WO2001034594A1 - Dipeptidyl peptidase iv inhibitors and methods of making and using dipeptidyl peptidase iv inhibitors - Google Patents

Dipeptidyl peptidase iv inhibitors and methods of making and using dipeptidyl peptidase iv inhibitors Download PDF

Info

Publication number
WO2001034594A1
WO2001034594A1 PCT/US2000/030836 US0030836W WO0134594A1 WO 2001034594 A1 WO2001034594 A1 WO 2001034594A1 US 0030836 W US0030836 W US 0030836W WO 0134594 A1 WO0134594 A1 WO 0134594A1
Authority
WO
WIPO (PCT)
Prior art keywords
amino
branched
inhibitor
cycloalkyl
group
Prior art date
Application number
PCT/US2000/030836
Other languages
French (fr)
Inventor
Paul Jackson
Joseph Steiner
Original Assignee
Guilford Pharmaceuticals, Inc.
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 Guilford Pharmaceuticals, Inc. filed Critical Guilford Pharmaceuticals, Inc.
Priority to AU19164/01A priority Critical patent/AU1916401A/en
Priority to JP2001536541A priority patent/JP2003535034A/en
Priority to CA002390231A priority patent/CA2390231A1/en
Priority to EP00982093A priority patent/EP1228061A4/en
Publication of WO2001034594A1 publication Critical patent/WO2001034594A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D257/00Heterocyclic compounds containing rings having four nitrogen atoms as the only ring hetero atoms
    • C07D257/02Heterocyclic compounds containing rings having four nitrogen atoms as the only ring hetero atoms not condensed with other rings
    • C07D257/04Five-membered rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • A61P13/08Drugs for disorders of the urinary system of the prostate
    • 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
    • A61P25/06Antimigraine agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/14Drugs for disorders of the nervous system for treating abnormal movements, e.g. chorea, dyskinesia
    • A61P25/16Anti-Parkinson drugs
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • 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
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C229/00Compounds containing amino and carboxyl groups bound to the same carbon skeleton
    • C07C229/46Compounds containing amino and carboxyl groups bound to the same carbon skeleton having amino or carboxyl groups bound to carbon atoms of rings other than six-membered aromatic rings of the same carbon skeleton
    • 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
    • C07C237/04Carboxylic 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 the carbon skeleton being acyclic and saturated
    • C07C237/12Carboxylic 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 the carbon skeleton being acyclic and saturated having the nitrogen atom of at least one of the carboxamide groups bound to an acyclic carbon atom of a hydrocarbon radical substituted by carboxyl groups
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C255/00Carboxylic acid nitriles
    • C07C255/45Carboxylic acid nitriles having cyano groups bound to carbon atoms of rings other than six-membered aromatic rings
    • C07C255/46Carboxylic acid nitriles having cyano groups bound to carbon atoms of rings other than six-membered aromatic rings to carbon atoms of non-condensed rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C327/00Thiocarboxylic acids
    • C07C327/38Amides of thiocarboxylic acids
    • C07C327/40Amides of thiocarboxylic acids having carbon atoms of thiocarboxamide groups bound to hydrogen atoms or to acyclic carbon atoms
    • C07C327/42Amides of thiocarboxylic acids having carbon atoms of thiocarboxamide groups bound to hydrogen atoms or to acyclic carbon atoms to hydrogen atoms or to carbon atoms of a saturated carbon skeleton
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D207/00Heterocyclic compounds containing five-membered rings not condensed with other rings, with one nitrogen atom as the only ring hetero atom
    • C07D207/02Heterocyclic compounds containing five-membered rings not condensed with other rings, with one nitrogen atom as the only ring hetero atom with only hydrogen or carbon atoms directly attached to the ring nitrogen atom
    • C07D207/04Heterocyclic compounds containing five-membered rings not condensed with other rings, with one nitrogen atom as the only ring hetero atom with only hydrogen or carbon atoms directly attached to the ring nitrogen atom having no double bonds between ring members or between ring members and non-ring members
    • C07D207/10Heterocyclic compounds containing five-membered rings not condensed with other rings, with one nitrogen atom as the only ring hetero atom with only hydrogen or carbon atoms directly attached to the ring nitrogen atom having no double bonds between ring members or between ring members and non-ring members with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
    • C07D207/16Carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D233/00Heterocyclic compounds containing 1,3-diazole or hydrogenated 1,3-diazole rings, not condensed with other rings
    • C07D233/02Heterocyclic compounds containing 1,3-diazole or hydrogenated 1,3-diazole rings, not condensed with other rings having no double bonds between ring members or between ring members and non-ring members
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D241/00Heterocyclic compounds containing 1,4-diazine or hydrogenated 1,4-diazine rings
    • C07D241/02Heterocyclic compounds containing 1,4-diazine or hydrogenated 1,4-diazine rings not condensed with other rings
    • C07D241/04Heterocyclic compounds containing 1,4-diazine or hydrogenated 1,4-diazine rings not condensed with other rings having no double bonds between ring members or between ring members and non-ring members
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D263/00Heterocyclic compounds containing 1,3-oxazole or hydrogenated 1,3-oxazole rings
    • C07D263/02Heterocyclic compounds containing 1,3-oxazole or hydrogenated 1,3-oxazole rings not condensed with other rings
    • C07D263/04Heterocyclic compounds containing 1,3-oxazole or hydrogenated 1,3-oxazole rings not condensed with other rings having no double bonds between ring members or between ring members and non-ring members
    • C07D263/06Heterocyclic compounds containing 1,3-oxazole or hydrogenated 1,3-oxazole rings not condensed with other rings having no double bonds between ring members or between ring members and non-ring members with hydrocarbon radicals, substituted by oxygen atoms, attached to ring carbon atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D277/00Heterocyclic compounds containing 1,3-thiazole or hydrogenated 1,3-thiazole rings
    • C07D277/02Heterocyclic compounds containing 1,3-thiazole or hydrogenated 1,3-thiazole rings not condensed with other rings
    • C07D277/04Heterocyclic compounds containing 1,3-thiazole or hydrogenated 1,3-thiazole rings not condensed with other rings having no double bonds between ring members or between ring members and non-ring members
    • C07D277/06Heterocyclic compounds containing 1,3-thiazole or hydrogenated 1,3-thiazole rings not condensed with other rings having no double bonds between ring members or between ring members and non-ring members with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D279/00Heterocyclic compounds containing six-membered rings having one nitrogen atom and one sulfur atom as the only ring hetero atoms
    • C07D279/101,4-Thiazines; Hydrogenated 1,4-thiazines
    • C07D279/121,4-Thiazines; Hydrogenated 1,4-thiazines not condensed with other rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D307/00Heterocyclic compounds containing five-membered rings having one oxygen atom as the only ring hetero atom
    • C07D307/02Heterocyclic compounds containing five-membered rings having one oxygen atom as the only ring hetero atom not condensed with other rings
    • C07D307/04Heterocyclic compounds containing five-membered rings having one oxygen atom as the only ring hetero atom not condensed with other rings having no double bonds between ring members or between ring members and non-ring members
    • C07D307/18Heterocyclic compounds containing five-membered rings having one oxygen atom as the only ring hetero atom not condensed with other rings having no double bonds between ring members or between ring members and non-ring members with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
    • C07D307/24Carbon atoms having three bonds to hetero atoms with at the most one bond to halogen
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings
    • C07D401/12Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings linked by a chain containing hetero atoms as chain links
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D403/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00
    • C07D403/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings
    • C07D403/04Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings directly linked by a ring-member-to-ring-member bond
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D403/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00
    • C07D403/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings
    • C07D403/12Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings linked by a chain containing hetero atoms as chain links
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D413/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms
    • C07D413/02Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing two hetero rings
    • C07D413/04Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing two hetero rings directly linked by a ring-member-to-ring-member bond
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D417/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00
    • C07D417/02Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00 containing two hetero rings
    • C07D417/04Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00 containing two hetero rings directly linked by a ring-member-to-ring-member bond
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07FACYCLIC, CARBOCYCLIC OR HETEROCYCLIC COMPOUNDS CONTAINING ELEMENTS OTHER THAN CARBON, HYDROGEN, HALOGEN, OXYGEN, NITROGEN, SULFUR, SELENIUM OR TELLURIUM
    • C07F9/00Compounds containing elements of Groups 5 or 15 of the Periodic System
    • C07F9/02Phosphorus compounds
    • C07F9/28Phosphorus compounds with one or more P—C bonds
    • C07F9/38Phosphonic acids RP(=O)(OH)2; Thiophosphonic acids, i.e. RP(=X)(XH)2 (X = S, Se)
    • C07F9/40Esters thereof
    • C07F9/4003Esters thereof the acid moiety containing a substituent or a structure which is considered as characteristic
    • C07F9/4006Esters of acyclic acids which can have further substituents on alkyl
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07FACYCLIC, CARBOCYCLIC OR HETEROCYCLIC COMPOUNDS CONTAINING ELEMENTS OTHER THAN CARBON, HYDROGEN, HALOGEN, OXYGEN, NITROGEN, SULFUR, SELENIUM OR TELLURIUM
    • C07F9/00Compounds containing elements of Groups 5 or 15 of the Periodic System
    • C07F9/02Phosphorus compounds
    • C07F9/28Phosphorus compounds with one or more P—C bonds
    • C07F9/48Phosphonous acids R—P(OH)2; Thiophosphonous acids including RHP(=O)(OH); Derivatives thereof
    • C07F9/4808Phosphonous acids R—P(OH)2; Thiophosphonous acids including RHP(=O)(OH); Derivatives thereof the acid moiety containing a substituent or structure which is considered as characteristic
    • C07F9/4833Cycloaliphatic acids or derivatives
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07FACYCLIC, CARBOCYCLIC OR HETEROCYCLIC COMPOUNDS CONTAINING ELEMENTS OTHER THAN CARBON, HYDROGEN, HALOGEN, OXYGEN, NITROGEN, SULFUR, SELENIUM OR TELLURIUM
    • C07F9/00Compounds containing elements of Groups 5 or 15 of the Periodic System
    • C07F9/02Phosphorus compounds
    • C07F9/547Heterocyclic compounds, e.g. containing phosphorus as a ring hetero atom
    • C07F9/553Heterocyclic compounds, e.g. containing phosphorus as a ring hetero atom having one nitrogen atom as the only ring hetero atom
    • C07F9/572Five-membered rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C2601/00Systems containing only non-condensed rings
    • C07C2601/06Systems containing only non-condensed rings with a five-membered ring
    • C07C2601/08Systems containing only non-condensed rings with a five-membered ring the ring being saturated

Definitions

  • the present invention relates to new and improved inhibitors of Dipepudyl Peptidase IV ("DPP IV"), and new and improved treatment methods and related uses.
  • DPP IV inhibitors according to the invention are useful for treating a wide variety of diseases and other abnormal conditions, including diseases impacting the central nervous system.
  • Dipeptidyl peptidase IV is a membrane-bound peptidase involved in the release of N-terminal dipeptides from proteins and other types or forms of peptides.
  • the enzyme is a type ⁇ membrane serine peptidase, and has a preference for removing proline-containing dipeptides from the N-terminus of the protein or peptide.
  • the enzyme contains 767 amino acids, and has been found in the kidney, epithelial cells, endothelial cells, small intestine, prostrate, seminal plasma and the brain.
  • the physiological roles of DPP IV have not been completely elucidated. It has been thought that DPP IV plays a role in the cleavage of various cytokines, growth factors and neuropeptides.
  • the enzyme also can cleave neuropeptides such as substance P and neuropeptide Y.
  • DPP IV is involved in cell adhesion and with the T-cell activation marker CD26.
  • DPP IV has been implicated in disease states such as HTV infection, diabetes, arthritis and certain cancers.
  • a DPP IV presence has been implicated in prostate and lung cancer, and DPP IV also has been found in patients having benign prostate hyperplasia.
  • DPP IV also is being investigated for its role in type II diabetes because the glucagon-like peptide (GLP-1) can be a substrate for DPP IV cleavage, and some DPP IV inhibitors have demonstrated efficacy in animal models for diabetes.
  • GLP-1 glucagon-like peptide
  • DPP IV has been implicated in HTV infection due to its association with CD 26. DPP IV also has been identified as a "research front" in an article about Alzheimer's disease. Shvaloff et al. , DIALOG FILE NO. 05335738/5.
  • DPP IV inhibition has been shown to increase release of TGF- ⁇ , a protein having neuroprotective properties. DPP IV inhibition itself, however, has not been implicated in a neuroprotective context.
  • DPP IV inhibition has been studied in the treatment of autoimmune diseases such as diabetes, arthritis and multiple sclerosis (a demyelination disease of the peripheral nerves). See PCT publications WO 97/40832 and WO 98/19998. Additionally, PCT publication WO 94/03055 discusses increasing production of hematopoietic cells with DPP IV inhibitors. PCT publication WO 95/11689 discloses the use of DPP IV inhibitors to block the entry of HIV into cells. U.S. Patent No. 5,543,396 discloses the use of inhibitors (certain proline phosphonate derivatives) to treat tumor invasion.
  • PCT publication WO 95/34538 mentions the use of certain serine protease inhibitors (such as certain DPP IV and PEP inhibitors) to treat peripheral neurological/autoimmune diseases like multiple sclerosis.
  • DPP IV inhibitors based upon molecules that bear a resemblance to proline have been investigated in the field.
  • PCT publication WO 95/11689 discloses c - amino boronic acid analogs of proline.
  • PCT publication WO 98/19998 discloses N- substituted 2-cyanopyrrolidines as DPP IV inhibitors.
  • PCT publication WO 95/34538 also discloses various proline containing compounds. Alexander et al., BIOSIS NO.
  • U.S. Patent Nos. 6,011,155; 6,110,949; and 6,124,305 discloses various N-substituted cyanopyrrolidines and cyanothiazolidines to inhibit DPP IV for the treatment of diabetes, and "conditions mediated by dipeptidyl peptidase- V inhibition. " The field, however, lacks appreciation of the usefulness of DPP IV inhibition for treating disease states, injuries and other abnormal conditions involving the central nervous system and other parts of the body, such as in the treatment of prostate.
  • inhibitors of dipeptidyl peptidase IV can include a proline mimetic and preferably possess an IC J0 of no more than about 1 ⁇ m, preferably no more than 100 nm, and have molecular weights of no more than 700, preferably no more than about 500.
  • the inhibitors are reversible.
  • the inhibitors preferably are sufficiently neutral and non-polar such that they can cross the blood-brain barrier via passive diffusion. In many cases, inhibitors that cannot cross by passive diffusion instead cross by active transport.
  • Inhibitors for use according to the invention include c-KPG and inhibitors according to Core Structures I, II , HI or IV, as shown below.
  • reversible inhibitors of dipeptidyl peptidase IV wherein the inhibitor is preferably reversible and preferably has a core structure of selected from the group consisting of Core Structure I, Core Structure II, Core Structure HI and Core Structure IV.
  • a given core structure can have functional and substitution groups, such as X, X,, A, Z and R, wherein X (if present) is CR2R3, O, S, or NR4; X, (if present) is CR2R3, O, S, or NR4 with the optional proviso that X and Xj cannot both be a heteroatom; A is H, COOH, or isosteres of carboxylic acids, such as one selected from the group consisting of CN, SO 3 H, CONOH, PO 3 R5R6, SO 2 NHR7, tetrazole, amides, esters, and acid anhydrides; Z (if present) is O or S; and the various R groups that are present are independently selected from the group of functional groups consisting of H, C r C 9 branched or straight chain alkyl, C 2 -C 9 branched or straight chain alkenyl, C 3 -C 8 cycloalkyl, C 5 -C 7 cycloalkenyl,
  • the inhibitors for use in such methods preferably should be reversible and preferably be able to cross the blood-brain barrier in amounts sufficient to treat the disorder.
  • the compounds according to the invention can be administered concurrently or sequentially with other compounds.
  • different compounds according to the invention e.g. , different compounds of one core structure group or compounds of two or more of the core structure groups
  • Uses of the compounds disclosed herein are provided (1) for treating disorders of the central nervous system and (2) for preparing compositions, formulations and medicaments for treating disorders of the central nervous system.
  • the inhibitors for use in such methods should be reversible and be able to penetrate or act upon the prostate.
  • the compounds according to the invention can be administered concurrently or sequentially with other compounds.
  • different compounds according to the invention e.g. , different compounds of one core structure group or compounds of two or more of the core structure groups
  • Uses of the compounds disclosed herein are provided (1) for treating disorders of the prostate and (2) for preparing compositions, formulations and medicaments for treating disorders of the prostate.
  • FIGURE 1 graphically depicts an assay employing organotypic spinal motor neurons and threohydroxyaspartate ("THA"). Exposure of neurons with THA alone resulted in death of 55-60% of the neurons. When the neurons were exposed to THA in combination with 10 ⁇ M c-KPG, the c-KPG spared greater than 50% of the neurons that would have otherwise been killed.
  • THA threohydroxyaspartate
  • the present invention provides DPP IV inhibitors that are useful for treating various disorders, including those of the central nervous system, among others.
  • the DPP IV inhibitors are pyrrolidine-based compounds, and more preferably constitute or include proline or proline mimetics.
  • the compounds according to the present invention preferably have sufficient stability, potency, selectivity, solubility and availability to be safe and effective in treating diseases, injuries and other abnormal conditions or insults to the central nervous system, the peripheral nerves and the prostate, for example.
  • treat in its various grammatical forms as used in relation to the present invention refers to preventing, curing, reversing, attenuating, alleviating, minimizing, suppressing, ameliorating or halting the deleterious effects of a disease state, disease progression, injury, wound, ischemia, disease causative agent (e.g. , bacteria, protozoans, parasites, fungi, viruses, viroids and/or prions), surgical procedure or other abnormal or detrimental condition (all of which are collectively referred to as "disorders,” as will be appreciated by the person of skill in the art).
  • a “therapeutically effective amount” of an inhibitor according to the invention is an amount that can achieve effective treatment, and such amounts can be determined in accordance with the present teachings.
  • DPP IV exhibits a preference for causing the removal of proline-containing dipeptides from the N-terminus of a protein or a peptide. Accordingly, proline has a structure that likely is recognized by or acted upon by the active site of DPP IV. Proline is unique among the 20 naturally-occurring amino acids in that it contains a cyclic secondary amino group, which as a result causes it to create interruptions in alpha- helical structures in proteins or peptides.
  • the DPP IV inhibitors according to the present invention can constitute or include proline or proline-like moieties, often referred to as "proline mimetics.”
  • a proline mimetic is a structure that sufficiently resembles proline such that its charge, polarity, shape and size are sufficiently duplicative of proline so as to participate in many of the molecular interactions involving proline.
  • a molecule or other compound that includes a proline moiety can itself be considered a proline mimetic. Accordingly, molecules that constitute or include proline or proline mimetics can interact with the natural interaction partners of proline, such as DPP IV.
  • a DPP IV inhibitor has the same or greater affinity for DPP IV than does the natural substrate of DPP IV, such as a protein containing a proline residue at its N-terminal end.
  • the inhibitor will have an equal or greater affinity to permit it to more effectively compete for the active site of DPP IV.
  • Inhibitors with lower affinities are still within the scope of the invention, and effective competition, and thus inhibition, can be ensured through dosing considerations.
  • the DPP IV inhibitor is used to treat disorders of the prostate, including, but not limited to, prostate cancer and post- prostatectomy nerve recovery.
  • erectile and voiding disorders are extremely common clinical conditions that result from diseases, injuries and trauma including complications associated with pelvic surgery. It is believed that local nerve injury during major pelvic surgeries account for complications such as erectile dysfunction and urinary incontinence. These complications might be caused by the trauma or the injury of the nerves (e.g. cavernous nerve) innervating the area during the surgery. Appropriate administration of a DPP IV inhibitors prior to, during or after surgery may be effective in blocking the nerve degeneration caused by pelvic surgery.
  • the inhibitor of the invention can be administered in the manner used with other prostate therapeutics, and can be combined with other products or methodologies for treating the prostate.
  • the DPP IV inhibitor can be used to treat disorders of the central nervous system (CNS) and the peripheral nerves.
  • CNS central nervous system
  • the DPP IV inhibitors according to the present invention can be used to treat CNS maladies such as strokes, tumors, ischemia, Parkinson's disease, memory loss, hearing loss, vision loss, migraines, brain injury, spinal cord injury, Alzheimer's disease and amyotrophic lateral sclerosis (which has a CNS component).
  • the DPP IV inhibitors can be used to treat disorders having a more peripheral nature, including multiple sclerosis and diabetic neuropathy.
  • the blood-brain barrier prevents many compounds in the circulation from crossing to the brain.
  • the brain is a complex biological structure that is susceptible to a variety of toxins. Additionally, being that the brain is composed primarily of nerves and related tissues, the brain lacks the natural regenerative capabilities of other organs and tissues. For example, the skin has extensive regeneration and restorative capabilities, and thus can withstand encounters with toxins and other physical insults, which it can be expected to encounter in daily life.
  • the brain itself is quite susceptible to toxins, and thus it is thought that the blood-brain barrier was an evolutionary development to protect the integrity of the brain.
  • the blood-brain barrier also can prevent the entry of beneficial compounds, such as drugs, that are needed to treat a disease, injury or other abnormal condition. Accordingly, the blood-brain barrier can be a complicating factor in developing therapeutics for the CNS.
  • a compound that crosses the blood-brain barrier via passive diffusion should have a log P between about 1 and about 4.
  • the log D which takes into consideration the charge of the compound.
  • polar and charged compounds are less amenable to crossing the blood-brain barrier by passive diffusion. Accordingly, a log D greater than about -2 is preferred.
  • the concepts of log P and log D are discussed in Waterbeemed, STRUCTURAL-PROPERTY CORRELATIONS IN DRUG RESEARCH (Academic Press).
  • the compound preferably has a molecular weight of about 700 or less, preferably about 500 or less.
  • a compound that is to cross the blood-brain barrier by passive diffusion should be "sufficiently neutral and non-polar" for its size that it can cross the blood-brain barrier in a therapeutically effective amount
  • potency of the compound as an inhibitor Besides efficiency of a compound in crossing the blood-brain barrier, another important consideration is the potency of the compound as an inhibitor.
  • potent inhibitors can have a lower efficiency in crossing the blood-brain barrier, but nevertheless can be effective due to their higher potencies. Conversely, a less potent inhibitor may require greater efficiency in crossing the blood-brain barrier in order to have a beneficial effect.
  • a therapeutically effective amount for treating a CNS disorder depends upon the potency of the inhibitor and its efficiency in crossing the blood-brain barrier or the administration route and approach employed to circumvent the blood-brain barrier.
  • the DPP IV inhibitors preferably have an IC 50 (for inhibition concentration where 50% of DPP IV is inhibited) value of less than about 1 ⁇ m, and preferably less than 100 ran.
  • IC 50 for inhibition concentration where 50% of DPP IV is inhibited
  • DPP IV inhibitors can have higher IC 50 values as long as their efficiency in crossing the blood-brain barrier is sufficient to treat the disease, injury or other abnormal condition.
  • the DPP IV inhibitor according to the invention is a reversible inhibitor. That is, the DPP IV inhibitor should be able to interact with the inhibitor without becoming permanently bound thereto in a manner that would denature or inactivate the DPP IV enzyme. The need for reversibility is due to the fact that DPP IV is a naturally- occurring enzyme that has normal physiologic functions.
  • An irreversible inhibitor can effectively eliminate functions of the enzyme, and thus result in cessation of normal physiologic processes.
  • the present invention utilizes the inhibition of DPP IV in certain contexts, such as in treating an ischemic event, for definite periods of time, such as during and after reperfusion in the ischemic area.
  • a reversible inhibitor would permit inhibited DPP IV molecules to resume normal function once the need for inhibition is gone.
  • the compounds according to the invention can be administered by a variety of systemic and CNS-targeted routes.
  • intra-arterial, intravenous intraventricular, intracavitary and intracranial administration routes can be employed.
  • Exemplary injection modalities can be by way of bolus, periodic injection and/or constant infusion.
  • the following routes can be employed for the compounds according to the invention, including parenteral, oral, nasal, inhalation spray, buccally, topically, transdermal, rectal, vaginal, via implanted reservoir or other routes available to the skilled person.
  • parenteral as used herein includes subcutaneous, intravenous, intramuscular, intraperitoneal, intrathecal, intraventricular, intrasternal, intracranial or intraosseous injection and infusion techniques.
  • the compounds of the present invention preferably penetrate the blood-train barrier when peripherally administered. Compounds which cannot sufficiently penetrate the blood-brain barrier can be effectively administered by an intraventricular route. It also is important to note that during the active phase of certain CNS disorders, blood-brain lineage is known to occur and will permit entry of the compounds of the invention to the central nervous system. Moreover, there are several other techniques that either physically break through the blood-brain barrier or circumvent it to deliver therapeutic agents. Examples of these techniques include intrathecal injections, surgical implants, and osmotic techniques. Invasive techniques often are employed, particularly direct administration to damaged neuronal tissue. One or more of the above can be employed according to the invention.
  • One embodiment for the administration of the compounds of the invention is by intrathecal injection, i.e., directly into the cerebrospinal fluid by puncturing the membranes surrounding the central nervous system is usually by lumbar puncture. Sustained dosages of agents directly into the cerebrospinal fluid can be attained by the use of infusion pumps that are implanted surgically.
  • Another embodiment for the administration of the compounds of the invention is by injection directly into the lumbar cerebrospinal fluid (intrathecally) or by injection intravenously.
  • the compounds according to the invention can be formulated with pharmaceutically- acceptable carriers and diluents, and can be used with methods and uses according to the invention.
  • the formulation will depend upon the disease state being treated and the administration route. See, for example, U.S. Patent No. 5,874,449, which is incorporated by reference.
  • Pharmaceutically acceptable carriers include aqueous solutions, non-toxic excipients, including salts, preservatives, buffers, such as phosphate buffers, and the like, as described in UNITED STATES PHARMACOPEIA AND NATIONAL FORMULARY (USP 24-NF 19); REMINGTON'S PHARMACEUTICAL SCIENCES; HANDBOOK ON PHARMACEUTICAL EXCIPIENTS (2d edition, Wade and Weller eds. 1994), the each of which are hereby incorporated by reference.
  • non-aqueous solvents are propylene glycol, polyethylene glycol, vegetable oil and injectable organic esters such as ethyloleate.
  • Aqueous carriers include water, alcoholic/aqueous solutions, saline solutions, parenteral vehicles, such as sodium chloride and Ringer's dextrose.
  • Intravenous vehicles include fluid and nutrient replenishers.
  • Preservatives include antimicrobials, anti-oxidants, chelating agents and inert gases.
  • the pH and exact concentration of the various components of the binding composition are adjusted according to routine skills in the art. See GOODMAN AND GILMAN'S THE PHARMACOLOGICAL BASIS FOR THERAPEUTICS (9th edition), the contents of which are hereby incorporated by reference.
  • Exemplary approaches include those where the compounds are to be administered in the form of sterile injectable preparations, for example, as sterile injectable aqueous or oleaginous suspensions. These suspensions can be formulated according to techniques known in the art using suitable dispersing or wetting agents and suspending agents.
  • the sterile injectable preparations may also be sterile injectable solutions or suspensions in non- toxic parenterally-acceptable diluents or solvents, for example, as solutions in 1,3- butanediol.
  • acceptable vehicles and solvents that may be employed are water, Ringer's solution and isotonic sodium chloride solution.
  • sterile fixed oils are conventionally employed as solvents or suspending mediums.
  • any blank fixed oil such as a synthetic mono- or di-glyceride may be employed.
  • Fatty acids such as a oleic acid and its glyceride derivatives, including olive oil and castor oil, especially in their polyoxyethylated forms, are useful in the preparation of injectables.
  • These oil solutions or suspensions may also contain long-chain alcohol diluents or dispersants.
  • the compounds may be administered orally in the form of capsules, tablets, aqueous suspensions or solutions; Tablets may contain carriers such as lactose and corn starch, and/or lubricating agents such as magnesium stearate.
  • Capsules may contain diluents including lactose and dried corn starch.
  • Aqueous suspensions may contain emulsifying and suspending agents combined with the active ingredient.
  • the oral dosage forms may further contain sweetening and/or flavoring and/or coloring agents.
  • compositions can be prepared by mixing the drug with suitable non-irritating excipients which are solid at room temperature, but liquid at rectal temperature such that they will melt in the rectum to release the drug.
  • suitable non-irritating excipients include cocoa butter, beeswax and polyethylene glycols.
  • the compounds may be administered topically, especially when the conditions addressed for treatment involve areas or organs readily accessible by topical application, including neurological disorders of the eye, the skin or the lower intestinal tract.
  • the compounds can be formulated as micronized suspensions in isotonic, pH adjusted sterile saline or, preferably, as a solution in isotonic, pH adjusted sterile saline, either with or without a preservative such as benzylalkonium chloride.
  • the compounds may be formulated into ointments, such as petrolatum.
  • the compounds can be formulated into suitable ointments containing the compounds suspended or dissolved in, for example, mixtures with one or more of the following: mineral oil, liquid petrolatum, white petrolatum, propylene glycol, polyoxyethylene polyoxypropylene compound, emulsifying wax and water.
  • the compounds can be formulated into suitable lotions or creams containing the active compound suspended or dissolved in, for example, a mixture of one or more of the following: mineral oil, sorbitan monostearate, polysorbate 60, cetyl ester wax, cetearyl alcohol, 2-octyldodecanol, benzyl alcohol and water.
  • the compounds of the present invention may be administered by a single dose, multiple discrete doses or continuous infusion. Because the compounds preferably are small, easily diffusible and relatively stable, they can be well-suited to continuous infusion. Dose levels on the order of about 0.1 mg to about 10,000 mg of the active ingredient are useful in the treatment of the above conditions, with preferred levels being about 0.1 mg to about 1,000 mg.
  • the specific dose level, and thus the therapeutically- effective amount, for any particular patient will vary depending upon a variety of factors, including the activity of the specific compound employed and its bioavailability at the site of drug action; the age, body weight, general health, sex and diet of the patient; the time of administration; the rate of excretion; drug combination; the severity of the particular disease being treated; and the form of administration.
  • in vitro dosage-effect results provide useful guidance on the proper doses for patient administration. Studies in animal models also are helpful. The considerations for determining the proper dose levels are available to the skilled person. See Example 5 below.
  • Certain compounds can administered in lyophilized form.
  • 1 to 100 mg of a compound of the present invention may be lyophilized in individual vials, together with a carrier and a buffer, such as mannitol and sodium phospshate.
  • the compound may be reconstituted in the vials with bacteriostatic water before administration.
  • the compounds of the present invention are preferably administered orally, rectally, parenterally or topically at least 1 to 6 times daily, and may follow an initial bolus dose of higher concentration.
  • Administration Regimen and Timing are preferably administered orally, rectally, parenterally or topically at least 1 to 6 times daily, and may follow an initial bolus dose of higher concentration.
  • any administration regimen regulating the timing and sequence of drug delivery can be used and repeated as necessary to effect treatment.
  • Such regimen may include pretreatment and/or co- administration with additional therapeutic agents.
  • the compounds should be administered to the affected cells as soon as possible. In situations where nervous insult is anticipated, the compounds should be administered before the expected nervous insult.
  • Such simations of increased likelihood of nervous insult include surgery (for example, carotid endarterectomy, cardiac, vascular, aortic, orthopedic); endovascular procedures such as arterial catherization (for example, carotid, vertebral, aortic, cardia, renal, spinal, Adamkiewicz); injections of embolic agents; coils or balloons for hemostasis; interruptions of vascularity for treatment of brain lesions; and predisposing medical conditions such as crescendo transient ischemic attacks, emboli and sequential strokes.
  • any compound for the treatment of stroke should adequately cross the blood-brain barrier and obtain sufficiently therapeutic levels within the brain and cerebral spinal fluid.
  • the compounds of the present invention may be administered (i) prior to surgery or radiation treatment to reduce the risk of metastasis; (ii) during surgery or in conjunction with radiation treatment; and/or (iii) after surgery or radiation therapy to reduce the risk of recurrence and to inhibit the growth of any residual tumorous cells.
  • the compounds of the present invention may be administered as a continuous supplement to, or as a replacement for, homonal ablation in order to slow tumor cell growth in both the untreated primary tumor and the existing metastatic lesions.
  • the compounds, methods and uses of the present invention are particularly useful where shed cells could not be removed by surgical intervention. After post-surgical recovery, the compounds, methods and uses of the present invention would be effective in reducing the chances of recurrence of a tumor engendered by such shed cells.
  • the compounds, methods and uses of the present invention also to provide combined preparation for simultaneous, separate, or sequential use which contain other biologically active agents.
  • biologically active agent can be either another compound of the present invention; steroids, for example hydrocortisomers such as methylprednisolone; anti- inflammatory or anti-immune drugs, such as methotrexate, azathioprine, cyclophosphamide or cyclosporin A; interferon- ⁇ ; antibodies, such as anti-CD4 antibodies; agents which can reduce the risk of a second ischemic event, such as ticlopidine; chemotherapeutic compositions; immunotherapeutic compositions; morphine for treating pain; or mixtures thereof.
  • the compounds according to the invention include various substitutions available to the skilled person and are to be employed in accordance with the teachings contained herein.
  • the Core Structures which constitute or include proline mimetics, can include a variety of functional groups as taught herein.
  • the inventions include isosteres of the compounds or the function groups contained therein. Guiding principles and illustrative examples of functional groups and isosteres are set forth in Smith et al. , INTRODUCTION TO THE PRINCIPLES OF DRUG DESIGN (John Wright & Sons, Ltd.), which is hereby incorporated by reference.
  • the compounds used according to the invention preferably are or contain moieties that resemble proline within their core strucmres. That is, these compounds are or contain proline mimetics.
  • One such compound that can be used according to the invention contains a proline mimetic and has the following structure:
  • c-KPG This compound, referred to as "c-KPG,” was tested in an assay employing organotypic spinal motor neurons and threohydroxyaspartate (“THA”), which is an inhibitor of the glutamate reuptake receptor. Synthesis protocols for c-KPG are disclosed in Nguyen et al., J. Med. Chem. 41: 2100-10 (1998).
  • Core structures which are DPP IV inhibitors and constitute or contain proline mimetics, are set forth below.
  • Exemplary core structures are depicted schematically, and the functional/substitution groups are set forth in text. All substitutions contemplated herein are permissive for various provisos, either alone or in any combination, such that if one group is included in a given position another group at the same or different position can be excluded.
  • Core Structure I is:
  • X is CR2R3, O, S, or NR4; optional proviso 1 that if X is S, then A cannot be CN; optional proviso 2 that if X is CH 2 and R is H, then A cannot be C; optional proviso 3 that if X is S, then Rl cannot be amino-substimted alkyl; optional proviso 4 that if X is CH 2 , then A cannot be COOH; optional proviso 5 that if X is S, or if X and XI are both CH 2 , and Z is O, and A is CN, and Rl is H, then R is not NH substituted with C1-C9 straight or branched chain alkyl, or NH substituted with C3-C7 cycloalkyl;
  • X is CR2R3, O, S, or NR4 with optional proviso 6 that X and XI cannot both be a heteroatom; optional proviso 7 if X and XI are both CH 2 , and Z is O, and Rl is NH 2 , then R is not 1-methylpropyl if A is COOH, and R is not cyclopentyl if A is CN.
  • A is H, COOH, or isosteres of carboxylic acids, such as one selected from the group consisting of CN, SO 3 H, CONOH, PO 3 R5R6, SO 2 NHR7, tetrazole, amides, esters, and acid anhydrides with optional proviso 8 that if A is CN, and Rl is NH 2 , and Z is O, and R is 1-methylpropyl, then X and XI are not both CH 2 , X and XI are not S, X is not O, and Z is O or S;
  • R and Rl are independently selected from the group of functional groups consisting of H, C r C 9 branched or straight chain alkyl, C 2 -C 9 branched or straight chain alkenyl, C 3 - C 8 cycloalkyl, C 5 -C 7 cycloalkenyl, aryl, heteroaryl and amino, wherein any of the functional groups can be substituted with one or more of C r C 9 straight or branched chain alkyl, aryl, heteroaryl, amino, halo, carbonyl, C r C 9 alkoxy, C 2 -C 9 alkenyloxy, phenoxy, benzyloxy, C 3 -C 8 cycloalkyl, cyano, amido, thiol, trifluromethyl, or hydroxy, wherein each of R and Rl can be the same or different; and
  • R2, R3, R4, R5, R6 and R7, if present, are independently selected from the group of functional groups consisting of H, C r C 9 branched or straight chain alkyl, C 2 -C 9 branched or straight chain alkenyl, C 3 -C 8 cycloalkyl, C 5 -C 7 cycloalkenyl, aryl, heteroaryl and amino, wherein any of the functional groups can be substituted with one or more of - C 9 straight or branched chain alkyl, aryl, heteroaryl, amino, halo, carbonyl, C t -C 9 alkoxy, C 2 -C 9 alkenyloxy, phenoxy, benzyloxy, C 3 -C 8 cycloalkyl, cyano, amido, thiol, trifluromethyl, or hydroxy, wherein each of R2, R3, R4, R5, R6 and R7, if present, can be the same or different.
  • core strucmres are provide according to the invention, such as those having ring modifications (II and HI): .
  • X is CR2R3, O, S, or NR4;
  • A is H, COOH, or isosteres of carboxylic acids, such as one selected from the group consisting of CN, SO 3 H, CONOH, PO 3 R5R6, SO 2 NHR7, tetrazole, amides, esters, and acid anhydrides;
  • R and Rl are independently selected from the group of functional groups consisting of H, C,-C 9 branched or straight chain alkyl, C 2 -C 9 branched or straight chain alkenyl, C 3 - C 8 cycloalkyl, C 5 -C 7 cycloalkenyl, aryl, heteroaryl and amino, wherein any of the functional groups can be substituted with one or more of C r C 9 straight or branched chain alkyl, aryl, heteroaryl, amino, halo, carbonyl, C r C 9 alkoxy, C 2 -C 9 alkenyloxy, phenoxy, benzyloxy, C 3 -C 8 cycloalkyl, cyano, amido, thiol, trifluromethyl, or hydroxy, wherein each of R and Rl can be the same or different; and
  • R2, R3, R4, R5, R6 and R7, if present, are independently selected from the group of functional groups consisting of H, C,-C 9 branched or straight chain alkyl, C 2 -C 9 branched or straight chain alkenyl, C 3 -C 8 cycloalkyl, C 5 -C 7 cycloalkenyl, aryl, heteroaryl and amino, wherein any of the functional groups can be substituted with one or more of C r C 9 straight or branched chain alkyl, aryl, heteroaryl, amino, halo, carbonyl, C,-C 9 alkoxy, C 2 -C 9 alkenyloxy, phenoxy, benzyloxy, C 3 -C 8 cycloalkyl, cyano, amido, thiol, trifluromethyl, or hydroxy, wherein each of R2, R3, R4, R5, R6 and R7, if present, can be the same or different.
  • Core Structure in is:
  • A is H, COOH, or isosteres of carboxylic acids, such as one selected from the group consisting of CN, SO 3 H, CONOH, PO 3 R5R6, SO 2 NHR7, tetrazole, amides, esters, and acid anhydrides;
  • R, Rl, R2 and R3 are independently selected from the group of functional groups consisting of H, C r C 9 branched or straight chain alkyl, C 2 -C 9 branched or straight chain alkenyl, C 3 -C 8 cycloalkyl, C 5 -C 7 cycloalkenyl, aryl, heteroaryl and amino, wherein any of the functional groups can be substituted with one or more of C C 9 straight or branched chain alkyl, aryl, heteroaryl, amino, halo, carbonyl, C r C 9 alkoxy, C 2 -C 9 alkenyloxy, phenoxy, benzyloxy, C 3 -C 8 cycloalkyl, cyano, amido, thiol, trifluromethyl, or hydroxy, wherein each of R, Rl, R2 and R3 can be the same or different; and
  • R4, R5, R6 and R7, if present, are independently selected from the group of functional groups consisting of H, C,-C 9 branched or straight chain alkyl, C 2 -C 9 branched or straight chain alkenyl, C 3 -C 8 cycloalkyl, C 5 -C 7 cycloalkenyl, aryl, heteroaryl and amino, wherein any of the functional groups can be substimted with one or more of C r C 9 straight or branched chain alkyl, aryl, heteroaryl, amino, halo, carbonyl, C r C 9 alkoxy, C 2 -C 9 alkenyloxy, phenoxy, benzyloxy, C 3 -C 8 cycloalkyl, cyano, amido, thiol, trifluromethyl, or hydroxy, wherein each of R4, R5, R6 and R7, if present, can be the same or different.
  • X is CR2R3, O, S, or NR4;
  • X is CR2R3, O, S, or NR4 with the proviso that X and XI cannot both be a heteroatom;
  • A is H, COOH, or isosteres of carboxylic acids, such as one selected from the group consisting of CN, SO 3 H, CONOH, PO 3 R5R6, SO 2 NHR7, tetrazole, amides, esters, and acid anhydrides;
  • R and Rl are independently selected from the group of functional groups consisting of H, C r C 9 branched or straight chain alkyl, C 2 -C 9 branched or straight chain alkenyl, C 3 - C 8 cycloalkyl, C 5 -C 7 cycloalkenyl, aryl, heteroaryl and amino, wherein any of the functional groups can be substituted with one or more of C r C 9 straight or branched chain alkyl, aryl, heteroaryl, amino, halo, carbonyl, C C 9 alkoxy, C 2 -C 9 alkenyloxy, phenoxy, benzyloxy, C 3 -C 8 cycloalkyl, cyano, amido, thiol, trifluromethyl, or hydroxy, wherein each of R and Rl can be the same or different; and
  • R2, R3, R4, R5, R6 and R7, if present, are independently selected from the group of functional groups consisting of H, C r C 9 branched or straight chain alkyl, C 2 -C 9 branched or straight chain alkenyl, C 3 -C 8 cycloalkyl, C 5 -C 7 cycloalkenyl, aryl, heteroaryl and amino, wherein any of the functional groups can be substituted with one or more of C r C 9 straight or branched chain alkyl, aryl, heteroaryl, amino, halo, carbonyl, C r C 9 alkoxy, C 2 -C 9 alkenyloxy, phenoxy, benzyloxy, C 3 -C 8 cycloalkyl, cyano, amido, thiol, trifluromethyl, or hydroxy, wherein each of R2, R3, R4, R5, R6 and R7, if present, can be the same or different.
  • the compounds of the core structures according to the present invention can administered in ester or salt forms according to the teachings provided herein. Acceptable formulations, dosages and administration regimens can be determined in accordance with the teachings contained herein.
  • R is NH-R 1 ;
  • R 1 is: C t - C 12 straight or branched chain alkyl; C 3 - C 7 cycloalkyl; CH 2 - CH 2 -NH-R U ;
  • R" is a pyridine ring optionally substimted in one or two positions with halo, trifluoromethyl, cyano or nitro; or a pyrimidine ring optionally substimted in one position with halo, trifluromethyl, cyano or nitro;
  • R ⁇ is a phenyl ring optionally substimted in one to three positions with halo or C, - C 3 alkoxy;
  • Each R w is independently a phenyl ring optionally substimted in one position with halo or C, - C 3 alkoxy; and R v is a 2-oxopyrrolidine group or a C 2 - C 4 alkoxy group.
  • R is NH-R 1 ;
  • R 1 is: Cj - C 12 straight or branched chain alkyl optionally substimted with hydroxy, acetyl,
  • adamantyl indanyl; piperidyl optionally substimted with benzyl; pyrrolidine optionally substimted with benzyl; bicycloheptyl optionally substituted in one to three positions with methyl; phenyl optionally substimted with in one to three positions with halo, methoxy, trifluoromethyl; pyridyl optionally substimted in one to three positions with halo, trifluoromethyl, nitro; or pyrimidyl optionally substimted with halo, trifluoromethyl, nitro;
  • R m is phenyl optionally substituted with CN, or pyridyl optionally substimted with CN;
  • R w is a group selected from phenyl, naphthyl, cyclohexenyl, pyridyl, pyrimidyl, adamantyl, phenoxy, wherein the group is optionally substimted in one to two positions with ethoxy, methoxy, halo, phenylsulfide, or phenylsulfide substimted with hydroxymethyl.
  • Prlnclpal Group carboxylic acid
  • Prlncloal Group carboxylic acid
  • Parent Hvdride pyrrolidine
  • EXAMPLE 5 EXEMPLARY NEURO ACTIVITY TESTING PROTOCOLS
  • DRG dorsal root ganglion
  • the spinal cord with attached DRGs from an adult mouse (15-10g) is removed.
  • Spinal nerves are cut away using micro-dissection scissors and any excess material is trimmed until the DRG is free.
  • Using sharp micro-dissecting scissors a transverse cut is made in the peripheral nerve, leaving 1-2 mm attached, and the explant is placed into Petri dish and covered with plating media.
  • the spinal nerve is trimmed to about ImM in length. Then, embed the explant in 30 ⁇ L of reduced growth factor Matrigel on a circular coverslip, and place in a 35 mM culture dish.
  • Primary antibody for example, Beta tubulin, Sigma Chemical Co.
  • secondary antibody Alexa 488 Goat Anti-Mouse
  • Cultures are derived from postnatal day 8 (P8) Sprague-Dawley rat lumbar spinal cord slices of 325 micron thickness. Each experiment consists of two 6- well plates with 5 slices from 4 different animals per well. Media changes are performed every 3 to 4 days. Cultures are treated with THA [L(-)-threo-3-hydroxyaspartic acid; Tocris Cookson Inc., Ballwin, Missouri] at 200 ⁇ M + compound (lO ⁇ M) after one week in culture.
  • the control is an untreated sample with 0.1 % DMSO as vehicle.
  • the THA control is a THA treated sample with 0.1 % DSMO as vehicle. Two wells are used per condition. One media change with new THA and compounds is performed.
  • the experiment is stopped 6 to 8 days following drug treatment (13-15 total days in vitro, DIV) as dictated by visual assessment of lesion, by fixation with 4% paraformaldehyde/0.1 M phosphate buffer for 30 minutes.
  • Slices are permeabilized with 100% cold methanol forlO minutes. Slices are transferred to staining wells. The slices are blocked with 10% HS/TBS. Primary antibody incubation is overnight at 4°C with SMI-32 antibody 1:5000 in 2% HS/TBS. SMI-32 was specific towards unphosphorylated H neurofilament subunit.
  • Vectastain ABC Elite Kit with rat absorbed anti-mouse secondary antibody is used with DAB to stain the slices.
  • the slices are mounted onto a slide and a coverslip is sealed with DPX mounting solution.
  • Quantification of surviving neurons is performed on a Ziess Axiovert microscope. Neuronal survival is determined by observing an intact neuronal cell body with processes located ventrally of the central canal in each hemisphere. This correlates to laminae VII, VIII and IX. Each hemisphere is counted individually. The statistics can be performed with StatView software on a minimum of three different experiments per condition and significance should be determined as compared to THA control. The percent of protection can be determined from the average number of living neurons by the following equation:
  • EXAMPLE 6 EXEMPLARY TESTING PROTOCOLS FOR PROSTATE
  • Protocols for testing efficacy, dosing, and administration schedules for post- prostatectomy nerve recovery can be performed in accordance with the teachings of Example 5.
  • DPP IV inhibitors in the treatment of prostate cancer, there are several cancer cell lines available of conducting in vitro assays. Appropriate cell lines include LNCaP, PC3, DU-145 and TSUPrl for use in cell proliferation assays.
  • a cell line can be propagated in a standard medium, such as RPMI

Abstract

The present invention provides the inhibitors of dipeptidyl peptidase IV based upon or including proline or similar moieties. The inhibitors are useful for treating various disorders, including those of the central nervous system and the prostate. Many of the inhibitors can be reversible, and can cross the blood-brain barrier. Methods of making and using the inhibitors and treatment methods also are provided.

Description

DIPEPTIDYL PEPTIDASE IV INHIBITORS AND METHODS OF MAKING AND USING DIPEPTIDYL PEPΏDASE TV INHIBITORS This application is a continuation-in-part of U.S. Application Serial No.
09/439,089, filed November 12, 1999, the entirety of which is hereby incorporated by reference.
BACKGROUND OF THE INVENTION
The present invention relates to new and improved inhibitors of Dipepudyl Peptidase IV ("DPP IV"), and new and improved treatment methods and related uses. The DPP IV inhibitors according to the invention are useful for treating a wide variety of diseases and other abnormal conditions, including diseases impacting the central nervous system.
Dipeptidyl peptidase IV is a membrane-bound peptidase involved in the release of N-terminal dipeptides from proteins and other types or forms of peptides. The enzyme is a type π membrane serine peptidase, and has a preference for removing proline-containing dipeptides from the N-terminus of the protein or peptide. The enzyme contains 767 amino acids, and has been found in the kidney, epithelial cells, endothelial cells, small intestine, prostrate, seminal plasma and the brain. The physiological roles of DPP IV have not been completely elucidated. It has been thought that DPP IV plays a role in the cleavage of various cytokines, growth factors and neuropeptides. The enzyme also can cleave neuropeptides such as substance P and neuropeptide Y. There also have been suggestions that DPP IV is involved in cell adhesion and with the T-cell activation marker CD26. DPP IV has been implicated in disease states such as HTV infection, diabetes, arthritis and certain cancers. For example, a DPP IV presence has been implicated in prostate and lung cancer, and DPP IV also has been found in patients having benign prostate hyperplasia. DPP IV also is being investigated for its role in type II diabetes because the glucagon-like peptide (GLP-1) can be a substrate for DPP IV cleavage, and some DPP IV inhibitors have demonstrated efficacy in animal models for diabetes. Additionally, DPP IV has been implicated in HTV infection due to its association with CD 26. DPP IV also has been identified as a "research front" in an article about Alzheimer's disease. Shvaloff et al. , DIALOG FILE NO. 05335738/5.
Inhibition of DPP IV has been shown to increase release of TGF-β, a protein having neuroprotective properties. DPP IV inhibition itself, however, has not been implicated in a neuroprotective context.
DPP IV inhibition has been studied in the treatment of autoimmune diseases such as diabetes, arthritis and multiple sclerosis (a demyelination disease of the peripheral nerves). See PCT publications WO 97/40832 and WO 98/19998. Additionally, PCT publication WO 94/03055 discusses increasing production of hematopoietic cells with DPP IV inhibitors. PCT publication WO 95/11689 discloses the use of DPP IV inhibitors to block the entry of HIV into cells. U.S. Patent No. 5,543,396 discloses the use of inhibitors (certain proline phosphonate derivatives) to treat tumor invasion. PCT publication WO 95/34538 mentions the use of certain serine protease inhibitors (such as certain DPP IV and PEP inhibitors) to treat peripheral neurological/autoimmune diseases like multiple sclerosis. DPP IV inhibitors based upon molecules that bear a resemblance to proline have been investigated in the field. For example, PCT publication WO 95/11689 discloses c - amino boronic acid analogs of proline. PCT publication WO 98/19998 discloses N- substituted 2-cyanopyrrolidines as DPP IV inhibitors. PCT publication WO 95/34538 also discloses various proline containing compounds. Alexander et al., BIOSIS NO. 199900218969 discusses research on prolylpyrrolidine phosphonates that are considered irreversible DPP IV inhibitors. U.S. Patent Nos. 6,011,155; 6,110,949; and 6,124,305 discloses various N-substituted cyanopyrrolidines and cyanothiazolidines to inhibit DPP IV for the treatment of diabetes, and "conditions mediated by dipeptidyl peptidase- V inhibition. " The field, however, lacks appreciation of the usefulness of DPP IV inhibition for treating disease states, injuries and other abnormal conditions involving the central nervous system and other parts of the body, such as in the treatment of prostate. Therefore, there exists needs for safe and effective compositions and methodologies for treating disease states, injuries and other abnormal conditions involving the central nervous system and other parts of the body by inhibiting DPP IV. These needs have gone unresolved until the development of the present inventions. SUMMARY OF THE INVENTIONS
In view of the needs of the art to provide new therapeutic products, methodologies, and uses, it is an object of the invention to provide inhibitors of dipetidyl peptidase.
In accomplishing this object and other objects, there are provided, in accordance with one aspect of the invention, inhibitors of dipeptidyl peptidase IV. The inhibitors according to the invention can include a proline mimetic and preferably possess an ICJ0 of no more than about 1 μm, preferably no more than 100 nm, and have molecular weights of no more than 700, preferably no more than about 500. Preferably, the inhibitors are reversible. Where the inhibitors are to be used to treat disorders involving the central nervous system, the inhibitors preferably are sufficiently neutral and non-polar such that they can cross the blood-brain barrier via passive diffusion. In many cases, inhibitors that cannot cross by passive diffusion instead cross by active transport. Of course, administration approaches also can be employed when treating the central nervous system to avoid adverse interference from the blood-brain barrier. Inhibitors for use according to the invention include c-KPG and inhibitors according to Core Structures I, II , HI or IV, as shown below.
In accordance with another aspect of the present invention, there are provided reversible inhibitors of dipeptidyl peptidase IV, wherein the inhibitor is preferably reversible and preferably has a core structure of selected from the group consisting of Core Structure I, Core Structure II, Core Structure HI and Core Structure IV. A given core structure can have functional and substitution groups, such as X, X,, A, Z and R, wherein X (if present) is CR2R3, O, S, or NR4; X, (if present) is CR2R3, O, S, or NR4 with the optional proviso that X and Xj cannot both be a heteroatom; A is H, COOH, or isosteres of carboxylic acids, such as one selected from the group consisting of CN, SO3H, CONOH, PO3R5R6, SO2NHR7, tetrazole, amides, esters, and acid anhydrides; Z (if present) is O or S; and the various R groups that are present are independently selected from the group of functional groups consisting of H, CrC9 branched or straight chain alkyl, C2-C9 branched or straight chain alkenyl, C3-C8 cycloalkyl, C5-C7 cycloalkenyl, aryl, heteroaryl and amino, wherein any of the functional groups can be substituted with one or more of CrC9 straight or branched chain alkyl, aryl, heteroaryl, amino, halo, carbonyl, CrC9 alkoxy, C2-C9 alkenyloxy, phenoxy, benzyloxy, C3-C8 cycloalkyl, cyano, amido, thiol, triflouromethyl, or hydroxy, wherein each of R and Rl can be the same or different; all substitutions contemplated herein are permissive for various provisos, either alone or in any combination, such that if one group is included in a given position another group at the same or different position can be excluded; and
In accordance with still another aspect of the invention, there are provided methods of treating patients having disorders involving the central nervous system with inhibitors of DPP IV. Preferably, the inhibitors for use in such methods preferably should be reversible and preferably be able to cross the blood-brain barrier in amounts sufficient to treat the disorder. The compounds according to the invention can be administered concurrently or sequentially with other compounds. Additionally, different compounds according to the invention (e.g. , different compounds of one core structure group or compounds of two or more of the core structure groups) can be administered concurrently or sequentially. Uses of the compounds disclosed herein are provided (1) for treating disorders of the central nervous system and (2) for preparing compositions, formulations and medicaments for treating disorders of the central nervous system.
In accordance with still another aspect of the invention, there are provided methods of treating patients having disorders of the prostate, including prostate abnormalities such as prostate cancer and post-prostatectomy nerve recovery. Preferably, the inhibitors for use in such methods should be reversible and be able to penetrate or act upon the prostate. The compounds according to the invention can be administered concurrently or sequentially with other compounds. Additionally, different compounds according to the invention (e.g. , different compounds of one core structure group or compounds of two or more of the core structure groups) can be administered concurrently or sequentially. Uses of the compounds disclosed herein are provided (1) for treating disorders of the prostate and (2) for preparing compositions, formulations and medicaments for treating disorders of the prostate.
These and other aspects of the invention will become apparent to the skilled person in view of the teachings contained herein. BRIEF DESCRIPTION OF THE DRAWINGS
FIGURE 1 graphically depicts an assay employing organotypic spinal motor neurons and threohydroxyaspartate ("THA"). Exposure of neurons with THA alone resulted in death of 55-60% of the neurons. When the neurons were exposed to THA in combination with 10 μM c-KPG, the c-KPG spared greater than 50% of the neurons that would have otherwise been killed.
DETAILED DESCRIPTION OF PREFERRED EMBODIMENTS
The present invention provides DPP IV inhibitors that are useful for treating various disorders, including those of the central nervous system, among others. Preferably, the DPP IV inhibitors are pyrrolidine-based compounds, and more preferably constitute or include proline or proline mimetics. The compounds according to the present invention preferably have sufficient stability, potency, selectivity, solubility and availability to be safe and effective in treating diseases, injuries and other abnormal conditions or insults to the central nervous system, the peripheral nerves and the prostate, for example. The word "treat" in its various grammatical forms as used in relation to the present invention refers to preventing, curing, reversing, attenuating, alleviating, minimizing, suppressing, ameliorating or halting the deleterious effects of a disease state, disease progression, injury, wound, ischemia, disease causative agent (e.g. , bacteria, protozoans, parasites, fungi, viruses, viroids and/or prions), surgical procedure or other abnormal or detrimental condition (all of which are collectively referred to as "disorders," as will be appreciated by the person of skill in the art). A "therapeutically effective amount" of an inhibitor according to the invention is an amount that can achieve effective treatment, and such amounts can be determined in accordance with the present teachings.
As explained above, DPP IV exhibits a preference for causing the removal of proline-containing dipeptides from the N-terminus of a protein or a peptide. Accordingly, proline has a structure that likely is recognized by or acted upon by the active site of DPP IV. Proline is unique among the 20 naturally-occurring amino acids in that it contains a cyclic secondary amino group, which as a result causes it to create interruptions in alpha- helical structures in proteins or peptides. Preferably, the DPP IV inhibitors according to the present invention can constitute or include proline or proline-like moieties, often referred to as "proline mimetics." A proline mimetic is a structure that sufficiently resembles proline such that its charge, polarity, shape and size are sufficiently duplicative of proline so as to participate in many of the molecular interactions involving proline. A molecule or other compound that includes a proline moiety can itself be considered a proline mimetic. Accordingly, molecules that constitute or include proline or proline mimetics can interact with the natural interaction partners of proline, such as DPP IV. Preferably, a DPP IV inhibitor has the same or greater affinity for DPP IV than does the natural substrate of DPP IV, such as a protein containing a proline residue at its N-terminal end. Preferably, the inhibitor will have an equal or greater affinity to permit it to more effectively compete for the active site of DPP IV. Inhibitors with lower affinities, however, are still within the scope of the invention, and effective competition, and thus inhibition, can be ensured through dosing considerations. In accordance with certain aspects of the invention, the DPP IV inhibitor is used to treat disorders of the prostate, including, but not limited to, prostate cancer and post- prostatectomy nerve recovery. For example, erectile and voiding disorders are extremely common clinical conditions that result from diseases, injuries and trauma including complications associated with pelvic surgery. It is believed that local nerve injury during major pelvic surgeries account for complications such as erectile dysfunction and urinary incontinence. These complications might be caused by the trauma or the injury of the nerves (e.g. cavernous nerve) innervating the area during the surgery. Appropriate administration of a DPP IV inhibitors prior to, during or after surgery may be effective in blocking the nerve degeneration caused by pelvic surgery. The inhibitor of the invention can be administered in the manner used with other prostate therapeutics, and can be combined with other products or methodologies for treating the prostate. A therapeutically effective amount of the inhibitor will depend upon its potency and its ability to enter or become available at the site of treatment, in this case the prostate and/or surrounding areas. The considerations for determining proper dose levels are available to the skilled person. See Example 6 below. In accordance with other aspects of the invention, the DPP IV inhibitor can be used to treat disorders of the central nervous system (CNS) and the peripheral nerves. For example, the DPP IV inhibitors according to the present invention can be used to treat CNS maladies such as strokes, tumors, ischemia, Parkinson's disease, memory loss, hearing loss, vision loss, migraines, brain injury, spinal cord injury, Alzheimer's disease and amyotrophic lateral sclerosis (which has a CNS component). Additionally, the DPP IV inhibitors can be used to treat disorders having a more peripheral nature, including multiple sclerosis and diabetic neuropathy.
When treating the CNS, a biological phenomenon known as the "blood-brain barrier" is encountered. The blood-brain barrier prevents many compounds in the circulation from crossing to the brain. The brain is a complex biological structure that is susceptible to a variety of toxins. Additionally, being that the brain is composed primarily of nerves and related tissues, the brain lacks the natural regenerative capabilities of other organs and tissues. For example, the skin has extensive regeneration and restorative capabilities, and thus can withstand encounters with toxins and other physical insults, which it can be expected to encounter in daily life. The brain itself, on the other hand, is quite susceptible to toxins, and thus it is thought that the blood-brain barrier was an evolutionary development to protect the integrity of the brain. The blood-brain barrier, however, also can prevent the entry of beneficial compounds, such as drugs, that are needed to treat a disease, injury or other abnormal condition. Accordingly, the blood-brain barrier can be a complicating factor in developing therapeutics for the CNS.
Compounds, such as molecules, cross the blood-brain barrier by two basic paths, referred to as "passive diffusion" and "active transport." Designing compounds to cross the blood-brain barrier via passive diffusion is somewhat easier than designing compounds to cross via active transport. Assays for evaluating the capability of a compound to cross the blood-brain barrier are disclosed in Boer et al., DRUG TRANSPORT ACROSS THE BLOOD- BRAIN BARRIER, (Harwood Academic Publishers).
Guidelines exist for creating compounds that cross the blood-brain barrier via passive diffusion. Typically, a compound that crosses the blood-brain barrier via passive diffusion should have a log P between about 1 and about 4. Related to this concept is the log D, which takes into consideration the charge of the compound. Typically, polar and charged compounds are less amenable to crossing the blood-brain barrier by passive diffusion. Accordingly, a log D greater than about -2 is preferred. The concepts of log P and log D are discussed in Waterbeemed, STRUCTURAL-PROPERTY CORRELATIONS IN DRUG RESEARCH (Academic Press). To further facilitate passive diffusion, the compound preferably has a molecular weight of about 700 or less, preferably about 500 or less. Thus, a compound that is to cross the blood-brain barrier by passive diffusion should be "sufficiently neutral and non-polar" for its size that it can cross the blood-brain barrier in a therapeutically effective amount
Larger and/or more highly charged and polar compounds also are within the scope of the present inventions. Typically, these compounds do not cross the blood-brain barrier via passive diffusion, but rather cross the barrier via active transport. There are guidelines for developing compound that will cross via active transport. Additionally, administration modalities, delivery vehicles and other formulation considerations can assist compounds according to the invention in crossing the blood-brain barrier. See, for example, U.S. Patent No. 5,874,449.
Besides efficiency of a compound in crossing the blood-brain barrier, another important consideration is the potency of the compound as an inhibitor. For example, potent inhibitors can have a lower efficiency in crossing the blood-brain barrier, but nevertheless can be effective due to their higher potencies. Conversely, a less potent inhibitor may require greater efficiency in crossing the blood-brain barrier in order to have a beneficial effect. Thus, a therapeutically effective amount for treating a CNS disorder depends upon the potency of the inhibitor and its efficiency in crossing the blood-brain barrier or the administration route and approach employed to circumvent the blood-brain barrier. In terms of potencies, the DPP IV inhibitors preferably have an IC50 (for inhibition concentration where 50% of DPP IV is inhibited) value of less than about 1 μm, and preferably less than 100 ran. Of course, DPP IV inhibitors can have higher IC50 values as long as their efficiency in crossing the blood-brain barrier is sufficient to treat the disease, injury or other abnormal condition. It is preferred that the DPP IV inhibitor according to the invention is a reversible inhibitor. That is, the DPP IV inhibitor should be able to interact with the inhibitor without becoming permanently bound thereto in a manner that would denature or inactivate the DPP IV enzyme. The need for reversibility is due to the fact that DPP IV is a naturally- occurring enzyme that has normal physiologic functions. An irreversible inhibitor can effectively eliminate functions of the enzyme, and thus result in cessation of normal physiologic processes. The present invention utilizes the inhibition of DPP IV in certain contexts, such as in treating an ischemic event, for definite periods of time, such as during and after reperfusion in the ischemic area. A reversible inhibitor would permit inhibited DPP IV molecules to resume normal function once the need for inhibition is gone.
Administration Routes and Formulations For treating the CNS, the compounds according to the invention can be administered by a variety of systemic and CNS-targeted routes. For example, intra-arterial, intravenous intraventricular, intracavitary and intracranial administration routes can be employed. Exemplary injection modalities can be by way of bolus, periodic injection and/or constant infusion. Depending upon the circumstance, the following routes can be employed for the compounds according to the invention, including parenteral, oral, nasal, inhalation spray, buccally, topically, transdermal, rectal, vaginal, via implanted reservoir or other routes available to the skilled person. The term parenteral as used herein includes subcutaneous, intravenous, intramuscular, intraperitoneal, intrathecal, intraventricular, intrasternal, intracranial or intraosseous injection and infusion techniques.
To be maximally effective as a therapeutic for central nervous system disorders, the compounds of the present invention preferably penetrate the blood-train barrier when peripherally administered. Compounds which cannot sufficiently penetrate the blood-brain barrier can be effectively administered by an intraventricular route. It also is important to note that during the active phase of certain CNS disorders, blood-brain lineage is known to occur and will permit entry of the compounds of the invention to the central nervous system. Moreover, there are several other techniques that either physically break through the blood-brain barrier or circumvent it to deliver therapeutic agents. Examples of these techniques include intrathecal injections, surgical implants, and osmotic techniques. Invasive techniques often are employed, particularly direct administration to damaged neuronal tissue. One or more of the above can be employed according to the invention. One embodiment for the administration of the compounds of the invention is by intrathecal injection, i.e., directly into the cerebrospinal fluid by puncturing the membranes surrounding the central nervous system is usually by lumbar puncture. Sustained dosages of agents directly into the cerebrospinal fluid can be attained by the use of infusion pumps that are implanted surgically.
Another embodiment for the administration of the compounds of the invention is by injection directly into the lumbar cerebrospinal fluid (intrathecally) or by injection intravenously.
The compounds according to the invention can be formulated with pharmaceutically- acceptable carriers and diluents, and can be used with methods and uses according to the invention. The formulation will depend upon the disease state being treated and the administration route. See, for example, U.S. Patent No. 5,874,449, which is incorporated by reference. Pharmaceutically acceptable carriers include aqueous solutions, non-toxic excipients, including salts, preservatives, buffers, such as phosphate buffers, and the like, as described in UNITED STATES PHARMACOPEIA AND NATIONAL FORMULARY (USP 24-NF 19); REMINGTON'S PHARMACEUTICAL SCIENCES; HANDBOOK ON PHARMACEUTICAL EXCIPIENTS (2d edition, Wade and Weller eds. 1994), the each of which are hereby incorporated by reference. Examples of non-aqueous solvents are propylene glycol, polyethylene glycol, vegetable oil and injectable organic esters such as ethyloleate. Aqueous carriers include water, alcoholic/aqueous solutions, saline solutions, parenteral vehicles, such as sodium chloride and Ringer's dextrose. Intravenous vehicles include fluid and nutrient replenishers. Preservatives include antimicrobials, anti-oxidants, chelating agents and inert gases. The pH and exact concentration of the various components of the binding composition are adjusted according to routine skills in the art. See GOODMAN AND GILMAN'S THE PHARMACOLOGICAL BASIS FOR THERAPEUTICS (9th edition), the contents of which are hereby incorporated by reference.
Exemplary approaches include those where the compounds are to be administered in the form of sterile injectable preparations, for example, as sterile injectable aqueous or oleaginous suspensions. These suspensions can be formulated according to techniques known in the art using suitable dispersing or wetting agents and suspending agents. The sterile injectable preparations may also be sterile injectable solutions or suspensions in non- toxic parenterally-acceptable diluents or solvents, for example, as solutions in 1,3- butanediol. Among the acceptable vehicles and solvents that may be employed are water, Ringer's solution and isotonic sodium chloride solution. In addition, sterile fixed oils are conventionally employed as solvents or suspending mediums. For this purpose, any blank fixed oil such as a synthetic mono- or di-glyceride may be employed. Fatty acids such a oleic acid and its glyceride derivatives, including olive oil and castor oil, especially in their polyoxyethylated forms, are useful in the preparation of injectables. These oil solutions or suspensions may also contain long-chain alcohol diluents or dispersants. Additionally, the compounds may be administered orally in the form of capsules, tablets, aqueous suspensions or solutions; Tablets may contain carriers such as lactose and corn starch, and/or lubricating agents such as magnesium stearate. Capsules may contain diluents including lactose and dried corn starch. Aqueous suspensions may contain emulsifying and suspending agents combined with the active ingredient. The oral dosage forms may further contain sweetening and/or flavoring and/or coloring agents.
The compounds may further be administered rectally in the form of suppositories. These compositions can be prepared by mixing the drug with suitable non-irritating excipients which are solid at room temperature, but liquid at rectal temperature such that they will melt in the rectum to release the drug. Such excipients include cocoa butter, beeswax and polyethylene glycols.
Moreover, the compounds may be administered topically, especially when the conditions addressed for treatment involve areas or organs readily accessible by topical application, including neurological disorders of the eye, the skin or the lower intestinal tract. For topical application to the eye, or ophthalmic use, the compounds can be formulated as micronized suspensions in isotonic, pH adjusted sterile saline or, preferably, as a solution in isotonic, pH adjusted sterile saline, either with or without a preservative such as benzylalkonium chloride. Alternatively, the compounds may be formulated into ointments, such as petrolatum. For topical application to the skin, the compounds can be formulated into suitable ointments containing the compounds suspended or dissolved in, for example, mixtures with one or more of the following: mineral oil, liquid petrolatum, white petrolatum, propylene glycol, polyoxyethylene polyoxypropylene compound, emulsifying wax and water. Alternatively, the compounds can be formulated into suitable lotions or creams containing the active compound suspended or dissolved in, for example, a mixture of one or more of the following: mineral oil, sorbitan monostearate, polysorbate 60, cetyl ester wax, cetearyl alcohol, 2-octyldodecanol, benzyl alcohol and water.
Dosing The compounds of the present invention may be administered by a single dose, multiple discrete doses or continuous infusion. Because the compounds preferably are small, easily diffusible and relatively stable, they can be well-suited to continuous infusion. Dose levels on the order of about 0.1 mg to about 10,000 mg of the active ingredient are useful in the treatment of the above conditions, with preferred levels being about 0.1 mg to about 1,000 mg. The specific dose level, and thus the therapeutically- effective amount, for any particular patient will vary depending upon a variety of factors, including the activity of the specific compound employed and its bioavailability at the site of drug action; the age, body weight, general health, sex and diet of the patient; the time of administration; the rate of excretion; drug combination; the severity of the particular disease being treated; and the form of administration. Typically, in vitro dosage-effect results provide useful guidance on the proper doses for patient administration. Studies in animal models also are helpful. The considerations for determining the proper dose levels are available to the skilled person. See Example 5 below.
Certain compounds can administered in lyophilized form. In this case, 1 to 100 mg of a compound of the present invention may be lyophilized in individual vials, together with a carrier and a buffer, such as mannitol and sodium phospshate. The compound may be reconstituted in the vials with bacteriostatic water before administration.
In treating CNS disorders resulting from global ischemia, for example, the compounds of the present invention are preferably administered orally, rectally, parenterally or topically at least 1 to 6 times daily, and may follow an initial bolus dose of higher concentration. Administration Regimen and Timing
For the compounds methods and uses of the present invention, any administration regimen regulating the timing and sequence of drug delivery can be used and repeated as necessary to effect treatment. Such regimen may include pretreatment and/or co- administration with additional therapeutic agents.
To maximize protection of nervous tissue from nervous insult, the compounds should be administered to the affected cells as soon as possible. In situations where nervous insult is anticipated, the compounds should be administered before the expected nervous insult. Such simations of increased likelihood of nervous insult include surgery (for example, carotid endarterectomy, cardiac, vascular, aortic, orthopedic); endovascular procedures such as arterial catherization (for example, carotid, vertebral, aortic, cardia, renal, spinal, Adamkiewicz); injections of embolic agents; coils or balloons for hemostasis; interruptions of vascularity for treatment of brain lesions; and predisposing medical conditions such as crescendo transient ischemic attacks, emboli and sequential strokes.
Where pretreatment for stroke or ischemia is impossible or impracticable, it is important to get the compounds to the affected cells as soon as possible during or after the event. In the time period between strokes, diagnosis and treatment procedures should be minimize to save the cells from further damage and death. It is clear that both in animal models of stroke and in humans, the effect of cerebral ischemia are manifest on the cerebral metabolism rapidly, with a time scale measured in minutes or hours. Any form of potential neuroprotective treatment should therefore be given by the most rapidly effective route, which in practice usually means intravenously. The optimal duration and route of administration of treatment will depend on the individual pharmacokinetic properties of the neuroprotective compound, on the adverse-effect profile of the drug, and on the nature of the insult that gave rise to the stroke. Excitotoxic injury following stroke evolves over at least 4 hours in rodents and possibly 48 hours in humans. Dyker et al, Stroke 29: 535-42 (1998). Thus, it would be desirable to provide neuroprotection throughout this critical time period. Ideally, any compound for the treatment of stroke should adequately cross the blood-brain barrier and obtain sufficiently therapeutic levels within the brain and cerebral spinal fluid. For patients with prostate cancer that is neither advanced nor metastatic, the compounds of the present invention may be administered (i) prior to surgery or radiation treatment to reduce the risk of metastasis; (ii) during surgery or in conjunction with radiation treatment; and/or (iii) after surgery or radiation therapy to reduce the risk of recurrence and to inhibit the growth of any residual tumorous cells.
For patients with advanced or metastatic prostate cancer, the compounds of the present invention may be administered as a continuous supplement to, or as a replacement for, homonal ablation in order to slow tumor cell growth in both the untreated primary tumor and the existing metastatic lesions. The compounds, methods and uses of the present invention are particularly useful where shed cells could not be removed by surgical intervention. After post-surgical recovery, the compounds, methods and uses of the present invention would be effective in reducing the chances of recurrence of a tumor engendered by such shed cells.
Combination with Other Treatments
The compounds, methods and uses of the present invention also to provide combined preparation for simultaneous, separate, or sequential use which contain other biologically active agents. Such biologically active agent can be either another compound of the present invention; steroids, for example hydrocortisomers such as methylprednisolone; anti- inflammatory or anti-immune drugs, such as methotrexate, azathioprine, cyclophosphamide or cyclosporin A; interferon-β; antibodies, such as anti-CD4 antibodies; agents which can reduce the risk of a second ischemic event, such as ticlopidine; chemotherapeutic compositions; immunotherapeutic compositions; morphine for treating pain; or mixtures thereof.
The compounds according to the invention include various substitutions available to the skilled person and are to be employed in accordance with the teachings contained herein. For example, the Core Structures, which constitute or include proline mimetics, can include a variety of functional groups as taught herein. Additionally, the inventions include isosteres of the compounds or the function groups contained therein. Guiding principles and illustrative examples of functional groups and isosteres are set forth in Smith et al. , INTRODUCTION TO THE PRINCIPLES OF DRUG DESIGN (John Wright & Sons, Ltd.), which is hereby incorporated by reference.
The compounds used according to the invention preferably are or contain moieties that resemble proline within their core strucmres. That is, these compounds are or contain proline mimetics. One such compound that can be used according to the invention contains a proline mimetic and has the following structure:
Figure imgf000016_0001
c-KPG This compound, referred to as "c-KPG," was tested in an assay employing organotypic spinal motor neurons and threohydroxyaspartate ("THA"), which is an inhibitor of the glutamate reuptake receptor. Synthesis protocols for c-KPG are disclosed in Nguyen et al., J. Med. Chem. 41: 2100-10 (1998).
As shown in Figure 1, exposure of neurons with THA alone resulted in death of 55- 60% of the neurons. Exposure of the neurons to THA in combination with 10 μM c-KPG (A DPP IV inhibitor), the c-KPG spared greater than 50% of the neurons that would have otherwise been killed. The results were highly significant (p == 0.004).
The invention includes other core structures as well. Core structures, which are DPP IV inhibitors and constitute or contain proline mimetics, are set forth below. Exemplary core structures are depicted schematically, and the functional/substitution groups are set forth in text. All substitutions contemplated herein are permissive for various provisos, either alone or in any combination, such that if one group is included in a given position another group at the same or different position can be excluded. For example, Core Structure I is:
(D
Figure imgf000017_0001
, which can be modified as set forth below, with the following numerically-identified optional provisos, which can be employed alone or in any combination:
X is CR2R3, O, S, or NR4; optional proviso 1 that if X is S, then A cannot be CN; optional proviso 2 that if X is CH2 and R is H, then A cannot be C; optional proviso 3 that if X is S, then Rl cannot be amino-substimted alkyl; optional proviso 4 that if X is CH2 , then A cannot be COOH; optional proviso 5 that if X is S, or if X and XI are both CH2, and Z is O, and A is CN, and Rl is H, then R is not NH substituted with C1-C9 straight or branched chain alkyl, or NH substituted with C3-C7 cycloalkyl;
X, is CR2R3, O, S, or NR4 with optional proviso 6 that X and XI cannot both be a heteroatom; optional proviso 7 if X and XI are both CH2, and Z is O, and Rl is NH2, then R is not 1-methylpropyl if A is COOH, and R is not cyclopentyl if A is CN.
A is H, COOH, or isosteres of carboxylic acids, such as one selected from the group consisting of CN, SO3H, CONOH, PO3R5R6, SO2NHR7, tetrazole, amides, esters, and acid anhydrides with optional proviso 8 that if A is CN, and Rl is NH2, and Z is O, and R is 1-methylpropyl, then X and XI are not both CH2, X and XI are not S, X is not O, and Z is O or S;
R and Rl are independently selected from the group of functional groups consisting of H, CrC9 branched or straight chain alkyl, C2-C9 branched or straight chain alkenyl, C3- C8 cycloalkyl, C5-C7 cycloalkenyl, aryl, heteroaryl and amino, wherein any of the functional groups can be substituted with one or more of CrC9 straight or branched chain alkyl, aryl, heteroaryl, amino, halo, carbonyl, CrC9 alkoxy, C2-C9 alkenyloxy, phenoxy, benzyloxy, C3-C8 cycloalkyl, cyano, amido, thiol, trifluromethyl, or hydroxy, wherein each of R and Rl can be the same or different; and
R2, R3, R4, R5, R6 and R7, if present, are independently selected from the group of functional groups consisting of H, CrC9 branched or straight chain alkyl, C2-C9 branched or straight chain alkenyl, C3-C8 cycloalkyl, C5-C7 cycloalkenyl, aryl, heteroaryl and amino, wherein any of the functional groups can be substituted with one or more of - C9 straight or branched chain alkyl, aryl, heteroaryl, amino, halo, carbonyl, Ct-C9 alkoxy, C2-C9 alkenyloxy, phenoxy, benzyloxy, C3-C8 cycloalkyl, cyano, amido, thiol, trifluromethyl, or hydroxy, wherein each of R2, R3, R4, R5, R6 and R7, if present, can be the same or different.
Other core strucmres are provide according to the invention, such as those having ring modifications (II and HI): .
OD
Figure imgf000018_0001
, which can be modified as follows: X is CR2R3, O, S, or NR4; A is H, COOH, or isosteres of carboxylic acids, such as one selected from the group consisting of CN, SO3H, CONOH, PO3R5R6, SO2NHR7, tetrazole, amides, esters, and acid anhydrides;
Z is O or S; R and Rl are independently selected from the group of functional groups consisting of H, C,-C9 branched or straight chain alkyl, C2-C9 branched or straight chain alkenyl, C3- C8 cycloalkyl, C5-C7 cycloalkenyl, aryl, heteroaryl and amino, wherein any of the functional groups can be substituted with one or more of CrC9 straight or branched chain alkyl, aryl, heteroaryl, amino, halo, carbonyl, CrC9 alkoxy, C2-C9 alkenyloxy, phenoxy, benzyloxy, C3-C8 cycloalkyl, cyano, amido, thiol, trifluromethyl, or hydroxy, wherein each of R and Rl can be the same or different; and
R2, R3, R4, R5, R6 and R7, if present, are independently selected from the group of functional groups consisting of H, C,-C9 branched or straight chain alkyl, C2-C9 branched or straight chain alkenyl, C3-C8 cycloalkyl, C5-C7 cycloalkenyl, aryl, heteroaryl and amino, wherein any of the functional groups can be substituted with one or more of Cr C9 straight or branched chain alkyl, aryl, heteroaryl, amino, halo, carbonyl, C,-C9 alkoxy, C2-C9 alkenyloxy, phenoxy, benzyloxy, C3-C8 cycloalkyl, cyano, amido, thiol, trifluromethyl, or hydroxy, wherein each of R2, R3, R4, R5, R6 and R7, if present, can be the same or different. Core Structure in is:
(ΠD
Figure imgf000019_0001
which can be modified as follows: A is H, COOH, or isosteres of carboxylic acids, such as one selected from the group consisting of CN, SO3H, CONOH, PO3R5R6, SO2NHR7, tetrazole, amides, esters, and acid anhydrides;
Z is O or S; R, Rl, R2 and R3 are independently selected from the group of functional groups consisting of H, CrC9 branched or straight chain alkyl, C2-C9 branched or straight chain alkenyl, C3-C8 cycloalkyl, C5-C7 cycloalkenyl, aryl, heteroaryl and amino, wherein any of the functional groups can be substituted with one or more of C C9 straight or branched chain alkyl, aryl, heteroaryl, amino, halo, carbonyl, CrC9 alkoxy, C2-C9 alkenyloxy, phenoxy, benzyloxy, C3-C8 cycloalkyl, cyano, amido, thiol, trifluromethyl, or hydroxy, wherein each of R, Rl, R2 and R3 can be the same or different; and
R4, R5, R6 and R7, if present, are independently selected from the group of functional groups consisting of H, C,-C9 branched or straight chain alkyl, C2-C9 branched or straight chain alkenyl, C3-C8 cycloalkyl, C5-C7 cycloalkenyl, aryl, heteroaryl and amino, wherein any of the functional groups can be substimted with one or more of CrC9 straight or branched chain alkyl, aryl, heteroaryl, amino, halo, carbonyl, CrC9 alkoxy, C2-C9 alkenyloxy, phenoxy, benzyloxy, C3-C8 cycloalkyl, cyano, amido, thiol, trifluromethyl, or hydroxy, wherein each of R4, R5, R6 and R7, if present, can be the same or different.
Other compounds according to the invention include amid bond isosteres, such as Core Structure IV. Core Structure IV is:
(IV)
Figure imgf000020_0001
, which can be modified as follows: X is CR2R3, O, S, or NR4;
X, is CR2R3, O, S, or NR4 with the proviso that X and XI cannot both be a heteroatom;
A is H, COOH, or isosteres of carboxylic acids, such as one selected from the group consisting of CN, SO3H, CONOH, PO3R5R6, SO2NHR7, tetrazole, amides, esters, and acid anhydrides;
R and Rl are independently selected from the group of functional groups consisting of H, CrC9 branched or straight chain alkyl, C2-C9 branched or straight chain alkenyl, C3- C8 cycloalkyl, C5-C7 cycloalkenyl, aryl, heteroaryl and amino, wherein any of the functional groups can be substituted with one or more of CrC9 straight or branched chain alkyl, aryl, heteroaryl, amino, halo, carbonyl, C C9 alkoxy, C2-C9 alkenyloxy, phenoxy, benzyloxy, C3-C8 cycloalkyl, cyano, amido, thiol, trifluromethyl, or hydroxy, wherein each of R and Rl can be the same or different; and
R2, R3, R4, R5, R6 and R7, if present, are independently selected from the group of functional groups consisting of H, CrC9 branched or straight chain alkyl, C2-C9 branched or straight chain alkenyl, C3-C8 cycloalkyl, C5-C7 cycloalkenyl, aryl, heteroaryl and amino, wherein any of the functional groups can be substituted with one or more of Cr C9 straight or branched chain alkyl, aryl, heteroaryl, amino, halo, carbonyl, CrC9 alkoxy, C2-C9 alkenyloxy, phenoxy, benzyloxy, C3-C8 cycloalkyl, cyano, amido, thiol, trifluromethyl, or hydroxy, wherein each of R2, R3, R4, R5, R6 and R7, if present, can be the same or different.
The compounds of the core structures according to the present invention can administered in ester or salt forms according to the teachings provided herein. Acceptable formulations, dosages and administration regimens can be determined in accordance with the teachings contained herein.
The invention is further described by the following examples, which are illustrative of the invention but do not limit the invention in any manner. EXAMPLE 1: SYNTHESIS OF COMPOUNDS ACCORDING TO CORE STRUCTURE I
Compounds according to Core Structure I can be produced according to a variety of approaches. Representative approaches are shown below:
Figure imgf000022_0001
Figure imgf000022_0002
4
Figure imgf000022_0003
5 Other approaches include:
H2/Pd/C
Figure imgf000022_0005
Figure imgf000022_0004
Figure imgf000023_0001
See, for example, Org. Lett. 1: 31-33 (1999).
Substiments can be placed on the ring by modification of starting materials as shown below:
Figure imgf000023_0002
Compounds containing sulfur in place of oxygen can be prepared following standard procedures, as shown below:
Figure imgf000023_0003
Further transformations can be performed by:
Lawβsson's Reagent
Figure imgf000023_0004
Figure imgf000023_0005
Other exemplary compounds are set forth below. Compound 1
Figure imgf000024_0001
4-oxazolidinecarboxylic acid, 3-[(ethylaminoacetyll-
Principal Group: Functionalized Hydride: carboxylic acid 4-oxazolidinecarboxylic acid Parent Hydride: Substituents:
Oxazolidine 3 acetyl amino ethyl
Compound 2
Figure imgf000024_0002
4-oxazolidinecarboxylic acid. 3-f2-(ethylamino)-3-methyl-1 -oxopentyll-
Principal Group: Functionalized Hydride: carboxylic acid 4-oxazolidinecarboxylic acid Parent Hydride: Substituents:
Oxazolidine 3 pentyl
2 amino ethyl
3 methyl 1 oxo Compound 3
Figure imgf000025_0001
4-oxazolidinecarboxylic acid. 3-f3-methyl- 1 -oxo-2-(phenylamino)pentvn-
Principal Group: Functionalized Hydride: carboxylic acid 4-oxazolidinecarboxylic acid Parent Hydride: Substituents:
Oxazolidine 3 pentyl 3 methyl
1 oxo
2 amino phenyl <-
Compound 4
Figure imgf000025_0002
4-thiazolidinecarboxylic acid, 3-f3-methyl-1 -oxo-2-(phenylamino)pentyll-
Principal Group: Functionalized Hydride: carboxylic acid 4-thiazolidinecarboxylic acid Parent Hydride: Substituents: thiazolidine 3 pentyl 3 methyl
1 oxo
2 amino phenyl Compound 5
Figure imgf000026_0001
2-thiazolidinecarboxylic acid. 3-f3-methyl- 1 -oxo-2-(phenylamino)pentvn-
Principal Group: Functionalized Hydride: carboxylic acid 2-thiazolidinecarboxylic acid Parent Hydride: Substituents: thiazolidine 3 pentyl 3 methyl
1 oxo
2 amino phenyl
Compound 6
Figure imgf000026_0002
2-oxazolidinecarboxylic acid. 3-f 3-methyl- 1 -oxo-2-(phenylamino)pentvH-
Principal Group: Functionalized Hydride: carboxylic acid 2-oxazolidinecarboxylic acid Parent Hydride: Substituents:
Oxazolidine 3 pentyl 3 methyl
1 oxo
2 amino phenyl Compound 7
Figure imgf000027_0001
3-oxazolidineethanamine. α-( ■ -methylpropyl)-β-oxo-N-phenyl-4-(2 -tetrazol-5-yl)-
Principal Group: Substituents: amine α propyl Conjunctive Parent: 1 methyl
3-oxazolidineethanamine β oxo
N phenyl
4 2/Y-tetrazol-5-yl
Compound 8
Figure imgf000027_0002
3-oxazolidineethanamine. β-oxo-N-phenyl-4-(2A -tetrazol-5-yl)-
Principal Group: Substituents: amine β oxo Conjunctive Parent: N phenyl
3-oxazolidineethanamine 4 2/y-tetrazol-5-yl Compound 9
Figure imgf000028_0001
3-thiazolidineethanamine. β-oxo-N-phenyl-4-(2/ -tetrazol-5-yl)- Principal Group: Substituents: amine β oxo Conjunctive Parent: N phenyl
3-thiazolidineethanamine 4 2//-tetrazol-5-yl Compound 10
Figure imgf000028_0002
3-thiazolidineethanamine. α-(2-methylpropyl)-β-oxo-N-phenyl-4-(2A -tetrazol-5-yl)-
Principal Group: Substituents: amine β oxo Conjunctive Parent: N phenyl
3-thiazolidineethanamine 4 2tf-tetrazol-5-yl α propyl
2 methyl Compound 1 1
Figure imgf000029_0001
3-thiazolidineethanamine. /V-ethyl-α-(2-methylpropyl)-β-oxo-2-(2A -tetrazol-5-yl)-
Principal Group: Substituents: amine β oxo Conjunctive Parent: N ethyl
3-thiazolidineethanamine 2 2#-tetrazol-5-yl α propyl
2 methyl
Compound 12
Figure imgf000029_0002
3-oxazolidineethanamine. N-ethyl-α-(2-methylpropyl)-β-oxo-2-(2 y-tetrazol-5-yl)-
Principal Group: Substituents: amine β oxo Conjunctive Parent: N ethyl
3-oxazolidineethanamine 2 2 -tetrazol-5-yl α propyl
2 methyl Compound 13
Figure imgf000030_0001
3-oxazolidineethanamine. N-ethyl-o.-(2-methylpropy|)-2-(2//-tetrazol-5-yl)-β-thioxo-
Principal Group: Substituents: amine β thioxo Conjunctive Parent: N ethyl
3-oxazolidineethanamine 2 2 V-tetrazol-5-yl α propyl
2 methyl
Compound 14
Figure imgf000030_0002
1 -imidazolidineethanamine. N-ethyl-α-(2-methylpropyl)-5-(2 -tetrazol-5-yl)-β-thioxo-
Principal Group: Substituents: amine β thioxo Conjunctive Parent: N ethyl
1 -imidazolidineethanamine 5 2A -tetrazol-5-yl α propyl
2 methyl Compound 15
Figure imgf000031_0001
1 -imidazolidineethanamine, N-ethyl-α-(2-methylpropyl)-β-oxo-5-(2/V-tetrazol-5-yl)
Principal Group: Substituents: amine β oxo Conjunctive Parent: N ethyl
1 -imidazolidineethanamine 5 2 V-tetrazol-5-yl a propyl
2 methyl
Compound 16
Figure imgf000031_0002
1-imidazolidineethanamine, α-(2-methylpropyl)-β-oxo-N-phenyl-5-(2A/-tetrazol-5-yl)
Principal Group: Substituents: amine β oxo Conjunctive Parent: N phenyl
1 -imidazolidineethanamine 5 2 V-tetrazol-5-yl α propyl
2 methyl Compound 1 7
Figure imgf000032_0001
4-imidazolidinecarboxylic acid. 3-.4-methyl-1 -oxo-2-(phenylamino)pentvπ-
Principal Group: Substituents:
Carboxylic acid 3 pentyl
Parent Hvdrid: 4 methyl
Imidazolidine 1 oxo
Functionalized Hydride: 2 amino
4-imidazolidinecarboxylic acid phenyl
Compound 18
Figure imgf000032_0002
4-imidazolidinecarboxylic acid, 3-[4-metriyl-2-(phenylamino)-1 -thioxopentyll-
Principal Group: Substituents:
Carboxylic acid 3 pentyl
Parent Hvdrid: 4 methyl
Imidazolidine 2 amino
Functionalized Hydride: 1 thioxo
4-imidazolidinecarboxylic acid phenyl * # * * #
Other compounds for use according to the invention include:
Compound 19: 1 -[2-[(5-chloropyridin-2-yl)amino]ethylamino]acetyl-2-cyano- (S)-pyrrolidine dihydrochloride
Compound 20: 1 -[2-[(5-trifluoromethylpyridin-2-yl)amino]ethylamino]acetyl- 2-cyano-(S)- pyrrolidine
Compound 21 1 -[2-[(5-cyanopyridin-2-yl)amino]ethylamino]acetyl-2-cyano-
(S)-pyrrolidine dihydrochloride
Compound 22: 1 -[2-[(pyrimidin-2-yl)amino]ethylamino]acetyl-2-cyano-(S)- pyrrolidine
Compound 23: 1 -[(1 -hydroxymethylcyclopent-1 -yl)amino]acetyl-2-cyano-(S)- pyrrolidine
Compound 24: 1 -_2-[(pyridin-2-yl)amino]ethylamino]acetyl-2-cyano-(S)- pyrrolidine
Compound 25: 1 -[2-[(4-chloropyrimidin-2-yl)amino]ethylamino]acetyl-2- cyano-(S)-pyrrolidine
Compound 26: 1 -[2-[(3-chloropyridin-2-yl)amino]ethylamino_acetyl-2-cyano- (S)-pγrrolidine
Compound 27: 1 -[2-_4-trifluoromethylpyrimidin-2-yl)amino]ethylamino]acetyl- 2-cyano-(S)- pyrrolidine
Compound 28: 1 -[(2-chlorophenyl)ethylamino]acetyl-2-cyano-(S)-pyrrolidine
Compound 29: 1 -[(3,3-diphenyl)propylamino]acetyl-2-cyano-(S)-pyrrolidine Compound 30: 1 -[2-[(5-nitropyridin-2-yl)amino]ethylamino]acetyl-2-cyano- (S)pyrrolidine
Compound 31 : 1 1 -[2-[(3-chloro-5-trifluoromethylpyridin-2- yl)amino]ethylamino]acetyl-2-cyano-( S)-pyrrolidine
Compound 32: 1 1-[2-[(3-trifluoromethylpyridin-2-yl)amino]ethylamino]acetyl- 2-cyano-(S) pyrrolidine
Compound 33: 1 1 -[2-[(3,5-dichloropyridin-2-yl)amino]ethylamino]acetyl-2- cyano-(S)-pyrrolidine
Compound 34: 1 1 -[(cyclopent-1 -yl)amino-acetyl-2-cyano-(S)-pyrrolidine monohydrochloride
Compound 35: 1 1 -[2-(2-bromo-4,5-dimethoxyphenyl)ethylamino]acetyl-2- cyano-(S)-pyrrolidine
Compound 36: 1 1 -[3-(isopropoxy)propylamino]acetyl-2-cyano-(S)-pyrrolidine monohydrochloride
Compound 37: 1 1 -[(2-hydroxy-1 ,1 -dimethylethylamino)]acetyl-2-cyano-(S)- pyrrolidine monohydrochloride
Compound 38: 1 1 -[3-(2-oxo-pyrrolidin-1-yl)propylamino]acetyl-2-cyano-(S)- pyrrolidine monohydrochloride
Compound 39: 3-[(cyclohexyl)amino]acetyl-4-cyano-R-thiazolidine monohydrochloride Compound 40: 3-[(3-isopropoxypropyl)amino]acetyl-4-cyano-(R)-thiazolidine monohydrochloride
Compound 41 : 3-[(isopropyl)amino]acetyl-4-cyano-(R)-thiazolidine monohydrochloride
Compound 42
Figure imgf000035_0001
"[( -hydroxymethylcyclohexyl)amino]acetyl-2-cyano-(S)-pyrrolidine
Compound 43
Figure imgf000035_0002
Pyrrolidine, l-[[2-(4-ethoxyphenyl)ethyl]amino]acetyl-2-cyano-,(S)-,monohydrochloride
Compound 44
Figure imgf000035_0003
Pyrrolidine, 1 -[(1 -phenylmethyl-3-prrolidinyl)amino]acetyl-2-cyano-,(S)-(R)-,dihydrochloride
Compound 45
Figure imgf000036_0001
Pyrrolidine, l-[[2-(4-memoxyphenyl)ethyl]amino]acetyl-2-cyano-,(S)-,monohydrochloride
Compound 46
Figure imgf000036_0002
Pyrrolidine, l-[[2-(3-methoxyphenyl)ethyl]amino]acetyl-2-cyano-,(S)-,monohydrochloride
Compound 47
Figure imgf000036_0003
Pyrrolidine, 1 -[[( 1 -naphthalenyl)methyl]amino]acetyl-2-cyano-(S)-,monohydrochloride
Compound 48
Figure imgf000036_0004
Pyrrolidine, l-[(3-phenylpropyl)amino]acetyl-2-cyano-,(S)-,monohydrochloride
Compound 49 2HC1
Figure imgf000037_0001
Pyrrolidine, l-[[3-[(pheny
Compound 50
Figure imgf000037_0002
Pyrrolidine, l-[2-[(3,4-dimethoxyphenyl)ethyl]amino]acetyl-2-cyano-,(S)-,monohydrochloride
Compound 51
Figure imgf000037_0003
Pyrrolidine, 1 -(acycloheptylamino)acetyl-2-cyano-,(S)-,monohydrochloride
Compound 52
Figure imgf000037_0004
Pyrrolidine, 1 -[[(6,6-dimethylbicyclo[3.1.1 ]hept-2-yl)methyl] amino] acetyl-2-cyano-[lS[lα,2α(S*),5α]]-(S)-,monohydrochloride
Compound 53
Figure imgf000038_0001
Pyrrolidine, l-[[2-(2,5-dimethoxyphenyl)ethyl]amino]acetyl-2-cyano-,(S)-,monohydrochloride
Compound 54
Figure imgf000038_0002
Pyrrolidine, 1 -[[2-(l -cyclohexen- 1 -yl)ethyl]amino]acetyl-2-cyano-,(S)-,monohydrochloride
Compound 55
Figure imgf000038_0003
Pyrrolidine, l-(cyclohexylamino)acetyl-2-cyano-,(S)-,monohydrochloride
Compound 56
Figure imgf000038_0004
Pyrrolidine, 1 -[(bicyclo[2.2.1 ]hept-2-yl)amino]acetyl-2- cyano-[lS[lα,2α(S*),5α]]-(S)-,monohydrochloride
Compound 57 2HC1
Figure imgf000039_0001
Pyrrolidine, l-[[2-(2-pyridinyl)ethyl]amino]acetyl-2-cyano-,(S)-,dihydrochloride
Compound 58
Figure imgf000039_0002
Pyrrolidine, 1 -[[(2-phenylamino)ethyl]amino]acetyl-2-cyano-,(S)-,dihydrochloride
Compound 59
HC1
Figure imgf000039_0003
Pyrrolidine, l-[(3,3-dimethylbutyl)amino]acetyl-2-cyano-,(S)-,monohydrochloride
Compound 60
HC1
Figure imgf000039_0004
Pyrrolidine, 1 -[(2,6,6-trimethylbicyclo[3.1. l]hept-3-yl)amino]acetyl-2-cyano-, (S)[lS[lα,2β,3α(S*),5α]]-monohydrochloride Compound 61
Figure imgf000040_0001
Pyrrolidine, 1 [[(l-hydroxymethyl)propyl]amino]acetyl-2-cyano-[S,S)]-
Compound 62
HC1
Figure imgf000040_0002
Pyrrolidine, l-[[[2-[(2-hydroxymethyl)phenyl]thio]phenylmethyl]amino]acetyl-2-cyano-,(S)- , monohydrochloride
Compound 63
HC1
Figure imgf000040_0003
Pyrrolidine, l-[[2-(2-methoxyphenyl)ethyl]amino]acetyl-2-cyano-,(S)- .monohydrochloride
Compound 64
HC1
Figure imgf000040_0004
Pyrrolidine, l-[(5-hydroxypentyl)amino]acetyl-2-cyano-,(S)-,monohydrochloride Compound 65
HC1
Figure imgf000041_0001
Pyrrolidine, l-(cyclobutylamino)acetyl-2-cyano-,(S)-monohydrochloride Compound 66
HC1
Figure imgf000041_0002
Pyrrolidine, l-[[2-(2,4-dicUorophenyl)ethyl]amino]acetyl-2-cyano-,(S),monohydrochloride
Compound 67
HC1
Figure imgf000041_0003
Pyrrolidine , 1 -[( 1 -hydroxymethyl]-3-methy lbutyl)amino] acetyl-2-cyano- , O- , (S)- ,
Compound 68
HC1
Figure imgf000041_0004
Pyrrolidine, l-[(2-hydroxy-2-phenylethyl)amino]acetyl-2-cyano-[2S-[lR*,2S*]- monohydrochloride Compound 69
HC1
Figure imgf000042_0001
Pyrrolidine, l-[[2-(2-fluorophenyl)ethyl]amino]acetyl-2-cyano-,(S)-,monohydrochloride
Compound 70
HC1
Figure imgf000042_0002
Pyrrolidine, l-(cyclopropylamino)acetyl-2-cyano-,(S)-,monohydrochloride
Compound 71
HC1
Figure imgf000042_0003
Pyrrolidine, l-[(2,6,6-trimethylbicyclo[3.1. l]hept-3-yl)amino]acetyl-2-cyano-, [1S[1 alpha, 2 alpha ,3 beta (S*),5 alpha ]] -monohydrochloride
Compound 72
HC1
Figure imgf000042_0004
Pyrrolidine , 1 -[[(2-phenoxy)ethyl] amino] acetyl-2-cyano- , (S)- .monohydrochloride Compound 73
HC1
Figure imgf000043_0001
Pyrrolidine, l-[2-[(3,5-dimemoxyphenyl)emyl]amino]acetyl-2-cyano-,(S)- .monohydrochloride
Compound 74
HC1
Figure imgf000043_0002
Pyrrolidine , 1 - [( 1 -adamanty l)amino] acety 1-2-cy ano- , (S)- , monohydrochloride
Compound 75
HC1
Figure imgf000043_0003
Pyrrolidine , 1 - [(1 , 1 , 3 , 3-tetramethylbutyl)amino] acety 1-2-cy ano- , (S)- .monohydrochloride
Compound 76
HC1
Figure imgf000043_0004
Pyrrolidine , 1 - [(2-adamantyl)amino] acety 1-2-cyano- , (S)- , monohydrochloride Compound 77
HC1
Figure imgf000044_0001
Pyrrolidine, 1-[(1 , l-dimethylpropyl)amino]acetyl-2-cyano-,(S)-,monohydrochloride Compound 78
HC1
Figure imgf000044_0002
Pyrrolidine, l-[(phenylmethyl)amino]acetyl-2-cyano-,(S)-,monohydrochloride
Compound 79
HC1
Figure imgf000044_0003
Pyrrolidine, 1-[(1 , l-dimethylethyl)amino]acetyl-2-cyano-,(S),monohydrochloride
Compound 80
Figure imgf000044_0004
Pyrrolidine, l-[[(2-adamantyl)methyl]amino]acetyl-2-cyano-,(S)-,monohydrochloride Compound 81
HCl
Figure imgf000045_0001
Pyrrolidine , 1 -[(2-phenylethyl)amino] acety 1-2-cyano- , (S)- .monohydrochloride Compound 82
HCl
Figure imgf000045_0002
Pyrrolidine, l-(pentylamino)acetyl-2-cyano-,(S)-,monohydrochloride
Compound 83
HCl
Figure imgf000045_0003
Pyrrolidine, l-(butylamino)acetyl-2-cyano-,(S)-,monohydrochloride Compound 84
HCl
Figure imgf000045_0004
Pyrrolidine , 1 -(cyclododecylamino)acety 1-2-cyano- , (S)- .monohydrochloride Compound 85
HCl
Figure imgf000046_0001
Pyrrolidine , 1 -(cycloocty lamino)acety 1-2-cyano-, (S)- .monohydrochloride Compound 86
HCl
Figure imgf000046_0002
Pyrrolidine , 1 -(propylamino)acety 1-2-cyano- , (S)- .monohydrochloride
Compound 87
HCl
Figure imgf000046_0003
Pyrrolidine, l-(ethylamino)acetyl-2-cyano-,(S)-, monohydrochloride
Compound 88
Figure imgf000046_0004
Pyrrolidine , 1 -(heptylamino)acety 1-2-cyano- , (S)- .monohydrochloride Compound 89
HCl
Figure imgf000047_0001
Pyrrolidine , 1 -(hexylamino)acety 1-2-cyano- , (S)- .monohydrochloride
Compound 90
2HC1
Figure imgf000047_0002
Pyrrolidine, l-[[3-[(5-cyano-2-pyridinyl)ammo]propyl]amino]acetyl-2-cyano-,(S)-, dihydrochloride
Compound 91
HCl
Figure imgf000047_0003
Pyrrolidine , 1 -[( 1 -ethy lpropyl)amino] acety 1-2-cyano- , (S)- .monohydrochloride Compound 92
HCl
Figure imgf000047_0004
Pyrrolidine, l-[(2,3-dmydro-lH-inden-2-yl)amino]acetyl-2-cyano-,(S)-,monohydrochloride Compound 93
HCl
Figure imgf000048_0001
Pyrrolidine, l-[(l-phenylmethyl-4-piperidinyl)amino]acetyl-2-cyano-,(S)-,- monohydrochloride
Other Compounds
Figure imgf000048_0002
wherein R is NH-R1 ;
R1 is: Ct - C12 straight or branched chain alkyl; C3 - C7 cycloalkyl; CH2- CH2-NH-RU;
CH2- CH2-R ; CH2- CHrCHR - R ; or CH2- CH -CH2-R ; R" is a pyridine ring optionally substimted in one or two positions with halo, trifluoromethyl, cyano or nitro; or a pyrimidine ring optionally substimted in one position with halo, trifluromethyl, cyano or nitro;
Rπι is a phenyl ring optionally substimted in one to three positions with halo or C, - C3 alkoxy;
Each Rw is independently a phenyl ring optionally substimted in one position with halo or C, - C3 alkoxy; and Rv is a 2-oxopyrrolidine group or a C2 - C4 alkoxy group.
Figure imgf000049_0001
wherein R is NH-R1;
R1 is: Cj - C12 straight or branched chain alkyl optionally substimted with hydroxy, acetyl,
Ct - C3 alkoxy, or - C3 hydroxy alkyl;
C3 - C12 cycloalkyl optionally substituted with hydroxyl, acetyl, C, - C3 alkoxy, or
0, - 03 hydroxy alkyl; adamantyl; indanyl; piperidyl optionally substimted with benzyl; pyrrolidine optionally substimted with benzyl; bicycloheptyl optionally substituted in one to three positions with methyl; phenyl optionally substimted with in one to three positions with halo, methoxy, trifluoromethyl; pyridyl optionally substimted in one to three positions with halo, trifluoromethyl, nitro; or pyrimidyl optionally substimted with halo, trifluoromethyl, nitro;
C, - C3 straight or branched chain alkyl substituted with Rw , and optionally substimted with hydroxy; or
(CH,)!., - NRπRra; R" is hydrogen or methyl;
Rm is phenyl optionally substituted with CN, or pyridyl optionally substimted with CN; and
Rw is a group selected from phenyl, naphthyl, cyclohexenyl, pyridyl, pyrimidyl, adamantyl, phenoxy, wherein the group is optionally substimted in one to two positions with ethoxy, methoxy, halo, phenylsulfide, or phenylsulfide substimted with hydroxymethyl.
EXAMPLE 2: SYNTHESIS OF COMPOUNDS ACCORDING TO CORE STRUCTURE II
Compounds according to Core Structure π can be produced according to a variety of approaches, including the approaches and methodologies provided above for Core Structure I. Appropriate starting materials include:
Figure imgf000050_0001
Other synthesis protocols also are available in the art, and are applicable in view of the teachings contained herein. Other exemplary compounds are set forth below.
Compound 1
Figure imgf000050_0002
3-morpholinecarboxylic acid. 4-.4-methyl-2-(phenylamino)-1 -thioxypentyll-
Principal Group: Substituents:
Carboxylic acid 4 pentyl Parent Hvdrid: 4 methyl morpholine 2 amino Functionalized Hydride: 1 thioxo
3-morpholinecarboxylic acid phenyl
Compound 2
Figure imgf000051_0001
3-thiomorpholinecarboxylic acid, 4-.4-methyl-2-(phenylamino)-1 -thioxypentyll-
Principal Group: Substituents:
Carboxylic acid 4 pentyl
Parent Hvdrid: 4 methyl thiomorpholine 2 amino
Functionalized Hydride: 1 thioxo
3-thiomorpholinecarboxylic acid phenyl
Compound 3
Figure imgf000051_0002
2-piperazinecarboxylic acid, 1-[4-methyl-2-(phenylamino)-1-thioxypentyll-
Principal Group: Substituents:
Carboxylic acid 1 pentyl Parent Hvdrid: 4 methyl piperazine 2 amino Functionalized Hydride: 1 thioxo
2-piperazinecarboxylic acid phenyl Compound 4
Figure imgf000052_0001
2-piperazinecarboxylic acid, 4-methyl-1 -r4-methyl-2-(phenylamino)-1 -thioxypentvH-
Principal Group: Substituents:
Carboxylic acid 1 pentyl
Parent Hvdrid: 4 methyl piperazine 2 amino
Functionalized Hydride: 1 thioxo
2-piperazinecarboxylic acid phenyl
4 methyl
Compound 5
Figure imgf000052_0002
2-piperazinecarboxylic acid. 4-methyl-1 -f4-methyl-1 -oxo-2-(phenylamino)pentyll-
Principal Group: Substituents: Carboxylic acid 1 pentyl Parent Hvdrid: 4 methyl piperazine 2 amino
Functionalized Hydride: 1 oxo 2-piperazinecarboxylic acid phenyl
4 methyl Compound 6
Figure imgf000053_0001
2-piperazinecarboxylic acid, 1 -f4-methyl-1 -oxo-2-(phenylamino)pentyl 1-
Principal Group: Substituents:
Carboxylic acid 1 pentyl Parent Hvdrid: 4 methyl piperazine 2 amino Functionalized Hvdride: 1 oxo
2-piperazinecarboxylic acid phenyl
Compound 7
Figure imgf000053_0002
3-morpholinecarboχylic acid. 4-f4-methyl-1 -oxo-2-(phenylamino)pentvπ-
Principal Group: Substituents:
Carboxylic acid 4 pentyl
Parent Hvdrid: 4 methyl morpholine 2 amino
Functionalized Hvdride: 1 oxo
3-morpholinecarboxylic acid phenyl Compound 8
Figure imgf000054_0001
3-thiomorpholinecarboxylic acid, 4-f4-methyl-1 -oxo-2-(phenylamino)pentvH-
Principal Group: Substituents:
Carboxylic acid 4 pentyl
Parent Hvdrid: 4 methyl thiomorpholine 2 amino
Functionalized Hvdride: 1 oxo
3-thiomorpholinecarboxylic acid phenyl
Compound 9
Figure imgf000054_0002
4-thiomorpholineethanamine, α-(2-methylpropyl)-β-oxo-N-phenyl-3-(2H-tetrazol-5-yl)-
Principal Group: Substituents: amine α propyl Conjunctive Parent: 2 methyl
4-thiomorpholineethanamine 3 2/7-tetrazol-5-yl β oxo
N phenyl Compound 10
Figure imgf000055_0001
4-morpholineethanamine. α-(2-methylpropyl)-β-oxo-N-phenyl-3-(2 Y-tetrazol-5-yl)-
Principal Group: Substituents: amine α propyl Conjunctive Parent: 2 methyl
4-morpholineethanamine 3 2 y-tetrazol-5-yl β oxo
N phenyl
Compound 1 1
Figure imgf000055_0002
1 -piperazineethanamine, 4-methyl-ct-(2-methylpropyl)-β-oxo-N-phenyl-2-(2/ -tetrazol-5-yl)-
Principal Group: Substituents: amine α propyl
Conjunctive Parent: 2 methyl 1 -piperazineethanamine 2 2/ -tetrazol-5-yl β oxo
N phenyl
4 methyl Compound 12
Figure imgf000056_0001
4-morpholineethanamine. N-ethyl-α-(2-methylpropyl)-β-oxo-3-(2/7-tetrazol-5-yl)-
Principal Group: Substituents: amine α propyl Conjunctive Parent: 2 methyl
4-morpholineethanamine 3 2A/-tetrazol-5-yl β oxo
N ethyl
Compound 13
Figure imgf000056_0002
4-thiomorpholineethanamine, N-ethyl-α-(2-methylpropyl)-β-oxo-3-(2/7-tetrazol-5-yl)-
Principal Group: Substituents: amine α propyl Conjunctive Parent: 2 methyl
4-thiomorpholineethanamine 3 2H-tetrazol-5-yl β oxo
N ethyl Compound 14
Figure imgf000057_0001
4-morpholineethanamine, Λt-ethyl-α-(2-methylpropyl)-β-oxo-3-(2λ -tetrazol-5-yl)-
Principal Group: Substituents: amine α propyl Conjunctive Parent: 2 methyl
4-morpholineethanamine 3 2 y-tetrazol-5-yl β oxo
N ethyl
EXAMPLE 3: SYNTHESIS OF COMPOUNDS ACCORDING TO CORE STRUCTURE m
Compounds according to Core Strucmre DI can be produced according to a variety of approaches. Representative approaches are shown below:
Figure imgf000058_0001
Figure imgf000058_0002
Figure imgf000058_0003
NaCNBH3
Figure imgf000058_0004
(As above)
Figure imgf000058_0005
Figure imgf000058_0006
See Oleksyszyn et al, Synthesis 479 (1978). Other exemplary compounds are depicted below. Compound 1
Figure imgf000059_0001
pentamide, 2-(ethylamino)-N,4-dimethyl-N-π-2 -tetrazol-5-yl)ethyll-
Principal Group: Substituents: amide 2 amino
Parent Hvdrid: ethyl pentane N,4-dimethyl
Functionalized Hvdride: N ethyl pentamide 1-2A/-tetrazol-5-yl
Compound 2
Figure imgf000059_0002
pentamide, N,4-dimethyl-2-(phenylamino)-N-f1-2/Y-tetrazol-5-yl)ethvH-
Principal Group: Substituents: amide 2 amino
Parent Hvdrid: phenyl pentane N,4-dimethyl
Functionalized Hvdride: N ethyl pentamide 1-2//-tetrazol-5-yl Compound 3
Figure imgf000060_0001
pentamide, 4-methyl-2-(phenylamino)-N-ρropyl-N-(2 - -tetrazol-5-yl methyl)-
Principal Group: Substituents: amide 4 methyl
Parent Hvdrid: 2 amino pentane phenyl
Functionalized Hvdride: N propyl pentamide N methyl
2 -tetrazol-5-yl
Compound 4
Figure imgf000060_0002
pentanethioamide, 4-methyl-2-(phenylamino)-N-propyl-N-(2//-tetrazol-5-yl methyl)-
Principal Group: Substituents: thioamide 4 methyl Parent Hvdrid: 2 amino pentane phenyl
Functionalized Hvdride: N propyl pentanethiomide N methyl
2/V-tetrazol-5-yl Compound 5
Figure imgf000061_0001
acetic acid, rf4-methyl-2-(phenylamino)-1 -thioxopentyllpropylaminol-
Principal Group: Substituents: oic acid ammo
Parent Hvdrid: pentyl ethane 4 methyl
Functionalized Hvdride: 2 amino
Acetic acid phenyl
1 thioxo propyl
Compound 6
Figure imgf000061_0002
acetic acid, ff4-methyl-1 -oxo-2-(phenylamino)pentyllpropylaminol
Principal Group: Substituents: oic acid ammo
Parent Hvdrid: pentyl ethane 4 methyl
Functionalized Hvdride: 2 amino
Acetic acid phenyl
1 oxo propyl Compound 7
Figure imgf000062_0001
acetic acid, [f2-ethylamino)-4-methyl-1 -oxopentyllpropylamino1
Principal Group: Substituents: oic acid ammo
Parent Hvdrid: pentyl ethane 2 amino
Functionalized Hvdride: ethyl
Acetic acid 4 methyl
1 oxo propyl
Compound 8
Figure imgf000062_0002
acetic acid. ff2-ethylamino)-1 -oxopropyll(phenylmethyl)aminol
Principal Group: Substituents: oic acid ammo
Parent Hvdrid: propyl ethane 2 amino
Functionalized Hvdride: ethyl
Acetic acid 1 oxo methyl phenyl Compound 9
Figure imgf000063_0001
acetic acid, ff2-ethylamino)-1 -thioxopropylKphenylmethvDaminol
Principal Group: Substituents: oic acid ammo
Parent Hvdrid: propyl ethane 2 amino
Functionalized Hvdride: ethyl
Acetic acid 1 thioxo methyl phenyl
Compound 10
Figure imgf000063_0002
acetic acid. fr2-ethylamino)-1 -oxopropyl1(phenylmethyl)amino1-
Principal Group: Substituents: oic acid amino
Parent Hvdrid: propyl ethane 2 amino
Functionalized Hvdride: ethyl
Acetic acid 1 oxo methyl phenyl Compound 1 1
Figure imgf000064_0001
phenylalanine, Λ/-.2-ethylamino)-1 -oxopropyll-N-tphenylmethyl)-
Principal Group: Substituents: oic acid ΛApropyl Conjunctive Parent: 2 amino phenylalanine ethyl 1 oxo N methyl Phenyl
Compound 12
Figure imgf000064_0002
phosphonic acid. [ff2-(ethylamino)-1 -oxopropyll(phenylmethyl)amino1methyll-, diphenyl ester-
Principal Group: Substituents: phosphonic acid methyl
Modifiers: amino diphenyl propyl 2 amino ethyl 1 oxo methyl phenyl Compound 13
Figure imgf000065_0001
phosphonic acid, ffr2-(ethylamino)-1 -oxopropynmethylamino1methyll-, diphenyl ester-
Principal Group: Substituents: phosphonic acid methyl Modifiers: amino diphenyl propyl
2 amino ethyl
1 oxo methyl
EXAMPLE 4: SYNTHESIS OF COMPOUNDS ACCORDING TO CORE
STRUCTURE IV
Methodologies for production of compounds according to Core Strucmre IV are disclosed in Lin et al, Proc. Nat'l Acad. Sci. USA 95: 14020-24 (1998). Exemplary compounds are set forth below.
Compound 1
Figure imgf000066_0001
Icv-lopentanecartionitnl-. 2.[1-Huoro-2-(propylamιno)et ylideπeK (2Z)-
Pr iπclpal Group: carbonitrilβ
Parent Hvdride: cyclopentanβ
Functionalized Hvdride: cyclopβntanecarbonitrilβ
Substituents:
2 ethytidβπe
1 fluoro
2 amino propyl
Compound 2
Figure imgf000067_0001
B-pyrrolidlnecarbonitπle. 3-H-(luoro-2-(propylamino)et yliden_l-. 13Z]-
Principal Group: carbonitπlβ
Parent Hvdride: pyrrolidine
Functionalized Hvdride:
2-pyrrolidinβcarbonrtnlβ
Substituents:
3 βthylldβnβ
1 fluoro
2 amino propyl
Compound 3
Figure imgf000067_0002
B-PVrrolidineearboxylic acid. 3-f1-H_oro-2-(propylamιno)ethylideπel-. (3^)7
Principal Group: carboxylic acid
Parent Hvdride: pyrrolidine
Functionalized Hvdride:
2-pyπrolidinacarboxylic acid
Substituents:
3 βthylidβnβ
1 fluoro
2 amino propyl Compound 4
Figure imgf000068_0001
B-furancarboxylιc acid. 3-11 -lluoro-2-forop ylaminolethvtideneltetrarivdro-. (3Z1-
Prlnclpal Group: carboxylic acid
Parent Hvdride: furan
Functionalized Hvdride:
2-furancarboxylic acid
Substituents:
3 ettiylidβnβ
1 fluoro
2 amino propyl tβtrahydro
Compound 5
Figure imgf000068_0002
P-furancaiboxylic acid. 4-H-fluoro-2-(propytamino)et ylldeneltetrahvdr_-. (4£1- '
Principal Group: carboxylic acid
Parent Hvdride: furan
Functionalized Hvdride:
3-furaπcarboxylic acid
Substituents:
4 ethylidenβ
1 fluoro
2 amino propyl tetrahydro Compound 6
Figure imgf000069_0001
B-pyrrolidinecarboxytic acid. 4-H-lluoro-2-(propytamino)etrιylidenel-. (4E -
Prlncloal Group: carboxylic acid
Parent Hvdride: pyrrolidine
Functionalized Hvdride:
3-pyrrolidinβcarboxylic acid
Subetltuents:
4 ethylidanβ
1 fluoro
2 amino propyl
Compoimd 7
Figure imgf000069_0002
B-pyrrolidinecarboxytic acid. 4-(2-amiπo-1-fluoro-3-pheπylpropytideπe)-. (4B-
Princlpal Group: carboxylic acid
Pqrent Hy ide; pyrrolidine
Functionalized Hvdride:
3-pyrrolidiπecar_oxyilc acid
Substituents:
4 propylldβnβ
2 amino 1 fluoro
3 phenyl Compound 8
Figure imgf000070_0001
kvclopantaπecarboxyllc add, 2-(2-amino-1-fluoro-3-PhenylpropylldeneK (2ZI-
Principal Groun: carboxylic acid
Parent Hvdride: cyclopentaπe
Functionalized Hvdride: cyclopβntanecarboxylic acid
Substituents:
2 propylidene
2 amino 1 fluoro
3 phenyl
Compound 9
Figure imgf000070_0002
B-pyrrolidinecaiboxylic acid.3-(2-amιno-1-fluoro-3-Phenylpropytιdene)-. (3Z ~
Principal Group: carboxylic acid Parent Hvdride: pyrrolidine
Functionalized Hvdride: 2-pyrrolidιnacarboxylιc acid Substituents: 3 propylidene
2 amino 1 fluoro
3 phenyl Compound 10
Figure imgf000071_0001
B-Pyrrolldinecaiboxylic acid. 3-(2-amiπo-1-fluoro-3-methylpentylidoπ9)-. (32)-
Prlncloal Group: carboxylic acid Parent Hvdride: pyrrolidine
Functionalized Hvdride: 2-pyrrolidinβcarboxylic acid Substituents: 3 peπtylidβne
2 amino 1 fluoro
3 methyl
Compoimd 11
Figure imgf000071_0002
fcvciopentanecarboxylic acid. 2-(2-amino-1-f1uoro-3-mothylpentyliden9)-. (221-
Principal Group: carboxylic acid
Parent Hvdride: cyclopentane
Functionalized Hvdride: cyciopentanecarboxylic acid
Substituents:
2 pentylidβne
2 amino 1 fluoro
3 methyl Compound 12
Figure imgf000072_0001
C-oyrrolidinecarboxylic acid. 4-(2-amino-1-fluoro-3-methylpentylldenel-. (4B-
Princlpal Group: carboxylic acid
Parent Hvdride: pyrrolidine
Functionalized Hvdride:
3-pyrrolidinβcaιboxylJc acid
Substituents:
4 pβntylidane
2 amino 1 fluoro
3 methyl
Compound 13
Figure imgf000072_0002
B-Pyrrolldlnecaibonitrile. 4-(2-amiπo-1-fluoro-3-methylpentylideπ9)-. (4B-
Princlpal Group: carbonitrile
Parent Hvdride: pyrrolidine
Functionalized Hvdride:
3-pyrrolidinecarbonitrilβ
Substituents:
4 pentylldane
2 amino 1 fluoro
3 methyl Compound 14
Figure imgf000073_0001
g-nvrrnliriln«πartιnnltrile.3-(2.Hmino-1-tluoro-3-methvlpentvlidene)-. (3Z
Principal Group: carbonitnle
Parent Hvdride: pyrrolidine
Functionalized Hvdride:
2-pyrrolldinecarbonitπlβ
Substituents:
3 pentylidβne
2 amino 1 fluoro
3 methyl
Compound 15
Figure imgf000073_0002
Principal Group: carbonitnle
Parent Hvdride: furan
Functionalized Hvdride:
2-furancarbonitrile Substituents: 3 pentylideπe
2 amino 1 fluoro
3 methyl tetrahydro Compound 16
Figure imgf000074_0001
Icvclopentanecarbonitrile. 2-(2-amino-1-tluorobutylideπ9)-. (2 T
Principal Group: carbonitnle
Parent Hvdride: cyclopentane
Functionalized Hvdride: cyelopentanecaibonitπle
Substituents:
2 butylidene
2 amino
1 fluoro
Compound 17
Figure imgf000074_0002
bhoaphonous acid. K2__-2-(2-amιno-1 -fluorobutylideπelcvclopentvH-. diphenyl ester
Parent Hvdride: phosphonous acid Substituents: cyclopeπtyl 2 butylidene 2 amino 1 fluoro Modifiers: diphenyl Compound 18
Figure imgf000075_0001
Iphosphoπous acid. r(32)-3-(2-amlno-1-lluorobutylidβπe)pyrrolldinyll-. diphenyl ester"
Parent Hvdride: phosphonous acid Substituents: pyrrolidinyl 3 butylidene 2 amino 1 fluoro Modifiers: diphenvl
Compoimd 19
Figure imgf000075_0002
Iphosphoπous acid. f(32)-3-(2-amιπo-1-fluoro-3-methylpentylid9ne)_yrrolldinyl1-. diphenyl ester
Parent Hvdride: phosphonous acid Substituents: pyrrolidinyl 3 pentylidene
2 amino 1 fluoro
3 methyl Modifiers: diphenyl Compound 20
Figure imgf000076_0001
Iphosphoπous acid. K2_n-2-(2-amino-1-fluoro-3-methylpentylidene)cvclopentyll-, diphenyl ester
Parent Hvdride: phosphonous acid Substituents: cyclopentyl 2 pentylidene
2 amino 1 fluoro
3 methyl Modifiers: diphenyl
EXAMPLE 5: EXEMPLARY NEURO ACTIVITY TESTING PROTOCOLS
There are a variety of protocols available for evaluating the neuroactivity of the above compounds and other compounds designed, made and used according to the invention. These assays can be in vivo or in vitro methods. The approaches below include assays measuring the ability of compounds to protect neuronal cells from toxic treatments, and the ability of the compounds to elicit neuronal cell growth, regeneration, neurite extension and the like.
Immunostaining and Neurite Outgrowth Quantitation Assays
Spinal cord and dorsal root ganglion (DRG) cells from adult mice can be isolated by micro-dissection. The spinal cord with attached DRGs from an adult mouse (15-10g) is removed. Spinal nerves are cut away using micro-dissection scissors and any excess material is trimmed until the DRG is free. Using sharp micro-dissecting scissors, a transverse cut is made in the peripheral nerve, leaving 1-2 mm attached, and the explant is placed into Petri dish and covered with plating media. When finished collecting all DRGs, the spinal nerve is trimmed to about ImM in length. Then, embed the explant in 30 μL of reduced growth factor Matrigel on a circular coverslip, and place in a 35 mM culture dish. Cover the sensory ganglion explant with 2 mis of media. Compounds, drugs or control solutions are added from 10X stocks, and are incubated at 37°C, 5% CO2, 95% humidity for 48 hrs. Wash cultures twice with PBS, and fix with 10% formalin for 30 minutes. Wash the fixed cultures twice with PBS and store refrigerated in PBS.
Place cultures in Block Buffer (5% Horse Serum, 5% Goat Serum, 1 % Triton X, PBS pH=7.4) overnight, while rotating, at a temperature of 4°C. Add primary antibody (for example, Beta tubulin, Sigma Chemical Co.) diluted in Block Buffer and incubate overnight at 4°C. Wash 5 times with PBS and apply secondary antibody (Alexa 488 Goat Anti-Mouse) diluted in block buffer. Incubate overnight at 4°C. Wash 5 times with PBS and leave overnight at 4°C. Coverslip the cultures and measure total neurite length from the end of the attached spinal nerve. Lengths of all neurites are quantitated and compared to those present in vehicle-treated control DRGs.
Neuroprotection Assays
Cultures are derived from postnatal day 8 (P8) Sprague-Dawley rat lumbar spinal cord slices of 325 micron thickness. Each experiment consists of two 6- well plates with 5 slices from 4 different animals per well. Media changes are performed every 3 to 4 days. Cultures are treated with THA [L(-)-threo-3-hydroxyaspartic acid; Tocris Cookson Inc., Ballwin, Missouri] at 200μM + compound (lOμM) after one week in culture. The control is an untreated sample with 0.1 % DMSO as vehicle. The THA control is a THA treated sample with 0.1 % DSMO as vehicle. Two wells are used per condition. One media change with new THA and compounds is performed. The experiment is stopped 6 to 8 days following drug treatment (13-15 total days in vitro, DIV) as dictated by visual assessment of lesion, by fixation with 4% paraformaldehyde/0.1 M phosphate buffer for 30 minutes. Slices are permeabilized with 100% cold methanol forlO minutes. Slices are transferred to staining wells. The slices are blocked with 10% HS/TBS. Primary antibody incubation is overnight at 4°C with SMI-32 antibody 1:5000 in 2% HS/TBS. SMI-32 was specific towards unphosphorylated H neurofilament subunit. Vectastain ABC Elite Kit with rat absorbed anti-mouse secondary antibody is used with DAB to stain the slices. The slices are mounted onto a slide and a coverslip is sealed with DPX mounting solution.
Quantification of surviving neurons is performed on a Ziess Axiovert microscope. Neuronal survival is determined by observing an intact neuronal cell body with processes located ventrally of the central canal in each hemisphere. This correlates to laminae VII, VIII and IX. Each hemisphere is counted individually. The statistics can be performed with StatView software on a minimum of three different experiments per condition and significance should be determined as compared to THA control. The percent of protection can be determined from the average number of living neurons by the following equation:
(drug treatment condition - THA control) / (Untreated control - THA control).
EXAMPLE 6: EXEMPLARY TESTING PROTOCOLS FOR PROSTATE
TREATMENT EFFICACY
Protocols for testing efficacy, dosing, and administration schedules for post- prostatectomy nerve recovery can be performed in accordance with the teachings of Example 5.
To evaluate DPP IV inhibitors in the treatment of prostate cancer, there are several cancer cell lines available of conducting in vitro assays. Appropriate cell lines include LNCaP, PC3, DU-145 and TSUPrl for use in cell proliferation assays.
For example, a cell line can be propagated in a standard medium, such as RPMI
1640 containing 10% fetal calf serum. Cells are first propagated and allowed to adhere. The cells can then be treated with one or more DPPP IV inhibitors at varying concentrations, and then pulsed with [3H] thymidine to evaluate incorporation, which is indicative of cell viability and proliferation. See U.S. Patent No. 5,804,602.
It is to be understood that the description, specific examples and data, while indicating exemplary embodiments, are given by way of illustration and are not intended to limit the present invention. Various changes and modifications within the present invention will become apparent to the skilled artisan from the discussion, disclosure and data contained herein, and thus are considered part of the invention.

Claims

WE CLAIM:
1. An inhibitor of dipeptidyl peptidase IV, wherein the inhibitor comprises a proline mimetic and possesses an IC50 of no more than 1 μm and has a molecular weight of no more than 500.
2. The inhibitor according to claim 1, wherein the IC50 is no more than 100 nm.
3. The inhibitor according to claim 1, wherein the inhibitor can be used to treat a central nervous system disorder selected from the group consisting of strokes, mmors, ischemia, Parkinson's disease, amyotrophic lateral sclerosis and migraines.
4. A reversible inhibitor of dipeptidyl peptidase IV, wherein the inhibitor has a core structure of:
(D
Figure imgf000080_0001
, wherein:
X is CR2R3, O, S, or NR4; with the proviso that if X is S, or if X and XI are both CH2, and Z is O, and A is CN, and Rl is H, then R is not NH substimted with C1-C9 straight or branched chain alkyl, or NH substimted with C3-C7 cycloalkyl;
Xj is CR2R3, O, S, or NR4 with the proviso that X and XI cannot both be a heteroatom, and with the proviso that if X and XI are both CH2, and Z is O, and Rl is NH2, then R is not 1-methylpropyl if A is COOH, and R is not cyclopentyl if A is CN; A is H, COOH, or isosteres of carboxylic acids, such as one selected from the group consisting of CN, SO3H, CONOH, PO3R5R6, SO2NHR7, tetrazole, amides, esters, and acid anhydrides, with the proviso that if A is CN, and Rl is NH2, and Z is O, and R is 1-methylpropyl, then X and XI are not both CH2; X and XI are not S; and X is not O;
Z is O or S;
R and Rl are independently selected from the group of functional groups consisting of H, CrC9 branched or straight chain alkyl, C2-C9 branched or straight chain alkenyl, C3- C8 cycloalkyl, C5-C7 cycloalkenyl, aryl, heteroaryl and amino, wherein any of the functional groups can be substituted with one or more of C C9 straight or branched chain alkyl, aryl, heteroaryl, amino, halo, carbonyl, CrC9 alkoxy, C2-C9 alkenyloxy, phenoxy, benzyloxy, C3-C8 cycloalkyl, cyano, amido, thiol, trifluromethyl, or hydroxy, wherein each of R and Rl can be the same or different; and
R2, R3, R4, R5, R6 and R7, if present, are independently selected from the group of functional groups consisting of H, CrC9 branched or straight chain alkyl, C2-C9 branched or straight chain alkenyl, C3-C8 cycloalkyl, C5-C7 cycloalkenyl, aryl, heteroaryl and amino, wherein any of the functional groups can be substimted with one or more of - C9 straight or branched chain alkyl, aryl, heteroaryl, amino, halo, carbonyl, CrC9 alkoxy, C2-C9 alkenyloxy, phenoxy, benzyloxy, C3-C8 cycloalkyl, cyano, amido, thiol, trifluromethyl, or hydroxy, wherein each of R2, R3, R4, R5, R6 and R7, if present, can be the same or different.
5. The reversible inhibitor according to claim 4, wherein the inhibitor possesses an IC50 of no more than 1 μm and has a molecular weight of no more than 500.
6. A reversible inhibitor of dipeptidyl peptidase IV, wherein the inhibitor has a core strucmre of:
(II)
Figure imgf000082_0001
, wherein:
X is CR2R3, O, S, or NR4;
A is H, COOH, or isosteres of carboxylic acids, such as one selected from the group consisting of CN, SO3H, CONOH, PO3R5R6, SO2NHR7, tetrazole, amides, esters, and acid anhydrides;
Z is O or S;
R and Rl are independently selected from the group of functional groups consisting of H, CrC9 branched or straight chain alkyl, C2-C9 branched or straight chain alkenyl, C3- Cg cycloalkyl, C5-C7 cycloalkenyl, aryl, heteroaryl and amino, wherein any of the functional groups can be substituted with one or more of C,-C9 straight or branched chain alkyl, aryl, heteroaryl, amino, halo, carbonyl, CrC9 alkoxy, C2-C9 alkenyloxy, phenoxy, benzyloxy, C3-C8 cycloalkyl, cyano, amido, thiol, trifluromethyl, or hydroxy, wherein each of R and Rl can be the same or different; and
R2, R3, R4, R5, R6 and R7, if present, are independently selected from the group of functional groups consisting of H, -C, branched or straight chain alkyl, C2-C9 branched or straight chain alkenyl, C3-C8 cycloalkyl, C5-C7 cycloalkenyl, aryl, heteroaryl and amino, wherein any of the functional groups can be substimted with one or more of Ct- C9 straight or branched chain alkyl, aryl, heteroaryl, amino, halo, carbonyl, C,-C9 alkoxy, C2-C9 alkenyloxy, phenoxy, benzyloxy, C3-C8 cycloalkyl, cyano, amido, thiol, trifluromethyl, or hydroxy, wherein each of R2, R3, R4, R5, R6 and R7, if present, can be the same or different.
7. The reversible inhibitor according to claim 6, wherein the inhibitor possesses an IC50 of no more than 1 μm and has a molecular weight of no more than 500.
8. A reversible inhibitor of dipeptidyl peptidase IV, wherein the inhibitor has a core strucmre of:
Figure imgf000083_0001
, wherein:
A is H, COOH, or isosteres of carboxylic acids, such as one selected from the group consisting of CN, SO3H, CONOH, PO3R5R6, SO2NHR7, tetrazole, amides, esters, and acid anhydrides;
Z is O or S;
R, Rl, R2 and R3 are independently selected from the group of functional groups consisting of H, CrC9 branched or straight chain alkyl, C2-C9 branched or straight chain alkenyl, C3-C8 cycloalkyl, C5-C7 cycloalkenyl, aryl, heteroaryl and amino, wherein any of the functional groups can be substimted with one or more of C,-C9 straight or branched chain alkyl, aryl, heteroaryl, amino, halo, carbonyl, CrC9 alkoxy, C2-C9 alkenyloxy, phenoxy, benzyloxy, C3-C8 cycloalkyl, cyano, amido, thiol, trifluromethyl, or hydroxy, wherein each of R, Rl, R2 and R3 can be the same or different; and R4, R5, R6 and R7, if present, are independently selected from the group of functional groups consisting of H, -Cg branched or straight chain alkyl, C2-C9 branched or straight chain alkenyl, C3-C8 cycloalkyl, C5-C7 cycloalkenyl, aryl, heteroaryl and amino, wherein any of the functional groups can be substimted with one or more of C,-C9 straight or branched chain alkyl, aryl, heteroaryl, amino, halo, carbonyl, C,-C9 alkoxy, C2-C9 alkenyloxy, phenoxy, benzyloxy, C3-C8 cycloalkyl, cyano, amido, thiol, trifluromethyl, or hydroxy, wherein each of R4, R5, R6 and R7, if present, can be the same or different.
9. The reversible inhibitor according to claim 8, wherein the inhibitor possesses an IC50 of no more than 1 μm and has a molecular weight of no more than 500.
10. A reversible inhibitor of dipeptidyl peptidase IV, wherein the inhibitor has a core structure of:
(TV)
Figure imgf000084_0001
, wherein:
X is CR2R3, O, S, or NR4;
X, is CR2R3, O, S, or NR4 with the proviso that X and XI cannot both be a heteroatom; A is H, COOH, or isosteres of carboxylic acids, such as one selected from the group consisting of CN, SO3H, CONOH, PO3R5R6, SO2NHR7, tetrazole, amides, esters, and acid anhydrides;
R and Rl are independently selected from the group of functional groups consisting of H, Cι-C9 branched or straight chain alkyl, C2-C9 branched or straight chain alkenyl, C3- Cg cycloalkyl, C5-C7 cycloalkenyl, aryl, heteroaryl and amino, wherein any of the functional groups can be substituted with one or more of CrC9 straight or branched chain alkyl, aryl, heteroaryl, amino, halo, carbonyl, CrC9 alkoxy, C2-C9 alkenyloxy, phenoxy, benzyloxy, C3-C8 cycloalkyl, cyano, amido, thiol, trifluromethyl, or hydroxy, wherein each of R and Rl can be the same or different; and
R2, R3, R4, R5, R6 and R7, if present, are independently selected from the group of functional groups consisting of H, CrC9 branched or straight chain alkyl, C2-C9 branched or straight chain alkenyl, C3-C8 cycloalkyl, C5-C7 cycloalkenyl, aryl, heteroaryl and amino, wherein any of the functional groups can be substimted with one or more of C,- C9 straight or branched chain alkyl, aryl, heteroaryl, amino, halo, carbonyl, C,-C9 alkoxy, C2-C9 alkenyloxy, phenoxy, benzyloxy, C3-C8 cycloalkyl, cyano, amido, thiol, trifluromethyl, or hydroxy, wherein each of R2, R3, R4, R5, R6 and R7, if present, can be the same or different.
11. The reversible inhibitor according to claim 10, wherein the inhibitor possesses an IC50 of no more than 1 μm and has a molecular weight of no more than 500.
12. A method of treating a patient having a disorder of the central nervous system, comprising administering to the patient a therapeutically effective amount of a reversible inhibitor of dipeptidyl peptidase IV, wherein the inhibitor has a core strucmre of:
CD
Figure imgf000086_0001
, wherein:
X is CR2R3, O, S, or NR4;
X, is CR2R3, O, S, or NR4 with the proviso that X and XI cannot both be a heteroatom;
A is H, COOH, or isosteres of carboxylic acids, such as one selected from the group consisting of CN, SO3H, CONOH, PO3R5R6, SO2NHR7, tetrazole, amides, esters, and acid anhydrides;
Z is O or S;
R and Rl are independently selected from the group of functional groups consisting of H, CrC9 branched or straight chain alkyl, C2-C9 branched or straight chain alkenyl, C3- C8 cycloalkyl, C5-C7 cycloalkenyl, aryl, heteroaryl and amino, wherein any of the functional groups can be substituted with one or more of CrC9 straight or branched chain alkyl, aryl, heteroaryl, amino, halo, carbonyl, CrC9 alkoxy, C2-C9 alkenyloxy, phenoxy, benzyloxy, C3-C8 cycloalkyl, cyano, amido, thiol, trifluromethyl, or hydroxy, wherein each of R and Rl can be the same or different; and
R2, R3, R4, R5, R6 and R7, if present, are independently selected from the group of functional groups consisting of H, CrC9 branched or straight chain alkyl, C2-C9 branched or straight chain alkenyl, C3-C8 cycloalkyl, C3-C7 cycloalkenyl, aryl, heteroaryl and amino, wherein any of the functional groups can be substituted with one or more of Cr C9 straight or branched chain alkyl, aryl, heteroaryl, amino, halo, carbonyl, C,-C9 alkoxy, C2-C9 alkenyloxy, phenoxy, benzyloxy, C3-C8 cycloalkyl, cyano, amido, thiol, trifluromethyl, or hydroxy, wherein each of R2, R3, R4, R5, R6 and R7, if present, can be the same or different.
13. The method according to claim 12, wherein the inhibitor possesses an IC50 of no more than 1 μm and has a molecular weight of no more than 500.
14. The method according to claim 12, wherein if X is S, or if X and XI are both CH2, and Z is O, and A is CN, and Rl is H, then R is not NH substimted with C1-C9 straight or branched chain alkyl, or NH substimted with C3-C7 cycloalkyl; and if X and XI are both CH2, and Z is O, and Rl is NH2, then R is not 1-methylpropyl if A is COOH, and R is not cyclopentyl if A is CN; and if A is CN, and Rl is NH2, and Z is O, and R is 1- methylpropyl, then X and XI are not both CH2; X and XI are not S; and X is not O;
15. A method of treating a patient having a disorder of the central nervous system, comprising administering to the patient a therapeutically effective amount of a reversible inhibitor of dipeptidyl peptidase IV, wherein the inhibitor has a core structure of:
Figure imgf000088_0001
, wherein:
X is CR2R3, O, S, or NR4;
A is H, COOH, or isosteres of carboxylic acids, such as one selected from the group consisting of CN, SO3H, CONOH, PO3R5R6, SO2NHR7, tetrazole, amides, esters, and acid anhydrides;
Z is O or S;
R and Rl are independently selected from the group of functional groups consisting of H, CrC9 branched or straight chain alkyl, C2-C9 branched or straight chain alkenyl, C3- Cg cycloalkyl, C5-C7 cycloalkenyl, aryl, heteroaryl and amino, wherein any of the functional groups can be substimted with one or more of CrC9 straight or branched chain alkyl, aryl, heteroaryl, amino, halo, carbonyl, C C9 alkoxy, -Cg alkenyloxy, phenoxy, benzyloxy, C3-C8 cycloalkyl, cyano, amido, thiol, trifluromethyl, or hydroxy, wherein each of R and Rl can be the same or different; and
R2, R3, R4, R5, R6 and R7, if present, are independently selected from the group of functional groups consisting of H, CrC9 branched or straight chain alkyl, C2-C9 branched or straight chain alkenyl, C3-C8 cycloalkyl, C5-C7 cycloalkenyl, aryl, heteroaryl and amino, wherein any of the functional groups can be substituted with one or more of Cr C9 straight or branched chain alkyl, aryl, heteroaryl, amino, halo, carbonyl, C,-C9 alkoxy, C2-C9 alkenyloxy, phenoxy, benzyloxy, C3-C8 cycloalkyl, cyano, amido, thiol, trifluromethyl, or hydroxy, wherein each of R2, R3, R4, R5, R6 and R7, if present, can be the same or different.
16. The method according to claim 15, wherein the inhibitor possesses an IC50 of no more than 1 μm and has a molecular weight of no more than 500.
17. A method of treating a patient having a disorder of the central nervous system, comprising administering to the patient a therapeutically effective amount of a reversible inhibitor of dipeptidyl peptidase IV, wherein the inhibitor has a core strucmre of:
(ffl)
Figure imgf000089_0001
, wherein
A is H, COOH, or isosteres of carboxylic acids, such as one selected from the group consisting of CN, SO3H, CONOH, PO3R5R6, SO2NHR7, tetrazole, amides, esters, and acid anhydrides;
Z is O or S;
R, Rl, R2 and R3 are independently selected from the group of functional groups consisting of H, CrC9 branched or straight chain alkyl, C2-C9 branched or straight chain alkenyl, C3-C8 cycloalkyl, C5-C7 cycloalkenyl, aryl, heteroaryl and amino, wherein any of the functional groups can be substimted with one or more of C,-C9 straight or branched chain alkyl, aryl, heteroaryl, amino, halo, carbonyl, C C9 alkoxy, C2-C9 alkenyloxy, phenoxy, benzyloxy, C3-C8 cycloalkyl, cyano, amido, thiol, trifluromethyl, or hydroxy, wherein each of R, Rl, R2 and R3 can be the same or different; and
R4, R5, R6 and R7, if present, are independently selected from the group of functional groups consisting of H, Cx-C9 branched or straight chain alkyl, C2-C9 branched or straight chain alkenyl, C3-C8 cycloalkyl, C5-C7 cycloalkenyl, aryl, heteroaryl and amino, wherein any of the functional groups can be substituted with one or more of C,-C9 straight or branched chain alkyl, aryl, heteroaryl, amino, halo, carbonyl, CrC9 alkoxy, C2-C9 alkenyloxy, phenoxy, benzyloxy, C3-C8 cycloalkyl, cyano, amido, thiol, trifluromethyl, or hydroxy, wherein each of R4, R5, R6 and R7, if present, can be the same or different.
18. The method according to claim 17, wherein the inhibitor possesses an IC50 of no more than 1 μm and has a molecular weight of no more than 500.
19. A method of treating a patient having a disorder of the central nervous system, comprising administering to the patient a therapeutically effective amount of a reversible inhibitor of dipeptidyl peptidase IV, wherein the inhibitor has a core strucmre of:
(IV)
Figure imgf000090_0001
, wherein:
X is CR2R3, O, S, or NR4;
Xi is CR2R3, O, S, or NR4 with the proviso that X and XI cannot both be a heteroatom; A is H, COOH, or isosteres of carboxylic acids, such as one selected from the group consisting of CN, SO3H, CONOH, PO3R5R6, SO2NHR7, tetrazole, amides, esters, and acid anhydrides;
R and Rl are independently selected from the group of functional groups consisting of H, CrC9 branched or straight chain alkyl, C2-C9 branched or straight chain alkenyl, C3- C8 cycloalkyl, C5-C7 cycloalkenyl, aryl, heteroaryl and amino, wherein any of the functional groups can be substituted with one or more of CrC9 straight or branched chain alkyl, aryl, heteroaryl, amino, halo, carbonyl, CrC9 alkoxy, C2-C9 alkenyloxy, phenoxy, benzyloxy, C3-C8 cycloalkyl, cyano, amido, thiol, trifluromethyl, or hydroxy, wherein each of R and Rl can be the same or different; and
R2, R3, R4, R5, R6 and R7, if present, are independently selected from the group of functional groups consisting of H, CrC9 branched or straight chain alkyl, C2-C9 branched or straight chain alkenyl, C3-C8 cycloalkyl, C3-C7 cycloalkenyl, aryl, heteroaryl and amino, wherein any of the functional groups can be substimted with one or more of Cr C9 straight or branched chain alkyl, aryl, heteroaryl, amino, halo, carbonyl, C,-C9 alkoxy, C2-C9 alkenyloxy, phenoxy, benzyloxy, C3-C8 cycloalkyl, cyano, amido, thiol, trifluromethyl, or hydroxy, wherein each of R2, R3, R4, R5, R6 and R7, if present, can be the same or different.
20. The method according to claim 19, wherein the inhibitor possesses an IC50 of no more than 1 μm and has a molecular weight of no more than 500.
21. A method of treating a patient having a disorder of the central nervous system, comprising administering to the patient a therapeutically effective amount of a inhibitor of dipeptidyl peptidase IV.
22. The method according to claim 21, wherein the inhibitor comprises a proline mimetic and possesses an IC50 of no more than 1 μm and has a molecular weight of no more than 700.
23. The method according to claim 21, wherein the inhibitor has a core strucmre selected from the group consisting of Core Strucmre I, Core Strucmre II, Core Strucmre HI and Core Strucmre IV.
24. The method according to claim 21, wherein the inhibitor is reversible.
25. The method according to claim 21, wherein the central nervous system disorder is selected from the group consisting of strokes, tumors, ischemia, Parkinson's disease, amyotrophic lateral sclerosis and migraines.
26. A method of treating a patient having a disorder selected from the group consisting of strokes, tumors, ischemia, Parkinson's disease, memory loss, hearing loss, vision loss, migraines, brain injury, spinal cord injury, Alzheimer's disease, amyotrophic lateral, multiple sclerosis, diabetic neuropathy and prostate abnormalities, wherein the method comprises administering to the patient a therapeutically effective amount of a inhibitor of dipeptidyl peptidase IV.
27. A method according to claim 26, wherein the inhibitor comprises a proline mimetic and possesses an IC50 of no more than 1 μm and has a molecular weight of no more than 700.
28. The method according to claim 26, wherein the inhibitor has a core strucmre selected from the group consisting of Core Strucmre I, Core Strucmre π, Core Strucmre HI and Core Structure IV.
29. A method of using a reversible inhibitor of DPP-IV, comprising administering to a human patient suffering from a central nervous system disorder a pharamceutically effective amount of the inhibitor, wherein the inhibitor is
Figure imgf000093_0001
wherein R is NH-R1 ;
R' is: Cj - C12 straight or branched chain alkyl; c3 - C7 cycloalkyl;
CH2- CH2-NH-R";
CH2- CH2-Rιπ;
CH2- CH2-CHRIV- RIV; or
CH2- H2- H2-R ;
R" is a pyridine ring optionally substimted in one or two positions with halo, trifluoromethyl, cyano or nitro; or a pyrimidine ring optionally substimted in one position with halo, trifluromethyl, cyano or nitro;
R is a phenyl ring optionally substimted in one to three positions with halo or C, - C3 alkoxy;
Each RIV is independently a phenyl ring optionally substituted in one position with halo or C, - C3 alkoxy; and
Rv is a 2-oxopyrrolidine group or a C2 - C4 alkoxy group.
30. A method of using a reversible inhibitor of DPP-IV, comprising administering to a human patient suffering from a central nervous system disorder a pharamceutically effective amount of the inhibitor, wherein the inhibitor is
Figure imgf000094_0001
wherein R is NH-R1;
R1 is: Cj - C12 straight or branched chain alkyl optionally substimted with hydroxy, acetyl, Ct - C3 alkoxy, or - C3 hydroxyalkyl;
C3 - C12 cycloalkyl optionally substituted with hydroxyl, acetyl, C, - C3 alkoxy, or Cj - C3 hydroxyalkyl;
adamantyl; indanyl; piperidyl optionally substimted with benzyl; pyrrolidine optionally substimted with benzyl; bicycloheptyl optionally substimted in one to three positions with methyl; phenyl optionally substimted with in one to three positions with halo, methoxy, trifluoromethyl; pyridyl optionally substimted in one to three positions with halo, trifluoromethyl, nitro; or pyrimidyl optionally substimted with halo, trifluoromethyl, nitro;
Ct - C3 straight or branched chain alkyl substituted with Rw , and optionally substimted with hydroxy; or
(CH,),., - NRπRιπ; Rπ is hydrogen or methyl; Rπι is phenyl optionally substituted with CN, or pyridyl optionally substimted with CN; and Rw is a group selected from phenyl, naphthyl, cyclohexenyl, pyridyl, pyrimidyl, adamantyl, phenoxy, wherein the group is optionally substituted in one to two positions with ethoxy, methoxy, halo, phenylsulfide, or phenylsulfide substituted with hydroxymethyl.
PCT/US2000/030836 1999-11-12 2000-11-13 Dipeptidyl peptidase iv inhibitors and methods of making and using dipeptidyl peptidase iv inhibitors WO2001034594A1 (en)

Priority Applications (4)

Application Number Priority Date Filing Date Title
AU19164/01A AU1916401A (en) 1999-11-12 2000-11-13 Dipeptidyl peptidase iv inhibitors and methods of making and using dipeptidyl peptidase iv inhibitors
JP2001536541A JP2003535034A (en) 1999-11-12 2000-11-13 Dipeptidyl peptidase IV inhibitors and methods for producing and using dipeptidyl peptidase IV inhibitors
CA002390231A CA2390231A1 (en) 1999-11-12 2000-11-13 Dipeptidyl peptidase iv inhibitors and methods of making and using dipeptidyl peptidase iv inhibitors
EP00982093A EP1228061A4 (en) 1999-11-12 2000-11-13 Dipeptidyl peptidase iv inhibitors and methods of making and using dipeptidyl peptidase iv inhibitors

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US43908999A 1999-11-12 1999-11-12
US09/439,089 1999-11-12

Publications (1)

Publication Number Publication Date
WO2001034594A1 true WO2001034594A1 (en) 2001-05-17

Family

ID=23743248

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2000/030836 WO2001034594A1 (en) 1999-11-12 2000-11-13 Dipeptidyl peptidase iv inhibitors and methods of making and using dipeptidyl peptidase iv inhibitors

Country Status (5)

Country Link
EP (1) EP1228061A4 (en)
JP (1) JP2003535034A (en)
AU (1) AU1916401A (en)
CA (1) CA2390231A1 (en)
WO (1) WO2001034594A1 (en)

Cited By (110)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP1175213A2 (en) * 1999-03-12 2002-01-30 Bristol-Myers Squibb Company Heterocyclic aromatic compounds useful as growth hormone secretagogues
WO2002083109A1 (en) * 2001-04-11 2002-10-24 Ferring Bv Treatment of type 2 diabetes with inhibitors of dipeptidyl peptidase iv
WO2003002596A2 (en) * 2001-06-27 2003-01-09 Probiodrug Ag Use of dipeptidyl peptidase iv inhibitors as therapeutics for neurological disorders
WO2003002553A2 (en) * 2001-06-27 2003-01-09 Smithkline Beecham Corporation Fluoropyrrolidines as dipeptidyl peptidase inhibitors
WO2003015775A1 (en) * 2001-08-17 2003-02-27 Probiodrug Ag New dipeptidyl peptidase iv inhibitors and their uses for lowering blood pressure levels
WO2003037327A1 (en) * 2001-10-26 2003-05-08 F. Hoffmann-La-Roche Ag N-substituted pyrrolidin derivatives as dipeptidyl peptidase iv inhibitors
WO2003045228A2 (en) * 2001-11-26 2003-06-05 Trustees Of Tufts College Methods for treating autoimmune disorders, and reagents related thereto
US6699871B2 (en) 2001-07-06 2004-03-02 Merck & Co., Inc. Beta-amino heterocyclic dipeptidyl peptidase inhibitors for the treatment or prevention of diabetes
WO2003106456A3 (en) * 2002-06-14 2004-03-04 Sanofi Synthelabo Azabicyclo-octane and nonane derivatives with ddp-iv inhibiting activity
US6710040B1 (en) 2002-06-04 2004-03-23 Pfizer Inc. Fluorinated cyclic amides as dipeptidyl peptidase IV inhibitors
WO2004098591A2 (en) 2003-05-05 2004-11-18 Probiodrug Ag Inhibitors of glutaminyl cyclase and their use in the treatment of neurological diseases
WO2004098625A2 (en) 2003-05-05 2004-11-18 Probiodrug Ag Medical use of inhibitors of glutaminyl and glutamate cyclases
US6844316B2 (en) 2001-09-06 2005-01-18 Probiodrug Ag Inhibitors of dipeptidyl peptidase I
WO2005012249A2 (en) 2003-08-01 2005-02-10 Bristol-Myers Squibb Company Adamantyglycine- based inhibitors of dipeptidyl peptidase iv for the treatment of diabetes
EP1506776A1 (en) 2003-08-12 2005-02-16 KeyNeurotek AG Use of enzyme inhibitors of the dipeptidylpeptidase IV and/or of the aminopeptidase n , and pharmaceutical preparations thereof for the prevention or therapy of neurodegenerative diseases
WO2005021536A2 (en) * 2003-08-29 2005-03-10 Sanofi-Aventis Adamantane and azabicyclo-octane and nonane derivatives, process of their preparation and their use as dpp-iv inhibitors
US6890905B2 (en) 2001-04-02 2005-05-10 Prosidion Limited Methods for improving islet signaling in diabetes mellitus and for its prevention
WO2005049027A2 (en) 2003-11-03 2005-06-02 Probiodrug Ag Combinations useful for the treatment of neuronal disorders
WO2005079795A2 (en) * 2004-02-20 2005-09-01 Novartis Ag Dpp-iv inhibitors for treating neurodegeneration and cognitive disorders
US6946480B2 (en) 2002-02-28 2005-09-20 Hans-Ulrich Demuth Glutaminyl based DPIV inhibitors
US6949515B2 (en) 1999-08-24 2005-09-27 Probiodrug Ag Effectors of dipeptidyl peptidase IV for topical use
US7078381B2 (en) 1998-02-02 2006-07-18 Trustees Of Tufts College Method of regulating glucose metabolism, and reagents related thereto
US7098239B2 (en) 2001-06-20 2006-08-29 Merck & Co., Inc Dipeptidyl peptidase inhibitors for the treatment of diabetes
US7101871B2 (en) 2002-10-18 2006-09-05 Merck & Co., Inc. Beta-amino heterocyclic dipeptidyl peptidase inhibitors for the treatment or prevention of diabetes
US7109347B2 (en) 2001-06-27 2006-09-19 Probiodrug Ag Dipeptidyl peptidase IV inhibitors and their uses as anti-cancer agents
US7122555B2 (en) 2003-06-20 2006-10-17 Hoffmann-La Roche Inc. Pyrido [2,1-a] isoquinoline derivatives
US7132443B2 (en) 2001-06-27 2006-11-07 Smithklinebeecham Corporation Fluoropyrrolidines as dipeptidyl peptidase inhibitors
US7157490B2 (en) 2002-11-07 2007-01-02 Merck & Co., Inc. Phenylalanine derivatives as dipeptidyl peptidase inhibitors for the treatment or prevention of diabetes
US7166579B2 (en) 1998-06-24 2007-01-23 Prosidion Limited Prodrugs of DP IV-inhibitors
US7169926B1 (en) 2003-08-13 2007-01-30 Takeda Pharmaceutical Company Limited Dipeptidyl peptidase inhibitors
EP1760076A1 (en) * 2005-09-02 2007-03-07 Ferring B.V. FAP Inhibitors
US7196201B2 (en) 2001-06-27 2007-03-27 Smithkline Beecham Corporation Pyrrolidines as dipeptidyl peptidase inhibitors
US7205409B2 (en) 2003-09-04 2007-04-17 Abbott Laboratories Pharmaceutical compositions as inhibitors of dipeptidyl peptidase-IV (DPP-IV)
US7208498B2 (en) 2002-07-15 2007-04-24 Merck & Co., Inc. Piperidino pyrimidine dipeptidyl peptidase inhibitors for the treatment of diabetes
EA008226B1 (en) * 2000-11-10 2007-04-27 Тайсо Фармасьютикал Ко., Лтд. Intermediates for the synthesis of cyanopyrrolidine derivatives
WO2007072083A1 (en) 2005-12-23 2007-06-28 Prosidion Limited Treatment of type 2 diabetes with a combination of dpiv inhibitor and metformin or thiazolidinedione
US7238724B2 (en) 2002-09-19 2007-07-03 Abbott Laboratories Pharmaceutical compositions as inhibitors of dipeptidyl peptidase-IV (DPP-IV)
US7253172B2 (en) 2001-06-20 2007-08-07 Merck & Co., Inc. Dipeptidyl peptidase inhibitors for the treatment of diabetes
US7259160B2 (en) 2003-07-31 2007-08-21 Merck & Co., Inc. Hexahydrodiazepinones as dipeptidyl peptidase-IV inhibitors for the treatment or prevention of diabetes
US7262207B2 (en) 2002-09-19 2007-08-28 Abbott Laboratories Pharmaceutical compositions as inhibitors of dipeptidyl peptidase-IV (DPP-IV)
US7265128B2 (en) 2003-01-17 2007-09-04 Merck & Co., Inc. 3-amino-4-phenylbutanoic acid derivatives as dipeptidyl peptidase inhibitors for the treatment or prevention of diabetes
WO2007120702A2 (en) 2006-04-11 2007-10-25 Arena Pharmaceuticals, Inc. Use of gpr119 receptor agonists for increasing bone mass and for treating osteoporosis, and combination therapy relating thereto
US7304086B2 (en) 2004-02-05 2007-12-04 Probiodrug Ag Inhibitors of glutaminyl cyclase
JPWO2005053695A1 (en) * 2003-12-04 2007-12-06 エーザイ・アール・アンド・ディー・マネジメント株式会社 Agents for preventing or treating multiple sclerosis
KR100792275B1 (en) * 2006-02-22 2008-01-08 영진약품공업주식회사 Cyclic hydrazide derivatives having ?-amino group, its pharmaceutical acceptable salts and preparation process thereof
US7332520B2 (en) 2003-06-06 2008-02-19 Merck & Co., Inc. Fused indoles as dipeptidyl peptidase inhibitors for the treatment or prevention of diabetes
US7368421B2 (en) 2001-06-27 2008-05-06 Probiodrug Ag Use of dipeptidyl peptidase IV inhibitors in the treatment of multiple sclerosis
WO2008055945A1 (en) 2006-11-09 2008-05-15 Probiodrug Ag 3-hydr0xy-1,5-dihydr0-pyrr0l-2-one derivatives as inhibitors of glutaminyl cyclase for the treatment of ulcer, cancer and other diseases
US7381537B2 (en) 2003-05-05 2008-06-03 Probiodrug Ag Use of inhibitors of glutaminyl cyclases for treatment and prevention of disease
WO2008065141A1 (en) 2006-11-30 2008-06-05 Probiodrug Ag Novel inhibitors of glutaminyl cyclase
US7388019B2 (en) 2003-01-31 2008-06-17 Merck & Co., Inc. 3-amino-4-phenylbutanoic acid derivatives as dipeptidyl peptidase inhibitors for the treatment or prevention of diabetes
US7390809B2 (en) 2002-10-07 2008-06-24 Merck & Co., Inc. Beta-amino heterocyclic dipeptidyl peptidase inhibitors for diabetes
US7456204B2 (en) 2003-06-17 2008-11-25 Merck & Co., Inc. Cyclohexylglycine derivatives as dipeptidyl peptidase inhibitors for the treatment or prevention of diabetes
US7462599B2 (en) 2003-10-15 2008-12-09 Probiodrug Ag Use of effectors of glutaminyl and glutamate cyclases
US7560455B2 (en) 2003-05-14 2009-07-14 Merck & Co., Inc. 3-Amino-4-phenylbutanoic acid derivatives as dipeptidyl peptidase inhibitors for the treatment or prevention of diabetes
US7608577B2 (en) 2001-10-12 2009-10-27 Osi Pharmaceuticals, Inc. Peptidyl ketones as inhibitors of DPIV
EP2116235A1 (en) 2005-01-10 2009-11-11 Arena Pharmaceuticals, Inc. Combination therapy for the treatment of diabetes and conditions related thereto and for the treatment of conditions ameliorated by increasing a blood GLP-1 level
US7622496B2 (en) 2005-12-23 2009-11-24 Zealand Pharma A/S Modified lysine-mimetic compounds
US7667044B2 (en) 2003-11-03 2010-02-23 Probiodrug Ag Compounds for the treatment of neurological disorders
US7671073B2 (en) 2004-05-18 2010-03-02 Merck Sharp & Dohme Corp. Cyclohexylalanine derivatives as dipeptidyl peptidase-IV inhibitors for the treatment or prevention of diabetes
US7678909B1 (en) 2003-08-13 2010-03-16 Takeda Pharmaceutical Company Limited Dipeptidyl peptidase inhibitors
EP2165703A2 (en) 2004-01-20 2010-03-24 Novartis Pharma AG. Direct compression formulation and process
US7687492B2 (en) 2004-05-04 2010-03-30 Merck Sharp & Dohme Corp. 1,2,4-Oxadiazole derivatives as dipeptidyl peptidase-IV inhibitors for the treatment or prevention of diabetes
US7687625B2 (en) 2003-03-25 2010-03-30 Takeda Pharmaceutical Company Limited Dipeptidyl peptidase inhibitors
US7687638B2 (en) 2004-06-04 2010-03-30 Takeda San Diego, Inc. Dipeptidyl peptidase inhibitors
US7718666B2 (en) 2003-06-20 2010-05-18 Hoffmann-La Roche Inc. Pyrido [2,1-a] isoquinoline derivatives
US7723344B2 (en) 2003-08-13 2010-05-25 Takeda San Diego, Inc. Dipeptidyl peptidase inhibitors
US7727964B2 (en) 2001-11-26 2010-06-01 Trustees Of Tufts College Peptidomimetic inhibitors of post-proline cleaving enzymes
US7728146B2 (en) 2006-04-12 2010-06-01 Probiodrug Ag Enzyme inhibitors
US7732446B1 (en) 2004-03-11 2010-06-08 Takeda Pharmaceutical Company Limited Dipeptidyl peptidase inhibitors
US7732162B2 (en) 2003-05-05 2010-06-08 Probiodrug Ag Inhibitors of glutaminyl cyclase for treating neurodegenerative diseases
US7754757B2 (en) 2004-02-05 2010-07-13 Kyorin Pharmaceutical Co., Ltd. Bicycloester derivative
US7781584B2 (en) 2004-03-15 2010-08-24 Takeda Pharmaceutical Company Limited Dipeptidyl peptidase inhibitors
US7790734B2 (en) 2003-09-08 2010-09-07 Takeda Pharmaceutical Company Limited Dipeptidyl peptidase inhibitors
US7825242B2 (en) 2004-07-16 2010-11-02 Takeda Pharmaceutical Company Limted Dipeptidyl peptidase inhibitors
WO2011005929A1 (en) 2009-07-09 2011-01-13 Arena Pharmaceuticals, Inc. Piperidine derivative and its use for the treatment of diabets and obesity
US7872124B2 (en) 2004-12-21 2011-01-18 Takeda Pharmaceutical Company Limited Dipeptidyl peptidase inhibitors
WO2011029920A1 (en) 2009-09-11 2011-03-17 Probiodrug Ag Heterocylcic derivatives as inhibitors of glutaminyl cyclase
US7915427B2 (en) 2006-03-08 2011-03-29 Kyorin Pharmaceuticals Co., Ltd. Process for producing aminoacetyl pyrrolidine carbonitrile derivative and intermediate for production thereof
US7960384B2 (en) 2006-03-28 2011-06-14 Takeda Pharmaceutical Company Limited Dipeptidyl peptidase inhibitors
WO2011107530A2 (en) 2010-03-03 2011-09-09 Probiodrug Ag Novel inhibitors
WO2011110613A1 (en) 2010-03-10 2011-09-15 Probiodrug Ag Heterocyclic inhibitors of glutaminyl cyclase (qc, ec 2.3.2.5)
WO2011127051A1 (en) 2010-04-06 2011-10-13 Arena Pharmaceuticals, Inc. Modulators of the gpr119 receptor and the treatment of disorders related thereto
WO2011131748A2 (en) 2010-04-21 2011-10-27 Probiodrug Ag Novel inhibitors
US8084605B2 (en) 2006-11-29 2011-12-27 Kelly Ron C Polymorphs of succinate salt of 2-[6-(3-amino-piperidin-1-yl)-3-methyl-2,4-dioxo-3,4-dihydro-2H-pyrimidin-1-ylmethy]-4-fluor-benzonitrile and methods of use therefor
US8093236B2 (en) 2007-03-13 2012-01-10 Takeda Pharmaceuticals Company Limited Weekly administration of dipeptidyl peptidase inhibitors
US8143427B2 (en) 2007-03-22 2012-03-27 Kyorin Pharmaceutical Co., Ltd. Method for producing aminoacetylpyrrolidinecarbonitrile derivative
WO2012040279A1 (en) 2010-09-22 2012-03-29 Arena Pharmaceuticals, Inc. Modulators of the gpr119 receptor and the treatment of disorders related thereto
US8222411B2 (en) 2005-09-16 2012-07-17 Takeda Pharmaceutical Company Limited Dipeptidyl peptidase inhibitors
WO2012123563A1 (en) 2011-03-16 2012-09-20 Probiodrug Ag Benz imidazole derivatives as inhibitors of glutaminyl cyclase
WO2012135570A1 (en) 2011-04-01 2012-10-04 Arena Pharmaceuticals, Inc. Modulators of the gpr119 receptor and the treatment of disorders related thereto
WO2012145604A1 (en) 2011-04-22 2012-10-26 Arena Pharmaceuticals, Inc. Modulators of the gpr119 receptor and the treatment of disorders related thereto
WO2012145361A1 (en) 2011-04-19 2012-10-26 Arena Pharmaceuticals, Inc. Modulators of the gpr119 receptor and the treatment of disorders related thereto
WO2012145603A1 (en) 2011-04-22 2012-10-26 Arena Pharmaceuticals, Inc. Modulators of the gpr119 receptor and the treatment of disorders related thereto
US8324383B2 (en) 2006-09-13 2012-12-04 Takeda Pharmaceutical Company Limited Methods of making polymorphs of benzoate salt of 2-[[6-[(3R)-3-amino-1-piperidinyl]-3,4-dihydro-3-methyl-2,4-dioxo-1(2H)-pyrimidinyl]methyl]-benzonitrile
WO2012170702A1 (en) 2011-06-08 2012-12-13 Arena Pharmaceuticals, Inc. Modulators of the gpr119 receptor and the treatment of disorders related thereto
WO2013055910A1 (en) 2011-10-12 2013-04-18 Arena Pharmaceuticals, Inc. Modulators of the gpr119 receptor and the treatment of disorders related thereto
WO2014074668A1 (en) 2012-11-08 2014-05-15 Arena Pharmaceuticals, Inc. Modulators of gpr119 and the treatment of disorders related thereto
WO2014113750A1 (en) 2013-01-18 2014-07-24 Dnj Pharma, Inc. Novel ddp-iv inhibitors
US8883714B2 (en) 2008-04-07 2014-11-11 Arena Pharmaceuticals, Inc. Pharmaceutical compositions comprising GPR119 agonists which act as peptide YY (PYY) secretagogues
US8906901B2 (en) 2005-09-14 2014-12-09 Takeda Pharmaceutical Company Limited Administration of dipeptidyl peptidase inhibitors
US8927590B2 (en) 2006-12-21 2015-01-06 Zealand Pharma A/S Synthesis of pyrrolidine compounds
EP2839832A2 (en) 2003-11-17 2015-02-25 Novartis AG Use of dipeptidyl peptidase IV inhibitors
EP2865670A1 (en) 2007-04-18 2015-04-29 Probiodrug AG Thiourea derivatives as glutaminyl cyclase inhibitors
WO2018162722A1 (en) 2017-03-09 2018-09-13 Deutsches Institut Für Ernährungsforschung Potsdam-Rehbrücke Dpp-4 inhibitors for use in treating bone fractures
EP3461819A1 (en) 2017-09-29 2019-04-03 Probiodrug AG Inhibitors of glutaminyl cyclase
US10555929B2 (en) 2015-03-09 2020-02-11 Coherus Biosciences, Inc. Methods for the treatment of nonalcoholic fatty liver disease and/or lipodystrophy
US11253508B2 (en) 2017-04-03 2022-02-22 Coherus Biosciences, Inc. PPARy agonist for treatment of progressive supranuclear palsy
US11324799B2 (en) 2017-05-05 2022-05-10 Zealand Pharma A/S Gap junction intercellular communication modulators and their use for the treatment of diabetic eye disease
EP4000630A1 (en) 2014-09-03 2022-05-25 Invex Therapeutics Ltd Elevated intracranial pressure treatment

Families Citing this family (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
TW583185B (en) * 2000-06-13 2004-04-11 Novartis Ag N-(substituted glycyl)-2-cyanopyrrolidines and pharmaceutical composition for inhibiting dipeptidyl peptidase-IV (DPP-IV) or for the prevention or treatment of diseases or conditions associated with elevated levels of DPP-IV comprising the same
RU2396269C2 (en) * 2005-12-01 2010-08-10 Ф.Хоффманн-Ля Рош Аг Heteroaryl-substituted piperidine derivatives as hepatic carnitine palmitoyltransferase (l-cpt1) inhibitors
EP1971862B1 (en) 2006-04-11 2010-11-10 Arena Pharmaceuticals, Inc. Methods of using gpr119 receptor to identify compounds useful for increasing bone mass in an individual
KR100848491B1 (en) * 2007-01-16 2008-07-28 영진약품공업주식회사 2-thiazolidine derivatives having beta;-amino group, pharmaceutical acceptable salts and preparation process thereof
CA2732984A1 (en) 2008-08-07 2010-02-11 Kyorin Pharmaceutical Co., Ltd. Process for production of bicyclo[2.2.2]octylamine derivative

Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4684662A (en) * 1983-01-07 1987-08-04 Hoechst Aktiengesellschaft Disubstituted proline derivatives
US4691022A (en) * 1983-06-23 1987-09-01 Hoechst Aktiengesellschaft Process for the preparation of monocyclic, bicyclic and tricyclic aminoacids
US4743616A (en) * 1984-07-31 1988-05-10 Suntory Limited Novel biologically active compound having anti-prolyl endopeptidase activity
US5118811A (en) * 1989-04-13 1992-06-02 Japan Tobacco Inc. Amino acid derivatives possessing prolyl endopeptidase-inhibitory activities
WO1998019998A2 (en) * 1996-11-07 1998-05-14 Novartis Ag N-substituted 2-cyanopyrrolidines

Family Cites Families (11)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
DD296075A5 (en) * 1989-08-07 1991-11-21 Martin-Luther-Universitaet Halle-Wittenberg,De PROCESS FOR THE PREPARATION OF NEW INHIBITORS OF DIPEPTIDYL PEPTIDASE IV
CA2121369C (en) * 1991-10-22 2003-04-29 William W. Bachovchin Inhibitors of dipeptidyl-aminopeptidase type iv
IL111785A0 (en) * 1993-12-03 1995-01-24 Ferring Bv Dp-iv inhibitors and pharmaceutical compositions containing them
US5543396A (en) * 1994-04-28 1996-08-06 Georgia Tech Research Corp. Proline phosphonate derivatives
WO1995034538A2 (en) * 1994-06-10 1995-12-21 Universitaire Instelling Antwerpen Purification of serine proteases and synthetic inhibitors thereof
DE19616486C5 (en) * 1996-04-25 2016-06-30 Royalty Pharma Collection Trust Method for lowering the blood glucose level in mammals
US6011155A (en) * 1996-11-07 2000-01-04 Novartis Ag N-(substituted glycyl)-2-cyanopyrrolidines, pharmaceutical compositions containing them and their use in inhibiting dipeptidyl peptidase-IV
DE19828114A1 (en) * 1998-06-24 2000-01-27 Probiodrug Ges Fuer Arzneim Produgs of unstable inhibitors of dipeptidyl peptidase IV
DE19828113A1 (en) * 1998-06-24 2000-01-05 Probiodrug Ges Fuer Arzneim Prodrugs of Dipeptidyl Peptidase IV Inhibitors
CO5150173A1 (en) * 1998-12-10 2002-04-29 Novartis Ag COMPOUNDS N- (REPLACED GLYCLE) -2-DIPEPTIDYL-IV PEPTIDASE INHIBITING CYANOPIRROLIDINS (DPP-IV) WHICH ARE EFFECTIVE IN THE TREATMENT OF CONDITIONS MEDIATED BY DPP-IV INHIBITION
US6110949A (en) * 1999-06-24 2000-08-29 Novartis Ag N-(substituted glycyl)-4-cyanothiazolidines, pharmaceutical compositions containing them and their use in inhibiting dipeptidyl peptidase-IV

Patent Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4684662A (en) * 1983-01-07 1987-08-04 Hoechst Aktiengesellschaft Disubstituted proline derivatives
US4691022A (en) * 1983-06-23 1987-09-01 Hoechst Aktiengesellschaft Process for the preparation of monocyclic, bicyclic and tricyclic aminoacids
US4743616A (en) * 1984-07-31 1988-05-10 Suntory Limited Novel biologically active compound having anti-prolyl endopeptidase activity
US5118811A (en) * 1989-04-13 1992-06-02 Japan Tobacco Inc. Amino acid derivatives possessing prolyl endopeptidase-inhibitory activities
WO1998019998A2 (en) * 1996-11-07 1998-05-14 Novartis Ag N-substituted 2-cyanopyrrolidines

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
See also references of EP1228061A4 *

Cited By (182)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8513190B2 (en) 1998-02-02 2013-08-20 Trustees Of Tufts College Method of regulating glucose metabolism, and reagents related thereto
US7078381B2 (en) 1998-02-02 2006-07-18 Trustees Of Tufts College Method of regulating glucose metabolism, and reagents related thereto
US9044424B2 (en) 1998-02-02 2015-06-02 1149336 Ontario, Inc. Methods of regulating glucose metabolism, and reagents related thereto
US7459428B2 (en) 1998-02-02 2008-12-02 Trustees Of Tufts College Method of regulating glucose metabolism, and reagents related thereto
US8318669B2 (en) 1998-02-02 2012-11-27 Trustees Of Tufts College Method of regulating glucose metabolism, and reagents related thereto
US7829530B2 (en) 1998-02-02 2010-11-09 Trustees Of Tufts College Method of regulating glucose metabolism, and reagents related thereto
US7166579B2 (en) 1998-06-24 2007-01-23 Prosidion Limited Prodrugs of DP IV-inhibitors
EP1175213A2 (en) * 1999-03-12 2002-01-30 Bristol-Myers Squibb Company Heterocyclic aromatic compounds useful as growth hormone secretagogues
EP1175213A4 (en) * 1999-03-12 2007-04-11 Bristol Myers Squibb Co Heterocyclic aromatic compounds useful as growth hormone secretagogues
US6949515B2 (en) 1999-08-24 2005-09-27 Probiodrug Ag Effectors of dipeptidyl peptidase IV for topical use
US7335645B2 (en) 1999-08-24 2008-02-26 Probiodrug Ag Effectors of dipeptidyl peptidase IV for topical use
EA008226B1 (en) * 2000-11-10 2007-04-27 Тайсо Фармасьютикал Ко., Лтд. Intermediates for the synthesis of cyanopyrrolidine derivatives
US6890905B2 (en) 2001-04-02 2005-05-10 Prosidion Limited Methods for improving islet signaling in diabetes mellitus and for its prevention
WO2002083109A1 (en) * 2001-04-11 2002-10-24 Ferring Bv Treatment of type 2 diabetes with inhibitors of dipeptidyl peptidase iv
US7253172B2 (en) 2001-06-20 2007-08-07 Merck & Co., Inc. Dipeptidyl peptidase inhibitors for the treatment of diabetes
US7098239B2 (en) 2001-06-20 2006-08-29 Merck & Co., Inc Dipeptidyl peptidase inhibitors for the treatment of diabetes
WO2003002553A3 (en) * 2001-06-27 2003-03-27 Smithkline Beecham Corp Fluoropyrrolidines as dipeptidyl peptidase inhibitors
US7132443B2 (en) 2001-06-27 2006-11-07 Smithklinebeecham Corporation Fluoropyrrolidines as dipeptidyl peptidase inhibitors
CN1321682C (en) * 2001-06-27 2007-06-20 前体生物药物股份有限公司 New dipeptidyl peptidase iv inhibitors and their uses as anti-cancer agents
US7109347B2 (en) 2001-06-27 2006-09-19 Probiodrug Ag Dipeptidyl peptidase IV inhibitors and their uses as anti-cancer agents
WO2003002595A2 (en) * 2001-06-27 2003-01-09 Probiodrug Ag Dipeptidyl peptidase iv inhibitors and their uses as anti-cancer agents
WO2003002553A2 (en) * 2001-06-27 2003-01-09 Smithkline Beecham Corporation Fluoropyrrolidines as dipeptidyl peptidase inhibitors
US7368421B2 (en) 2001-06-27 2008-05-06 Probiodrug Ag Use of dipeptidyl peptidase IV inhibitors in the treatment of multiple sclerosis
US7183290B2 (en) 2001-06-27 2007-02-27 Smithkline Beecham Corporation Fluoropyrrolidines as dipeptidyl peptidase inhibitors
WO2003002595A3 (en) * 2001-06-27 2003-05-08 Probiodrug Ag Dipeptidyl peptidase iv inhibitors and their uses as anti-cancer agents
US7368576B2 (en) 2001-06-27 2008-05-06 Probiodrug Ag Dipeptidyl peptidase IV inhibitors and their uses as anti-cancer agents
WO2003002596A2 (en) * 2001-06-27 2003-01-09 Probiodrug Ag Use of dipeptidyl peptidase iv inhibitors as therapeutics for neurological disorders
US7196201B2 (en) 2001-06-27 2007-03-27 Smithkline Beecham Corporation Pyrrolidines as dipeptidyl peptidase inhibitors
WO2003002596A3 (en) * 2001-06-27 2003-07-31 Probiodrug Ag Use of dipeptidyl peptidase iv inhibitors as therapeutics for neurological disorders
US7125873B2 (en) 2001-07-06 2006-10-24 Merck & Co., Inc. Beta-amino tetrahydroimidazo (1, 2-a) pyrazines and tetrahydrotrioazolo (4, 3-a) pyrazines as dipeptidyl peptidase inhibitors for the treatment or prevention of diabetes
US8440668B2 (en) 2001-07-06 2013-05-14 Merck Sharp & Dohme Corp Beta-amino heterocyclic dipeptidyl peptidase inhibitors for the treatment of type 2 diabetes
US6699871B2 (en) 2001-07-06 2004-03-02 Merck & Co., Inc. Beta-amino heterocyclic dipeptidyl peptidase inhibitors for the treatment or prevention of diabetes
US8168637B2 (en) 2001-07-06 2012-05-01 Merck Sharp & Dohme Corp. Beta-amino heterocyclic dipeptidyl peptidase inhibitors for the treatment of diabetes
WO2003015775A1 (en) * 2001-08-17 2003-02-27 Probiodrug Ag New dipeptidyl peptidase iv inhibitors and their uses for lowering blood pressure levels
US6844316B2 (en) 2001-09-06 2005-01-18 Probiodrug Ag Inhibitors of dipeptidyl peptidase I
US7144856B2 (en) 2001-09-06 2006-12-05 Probiodrug Ag Inhibitors of dipeptidyl peptidase I
US7608577B2 (en) 2001-10-12 2009-10-27 Osi Pharmaceuticals, Inc. Peptidyl ketones as inhibitors of DPIV
WO2003037327A1 (en) * 2001-10-26 2003-05-08 F. Hoffmann-La-Roche Ag N-substituted pyrrolidin derivatives as dipeptidyl peptidase iv inhibitors
US6861440B2 (en) 2001-10-26 2005-03-01 Hoffmann-La Roche Inc. DPP IV inhibitors
US7314884B2 (en) 2001-10-26 2008-01-01 Hoffmann-La Roche Inc. DPP IV inhibitors
US7803819B2 (en) 2001-10-26 2010-09-28 Hoffmann-La Roche Inc. DPP IV inhibitors
EA007968B1 (en) * 2001-10-26 2007-02-27 Ф. Хоффманн-Ля Рош Аг N-substituted pyrrolidin derivatives as dipeptidyl peptidase iv inhibitors
US8410053B2 (en) 2001-11-26 2013-04-02 Trustees Of Tufts College Methods for treating autoimmune disorders, and reagents related thereto
WO2003045228A3 (en) * 2001-11-26 2004-10-21 Tufts College Methods for treating autoimmune disorders, and reagents related thereto
US7727964B2 (en) 2001-11-26 2010-06-01 Trustees Of Tufts College Peptidomimetic inhibitors of post-proline cleaving enzymes
US9757400B2 (en) 2001-11-26 2017-09-12 Trustees Of Tufts College Peptidomimetic inhibitors of post-proline cleaving enzymes
WO2003045228A2 (en) * 2001-11-26 2003-06-05 Trustees Of Tufts College Methods for treating autoimmune disorders, and reagents related thereto
EP2769715A3 (en) * 2001-11-26 2014-09-17 Trustees Of Tufts College Methods for treating autoimmune disorders, and reagents related thereto
US6946480B2 (en) 2002-02-28 2005-09-20 Hans-Ulrich Demuth Glutaminyl based DPIV inhibitors
US6710040B1 (en) 2002-06-04 2004-03-23 Pfizer Inc. Fluorinated cyclic amides as dipeptidyl peptidase IV inhibitors
EA008166B1 (en) * 2002-06-14 2007-04-27 Санофи-Авентис Azabicyclo-octane and nonane derivatives with ddp-iv inhibiting activity
CN100347170C (en) * 2002-06-14 2007-11-07 赛诺菲-安万特公司 Azabicyclo-octane and nonane derivatives with DPP-IV inhibiting activity
WO2003106456A3 (en) * 2002-06-14 2004-03-04 Sanofi Synthelabo Azabicyclo-octane and nonane derivatives with ddp-iv inhibiting activity
US7781455B2 (en) 2002-06-14 2010-08-24 Sanofi-Aventis Compounds
JP2005532369A (en) * 2002-06-14 2005-10-27 サノフイ−アベンテイス Azabicyclo-octane and nonane derivatives having DDP-IV inhibitory activity
US7208498B2 (en) 2002-07-15 2007-04-24 Merck & Co., Inc. Piperidino pyrimidine dipeptidyl peptidase inhibitors for the treatment of diabetes
US7238724B2 (en) 2002-09-19 2007-07-03 Abbott Laboratories Pharmaceutical compositions as inhibitors of dipeptidyl peptidase-IV (DPP-IV)
US8637546B2 (en) 2002-09-19 2014-01-28 Abbvie Inc. Pharmaceutical compositions as inhibitors of dipeptidyl peptidase-IV (DDP-IV)
US7262207B2 (en) 2002-09-19 2007-08-28 Abbott Laboratories Pharmaceutical compositions as inhibitors of dipeptidyl peptidase-IV (DPP-IV)
US8119664B2 (en) 2002-09-19 2012-02-21 Abbott Laboratories Pharmaceutical compositions as inhibitors of dipeptidyl peptidase-IV (DPP-IV)
US7390809B2 (en) 2002-10-07 2008-06-24 Merck & Co., Inc. Beta-amino heterocyclic dipeptidyl peptidase inhibitors for diabetes
US7101871B2 (en) 2002-10-18 2006-09-05 Merck & Co., Inc. Beta-amino heterocyclic dipeptidyl peptidase inhibitors for the treatment or prevention of diabetes
US7157490B2 (en) 2002-11-07 2007-01-02 Merck & Co., Inc. Phenylalanine derivatives as dipeptidyl peptidase inhibitors for the treatment or prevention of diabetes
US7265128B2 (en) 2003-01-17 2007-09-04 Merck & Co., Inc. 3-amino-4-phenylbutanoic acid derivatives as dipeptidyl peptidase inhibitors for the treatment or prevention of diabetes
US7388019B2 (en) 2003-01-31 2008-06-17 Merck & Co., Inc. 3-amino-4-phenylbutanoic acid derivatives as dipeptidyl peptidase inhibitors for the treatment or prevention of diabetes
US7687625B2 (en) 2003-03-25 2010-03-30 Takeda Pharmaceutical Company Limited Dipeptidyl peptidase inhibitors
EA009291B1 (en) * 2003-05-05 2007-12-28 Пробиодруг Аг Use of effectors of glutaminyl and glutamate cyclases
AU2004237408C9 (en) * 2003-05-05 2010-04-15 Probiodrug Ag Medical use of inhibitors of glutaminyl and glutamate cyclases
WO2004098591A2 (en) 2003-05-05 2004-11-18 Probiodrug Ag Inhibitors of glutaminyl cyclase and their use in the treatment of neurological diseases
US7732162B2 (en) 2003-05-05 2010-06-08 Probiodrug Ag Inhibitors of glutaminyl cyclase for treating neurodegenerative diseases
EP2206496A1 (en) 2003-05-05 2010-07-14 Probiodrug AG Medical use of inhibitors of glutaminyl and glutamate cyclases
WO2004098625A3 (en) * 2003-05-05 2005-06-23 Probiodrug Ag Medical use of inhibitors of glutaminyl and glutamate cyclases
AU2004237408C1 (en) * 2003-05-05 2010-04-01 Probiodrug Ag Medical use of inhibitors of glutaminyl and glutamate cyclases
DE202004021723U1 (en) 2003-05-05 2010-07-15 Probiodrug Ag Medical use of inhibitors of glutaminyl and glutamate cyclases
US7381537B2 (en) 2003-05-05 2008-06-03 Probiodrug Ag Use of inhibitors of glutaminyl cyclases for treatment and prevention of disease
US8685664B2 (en) 2003-05-05 2014-04-01 Probiodrug Ag Use of inhibtors of glutaminyl cyclases for treatment and prevention of disease
AU2004237408B2 (en) * 2003-05-05 2009-10-01 Probiodrug Ag Medical use of inhibitors of glutaminyl and glutamate cyclases
US8809010B2 (en) 2003-05-05 2014-08-19 Probiodrug Ag Method for prophylactic treatment of alzheimer's disease using inhibitors of glutaminyl cyclase and glutamate cyclases
EP1961416A1 (en) 2003-05-05 2008-08-27 Probiodrug AG Medical use of inhibitors of glutaminyl and glutamate cyclases
WO2004098625A2 (en) 2003-05-05 2004-11-18 Probiodrug Ag Medical use of inhibitors of glutaminyl and glutamate cyclases
US7560455B2 (en) 2003-05-14 2009-07-14 Merck & Co., Inc. 3-Amino-4-phenylbutanoic acid derivatives as dipeptidyl peptidase inhibitors for the treatment or prevention of diabetes
US7332520B2 (en) 2003-06-06 2008-02-19 Merck & Co., Inc. Fused indoles as dipeptidyl peptidase inhibitors for the treatment or prevention of diabetes
US7456204B2 (en) 2003-06-17 2008-11-25 Merck & Co., Inc. Cyclohexylglycine derivatives as dipeptidyl peptidase inhibitors for the treatment or prevention of diabetes
US7718666B2 (en) 2003-06-20 2010-05-18 Hoffmann-La Roche Inc. Pyrido [2,1-a] isoquinoline derivatives
US7122555B2 (en) 2003-06-20 2006-10-17 Hoffmann-La Roche Inc. Pyrido [2,1-a] isoquinoline derivatives
US7259160B2 (en) 2003-07-31 2007-08-21 Merck & Co., Inc. Hexahydrodiazepinones as dipeptidyl peptidase-IV inhibitors for the treatment or prevention of diabetes
EP1997489A1 (en) * 2003-08-01 2008-12-03 Bristol-Myers Squibb Company Adamantylglycine derivatives as inhibitors of dipeptidyl peptidase IV for the treatment of diabetes
WO2005012249A3 (en) * 2003-08-01 2005-05-06 Bristol Myers Squibb Co Adamantyglycine- based inhibitors of dipeptidyl peptidase iv for the treatment of diabetes
WO2005012249A2 (en) 2003-08-01 2005-02-10 Bristol-Myers Squibb Company Adamantyglycine- based inhibitors of dipeptidyl peptidase iv for the treatment of diabetes
US7291599B2 (en) 2003-08-12 2007-11-06 Keyneurotek Ag Use of inhibitors of enzymes having activities of amino peptidase N and/or dipeptidyl peptidase IV and of pharmaceutical preparations thereof for a therapy and prevention of chronical neurodegenerative diseases
EP1506776A1 (en) 2003-08-12 2005-02-16 KeyNeurotek AG Use of enzyme inhibitors of the dipeptidylpeptidase IV and/or of the aminopeptidase n , and pharmaceutical preparations thereof for the prevention or therapy of neurodegenerative diseases
US7790736B2 (en) 2003-08-13 2010-09-07 Takeda Pharmaceutical Company Limited Dipeptidyl peptidase inhibitors
US7678909B1 (en) 2003-08-13 2010-03-16 Takeda Pharmaceutical Company Limited Dipeptidyl peptidase inhibitors
US7723344B2 (en) 2003-08-13 2010-05-25 Takeda San Diego, Inc. Dipeptidyl peptidase inhibitors
US7169926B1 (en) 2003-08-13 2007-01-30 Takeda Pharmaceutical Company Limited Dipeptidyl peptidase inhibitors
US7652021B2 (en) 2003-08-29 2010-01-26 Sanofi-Aventis Compounds useful for DPP-IV enzyme inhibition
WO2005021536A2 (en) * 2003-08-29 2005-03-10 Sanofi-Aventis Adamantane and azabicyclo-octane and nonane derivatives, process of their preparation and their use as dpp-iv inhibitors
AU2004268832B2 (en) * 2003-08-29 2011-04-28 Sanofi-Aventis Adamantane and azabicyclo-octane and nonane derivatives, process of their preparation and their use as DPP-IV inhibitors
WO2005021536A3 (en) * 2003-08-29 2005-10-13 Sanofi Aventis Adamantane and azabicyclo-octane and nonane derivatives, process of their preparation and their use as dpp-iv inhibitors
US7205409B2 (en) 2003-09-04 2007-04-17 Abbott Laboratories Pharmaceutical compositions as inhibitors of dipeptidyl peptidase-IV (DPP-IV)
US7790734B2 (en) 2003-09-08 2010-09-07 Takeda Pharmaceutical Company Limited Dipeptidyl peptidase inhibitors
EP2289498A1 (en) 2003-10-15 2011-03-02 Probiodrug AG Use of inhibitors of glutaminyl clyclase
US7462599B2 (en) 2003-10-15 2008-12-09 Probiodrug Ag Use of effectors of glutaminyl and glutamate cyclases
US7667044B2 (en) 2003-11-03 2010-02-23 Probiodrug Ag Compounds for the treatment of neurological disorders
WO2005049027A2 (en) 2003-11-03 2005-06-02 Probiodrug Ag Combinations useful for the treatment of neuronal disorders
EP2338490A2 (en) 2003-11-03 2011-06-29 Probiodrug AG Combinations Useful for the Treatment of Neuronal Disorders
EP2839832A2 (en) 2003-11-17 2015-02-25 Novartis AG Use of dipeptidyl peptidase IV inhibitors
JPWO2005053695A1 (en) * 2003-12-04 2007-12-06 エーザイ・アール・アンド・ディー・マネジメント株式会社 Agents for preventing or treating multiple sclerosis
EP2165703A2 (en) 2004-01-20 2010-03-24 Novartis Pharma AG. Direct compression formulation and process
EP3023095A1 (en) 2004-01-20 2016-05-25 Novartis AG Direct compression formulation and process
EP3738585A1 (en) 2004-01-20 2020-11-18 Novartis Ag Direct compression formulation and process
EP3366283A1 (en) 2004-01-20 2018-08-29 Novartis AG Direct compression formulation and process
US8053465B2 (en) 2004-02-05 2011-11-08 Kyorin Pharmaceutical Co., Ltd. Bicycloester derivative
US7304086B2 (en) 2004-02-05 2007-12-04 Probiodrug Ag Inhibitors of glutaminyl cyclase
US7754757B2 (en) 2004-02-05 2010-07-13 Kyorin Pharmaceutical Co., Ltd. Bicycloester derivative
WO2005079795A3 (en) * 2004-02-20 2005-11-10 Novartis Ag Dpp-iv inhibitors for treating neurodegeneration and cognitive disorders
WO2005079795A2 (en) * 2004-02-20 2005-09-01 Novartis Ag Dpp-iv inhibitors for treating neurodegeneration and cognitive disorders
US7732446B1 (en) 2004-03-11 2010-06-08 Takeda Pharmaceutical Company Limited Dipeptidyl peptidase inhibitors
US8188275B2 (en) 2004-03-15 2012-05-29 Takeda Pharmaceutical Company Limited Dipeptidyl peptidase inhibitors
US7906523B2 (en) 2004-03-15 2011-03-15 Takeda Pharmaceutical Company Limited Dipeptidyl peptidase inhibitors
US8173663B2 (en) 2004-03-15 2012-05-08 Takeda Pharmaceutical Company Limited Dipeptidyl peptidase inhibitors
US7781584B2 (en) 2004-03-15 2010-08-24 Takeda Pharmaceutical Company Limited Dipeptidyl peptidase inhibitors
US8329900B2 (en) 2004-03-15 2012-12-11 Takeda Pharmaceutical Company Limited Dipeptidyl peptidase inhibitors
US8288539B2 (en) 2004-03-15 2012-10-16 Takeda Pharmaceutical Company Limited Dipeptidyl peptidase inhibitors
US7807689B2 (en) 2004-03-15 2010-10-05 Takeda Pharmaceutical Company Limited Dipeptidyl peptidase inhibitors
US7687492B2 (en) 2004-05-04 2010-03-30 Merck Sharp & Dohme Corp. 1,2,4-Oxadiazole derivatives as dipeptidyl peptidase-IV inhibitors for the treatment or prevention of diabetes
US7671073B2 (en) 2004-05-18 2010-03-02 Merck Sharp & Dohme Corp. Cyclohexylalanine derivatives as dipeptidyl peptidase-IV inhibitors for the treatment or prevention of diabetes
US7687638B2 (en) 2004-06-04 2010-03-30 Takeda San Diego, Inc. Dipeptidyl peptidase inhibitors
US7825242B2 (en) 2004-07-16 2010-11-02 Takeda Pharmaceutical Company Limted Dipeptidyl peptidase inhibitors
US7872124B2 (en) 2004-12-21 2011-01-18 Takeda Pharmaceutical Company Limited Dipeptidyl peptidase inhibitors
EP2116235A1 (en) 2005-01-10 2009-11-11 Arena Pharmaceuticals, Inc. Combination therapy for the treatment of diabetes and conditions related thereto and for the treatment of conditions ameliorated by increasing a blood GLP-1 level
WO2007085895A3 (en) * 2005-09-02 2008-04-17 Ferring Bv Fap inhibitors
EP1760076A1 (en) * 2005-09-02 2007-03-07 Ferring B.V. FAP Inhibitors
WO2007085895A2 (en) * 2005-09-02 2007-08-02 Ferring B.V. Fap inhibitors
US8183280B2 (en) 2005-09-02 2012-05-22 Vantia Limited FAP inhibitors
US8906901B2 (en) 2005-09-14 2014-12-09 Takeda Pharmaceutical Company Limited Administration of dipeptidyl peptidase inhibitors
US8222411B2 (en) 2005-09-16 2012-07-17 Takeda Pharmaceutical Company Limited Dipeptidyl peptidase inhibitors
US8431540B2 (en) 2005-12-23 2013-04-30 Zealand Pharma A/S Modified lysine-mimetic compounds
WO2007072083A1 (en) 2005-12-23 2007-06-28 Prosidion Limited Treatment of type 2 diabetes with a combination of dpiv inhibitor and metformin or thiazolidinedione
US7622496B2 (en) 2005-12-23 2009-11-24 Zealand Pharma A/S Modified lysine-mimetic compounds
KR100792275B1 (en) * 2006-02-22 2008-01-08 영진약품공업주식회사 Cyclic hydrazide derivatives having ?-amino group, its pharmaceutical acceptable salts and preparation process thereof
US7915427B2 (en) 2006-03-08 2011-03-29 Kyorin Pharmaceuticals Co., Ltd. Process for producing aminoacetyl pyrrolidine carbonitrile derivative and intermediate for production thereof
US7960384B2 (en) 2006-03-28 2011-06-14 Takeda Pharmaceutical Company Limited Dipeptidyl peptidase inhibitors
EP2253311A2 (en) 2006-04-11 2010-11-24 Arena Pharmaceuticals, Inc. Use of GPR119 receptor agonists for increasing bone mass and for treating osteoporosis, as well as combination therapy relating thereto
WO2007120702A2 (en) 2006-04-11 2007-10-25 Arena Pharmaceuticals, Inc. Use of gpr119 receptor agonists for increasing bone mass and for treating osteoporosis, and combination therapy relating thereto
US7728146B2 (en) 2006-04-12 2010-06-01 Probiodrug Ag Enzyme inhibitors
US8324383B2 (en) 2006-09-13 2012-12-04 Takeda Pharmaceutical Company Limited Methods of making polymorphs of benzoate salt of 2-[[6-[(3R)-3-amino-1-piperidinyl]-3,4-dihydro-3-methyl-2,4-dioxo-1(2H)-pyrimidinyl]methyl]-benzonitrile
WO2008055945A1 (en) 2006-11-09 2008-05-15 Probiodrug Ag 3-hydr0xy-1,5-dihydr0-pyrr0l-2-one derivatives as inhibitors of glutaminyl cyclase for the treatment of ulcer, cancer and other diseases
US8084605B2 (en) 2006-11-29 2011-12-27 Kelly Ron C Polymorphs of succinate salt of 2-[6-(3-amino-piperidin-1-yl)-3-methyl-2,4-dioxo-3,4-dihydro-2H-pyrimidin-1-ylmethy]-4-fluor-benzonitrile and methods of use therefor
WO2008065141A1 (en) 2006-11-30 2008-06-05 Probiodrug Ag Novel inhibitors of glutaminyl cyclase
US9469609B2 (en) 2006-12-21 2016-10-18 Zealand Pharma A/S Synthesis of pyrrolidine compounds
US8927590B2 (en) 2006-12-21 2015-01-06 Zealand Pharma A/S Synthesis of pyrrolidine compounds
US8093236B2 (en) 2007-03-13 2012-01-10 Takeda Pharmaceuticals Company Limited Weekly administration of dipeptidyl peptidase inhibitors
US8143427B2 (en) 2007-03-22 2012-03-27 Kyorin Pharmaceutical Co., Ltd. Method for producing aminoacetylpyrrolidinecarbonitrile derivative
EP2865670A1 (en) 2007-04-18 2015-04-29 Probiodrug AG Thiourea derivatives as glutaminyl cyclase inhibitors
US8883714B2 (en) 2008-04-07 2014-11-11 Arena Pharmaceuticals, Inc. Pharmaceutical compositions comprising GPR119 agonists which act as peptide YY (PYY) secretagogues
WO2011005929A1 (en) 2009-07-09 2011-01-13 Arena Pharmaceuticals, Inc. Piperidine derivative and its use for the treatment of diabets and obesity
WO2011029920A1 (en) 2009-09-11 2011-03-17 Probiodrug Ag Heterocylcic derivatives as inhibitors of glutaminyl cyclase
WO2011107530A2 (en) 2010-03-03 2011-09-09 Probiodrug Ag Novel inhibitors
WO2011110613A1 (en) 2010-03-10 2011-09-15 Probiodrug Ag Heterocyclic inhibitors of glutaminyl cyclase (qc, ec 2.3.2.5)
WO2011127051A1 (en) 2010-04-06 2011-10-13 Arena Pharmaceuticals, Inc. Modulators of the gpr119 receptor and the treatment of disorders related thereto
WO2011131748A2 (en) 2010-04-21 2011-10-27 Probiodrug Ag Novel inhibitors
WO2012040279A1 (en) 2010-09-22 2012-03-29 Arena Pharmaceuticals, Inc. Modulators of the gpr119 receptor and the treatment of disorders related thereto
EP3323818A1 (en) 2010-09-22 2018-05-23 Arena Pharmaceuticals, Inc. Modulators of the gpr119 receptor and the treatment of disorders related thereto
WO2012123563A1 (en) 2011-03-16 2012-09-20 Probiodrug Ag Benz imidazole derivatives as inhibitors of glutaminyl cyclase
WO2012135570A1 (en) 2011-04-01 2012-10-04 Arena Pharmaceuticals, Inc. Modulators of the gpr119 receptor and the treatment of disorders related thereto
WO2012145361A1 (en) 2011-04-19 2012-10-26 Arena Pharmaceuticals, Inc. Modulators of the gpr119 receptor and the treatment of disorders related thereto
WO2012145603A1 (en) 2011-04-22 2012-10-26 Arena Pharmaceuticals, Inc. Modulators of the gpr119 receptor and the treatment of disorders related thereto
WO2012145604A1 (en) 2011-04-22 2012-10-26 Arena Pharmaceuticals, Inc. Modulators of the gpr119 receptor and the treatment of disorders related thereto
WO2012170702A1 (en) 2011-06-08 2012-12-13 Arena Pharmaceuticals, Inc. Modulators of the gpr119 receptor and the treatment of disorders related thereto
WO2013055910A1 (en) 2011-10-12 2013-04-18 Arena Pharmaceuticals, Inc. Modulators of the gpr119 receptor and the treatment of disorders related thereto
US9115082B2 (en) 2012-01-18 2015-08-25 Catherine Yang Dipeptidyl-peptidase-IV inhibitors for treatment of type 2 diabetes complex with hypertension
WO2014074668A1 (en) 2012-11-08 2014-05-15 Arena Pharmaceuticals, Inc. Modulators of gpr119 and the treatment of disorders related thereto
WO2014113750A1 (en) 2013-01-18 2014-07-24 Dnj Pharma, Inc. Novel ddp-iv inhibitors
EP4000630A1 (en) 2014-09-03 2022-05-25 Invex Therapeutics Ltd Elevated intracranial pressure treatment
US10555929B2 (en) 2015-03-09 2020-02-11 Coherus Biosciences, Inc. Methods for the treatment of nonalcoholic fatty liver disease and/or lipodystrophy
US10772865B2 (en) 2015-03-09 2020-09-15 Coherus Biosciences, Inc. Methods for the treatment of nonalcoholic fatty liver disease and/or lipodystrophy
US11400072B2 (en) 2015-03-09 2022-08-02 Coherus Biosciences, Inc. Methods for the treatment of nonalcoholic fatty liver disease and/or lipodystrophy
WO2018162722A1 (en) 2017-03-09 2018-09-13 Deutsches Institut Für Ernährungsforschung Potsdam-Rehbrücke Dpp-4 inhibitors for use in treating bone fractures
US11253508B2 (en) 2017-04-03 2022-02-22 Coherus Biosciences, Inc. PPARy agonist for treatment of progressive supranuclear palsy
US11324799B2 (en) 2017-05-05 2022-05-10 Zealand Pharma A/S Gap junction intercellular communication modulators and their use for the treatment of diabetic eye disease
EP3461819A1 (en) 2017-09-29 2019-04-03 Probiodrug AG Inhibitors of glutaminyl cyclase

Also Published As

Publication number Publication date
CA2390231A1 (en) 2001-05-17
AU1916401A (en) 2001-06-06
JP2003535034A (en) 2003-11-25
EP1228061A1 (en) 2002-08-07
EP1228061A4 (en) 2004-12-15

Similar Documents

Publication Publication Date Title
EP1228061A1 (en) Dipeptidyl peptidase iv inhibitors and methods of making and using dipeptidyl peptidase iv inhibitors
US20040152745A1 (en) Dipeptidyl peptidase IV inhibitors and methods of making and using dipeptidyl peptidase IV inhibitors
JP6892218B2 (en) How to treat diseases caused by drug delivery conjugates and PSMA-expressing cells
US7553830B2 (en) Compositions for the delivery of negatively charged molecules
TW201034690A (en) Technetium-and rhenium-bis (heteroaryl) complexes and methods of use thereof for inhibiting PSMA
JP2009500423A (en) Fibroblast activation protein alpha inhibitor
KR20000035925A (en) Phosphinic acid amides as matrix metalloprotease inhibitors
CZ2004973A3 (en) Glutaminyl based DPIV inhibitors
IL139862A (en) Thiazolidine or pyrrolidine
JP2000508319A (en) αvβ3 antagonist
CN102159553A (en) Targeted nitroxide agents
JP2018048178A (en) Oxabicycloheptanes, and oxabicycloheptanes for treatment of reperfusion injury
AU2014203872A1 (en) Use of fatty acid niacin conjugates for treating diseases
JP3832229B2 (en) Phenylethenesulfonamide derivative-containing medicine
JP2021521106A (en) HSP90 targeted conjugate and its formulation
US20130338372A1 (en) Substituted Imidazoline Compounds
JP2004026678A (en) Therapeutic agent for type 2 diabetes
JP2016537353A (en) Fatty acid niacin complex
AU3171293A (en) Use of renin inhibitors for the treatment of glaucoma
US11136295B2 (en) D-amphetamine compounds, compositions, and processes for making and using the same
KR20010083122A (en) Novel remedies for allergic diseases
JP2003503476A (en) Cyclic amide derivative
EP3607948A1 (en) Tissue transglutaminase modulators for medicinal use
CA3123368A1 (en) Mitochondria-targeting peptides
MXPA02004679A (en) Dipeptidyl peptidase iv inhibitors and methods of making and using dipeptidyl peptidase iv inhibitors

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A1

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

AL Designated countries for regional patents

Kind code of ref document: A1

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

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

Ref document number: 2390231

Country of ref document: CA

WWE Wipo information: entry into national phase

Ref document number: 19164/01

Country of ref document: AU

WWE Wipo information: entry into national phase

Ref document number: 2000982093

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: PA/a/2002/004679

Country of ref document: MX

ENP Entry into the national phase

Ref country code: JP

Ref document number: 2001 536541

Kind code of ref document: A

Format of ref document f/p: F

WWP Wipo information: published in national office

Ref document number: 2000982093

Country of ref document: EP

REG Reference to national code

Ref country code: DE

Ref legal event code: 8642

WWW Wipo information: withdrawn in national office

Ref document number: 2000982093

Country of ref document: EP

DPE2 Request for preliminary examination filed before expiration of 19th month from priority date (pct application filed from 20040101)