to which they are attached forms piperazin-1-yl or piperidin-1-yl which is substituted at the 4-position with an Rc group.
[0117] (d) Within the above preferred groups (A), A(I -4), A(l-viii) and A(i-4)(i-vhϊ), and more preferred groups contained therein, R5 is -alkylene-SO2NRuR12 where R11 and R12 together with the nitrogen atom to which they are attached form a bridged azabicyclic ring optionally substituted with one, two, or three Ra independently selected from alkyl, haloalkyl, alkoxy, hydroxy, haloalkoxy, or halo; or optionally substituted with one or two Rb independently selected from hydrogen, alkyl, haloalkyl, alkoxy, hydroxy, haloalkoxy, halo, carboxy, or alkoxycarbonyl and one R0 selected from hydroxyalkyl, alkoxyalkyl, aminoalkyl, aryl, heteroaryl, aralkyl, heteroaralkyl, cycloalkyl, cycloalkylalkyl, heterocycloalkylalkyl, acyl, acylalkyl, aryloxycarbonyl, aralkyloxycarbonyl, heteroaryloxycarbonyl, heteroaralkyloxycarbonyl, heterocycloalkyloxycarbonyl, cycloalkyloxycarbonyl, heterocycloalkylaminocarbonyl, aryloxy, heteroaryloxy, aralkyloxy, heteroaralkyloxy, aminocarbonyl, aminosulfonyl, or -SO2R13 (where R13 is alkyl, aryl, heteroaryl, or heterocycloalkyl); and further wherein the aromatic or alicyclic ring in Rc is optionally substituted with one, two, or three Rd independently selected from alkyl, haloalkyl, alkoxy, hydroxy, haloalkoxy, or halo.
[0118] (e) Within the above preferred groups (A), A(l-4), A(l-viii) and A(i-4)(i-vϋi), and more preferred groups contained therein, R5 is -alkylene-S02NRnR12 where R11 and R12 together with the nitrogen atom to which they are attached form heterocycloamino optionally substituted with one, two, or three Ra independently selected from alkyl, haloalkyl, alkoxy, hydroxy, haloalkoxy, or halo. Preferably, R11 and R12 together with the nitrogen atom to which they are attached form 2,3-dihydroindol-l-yl, 3,4-dihydroisoquinolin-2-yl, morpholinyl, piperidin-1-yl or piperazin-1-yl optionally substituted with one, two, or three Ra independently selected from methyl, ethyl, trifluoromethyl, methoxy, hydroxyl, ethoxy, trifluoromethoxy, or fluoro. [0119] (f) Within the above preferred groups (A)5 A(I -4), A(I -viii) and A(i-4)(i-viii), and more preferred groups contained therein, R5 is -alkylene-SO2NRuR12 where R11 and R12 together with the nitrogen atom to which they are attached form heterocycloamino optionally substituted with one or two Rb independently selected from hydrogen, alkyl, haloalkyl, alkoxy, hydroxy, haloalkoxy, halo, carboxy, or alkoxycarbonyl and one Rc selected from aryl, heteroaryl, aralkyl, heteroaralkyl, cycloalkyl, cycloalkylalkyl, acyl, or alkoxycarbonyl; and further wherein the aromatic or alicyclic ring in Rc is optionally substituted with one, two, or three R independently selected from alkyl, haloalkyl, alkoxy, hydroxy, haloalkoxy, or halo. Preferably, R11 and R12 together with the nitrogen atom to which they are attached form 2,3- dihydroindol- 1 -yl, 3 ,4-dihydroisoquinolin-2-yl, morpholinyl, piperidin- 1 -yl or piperazin- 1 -yl optionally substituted with with one or two Rb independently selected from hydrogen, methyl, trifiuoromethyl, methoxy, hydroxyl, trifluoromethoxy, carboxy, fluoro, or methoxycarbonyl and one Rc selected from phenyl, pyridinyl, pyrimidinyl, benzyl, cyclopropyl, cyclopropylmethyl, benzoyl, acetyl, or trifluoroacetyl; and further wherein the aromatic or alicyclic ring in Rc is optionally substituted with one, two, or three Rd independently selected from methyl, ethyl, trifiuoromethyl, methoxy, hydroxyl, ethoxy, trifluoromethoxy, or fluoro.
[0120] (g) Within the above preferred groups (A)5 A( 1 -4), A( 1 -viii) and A(i-4)(i-viii), and more preferred groups contained therein, R5 is -alkylene-SO2NRuR12 where R11 is 2- imidazol-4-ylethyl5 imidazol-4-ylmethyl, l-methylimidazol-4-ylmethyl, 2-(l-methylimidazol- 4-yl)ethyl, 4-CF3pyridin-3-yl, 4-CNpyridin-3-yl, 3-CF3pyridin-2-yl, 3-CNρyridin-2-yl, 3- CF3pyridin-4-yl, 3-CNρyridin-4-yl, 2-CF3pyridin-3-yl, 2-CNpyridin-3-yl, 2- JV- methylaminoethyl, 2-JV,JV-dimethylaminoethyl, 2-iV-ethyl-iV-methylaminoethyl, 2-JV- isopropyl-TV-methylaminoethyl, 2-(JV -cyclopropyl-JV-methylamino)ethyl5 2-(JV-cyclobutyl-JV- methylamino)ethyl, 2-[JV-(oxetan-3-yl)-JV-methylamino]ethyl, 2-[JV-(azetidin-3-yl)-JV- methylaminojethyl, 2-[ JV-(I, l-dioxo-lλ6-thietan-3-yl)methylamino]ethyl, 2-(JV-cyclopentyl- JV-methylamino)ethyl5 2-[JV-(3-CH3Ocycloρentyl)-JV-methylamino]ethyl5 2-[JV-(3- CHF2Ocyclopentyl)-JV-methylamino]ethyl52-[JV-(3-CF3Ocycloρentyl)-JV-methylamino]ethyl5 2-[JV-(3-phenoxycyclopentyl)-JV-methylamino)ethyl, 2-{JV-[3-(4-Fphenoxy)cyclopentyl]-JV- methylamino} ethyl, 2-{JV-[3-(4-Clphenoxy)cyclopentyl]-JV-methylamino}ethyl, 2-{JV-[3-(4- Brphenoxy)cyclopentyl]-JV-methylamino}ethyl, 2-{JV-[3-(4-COOH-phenoxy)cyclopentyl]-JV- methylamino } ethyl, 2- (JV- [3 -(4-CN-phenoxy)cyclopentyl] -JV-methylamino } ethyl, 2- (JV- [3 - (4-CONH2-phenoxy)cyclopentyl]-N-methylamino}ethyl, 2-(JV-cyclohexyl-JV- methylamino)ethyl, 2-[JV-(tetrahydropyran-4-yl)-JV-methylamino]ethyl, 2-[JV-(piperidin-4-yl)- N-methylamino]ethyl, 2-[iV-(l -acetylpiperidin-4-yl)-iV-methylamino]ethyl, 2-[JV-(I - CF3COpiperidin-4-yl)-Λr-methylaniino]ethyl, 2-[iV-(tetrahydrothiopyran-4-yl)-iV- methylamino] ethyl, 2-[7V-(1 , 1 -dioxo- 1 λ6-hexahydrottøopyran-4-yl)-N-methylamino]ethyl, 2- [/V-(l-CH3SO2piperidin-4-yl)-iV-methylamino]ethyl, 2-[7V-(tetrahydropyran-3-yl)-iV- methylamino] ethyl, 2-[7V-(tetrahydrothiopyran-3-yl)-iV-methylammo]ethyl, 2-[N-(1, 1-dioxo- lλ6-hexahydrothiopyran-3-yl)-N-methylamino]ethyl, 2-[iV-(piperidin-3-yl)-7V- methylamino]ethyl5 2-[7V-(l-CH3COpiperidin-3-yl)-7V-methylamino]ethyl, 2-[N-(I- CF3COpiperidin-3-yl)-iV-methylamino]ethyl, 2-[7V-(l-CH3SO2ρiperidin-3-yl)-7V- methylamino]ethyl, 2-(N-CH3SO2-iV-methylamino)ethyl, 2-(iV-CF3SO2-N-methylamino)ethyl, 2-(7V-C2H5SO2-7V-methylamino)ethyl, 2-[iV-(CH3)2CHSO2-7V-methylamino]ethyl, 2-(N- cyclopropylSO2-N-methylamino)ethyl, 2-(N-cyclobutylSO2-7V-methylamino)ethyl, 2-(N- cyclopentylSO2-N-methylamino)ethyl, 2-(N-cyclohexylSO2-N-methylamino)ethyl, 2-(N- phenylSO2-N-methylamino)ethyl, 2-[N-(2-CH3ρhenylSO2)-N-methylamino]ethyl, 2-[N-(3- CH3phenylSO2)-N-methylamino]ethyl5 2-[N-(4-CH3phenylSO2)-N-methylamino]ethyl, 2-[N- (2-FphenylSO2)-N-methylamino]ethyl, 2-[N-(3-FphenylSO2)-N-methylamino]ethyl, 2-[N-(4- FphenylSO2)-N-methylamino]ethyl, 2-[N-(2-OHphenylSO2)-N-methylamino]ethyl, 2-[N-(3- OHphenylSO2)-N-methylamino]ethyl, 2-[N-(4-OHphenylSO2)-N-methylamino]ethyl, 2-[N- (2-CH3OρhenylSO2)-N-methylamino]ethyl, 2-[N-(3-CH3OphenylSO2)-N-methylamino]ethyl, 2-[iV-(4-CH3OphenylSO2)-N-methylamino]ethyl, 2-[N-(2-CO2HphenylSO2)-N- methylamino]ethyl, 2-[N-(3-CO2HphenylSO2)-N-methylamino]ethyl, 2-[N-(4-
CO2HphenylSO2)-N-methylamino]ethyl, 2-[N-(2-COΝH2phenylSO2)-N-methylamino]ethyl, 2-[N-(3-COΝH2phenylSO2)-N-methylamino]ethyl, 2-[N-(4-COΝH2phenylSO2)-N- methylamino]ethyl, 2-[N-(2-COΝ(CH3)2phenylSO2)-N-methylamino]ethyl, 2-[N-(3- COΝ(CH3)2phenylSO2)-N-methylamino]ethyl, 2-[N-(4-COΝ(CH3)2phenylSO2)-N- methylamino] ethyl, 2-[N-(methylphenylaminocarbonyl)-N-methylamino]ethyl, 2-{N-[(3-
COΝH2phenyl)methylΝCO]-N-methylamino}ethyl, 2-{N-[(4-COΝH2ρhenyl)methylΝCO]-N- methylamino}ethyl, 2-{N-[(2-COΝH2phenyl)methylΝCO]-N-methylamino}ethyl, 2-{N-[(3- Fphenyl)methylΝCO]-N-methylamino} ethyl, 2-{N-[(4-Fphenyl)methylΝCO]-N- methylamino} ethyl, 2-{N-[(2-Fphenyl)methylΝCO]-N-methylamino}ethyl, 2-{N-[(2- CH3Ophenyl)methylΝCO]-N-methylamino}ethyl5 2-{N-[(3-CΝphenyl)methylΝCO]-N- methylamino} ethyl, 2- {N-[(4-CΝphenyl)methylΝCO]-N-methylamino} ethyl, 2- {N-[(2- CΝphenyl)methylΝCO]-N-methylamino}ethyl, 2-{N-[(3-OHphenyl)methylΝCO]-N- methylamino}ethyl, 2-{N-[(4-OHphenyl)methylΝCO]-N-methylamino}ethyl, 2-{N-[(2- OHphenyl)methylNCO]-iV-methylamino}ethyl, 2-{iV-[(3-CH3Ophenyl)methylNCO]-iV- methylamino } ethyl, 2- (N- [(4-CH3Ophenyl)methylNCO]-N-methylamino } ethyl, 2- {iV- [(pyridin-2-yl)methylNCO]-N-methylamino}ethyl, 2-{N-[(pyridin-3-yl)methylNCO]-N- methylamino} ethyl, 2-{N-[φyridin-4-yl)methylNCO]-N-methylamino}ethyl, 2-{N- [(pyrimidin-2-yl)methylNCO]-iV-methylammo}ethyl, 2-{N-[(pyrimidin-4-yl)methylNCO]-N- methylamino} ethyl, 2-{N-[(pyrimidm-5yl)methylNCO]-N-methylamino}ethyl, 2-{iV- [([LS^J-triazin^-yOmethylNCOJ-iV-methylaminoJethyl^-fiV-tCS- CO2Hphenyl)methylNCO]-Λr-methylamino}ethyl, 2-{Λr-[(4-CO2Hphenyl)methylNCO]-iV- methylamino}ethyl, 2-{iV-[(2-CO2Hphenyl)methylNCO]-N-methylammo}ethyl, 2-(N- CH3OCO-N-methylamino)ethyl, 2-(N-C2H5OCO-N-methylamino)ethyl, 2-[N-
(CH3)2CHOCO-N-methylamino]ethyl, 2-[N-(CH3)3COCO-N-methylamino]ethyl, 2-(N- cyclopropylOCO-N-methylamino)ethyl, 2-(N-cyclobutylOCO-N-methylamino)ethyl, 2-[N- (oxetan-3-ylOCO)-N-methylamino]ethyl, 2-[N-(l-acetylazetidin-3-yl)-N-methylamino]ethyl, 2-[N-(CF3COazetidin-3-yl)-N-methylammo]ethyl5 2-[N-(l-CH3SO2azetidin-3-yl)-N- methylaminojethyl, 2-[ N-(l,l-dioxo-lλ6-thietan-3-ylOCO)methylamino]ethyl, 2-[ N-
(tetrahydrofuran-3-ylOCO)-N-methylamino]ethyl, 2-[ N-(tetrahydrothiophen-3-ylOCO)-N- methylamino] ethyl, 2-[N-(I, l-dioxo-lλ6-tetrahydrothiophen-3-ylOCO)-N- methylamino] ethyl, 2-[ N-(pyrrolidin-3-ylOCO)-N-methylamino] ethyl, 2-[ N-(I- CH3COpyrrolidin-3-ylOCO)-N-methylamino]ethyl, 2-[ N-(I -CFsCOpyrrolidin-3-ylOCO)- methylaminojethyl, 2-[ N-(l-CH3SO2pyrrolidin-3-ylOCO)-N-methylamino]ethyl, 2-[ N- (cyclohexylOCO)-N-methylamino]ethyl, 2-[ N-(tetrahydropyran-4-ylOCO)-N- methylamino] ethyl, 2-[ N-(tetrahydrothiopyran-4-ylOCO)-N-methylamino]ethyl, 2-[N-(I5I- dioxo-lλ6-hexahydrothiopyran-3-ylOCO)-N-methylamino]ethyl, 2-[ N-(piperidin-4-ylOCO)- N-methylamino]ethyl, 2-[ N-(l-CH3COpiperidin-4-ylOCO)-N-methylamino]ethyl, 2-[ N-(I- CF3COpiperidin-4-ylOCO)-N-methylamino]ethyl, 2-[ N-(I -CH3SO2piperidin-4-ylOCO)-N- methylamino]ethyl, 2-[ N-(benzylOCO)-N-methylamino]ethyl, 2-[ N-(2-CH3phenylmethyl- OCO)-N-methylamino]ethyl, 2-[ N-(3-CH3phenylmethylOCO)-N-methylamino]ethyl, 2-[ N- (4-CH3phenylmethylOCO)-N-methylamino]ethyl, 2-[ N-(2-Fphenylmethyl OCO)-N- methylamino]ethyl, 2-[ N-(3-FphenylmethylOCO)-N-methylamino] ethyl, 2-[ N-(4-Fphenyl- methylOCO)-N-methylamino]ethyl, 2-[ N-(2-OHphenylmethyl OCO)-N-methylamino]ethyl, 2-[ N-(3-OHphenylmethylOCO)-N-methylamino]ethyl, 2-[ N-(4-OHphenylmethylOCO)-N- methylamino]ethyl, 2-[ N-(2-CH3OphenylmethylOCO)-N-methylamino]ethyl, 2-[ N-(3- CH3OρhenylmethylOCO)-N-methylamino]ethyl, 2-[ N-(4-CH3OphenylmethylOCO)-N- methylaminojethyl, 2-[ N-(2-CNphenylmethyl OCO)-N-methylamino] ethyl, 2-[ iV-(3- CNphenylmethylOCO)-N-methylamino]ethyl, 2-[ N-(4-CNphenylmethylOCO)-iV- methylamino]ethyl, 2-[ N-(2-CO2HphenylmethylOCO)-N-methylaminoJethyl, 2-[ N-(3- CO2HphenylmethylOCO)-N-methylaminoJethyl, 2-[N-(4-CO2HphenylmethylOCO)-N- methylamino]ethyl, 2-[iV-(2-CONH2phenylmethylOCO)-iV-methylamino]ethyl, 2-[N-(3- CONH2phenylmethylOCO)-N-methylamino]ethyl, 2-[7V-(4-CONH2phenylmethylOCO)-7V- methylaminojethyl, 2-[N-(pyridin-2-ylmethyl OCO)-iV-methylamino] ethyl, 2-[iV-(pyridin-3- ylmethylOCO)-iV-methylamino]ethyl, 2-[N-(pyridin-4-ylmethylOCO)-iV-methylamino]ethyl, 2- [iV-(ρyrimidin-2-ylmethyl OCO)-N-methylamino]ethyl, 2- [iV-(pyrimidin-4-ylmethylOCO)- iV-methylamino] ethyl, 2-[iV-(pyrimidin-5-ylmethylOCO)-iV-methylamino]ethyl, 2-[iV- (pyrazin-2-ylmethyl OCO)-iV-methylamino]ethyl, 2-[iV-(pyridazin-3-ylmethylOCO)-N- methylamino] ethyl, 2-[N-(pyridazin-4-ylmethylOCO)-N-methylamino]ethyl, 2-[N- ([1.3.5]triazin-2-ylmethyl OCO)-N-methylamino]ethyl, 2-[N-(2-CH3phenylmethylΝHCO)-N- methylamino]ethyl, 2-[N-(3-CH3phenylmethylΝHCO)-N-methylamino]ethyl, 2-[N-(4- CH3ρhenylmethylΝHCO)-N-methylamino]ethyl, 2-[N-(2-FphenylmethylNHCO)-N- methylamino]ethyl, 2-[iV-(3-FphenylmethylΝHCO)-jV-methylamino]ethyl, 2-[N-(4- FphenylmethylNHCO)-N-methylamino]ethyl, 2-[N-(2-OHphenylmethylNHCO) -N- methylamino]ethyl, 2-[N-(3-OHρhenylmethylΝHCO)-N-methylamino]ethyl, 2-[N-(4- OHphenylmethylΝHCO)-N-methylamino]ethyl, 2-[N-(2-CH3OphenylmethylΝHCO)-N- methylaminojethyl, 2-[N-(3-CH3OphenylmethylΝHCO)-N-methylamino]ethyl, 2-[N-(4- CH3OphenylmethylΝHCO)-N-methylamino]ethyl, 2-[N-(2-CΝphenylmethylΝHCO)-N- methylaminojethyl, 2-[N-(3-CΝphenylmethylΝHCO)-N-methylamino]ethyl, 2-[N-(4- CΝphenylmethylΝHCO)-N-methylamino]ethyl, 2-[N-(2-CO2HphenylmethylΝHCO)-N- methylamino]ethyl, 2-[N-(3-CO2HphenylmethylΝHCO)-N-methylamino]ethyl, 2-[N-(4- CO2HphenylmethylΝHCO)-N-methylamino]ethyl, 2-[N-(2-COΝH2ρhenylmethylΝHCO)-N- methylamino]ethyl, 2-[N-(3-COΝH2phenylmethylΝHCO)-N-methylamino]ethyl, 2-[N-(4- COΝH2phenylmethylΝHCO)-N-methylamino]ethyl, 2-[N-(pyridin-2-ylmethylΝHCO)-N- methylamino]ethyl, 2-[N-(pyridin-3-ylmethylΝHCO)-N-methylamino]ethyl, 2-[N-(pyridin-4- ylmethylΝHCO)-N-methylamino]ethyl, 2-[N-(pyrimidin-2-ylmethylΝHCO)-N- methylaminojethyl, 2-[N-(pyrimidin-4-ylmethylΝHCO)-N-methylamino]ethyl, 2-[N-
(pyrimidin-5-ylmethylΝHCO)-N-methylamino]ethyl, 2-[N-(pyrazm-2-ylmethylΝHCO)-N- methylamino] ethyl, 2- [N-(pyridazin-3 -ylmethylΝHCO)-N-methylamino] ethyl, 2- [N- (pyridazin-4-ylmethylΝHCO)-N-methylamino]ethyl, 2-[N-([l .3.5]triazin-2-ylmethylΝHCO)- N-methylamino]ethyl, 2-[N-(2-CH3phenylmethylN(CH3)CO)-N-methylamino]ethyl, 2-[N-(3- CH3phenylmethylN(CH3)CO)-iV-methylamino]ethyl, 2-[iV-(4-CH3phenylmethylN(CH3)CO)- N-methylamino]ethyl, 2-[iV-(2-FphenylmethylN(CH3)CO)-iV-methylamino]ethyl, 2-[7V-(3- FphenylmethylN(CH3)CO)-7V-methylamino]ethyl, 2-[N-(4-FphenylmethylN(CH3)CO)-iV- methylamino]ethyl, 2-[7V-(2-OHphenylmethylN(CH3)CO)-N-methylammo]etliyl, 2-[iV-(3- OHplienylmethylN(CH3)CO)-N-methylamino]ethyl, 2-[iV-(4-OHphenylmethylN(CH3)CO)- N-methylamino]ethyl, 2-[N-(2-CH3OρhenylmethylN(CH3)CO)-iV-methylamino]etliyl, 2-[N- (3-CH3OphenylmethylN(CH3)CO)-iV-methylammo]ethyl, 2-[iV-(4-
CH3OphenylmethylN(CH3)CO)-iV-methylamino]ethyl, 2-[N-(2-CNphenylmethylN(CH3)CO)- N-methylamino]ethyl, 2-[Λr-(3-CNphenylmethylN(CH3)CO)-N-methylaniino]ethyl, 2-[N-(A- CNphenylmethylN(CH3)CO)-N-methylamino]ethyl, 2-[iV-(2-CO2HphenylmethylN(CH3)CO)- N-methylamino]ethyl, 2-[iV-(3-CO2HphenylmethylN(CH3)CO)-iV-metliylammo]ethyl, 2-[iV- (4-CO2HphenylmethylN(CH3)CO) -N-methylamino]ethyl, 2-[N-(2-CONH2ρhenylmethyl- N(CH3)CO)-N-methylamino]ethyl, 2-[iV-(3-CONH2phenylmethylN(CH3)CO)-iV-methyl- aminojethyl, 2-[N-(4-CONH2phenylmethylN(CH3)CO)-iV-methylamino]etliyl, 2-[N-(pyridin- 2-ylmethylN(CH3)CO)-iV-methylamino]ethyl, 2-[iV-(pyridin-3-ylmethylN(CH3)CO)-N- methylamino]ethyl, 2-[N-(pyridin-4-ylmethylN(CH3)CO)-iV-methylamino]ethyl, 2-[iV- (pyrimidin-2-ylmethylN(CH3)CO)-Λ/-methylamino]ethyl, 2-[N-(pyrimidin-4-ylmethyl- N(CH3)CO)-iV-methylamino]ethyl, 2-[iV-(pyrimidin-5-ylmethylN(CH3)CO)-iV-methyl- amino]ethyl, 2-[iV-(pyrazin-2-ylmethylN(CH3)CO)-iV-methylamino]ethyl, 2-[iV-(pyridazin-3- ylmethylN(CH3)CO)-N-methylamino]ethyl, 2-[iV-(pyridazin-4-ylmetliylN(CH3)CO) JV- methylamino]ethyl, 2-[iV-([l .3.5]triazin-2-ylmethyl-iV-(CH3)CO)-iV-methylamino]ethyl, 2-[iV- (pyridin-4-yl)-iV-methylamino] ethyl, 2- [iV-(pyridin-3 -yl)-jV-methylamino]ethyl, 2- [iV- (pyridin-2-yl)-N-methylamino]ethyl, 2-[N-(pyrimidin-4-yl)-iV-methylamino]ethyl, 2-[N- (pyrimidin-2-yl)-N-methylamino]ethyl, 2-[N-(pyrimidin-5-yl)-N-niethylamino]ethyl, 2-[N- ([1.3.5]triazin-2-yl)-N-methylamino]ethyl5 2-[N-(phenyl)-N-methylamino]ethyl, 2-[N- (pyrazin-2-yl)-N-methyl-amino]ethyl, 2-[N-(pyridazin-4-yl)-N-metliylammo]ethyl, 2-[N- (pyridazin-3-yl)-N-methyl-amino] ethyl, 2-[N-(4-F-phenyl)-N-methylamino]ethyl, 2-[N-(3-F- phenyl)-N-methylamino]ethyl 2-[N-(2-F-phenyl)-N-methylamino]ethylJ 2-[N-(2,4-diF- ρhenyl)-N-methylamino]ethyl, 2- [N-(2,3 -diF-phenyl)-N-methylamino]ethyl, 2- [N-(2,5-diF- phenyl)-N-methylamino]ethyl, 2-[N-(2,6-diF-phenyl)-N-methylamino]ethyl, 2-[N-(2,4,6-triF- phenyl)-N-methylamino] ethyl, 2-[N-(2,3,6-triF-phenyl)-N-methylamino]ethyl, 2-[N-(2,3,4- triF-phenyl)-N-methylamino]ethyl, 2-[N-(4-CH3O-phenyl)-N-methylamino]ethyl, 2-[N-(3- CH3O-phenyl)-N-methylamino]ethyl, 2-[N-(2-CH3O-phenyl)-N-methylamino]ethyl, 2-[N-(4- CΝ-phenyl)-N-methylamino]ethyl5 2-[N-(3-CΝ-phenyl)-N-methylamino]ethyl, 2-[N-(2-CΝ- phenyl)-N-methylamino]ethyl, 2-[N-(4-CO2H-phenyl)-N-methylamino]ethyl, 2-[N-(3-CO2H- phenyl)-N-methylamino]ethyl, 2-[N-(2-CO2H-phenyl)-N-methylammo]ethyl, 2-[N-(4- CONH2-phenyl)-N-methylamino]ethyl5 2-[iV-(3-CONH2-phenyl)-iV-metliylamino]ethyl, 2-[iV- (2-CONH2-phenyl)-N-methylamino]ethyl, 2-(iV-CH3CO-N-methylamino)ethyl, 2-(N-CF3CO- N-methylamino)ethyl, 2-(N-C2H5CO-N-methylamino)ethyl5 2-[N-(CH3)2CHCO-N- methylaminojethyl, 2-[N-(CH3)3CCO-N-methylamino]ethyl, 2-(iV-cyclopropylCO-N- methylamino)ethyl, 2-(N-cyclobutylCO-N-methylamino)ethyl, 2-[N-(oxetan-3-ylCO)-N- methylamino]ethyl, 2-[N-(azetidin-l-ylCO)-iV-methylamino]ethyl, 2-[N-(1, 1-dioxo-lλ6- tetrahydrothiophen-3-ylCO)-N-methylamino]ethyl, 2-[N-(azetidin-3-ylCO)-N- methylatnino]ethyl, 2-[N-(I -acetylazetidin-3-ylCO)-N-methylamino]ethyl, 2-[N-
(CF3COazetidin-3-ylCO)-N-methylamino]ethyl, 2-[N-(l-CH3SO2azetidin-3-ylCO)-N- methylamino] ethyl, 2-(N-cyclopentylCO-N-methylammo)ethyl, 2-[N-(3-CH3θpyrrolidin-l- ylCO)-N-methylamino]ethyl, 2-[N-(3-CF3OρyπOlidin-l-ylCO)-N-methylamino]ethyl5 2-[N- (3-CHF2Opyrrolidin-l -ylCO)-N-methylamino]ethyl, 2-[N-(3-phenoxypyrrolidin-l -ylCO)-N- methylamino] ethyl, 2-{N-[3-(4-Fphenoxy)pyrrolidin-l-ylCO]-N-methylamino}ethyl, 2-{N- [3-(4-Clphenoxy)pyrrolidin- 1 -ylCO] -N-methylamino } ethyl, 2- {N-[3 -(4- Brphenoxy)pyrrolidin- 1 -ylCO] -N-methylamino } ethyl, 2- {N-[3 -(COOH-pyrrolidin- 1 -ylCO]- N-methylamino } ethyl, 2- {N-[3 -(CΝ-pyrrolidin- 1 -ylCO] -N-methylamino} ethyl, 2- {N- [3 - (COΝH2-phenoxy)cyclopentylCO] -N-methylamino } ethyl, 2- {N-[3 -(COΝHCHrpyrrolidin- 1 - ylCOj-N-methylaminoJethyl^-IN-P^COΝ^Hs^-pyrrolidin-l-ylCOJ-N- methylamino} ethyl 2-[N-(pyrrolidin- 1 -ylCO)-N-methylamino]ethyl, 2-[N-(morpholin-4- ylCO)-N-methylamino]-ethyl, 2-[N-(l-oxazolidin-3-ylCO)-N-methylamino]ethyl, 2-[N- (tetrahydrofuran-3-ylCO)-N-methylamino]ethyl, 2-[N-(tetraliydrothiophen-3-ylCO)-N- methylamino] ethyl, 2-[N-(I, l-dioxo-lλ6-tetrahydrothiophen-3-ylCO)-N-methylamino]ethyl, 2-(N-cyclohexylCO-N-methylamino)-ethyl, 2-[N-(tetrahydropyran-4-ylCO)-N- methylaminojethyl, 2-[N-(piperidin-4-ylCO)-N-methylamino]ethyl, 2-[N-(I- CH3COpiperidm-4-ylCO)-N-methylamino]ethyl, 2-[N-(l-CF3COpiperidin-4-ylCO)-N- methylamino]ethyl, 2-[N-(I -CH3SO2piperidin-4-ylCO)-N-methylamino]ethyl, 2-[N- (tetrahydrothiopyran-4-ylCO)-N-methylamino] ethyl, 2- [N-( 1 , 1 -dioxo- 1 λ6- hexahydrothiopyran-4-ylCO)-N-methylamino]ethyl5 2-[N-(piperidin-l-ylCO)-N- methylamino]ethyl, 2-[N-(thiomorpholin-4-ylCO)-N-methylamino]ethyl, 2-[N-(1 , 1- dioxothiomorpholin-4-ylCO)-N-methylamino]ethyl, 2-[N-(piperazin-4-ylCO)-N- methylaminojethyl, 2-[N-(4-CH3COpiperazin-l-ylCO)-N-methylamino]ethyl, 2-[N-(4- CF3COpiperazin-l-ylCO)-N-methylamino]ethyl3 2-[N-(CH3SO2piperazin-l-ylCO)-N- methylaminojethyl, 2-[iV-(phenylCO)-iV-methylammo]ethyl, 2-[iV-(2~FphenylCO)-iV- methylaminojethyl, 2-[JV-(3-FphenylCO)-JV-methylamino]ethyl, 2-[iV-(4-FphenylCO)-iV- methylammo]ethyl, 2-[N-(2-OHphenylCO)-N-methylamino]ethyl, 2-[N-(3-OHphenylCO)-N- methylamino]ethyl, 2-[N-(4-OHphenylCO)-N-methylamino]ethyl, 2-[JV-(2-CH3OphenylCO)- iV-methylamino]ethyl, 2-[N-(3-CH3OphenylCO)-JV-methylamino]ethyl, 2-[iV-(4- CH3OphenylCO)-N-methylamino]ethyl, 2-[N-(2-CO2HphenylCO)-N-methylamino]ethyl, 2- [N-(3-CO2HphenylCO)-N-methylamino]ethyl, 2-[7V-(4-CO2HphenylCO)-iV- methylamino]ethyl, 2-[iV-(2-CNphenylCO)-iV-methylammo]ethyl3 2-[N-(3-CNphenylCO)-iVr- methylamino]ethyl, 2-[iV-(4-CNphenylCO)-iV-methylamino]ethyl, 2-[iV-(2-CH3phenylCO)-iV- methylamino]ethyl, 2-[N-(3-CH3phenylCO)-N-methylamino]ethyl, 2-[iV-(4-CH3phenylCO)- iV-methylamino] ethyl, 2-[N-(2-CONH2phenylCO)-iV-methylamino]ethyl, 2-[N-(3- CONH2phenylCO)-N-methylamino]ethyl, 2-[N-(4-CONH2ρhenylCO)-N-methylamino]ethyl, 2-[Λr-(pyridin-2-ylCO)-Λr-ttiethylamino]ethyl, 2-[iV-(pyridm-3-ylCO)-iV-methylamino]ethyl, 2-[Λr-(pyridin-4-ylCO)-iV-methylamino]ethyl, 2-[iV-(pyrimidin-4-ylCO)-Λr- methylamino]ethyl, 2-[iV-(pyrimidin-2-ylCO)-iV-methylamino]ethyl, 2-[iV-(pyrimidin-5- ylCO)-iV-methylamino] ethyl, 2-[iV-(benzylCO)-iV-methylamino]ethyl, 2-[N-(3- CONH2phenylmethylCO)-iV-methylamino]ethyl, 2-[iV-(4-CONH2phenylmethylCO)-N- methylamino]ethyl, 2-[iV-(2-CONH2phenylmethylCO)-N-methylamino]ethyl, 2-[N-(3- FphenylmethylCO)-N-methylamino]ethyl, 2-[iV-(4-FphenylmethylCO)-N-methylamino]ethyl, 2-[N-(2-FphenylmethylCO)-//-methylamino]ethyl, 2-[iV-(2-CH3OphenylmethylCO)-iV- methylaminojethyl, 2-[iV-(3-CH3OphenylmethylCO)-iV-methylamino]ethyl, 2-[iV-(4- CH3OρhenylmethylCO)-7V-methylamino]ethyl, 2-[N-(2-CNphenylmethylCO)-N- methylamino]ethyl, 2-[iV"-(3-CNphenylmethylCO)-N-methylamino]ethyl, 2-[iV-(4- CNphenylmethylCO)-iV-inethylamino]ethyl, 2-[iV-(2-OHphenylmethylCO)-iV- methylaminojethyl, 2-[N-(3-OHphenylmethylCO)-N-methylamino]ethyl, 2-[iV-(4- OHphenylmethylCO)-iV-rQethylammo]ethyl, 2-[N-(2-CO2HphenylmethylCO)-N- methylamino]ethyl, 2-[N-(3-CO2HphenylmethylCO)-iV-methylamino]ethyl, 2-[N-(4- CO2HphenyImethylCO)-iV-methylamino]ethyl, 2-[7V-(pyridin-2-ylmethylCO)-N- methylaminojethyl, 2-[N-(pyridin-3-ylmethylCO)-N-methylamino]ethyl, 2-[N-(pyridin-4- ylmethylCO)-N-methylamino]ethyl, 2-[iV-(pyrimidin-2-ylmethylCO)-iV-methylamino]ethyl, 2-[N-(pyrimidin-4-ylmethylCO)-iV-methylamino]ethyl, 2-[N-(pyrimidin-5-ylmethylCO)-N- methylamino]ethyl; 2-[N-(pyrazin-2-ylmethylCO)-iV-methylanimo]ethyl 2-[iV- (phenylNHCO)-N-methylamino]ethyl, 2-[N-(3-CONH2phenylNHCO)-iV-methylamino]ethyl, 2-[iV-(4-CONH2phenylNHCO)-iV-methylamino]ethyl, 2-[iV-(2-CONH2phenylNHCO)-iV- methylaminojethyl, 2-[iV-(3-FphenylNHCO)-iV-methylamino]ethyl, 2-[N-(4-FphenylNHCO)- 7V-methylamino]ethyl, 2-[N-(2-FphenylNHCO)-N-methylamino]ethyl, , 2-[iV-(2- CH3OphenylNHCO)-iV-methylammo]ethyl, 2-[iV-(3-CH3OphenylNHCO)-iV- methylamino]ethyl, 2-[iV-(4-CH3OphenylNHCO)-iV-metliylammo]ethyl5 2-[N-(2-
CNphenylNHCO)-N-methylaniino]ethyl, 2-[iV-(3-CNphenylNHCO)-iV-methylamino]ethyl, 2- [iV-(4-CNphenylNHCO)-iV-methylamino]ethyl, 2-[N-(2-OHphenylNHCO)-iV- methylaminojethyl, 2-[7V-(3-OHphenylNHCO)-iV-methylamino]ethyl, 2-[iV-(4-OHphenyl- NHCO)-iV-methylamino]ethyl, 2-[iV-(2-CO2HplienylNHCO)-iV-methylamino]ethyl, 2-[N-(3- CO2HphenylNHCO)-iV-metliylamino]ethyl, 2-[iV-(4-CO2HphenylNHCO)-iV- methylamino]ethyl, 2-[N-(pyridin-2-ylNHCO)-iV-methylammo]ethyls 2-[N-(pyridin-3- ylNHCO)-iV-methylamino]ethyl, 2-[iV-(pyridin-4-ylNHCO)-iV-methylamino]ethyl, 2-[N- (ρyrimidin-2-ylNHCO)-N-methylamino] ethyl, 2- [N-(pyrimidin-4-ylNHCO)-iV- methylamino]ethyl, 2-[iV-(ρyrimidin-5-ylNHCO)-iV-methylamino]ethyl, 2-[N-([\ .3.5]triazin- 2-ylNHCO)-N-methylamino]ethyl, 2-[ iV-(l,2,3,4-tetraliydroquinolin-l-ylCO)-iV- methylamino]ethyl, 2-[N-(7-CONH2- 1 ,2,3,4-tetrahydroquinolin- 1 -ylCO)-N- methylamino]ethyl, 2-[iV-(6-CONH2-l,2,3,4-tetrahydroquinolin-l-ylCO)-iV- methylamino]ethyl, 2-[N-(8-CONH2-l,2,3,4-tetrahydroquinolin-l-ylCO)-N- methylamino]ethyl, 2-[N-(7-F-l,2,3,4-tetrahydroquinolin-l-ylCO)-N-methylamino]ethyl, 2- [N-(6-F-l,2,3,4-tetrahydroquinolin-l-ylCO)-N-methylamino]ethyl3 2-[N-(8-F-l,2,3,4- tetrahydroquinolin-1 -ylCO)-iV-methylamino]ethyl, 2-[iV-(7-CH3O-l ,2,3,4-tetraliydroquinolin- 1 -ylCO)-iV-methylamino]ethyl, 2-[iV-(6-CH3O- 1 ,2,3 ,4-tetrahydroquinolin- 1 -ylCO)-iV- methylamino]ethyl, 2-[N-(8-CH3O- 1 ,2,3 ,4-tetrahydroquinolin- 1 -ylCO)-N-methylamino]ethyl, 2-[N-(7-CN-l,2,3,4-tetrahydroquinolin-l-ylCO)-iV-methylamino]ethyl5 2-[N-(6-CN-l,2,3,4- tetrahydroquinolin-l-ylCO)-Λ/-methylamino]ethyl, 2-[iV-(8-CN-l,2,3,4-tetrahydroquinolin-l- ylCO)-iV-methylamino]ethyl, 2-[iV-(7-OH-l,253,4-tetrahydroquinolin-l-ylCO)-N- methylaminojethyl, 2-[7vr-(6-OH-l,2,3,4-tetrahydroquinolin-l-ylCO)-N-methylarnino]ethyl, 2-[/V-(8-OH- 1 ,2,3 ,4-tetrahydroquinolin- 1 -ylCO)-N-methylamino]ethyl, 2-[iV-(7-CO2H- 1 ,2,3 ,4-tetrahydroquinolin- 1 -ylCO)-iV-methylamino] ethyl, 2- [JV-(6-CO2H- 1 ,2,3 ,4- tetrahydroquinolin-1 -ylCO)-N-methylamino]ethyl, 2-[JV-(8-CO2H-l ,2,3,4-tetrahydroquinolin- l-ylCO)-iV-methylamino]ethyl, 2-[iV-(l,2,3,4-tetrahydro-[l,8]naphthyridin-l-ylCO)-N- methylamino]ethyl, 2-[N-(1 ,2,3,4-tetrahydro-[l ,7]naphthyridin-l -ylCO)-iV- methylamino] ethyl, 2-[N-(l,2,3,4-tetrahydro-[l,6]naphthyridin-l-ylCO)-iV- methylamino]ethyl, 2-[iV-(5,6,7,8-tetrahydropyrido[2,3-b]pyrazin-5-ylCO)-jV- methylamino]ethyl, 2-[N-(5,6,7,8-tetrahydropyrido[2,3-d]pyrimidin-8-ylCO)-iV- methylaminojethyl, 2-[N-(5,6,758-tetrahydropyrido[3,2-d]pyrimidin-5-ylCO)-N- methylaminojethyl, 2-[iV-(5,6,7,8-tetrahydropyrido[2,3-d]pyridazin-l-ylCO)-N- methylamino]ethyl, 2-[ N-(2,3-dihydro-lH-indol-l-ylCO)methylamino]ethyl, 2-[iV-(7- CONH2-2,3-dihydro-lH-indol-l-ylCO)-iV-methylammo]-ethyl, 2-[iV-(6-CONH2-2,3-dihydro- lH-indol-l-ylCO)-iV-methylammo]ethyl, 2-[iV-(5-CONH2-253-dihydro-lH'-indol-l-ylCO)-iV- methylamino]ethyl, 2-[N-(7-F-2,3-dihydro-lH-indol-l-ylCO)-N-methylamino]ethyl, 2-[N-(6- F-2,3-dihydro-lH-indol-l-ylCO)-iV-methylamino]ethyl, 2-[iV-(5-F-2,3-dihydro-lH-indol-l- ylCO)-iV-methylamino]ethyl, 2-[iV-(7-CH3O-2,3-dihydro-l/f-indol-l-ylCO)-iV- methylaminojethyl, 2-[N-(6-CH3O-2,3-dihydro-lH-indol-l-ylCO)-iV-methyl-ammo]ethyl, 2- [N-(5-CH3O-2,3-dihydro-lif-indol-l-ylCO)-iV-methylammo]ethyl, 2-[iV-(7-CN-2,3-dihydro- lH-indol-l-ylCO)-iV-methylamino]ethyl, 2-[N-(6-CN-2,3-dihydro-l/f-mdol-l-ylCO)-N- methylaminojethyl, 2-[N-(5-CN-2,3-dihydro-lH-indol-l-ylCO)-N-methylamino]ethyl, 2-[iV- (7-OH-2,3-dihydro-l//-indol-l-ylCO)-7V-methylamino]ethylJ 2-[iV-(6-OH-2,3-dihydro-l//- indol- 1 -ylCO)-Λr-methylamino]ethyl, 2-[iV-(5-OH-2,3-dihydro- 1/f-indol- 1 -ylCO)-iV- methylaminojethyl, 2-[iV-(7-CO2H-2,3-dihydro-lH-indol-l-ylCO)-iV-methylamino]ethyl, 2- [iV-(6-CO2H-2,3-dihydro-lH-indol-l-ylCO)-iV-methylamino]ethyl, 2-[iV-(5-CO2H-2,3- dihydro- 1/f-indol- 1 -ylCO)-N-methylamino] ethyl, 2-[iV-(2,3 -dihydro- 1 H-pyrrolo [2,3 - b]pyridin- 1 -ylCO)-iV-methylamino] ethyl, 2- [N-(2,3 -dihydro- 1 H-pyrrolo[2,3 -c]pyridin- 1 - ylCO)-iV-methylamino]ethyl, 2-[iV-(2,3-dihydro-lH-pyrrolo[3,2-c]pyridin-l-ylCO)-N- methylamino]ethyl, 2-[iV-(6,7-dihydro-51H-pyrrolo[2,3-b]pyrazin-5-ylCO)-iVr- methylaminojethyl, 2-[iV-(6,7-dihydro-5H-pyrrolo[2,3-d]pyrimidin-7-ylCO)-jV- methylamino]ethyl, 2-[N-(6,7-dihydro-5H-pyrrolo[3,2-d]pyrimidin-5-ylCO)-iV- methylamino]ethyl, 2-[iV-(2,3 -dihydro- 1 H-pyrrolo [2,3 -d]pyridazin- 1 -ylCO)-JV- methylamino]ethyl, 2-aminoethyl, 2-iV-methylaminoethyl 2-iV-ethylaminoethyl, 2-(2,2,2- trifluoroethylamino)ethyl, 2-iV-isopropylaminoethyl, 2-[2-CF3-2,2,2- trifluoroethylamino)ethyl, 2-(iV-cyclopropylammo)ethyl, 2-(iV-cyclobutylamino)ethyl, 2-[N- (oxetan-3-yl)amino]ethyl, 2-[N-(azetidin-3-yl)amino]ethyl, 2-[ 7V-(l,l-dioxo-lλ6-thietan-3- yl)amiiio]ethyl, 2-(iV-cyclopentylamino)ethyl, 2-[iV-(3-CH3Ocyclopentyl)amino]ethyl, 2-[N- (3-CHF2Ocyclopentyl)amino]ethyl, 2-[JV-(3-CF3Ocycloρentyl)amino]ethyl, 2-[N-(3-phenoxy- cyclopentyl)amino)ethyl, 2-{iV-[3-(4-Fphenoxy)cyclopentyl]amino}ethyl, 2-{iV-[3-(4- Clphenoxy)cyclopentyl]amino}ethyl, 2-{iV-[3-(4-Brphenoxy)cyclopentyl]ammo}ethyl, 2-{N- [3-(COOH-phenoxy)cyclopentyl]amino}ethyl, 2-{iV-[3-(CN- phenoxy)cyclopentyl] amino } ethyl, 2- (N- [3 -(CONH2-phenoxy)cyclopentyl] amino } ethyl, 2- (N-cyclohexylamino)ethyl, 2-[iV-(tetrahydropyran-4-yl)amino]ethyl, 2-[N-(piperidin-4- yl)amino]ethyl, 2-[N-(I -acetylpiperidin-4-yl)amino]ethyl, 2-[N-(I -CF3COpiperidin-4- yl)amino]ethyl, 2-[N-(tetrahydrothiopyran-4-yl)amino]ethyl, 2-[N-(1, 1-dioxo-lλ6- hexahydrothiopyran-4-yl)amino]ethyl, 2-[N-(I -CH3 SO2piperidin-4-yl)amino] ethyl, 2-[N- (tetrahydropyran-3 -yl)amino] ethyl, 2- [N-(tetrahydrothiopyran-3 -yl)amino] ethyl, 2- [N-( 1 , 1 - dioxo-lλ6-hexahydrotm'opyran-3-yl)ammo]ethyl, 2-[N-(piperidin-3-yl)amino]ethyl, 2-[N-(I- CH3COpiperidin-3-yl)ammo]ethyl5 2-[N-(l-CF3COpiperidin-3-yl)ammo]ethyl, 2-[N-(I- CH3SO2piρeridin-3-yl)amino]ethyl, 2-(N-CH3 SO2amino)ethyl, 2-(N-CF3SO2amino)ethyl, 2- (N-C2H5SO2amino)ethyl, 2-[N-(CH3)2CHSO2amino]ethyl, 2-[N-(CH3)3CSO2amino]ethyl, 2- (N-cyclopropylSO2amino)ethyl, 2-(N-cyclobutylSO2amino)ethyl, 2-(N- cyclopentylSO2amino)ethyl, 2-(N-cyclohexylSO2-amino)ethyl, 2-(N-phenylamino)ethyl, 2- [N-(2-CH3phenylSO2)amino]ethyl, 2-[N-(3-CH3ρhenylSO2)amino]ethyl, 2-[N-(4- CH3phenylSO2)amino]ethyl, 2-[N-(2-FphenylSO2)-amino]ethyl, 2-[N-(3- FphenylSO2)amino]ethyl, 2-[N-(4-FphenylSO2)amino]ethyl, 2-[N-(2-
OHphenylSO2)amino]ethyl, 2-[N-(3-OHphenylSO2)amino]ethyl, 2-[N-(4-OHphenylSO2)- amino]ethyl, 2-[N-(2-CH3OphenylSO2)amino]ethyl, 2-[N-(3-CH3OphenylSO2)amino]ethyl, 2-[N-(4-CH3OphenylSO2)amino]ethyl, 2-[N-(2-CO2HphenylSO2)amino]ethyl, 2-[N-(3- CO2HphenylSO2)amino]ethyl, 2-[N-(4-CO2HphenylSO2)amino]ethyl, 2-[N-(2- COΝH2phenyl-SO2)amino]ethyl, 2-[N-(3-CONH2phenylSO2)amino]ethyl, 2-[N-(4-
COΝH2phenylSO2)-amino]ethyl, 2-[N-(2-COΝ(CH3)2phenylSO2)amino]ethyl, 2-[N-(3- COΝ(CH3)2phenylSO2)-ammo]ethyl, 2-[N-(4-COΝ(CH3)2phenylSO2)amino]ethyl, 2-[N- (methylphenylaminocarbonyl)-amino]ethyl, 2-{N-[(3- COΝH2phenyl)methylΝCO]amino}ethyl, 2-{N-[(4-COΝH2phenyl)- methylNCO]amino} ethyl, 2-{N-[(2-CONH2phenyl)methylNCO]amino}ethyl, 2-{N-[(3- Fphenyl)methylΝCO]amino}ethyl, 2-{N-[(4-Fphenyl)methylΝCO]amino}ethyl, 2-{N-[(2- Fphenyl)methylΝCO]amino}ethyl, 2-{N-[(2-CH3Ophenyl)methylΝCO]amino}ethyl, 2-{N- [(3-CΝphenyl)methylΝCO]amino}ethyl, 2-{N-[(4-CΝphenyl)methyIΝCO]amino}ethyl, 2- {N-[(2-CNphenyl)methylNCO]amino}ethyl, 2-{N-[(3-OHphenyl)methylNCO]amino}ethyl, 2-{N-[(4-OHphenyl)methylNCO]amino}ethyl, 2-{N-[(2-
OHphenyl)methylΝCO]amino}ethyl, 2-{N-[(3-CH3Ophenyl)methylΝCO]amino}ethyl, 2-{N- [(4-CH3Oρhenyl)methylΝCO]amino}ethyl, 2-{N-[(pyridin-2-yl)methylNCO]amino}ethyl, 2- {N- [(pyridin-3 -yl)methylNCO] amino } ethyl, 2- {N- [(pyridin-4-yl)methylΝCO]amino } ethyl, 2-{7V-[(pyrimidin-2-yl)methylNCO]amino}ethyl, 2-{iV-[(pyrimidin-4- yl)methylNCO]amino}ethyl5 2-{τV-[(pyrimidin-5yl)methylNCO]amino}ethyl, 2-{N-[([l .3.5]- triazin-2-yl)methylNCO]ammo}ethyl, 2-{iV-[(3-CO2Hphenyl)methyl-NCO]amino}ethyl, 2- {iV-[(4-CO2Hphenyl)methylNCO]amino}ethyl, 2-{N-[(2- CO2Hρhenyl)methylNCO]amino} ethyl, 2-(N-CH3OCOamino)ethyl, 2-(N-
C2H5OCOamino)ethyl 2-[N-(CH3)2CHOCOamino]ethyl, 2-[N-(CH3)3COCOamino]ethyl, 2- (N-cyclopropylOCO-amino)ethyl, 2-(7V-cyclobutylOCOamino)ethyl, 2-[N-(oxetan-3-ylOCO) amino]ethyl, 2-[N-(azetidin-3-yl)amino]ethyl, 2-[N~(l-acetylazetidin-3-yl)amino]ethyl, 2-[N- (CF3COazetidin-3-yl)amino]ethyl, 2-[N-(l-CH3SO2azetidin-3-yl)amino]ethyl, 2-[ N-(I3I- dioxo-lλ6-thietan-3-ylOCO)amino]ethyl, 2-[ N-(tetrahydrofuran-3-ylOCO)amino]ethyl, 2-[ N-(tetrahydrothiophen-3-ylOCO)amino]ethyl, 2-[iV-(l,l-dioxo-lλ6-tetrahydrothiophen-3- ylOCO)amino]ethyl 2-[ 7V-(pyrrolidin-3-ylOCO)amino]ethyl, 2-[ N-(I -CH3 COpyrrolidin-3- ylOCO)amino]ethyl 2-[ N-(l-CF3COpyrrolidin-3-ylOCO)amino]ethyl, 2-[ N-(I- CH3SO2pyrrolidin-3-ylOCO)amino]ethyl, 2-[ N-(cyclohexylOCO)amino]ethyl, 2-[ N- (tetrahydropyran-4-ylOCO)amino]ethyl, 2-[ N-(tetrahydrothiopyran-4-ylOCO)amino]ethyl, 2- [N-( 1 , 1 -dioxo- 1 λ6-hexahydrothiopyran-3 -ylOCO)-amino]ethyl, 2- [ N-(piperidin-4- ylOCO)amino]ethyl, 2-[ N-(l-CH3COpiperidin-4-ylOCO)amino]ethyl, 2-[ N-(I- CF3COpiperidin-4-ylOCO)amino]ethyl, 2-[ N-(l-CH3SO2piperidin-4-ylOCO)amino]ethyl, 2- [ N-(benzylOCO)amino]ethyl, 2-[ N-(2-CH3phenylmethylOCO)amino]ethyl, 2-[ N-(3- CH3phenylmethylOCO)amino]ethyl, 2-[ N-(4-CH3phenylmethylOCO)amino]ethyl, 2-[ N-(2- Fphenylmethyl OCO)amino] ethyl, 2-[ N-(3-FphenylmethylOCO)amino]ethyl, 2-[ N-(4- FphenylmethylOCO)amino] ethyl, 2-[ N-(2-OHphenylmethyl OCO)amino]ethyl5 2-[ N-(3- OHphenylmethylOCO)amino] ethyl, 2-[ N-(4-OHphenylmethylOCO)amino]ethyl, 2-[ N-(2- CH3OρhenylmethylOCO)amino]ethyl, 2-[ N-(3-CH3OphenylmethylOCO)amino]ethyl, 2-[ N- (4-CH3OphenylmethylOCO)amino]ethyl; 2-[ N-(2-CΝρhenylmethyl OCO)amino] ethyl, 2-[ N-(3-CNphenylmethylOCO)amino]ethyl, 2-[ N-(4-CNphenylmethylOCO)amino]ethyl, 2-[ N- (2-CO2HphenylmethylOCO)amino]ethyl, 2-[ N-(3-CO2HphenylmethylOCO)amino]ethyl, 2- [N-(4-C02HphenylmethylOCO)amino]ethyl, 2-[N-(2- COΝH2phenylmethylOCO)amino]ethyl, 2-[N-(3-COΝH2phenylmethylOCO)amino]ethyl, 2- [N-(4-CONH2phenylmethylOCO)amino]ethyl, 2-[N-(pyridin-2-ylmethyl OCO)amino]ethyl, 2-[N-(ρyridin-3-ylmethylOCO)amino]ethyl, 2-[N-(pyridin-4-ylmethylOCO)amino]ethyl, 2- [N-(pyrimidin-2-ylmethyl OCO)amino]ethyl, 2-[N-(pyrimidin-4-ylmethylOCO)amino]ethyl, 2- [N-(pyrimidin-5-ylmethylOCO)amino]ethyl, 2- [N-(ρyrazin-2-ylmethyl OCO)amino]ethyl, 2-[iV-(pyridazin-3-ylmethylOCO)amino]ethyl, 2-[N-(pyridazin-4-ylmethylOCO)amino]ethyl, 2-[JV-([1.3.5]triazin-2-ylmethyl OCO)amino]ethyl, 2-[iV-(2-
CH3phenylmethylΝHCO)amino]ethyl, 2-[iV-(3-CH3plienylmethyIΝHCO)amino]ethyl, 2-[iV- (4-CH3phenylmethylNHCO)amino]ethyl, 2-[N-(2-FphenylmethylNHCO)amino]ethyl, 2-[iV- (3-FphenylmethylNHCO)amino]ethyl, 2-[N-(4-FphenylmethylNHCO)amino]ethyl, 2-[iV-(2- OHphenylmethylNHCO)amino]ethyl, 2-[N-(3-OHphenylmethylNHCO)amino]ethyl, 2-[iV-(4- OHphenylmethylNHCO)amino]ethyl, 2-[iV-(2-CH3OphenylmethylNHCO)amino]ethyl, 2-[7V- (3-CH3OρhenylmethylNHCO)amino]ethyl, 2-[N-(4-CH3OρhenylmethylNHCO) amino]ethyl, 2-[iV-(2-CNphenylmethylNHCO) amino]ethyl, 2-[iV-(3-CNphenylmethylNHCO)amino]ethyl, 2-[N-(4-CNphenylmethylNHCO)amino]ethyl, 2-[N-(2-
C02HphenylmethylNHCO)amino]ethyl, 2-[iV-(3-C02HphenylmethylNHCO)ammo]ethyl, 2- [iV-(4-CO2HphenylmethylNHCO)amino]ethyl, 2-[iV-(2-CONH2phenylmethylNHCO)- ammojethyl, 2-[N-(3-CONH2ρhenylmethylNHCO)ammo]ethyl, 2-[N-(4- CONH2phenylmethyl-NHCO)amino]ethyl, 2-[iV-(pyridin-2-ylmethylNHCO)amino]ethyl, 2- [N-(ρyridin-3-ylmethylNHCO)amino]ethyl, 2-[iV-(pyridin-4-ylmethylNHCO)amino]ethyl, 2- [iV-(pyrimidin-2-ylmethylNHCO)ammo]etliyl, 2- [N-(pyrimidin-4- ylmethylNHCO)amino]ethyl, 2-[N-(pyrimidin-5-ylmetliylNHCO)amino]ethyl, 2-[N-(pyrazin- 2-ylmethylNHCO)amino]ethyl, 2-[iV-(pyridazin-3-ylmethylNHCO)ammo]ethyl, 2-[iV- (pyridazin-4-ylmethylNHCO)amino]ethyl, 2-[N-([ 1.3.5]triazin-2- ylmethylNHCO)amino]ethyl, 2-[iV-(2-CH3phenylmethylN(CH3)CO)-amino]ethyl3 2-[iV-(3- CH3phenylmethylN(CH3)CO)amino]ethyl, 2-[iV-(4-CH3phenylmethyl- N(CH3)CO)amino]ethyl, 2-[iV-(2-FphenylmethylN(CH3)CO)ammo]ethyl, 2-[iV-(3- FphenylmethylN(CH3)CO)amino]ethyl, 2-[N-(4-FρhenylmethylN(CH3)CO)amino]ethyl, 2- [N-(2-OHphenylmethylN(CH3)CO)amino]ethyl, 2-[iV-(3-OHphenylmethylN(CH3)CO)- aminojethyl, 2-[N-(4-OHphenylmethylN(CH3)CO)amino]ethyl, 2-[N-(2-CH30phenylmethyl- N(CH3)CO)amino]ethyl, 2-[7V-(3-CH3OphenylmethylN(CH3)CO)amino]ethyl, 2-[iV-(4- CH3OphenylmethylN(CH3)CO)amino]ethyl, 2-[N-(2-
CNphenylmethylN(CH3)CO)amino]ethyl, 2-[jV-(3-CNphenylmethylN(CH3)CO)amino]ethyl, 2-[JV-(4-CNphenylmethylN(CH3)CO)-amino]ethyl, 2-[iV-(2- ' CO;2HphenylmethylN(CH3)CO)aiΩino]etliyl, 2-[N-(3-CO2Hphenylmethyl-
N(CH3)CO)amino]ethyl, 2-[iV-(4-CO2HphenylmethylN(CH3)CO)amino]ethyl, 2-[iV-(2- CONH2phenylmethylN(CH3)CO)amino]ethyl, 2-[N-(3-CONH2phenylmethylN(CH3)CO)- amino]ethyl, 2-[N-(4-CONH2ρhenylmethylN(CH3)CO)amino]ethyl 2-[iV-(pyridin-2- ylmethylN(CH3)CO)amino]ethyl, 2-[iV-(pyridin-3-ylmethylN(CH3)CO)amino]ethyl, 2-[N- (pyridin-4-ylmethylN(CH3)CO)amino]ethyl, 2-[N-(pyrimidin-2-ylmethylN(CH3)CO)- amino]ethyl, 2-[iV-(pyrimidin-4-ylmethylN(CH3)CO)amino]ethyl5 2-[N-(pyrimidin-5- ylmethylN(CH3)CO)amino]ethyl, 2-[iV-(pyrazin-2-ylmethylN(CH3)CO)amino]ethyl, 2-[N- (pyridazin-3 -ylmethylN(CH3)CO)amino] ethyl, 2- [iV-(pyridazin-4-ylmethylN(CH3)CO)- amino]ethyl, 2-[JV-([l .3.5]triazin-2-ylmethy W(CH3)CO)amino]ethyl, 2-[N-(pyridin-4- yl)amino]ethyl, 2-[iV-(pyridin-3-yl)amino]ethyl, 2-[iV-(pyridin-2-yl)amino]ethyl, 2-[N- (pyrimidin-4-yl)amino] ethyl, 2- [JV-(pyrimidin-2-yl)amino] ethyl, 2- [7V-(pyrimidin-5 - yl)amino]ethyl, 2-[iV-([1.3.5]triazin-2-yl)amino]ethyl, 2-[iV-(phenyl)amino]ethyl, 2-[iV- (pyrazin-2-yl)amino]ethyl, 2-[iV-(pyridazin-4-yl)amino]ethyl, 2-[iV-(ρyridazm-3- yl)amino] ethyl, 2-[N-(4-F-phenyl)amino]ethyl, 2-[iV-(3-F-phenyl)amino]ethyl3 2-[N-(2-F- phenyl)amino]ethyl, 2-[iV-(2,4-diF-phenyl)amino]ethyl, 2-[Λr-(2,3-diF-phenyl)amino]ethyl, 2- [JV-(2,5-diF-phenyl)amino] ethyl, 2-[N-(2,6-diF-phenyl)amino]ethyl, 2-[N-(2,4,6-triF- ρhenyl)amino]ethyl, 2-[JV-(2,3,6-triF-phenyl)amino]ethyl, 2-[N-(2,3,4-triF- phenyl)amino]ethyl, 2-[N-(4-CH3O-phenyl)amino]ethyl, 2-[iV-(3-CH3O-phenyl)amino]ethyl, 2-[7V-(2-CH3O-phenyl)amino]ethyl, 2-[N-(4-CN-phenyl)amino]ethyl, 2-[iV-(3-CN- phenyl)amino]ethyl, 2-[iV-(2-CN-phenyl)amino]ethyl, 2-[N-(4-CO2H-ρhenyl)amino]ethyl, 2- [N-(3-CO2H-phenyl)amino]ethyl, 2-[iV-(2-CO2H-phenyl)amino]ethyl, 2-[N-(4-CONH2- phenyl)amino]ethyl, 2-[iV-(3-CONH2-phenyl)amino]ethyl 2-[iV-(2-CONH2- phenyl)amino]ethyl, 2-(iV-CH3COamino)ethyl, 2-(iV-CF3COmino)ethyl, 2-(JV- C2H5COamino)ethyl, 2-[7V-(CH3)2CHCOamino]ethyl, 2-[iV-(CH3)3CCOamino]ethyl, 2-(JV- cyclopropylCOamino)ethyl, 2-(7V-cyclobutylCOamino)ethyl, 2-[iV-(oxetan-3 - ylCO)amino]ethyl 2-[iV-(azetidin-l-ylCO)amino]ethyl, 2-[iV-(l,l-dioxo-lλ6- tetrahydrothiophen-3-ylCO)-amino]ethyl, 2-[iV-(azetidin-3-ylCO)amino]ethyl, 2-[7V-(I- acetylazetidin-4-ylCO)amino]ethyl, 2-[7V-(CF3COazetidin-4-ylCO)amino]ethyl, 2-[2V-(I- CH3SO2azetidin-4-ylCO)amino]ethyl, 2-(7V-cyclopentylCOamino)ethyl, 2-[7V-(3-
CH3Opyrrolidin-l-ylCO)amino]ethyl, 2-[iV-(3-CF3Opyrrolidm-l-ylCO)aiBino]ethyl, 2-[iV-(3- CHF2Opyrrolidin-l-ylCO)amino]ethyl, 2-[iV-(3-phenoxyρyrrolidin-l -ylCO)amino] ethyl, 2- {7V- [3 -(4-Fphenoxy)pyπ.Olidin- 1 -ylCO] amino } ethyl, 2- {N- [3 -(4-Clphenoxy)pyrrolidin- 1 - ylCO] amino } ethyl, 2- {7V-[3 -(4-Brphenoxy)pyrrolidin- 1 -ylCO]amino } ethyl, 2- {N- [3 - (COOH)pyrrolidin-l-ylCO]amino}ethyl, 2-{/V-[3-(CN) pyrrolidin-1-ylCO] amino} ethyl, 2- {iV-[3-(CONH2) pyrrolidin-l-ylCO]amino} ethyl, 2- {N- [3 -(CONHCH3) pyrrolidin-1- ylCO]amino}ethyl, 2-{7V-[3-(CON(CH3)2) pyrrolidin-1-ylCO] amino} ethyl, 2-[7V-(pyrrolidin- 1 -ylCO)amino] ethyl, 2-[iV-(tetrahydrofLiran-3-ylCO)amino]ethyl, 2-[7V-(tetrahydrothiophen- 3 -ylCO)amino] ethyl, 2-[N-(oxazolidin-3 -ylCO)amino] ethyl, 2-[N-(morpholin-4- yllCO)amino]ethyl, 2-(N-cyclopentylCOamino)ethyl, 2- [iV-(tetrahydropyran-4- ylCO)amino]ethyl5 2-[N-(piperidin-4-ylCO)amino]ethyI, 2-[iV-(l-CH3COpiperidin-4- ylCO)amino]ethyl, 2-[N-(l-CF3COpiperidin-4-ylCO)amino]ethyl, 2-[iV-(l-CH3S02piperidin- 4-ylCO)amino] ethyl, 2-[N-(tetrahydrothiopyran-4-ylCO)amino]ethyl, 2-[N-(1, 1-dioxo-lλ6- hexahydrothiopyran-4-ylCO)amino]ethyl, 2-[N-(piperidin-l-ylCO)amino] ethyl, 2-[N- (thiomorpholin-4-ylCO)amino]ethyl, 2-[N-(1 , 1 -dioxothiomorpholin-4-ylCO)amino]ethyl, 2- [N-φiperazin-4-ylCO)amino]ethyl, 2-[N-(4-CH3COpiperazin-l-ylCO)amino]ethyl, 2-[N-(4- CF3COpiperazin-l-ylCO)amino]ethyl, 2-[7V-(CH3SO2piperazin-l-ylCO)amino]ethyl, 2-[N- (phenylCO)amino] ethyl, 2-[N-(2-FphenylCO)amino]ethyl, 2-[N-(3-FρhenylCO)amino]-ethyl, 2-[N-(4-FphenylCO)amino]ethyl, 2-[N-(2-OHphenylCO)amino]ethyl, 2-[N-(3- OHphenylCO)amino]ethyl, 2-[N-(4-OHphenylCO)amino]ethyl, 2-[N-(2-CH3OphenylCO)- amino]ethyl, 2-[N-(3-CH3OphenylCO)amino]ethyl, 2-[N-(4-CH3OphenylCO)amino]ethyl, 2- [N-(2-C02HphenylCO)amino]ethyl, 2-[N-(3-CO2HphenylCO)amino]ethyl, 2-[N-(4- CO2Hphenyl-CO)amino]ethyl, 2-[N-(2-CΝphenylCO)amino]ethyl, 2-[N-(3- CΝphenylCO)amino]ethyl, 2-[N-(4-CΝphenylCO)amino]ethyl, 2-[N-(2- CH3phenylCO)amino]ethyl, 2-[N-(3-CH3phenylCO)-amino]ethyl, 2-[N-(4- CH3phenylCO)amino]ethyl, 2-[N-(2-COΝH2phenylCO)amino]ethyl, 2-[N-(3- COΝH2phenylCO)amino]ethyl, 2-[N-(4-COΝH2phenylCO)amino]ethyl, 2-[N-(pyridin-2- ylCO)amino]ethyl, 2-[N-(pyridin-3-ylCO)amino]ethyl, 2-[N-(pyridin-4-ylCO)amino]ethyl, 2- [N-(pyrimidin-4-ylCO)amino]ethyl, 2-[N-(pyrimidin-2-ylCO)amino]ethyl, 2-[N-(pyrimidin- 5-ylCO)amino]ethyl, 2-[ N-(benzylCO)amino]ethyl, 2-[N-(3-COΝH2phenylmethylCO)- amino]ethyl, 2-[N-(4-CONH2phenylmethylCO)amino]ethyl, 2-[N-(2- COΝH2phenylmethylCO)-amino]ethyl, 2-[N-(3-FphenylmethylCO)amino]ethyl, 2-[N-(4- FphenylmethylCO)amino]ethyl, 2-[N-(2-FphenylmethylCO)amino]ethyl, 2-[N-(2-
CH3OphenylmethylCO)amino]ethyl, 2-[N-(3-CH3OphenylmethylCO)amino]ethyl, 2-[N-(4- CH3OphenylmethylCO)amino]ethyl, 2-[N-(2-CΝphenylmethylCO)amino]ethyl, 2-[N-(3- CΝphenylmethylCO)amino]ethyl, 2-[N-(4-CΝphenylmethylCO)amino]ethyl, 2-[N-(2- OHphenylmethylCO)amino] ethyl, 2-[N-(3-OHphenylmethylCO)amino]ethyl, 2-[N-(4- OHphenylmethylCO)amino]ethyl, 2-[N-(2-CO2HphenylmethylCO)amino]ethyl, 2-[N-(3- CO2HphenylmethylCO)amino]ethyl, 2-[N-(4-CO2HphenylmethylCO)amino]ethyl, 2-[N- (pyridin-2-ylmethylCO)amino]ethyl, 2-[N-(pyridin-3-ylmethylCO)amino]ethyl, 2-[N- (pyridin-4-ylmethylCO)amino]ethyl, 2-[N-(pyrimidin-2-ylmethylCO)amino]ethyl, 2-[N- (pyrimidin-4-ylmethylCO)amino]ethyl, 2-[N-(pyrimidin-5-ylmethylCO)amino]ethyl, 2-[N- (pyrazin-2-ylmethylCO)amino]ethyl, 2-[ N-(phenylNHCO)-atnino]ethyl, 2-[iV-(3- CONH2phenylNHCO)amino]ethyl5 2-[iV-(4-CONH2plienylNHCO)-amino]ethyl, 2-[iV-(2- CONH2phenylNHCO)amino]ethyl5 2-[7V-(3-FphenylNHCO)amino]ethyl, 2-[iV-(4- FphenylNHCO)amino]ethyl, 2-[N-(2-FphenylNHCO)amino]ethyl, 2-[N-(2-
CH3OphenylNHCO)amino]ethyl, 2-[iV-(3-CH3OphenylNHCO)amino]ethyl, 2-[iV-(4- CH3OphenylNHCO)amino]ethyl, 2-[iV-(2-CNphenylNHCO)amino]ethyl, 2-[iV-(3- CNphenylNHCO)amino]ethyl, 2-[iV-(4-CNphenylNHlCO)amino]ethyl, 2-[iV-(2-OHphenyl- NHCO)amino]ethyl, 2-[iV-(3-OHphenylNHCO)ammo]ethyl, 2-[N-(4-OHphenylNHlCO)- amino]ethyl, 2-[N-(2-CO2HphenylNHCO)amino]ethyl, 2-[N-(3-CO2HphenylNHCO)- amino]ethyl5 2-[N-(4-CO2HphenylNHCO)amino]ethyl, 2-[N-(pyridin-2- ylNHCO)amino]ethyl, 2-[N-(pyridin-3-ylNHlCO)amino]ethyl, 2-[N-(pyridin-4- ylNHCO)amino] ethyl, 2-[N-(pyrimidin-2-ylNHCO)amino]ethyl, 2-[N-(pyrimidin-4- ylNHCO)amino]ethyl, 2-[N-(pyrimidin-5-ylNHCO)amino]ethyl, 2-[iV-([l .3.5]triazin-2- ylNHCO)amino]ethyl, 2- [N-(1, 2,3, 4-tetrahydroquinolin-l-ylCO)amino]ethyl, 2-[iV-(7- CONH2-1 ,2,3,4-tetrahydroquinolin-l -ylCO)amino] ethyl, 2-[N-(6-CONH2- 1 ,2,3,4- tetrahydroquinolin- 1 -ylCO)amino]ethyl, 2-[N-(8-CONH2- 1 ,2,3 ,4-tetrahydroquinolin- 1 - ylCO)amino]ethyl, 2-[iV-(7-F-l,2,3,4-tetrahydroquinolin-l-ylCO)amino]ethyl, 2-[N-(6-F- 1 ,2,3,4-tetrahydroquinolin- 1 -ylCO)-aiϊiino]ethyl, 2-[iV-(8-F-l ,2,3 ,4-tetrahydroquinolin- 1 - ylCO)amino] ethyl, 2-[iV-(7-CH30-l,2,3,4-tetrahydroquinolin-l-ylCO)amino]ethyl, 2-[N-(6- CH3O-l,2,3,4-tetrahydroquinolin-l-ylCO)amino]ethyl, 2-[7V-(8-CH3O-l,2,3,4- tetrahydroquinolin- 1 -ylCO)amino]ethyl, 2-[iV-(7-CN- 1 ,2,3 ,4-tetrahydroquinolin- 1 - ylCO)amino]ethyl, 2-[iV-(6-CN-l,2,3,4-tetrahydroquinolm-l-ylCO)amino]ethyl, 2-[N-(8-CN- 1 ,2,3 ,4-tetrahydroquinolin- 1 -ylCO)amino] ethyl, 2-[JV-(7-OH- 1 ,2,3 ,4-tetrahydroquinolin- 1 - ylCO)amino]etriyl, 2-[N-(6-OH-l,2,3,4-tetrahydroquinolin-l-ylCO)amino]ethyl, 2-[N-(8-OH- l,2,3,4-tetrahydroquinolin-l-ylCO)amino]ethyl, 2-[N-(7-CO2H-l,2,3,4-tetrahydroquinolin-l- ylCO)amino]ethyl, 2-[N-(6-C02H-l,2,3,4-tetrahydroqmnolin-l-ylCO)amino]ethyl, 2-[iV-(8- CO2H- 1 ,2,3,4-tetrahydroquinolin- l-ylCO)amino]ethyl, 2-[N-(1 ,2,3 ,4-tetrahydro- [l,8]naphthyridin-l-ylCO)amino]ethyI, 2-[N-(l,2,3,4-tetrahydro-[l,7]naphthyridin-l- ylCO)amino]ethyl, 2-[iV-(l,2,3,4-tetrahydro-[l,6]naphthyridin-l-ylCO)amino]ethyl, 2-[N- (5,6,7,8-tetrahydropyrido[2,3-b]pyrazin-5-ylCO)amino]ethyl, 2-[N-(5,6,7,8- tetrahydropyrido[2,3-d]pyrimidin-8-ylCO)amino]ethyl, 2-[N-(5,6,7,8-tetrahydropyrido[3,2- d]pyrimidin-5-ylCO)amino]ethyl, 2-[N-(5,6,7,8-tetrahydropyrido[2,3-d]pyridazin-l- ylCO)aniino]ethyl, 2-[ N-(2,3-dihydro-lH-indol-l-ylCO) aminojethyl, 2-[N-(7-COΝH2-2,3- dihydro-l//-indol-l-ylCO)amino]ethyl, 2-[N-(6-CONH2-2,3-dihydro-li7-indol-l- ylCO)amino]ethyl, 2-[Λr-(5-CONH2-2,3-dihydro-lH-indol-l-ylCO)amino]ethyl, 2-[iV-(7-F- 2,3-dihydro-lH"-indol-l-ylCO)amino]ethyl, 2-[iV-(6-F-2,3-dihydro-lH"-indol-l- ylCO)amino]ethyl, 2-[N-(5-F-2,3-dihydro-lJff-indol-l-ylCO)amino]etliyl, 2-[iV-(7-CH30-2,3- dihydiO-lH-indol-l-ylCO)amino]etliyl, 2-[7V-(6-CH3O-2,3-dihydro-lH-indol-l- ylCO)amino]ethyl, 2-[7V-(5-CH3O-2,3-dihydro-lH-indol-l-ylCO)amino]ethyl, 2-[iV-(7-CN- 2,3-dihydro-lH-indol-l-ylCO)amino]ethyl, 2-[N-(6-CN-2;3-dihydro-lH-indol-l- ylCO)amino]ethyl, 2-[N-(5-CN-2,3-dihydro-li/-indol-l -ylCO)amino] ethyl, 2-[iV-(7-OH-2,3- dihydro-lH-indol-l-ylCO)amino]ethyl, 2-[N-(6-OH-2,3-dihydro-lH-indol-l- ylCO)amino]ethyl, 2-[iV-(5-OH-2,3-dihydro-lH-indol-l-ylCO)amino]ethyl, 2-[iV-(7-CO2H- 2,3-dihydro-lH-indol-l-ylCO)ammo]ethyl, 2-[iV-(6-CO2H-2,3-diliydro-lH-mdol-l- ylCO)amino]ethyl, 2-[N-(5-CO2H-2,3-dihydro-lH-mdol-l-ylCO)amino]ethyl, 2-[N-(2,3- dihydro-lΗ-pyrrolo[2,3-b]pyridm-l-ylCO)-amino]ethyl, 2-[iV-(2,3-dihydro-lΗ-pyrrolo[2,3- c]pyridin- 1 -ylCO)-amino] ethyl, 2-[JV-(2,3 -dihydro- 1 H-pyrrolo [3 ,2-c]pyridin- 1 -ylCO)- amino]ethyl, 2-[N-(6,7-dihydro-5H-pyrrolo[2,3-b]pyrazin-5-ylCO)-amino]ethyl, 2-[N-(6,7- dihydro-5H-pyrrolo[2,3-d]pyrimidin-7-ylCO)-amino]ethyl 2-[iV-(6,7-dihydro-5H- pyrrolo[3 ,2-d]pyrimidin-5 -ylCO)-amino] ethyl, or 2-[iV-(2,3 -dihydro- 1 H-pyrrolo [2,3 - d]pyridazin-l-ylCO) amino]ethyl and R10 is methyl or ethyl.
[0121] (h) Within the above preferred groups (A)5 A(l-4), A(l-viii) and A(i-4)(i-viii), and more preferred groups contained therein, R5 is -alkylene-SC^NR11R12 where here R11 and R12 together with the nitrogen atom to which they are attached form piperidin-1-yl, A- methylpiperidin-1-yl, 4-ethylpiperidin-l-yl, 4-(2,2,2-trifluoroethyl)piperidin-l-yl, 4-(2- isopropyl)piperidin- 1 -yl, 4-(2-trifluoromethyl-2,2,2-trifluoroethyl)ρiperidin- 1 -yl A- (cyclopropyl)piperidin-l-yl, 4-(cyclobutyl)piperidin-l-yl, 4-(oxetan-3-yl)piperidin-l-yl A- (azetidin-3 -yl)piperidin- 1 -yl, 4-( 1 , 1 -dioxo- 1 λ6-thietan-3 -yl)piperidin- 1 -yl, 4-(cyclopentyl)- piperidin- 1 -yl, 4-(3-CH3Ocyclopentyl)piperidin- 1 -yl, 4-(3-CHF2Ocyclopentyl)piperidin- 1 -yl, 4-(3 -CF3Ocyclopentyl)piperidin- 1 -yl, 4-(3 -phenoxycyclopentyl)piperidin- 1 -yl, 4-[3 -(4- Fphenoxy)cyclopentyl]piperidin- 1 -yl, 4-[3 -(4-Clphenoxy)cyclopentyl]piperidin- 1 -yl, 4- [3 -(4- Brphenoxy)cyclopentyl]piperidin-l-yl, 4-[3-(4-CO2Hphenoxy)cyclopentyl]piperidin-l-yl, 4- [3-(4-CNphenoxy)cyclopentyl]piperidin-l -yl, 4-[3-(4-CONH2phenoxy)cyclopentyl]piperidin- 1-yl, 4-(cyclohexyl)piperidin-l-yl, 4-(tetrahydropyran-4-yl)piperidin-l-yl, 4-(piperidin-4- yl)piperidin- 1 -yl, 4-(tetrahydropyran-3 -yl)piperidin- 1 -yl, 4-(tetrahydrothiopyran-3 - yl)piperidin- 1 -yl, 4-(l , 1 -dioxo- 1 λ6-hexahydrothiopyran-3-yl)piperidin- 1 -yl, 4-(piperidin-3 - yl)piperidin-l-yl, 4-(methylsulfonyl)piperidin-l-yl, 4-(ethylsulfonyl)piperidin-l-yl, 4- (isopropylsulfonyl)piperidin- 1 -yl, 4-(ferr-butylsulfonyl)piperidin- 1 -yl, A- (cyclopropylsulfonyl)piperidin- 1 -yl, 4-(cyclobutyl-sulfonyl)piperidin- 1 -yl, A- (cyclopentylsulfonyl)piperidin- 1 -yl, 4-(cyclohexylsulfonyl)piperidin- 1 -yl, A- (benzenesulfonyl)piperidin- 1 -yl, 4-(2-CH3phenylsulfonyl)piperidin- 1 -yl, 4-(3 -
CH3phenylsulfonyl)piperidin- 1 -yl, 4-(4-CH3phenylsulfonyl)piperidin- 1 -yl, 4-(2-Fphenyl- sulfonyl)piperidin- 1 -yl, 4-(3 -Fphenylsulfonyl)piperidin- 1 -yl, 4-(4-Fphenylsulfonyl)piρeridin- 1-yl, 4-(2-OHphenylsulfonyl)piperidin-l-yl, 4-(3-OHphenylsulfonyl)piperidin-l-yl, 4-(4- OHphenylsulfonyl)piperidin-l-yl, 4-(2-CH3θphenylsulfonyl)piperidin-l-yl, 4-(3- CH3Ophenylsulfonyl)piperidin- 1 -yl, 4-(4-CH3Ophenylsulfonyl)piperidin- 1 -yl, 4-(2- CO2Hphenylsulfonyl)piperidin- 1 -yl, 4-(3 -CO2Hphenylsulfonyl)piperidin- 1 -yl, 4-(4- CO2Hphenylsulfonyl)piperidin- 1 -yl, 4-(2-CONH2phenylsulfonyl)piperidin- 1 -yl, 4-(3 - CONH2phenylsulfonyl)piperidin- 1 -yl, 4-(4-CONH2phenylsulfonyl)piperidin- 1 -yl, 4-(2- CON(CH3)2phenylsulfonyl)piperidin-l-yl, 4-(3-CON(CH3)2phenylsulfonyl)piperidin-l-yl, 4- (4-CON(CH3)2phenylsulfonyl)piperidin- 1 -yl, 4-(methylphenylNCO)piperidin- 1 -yl, 4-[(3- CONH2phenyl)methylNCO]piperidin- 1 -yl, 4- [(4-CONH2phenyl)methylNCO]piperidin- 1 -yl, 4-[(2-CONH2phenyl)methylNCO]piρeridin-l-yl, 4-[(3-Fphenyl)methylNCO]piperidin-l-yl, 4-[(4-Fphenyl)methylNCO]ρiperidin-l-yl, 4-[(2-Fphenyl)methylNCO]piperidin-l-yl, 4-[(3- OCH3phenyl)methylNCO]piρeridin-l-yl, 4-[(4-OCH3phenyl)methylNCO]piperidin-l-yl, 4- [(2-OCH3phenyl)methylNCO]piperidin-l-yl, 4-[(3-CNρhenyl)methylNCO]piperidin-l-yl, 4- [(4-CNphenyl)methylNCO]piρeridin-l -yl, 4-[(2-CNphenyl)methylNCO]piperidin-l -yl, 4-[(3- OHphenyl)methylNCO]piperidin-l-yl, 4-[(4-OHρhenyl)methylNCO]piperidin-l-yl, 4-[(2- OHphenyl)methylNCO]piperidin- 1 -yl, 4- [(pyridin-2-yl)methylNCO]piperidin- 1 -yl, A- [(pyridin-3 -yl)methylNCO]piperidin- 1 -yl, 4-[(pyridin-4-yl)methylNCO]piperidin- 1 -yl, A- [(pyrimidin-2-yl)methylNCO]piperidin- 1 -yl, 4-[(pyrimidin-4-yl)methylNCO]piperidin- 1 -yl, 4-[(pyrimidin-5-yl)methylNCO]piperidm-l-yl, 4-[(3-CO2Hphenyl)methylNCO]piperidin-l- yl, 4-[(4-CO2Hphenyl)methylNCO]piperidin- 1 -yl, 4-[(2-CO2Hphenyl)methylNCO]piperidin- 1-yl, 4-[(3-CH3phenylmethyl)NHCO]piperidin-l-yl, 4-[(4- CH3phenylmethyl)NHCO]piperidin- 1 -yl, 4-[(2-CH3ρhenylmethyl)NHCO]ρiperidin- 1 -yl, 4- [(3-CONH2phenylmethyl)NHCO]piperidin-l-yl, 4-[(4-
CONH2phenylmethyl)NHCO]piperidin- 1 -yl, 4-[(2-CONH2phenylmethyl)NHCO]piperidin- 1 - yl, 4-[(3-Fphenylmethyl)NHCO]ρiperidin-l-yl, 4-[(4-Fρhenylmethyl)NHCO]piperidin-l-yl, 4-[(2-Fphenylmethyl)NHCO]piperidin-l-yl, 4-[(3-OCH3phenylniethyl)NHCO]piperidin-l-yl, 4-[(4-OCH3phenylmethyl)NHCO]piperidin-l-yl, 4-[(2-OCH3phenylmethyl)NHCO]pipeπdin- 1-yl, 4-[(3-CNphenylmethyl)NHCO]piperidin-l-yl, 4-[(4-CNphenylmethyl)NHCO]piperidin- 1 -yl, 4-[(2-CNphenylmethyl)NHCO]piperidin- 1 -yl, 4-[(3-OHphenylmethyl)NHCO]piperidin- 1-yl, 4-[(4-OHphenylmethyl)NHCO]piperidin-l-yl, 4-[(2-
OHphenylmethyl)NHCO]piperidin- 1 -yl, 4-[(pyridin-2-ylmethyl)NHCO]piperidin- 1 -yl, 4- [(pyridin-3-ylmethyl)NHCO]ρiperidin-l-yl, 4-[(pyridin-4-ylmethyl)NHCO]piperidin-l-yl, 4- [(pyrimidin-2-ylmethyl)NHCO]piperidin- 1 -yl, 4- [(pyrimidin-4-ylmethyl)NHCO]piperidin- 1 - yl, 4-[(pyrimidm-5-ylmethyl)NHCO]piperidin-l-yl, 4-[(3-
CO2Hphenylmethyl)NHCO]piperidin-l-yl, 4-[(4-CO2Hphenylmethyl)NHNCO]-piperidin-l- yl, 4-[(2-CO2Hphenylmethyl)NHCO]piperidm-l-yl, 4-[(pyrazin-2-ylmethyl)- NHCO]piperidin- 1 -yl, 4- [(pyridazin-3 -ylmethyl)NHCO]piperidin- 1 -yl, 4- [(pyridazin-4- ylmethyl)NHCO]ρiperidin-l-yl, 4-[([l .3.5]triazin-2-ylmethyl)NHCO]piperidin-l-yl, 4-[JV-(3- CH3phenylmethyl)N(CH3)CO]piperidin-l-yl, 4-[ JV-(4- CH3phenylmethyl)N(CH3)CO]piperidin-l-yl, 4-[ JV-(2- CH3phenylmethyl)N(CH3)CO]piperidin-l-yl, 4-[ 7V-(3-CONH2phenyl- methyl)N(CH3)CO]piperidin-l-yl, 4-[ Λr-(4-CONH2phenylmethyl)N(CH3)CO]piperidin-l-yl, 4-[ iV-(2-CONH2phenylmethyl)N(CH3)CO]piperidin-l-yl3 4-[ iV-(3-
Fphenylmethyl)N(CH3)CO]-ρiperidin-l-yl, 4-[ N-(4-Fρhenylmethyl)isf(CH3)CO]piperidin-l- yl, 4-[ iV-(2-Fphenylmethyl)-N(CH3)CO]piperidin-l-yl, 4-[ JV-(3- OCH3phenylmethyl)N(CH3)CO]piperidin-l-yl, 4-[ JV-(4- OCH3phenylmethyl)N(CH3)CO]piperidin-l-yl, 4-[ iV-(2-OCH3phenylmethyl)N(CH3)CO]- piperidin-1-yl, 4-[ N-(3-CNphenylmethyl)N(CH3)CO]piperidin-l-yl, 4-[ JV-(4-CNphenyl- methyl)N(CH3)CO]piperidin-l-yl, 4-[ iV-(2-CNphenylmethyl)N(CH3)CO]piperidin-l-yl, 4-[ iV-(3-OHphenylmethyl)N(CH3)CO]piperidin-l-yl, 4-[ 7V-(4-OHphenylmethyl)N(CH3)CO]- piperidin-1-yl, 4-[ N-(2-OHphenylmethyl)N(CH3)CO]piρeridin-l-yl, 4-[ 7V-(pyridin-2- ylmethyl)N(CH3)CO]piperidin-l-yl, 4-[ N-(pyridin-3-ylmethyl)N(CH3)CO]piperidin-l-yl, 4-[ N-(pyridin-4-ylmethyl)N(CH3)CO]piperidin-l-yl, 4-[ iV-(pyriinidm-2-ylmethyl)N(CH3)CO]- piperidin-1-yl, 4-[ iV-(pyrimidin-4-ylmethyl)N(CH3)CO]piperidin-l-yl, 4-[ iV-(pyrimidin-5- ylmethyl)N(CH3)CO]piperidin-l-yl, 4-[ iV-(3-Cθ2Hphenylmethyl)N(CH3)CO]piperidin-l-yl, 4-[ iV-(4-CO2Hphenylmethyl)N(CH3)NCO]piperidin-l-yl, 4-[ iV-(2-CO2Hphenylmethyl)- N(CH3)CO]piperidin-l-yl, 4-[ iV-(pyrazin-2-ylmethyl)N(CH3)CO]piperidin-l-yl, 4-[ JV-
(pyridazin-3-ylmethyl)N(CH3)CO]piperidin-l -yl, 4-[ iV-(pyridazin-4-ylmethyl)N(CH3)CO]- piperidin-1-yl, 4-[ iV-([1.3.5]triazin-2-ylmethyl)N(CH3)CO]ρiρeridin-l-yl, 4-(pyridin-4- yl)piperidin-l-yl, 4-(ρyridin-3-yl)piperidin-l-yl, 4-(pyridin-2-yl)piperidin-l-yl, 4-(pyrimidin- 4-yl)piperidin-l-yl, 4-(pyrimidin-2-yl)piperidin-l-yl, 4-(pyrimidin-5-yl)piρeridin-l-yl, 4- ([1.3.5]triazin-2-yl)piperidin-l-yl, 4-(phenyl)piρeridin-l-yl, 4-(pyrazin-2-yl)piperidin-l-yl, A- (pyridazin-3-yl)piperidin- 1 -yl, 4-(pyridazin-4-yl)ρiperidin- 1 -yl, 4-(4-Fphenyl)piperidin- 1 -yl, 4-(3 -Fphenyl)piperidin- 1 -yl, 4-(2-Fphenyl)piperidin- 1 -yl, 4-(2,4-diFphenyl)piperidin- 1 -yl, A- (2,3 -diFphenyl)piperidin- 1 -yl, 4-(2,5 -diFphenyl)piperidin- 1 -yl, 4-(2,6-diFphenyl)piperidin- 1 - yl, 4-(2,4,6-triFphenyl)piperidin-l-yl, 4-(2,3,6-triFphenyl)piperidin-l-yl, 4-(2,3,4- triFphenyl)-piperidin- 1 -yl, 4-(4-CH3Ophenyl)piperidin-l -yl, 4-(3-CH3Ophenyl)piperidin- 1 - yl, 4-(2-CH3Ophenyl)piperidin-l-yl, 4-(4-CNphenyl)piperidin-l-yl, 4-(3- CNphenyl)piperidin-l-yl, 4-(2-CNphenyl)piperidin-l-yl, 4-(4-CO2Hphenyl)piperidin-l-yl, 4- (3 -CO2Hρhenyl)ρiperidin- 1 -yl, 4-(2-CO2Hphenyl)piperidin- 1 -yl, 4-(4- CONH2phenyl)piperidin-l-yl, 4-(3-CONH2phenyl)-ρiperidin-l-yl, 4-(2- CONH2phenyl)piperidin- 1 -yl, 4-(methylcarbonyl)piperidin- 1 -yl, A- (trifluoromethylcarbonyl)piperidin- 1 -yl, 4-(ethylcarbonyl)piperidin- 1 -yl, A- (isopropylcarbonyl)-piperidin- 1 -yl, 4-(/ert-butylcarbonyl)piperidin- 1 -yl, A- (cyclopropylCO)piperidin- 1 -yl, 4-(cyclobutylCO)piperidin- 1 -yl, 4-(cyclopentylCO)piperidin- 1 -yl, 4-(azetidin-3-ylCO)piperidin- 1 -yl, 4-(3-CH3Opyrrolidin- 1 -ylCO)piperidin- 1 -yl, 4-(3- CF3Opyrrolidin- 1 -ylCO)piperidin- 1 -yl, 4-(3-CHF2Opyrrolidin- 1 -ylCO)piperidin- 1 -yl, 4-(3 - CO2Hpyrrolidin-l-ylCO)piperidin-l-yl, 4-(3-CNpyrrolidin-l-ylCO)piperidin-l-yl, 4-(3- CONH2pyrrolidin- 1 -ylCO)piperidin- 1 -yl, 4-(pyrrolidin- 1 -ylCO)piperidin- 1 -yl, 4(oxazolidin- 3 -ylCO)piperidin- 1 -yl, 4-(tetrahydrofuran-3-ylCO)piperidin- 1 -yl, 4-(tetrahydrothiophen-3 - ylCO)piperidin- 1 -yl, 4-( 1 , 1 -dioxo- 1 λ6-tetrahydrothiophen-3 -ylCO)piperidin- 1 -yl, A-
(cyclohexylCO)piperidin- 1 -yl, 4-(tetrahydropyran-4-ylCO)piperidin- 1 -yl, 4-(piperidin-4- ylCO)piperidin- 1 -yl, 4-(tetrahydrothiopyran-4-ylCO)piperidin- 1 -yl, 4-(l , 1 -dioxo- 1 λ6- hexahydrothiopyran-4-ylCO)piperidin- 1 -yl, 4-(piperidin- 1 -ylCO)piperidin- 1 -yl, 4- (morpholin-4-ylCO)piperidin- 1 -yl, 4-(thiomorpholin-4-ylCO)piperidin- 1 -yl, 4-(l , 1 -dioxo- 1 λ6-thiomorpholin-4-ylCO)piperidin- 1 -yl, 4-(piperazin- 1 -ylCO)piperidin- 1 -yl, A-
(phenylCO)ρiperidin- 1 -yl, 4-(2-CH3phenylCO)ρiperidin- 1 -yl, 4-(3 -CH3phenylCO)piperidin- 1-yl, 4-(4-CH3phenylCO)piperidin-l-yl, 4-(2-FphenylCO)ρiperidin-l-yl, 4-(3- FphenylCO)piperidin- 1 -yl, 4-(4-FphenylCO)piperidin- 1 -yl, 4-(2-OHpheny lCO)-piperidin- 1 - yl, 4-(3-OHphenylCO)piρeridin-l-yl, 4-(4-OHphenylCO)piρeridin-l-yl, 4-(2- CH3OphenylCO)piperidin-l-yl, 4-(3-CH3OphenylCO)piperidin-l-yl, 4-(4-CH3OphenylCO)- piperidin-1-yl, 4-(2-CO2HphenylCO)ρiperidin-l-yl, 4-(3-CO2HphenylCO)piperidin-l-yl, A- (4-CO2HphenylCO)piperidin-l-yl, 4-(2-CONH2phenylCO)piperidin-l-yl, 4-(3- CONH2phenylCO)-piperidin- 1 -yl, 4-(4-CONH2phenylCO)piperidin- 1 -yl, 4-(2- CNphenylCO)piperidin-l-yl, 4-(3-CNphenylCO)piρeridin-l-yl, 4-(4-CNphenylCO)piperidin- 1-yl, 4-(pyridin-4-ylCO)piperidin-l-yl, 4-(pyridin-3-ylCO)piperidin-l-yl, 4-(pyridin-2- ylCO)piperidin- 1 -yl, 4-(pyrimidin-4-ylCO)piperidin- 1 -yl, 4-(pyrimidin-2-ylCO)piperidin- 1 - yl, 4-(pyrimidin-5-ylCO)piperidin-l-yl 4-(benzylCO)piperidin-l-yl, 4-(2- FphenylmethylCO)piperidin- 1 -yl, 4-(3 -FphenylmethylCO)-piperidin- 1 -yl, 4-(4- FphenylmethylCO)piperidin- 1 -yl, 4-(2-OHphenylmethylCO)piperidin- 1 -yl, 4-(3 - OHphenylmethylCO)piperidin- 1 -yl, 4-(4-OHphenylmethylCO)piperidin- 1 -yl, 4-(2- CH3OphenylmethylCO)piperidin-l -yl, 4-(3-CH3OphenylmethylCO)ρiperidm-l -yl, 4-(4- CH3OphenylmethylCO)piperidin- 1 -yl, 4-(2-CO2HphenylmethylCO)piρeridin- 1 -yl, 4-(3 - CO2HphenylmethylCO)piperidin- 1 -yl, 4-(4-CO2HphenylmethylCO)piperidin- 1 -yl, 4-(2- CONH2phenylmethylCO)piρeridin- 1 -yl, 4-(3 -CONH2phenylmethylCO)piperidin- 1 -yl, 4-(4- CONH2phenylmethylCO)piperidin- 1 -yl, 4-(2-CNphenylmethylCO)piperidin- 1 -yl, 4-(3 - CNphenylmethylCO)piperidin- 1 -yl, 4-(4-CNphenylmethylCO)piperidin- 1 -yl, 4-(pyridin-4- ylmethylCO)piperidin- 1 -yl, 4-(pyridin-3 -ylmethylCO)piperidin- 1 -yl, 4-(pyridin-2- ylmethylCO)piperidin- 1 -yl, 4-(pyrimidin-4-ylmethylCO)piperidin- 1 -yl, 4-(pyrimidin-2- ylmethylCO)piperidin- 1 -yl, 4-(pyrimidin-5-ylmethylCO)piperidin- 1 -yl, 4-(pyriazin-2- ylmethylCO)piρeridin-l -yl, 4-(phenylNHCO)piperidin-l -yl, 4-(2-FphenylNHCO)piperidin- 1-yl, 4-(3-FphenylNHCO)piperidin-l-yl, 4-(4-FphenylNHCO)piperidin-l-yl, 4-(2- OHphenylNHCO)-piperidin-l-yl, 4-(3-OHphenylNHCO)piperidin-l-yl, 4-(4- OHphenylNHCO)piperidin-l-yl, 4-(2-CH3OphenylNHCO)piperidin-l-yl, 4-(3- CH3OphenylNHCO)piperidin-l-yl, 4-(4-CH3OphenylNHCO)piperidin-l-yl, 4-(2- CO2HphenylNHCO)piperidin- 1 -yl, 4-(3 -CO2Hphenyl-NHCO)piperidin- 1 -yl, 4-(4- CO2HphenylNHCO)piperidin- 1 -yl, 4-(2-CONH2phenylNHCO)-piperidin- 1 -yl, 4-(3 - CONH2phenylNHCO)piperidin-l-yl, 4-(4-CONH2phenylNHCO)piperidin-l-yl, 4-(2- CNphenylNHCCOpiperidin-l-yl, 4-(3-CNphenylNHCO)piperidin-l-yl, 4-(4-
CNphenylNHCO)piperidin- 1 -yl, 4-(pyridin-4-ylNHCO)piρeridin- 1 -yl, 4-(pyridin-3 - ylNHCO)-piperidin- 1 -yl, 4-(pyridin-2-ylNHCO)piperidin- 1 -yl, 4-(pyrimidin-4- ylNHCO)piperidin- 1 -yl, 4-(pyrimidin-2-ylNHCO)piperldin- 1 -yl, 4-(pyrimidin-5 - ylNHCO)piperidin-l-yl, 4-([1.3.5]triazin-2-ylNHCO)piperidin-l-yl, 4-(l,2,3,4- tetrahydroquinolin- 1 -ylCO)piperidin- 1 -yl, 4-(7-CONH2- 1 ,2,3 ,4-tetrahydroquinolin- 1 - ylCO)piperidin-l-yl, 4-(6-CONH2-l,2,3,4-tetrahydroquinolin-l-ylCO) ρiperidin-1-yl, 4-(8- CONH2- 1 ,2,3 ,4-tetrahydroquinolin- 1 -ylCO) piperidin- 1 -yl, 4-(7-F- 1 ,2,3 ,4- tetrahydroquinolin- 1 -ylCO)piperidin- 1 -yl, 4-(6-F- 1 ,2,3 ,4-tetrahydroquinolin- 1 - ylCO)piperidin-l -yl, 4-(8-F-1 ,2,3,4-tetrahydroquinolin-l -ylCO)piperidin-l -yl, 4-(7-CH3O- 1 ,2,3,4-tetrahydroquinolin-l -ylCO)piperidin-l -yl, 4-(6-CH3O-1 ,2,3,4-tetrahydroquinolin-l - ylCO) piperidin-1-yl, 4-(8-CH3O-1, 2,3, 4-tetrahydroquinolin-l-ylCO) ρiperidin-1-yl, 4-(7- CN- 1 ,2,3 ,4-tetrahydroquinolin- 1 -ylCO)piperidin- 1 -yl, 4-(6-CN- 1 ,2,3 ,4-tetrahydroquinolin- 1 - ylCO)piperidin- 1 -yl, 4-(8-CN- 1 ,2,3 ,4-tetrahydroquinolin- 1 -ylCO)-piperidin- 1 -yl, 4-(7-OH- 1 ,2,3 ,4-tetrahydroquinolin- 1 -ylCO)piperidin- 1 -yl, 4-(6-OH- 1 ,2,3 ,4-tetrahydroquinolin- 1 - ylCO)piperidin- 1 -yl, 4-(8-OH- 1 ,2,3 ,4-tetrahydroquinolin- 1 -ylCO)piperidin- 1 -yl, 4-(7-CO2H- 1 ,2,3,4-tetrahydroquinolin- 1 -ylCO)piperidin- 1 -yl, 4-(6-CO2H- 1 ,2,3 ,4-tetrahydroquinolin- 1 - ylCO)piperidin-l-yl, 4-(8-CO2H-l,2,3,4-tetrahydroquinolin-l-ylCO)piρeridin-l-yl, 4- ( 1 ,2,3 ,4-tetrahydro- [ 1 , 8]naphthyridin- 1 -ylCO)piρeridin- 1 -yl, 4-( 1 ,2,3 ,4-tetrahydro- [1 ,7]naphthyridin- 1 -ylCO)piperidin-l -yl, 4-(l ,2,3 ,4-tetrahydro-[l ,6]naphthyridin-l - ylCO)piperidin-l-yl, 4-(5,657,8-tetrahydropyrido[2,3-b]pyrazin-5-ylCO)piperidin-l-yl, 4- (5,6,7,8-tetrahydropyrido[2,3-d]pyrimidin-8-ylCO)piperidin-l-yl, 4-(5,6,7,8- tetrahydropyrido[3,2-d]pyrimidin-5-ylCO)piperidin-l-yl, 4-(5,6,7,8-tetrahydropyrido[2,3- d]pyridazin- 1 -ylCO)ρiperidin- 1 -yl, 4-(2,3 -dihydro- lH-indol- 1 -ylCO)piperidin- 1 -yl, 4-(7- CONH2-2,3 -dihydro- 1 H-indol- 1 -ylCO)piperidin- 1 -yl, 4-(6-CONH2-2,3-dihydro- lH-indol- 1 - ylCO)piperidin- 1 -yl, 4-(5-CONH2-2,3 -dihydro- 1 H-indol- 1 -ylCO)piperidin- 1 -yl, 4-(7-F-2,3 - dihydro- 1 H-indol- 1 -ylCO)piperidin- 1 -yl, 4-(6-F-2,3 -dihydro- 1 H-indol- 1 -ylCO)piperidin- 1 -yl 4-(5-F-2,3-dihydro-lH-indol-l-ylCO)piperidin-l-yl, 4iV-(7-CΗ30-2,3-dihydro-lH-mdol-l- ylCO)piperidin-l-yl, 4-(6-CΗ3O-2,3-dihydro-lH-indol-l-ylCO)piperidin-l-yl, 4-(5-CH3O- 2,3 -dihydro- 1 H-indol- 1 -ylCO)piperidin- 1 -yl, 4-(7-CN-2,3 -dihydro- 1 H-indol- 1 - ylCO)pipeπdin-l-yl, 4-(6-CN-2,3-dihydro-lH-indol-l-ylCO)piperidin-l-yl, 4-(5-CN-2,3- dihydro- 1 H-indol- 1 -ylCO)piperidin- 1 -yl, 4-(7-OΗ-2,3 -dihydro- lH-indol- 1 -ylCO)piperidin- l.yl5 4.(6-OΗ-2,3-dihydiO-lH-indol-l-ylCO)piperidin-l-yl, 4-(5-OΗ-2,3-dihydro-lH-indol- l-ylCO)piperidin-l-yl, 4-(7-CO2Η-2,3-dihydro-lH-indol-l-ylCO)piperidin-l-yl, 4-(6-CO2H- 2,3-dihydro-lH-indol-l-ylCO)piperidin-l-yl, 4-(5-CO2H-2,3-dihydro-lH-indol-l- ylCO)piperidin- 1 -yl, 4-(2,3 -dihydro- 1 Η-pyrrolo [ 2,3 -b]pyridin- 1 -ylCO)piperidin- 1 -yl, 4-(2,3 - dihydro-lΗ-pyrrolo[2,3-c]pyridin-l -ylCO)piperidin-l -yl, 4-(2,3-dihydro-lH-pyrrolo[3,2- c]pyridin- 1 -ylCO)piperidin- 1 -yl, 4-(6,7-dihydro-5H-pyrrolo [2,3-b]pyrazin-5-ylCO)piperidin- 1-yl, 4-(6,7-dihydro-5H-ρyrrolo[2,3-d]pyrimidin-7-ylCO)ρiperidin-l-yl, 4-(6,7-dihydro-5H- pyrrolo[3,2-djpyrimidin-5-ylCO)piperidin-l-yl, or 4-(2,3-dihydro-lH-pyrrolo[2,3- d]pyridazin- 1 -ylCO)piperidin- 1 -yl.
[0122] (i) Within the above preferred groups (A), A(l-4), A(l-viii) and A(i-4)(i-viii), and more preferred groups contained therein, R5 is -alkylene-SO2NR R where here R11 and R12 together with the nitrogen atom to which they are attached form piperazin-1-yl, A- methylpiperazin-1-yl, 4-ethylpiperazin-l-yl, 4-(2,2,2-trifluoroethyl)piperazin-l-yl, 4-(2- isopropyl)piperazin- 1 -yl, 4-(2-trifluoromethyl-2,2,2-triiluoroethyl)piperazin- 1 -yl A- (cyclopropyl)piperazin-l-yl, 4-(cyclobutyl)piperazin-l-yl, 4-(oxetan-3-yl)piperazin-l-yl A- (azetidin-3-yl)piperazin-l-yl, 4-(l,l-dioxo-lλ6-thietan-3-yl)piperazin-l-yl, 4-(cyclopentyl)- piperazin- 1 -yl, 4-(3 -CH3Ocyclopentyl)piperazin- 1 -yl, 4-(3 -CHF2Ocyclopentyl)piperazin- 1 - yl, 4-(3-CF3Ocyclopentyl)piperazin-l-yl, 4-(3-phenoxycyclopentyl)ρiρerazin-l-yl, 4-[3-(4- Fphenoxy)cyclopentyl]piperazin-l-yl, 4-[3-(4-Clphenoxy)cyclopentyl]piperazin-l-yl, 4-[3- (4-Brphenoxy)cyclopentyl]piperazin-l-yl, 4-[3-(4-CO2Hphenoxy)cyclopentyl]piperazin-l-yl, 4-[3-(4-CNphenoxy)cyclopentyl]piperazin-l-yl, 4-[3-(4-
CONH2phenoxy)cyclopentyl]piperazin- 1 -yl, 4-(cyclohexyl)piperazin- 1 -yl, A- (tetrahydropyran-4-yl)piperazin- 1 -yl, 4-(piperidin-4-yl)piperazin- 1 -yl, 4-(tetrahydropyran-3 - yl)piperazin- 1 -yl, 4-(tetrahydrothiopyran-3 -yl)piperazin- 1 -yl, 4-( 1 , 1 -dioxo- 1 λ6- hexahydrothiopyran-3 -yl)piperazin- 1 -yl, 4-(piperidin-3 -yl)piperazin- 1 -yl, A- (methylsulfonyl)piperazin-l-yl, 4-(ethylsulfonyl)piperazin-l-yl, 4-(isopropylsulfonyl)- piperazin- 1 -yl, 4-(ter£-butylsulfonyl)piperazin- 1 -yl, 4-(cyclopropylsulfonyl)piperazin- 1 -yl, 4- (cyclobutyl-sulfonyl)piperazin- 1 -yl, 4-(cyclopentylsulfonyl)piperazin- 1 -yl, 4-(cyclohexyl- sulfonyl)piperazin- 1 -yl, 4-(benzenesulfonyl)piperazin- 1 -yl, 4-(2- CH3phenylsulfonyl)piperazin- 1 -yl, 4-(3-CH3phenylsulfonyl)piperazin- 1 -yl, 4-(4- CH3phenylsulfonyl)piperazin-l-yl, 4-(2-Fphenyl-sulfonyl)piperazin-l-yl, 4-(3- Fphenylsulfonyl)piperazin- 1 -yl, 4-(4-Fphenyl-sulfonyl)piperazin- 1 -yl, 4-(2- OHphenylsulfonyl)piperazin- 1 -yl, 4-(3 -OHphenyIsulfonyl)-piperazin- 1 -yl, 4-(4- OHphenylsulfonyl)piperazin- 1 -yl, 4-(2-CH3Ophenylsulfonyl)piperazin- 1 -yl, 4-(3 - CH3Ophenylsulfonyl)piperazin- 1 -yl, 4-(4-CH3Ophenylsulfonyl)piperazin- 1 -yl, 4-(2-CO2H- phenylsulfonyl)piperazin- 1 -yl, 4-(3 -CO2Hphenylsulfonyl)piperazin- 1 -yl, 4-(4-CO2Hphenyl- sulfonyl)piperazin-l -yl, 4-(2-CONH2phenylsulfonyl)piperazin-l -yl, 4-(3-CONH2phenyl- sulfonyl)piperazin- 1 -yl, 4-(4-CONH2phenylsulfonyl)piperazin- 1 -yl, 4-(2-CON(CH3)2phenyl- sulfonyl)piperazin- 1 -yl, 4-(3 -CON(CH3)2phenylsulfonyl)piperazin- 1 -yl, 4-(4- CON(CH3)2phenylsulfonyl)piperazin-l-yl, 4-(methylphenylNCO)piperazin-l-yl, 4-[(3- CONH2phenyl)methylNCO]piρerazin- 1 -yl, 4-[(4-CONH2phenyl)methylNCO]piρerazin- 1 -yl, 4-[(2-CONH2phenyl)methylNCO]ρiperazin-l-yl, 4-[(3-Fphenyl)methylNCO]ρiperazin-l-yl, 4-[(4-Fphenyl)methylNCO]piperazin-l-yl, 4-[(2-Fρhenyl)methylNCO]ρiperazin-l-yl, 4-[(3- OCH3phenyl)methylNCO]ρiperazin- 1 -yl, 4-[(4-OCH3phenyl)methylNCO]piperazin- 1 -yl, A-
A6 [(2-OCH3phenyl)methylNCO]piperazin-l-yl, 4-[(3-CNphenyl)methylNCO]piperazin-l-yl, 4- [(4-CNphenyl)methylNCO]piperazin- 1 -yl, 4- [(2-CNphenyl)methylNCO]piperazin- 1 -yl, 4- [(3-OHphenyl)methylNCO]piperazin-l-yl, 4-[(4-OHphenyl)methylNCO]piperazin-l-yl, 4- [(2-OHphenyl)methylNCO]piperazin- 1 -yl, 4-[(pyridin-2-yl)methylNCO]piperazin- 1 -yl, 4- [(pyridin-3-yl)methylNCO]piperazin-l-yl, 4-[(pyridin-4-yl)methylNCO]piperazin-l-yl, 4- [(pyrimidin-2-yl)methylNCO]piperazin- 1 -yl, 4-[(pyrimidin-4-yl)methylNCO]piperazin- 1 -yl, 4-[(pyrimidin-5-yl)methylNCO]piperazin-l-yl, 4-[(3-CO2Hphenyl)methylNCO]piperazin-l- yl, 4-[(4-CO2Hphenyl)methylNCO]piperazin-l-yl, 4-[(2-CO2Hρhenyl)methylNCO]piρerazin- 1-yl, 4-[(3-CH3phenylmethyl)NHCO]piperazin-l-yl, 4-[(4- CH3phenylmethyl)NHCO]piperazin- 1 -yl, 4-[(2-CH3phenylmethyl)NHCO]piρerazin- 1 -yl, 4- [(3-CONH2phenylmethyl)NHCO]piperazin-l-yl, 4-[(4-
CONH2phenylmethyl)NHCO]piρerazin-l-yl, 4-[(2-CONH2phenylmethyl)NHCO]piρerazin- 1 -yl, 4-[(3 -Fphenylmethyl)NHCO]piperazin- 1 -yl, 4-[(4-Fphenylmethyl)NHCO]piperazin- 1 - yl, 4-[(2-Fphenylmethyl)NHCO]ρiperazin-l-yl, 4-[(3-OCH3phenylmethyl)NHCO]piperazin- 1-yl, 4-[(4-OCH3phenylmethyl)NHCO]piperazin-l-yl, 4-[(2-
OCH3phenylmethyl)NHCO]piperazin-l-yl, 4-[(3-CNphenylmethyl)NHCO]piperazin-l-yl, 4- [(4-CNρhenylmethyl)NHCO]piρerazin- 1 -yl, 4- [(2-CNρhenylmethyl)NHCO]ρiperazin- 1 -yl, 4-[(3 -OHphenylmethyl)NHCO]piperazin- 1 -yl, 4- [(4-OHphenylmethyl)NHCO]piperazin- 1 - yl, 4-[(2-OHphenylmethyl)NHCO]piperazin-l-yl, 4-[(pyridin-2-ylmethyl)NHCO]piperazin- 1 -yl, 4-[(pyridin-3 -ylmethyl)NHCO]piperazin- 1 -yl, 4-[(pyridin-4-ylmethyl)NHCO]piperazin- 1 -yl, 4- [(pyrimidin-2-ylmethyl)NHCO]piperazin- 1 -yl, 4-[(pyrimidin-4- ylmethyl)NHCO]piperazin- 1 -yl, 4- [(pyrimidin-5-ylmethyl)NHCO]piperazin- 1 -yl, 4- [(3- CO2Hphenylmethyl)NHCO]piperazin- 1 -yl, 4-[(4-CO2Hphenylmethyl)NHNCO]-piperazin- 1 - yl, 4-[(2-CO2Hphenylmethyl)NHCO]piperazin-l-yl, 4-[(pyrazin-2-ylmethyl)- NHCO]piperazin-l-yl, 4-[(pyridazin-3-ylmethyl)NHCO]piperazin-l-yl, 4-[(pyridazin-4- ylmethyl)NHCO]piperazin- 1 -yl, 4-[([ 1.3.5]triazin-2-ylmethyl)NHCO]ρiperazin-l -yl, 4-[N- (3-CH3phenylmethyl)N(CH3)CO]piperazin-l-yl, 4-[ JV-(4- CH3phenylmethyl)N(CH3)CO]piperazin-l-yl, 4-[ N-(2- CH3phenylmethyl)Ν(CH3)CO]piperazin-l-yl, 4-[ N-(3-COΝH2phenyl- methyl)N(CH3)CO]piperazin-l-yl, 4-[ N-(4-CONH2phenylmethyl)N(CH3)CO]piperazin-l-yl, 4-[ N-(2-CONH2phenylmethyl)N(CH3)CO]piperazin-l-yl, 4-[ N-(3-
Fphenylmethyl)Ν(CH3)CO]-piperazin-l-yl, 4-[N-(4-Fphenylmethyl)Ν(CH3)CO]piperazin-l- yl, 4-[ N-(2-Fphenylmethyl)-N(CH3)CO]piperazin-l-yl, 4-[ N-(3- OCH3phenylmethyl)Ν(CH3)CO]piperazin-l-yl, 4-[ N-(4- OCH3phenylmethyl)N(CH3)CO]ρiperazin-l -yl, 4-[ N-(2-OCH3ρhenylmethyl)N(CH3)CO]- piρerazin-1-yl, 4-[ N-(3-CNphenylmethyl)N(CH3)CO]piperazin-l-yl, 4-[ N-(4-CNphenyl- methyl)N(CH3)CO]piperazin-l-yl, 4-[ iV-(2-CNphenylmethyl)N(CH3)CO]piperazin-l-yl, 4-[ iV-(3-OHphenylmethyl)N(CH3)CO]piperazm-l-yl, 4-[ N-(4-OHphenylmethyl)N(CH3)CO]- piperazin-1-yl, 4-[ N-(2-OHphenylmethyl)N(CH3)CO]piperazin-l-yl, 4-[ N-(pyridin-2- ylmethyl)N(CH3)CO]ρiperazin-l-yl, 4-[ N-(pyridin-3-ylmethyl)N(CH3)CO]piperazin-l-yl, 4- [ N-(pyridin-4-ylmethyl)N(CH3)CO]piperazin-l-yl, 4-[ iV-(pyrimidin-2- ylmethyl)N(CH3)CO]-piρerazin-l-yl, 4-[ N-(pyrimidin-4-ylmethyl)N(CH3)CO]piperazin-l- yl, 4-[ iV-(pyrimidin-5-ylmethyl)N(CH3)CO]ρiperazin-l-yl, 4-[ JV-(3- CO2Hphenylmethyl)N(CH3)CO]piperazin-l-yl, 4-[ N-(4-
CO2Hphenylmethyl)N(CH3)NCO]piperazin- 1 -yl, 4-[ N-(2-CO2Hphenylmethyl)- N(CH3)CO]piperazin-l-yl, 4-[ N-(pyrazm-2-ylmethyl)N(CH3)CO]piperazin-l-yl, 4-[ N- (pyridazin-3-ylmethyl)N(CH3)CO]piperazin-l-yl, 4-[ N-(pyridazin-4-ylmethyl)N(CH3)CO]- piperazin-1-yl, 4-[ N-([1.3.5]triazin-2-ylmethyl)N(CH3)CO]piperazin-l-yl, 4-(pyridin-4- yl)piperazin-l-yl, 4-(pyridin-3-yl)piperazin-l-yl, 4-(pyridin-2-yl)piperazin-l-yl, 4- (pyrimidin-4-yl)piperazin- 1 -yl, 4-(pyrimidin-2-yl)piperazin- 1 -yl, 4-(pyrimidin-5- yl)piperazin-l-yl, 4-([1.3.5]triazm-2-yl)piperazin-l-yl, 4-(phenyl)piperazin-l-yl, 4-(pyrazin- 2-yl)piperazin-l-yl, 4-(pyridazin-3-yl)piperazin-l-yl, 4-(pyridazin-4-yl)piperazin-l-yl, 4-(4- Fphenyl)piperazin-l-yl, 4-(3-Fphenyl)piperazin-l-yl, 4-(2-Fphenyl)piperazin-l-yl, 4-(2,4- diFphenyl)piperazin-l-yl, 4-(2,3-diFphenyl)piperazin-l-yl, 4-(2,5-diFphenyl)piperazin-l-yl, 4-(2,6-diFphenyl)piperazin-l-yl, 4-(2,4,6-triFphenyl)piperazin-l-yl, 4-(2,3,6- triFphenyl)piperazin- 1 -yl, 4-(2,3 ,4-triFphenyl)-piperazin- 1 -yl, 4-(4-CH3Ophenyl)piperazin- 1 - yl, 4-(3-CH3Ophenyl)piperazin-l-yl, 4-(2-CH3Ophenyl)piperazin-l-yl, 4-(4- CNphenyl)piperazin-l-yl, 4-(3-CNphenyl)piperazin-l-yl, 4-(2-CNphenyl)piperazin-l-yl, 4- (4-CO2Hphenyl)piperazin-l-yl, 4-(3-CO2Hphenyl)piperazin-l-yl, 4-(2-
CO2Hphenyl)piperazin-l-yl, 4-(4-CONH2phenyl)piperazin-l-yl, 4-(3-CONH2phenyl)- piperazin-1-yl, 4-(2-CONH2phenyl)piperazin-l-yl, 4-(methylcarbonyl)piperazin-l-yl, A- (trifluoromethylcarbonyl)piperazin- 1 -yl, 4-(ethylcarbonyl)piperazin- 1 -yl, A- (isopropylcarbonyl)-piperazin- 1 -yl, 4-(ført-butylcarbonyl)piperazin- 1 -yl, A- (cyclopropylCO)piperazin- 1 -yl, 4-(cyclobutylCO)piperazin- 1 -yl, A-
(cyclopentylCO)piperazin- 1 -yl, 4-(azetidin-3 -ylCO)piperazin- 1 -yl, 4-(3 -CHOpyrrolidin- 1 - ylCO)piperazin- 1 -yl, 4-(3 -CFsOpyrrolidin- 1 -ylCO)ρiperazin- 1 -yl, 4-(3 -CHF2Opyrrolidin- 1 - ylCO)piperazin- 1 -yl, 4-(3 -COaHpyrrolidin- 1 -ylCO)piperazin- 1 -yl, 4-(3 -CNpyrrolidin- 1 - ylCO)piperazin- 1 -yl, 4-(3 -CONH^yrrolidin- 1 -ylCO)piperazin- 1 -yl, 4-(pyrrolidin- 1 - ylCO)piperazin-l -yl, 4(oxazolidin-3-ylCO)piperazin-l -yl, 4-(tetrahydrofuran-3- ylCO)piperazin- 1 -yl, 4-(tetrahydrothiophen-3 -ylCO)piperazin- 1 -yl, 4-( 1 , 1 -dioxo- 1 λ6- tetrahydrothiophen-3 -ylCO)piperazin- 1 -yl, 4-(cyclohexylCO)piperazin- 1 -yl, 4- (tetrahydropyran-4-ylCO)piperazin- 1 -yl, 4-(piperdin- 1 -ylCO)piperazin- 1 -yl, A- (tetrahydrothiopyran-4-ylCO)piperazin- 1 -yl, 4-(l , 1 -dioxo- 1 λ6-hexahydrothiopyran-4- ylCO)piperazin- 1 -yl, 4-(piperidin- 1 -ylCO)piperazin- 1 -yl, 4-(morpholin-4-ylCO)piperazin- 1 - yl, 4-(thiomorpholm-4-ylCO)piperazin- 1 -yl, 4-(l,l -dioxo- 1 λ6-thiomorpholin-4- ylCO)piperazin-l-yl, 4-(piperazin-l-ylCO)piperazin-l-yl, 4-(phenylCO)piperazin-l-yl, 4-(2- CH3phenylCO)piperazin-l-yl, 4-(3-CH3ρhenylCO)piperazin-l-yl, 4-(4- CH3phenylCO)piperazin- 1 -yl, 4-(2-FphenylCO)piperazin- 1 -yl, 4-(3 -FphenylCO)piperazin- 1 - yl, 4-(4-FρhenylCO)piperazin-l-yl, 4-(2-OHphenylCO)-piperazin-l-yl, 4-(3- OHphenylCO)piρerazin- 1 -yl, 4-(4-OHρhenylCO)piρerazin- 1 -yl, 4-(2- CH3OphenylCO)piperazin- 1 -yl, 4-(3-CH3OphenylCO)piperazin- 1 -yl, 4-(4-CH3OphenylCO)- piperazin-1-yl, 4-(2-CO2HphenylCO)piperazin-l-yl, 4-(3-CO2HphenylCO)piperazin-l-yl, 4- (4-CO2HphenylCO)piperazin-l-yl, 4-(2-CONH2phenylCO)piperazin-l-yl, 4-(3- CONH2phenylCO)-piρerazin- 1 -yl, 4-(4-CONH2phenylCO)piperazin- 1 -yl, 4-(2- CNphenylCO)piperazin- 1 -yl, 4-(3-CNphenylCO)piperazin-l -yl, 4-(4- CNphenylCO)piperazin- 1 -yl, 4-(pyridin-4-ylCO)piperazin- 1 -yl, 4-(pyridin-3 - ylCO)piperazin- 1 -yl, 4-(pyridin-2-ylCO)piρerazin- 1 -yl, 4-(pyrimidin-4-ylCO)piperazin- 1 -yl, 4-(pyrimidin-2-ylCO)piperazin- 1 -yl, 4-(pyrimidin-5-ylCO)piperazin- 1 -yl 4-
(benzylCO)piperazin- 1 -yl, 4-(2-FphenylmethylCO)piperazin- 1 -yl, 4-(3 -FphenylmethylCO)- piperazin- 1 -yl, 4-(4-FphenylmethylCO)piperazin- 1 -yl, 4-(2-OHphenylmethylCO)piperazin- 1-yl, 4-(3-OHphenylmethylCO)piperazin-l-yl, 4-(4-OHphenylmethylCO)piperazin-l-yl, 4- (2-CH3OphenylmethylCO)piperazin- 1 -yl, 4-(3 -CH3OphenylmethylCO)piperazin- 1 -yl, 4-(4- CH3OphenylmethylCO)piperazin- 1 -yl, 4-(2-CO2HphenylmethylCO)piperazin- 1 -yl, 4-(3 - CO2HphenylmethylCO)piperazin-l-yl, 4-(4-CO2HphenylmethylCO)piperazin-l-yl, 4-(2- CONH2phenylmethylCO)piperazin- 1 -yl, 4-(3 -CONH2phenylmethylCO)piperazin- 1 -yl, 4-(4- CONH2phenylmethylCO)piρerazin- 1 -yl, 4-(2-CNphenylmethylCO)piperazin- 1 -yl, 4-(3- CNphenylmethylCO)piperazin- 1 -yl, 4-(4-CNphenylmethylCO)piperazin- 1 -yl, 4-(pyridin-4- ylmethylCO)piperazin-l-yl, 4-(pyridin-3-ylmethylCO)piperazin-l-yl, 4-(pyridin-2- ylmethylCO)piperazin- 1 -yl, 4-(pyrimidin-4-ylmethylCO)piperazin- 1 -yl, 4-(pyrimidin-2- ylmethylCO)piperazin- 1 -yl, 4-(pyrimidin-5 -ylmethylCO)piperazin- 1 -yl, 4-(pyriazin-2- ylmethylCO)piperazin- 1 -yl, 4-(phenylNHCO)piperazin- 1 -yl, 4-(2-FphenylNHCO)piperazin- 1-yl, 4-(3-FphenylNHCO)piperazin-l-yl, 4-(4-FphenylNHCO)piperazin-l-yl, 4-(2- OHρhenyl-NHCO)-piperazin- 1 -yl, 4-(3 -OHphenylNHCO)piperazin- 1 -yl, 4-(4- OHphenylNHCO)piρerazin-l-yl, 4-(2-CH3OphenylNHCO)piperazin-l-yl, 4-(3- CH3OphenylNHCO)piperazin- 1 -yl, 4-(4-CH3OphenylNHCO)piperazin- 1 -yl, 4-(2- CO2HphenylNHCO)piperazin-l-yl, 4-(3-CO2Hphenyl-NHCO)piperazin-l-yl, 4-(4- CO2HphenylNHCO)piperazin- 1 -yl, 4-(2-CONH2phenylNHCO)-piperazin- 1 -yl, 4-(3- CONH2phenylNHCO)piperazin- 1 -yl, 4-(4-CONH2phenylNHCO)piperazin- 1 -yl, 4-(2- CNρhenylNHCO)piρerazin-l-yl, 4-(3-CNρhenylNHCO)piρerazin-l-yl5 4-(4- CNphenylNHCO)piperazin- 1 -yl, 4-(pyridin-4-ylNHCO)piperazin- 1 -yl, 4-(pyridin-3 - ylNHCO)-piperazin- 1 -yl, 4-(pyridin-2-ylNHCO)piperazin- 1 -yl, 4-(pyrimidin-4- ylNHCO)piperazin- 1 -yl, 4-(pyrimidin-2-ylNHCO)piperazin- 1 -yl, 4-(pyrimidin-5- ylNHCO)piperazin-l-yl, 4-([l .3.5]triazin-2-ylNHCO)piperazin-l-yl, 4-(l,2,3,4- tetrahydroquinolin-l-ylCO)piperazin-l-yl, 4-(7-CONH2-l,2,3,4-tetrahydroquinolin-l- ylCO)piperazin-l-yl, 4-(6-CONH2-1, 2,3, 4-tetrahydroquinolin-l-ylCO) piperazin-1-yl, 4-(8- CONH2-1, 2,3, 4-tetrahydroquinolin-l-ylCO) piperazin-1-yl, 4-(7-F- 1,2,3, 4- tetrahydroquinolin- 1 -ylCO)piperazin- 1 -yl, 4-(6-F- 1 ,2,3 ,4-tetrahydroquinolin- 1 - ylCO)piρerazin- 1 -yl, 4-(8-F- 1 ,2,3 ,4-tetrahydroquinolin- 1 -ylCO)ρiperazin- 1 -yl, 4-(7-CH3O- 1 ,2,3 ,4-tetrahydroquinolin- 1 -ylCO)piperazin- 1 -yl, 4-(6-CH3O-1 ,2,3 ,4-tetrahydroquinolin- 1 - ylCO) piperazin-1-yl, 4-(8-CH3O-1, 2,3,4-tetrahydroquinolin-l-ylCO) piperazin-1-yl, 4-(7- CN- 1 ,2,3 ,4-tetrahydroquinolin- 1 -ylCO)ρiperazin- 1 -yl, 4-(6-CN- 1 ,2,3 ,4-tetrahydroquinolin- 1 - ylCO)ρiperazin- 1 -yl, 4-(8-CN- 1 ,2,3 ,4-tetrahydroquinolin- 1 -ylCO)-piperazin- 1 -yl, 4-(7-OH- 1 ,2,3,4-tetrahydroquinolin- 1 -ylCO)piperazin- 1 -yl, 4-(6-OH- 1 ,2,3 ,4-tetrahydroquinolin- 1 - ylCO)ρiperazin- 1 -yl, 4-(8-OH- 1 ,2,3 ,4-tetrahydroquinolin- 1 -ylCO)ρiperazin- 1 -yl, 4-(7-CO2H- 1 ,2,3 ,4-tetrahydroquinolin- 1 -ylCO)piperazin- 1 -yl, 4-(6-CO2H- 1 ,2,3 ,4-tetrahydroquinolin- 1 - ylCO)piperazin-l-yl, 4-(8-CO2H-l,2,3,4-tetrahydroquinolin-l-ylCO)piperazin-l-yl, 4- ( 1 ,2,3 ,4-tetrahydro- [ 1 ,8]naphthyridin- 1 -ylCO)piperazin- 1 -yl, 4-( 1 ,2,3 ,4-tetrahydro- [l,7]naphthyridin-l-ylC0)piperazin-l-yl, 4-(l,2,3,4-tetrahydro-[l,6]naphthyridin-l- ylCO)piperazin-l-yl, 4-(5,6J7,8-tetraliydropyrido[2,3-b]pyrazin-5-ylCO)piperazin-l-yl, 4- (5,6,7,8-tetrahydropyrido[2,3-d]pyrimidin-8-ylCO)piperazin-l-yl, 4-(5,6,7,8- tetrahydropyrido[3,2-d]pyrimidin-5-ylCO)piperazin-l-yl, 4-(5,6,7,8-tetrahydropyrido[2,3- djpyridazin- 1 -ylCO)piperazin- 1 -yl, 4-(2,3 -dihydro- lH-indol- 1 -ylCO)piperazin- 1 -yl, 4-(7- CONH2-2,3 -dihydro- lH-indol- 1 -ylCO)piperazin- 1 -yl, 4-(6-CONH2-2,3 -dihydro- lH-indol- 1 - ylCO)piperazin-l-yl, 4-(5-CONH2-2,3-dihydro-lH-indol-l-ylCO)piρerazin-l-yl, 4-(7-F-2,3- dihydro- 1 H-indol- 1 -ylCO)piρerazin- 1 -yl, 4-(6-F-2,3 -dihydro- 1 /i-indol- 1 -ylCO)piperazin- 1 - yl, 4-(5-F-2,3-dihydro-lH-indol-l-ylCO)piperazin-l-yl, 4iV-(7-CH3O-2,3-dihydro-lH-indol- l-ylCO)piperazin-l-yl, 4-(6-CH3O-2,3-dihydro-lH-indol-l-ylCO)piperazin-l-yl, 4-(5-CH3O- 2,3-dihydro-li/-indol-l-ylCO)piperazm-l-yl5 4-(7-CN-2,3-dihydro-lH-indol-l- ylCO)piperazin- 1 -yl, 4-(6-CN-2,3 -dihydro- lH-indol- 1 -ylCO)piperazin- 1 -yl, 4-(5-CN-2,3 - dihydro-lH-indol-l-ylCO)piperazin-l-yl, 4-(7-OH-2,3-dihydro-lH-indol-l-ylCO)piperazin- l-yl5 4.(6-OH-2,3-dihydro-lH-indol-l-ylCO)piperazin-l-yl, 4-(5-OH-2,3-dihydro-lH-indol- 1 -ylCO)piperazin- 1 -yl, 4-(7-CO2H-2,3 -dihydro- 1 H-indol- 1 -ylCO)piperazin- 1 -yl, 4-(6-CO2H- 2,3-dihydro-lH-indol-l-ylCO)piperazin-l-yl5 4-(5-CO2H-2,3-dihydro-lH-indol-l- ylCO)piperazin- 1 -yl, 4-(2,3 -dihydro- 1 H-pyrrolo [2,3 -bjpyridin- 1 -ylCO)piperazin- 1 -yl, 4-(2,3 - dihydro- 1 H-pyrrolo [2,3-c]pyridin- 1 -ylCO)piperazin- 1 -yl, 4-(2,3 -dihydro- 1 H-pyrrolo [3 ,2- c]pyridin-l -ylCO)piperazin-l -yl, 4-(6,7-dihydro-5H-pyrrolo[2,3-b]pyrazin-5- ylCO)piperazin-l-yl, 4-(6,7-dihydro-5H-pyrrolo[2,3-d]pyrimidin-7-ylCO)piperazin-l-yl, 4- (6,7-dihydro-5H-pyrrolo[3,2-d]pyrimidin-5-ylCO)piperazin-l-yl, or 4-(2,3 -dihydro- IH- pyrrolo [2,3 -d]pyridazin- 1 -ylCO)piperazin- 1 -yl.
[0123] Preferably, the stereochemistry at the carbon to which R5 is attached is (R) and to which R4 and R6 are attached is (S).
[0124] Preferably, the stereochemistry at the carbon to which R5 and R6 are attached is (R) and to which R4 is attached is (S).
[0125] (B) Another preferred group of compounds of Formula (I) is that wherein:
R3 is alkyl, preferably methyl or ethyl and R4 is alkyl, preferably methyl, ethyl, propyl or butyl, more preferably R4 is methyl. Preferably, R3 and R4 are methyl. [0126] (C) Yet another preferred group of compounds of Formula (I) is that wherein R3 and R4 together with the carbon atom to which they are attached form cycloalkylene, preferably cyclopropylene, cyclopentylene, or cyclohexylene, more preferably cyclopropylene.
[0127] (D) Yet another preferred group of compounds of Formula (I) is that wherein R3 and R4 together with the carbon atom to which they are attached form piperidin-4-yl substituted at the nitrogen atom with ethyl, 2,2,2-trifluoroethyl or cyclopropyl, tetrahydropyran-4-yl, tetrahydrothiopyran-4-yl, or l,l-dioxotetrahydrothiopyran-4-yl. [0128] (E) Yet another preferred group of compounds of Formula (I) is that wherein R6 is haloalkyl, preferably, difluoromethyl, trifluoromethyl, 2,2,2-trifluoroethyl, or 2,2,3,3,3- pentafluoropropyl and R7 and R8 are hydrogen.
[0129] (F) Yet another preferred group of compounds of Formula (I) is that wherein R6 is haloalkyl, preferably, difluoromethyl, trifluoromethyl, 2,2,2-trifluoroethyl, or 2,2,3,3,3- pentafluoropropyl, R7 is haloalkyl, preferably, trifluoromethyl, 2,2,2-trifluoroethyl, or 2,2,3,3,3-pentafluoropropyl, and R8 are hydrogen.
[0130] (G) Yet another preferred group of compounds of Formula (I) is that wherein R6 is haloalkyl, preferably, difluoromethyl, trifluoromethyl, 2,2,2-trifluoroethyl, or 2,2,3,3,3- pentafluoropropyl, R7 is alkyl, preferably, methyl, ethyl, or propyl, and R8 are hydrogen.
[0131] With the preferred groups (B)-(C), more preferred groups of compounds are those wherein R1, R2, R5, R6, R7 and R8 are as defined for group (A) above.
[0132] With the preferred groups (D)-(G), more preferred groups of compounds are those wherein R1, R2, R3, R4, and R5 are as defined for group (A) above.
[0133] It should be noted that reference to the preferred embodiments set forth above includes all combinations of particular and preferred groups unless stated otherwise.
GENERAL SYNTHETIC SCHEME
[0134] Compounds of this invention can be made by the methods depicted in the reaction schemes shown below.
[0135] The starting materials and reagents used in preparing these compounds are either available from commercial suppliers such as Aldrich Chemical Co., (Milwaukee, Wis.), Bachem (Torrance, Calif), or Sigma (St. Louis, Mo.) or are prepared by methods known to those skilled in the art following procedures set forth in references such as Fieser and Fieser's Reagents for Organic Synthesis, Volumes 1-17 (John Wiley and Sons, 1991); Rodd's Chemistry of Carbon Compounds, Volumes 1-5 and Supplementals (Elsevier Science Publishers, 1989); Organic Reactions, Volumes 1-40 (John Wiley and Sons, 1991), March's Advanced Organic Chemistry, (John Wiley and Sons, 4th Edition) and Larock's Comprehensive Organic Transformations (VCH Publishers Inc., 1989). These schemes are merely illustrative of some methods by which the compounds of this invention can be synthesized, and various modifications to these schemes can be made and will be suggested to one skilled in the art having referred to this disclosure.
[0136] The starting materials and the intermediates of the reaction may be isolated and purified if desired using conventional techniques, including but not limited to filtration, distillation, crystallization, chromatography and the like. Such materials may be characterized using conventional means, including physical constants and spectral data.
[0137] Unless specified to the contrary, the reactions described herein take place at atmospheric pressure over a temperature range from about -78 0C to about 150 0C, more preferably from about 0 0C to about 125 0C and most preferably at about room (or ambient) temperature, e.g., about 200C.
[0138] In the reactions described hereinafter it may be necessary to protect reactive functional groups, for example hydroxy, amino, imino, thio or carboxy groups, where these are desired in the final product, to avoid their unwanted participation in the reactions. Conventional protecting groups may be used in accordance with standard practice, for examples see T. W. Greene and P. G. M. Wuts in "Protective Groups in Organic Chemistry" John Wiley and Sons, 1999.
[0139] Compounds of Formula (I) where R1, R2, R3, R4, R5, R6 and R8 are as defined in the Summary of the Invention and R7 is hydrogen can be prepared by proceeding as in the following Reaction Scheme 1 below.
Scheme 1
[0140] Reaction of a ketone of formula 1 where R6 and R8 are as defined in the Summary of the Invention with an α-amino ester of formula 2 where R is a carboxy protecting group, preferably an alkyl group, preferably methyl, and R5 is as defined in the Summary of the Invention under reductive amination reaction conditions provide a compound of formula 3. The reaction is carried out in the presence of a suitable dehydrating agent such as TiCl4, magnesium sulfate, isopropyl trifluoroacetate, in the presence of a base such as diisopropylethylamine, pyridine, and the like and in a suitable organic solvent such as methylene chloride to give an imine. The imine is reduced with a suitable reducing agent such as sodium borohydride, sodium cyanoborohydride, and the like in a suitable organic solvent such as methanol, ethanol, and the like.
[0141] Compounds of formula 1 such as 2,2,2-trifluoromethylacetophenone is commercially available. Others can be prepared by methods well known in the art. α- Amino esters of formula 2 can be prepared by methods well known in the art. For example, a compound of formula 2 where R5 is -alkylene-SO2NRHR12 where R11 and R12 are as defined in the Summary of the Invention can be prepared by the procedure described in Ross, D.L.; Skinner, C.G.; Shive, W. J Org. Chem. 1959, 24, 1372-1374; b) Byrnes, S.; Burckart, G.J.; Mokotoff, M. J. Med. Chem. 1978, 21, 45-49.
[0142] Hydrolysis of the ester group in compound 3 provides a compound of formula 4. The hydrolysis conditions depend on the nature of the protecting group. For example, when R is alkyl the hydrolysis is carried out under aqueous basic hydrolysis reaction conditions to give the corresponding acid of formula 4. The reaction is typically carried out with cesium carbonate, lithium hydroxide, and the like in an aqueous alcohol such as methanol, ethanol, and the like.
[0143] Compound 4 is then reacted with an α-hydroxyketoamide of formula 5 to give a compound of Formula 6. The reaction is typically carried out in the presence of a suitable coupling agent e.g., benzotriazole-1-yloxytrispyrrolidinophosphonium hexafluorophosphate (PyBOP®), O-benzotriazol-l-yl-iV^N'jiV'-tetramethyl-uronium hexafluorophosphate (HBTU), <9-(7-azabenzotriazol- 1 -yl)- 1 , 1 ,3,3 -tetramethyl-uronium hexafluorophosphate (HATU), l-(3-dimethylaminopropyl)-3-ethylcarbodiimide hydrochloride (EDC), or l^-dicyclohexyl-carbodiimide (DCC), optionally in the presence of 1-hydroxy-benzotriazole (HOBT), and a base such as AyV-diisopropylethylamine, triethylamine, iV-methylmorpholine, and the like. The reaction is typically carried out at 20 to 300C, preferably at about 25 0C, and requires 2 to 24 h to complete. Suitable reaction solvents are inert organic solvents such as halogenated organic solvents (e.g., methylene chloride, chloroform, and the like), acetonitrile, iV,7V-dimethylformamide, ethereal solvents such as tetrahydrofuran, dioxane, and the like.
[0144] Alternatively, the above coupling step can be carried out by first converting 4 into an active acid derivative such as succinimide ester and then reacting it with an α- hydroxyketoamide of formula 5. The reaction typically requires 2 to 3 h to complete. The conditions utilized in this reaction depend on the nature of the active acid derivative. For example, if it is an acid chloride derivative of 4, the reaction is carried out in the presence of a suitable base (e.g. triethylamine, diisopropylethylamine, pyridine, and the like). Suitable reaction solvents are polar organic solvents such as acetonitrile, Λζ,Λf-dimethylformarnide, dichloromethane, or any suitable mixtures thereof. Compounds of formula 5 can be prepared by methods well known in the art e.g., they can be prepared by the procedures described in PCT application publication No. WO 02/18369, the disclosure of which is incorporated herein by reference in its entirety.
[0145] Oxidation of the hydroxyl group in compound 6 with a suitable oxidizing agent such as OXONE® provides a compound of Formula (I).
[0146] Alternatively, compounds of Formula (I) where R1, R2, R3, R4, R5, R6 and R8 are as defined in the Summary of the Invention and R7 is hydrogen can be prepared by proceeding as in the following Reaction Scheme 2 below.
Scheme 2
7 8 9 10
[0147] Reaction of a compound of formula 8 where R5 is as defined in the Summary of the
Invention and PG is a suitable oxygen protecting group with a hemiacetal of formula 7 where R6 is as defined in the Summary of the Invention provides an imine compound of of formula 9. Treatment of 9 with an organolithium compound of formula R Li where R is not hydrogen provides compound 10. Removal of the oxygen protecting group, followed by oxidation of the resulting alcohol 11 provides a compound of formula 4 which is then converted to a compound of Formula (I) as described in Scheme 1 above. Suitable oxygen protecting groups and reaction conditions for putting them on and removing them can be found in Greene, T. W.; and Wuts, P. G. M.; Protecting Groups in Organic Synthesis; John Wiley & Sons, Inc. 1999.
[0148] Alternatively, compounds of Formula (I) where R1, R2, R3, R4, R5, R6 and R8 are as defined in the Summary of the Invention and R7 is hydrogen can be prepared by proceeding as in the following Reaction Scheme 3 below.
Scheme 3
- (I)
[0149] Reaction of an amino acid compound of formula 2 where R is alkyl and R5 is as defined in the Summary of the Invention with a hemiacetal compound of formula 7 provides a 2-(l-hydroxy-2,2,2-trifluoroethylarnino)acetate compound of formula 12. The reaction is carried out in the presence of a catalytic amount of an acid such as^>-toluenesulfonic acid and in an aromatic hydrocarbon solvent such as toluene, benzene, and the like.
[0150] Treatment of 12 with a compound of formula R8H where R8 is aryl or heteroaryl under Friedel-Crafts reaction conditions or trialkylaluminum in toluene (to give 3 where R8 is alkyl) provides a compound of formula 3 which is then converted to a compound of Formula (I) as described above.
[0151] Alternatively, compounds of Formula (I) where R1, R2, R3, R4, R5, R6 and R8 are as defined in the Summary of the Invention and R7 is hydrogen can be prepared by proceeding as in the following Reaction Scheme 4 below.
Scheme 4 optional
— *" activated acid deriv.
[0152] Reaction of a compound of formula 13 where R is as defined in Summary of the Invention with a compound of formula 14 where R' is hydrogen or a carboxy protecting group and Rz is R5 or a precursor group (e.g., -alkylene-S-trityl, and the like) to R5 group provides a compound of formula 15. The reaction is carried out in a suitable organic solvent, including but not limited to, diethyl ether, tetrahydrofuran, acetonitrile, benzene, toluene, xylene, and the like, or mixtures thereof and optionally in the presence of an organic or inorganic base. Preferably, the organic base is triethylamine, pyridine, JV-methylmorpholine, collidine, diisopropylethylamine, and the like. Preferably, the inorganic base is cesium carbonate, sodium carbonate, sodium bicarbonate, and the like. The reaction is optionally carried out in the presence of a drying agent such as molecular sieves. Preferably, the reaction is carried out at room temperature.
[0153] Compounds of formula 13 can be prepared by methods well known in the art. For example, a compound of formula 13 where R8 is phenyl or 4-fluorophenyl and R6 is trifluoromethyl can be readily prepared from commercially available 2,2,2- trifluoroacetophenone or 2,2,2,4'-tetrafluoroacetophenone respectively, by reducing the keto group to an alcoholic group by suitable reducing agent such as sodium borohydride, lithium aluminum hydride, and the like. The solvent used depends on the type of reducing agent. For example, when sodium borohydride is used the reaction is carried out in an alcoholic organic solvent such as methanol, ethanol, and the like. When lithium aluminum hydride is used the reaction is carried out in an ethereal solvent such as tetrahydrofuran, and the like. Reaction of 2,2,2-trifluoro-l-phenylethanol or 2,2,2-trifluoro-l-(4-fiuorophenyl)ethanol with triflic anhydride or trifluoromethanesulfonyl chloride provides the desired compound. Compounds of formula 13 where R7 and R8 are hydrogen and R6 is 2,2,3,3,3- pentafluoropropyl can be prepared from commercially available 2,2,3, 3,3 -pentafluoropropan- l-ol can as described above. Optically enriched compound of formula 15 can be obtained by reduction of the corresponding halogenated acetophenone with a suitable reducing agent such as catecholborane or BH3-DMS complex in the presence of a suitable catalyst such as (S) or (i?)-methyl CBS oxazaborolidine catalyst or (S) or (R)-a,a -diphenyl-2-pyrrolidine-methanol in the presence of BBN to provide chiral alcohol which is then converted to compound 13 as described above. Compounds of formula 14 are either commercially available or they can be prepared by methods well known in the art.
[0154] Removal of the carboxy protecting group from a compound of formula 15 where R' is a protecting group provides a compound of formula 16. The conditions used to remove the carboxy protecting group depend on the nature of the carboxy protecting group. For example, if R' is allcyl, it is removed under basic hydrolysis reaction conditions utilizing aqueous base such as aqueous lithium hydroxide, sodium hydroxide, and the like in an alcoholic solvent such as methanol, ethanol, and the like. Additionally, if the Rz group in compound 14 is a precursor group to R5, it can be converted to R5 prior or after the ester hydrolysis step.
[0155] Compound 15 (where R' is hydrogen) or 16 is then converted to an activated acid derivative 17 (X is a leaving group) and which upon reaction with an aminoacetonitrile compound of formula 5 provides a compound of Formula (I) when Rz is R5 or a precursor compound to (I) when Rz is a precursor group to R5. The activated acid derivative can be prepared and then reacted with compound 5 in a stepwise manner or the activated acid derivative can be generated in situ in the presence of compound 5. For example, if the activated acid is acid halide it is first prepared by reacting 16 with a halogenating agent such as thionyl chloride, oxalyl chloride and the like and then reacted with compound 5. Alternatively, the activated acid derivative is generated in situ by reacting compound 16 and 5 in the presence of a suitable coupling agent e.g., benzotriazole- 1 -yloxytrispyrrolidinophosphonium hexafluorophosphate (PyBOP®), O-benzotriazol- 1 -yl-iV,iV,iV',iV'-tetramethyl-uronium hexafluorophosphate (HBTU), (9-(7-azabenzotriazol- 1 -yl)- 1 , 1 ,3 ,3 -tetramethyl-uronium hexafluorophosphate (HATU), l-(3-dimethylaminopropyl)-3-ethylcarbodiimide hydrochloride (EDC), 1,3-dicyclohexyl- carbodiimide (DCC), an the like, optionally in the presence of 1-hydroxybenzotriazole (HOBT), and in the presence of a base such as JV,JV-diisopropylethyIamine, triethylamine, N- methylmorpholine, and the like. Suitable reaction solvents are inert organic solvents such as halogenated organic solvents (e.g., methylene chloride, chloroform, and the like), acetonitrile, ΛζΛf-dimethylformamide, ethereal solvents such as tetrahydrofuran, dioxane, and the like. Alternatively, the activated acid can be reacted with CR1R2CNH2)CONH2 where R1 and R2 are as described in the Summary of the Invention, followed by conversion of the -CONH2 group to the cyano group by methods well known in the art. IfRz is a precursor group to R5, it is converted to R5 group to provide a compound of Formula (I) e.g, conversion of — alkylene-S-NRπR12 to -alkylene-SO2-NRπR12 under oxidation reaction conditions.
[0156] Alternatively, the compound of Formula (I) where R3 is -alkylene-SO2NRnR12 where R11 and R12 are as defined in the Summary of the Invention and where R1, R2, R3, R4, R6 and R8 are as defined in the Summary of the Invention can be prepared as illustrated and described in Scheme 5 below.
Scheme 5
19
[O]
(I)
[0157] Treatment of a compound of formula 18, prepared as described in Scheme 4 above, where Rz is -CH2-S-trityl with an oxidizing agent such as iodine in methanol to give the disulfide compound of formula 19. Oxidation of 19 with chlorine in the presence of water followed by treatment with an amine of formula NHR11R12 in the presence of a suitable organic base such as triethylamine, diisopropylamine, pyridine, and the like provides a compound of formulal 20 which upon oxidation provides a compound of Formula (I).
[0158] Amines of formula NHR11R12 are either commercially available or they can be prepared by methods known in the art. For example, 1-cyclopropylpiperazine was prepared according to Gillaspy, M. A.; Lefker, B. A.; Hada, W. A.; Hoover, DJ Tetrahedron Lett. 1995, 36, 7399-7402. Other cyclic amines can be prepared from commercially available starting materials. For example, analogs of piperazine can be prepared from 1-tert- butoxycarbonylpiperazine or 1-benzyloxy-carbonylpiperazine utilizing procedures well known in the art. For example, acylation of the 4-position can be performed by treatment with an acyl chloride (e.g. benzoyl chloride) or sulfonylation can be achieved by treatment with a sulfonyl chloride (e.g. methane sulfonyl chloride) in the presence of triethylamine or diisopropylethylamine in a suitable solvent such as, but not limited to, methylene chloride. Urea formation was achieved by treatment with an isocyanate (e.g. isopropylisocyanate) in a suitable solvent such as methylene chloride. Alkylation was achieved using alkyl electrophiles bearing a suitable leaving group such as halide, tosylate, or triflate (e.g. 2,2,2- trifluoroethyl trifluoromethanesulfonate, prepared by the treatment of 2,2,2-trifuoroethanol with triflic anhydride in the presence of diisopropylethylamine in methylene chloride) in a suitable solvent such as methylene chloride or diethyl ether in the presence of triethylamine or diisopropylamine if necessary. Alkylation can also be achieved via reductive animation using a suitable aldehyde in the presence of an acid catalyst and sodium cyanoborohydride in an acceptable solvent such as methanol. Removal of the tert-butyloxycarbonyl protection group can be achieved using trifluoroacetic acid in methylene chloride to produce the trifluoroacetate salt or 4 M hydrochloric acid in dioxane (Aldrich) to produce the HCl salt after solvent removal. The benzyloxycarbonyl group can be removed using 30% hydrobromic acid in acetic acid (Aldrich) in methylene chloride or by hydrogenation utilizing 10% Pd/C under an atmosphere of hydrogen gas in a suitable solvent such as ethanol. These examples are merely illustrative of some methods by which amines (HNR11R12) were made, and various modifications or additional procedures can be utilized to synthesize desirable amines and will be suggested to one skilled in the art having referred to this disclosure.
[0159] A compound of Formula (I) can be converted to other compounds of Formula (I). For example:
[0160] A compound of Formula (I) containing a hydroxy group may be prepared by de- alkylation/benzylation of an alkoxy/benzyloxy substituent; those containing an acid group, by hydrolysis of an ester group; and those containing a cyano, by displacement of a bromine atom on the corresponding compounds of Formula (I). A compound of Formula (I) containing a cyano group can be converted to a corresponding carboxy containing compound by hydrolysis of the cyano group. The carboxy group, in turn, can be converted to an ester group. ' [0161] A compound of Formula (I) can be prepared as a pharmaceutically acceptable acid addition salt by reacting the free base form of the compound with a pharmaceutically acceptable inorganic or organic acid. Alternatively, a pharmaceutically acceptable base addition salt of a compound of Formula (I) can be prepared by reacting the free acid form of the compound with a pharmaceutically acceptable inorganic or organic base. Inorganic and organic acids and bases suitable for the preparation of the pharmaceutically acceptable salts of compounds of Formula (I) are set forth in the definitions section of this Application. Alternatively, the salt forms of the compounds of Formula (I) can be prepared using salts of the starting materials or intermediates.
[0162] The free acid or free base forms of the compounds of Formula (I) can be prepared from the corresponding base addition salt or acid addition salt form. For example, a compound of Formula (I) in an acid addition salt form can be converted to the corresponding free base by treating with a suitable base (e.g., ammonium hydroxide solution, sodium hydroxide, and the like). A compound of Formula (I) in a base addition salt form can be converted to the corresponding free acid by treating with a suitable acid (e.g., hydrochloric acid, etc).
[0163] The JV-oxides of compounds of Formula (I) can be prepared by methods known to those of ordinary skill in the art. For example, iV-oxides can be prepared by treating an unoxidized form of the compound of Formula (I) with an oxidizing agent (e.g., trifluoroperacetic acid, permaleic acid, perbenzoic acid, peracetic acid,
/røetø-chloroperoxybenzoic acid, or the like) in a suitable inert organic solvent (e.g., a halogenated hydrocarbon such as dichloromethane) at approximately O0C. Alternatively, the JV~oxides of the compounds of Formula (I) can be prepared from the iV-oxide of an appropriate starting material.
[0164] Compounds of Formula (I) in unoxidized form can be prepared from iV-oxides of compounds of Formula (I) by treating with a reducing agent (e.g., sulfur, sulfur dioxide, triphenyl phosphine, lithium borohydride, sodium borohydride, phosphorus trichloride, tribromide, or the like) in a suitable inert organic solvent (e.g., acetonitrile, ethanol, aqueous dioxane, or the like) at 0 to 8O0C.
[0165] Prodrug derivatives of the compounds of Formula (I) can be prepared by methods known to those of ordinary skill in the art (e.g., for further details see Saulnier et α/.(1994), Bioorganic and Medicinal Chemistry Letters, Vol. 4, p. 1985). For example, appropriate prodrugs can be prepared by reacting a non-derivatized compound of Formula (I) with a suitable carbamylating agent (e.g., ljl-acyloxyalkylcarbonochloridatejpαra-nitrophenyl carbonate, or the like).
[0166] those of ordinary skill in the art. A detailed description of the techniques applicable to the creation of protecting groups and their removal can be found in T. W. Greene, Protecting Groups in Organic Synthesis, 3rd edition, John Wiley & Sons, Inc. 1999.
[0167] Compounds of the present invention may be conveniently prepared or formed during the process of the invention, as solvates (e.g. hydrates). Hydrates of compounds of the present invention may be conveniently prepared by recrystallisation from an aqueous/organic solvent mixture, using organic solvents such as dioxin, tetrahydrofuran or methanol.
[0168] Compounds of Formula (I) can be prepared as their individual stereoisomers by reacting a racemic mixture of the compound with an optically active resolving agent to form a pair of diastereoisomeric compounds, separating the diastereomers and recovering the optically pure enantiomer. While resolution of enantiomers can be carried out using covalent diasteromeric derivatives of compounds of Formula (I), dissociable complexes are preferred (e.g., crystalline diastereoisomeric salts). Diastereomers have distinct physical properties (e.g., melting points, boiling points, solubilities, reactivity, etc.) and can be readily separated by taking advantage of these dissimilarities. The diastereomers can be separated by chromatography or, preferably, by separation/resolution techniques based upon differences in solubility. The optically pure enantiomer is then recovered, along with the resolving agent, by any practical means that would not result in racemization. A more detailed description of the techniques applicable to the resolution of stereoisomers of compounds from their racemic mixture can be found in Jean Jacques Andre Collet, Samuel H. Wilen, Enantiomers, Racemates and Resolutions, John Wiley & Sons, Inc. (1981).
PREPARATION OF BIOLOGICAL AGENTS
[0169] In practicing this invention several processes for the generation or purification of biological agents are used. Methods for preparing the biologies are well known in the art as discussed below.
[0170] Monoclonal antibodies can be prepared using standard techniques well known in the art such as by the method of Kohler and Milstein, Nature 1975, 256:495, or a modification thereof, such as described by Buck et al. 1982, In Vitro 18:377. Typically, a mouse or rat is immunized with the MenB PS derivative conjugated to a protein carrier, boosted and the spleen (and optionally several large lymph nodes) removed and dissociated into single cells. If desired, the spleen cells may be screened (after removal of non- specifically adherent cells) by applying a cell suspension to a plate or well coated with the antigen. B-cells, expressing membrane-bound immunoglobulin specific for the antigen, will bind to the plate, and will not be rinsed away with the rest of the suspension. Resulting B- cells, or all dissociated spleen cells, are then induced to fuse with myeloma cells to form hybridomas. Representative murine myeloma lines for use in the hybridizations include those available from the American Type Culture Collection (ATCC).
[0171] Chimeric antibodies composed of human and non-human amino acid sequences may be formed from the mouse monoclonal antibody molecules to reduce their immunogenicity in humans (Winter et al. Nature 1991 349:293; Lobuglio et al. Proc. Nat. Acad. ScL USA 1989 86:4220; Shaw et al. J Immunol. 1987 138:4534; and Brown et al. Cancer Res. 1987 47:3577; Riechmann et al. Nature 1988 332:323; Verhoeyen et al. Science 1988 239:1534; and Jones et al. Nature 1986 321:522; EP Publication No.519, 596, published Dec. 23, 1992; and U.K. Patent Publication No. GB 2,276,169, published Sep. 21, 1994).
[0172] Antibody molecule fragments, e.g., F(ab')2, FV, and sFv molecules, that are capable of exhibiting immunological binding properties of the parent monoclonal antibody molecule can be produced using known techniques. Inbar et al. Proc. Nat. Acad. Sd. USA 1972 69:2659; Hochman et al. Biochem. 1976 15:2706; Ehrlich et al. Biochem. 1980 19:4091; Huston et al. Proc. Nat. Acad. ScI USA 1988 85(16):5879; and U.S. Pat. Nos. 5,091,513 and 5,132,405, and U.S. Pat. No. 4,946,778.
[0173] In the alternative, a phage-display system can be used to expand the monoclonal antibody molecule populations in vitro. Saiki, et al. Nature 1986 324:163; Scharf et al. Science 1986 233:1076; U.S. Pat. Nos. 4,683,195 and 4,683,202; Yang et al. J. MoI. Biol. 1995 254:392; Barbas, III et al Methods: Comp. Meth Enzymol. 1995 8:94; Barbas, III et al. Proc. Natl. Acad. Sci. USA 1991 88:7978.
[0174] The coding sequences for the heavy and light chain portions of the Fab molecules selected from the phage display library can be isolated or synthesized, and cloned into any suitable vector or replicon for expression. Any suitable expression system can be used, including, for example, bacterial, yeast, insect, amphibian and mammalian systems. Expression systems in bacteria include those described in Chang et al. Nature 1978 275:615, Goeddel et al Nature 1979 281:544, Goeddel et al. Nucleic Acids Res. 1980 8:4057, European Application No. EP 36,776, U.S. Pat. No. 4,551,433, deBoer et al Proc. Natl. Acad. Sci. USA 1983 80:21-25, and Siebenlist et al Cell 1980 20:269.
[0175] Expression systems in yeast include those described in Hinnen et al Proc. Natl.
Acad. Sci. USA 1978 75:1929, Ito et al J. Bacteriol. 1983 153:163, Kurtz et al MoI Cell.
Biol. 1986 6:142, Kunze et al J. Basic Microbiol. 1985 25:141, Gleeson et al J. Gen.
Microbiol. 1986 132:3459, Roggenkamp et al MoI. Gen. Genet. 1986 202:302, Das et al. J.
Bacteriol. 1984 158:1165, De Louvencourt et al. J. Bacteriol. 1983 154:737, Van den Berg et al Bio/Technology 1990 8:135, Kunze et al J. Basic Microbiol 1985 25:141, Cregg et al
MoI. Cell. Biol. 1985 5:3376, U.S. Pat. Nos. 4,837,148 and 4,929,555, Beach et al Nature
1981 300:706, Davidow et α/. Curr. Genet. 1985 10:380, Gaillardin et al Curr. Genet. 1985
10:49, Ballance et al. Biochem. Biophys. Res. Commun. 1983 112:284-289, Tilburn et α/.
Gene 1983 26:205-221, Yelton et al Proc. Natl. Acad. Sci. USA 1984 81 :1470-1474, Kelly et al EMBO J. 1985 4:475479; European Application No. EP 244,234, and International
Publication No. WO 91/00357.
[0176] Expression of heterologous genes in insects can be accomplished as described in U.S. Pat. No. 4,745,051, European Application Nos. EP 127,839 and EP 155,476, Vlak et al J. Gen. Virol 1988 69:765-776, Miller et al Ann. Rev. Microbiol. 1988 42:177, Carbonell et al. Gene 1988 73:409, Maeda et al Nature 1985 315:592-594, Lebacq-Verheyden et al MoI. Cell. Biol. 1988 8:3129, Smith et al Proc. Natl. Acad. Sci. USA 1985 82:8404, Miyajima et al. Gene 1987 58:273, and Martin et al DNA 1988 7:99. Numerous baculoviral strains and variants and corresponding permissive insect host cells from hosts are described in Luckow et al Bio/Technology 1988 6:47-55, Miller et al GENETIC ENGINEERING, Setlow, J. K. et al eds., Vol. 8, Plenum Publishing, pp. 1986 277-279, and Maeda et al. Nature 1985 315:592- 594.
[0177] Mammalian expression can be accomplished as described in Dijkema et al EMBO J. 1985 4:761, Gorman et al Proc. Natl. Acad. Sci. USA 1982 79:6777, Boshart et al Cell 1985 41:521, and U.S. Pat. No. 4,399,216. Other features of mammalian expression can be facilitated as described in Ham et al. Meth. Enz. 1979 58:44, Barnes et al. Anal. Biochem. 1980 102:255, U.S. Pat. Nos. 4,767,704, 4,657,866, 4,927,762, 4,560,655 and Reissued U.S. Pat. No. RE 30,985, and in International Publication Nos. WO 90/103430, WO 87/00195. [0178] The production of recombinant adenoviral vectors are described in U.S. Pat. No. 6,485,958.
[0179] Botulinum toxin type A can be obtained by establishing and growing cultures of Clostridium botulinum in a fermenter and then harvesting and purifying the fermented mixture in accordance with known procedures.
[0180] Any of the above-described protein production methods can be used to provide the biologic that would benefit from the present invention.
PHARMACOLOGY AND UTILITY [0181] The compounds of the invention are selective inhibitors of cysteine proteases such as cathepsin S, K, B, and/or F, and in particular cathepsin S, and accordingly are useful for treating diseases in which cysteine protease activity contributes to the pathology and/or symptomatology of the disease. For example, the compounds of the invention are useful in treating autoimmune disorders, including, but not limited to, juvenile onset diabetes, psoriasis, multiple sclerosis, pemphigus vulgaris, Graves' disease, myasthenia gravis, systemic lupus erythemotasus, rheumatoid arthritis and Hashimoto's thyroiditis, allergic disorders, including, but not limited to, asthma, allogeneic immune responses, including, but not limited to, organ transplants or tissue grafts and endometriosis.
[0182] Cathepsin S is also implicated in disorders involving excessive elastolysis, such as chronic obstructive pulmonary disease (e.g., emphysema), bronchiolitis, excessive airway elastolysis in asthma and bronchitis, pneumonities and cardiovascular disease such as plaque rupture and atheroma. Cathepsin S is implicated in fibril formation and, therefore, inhibitors of cathepsins S are of use in treatment of systemic amyloidosis.
[0183] The cysteine protease inhibitory activities of the compounds of Formula (I) can be determined by methods known to those of ordinary skill in the art. Suitable in vitro assays for measuring protease activity and the inhibition thereof by test compounds are known. Typically, the assay measures protease-induced hydrolysis of a peptide-based substrate. Details of assays for measuring protease inhibitory activity are set forth in Biological Examples 1-5, infra. ADMINISTRATION AND PHARMACEUTICAL COMPOSITIONS [0184] In general, compounds of Formula (I) will be administered in therapeutically effective amounts via any of the usual and acceptable modes known in the art, either singly or in combination with one or more therapeutic agents. A therapeutically effective amount may vary widely depending on the severity of the disease, the age and relative health of the subject, the potency of the compound used and other factors. For example, therapeutically effective amounts of a compound of Formula (I) may range from about 10 micrograms per kilogram body weight (μg/kg) per day to about 100 milligram per kilogram body weight (mg/kg) per day, typically from about 100 μg/kg/day to about 10 mg/kg/day. Therefore, a therapeutically effective amount for an 80 kg human patient may range from about 1 mg/day to about 8 g/day, typically from about 1 mg/day to about 800 mg/day. In general, one of ordinary skill in the art, acting in reliance upon personal knowledge and the disclosure of this Application, will be able to ascertain a therapeutically effective amount of a compound of Formula (I) for treating a given disease.
[0185] The compounds of Formula (I) can be administered as pharmaceutical compositions by one of the following routes: oral, systemic (e.g., transdermal, intranasal or by suppository) or parenteral (e.g., intramuscular, intravenous or subcutaneous). Compositions can take the form of tablets, pills, capsules, semisolids, powders, sustained release formulations, solutions, suspensions, elixirs, aerosols, or any other appropriate composition and are comprised of, in general, a compound of Formula (I) in combination with at least one pharmaceutically acceptable excipient. Acceptable excipients are non-toxic, aid administration, and do not adversely affect the therapeutic benefit of the active ingredient. Such excipient may be any solid, liquid, semisolid or, in the case of an aerosol composition, gaseous excipient that is generally available to one of skill in the art.
[0186] Solid pharmaceutical excipients include starch, cellulose, talc, glucose, lactose, sucrose, gelatin, malt, rice, flour, chalk, silica gel, magnesium stearate, sodium stearate, glycerol monostearate, sodium chloride, dried skim milk, and the like. Liquid and semisolid excipients may be selected from water, ethanol, glycerol, propylene glycol and various oils, including those of petroleum, animal, vegetable or synthetic origin (e.g., peanut oil, soybean oil, mineral oil, sesame oil, and the like). Preferred liquid carriers, particularly for injectable solutions, include water, saline, aqueous dextrose and glycols. [0187] The amount of a compound of Formula (I) in the composition may vary widely depending upon the type of formulation, size of a unit dosage, kind of excipients and other factors known to those of skill in the art of pharmaceutical sciences. In general, a composition of a compound of Formula (I) for treating a given disease will comprise from 0.01 %w to 90%w, preferably 5%w to 50%w, of active ingredient with the remainder being the excipient or excipients. Preferably the pharmaceutical composition is administered in a single unit dosage form for continuous treatment or in a single unit dosage form ad libitum when relief of symptoms is specifically required. Representative pharmaceutical formulations containing a compound of Formula (I) are described below.
Examples
[0188] The present invention is further exemplified, but not limited by, the following examples that illustrate the preparation of compounds of Formula (I) (Examples) and intermediates (References) according to the invention.
Reference A
[0189] Synthesis of trifluoromethanesulfonic acid 2,2,2-trifluoro-l-(4-fluorophenyl)ethyl ester
Step 1
[0190] To a stirred solution of 2,2,2,4' -tetrafluoroacetophenone (10 g, 52.1 mmol) in methanol (50 niL) was added NaBH4 (0.98 g, 26.5 mmol) at 0° C. After stirring at 25° C for 2 h, the reaction mixture was quenched by adding IN HCl (100 mL) and then extracted with ethyl ether. The ether extract was washed with brine, dried with MgSO4, and concentrated to give 2,2,2-trifluoro-l-(4-fluorophenyl)ethanol (11.32 g) which was used in next step without further purification.
Step 2
[0191] NaH (640 mg, 16mmol, 60% in mineral oil) was washed twice with hexane (20 mL) and then suspended in dried diethyl ether (20 mL). A solution of 2,2,2-trifluoro-l-(4-fluoro- phenyl)ethanol (1.94 g, 10 mmol) in diethyl ether (10 mL) was added at O0C. After stirring for 2 h at room temperature, a solution of trifluoromethanesulfonyl chloride (1.68 g, 10 mmol) in diethyl ether (10 mL) was added. After 2 h, the reaction mixture was quenched by adding a solution OfNaHCO3 and the product was extracted with diethyl ether. The extracts were washed with brine and dried, and the solvent was removed to yield trifluoromethanesulfonic acid 2,2,2-trifluoro-l-(4-fiuorophenyl)ethyl ester (3.3 g).
Reference B [0192] Synthesis of 2,2,2-triftuoro-l (R) -(4-fluorophenyl)ethanol
[0193] To a -78 0C toluene (25 mL)/dichloromethane (25 mL) solution of 2,2,2,4'- tetrafluoroacetophenone (2.5 g, 13.01 mmol) and IM S-methyl CBS oxazaborolidine catalyst (1.3 mL, 1.3 mmol) was added freshly distilled catecholborane (1.66 mL, 15.62 mmol). The reaction mixture was maintained at -78 0C for 16 h at which time 4N HCl (5 mL in dioxane) was added and the reaction mixture was allowed to warm to room temperature. The reaction mixture was diluted with ethyl acetate and washed with a saturated brine solution. The organic layer was dried over magnesium sulfate, filtered and concentrated to provide a solid. The solid was suspended in hexanes and filtered off. The hexanes filtrate containing the desired product was concentrated and the residue subjected to flash chromatography (10 hexanes: 1 ethylacetate) to provide the title compound as colorless oil (2.2g, 87% yield). The ratio of enantiomers was determined to be 95:5 by chiral HPLC (Chiralcel OD column, 95 hexanes: 5 isopropanol mobile phase. Ret. time major product 6.757 min. Ret. time minor isomer 8.274 min.).
Reference D
[0194] Synthesis of 2(i?)-[2,2,2-trifluoro-l (S)-(4-fluorophenyl)ethylamino]-3- tritylsulfanylpropionic acid
[0195] To a slurry of S-trityl-L-cysteine (4.86 g, 13.37 mmol) in dichloromethane (97 mL, 20 niL/g AA) at room temperature was added diisopropylethylamine (9.32 mL, 53.48 mmol) followed by a solution of trifluoromethanesulfonic acid 2,2,2-trifluoro- 1 (i?5)-phenylethyl ester (5.32 g, 16.04 mmol) (major enantiomer (S), 90 ee) in dichloromethane (15 mL) via syringe all at once. After 19 h, the reaction mixture was concentrated on the rotovap to give an oil. Diethyl ether was added and the solution was washed with IN HCl and brine. The organic layer was dried over MgSO4, filtered, and concentrated. Flash chromatography of the residue with 2 hexanes/1 ethyl acetate/.25% acetic acid as the eluent provided 2(i?)-[2,2,2- trifluoro-l(i?S)-(4-fluorophenyl)ethylamino]-3-tritylsulfanyl-propionic acid (6 g) (major diastereomer (R,S), 90 de) as an oil/foam.
Reference E [0196] Synthesis of 2-(l-aminocyclopropyl)-iV-cyclopropyl-2-hydroxyacetamide
Stepl [0197] 1-Aminocyclopropanecarbonitrile chlorohydrate (6.1 g, 51.4 mmol) was refluxed in 6N hydrochloric acid (500 mL) for 7 h and then concentrated to yield 1-aminocyclopropane- carboxylic acid chlorohydrate as an off-white solid which was used in the next step without further purification:
Step 2 [0198] A solution of 1-aminocyclopropanecarboxylic acid chlorohydrate (3.6 g, 26.2 mmol) in MeOH (100 mL), containing potassium carbonate (4.0 g, 28.94 mmol) was stirred at room temperature for 48 h. After filtration, MeOH was removed under reduced pressure to yield 1-aminocyclopropanecarboxylic acid (2.64 g) which was used in the next step without further purification.
Step 3
[0199] 1-Aminocyclopropanecarboxylic acid (2.64 g, 26.1 mmol ) and tetramethylammonium hydroxide (2.38 g, 26.1 mmol) was added to acetonitrile (150 mL). The reaction mixture became homogeneous after stirring at room temperature for about an hour. Boc2O (8.54 g, 39.2 mmol) was then added and stirring was continued for 2 days. On the 3rd day, another portion of BoC2O (2.85 g, 13.1 mmol) was added and the reaction mixture stirred an additional day. Acetonitrile was removed under reduced pressure and the residue was partitioned between H2O and Et2O. The aqueous layer was washed with Et2O and then acidified with solid citric acid to pH ~3. The aqueous solution was extracted with EtOAc. The combined EtOAc extracts were washed with brine, dried (Na2SO4), and the EtOAc was removed under reduced pressure to give 1-tert-butoxycarbonylaminocyclopropanecarboxylic acid as a white solid (2.32 g) which was used in the next step without further purification.
Step 4
[0200] To a solution of 1-fe/Y-butoxycarbonylaminocyclopropanecarboxylic acid (2.32 g, 11.5 mmol) in CH2Cl2 (25 mL) at 0 0C was added N,O-dimethylhydroxylamine hydrochloride (1.24 g, 12,7 mmol), triethylamine (2.57 g, 3.54 mL, 25.4 mmol), and HATU (4.82 g, 12.7 mmol). The reaction mixture was stirred at room temperature for 4 h. The reaction mixture was concentrated under reduced pressure and then partitioned between Et2O and water. The water layer was extracted with Et2O. The combined organic layer was washed with brine, dried (Na2SO4), and concentrated under reduced pressure to yield [l-(methoxy-methyl- carbamoyl)-cyclopropyl]carbamic acid tert-butyl ester which was used in the next step without further purification.
Step 5
[0201] To a 0.05 M solution of [l-(methoxy-methyl-carbamoyl)cyclopropyl]carbamic acid tert-butyl ester in Et2O (80 mL, 4.0 mmol) at room temperature was added dropwise lithium aluminum hydride (1.0 M in Et2O, 5 mL, 5.0 mmol). The reaction mixture was stirred for another 20 min and then quenched with 6 mL of a solution of KHSO4 in water. The layers were separated and the aqueous layer was extracted with Et2O. The combined organic layers were washed with 1 N HCl, saturated NaHCO3, and brine, dried (Na2SO4), and concentrated to yield (l-formylcyclopropyl)carbamic acid tert-butyl ester as a colorless oil (393 mg) which was used immediately in the next step without further purification.
Step 6
[0202] To a solution of (l-formylcyclopropyl)carbamic acid tert-butyl ester (393 mg, 2.12 mmol) in CH2Cl2 (4 niL) was added acetic acid (191 mg, 0.182 mL, 3.18 mmol), and cyclopropyl isocyanide (142 mg, 2.12 mmol). The reaction mixture was stirred overnight at room temperature and then concentrated under reduced pressure to yield crude acetic acid (1- tert-butoxycarbonylammocyclopropyi)cyclopropylcarbamoyl methyl ester which was used in the next step without further purification. Step 7
[0203] To a solution of the acetic acid (l-tert-butoxycarbonylaminocyclopropyl)- cyclopropyl-carbamoyl-methyl ester in MeOH (5 mL) was added 10% NaOH (1 mL). The reaction mixture was stirred at room temperature for 2 h and then acidified with 2.5N HCl to pH 7. The solution was extracted with EtOAc, washed with brine, dried (Na2SO4), and concentrated under reduced pressure to yield [1-
(cyclopropylcarbamoylhydroxymethyl)cyclopropylj-carbamic acid tert-butyl ester as a yellow oil which was used in the next step without further purification.
Step 8
[0204] A solution of [1 -(cyclopropylcarbamoylhydroxymethyl)cyclopropyl]carbamic acid tert-butyl ester in CH2Cl2 (5 mL) and TFA (5 mL) was stirred at room temperature for 2.5 h. The reaction mixture was concentrated and chased with toluene to yield 2-(l- aminocyclopropyl)-N-cyclopropyl-2-hydroxyacetamide.
Reference F [0205] Synthesis of 3-amino-N-cyclopropyl-2-hydroxy-3-methylbutyramide
Step l [0206] To a solution of (2-2ydroxy-l,l-dimethylethyl)-carbamic acid tert-hutyl ester (284 mg, 1.5 mmol) in CH2Cl2 (5 mL) was added at 0 0C Dess-Martin periodane (763 mg, 1.8 mmol). After 1.5 h, a solution of 0.26M Na2S2O3 in saturated NaHCO3 (6 mL) was added and the resulting mixture was stirred for 15 min. The layers were separated and the aqueous layer was extracted with CH2Cl2. The combined organic layers were dried (Na2SO4) and concentrated to yield (l,l-dimethyl-2-oxo-ethyl)carbamic acid tert-buty\ ester as a white solid which was used in the next step without further purification.
Step 2
[0207] To a solution of (l,l-dimethyl-2-oxo-ethyl)-carbamic acid fert-butyl ester in CH2Cl2 was added acetic acid (180 mg, 0.172 mL, 3.0 mmol) and cyclopropyl isocyanide (101 mg, 1.5 mmol). The reaction mixture was stirred overnight at room temperature and then concentrated to yield crude acetic acid 2-før^butoxycarbonylamino-l-cyclopropylcarbamoyl- 2-methylpropyl ester which was used in the next step without further purification.
Step 3 [0208] To a solution of acetic acid 2-fert-butoxycarbonylamino- 1 -cyclopropylcarbamoyl-2- methylpropyl ester in MeOH (10 mL) was added 10% NaOH (1.5 mL). The reaction mixture was stirred at room temperature for 3 h and then acidified with IN Hydrochloric acid to pH 7. The reaction mixture was extracted with EtOAc. The organic layer was dried (Na2SO4) concentrated to yield (2-cyclopropylcarbamoyl-2-hydroxy-l,l-dimethylethyl)-carbamic acid tert-hutyl ester which was used in the next step without further purification.
Step 4
[0209] A solution of (2-cyclopropylcarbamoyl-2-hydroxy- 1 , 1 -dimethylethyl)-carbamic acid tert-butyl ester in CH2Cl2 (10 mL) and TFA (5 mL) was stirred at room temperature for 4 h. The reaction mixture was then concentrated and chased with toluene to yield 3-amino-N- cyclopropyl -2-hydroxy-3 -methylbutyramide.
Reference G [0210] Synthesis of 3 -amino-JV-benzyl-2-hydroxy-3 -methylbutyramide
[0211] 3-Amino-iV-benzyl-2-hydroxy-3-methylbutyramide was made by the procedure described for 3-amino-N-cyclopropyl-2-hydroxy-3-methylbutyramide by substituting cyclopropyl isocyanide with benzyl isocyanide.
Biological Examples
Example 1 Cathepsin B Assay [0212] Solutions of test compounds in varying concentrations were prepared in 10 μL of dimethyl sulfoxide (DMSO) and then diluted into assay buffer (40 μL, comprising: N,N- bis(2-hydroxyethyl)-2-aminoethanesulfonic acid (BES), 50 mM (pH 6); polyoxyethylenesorbitan monolaurate, 0.05%; and dithiothreitol (DTT), 2.5 mM). Human cathepsin B (0.025 pMoles in 25 μL of assay buffer) was added to the dilutions. The assay solutions were mixed for 5-10 seconds on a shaker plate, covered and incubated for 30 min at room temperature. Z-FR-AMC (20 nMoles in 25 μL of assay buffer) was added to the assay solutions and hydrolysis was followed spectrophotometrically at (λ 460 nm) for 5 min. Apparent inhibition constants (K1) were calculated from the enzyme progress curves using standard mathematical models.
[0213] Compounds of the invention were tested by the above-described assay and observed to exhibit cathepsin B inhibitory activity.
Example 2 Cathepsin K Assay [0214] Solutions of test compounds in varying concentrations were prepared in 10 μL of dimethyl sulfoxide (DMSO) and then diluted into assay buffer (40 μL, comprising: MES5 50 mM (pH 5.5); EDTA, 2.5 mM; and DTT, 2.5 mM). Human cathepsin K (0.0906 pMoles in 25 μL of assay buffer) was added to the dilutions. The assay solutions were mixed for 5-10 seconds on a shaker plate, covered and incubated for 30 min at room temperature. Z-Phe- Arg-AMC (4 nMoles in 25 μL of assay buffer) was added to the assay solutions and hydrolysis was followed spectrophotometrically at (λ 460 nm) for 5 min. Apparent inhibition constants (Kj) were calculated from the enzyme progress curves using standard mathematical models.
[0215] Compounds of the invention were tested by the above-described assay and observed to exhibit cathepsin K inhibitory activity.
Example 3 Cathepsin L Assay
[0216] Solutions of test compounds in varying concentrations were prepared in 10 μL of dimethyl sulfoxide (DMSO) and then diluted into assay buffer (40 μL, comprising: MES, 50 niM (pH 5.5); EDTA, 2.5 niM; and DTT, 2.5 mM). Human cathepsin L (0.05 pMoles in 25 μL of assay buffer) was added to the dilutions. The assay solutions were mixed for 5-10 seconds on a shaker plate, covered and incubated for 30 min at room temperature. Z-Phe- Arg-AMC (1 nMoles in 25 μL of assay buffer) was added to the assay solutions and hydrolysis was followed spectrophotometrically at (λ 460 nm) for 5 min. Apparent inhibition constants (K;) were calculated from the enzyme progress curves using standard mathematical models.
[0217] Compounds of the invention were tested by the above-described assay and observed to exhibit cathepsin L inhibitory activity.
Example 4
Cathepsin S Assay
[0218] Solutions of test compounds in varying concentrations were prepared in 10 μL of dimethyl sulfoxide (DMSO) and then diluted into assay buffer (40 μL, comprising: MES, 50 mM (pH 6.5); EDTA, 2.5 mM; and NaCl, 100 mM); β-mercaptoethanol, 2.5 mM; and BSA, 0.00%. Human cathepsin S (0.05 pMoles in 25 μL of assay buffer) was added to the dilutions. The assay solutions were mixed for 5-10 seconds on a shaker plate, covered and incubated for 30 min at room temperature. Z-Val-Val-Arg-AMC (4 nMoles in 25 μL of assay buffer containing 10% DMSO) was added to the assay solutions and hydrolysis was followed spectrophotometrically (at λ 460 nm) for 5 min. Apparent inhibition constants (Kj) were calculated from the enzyme progress curves using standard mathematical models. [0219] Compounds of the invention were tested by the above-described assay and observed to exhibit cathepsin S inhibitory activity of < or = 100 nm.
Example 5 Cathepsin F Assay
[0220] Solutions of test compounds in varying concentrations were prepared in 10 μL of dimethyl sulfoxide (DMSO) and then diluted into assay buffer (40 μL, comprising: MES, 50 mM (pH 6.5); EDTA, 2.5 mM; and NaCl, 100 raM); DTT, 2.5 mM; and BSA, 0.01%. Human cathepsin F (0.1 pMoles in 25 μL of assay buffer) was added to the dilutions. The assay solutions were mixed for 5-10 seconds on a shaker plate, covered and incubated for 30 min at room temperature. Z-Phe-Arg-AMC (2 nMoles in 25 μL of assay buffer containing 10% DMSO) was added to the assay solutions and hydrolysis was followed spectrophotometrically (at λ 460 nm) for 5 min. Apparent inhibition constants (Kj) were calculated from the enzyme progress curves using standard mathematical models.
[0221] Compounds of the invention were tested by the above-described assay and observed to exhibit cathepsin F inhibitory activity.
Example 1 [0222] Representative pharmaceutical formulations Containing a Compound of Formula (I)
ORAL FORMULATION
Compound of Formula (I) 10- 100 mg
Citric Acid Monohydrate 105 mg
Sodium Hydroxide 18 mg Flavoring
Water q.s. to lOO mL
INTRAVENOUS FORMULATION Compound of Formula (I) 0.1-10 mg Dextrose Monohydrate q.s. to make isotonic
Citric Acid Monohydrate 1.05 mg Sodium Hydroxide 0.18 mg
Water for Injection q.s. to 1.0 niL
TABLET FORMULATION Compound of Formula (I) 1%
Microcrystalline Cellulose 73%
Stearic Acid 25%
Colloidal Silica 1%
[0223] The foregoing invention has been described in some detail by way of illustration and example, for purposes of clarity and understanding. It will be obvious to one of skill in the art that changes and modifications may be practiced within the scope of the appended claims. Therefore, it is to be understood that the above description is intended to be illustrative and not restrictive. The scope of the invention should, therefore, be determined not with reference to the above description, but should instead be determined with reference to the following appended claims, along with the full scope of equivalents to which such claims are entitled.

Claims

WE CLAIM:
1. A compound of Formula (I):
where: R1 is hydrogen or alkyl; R2 is cycloalkyl, cycloalkylalkyl, aralkyl, heteroaryl, or heteroaralkyl optionally substituted with one or two substitutents independently selected from alkyl, alkoxy, or halo; R3 is hydrogen, alkyl or alkoxyalkyl; R4 is alkyl; or R and R together with the carbon atom to which they are attached form cycloalkylene optionally substituted with one to four fluoro or heterocycloalkylene optionally substituted with alkyl, alkoxyalkyl, hydroxyalkyl, acyl, cycloalkyl, cycloalkylalkyl, or haloalkyl; R5 is -alkylene-SO2NRnR12 where Rn is hydrogen, alkyl, haloalkyl, alkoxyalkyl, hydroxyalkyl, aminoalkyl, carboxyalkyl, alkoxycarbonylalkyl, acyl, cycloalkyl, cycloalkylalkyl, aryl, heteroaryl, aralkyl, heteroaralkyl, heterocycloalkylalkyl, acylalkyl, or heterocycloalkylaminocarbonyl and R12 is hydrogen, alkyl, hydroxyalkyl, alkoxyalkyl, aryl, heteroaryl, aralkyl, heteroaralkyl, cycloalkyl, cycloalkylalkyl, heterocycloalkyl, or heterocycloalkylalkyl); or R11 and R12 together with the nitrogen atom to which they are attached form heterocycloamino or bridged azabicyclic ring, wherein the aromatic or alicyclic ring in R5 is optionally substituted with one, two, or three Ra independently selected from alkyl, haloalkyl, alkoxy, hydroxy, haloalkoxy, cyano, halo, carboxy or alkoxycarbonyl; or optionally substituted with one or two Rb independently selected from hydrogen, alkyl, haloalkyl, alkoxy, hydroxy, haloalkoxy, halo, carboxy, or alkoxycarbonyl and one Rc selected from cyano, hydroxyalkyl, alkoxyalkyl, aminoalkyl, aryl, heteroaryl, aralkyl, heteroaralkyl, cycloalkyl, cycloalkylalkyl, heterocycloalkyl, heterocycloalkylalkyl, acyl, acylalkyl, alkoxycarbonyl, aryloxycarbonyl, aralkyloxycarbonyl, heteroaryloxycarbonyl, heteroaralkyloxycarbonyl, heterocycloalkyloxycarbonyl, heterocycloalkylalkyloxycarbonyl, cycloalkyloxycarbonyl, cycloalkylalkyloxycarbonyl, aryloxy, heteroaryloxy, aralkyloxy, heteroaralkyloxy, aminocarbonyl, aminosulfonyl, or -SO2R13 (where R13 is alkyl, cycloalkyl, aryl, heteroaryl, or heterocycloalkyl); and further wherein the aromatic or alicyclic ring in Rc is optionally substituted with one, two, or three Rd independently selected from alkyl, haloalkyl, alkoxy, hydroxy, haloalkoxy, halo, cyano, -CONH2, alkylaminocarbonyl, dialkylaminocarbonyl, alkylcarbonyl, alkylsulfonyl, or alkylsulfonylamino; R6 is haloalkyl; R7 is hydrogen, alkyl, or haloalkyl; and R8 is hydrogen, alkyl, haloalkyl, cycloalkyl, aryl, heteroaryl, heterocycloalkyl attached via a carbon atom wherein the aromatic or alicyclic ring in R8 is optionally substituted with one, two, or three Re independently selected from alkyl, halo, haloalkyl, hydroxy, alkoxy, haloalkoxy, alkoxycarbonyl, carboxy, cyano, alkylcarbonyl, alkylsulfonyl, alkylsulfonylamino, alkylaminocarbonyl, dialkylaminocarbonyl, or aminosulfonyl; or a pharmaceutically acceptable salts thereof.
2. The compound of Claim 1 wherein R1 is hydrogen and R is cyclopropyl, 1- phenylethyl, or lH-pyrazol-5-yl.
3. The compound of Claim 1 wherein R is hydrogen and R is cyclopropyl.
4. The compound ofClaim 2 or 3 wherein R3 is hydrogen and R4 is alkyl.
5. The compound of Claim 2 or 3 wherein R3 is hydrogen and R4 is methyl, ethyl, propyl or butyl.
6. The compound of Claim 2 or 3 wherein R3 is hydrogen and R4 ethyl or rø-propyl.
7. The compound of Claim 2 or 3 wherein R3 and R are alkyl.
8. The compound of Claim 2 or 3 wherein R3 and R are independently methyl or ethyl.
9. The compound of Claim 2 or 3 wherein R3 and R4 are methyl.
10. The compound of Claim 2 or 3 wherein R3 and R4 together with the carbon atom to which they are attached form cycloalkylene.
11. The compound of Claim 2 or 3 wherein R3 and R4 together with the carbon atom to which they are attached form cyclopropylene.
12. The compound of any of the Claims 2-11 wherein R6 is haloalkyl and R7 and R8 are hydrogen.
13. The compound of any of the Claims 2-11 wherein R6 is 2,2,2-trifluoroethyl or 2,2,3,3,3-pentafluoropropyl and R7 and R8 are hydrogen.
14. The compound of any of the Claims 2-11 wherein R6 is haloalkyl, R7 is haloalkyl, and R8 is hydrogen.
15. The compound of any of the Claims 2-11 wherein R6 is haloalkyl, R7 is alkyl, and R8 is hydrogen.
16. The compound of any of the Claims 2-11 wherein R6 is haloalkyl, R7 is hydrogen, and R8 is aryl optionally substituted with one, two, or three Re.
17. The compound of any of the Claims 2-11 wherein R6 is trifluoromethyl or difluoromethyl, R7 is hydrogen, and R8 is 4-fluorophenyl, 2,3-, 2,4-, 2,5-, 2,6-, 3,4, or 3,5- difluorophenyl.
18. The compound of any of the Claims 2-11 wherein R6 is haloalkyl, R7 is hydrogen, and R8 and R8 is heteroaryl optionally substituted with one, two, or three Re.
19. The compound of any of the Claims 2-18 wherein R5 is -alkylene-SC^NR11R12 where: R11 is hydrogen, alkyl, haloalkyl, alkoxyalkyl, hydroxyalkyl, aminoalkyl, carboxyalkyl, alkoxycarbonylalkyl, acyl, cycloalkyl, cycloalkylalkyl, aryl, heteroaryl, aralkyl, heteroaralkyl, heterocycloalkylalkyl, acylalkyl, or heterocycloalkylaminocarbonyl; and R12 is hydrogen, alkyl, aryl, heteroaryl, aralkyl, heteroaralkyl, cycloalkyl, cycloalkylalkyl, heterocycloalkyl, or heterocycloalkylalkyl; wherein the aromatic or alicyclic ring are optionally substituted with one, two, or three Ra independently selected from alkyl, haloalkyl, alkoxy, hydroxy, haloalkoxy, cyano, halo, carboxy or alkoxycarbonyl; or optionally substituted with one or two Rb independently selected from hydrogen, alkyl, haloalkyl, alkoxy, hydroxy, haloalkoxy, halo, carboxy, or alkoxycarbonyl and one Re selected from cyano, hydroxyalkyl, alkoxyalkyl, aminoalkyl, aryl, heteroaryl, aralkyl, heteroaralkyl, cycloalkyl, cycloalkylalkyl, heterocycloalkyl, heterocycloalkylalkyl, acyl, acylalkyl, alkoxycarbonyl, aryloxycarbonyl, aralkyloxycarbonyl, heteroaryloxycarbonyl, heteroaralkyloxycarbonyl, heterocycloalkyloxycarbonyl, heterocycloalkylalkyloxycarbonyl, cycloalkyloxycarbonyl, cycloalkylalkyloxycarbonyl, aryloxy, heteroaryloxy, aralkyloxy, heteroaralkyloxy, aminocarbonyl, aminosulfonyl, or - SO2R13 (where R13 is alkyl, cycloalkyl, aryl, heteroaryl, or heterocycloalkyl); and further wherein the aromatic or alicyclic ring in R0 is optionally substituted with one, two, or three Rd independently selected from alkyl, haloalkyl, alkoxy, hydroxy, haloalkoxy, halo, cyano, - CONH2, alkylaminocarbonyl, dialkylaminocarbonyl, alkylcarbonyl, alkylsulfonyl, or alkylsulfonylamino.
20. The compound of any of the Claims 2-18 wherein R5 is -alkylene-SO^R11R12 where here R11 and R12 together with the nitrogen atom to which they are attached form heterocycloamino substituted with one, two, or three Ra independently selected from alkyl, haloalkyl, alkoxy, hydroxy, haloalkoxy, cyano, halo, carboxy or alkoxycarbonyl; or optionally substituted with one or two Rb independently selected from hydrogen, alkyl, haloalkyl, alkoxy, hydroxy, haloalkoxy, halo, carboxy, or alkoxycarbonyl and one Rc selected from cyano, hydroxyalkyl, alkoxyalkyl, aminoalkyl, aryl, heteroaryl, aralkyl, heteroaralkyl, cycloalkyl, cycloalkylalkyl, heterocycloalkyl, heterocycloalkylalkyl, acyl, acylalkyl, alkoxycarbonyl, aryloxycarbonyl, aralkyloxycarbonyl, heteroaryloxycarbonyl, heteroaralkyloxycarbonyl, heterocycloalkyloxycarbonyl, heterocycloalkylalkyloxycarbonyl, cycloalkyloxycarbonyl, cycloalkylalkyloxycarbonyl, aryloxy, heteroaryloxy, aralkyloxy, heteroaralkyloxy, aminocarbonyl, aminosulfonyl, or -SO2R13 (where R13 is alkyl, cycloalkyl, aryl, heteroaryl, or heterocycloalkyl); and further wherein the aromatic or alicyclic ring in Rc is optionally substituted with one, two, or three Rd independently selected from alkyl, haloalkyl, alkoxy, hydroxy, haloalkoxy, halo, cyano, -CONH2, alkylaminocarbonyl, dialkylaminocarbonyl, alkylcarbonyl, alkylsulfonyl, or alkylsulfonylamino.
21. The compound of any of the Claims 2-18 wherein R5 is -alkylene-SO^R11R12 where here R1 ' and R12 together with the nitrogen atom to which they are attached form piperazin-4- yl or piperidin-1-yl substituted at the 4-position with one, two, or three Ra independently selected from alkyl, haloalkyl, alkoxy, hydroxy, haloalkoxy, cyano, halo, carboxy or alkoxycarbonyl; or optionally substituted with one or two Rb independently selected from hydrogen, alkyl, haloalkyl, alkoxy, hydroxy, haloalkoxy, halo, carboxy, or alkoxycarbonyl and one Rc selected from cyano, hydroxyalkyl, alkoxyalkyl, aminoalkyl, aryl, heteroaryl, aralkyl, heteroaralkyl, cycloalkyl, cycloalkylalkyl, heterocycloalkjd, heterocycloalkylalkyl, acyl, acylalkyl, alkoxycarbonyl, aryloxycarbonyl, aralkyloxycarbonyl, heteroaryloxycarbonyl, heteroaralkyloxycarbonyl, heterocycloalkyloxycarbonyl, heterocycloalkylalkyloxycarbonyl, cycloalkyloxycarbonyl, cycloalkylalkyloxycarbonyl, aryloxy, heteroaryloxy, aralkyloxy, heteroaralkyloxy, aminocarbonyl, aminosulfonyl, or - SO2R13 (where R13 is alkyl, cycloalkyl, aryl, heteroaryl, or heterocycloalkyl); and further wherein the aromatic or alicyclic ring in Rc is optionally substituted with one, two, or three Rd independently selected from alkyl, haloalkyl, alkoxy, hydroxy, haloalkoxy, halo, cyano, - CONH2, alkylaminocarbonyl, dialkylaminocarbonyl, alkylcarbonyl, alkylsulfonyl, or alkylsulfonylamino.
22. A pharmaceutical composition comprising a compound of any of the Claims 1-21 in admixture with one or more suitable excipients.
23. A method for treating a disease in an animal mediated by Cathepsin S which method comprises administering to the animal a pharmaceutical composition comprising a compound of any of the Claims 1-21 in admixture with one or more suitable excipients.
24. The method of Claim 22 wherein the disease is rheumatoid arthritis, multiple sclerosis, myasthenia gravis, psoriasis, pemphigus vulgaris, Graves' disease, myasthenia gravis, systemic lupus erythemotasus, asthma, pain, and atherosclerosis.
25. A method of treating a patient undergoing a therapy wherein the therapy causes an immune response in the patient comprising administering to the patient a compound of any of Claims 1-21.
26. The compound of Claim 1 wherein: R3 is alkyl and R4 is alkyl.
27. The compound of Claim 1 wherein: R3 and R4 together with the carbon atom to which they are attached form cycloalkylene.
28. The compound of Claim 1 wherein: R3 and R4 together with the carbon atom to which they are attached form pipeiϊdin-4- yl substituted at the nitrogen atom with ethyl, 2,2,2-trifluoroethyl or cyclopropyl, tetrahydropyran-4-yl, tetrahydrothiopyran-4-yl, or l,l-dioxotetraliydrothiopyran-4-yl.
29. The compound of Claim 1 wherein: R6 is haloalkyl and R7 and R8 are hydrogen.
30. The compound of Claim 1 wherein: R6 is haloalkyl, R7 is haloalkyl, and Rs are hydrogen.
31. The compound of Claim 1 wherein: R6 is haloalkyl, R7 is alkyl, and R8 are hydrogen.
EP06739282A 2005-03-21 2006-03-21 Alpha ketoamide compounds as cysteine protease inhibitors Withdrawn EP1866276A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US66404105P 2005-03-21 2005-03-21
PCT/US2006/010426 WO2006102423A1 (en) 2005-03-21 2006-03-21 Alpha ketoamide compounds as cysteine protease inhibitors

Publications (1)

Publication Number Publication Date
EP1866276A1 true EP1866276A1 (en) 2007-12-19

Family

ID=37024161

Family Applications (1)

Application Number Title Priority Date Filing Date
EP06739282A Withdrawn EP1866276A1 (en) 2005-03-21 2006-03-21 Alpha ketoamide compounds as cysteine protease inhibitors

Country Status (4)

Country Link
EP (1) EP1866276A1 (en)
JP (1) JP2008533205A (en)
CA (1) CA2603305A1 (en)
WO (1) WO2006102423A1 (en)

Families Citing this family (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CA2743749A1 (en) * 2008-11-13 2010-05-20 Virobay, Inc. Haloalkyl containing compounds as cysteine protease inhibitors
JP5666468B2 (en) * 2008-12-19 2015-02-12 メディヴィル・ユーケイ・リミテッド Cysteine protease inhibitor
CN102858751A (en) * 2009-12-10 2013-01-02 美迪维尔英国有限公司 Cysteine protease inhibitors
AU2011261375B2 (en) 2010-06-04 2016-09-22 Albany Molecular Research, Inc. Glycine transporter-1 inhibitors, methods of making them, and uses thereof
KR20130087504A (en) * 2010-06-16 2013-08-06 메디버 유케이 리미티드 New cathepsin s protease inhibitors, useful in the treatment of e.g. autoimmune disorders, allergy and chronic pain conditions
WO2012172473A1 (en) * 2011-06-13 2012-12-20 Medivir Uk Ltd Cysteine protease inhibitors
EP3946332A1 (en) 2019-04-05 2022-02-09 Université de Bretagne Occidentale Protease-activated receptor-2 inhibitors for the treatment of sensory neuropathy induced by a marine neurotoxic poisoning

Family Cites Families (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6150378A (en) * 1997-10-07 2000-11-21 Cephalon, Inc. Peptidyl-containing α-ketoamide cysteine and serine protease inhibitors

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
See references of WO2006102423A1 *

Also Published As

Publication number Publication date
CA2603305A1 (en) 2006-09-28
WO2006102423A1 (en) 2006-09-28
JP2008533205A (en) 2008-08-21

Similar Documents

Publication Publication Date Title
EP1663958B1 (en) Haloalkyl containing compounds as cysteine protease inhibitors
EP2511257B1 (en) Composition comprising an alpha ketoamide compound as cysteine protease inhibitor
WO2006102423A1 (en) Alpha ketoamide compounds as cysteine protease inhibitors
US8748649B2 (en) Di-fluoro containing compounds as cysteine protease inhibitors
CA2602112A1 (en) Sulfonyl containing compounds as cysteine protease inhibitors
EP2079683B1 (en) Di-fluoro containing compounds as cysteine protease inhibitors
US8163735B2 (en) Sulfonamide compounds as cysteine protease inhibitors
WO2009123623A1 (en) Di-fluoro containing compounds as cysteine protease inhibitors

Legal Events

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

Free format text: ORIGINAL CODE: 0009012

17P Request for examination filed

Effective date: 20071018

AK Designated contracting states

Kind code of ref document: A1

Designated state(s): AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HU IE IS IT LI LT LU LV MC NL PL PT RO SE SI SK TR

AX Request for extension of the european patent

Extension state: AL BA HR MK YU

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

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

18D Application deemed to be withdrawn

Effective date: 20081001