WO2008119005A1 - 3,4-dehydro-proline-containing inhibitors of dipeptidylpeptidase iv - Google Patents

3,4-dehydro-proline-containing inhibitors of dipeptidylpeptidase iv Download PDF

Info

Publication number
WO2008119005A1
WO2008119005A1 PCT/US2008/058378 US2008058378W WO2008119005A1 WO 2008119005 A1 WO2008119005 A1 WO 2008119005A1 US 2008058378 W US2008058378 W US 2008058378W WO 2008119005 A1 WO2008119005 A1 WO 2008119005A1
Authority
WO
WIPO (PCT)
Prior art keywords
compound
group
alkyl
alkenyl
alkynyl
Prior art date
Application number
PCT/US2008/058378
Other languages
French (fr)
Inventor
William W. Bachovchin
Hung-Sen Lai
Wengen Wu
Original Assignee
Trustees Of Tufts College
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 Trustees Of Tufts College filed Critical Trustees Of Tufts College
Publication of WO2008119005A1 publication Critical patent/WO2008119005A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07FACYCLIC, CARBOCYCLIC OR HETEROCYCLIC COMPOUNDS CONTAINING ELEMENTS OTHER THAN CARBON, HYDROGEN, HALOGEN, OXYGEN, NITROGEN, SULFUR, SELENIUM OR TELLURIUM
    • C07F5/00Compounds containing elements of Groups 3 or 13 of the Periodic System
    • C07F5/02Boron compounds
    • C07F5/025Boronic and borinic acid compounds
    • 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

Definitions

  • Proteases are enzymes that cleave proteins at specific peptide bonds. Proteases can be classified into four generic classes: serine; thiol or cysteinyl; acid or aspartyl; and metalloproteases (Cuypers et al, J. Biol. Chem. 1982, 257, 7086. Proteases are essential to a variety of biological activities, such as digestion, formation and dissolution of blood clots, reproduction, and immune reaction to foreign cells and organisms. However, aberrant proteolysis is associated with a number of diseases in humans and other mammals. Accordingly, it is often beneficial to disrupt the function of one or more proteolytic enzymes in the course of treating a patient.
  • the binding site for a peptide substrate consists of a series of "specificity subsites" across the surface of the enzyme.
  • the term "specificity subsite” refers to a pocket or other site on the enzyme capable of interacting with a portion of a substrate for the enzyme.
  • proteases e.g., serine and cysteine proteases
  • the present application utilizes the nomenclature of Schechter and Berger (Biochem. Biophys. Res. Commun. 1967, 27, 157-162).
  • the individual amino acid residues of a substrate or inhibitor are designated Pl, P2, etc. and the corresponding subsites of the enzyme are designated Sl, S2, etc., starting with the carboxy terminal residue produced in the cleavage reaction.
  • the scissile bond of the substrate is the amide bond between Pl-Pl ' of the substrate.
  • the Xaa3 residue is referred to as the Pl residue and binds to the Sl subsite of the enzyme
  • Xaa2 is referred to as the P2 residue and binds to the S2 subsite, and so forth.
  • Dipeptidyl peptidase IV is a serine protease that cleaves N- terminal dipeptides from a peptide chain containing, preferably, a pro line residue in the penultimate position, i.e., in the Pl position.
  • DPIV belongs to a group of cell-membrane- associated peptidases and, like the majority of cell-surface peptidases, is a type II integral membrane protein, being anchored to the plasma membrane by its signal sequence.
  • DPIV is found in a variety of differentiated mammalian epithelia, endothelia and hematopoetic cells and tissues, including those of lymphoid origin where it is found specifically on the surface of CD4 + T cells.
  • DPIV has been identified as leukocyte differentiation marker CD26.
  • One aspect of the present invention relates to inhibitors of post-pro line protease enzymes, such as inhibitors of dipeptidyl peptidase IV (DPIV), as well as pharmaceutical compositions thereof, and methods for using such inhibitors.
  • the inhibitors of the present invention include a dehydroproline residue having a functional group that covalently reacts with the active site of DPIV.
  • One aspect of the invention is a protease inhibitor represented by Formula (I):
  • R 4 represents H, alkyl, alkenyl, alkynyl, -C(X 1 )(X 2 )(X 3 ), -(CH 2 )Di-R 43 , -(CH 2 )Ii-OH, -(CH 2 )n-O-alkyl, -(CH 2 )n-O-alkenyl, -(CH 2 )n-O-alkynyl, -(CH 2 )U-O-(CH 2 )In-R 43 , -(CH 2 )n-SH, -(CH 2 )n-S-alkyl, -(CH 2 )n-S-alkenyl,
  • R43 represents independently for each occurrence aryl, aralkyl, or non-aromatic hetereocyclyl
  • R-44 represents independently for each occurrence H, alkyl, alkenyl, aryl, aralkyl, or non-aromatic heterocyclyl
  • Yi and Y 2 independently represent OH, or a group capable of being hydro lyzed to a hydroxyl group; or taken together with the boron to which they are bonded form a 5- membered to 8-membered ring comprising said boron and two oxygen atoms bonded to said boron;
  • R41 is selected from the group consisting of N 3 , SH, NH 2 , NO 2 , and -ORg ' ;
  • R4 2 is selected from the group consisting of hydrogen, lower alkyl, amine, and -OR 44 , or a pharmaceutically acceptable salt; or R 41 and R 42 taken together with the phosphorous atom to which they are attached complete a heterocyclic ring having from 5 to 8 atoms in the ring structure;
  • X l is halogen;
  • X 2 and X 3 is hydrogen or halogen; Ri is selected from a C-terminally linked amino acid residue or amino acid analog, a
  • R 2 is absent or represents one or more substituents attached at positions 3, 4, or 5 of the dehydroproline ring, each of which is independently selected from the group consisting of hydrogen, halogen, hydroxyl, lower alkyl, lower alkenyl, lower alkynyl, lower alkoxy, lower hydroxyalkyl, lower alkoxyalkyl, carbonyl, carboxyl, ester, formate, ketone, thiocarbonyl, thioester, thioacetate, thioformate, amino, acylamino, amido, nitro, sulfate, sulfonate, sulfonamido, -(CH 2 X n -R 51 , -(CH 2 X n -OH, -(CH 2 ) m -O-lower alkyl, -(CH 2 X n -O- lower alkenyl, -(CH 2 ) n -O-(CH 2 ) m
  • R 3 is selected from the group consisting of hydrogen, halogen, lower alkyl, lower alkenyl, lower alkynyl, carbonyl, thiocarbonyl, amino, acylamino, amido, nitro, sulfate, sulfonate, a sulfonamido, -(CH 2 X n -R 51 , -(CH 2 X n -OH, -(CH 2 ) m -O-lower alkyl, -(CH 2 X n -O- lower alkenyl, -(CH 2 ) n -O-(CH 2 ) m -R 5 ⁇ , -(CH 2 X n -SH, -(CH 2 ) m -S-lower alkyl, -(CH 2 X n -S- lower alkenyl, or -(CH 2 ) n -S-(CH 2 ) m -R 5
  • R 5 is selected from the group consisting of hydrogen, halogen, alkyl, alkenyl, alkynyl, aryl, -(CH 2 ) m -R 51 , -(CH 2 ) m -OH, -(CH 2 ) m -O-alkyl, -(CH 2 ) m -O-alkenyl, -(CH 2 ) m -O-alkynyl, -(CH 2 ) m -O-(CH 2 ) m -R 51 , -(CH 2 ) m -SH, -(CH 2 ) m -S-alkyl, -(CH 2 ) m -S- alkenyl, -(CH 2 ) m -S-alkynyl, -(CH 2 ) m -S-(CH 2 ) m -R 5 i,
  • R51 represents for each occurrence aryl, aralkyl, or non-aromatic heterocyclyl;
  • R 52 and R 53 independently represent hydrogen, alkyl, alkenyl, -(CH 2 ) m -R 5 i,
  • W is a group of formula -B(Yi)(Y 2 ).
  • Ri is a C-terminally linked amino acid residue or amino acid analog or a C-terminally linked peptide or peptide analog. In certain embodiments, such as when W and Ri have the values described above,
  • R 3 is hydrogen or a lower alkyl group.
  • the compound is represented by:
  • Another aspect of the invention provides a protease inhibitor having a structure represented by Formula (II):
  • R 2 is absent or represents one or more substituents attached at positions 3, 4, or 5 of the dehydroproline ring, each of which is independently selected from the group consisting of hydrogen, halogen, hydroxyl, lower alkyl, lower alkenyl, lower alkynyl, lower alkoxy, lower hydroxyalkyl, lower alkoxyalkyl, carbonyl, carboxyl, ester, formate, ketone, thiocarbonyl, thioester, thioacetate, thioformate, amino, acylamino, amido, nitro, sulfate, sulfonate, sulfonamido, -(CH 2 X n -R 43 , -(CH 2 X n -OH, -(CH 2 ) m -O-lower alkyl, -(CH 2 X n -O- lower alkenyl, -(CH 2 X 1 -O-(CH 2 X n
  • Rioi is selected from the group consisting of H, alkyl, alkoxy, alkenyl, alkynyl, amino, alkylamino, acylamino, cyano, sulfonylamino, acyloxy, aryl, non-aromatic heterocyclyl, heteroaryl, and a polypeptide chain of 1 to 8 amino acid residues;
  • Rio2 is selected from the group consisting of H, lower alkyl, and aralkyl
  • Rio3 is selected from the group consisting of H, halogen, and alkyl, or R102 and R103 together with the atoms to which they are attached, form a 3- to 6-membered heterocyclic ring;
  • R105 is selected from the group consisting of H, halogen, lower alkyl, and aralkyl;
  • R 4 represents H, alkyl, alkenyl, alkynyl, -C(X 1 )(X 2 )(X 3 ), -(CH 2 )Hi-R 43 , -(CH 2 )Ii-OH, -(CH 2 )n-O-alkyl, -(CH 2 )n-O-alkenyl, -(CH 2 )n-O-alkynyl, -(CH 2 )H-O-(CH 2 )Hi-R 43 , -(CH 2 )n-SH, -(CH 2 )n-S-alkyl, -(CH 2 )n-S-alkenyl, -(CH 2 )n-S-alkynyl, -(CH 2 )H-S-(CH 2 )Hi-R 43 , -C(O)C(O)NH 2 , or -C(O)C(O)OR 44 ;
  • R 43 represents independently for each occurrence aryl, aralkyl, or non-aromatic hetereocyclyl
  • R 44 independently for each occurrence is selected from the group consisting of H, alkyl, alkenyl, aryl, aralkyl, and non-aromatic heterocyclyl;
  • Yi and Y 2 independently represent OH, or a group capable of being hydro lyzed to a hydroxyl group; or taken together with the boron to which they are bonded form a 5- membered to 8-membered ring comprising said boron and two oxygen atoms bonded to said boron;
  • R 40 is O or S;
  • R41 is selected from the group consisting of N 3 , SH, NH 2 , NO 2 and -OR ⁇ ;
  • R 42 is selected from the group consisting of H, lower alkyl, amine, and -OR 44 , or a pharmaceutically acceptable salt; or R 41 and R 42 taken together with the phosphorous atom to which they are attached complete a heterocyclic ring having from 5 to 8 atoms in the ring structure;
  • X 1 is halogen;
  • X 2 and X 3 is hydrogen or halogen; and n is an integer in the range of 2 to 6;
  • R107 is selected from the group consisting of H, aryl, alkyl, aralkyl, non-aromatic heterocyclyl, heteroaryl, heteroaralkyl, and polypeptide chains of 1 to 8 amino acid residues;
  • L is absent or is selected from the group consisting of alkyl, alkenyl, alkynyl, -(CH 2 ) m O(CH 2 ) m -, -(CH 2 ) m NR 2 (CH 2 ) m -, and -(CH 2 ) m S(CH 2 ) m -;
  • X is absent or is selected from the group consisting of -N(R 107 )-, -O-, and -S-;
  • Rio ⁇ is a group of formula -B(Yi)(Y 2 ).
  • one or more of L, X and Y is absent.
  • two or more of L, X and Y are absent.
  • each of L, X and Y is absent.
  • Rioi is generally -H.
  • one or both of Rio 3 and R105 are -H in compound represented by Formula (II). More typically, both Ri 03 and R 105 are -H.
  • the protease inhibitor inhibits DPIV with a K 1 of 50 nm or less. In other embodiments, the protease inhibitor inhibits dipeptidyl peptidase VIII with a K 1 of 100 ⁇ M or greater, such as 200 ⁇ M or greater, 500 ⁇ M or greater or even 1 mM or greater.
  • the inhibitor is orally active.
  • the inhibitor has a therapeutic index in humans of at least 2, at least 5, at least 10, or at least 100; e.g., such as a therapeutic index for regulating glucose metabolism.
  • a pharmaceutical composition comprising a pharmaceutically acceptable carrier; and a compound of the present invention or a pharmaceutically acceptable salt thereof.
  • Another aspect of the invention provides for use of one or more of the compounds of the present invention in the manufacture of a medicament for inhibiting a post-pro line protease enzyme in vivo.
  • the subject inhibitors may be used to manufacture medicaments for increasing plasma concentrations of one or more peptide hormones processed by post-proline protease enzymes (e.g., DP-IV and the like).
  • Exemplary medicaments are useful in increasing plasma concentrations of such hormones as glucagons-like peptide, NPY, PPY, secretin, GLP-I, GLP-2, and GIP.
  • the subject inhibitors may be used to manufacture medicaments for regulating glucose metabolism, such as for use in treating patients suffering from Type II diabetes, insulin resistance, glucose intolerance, hyperglycemia, hypoglycemia, hyperinsulinemia, obesity, hyperlipidemia, or hyperlipoproteinemia.
  • Yet another aspect of the invention provides a packaged pharmaceutical, comprising a preparation of a compound of the present invention; optionally a pharmaceutically acceptable carrier; and written and/or pictorial instructions describing the use of the preparation for inhibiting a post-proline protease enzyme in vivo, such as for regulating glucose metabolism.
  • the packaged pharmaceutical can also include, e.g., as a co-formulation with the protease inhibitor or simply co-packaged with the protease inhibitor, insulin and/or an insulinotropic agent.
  • the packaged pharmaceutical can also include, e.g., as a co-formulation with the protease inhibitor or simply co-packaged with the protease inhibitor, an Ml receptor antagonist, a prolactin inhibitor, agents acting on the ATP-dependent potassium channel of ⁇ -cells, metformin, and/or glucosidase inhibitors.
  • the present invention also relates to improved methods for the long-term reduction and abatement of at least one of the foregoing disorders based on a therapeutic regimen administered over the short-term.
  • the present invention further provides a method for regulating and altering on a long-term basis the glucose and lipogenic responses of vertebrate animals, including humans.
  • the compounds of the invention may produce long-lasting beneficial changes in one or more of the following: the sensitivity of the cellular response of a species to insulin (reduction of insulin resistance), blood insulin levels, hyperinsulinemia, blood glucose levels, the amount of body fat stores, and blood lipoprotein levels, thus providing effective treatments for diabetes, obesity and/or atherosclerosis.
  • the present invention relates to inhibitors of post-pro line protease enzymes, such as inhibitors of dipeptidyl peptidase IV, as well as pharmaceutical compositions thereof, and methods for using such inhibitors.
  • Advantageous features for compounds of the present invention may include: better therapeutic indices, owing in part to reduced toxicity and/or improved specificity for the targeted protease; better oral availability; increased shelf-life; and/or increased duration of action (such as single oral dosage formulations which are effective for more than 4 hours, and even more preferably for more than 8, 12, or 16 hours).
  • the compounds of the present invention may be used as part of treatments for a variety of disorders/conditions, such as those which are mediated by DPIV.
  • the subject inhibitors may be used to up-regulate GIP and GLP-I activities, e.g., by increasing the half- life of those hormones, as part of a treatment for regulating glucose levels and/or metabolism, e.g., to reduce insulin resistance, treat hyperglycemia, hyperinsulinemia, obesity, hyperlipidemia, hyperlipoproteinemia (such as chylomicrons, VLDL, and LDL), and to regulate body fat and more generally lipid stores, and, more generally, for the improvement of metabolism disorders, especially those associated with diabetes, obesity and/or atherosclerosis.
  • Compounds which inhibit DPIV are, correlatively, able to improve glucose tolerance, though not necessarily through mechanisms involving DPIV inhibition per se.
  • the subject method utilizes an agent with a K 1 for DPIV inhibition of 50.0 nM or less, 10.0 nM or less, or 1.0, 0.1, or even 0.01 nM or less.
  • K 1 for DPIV inhibition of 50.0 nM or less, 10.0 nM or less, or 1.0, 0.1, or even 0.01 nM or less.
  • inhibitors with K 1 values in the picomolar and even femtomolar range are contemplated.
  • active agents are described herein, for convenience, as "DPIV inhibitors", it will be understood that such nomenclature is not intending to limit the subject invention to a particular mechanism of action.
  • the inhibitor(s) is selected, and the amount of inhibitor formulated, to provide a dosage which inhibits serum post-pro line protease enzymes (e.g., DPIV) levels by at least 50 % for at least 4 hours after a single dose, for at least 8 hours, or for at least 12 or
  • serum post-pro line protease enzymes e.g., DPIV
  • the method involves administration of a DPIV inhibitor, preferably at a predetermined time(s) during a 24-hour period, in an amount effective to improve one or more aberrant indices associated with glucose metabolism disorders (e.g., glucose intolerance, insulin resistance, hyperglycemia, hyperinsulinemia, and Type I and II diabetes).
  • a DPIV inhibitor preferably at a predetermined time(s) during a 24-hour period, in an amount effective to improve one or more aberrant indices associated with glucose metabolism disorders (e.g., glucose intolerance, insulin resistance, hyperglycemia, hyperinsulinemia, and Type I and II diabetes).
  • the method involves administration of a DPIV inhibitor in an amount effective to improve aberrant indices associated with obesity.
  • Fat cells release the hormone leptin, which travels in the bloodstream to the brain and, through leptin receptors there, stimulates production of GLP-I.
  • GLP-I in turn, produces the sensation of being full.
  • the leading theory is that the fat cells of most obese people probably produce enough leptin, but the leptin may not be able to engage properly the leptin receptors in the brain, and so does not stimulate production of GLP-I. Accordingly, a great deal of research has been focused on utilizing preparations of GLP-I as an appetite suppressant.
  • the subject method provides a means for increasing the half- life of both endogenous and ectopically added GLP-I in the treatment of disorders associated with obesity.
  • the present invention provides methods and compositions for altering the pharmacokinetics of a variety of different polypeptide hormones by inhibiting the proteolysis by DPIV or some other proteolytic activity of one or more peptide hormones. Post-secretory metabolism is an important element in the overall homeostasis of regulatory peptides, and the other enzymes involved in these processes may be suitable targets for pharmacological intervention by the subject compounds and methods.
  • the subject method may be used to increase the half-life of other proglucagon-derived peptides, such as glicentin (corresponding to PG 1-69), oxyntomodulin (PG 33-69), glicentin-related pancreatic polypeptide (GRPP, PG 1-30), intervening peptide-2 (IP-2, PG 111-122 amide), and glucagon-like peptide-2 (GLP-2, PG 126-158).
  • GLP-2 for example, has been identified as a factor responsible for inducing proliferation of intestinal epithelium. See, e.g., Drucker et al. Proc. Natl. Acad. Sci. USA 1996, 93, 7911.
  • the DPP IV inhibitors can also be used as part of a regimen for treating injury, inflammation or resection of intestinal tissue, e.g., where enhanced growth and repair of the intestinal mucosal epithelial is desired, such as in the treatment of Crohn's disease or Inflammatory Bowel Disease (IBD).
  • IBD Inflammatory Bowel Disease
  • GHRF growth hormone- releasing factor
  • VIP vasoactive intestinal peptide
  • PHI peptide histidine isoleucine
  • PACAP pituitary adenylate cyclase activating peptide
  • GIP gastric inhibitory peptide
  • GHRF is secreted by the hypothalamus, and stimulates the release of growth hormone (GH) from the anterior pituitary.
  • the subject method can also be used in veterinary practice, for example, to develop higher yield milk production and higher yield, leaner livestock.
  • the DPP IV inhibitors of the invention may be used to alter the plasma half-life of secretin, VIP, PHI, PACAP, GIP and/or helodermin.
  • the inhibitors may also be used to alter the pharmacokinetics of Peptide YY and neuropeptide Y, both members of the pancreatic polypeptide family, because DPP IV has been implicated in the processing of those peptides in a manner which alters receptor selectivity.
  • the DPP IV inhibitors may be used to stimulate hematopoiesis.
  • the DPP IV inhibitors may be used to inhibit growth or vascularization of transformed cells/tissues, e.g., to inhibit cell proliferation such as that associated with tumor growth and metastasis, and for inhibiting angiogenesis in an abnormal proliferative cell mass.
  • the subject DPP IV inhibitors may be used as an immunosuppressant.
  • the DPP IV inhibitors according to the present invention may 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 may be used to treat disorders having a more peripheral nature, including multiple sclerosis and diabetic neuropathy.
  • Another aspect of the present invention provides a method for stimulating hematopoietic cells in culture or in vivo.
  • the subject DPP IV pro- inhibitors include an address moiety that is a substrate for a protease that is expressed in bone marrow.
  • the DPP IV inhibitors of the invention may be used to restore or prevent a deficiency in hematopoietic cell number in a subject. Such deficiencies can arise, for example, from genetic abnormalities, disease, stress, chemotherapy, and radiation treatment.
  • the present invention provides a method of reducing the rate of degradation of tumor suppressors.
  • compounds of the present invention inhibit the growth of cancer cells.
  • the compounds of the present invention may be formulated in topical form for treatment of skin disorders. Such inhibitors are contemplated as possessing important practical application in treating cell proliferative diseases, such as cancer, restenosis, and psoriasis.
  • the subject inhibitors may be used to inhibit growth or vascularization of transformed cells/tissues, e.g., to inhibit cell proliferation, such as that associated with tumor growth and metastasis, and for inhibiting angiogenesis in an abnormal proliferative cell mass.
  • the subject inhibitors may be used as an immunosuppressant.
  • Another aspect of the present invention relates to pharmaceutical compositions of the subject post-proline protease enzyme inhibitors, particularly DPIV inhibitors, and their uses in treating and/or preventing disorders which can be improved by altering the homeostasis of peptide hormones.
  • the inhibitors have hypoglycemic and antidiabetic activities, and may be used in the treatment of disorders marked by aberrant glucose metabolism (including storage).
  • the compositions of the subject methods are useful as insulinotropic agents, or to potentiate the insulinotropic effects of such molecules as GLP-I.
  • compositions may be useful for the treatment and/or prophylaxis of a variety of disorders, including one or more of: hyperlipidemia, hyperglycemia, obesity, glucose tolerance insufficiency, insulin resistance, and diabetic complications.
  • the inhibitors of the subject method are small molecules, e.g., with molecular weights less than 7500 amu, less than 5000 amu, less than 2000, or less than 1000 amu. In certain embodiments, the inhibitors are orally active. //. Definitions
  • high affinity means strong binding affinity between molecules with a dissociation constant K D of no greater than 1 ⁇ M.
  • K D is less than 100 nM, 10 nM, 1 nM, 100 pM, or even 10 pM or less.
  • the two molecules can be covalently linked (K D is essentially 0).
  • boro-Ala refers to the analog of alanine in which the carboxylate group (COOH) is replaced with a boronyl group (B(OH) 2 ).
  • boro-Pro refers to the analog of pro line in which the carboxylate group (COOH) is replaced with a boronyl group (B(OH) 2 ).
  • boro-Xaa where Xaa is an amino acid residue, refers to the analog of an amino acid in which the carboxylate group (COOH) is replaced with a boronyl group (B(OH) 2 ).
  • a "patient” or “subject” to be treated by the subject method can mean either a human or non-human subject.
  • ED50 means the dose of a drug that, in 50% of patients, will provide a clinically relevant improvement or change in a physiological measurement, such as glucose responsiveness, increase in hematocrit, decrease in tumor volume, etc.
  • IC 50 means the dose of a drug that inhibits a biological activity by 50%, e.g., the amount of inhibitor required to inhibit at least 50% of DPIV (or other PPCE) activity in vivo.
  • inhibitor is meant to describe a compound that blocks or reduces an activity of an enzyme (for example, inhibition of proteolytic cleavage of standard fluorogenic peptide substrates such as suc-LLVY-AMC, Box-LLR-AMC and Z- LLE-AMC, inhibition of various catalytic activities of the 2OS proteasome).
  • An inhibitor can act with competitive, uncompetitive, or noncompetitive inhibition.
  • An inhibitor can bind reversibly or irreversibly, and therefore the term includes compounds that are suicide substrates of an enzyme.
  • An inhibitor can modify one or more sites on or near the active site of the enzyme, or it can cause a conformational change elsewhere on the enzyme.
  • a compound is said to have an "insulinotropic activity" if it is able to stimulate, or cause the stimulation of, the synthesis or expression of the hormone insulin.
  • interact as used herein is meant to include all interactions (e.g., biochemical, chemical, or biophysical interactions) between molecules, such as protein- protein, protein-nucleic acid, nucleic acid-nucleic acid, protein-small molecule, nucleic acid-small molecule, or small molecule-small molecule interactions.
  • LD 50 means the dose of a drug that is lethal in 50% of test subjects.
  • prophylactic or therapeutic treatment is art-recognized and includes administration to the host of one or more of the subject compositions. If it is administered prior to clinical manifestation of the unwanted condition (e.g., disease or other unwanted state of the host animal) then the treatment is prophylactic, (i.e., it protects the host against developing the unwanted condition), whereas if it is administered after manifestation of the unwanted condition, the treatment is therapeutic, (i.e., it is intended to diminish, ameliorate, or stabilize the existing unwanted condition or side effects thereof).
  • the unwanted condition e.g., disease or other unwanted state of the host animal
  • preventing is art-recognized, and when used in relation to a condition, such as a local recurrence (e.g., pain), a disease such as cancer, a syndrome complex such as heart failure or any other medical condition, is well understood in the art, and includes administration of a composition which reduces the frequency of, or delays the onset of, symptoms of a medical condition in a subject relative to a subject which does not receive the composition.
  • a condition such as a local recurrence (e.g., pain)
  • a disease such as cancer
  • a syndrome complex such as heart failure or any other medical condition
  • prevention of cancer includes, for example, reducing the number of detectable cancerous growths in a population of patients receiving a prophylactic treatment relative to an untreated control population, and/or delaying the appearance of detectable cancerous growths in a treated population versus an untreated control population, e.g., by a statistically and/or clinically significant amount.
  • Prevention of an infection includes, for example, reducing the number of diagnoses of the infection in a treated population versus an untreated control population, and/or delaying the onset of symptoms of the infection in a treated population versus an untreated control population.
  • Prevention of pain includes, for example, reducing the magnitude of, or alternatively delaying, pain sensations experienced by subjects in a treated population versus an untreated control population.
  • therapeutic index refers to the therapeutic index of a drug defined as LD 50 /ED 50 .
  • a “therapeutically effective amount” of a compound, e.g., such as a DPIV inhibitor of the present invention, with respect to the subject method of treatment refers to an amount of the compound(s) in a preparation which, when administered as part of a desired dosage regimen (to a mammal, preferably a human) alleviates a symptom, ameliorates a condition, or slows the onset of disease conditions according to clinically acceptable standards for the disorder or condition to be treated or the cosmetic purpose, e.g., at a reasonable benefit/risk ratio applicable to any medical treatment.
  • a “single oral dosage formulation” is a dosage which provides an amount of drug to produce a serum concentration at least as great as the EC50 for that drug, but less than the LD50.
  • a single oral dosage formulation provides an amount of drug necessary to produce a serum concentration at least as great as the IC50 for that drug, but less than the LD 50 .
  • a single oral dosage formulation is preferably an amount of drug which produces a serum concentration at least 10 percent less than the
  • LD 50 and even more preferably at least 50 percent, 75 percent, or even 90 percent less than the drug's the LD 50 .
  • inhibitor is meant to describe a compound that blocks or reduces an activity of an enzyme (for example, inhibition of proteolytic cleavage of standard fluorogenic peptide substrates such as suc-LLVY-AMC, Box-LLR-AMC and Z- LLE-AMC, inhibition of various catalytic activities of the 2OS proteasome).
  • An inhibitor can act with competitive, uncompetitive, or noncompetitive inhibition.
  • An inhibitor can bind reversibly or irreversibly, and therefore the term includes compounds that are suicide substrates of an enzyme.
  • An inhibitor can modify one or more sites on or near the active site of the enzyme, or it can cause a conformational change elsewhere on the enzyme.
  • C x _ y alkyl refers to substituted or unsubstituted saturated hydrocarbon groups, including straight-chain alkyl and branched-chain alkyl groups that contain from x to y carbons in the chain, including haloalkyl groups, such as trifluoromethyl and 2,2,2- tirfluoroethyl, etc.
  • Co alkyl indicates a hydrogen where the group is in a terminal position, a bond if internal.
  • C 2 - y alkenyl and “C 2 _ y alkynyl” refer to substituted or unsubstituted unsaturated aliphatic groups analogous in length and possible substitution to the alkyls described above, but that contain at least one double or triple bond respectively.
  • "lower alkyl”, as used herein, means an alkyl group, as defined above, but having from one to ten carbons, more preferably from one to six carbon atoms in its backbone structure such as methyl, ethyl, n- propyl, isopropyl, n-butyl, isobutyl, sec-butyl, and tert-butyl.
  • “lower alkenyl” and “lower alkynyl” have similar chain lengths.
  • a substituent designated herein as alkyl is a lower alkyl.
  • alkoxyl refers to an alkyl group, as defined below, having an oxygen moiety attached thereto.
  • Representative alkoxyl groups include methoxy, ethoxy, propoxy, tert-butoxy, and the like.
  • An "ether” is two hydrocarbons covalently linked by an oxygen. Accordingly, the substituent of an alkyl that renders that alkyl an ether is or resembles an alkoxyl, such as can be represented by one of -O-alkyl, -O- alkenyl, -O-alkynyl, -O-(CH 2 ) m -Ri, where m and Ri are described below.
  • amine and “amino” are art-recognized and refer to both unsubstituted and substituted amines, e.g., a moiety that can be represented by the general formulae:
  • R 35 Rs, and R 6 each independently represent a hydrogen, an alkyl, an alkenyl, -(CH 2 ) m -Ri, or R 3 and R 5 taken together with the N atom to which they are attached complete a heterocycle having from 4 to 8 atoms in the ring structure;
  • Ri represents an alkenyl, aryl, cycloalkyl, a cycloalkenyl, a heterocyclyl, or a polycyclyl; and m is zero or an integer in the range of 1 to 8.
  • only one of R 3 or R5 can be a carbonyl, e.g., R 3 , R 5 , and the nitrogen together do not form an imide.
  • R 3 and R 5 each independently represent a hydrogen, an alkyl, an alkenyl, or -(CH 2 ) m -Ri.
  • alkylamine as used herein means an amine group, as defined above, having a substituted or unsubstituted alkyl attached thereto, i.e., at least one of R 3 and R 5 is an alkyl group.
  • an amino group or an alkylamine is basic, meaning it has a conjugate acid with a pKa >7.00, i.e., the protonated forms of these functional groups have pKas relative to water above about 7.00..
  • carbonyl is art-recognized and includes such moieties as can be represented by the general formula:
  • X is a bond or represents an oxygen or a sulfur
  • R 7 represents a hydrogen, an alkyl, an alkenyl, -(CH 2 ) m -Ri or a pharmaceutically acceptable salt
  • R 8 represents a hydrogen, an alkyl, an alkenyl or -(CH 2 ) m -Ri, where m and Ri are as defined above.
  • heterocyclyl or “heterocyclic group” refer to substituted or unsubstituted non-aromatic 3- to 10-membered ring structures, more preferably 3- to 7- membered rings, whose ring structures include one to four heteroatoms.
  • heterocyclyl or “heterocyclic group” also include polycyclic ring systems having two or more cyclic rings in which two or more carbons are common to two adjoining rings wherein at least one of the rings is heterocyclic, e.g., the other cyclic rings can be cycloalkyls, cycloalkenyls, cycloalkynyls, aryls, heteroaryls, and/or heterocyclyls.
  • Heterocyclyl groups include, for example, piperidine, piperazine, pyrrolidine, morpholine, lactones, lactams, and the like.
  • substituted refers to moieties having substituents replacing a hydrogen on one or more carbons of the backbone. It will be understood that “substitution” or
  • substituted with includes the implicit proviso that such substitution is in accordance with permitted valence of the substituted atom and the substituent, and that the substitution results in a stable compound, e.g., which does not spontaneously undergo transformation such as by rearrangement, cyclization, elimination, etc.
  • substituted is contemplated to include all permissible substituents of organic compounds.
  • the permissible substituents include acyclic and cyclic, branched and unbranched, carbocyclic and heterocyclic, aromatic and non-aromatic substituents of organic compounds.
  • the permissible substituents can be one or more and the same or different for appropriate organic compounds.
  • the heteroatoms such as nitrogen may have hydrogen substituents and/or any permissible substituents of organic compounds described herein which satisfy the valences of the heteroatoms.
  • Substituents can include, for example, a halogen, a hydroxyl, a carbonyl (such as a carboxyl, an alkoxycarbonyl, a formyl, or an acyl), a thiocarbonyl (such as a thioester, a thioacetate, or a thioformate), an alkoxyl, a phosphoryl, a phosphate, a phosphonate, a phosphinate, an amino, an amido, an amidine, an imine, a cyano, a nitro, an azido, a sulfhydryl, an alkylthio, a sulfate, a sulfonate, a sulfamoyl, a s
  • abbreviations used herein for designating the amino acids and the protective groups are based on recommendations of the IUPAC-IUB Commission on Biochemical Nomenclature (see Biochemistry 1972, 11, 1726-1732).
  • Residues represent "residues" of methionine, isoleucine, leucine, alanine, and glycine, respectively.
  • Residue means a moiety derived from the corresponding ⁇ -amino acid by eliminating the OH portion of the carboxyl group and the H portion of the ⁇ -amino group.
  • amino acid side chain is that part of an amino acid exclusive of the -CH(NH 2 )COOH portion, as defined by K. D. Kopple, Peptides and Amino Acids; Benjamin: New York, 1966; pp.
  • side chains of the common amino acids are -CH 2 CH 2 SCH 3 (the side chain of methionine), -CH 2 (CH 3 )-CH 2 CH 3 (the side chain of isoleucine), -CH 2 CH(CH 3 ) 2 (the side chain of leucine) or H-(the side chain of glycine).
  • amino acids used in the present invention are those naturally occurring amino acids found in native proteins, or the naturally occurring anabolic or catabolic products of such amino acids which contain amino and carboxyl groups.
  • Particularly suitable amino acid side chains include side chains selected from those of the following amino acids: glycine, alanine, valine, cysteine, leucine, isoleucine, serine, threonine, methionine, glutamic acid, aspartic acid, glutamine, asparagine, lysine, arginine, proline, histidine, phenylalanine, tyrosine, and tryptophan, and those amino acids and amino acid analogs which have been identified as constituents of peptidylglycan bacterial cell walls.
  • amino acid residue further includes analogs, derivatives and congeners of any specific amino acid referred to herein, as well as C-terminal or N-terminal protected amino acid derivatives (e.g., modified with an N-terminal or C-terminal protecting group).
  • the present invention contemplates the use of amino acid analogs wherein a side chain is lengthened or shortened while still providing a carboxyl, amino or other reactive precursor functional group for cyclization, as well as amino acid analogs having variant side chains with appropriate functional groups).
  • the subject compound can include an amino acid analog such as, for example, cyanoalanine, canavanine, djenkolic acid, norleucine, 3-phosphoserine, homoserine, dihydroxy-phenylalanine, 5- hydroxytryptophan, 1-methylhistidine, 3-methylhistidine, diaminopimelic acid, ornithine, or diaminobutyric acid.
  • amino acid analog such as, for example, cyanoalanine, canavanine, djenkolic acid, norleucine, 3-phosphoserine, homoserine, dihydroxy-phenylalanine, 5- hydroxytryptophan, 1-methylhistidine, 3-methylhistidine, diaminopimelic acid, ornithine, or diaminobutyric acid.
  • amino acid analog such as, for example, cyanoalanine, canavanine, djenkolic acid, norleucine, 3-phosphoserine, homoserine, dihydroxy-phenyla
  • (D) and (L) stereoisomers of such amino acids when the structure of the amino acid admits of stereoisomeric forms.
  • the configuration of the amino acids and amino acid residues herein are designated by the appropriate symbols (D), (L) or (DL), furthermore when the configuration is not designated, the amino acid or residue can have the configuration (D), (L), or (DL).
  • the structure of some of the compounds of this invention includes asymmetric carbon atoms. It is to be understood accordingly that the isomers arising from such asymmetry are included within the scope of this invention. Such isomers can be obtained in substantially pure form by classical separation techniques and by sterically controlled synthesis.
  • a named amino acid shall be construed to include both the (D) and (L) stereoisomers.
  • peptide analogue refers to a peptide comprising one or more non-naturally occurring amino acid.
  • non-naturally occurring amino acids include, but are not limited to, D-amino acids (i.e., an amino acid of an opposite chirality to the naturally occurring form), JV- ⁇ -methyl amino acids, C- ⁇ -methyl amino acids, ⁇ -methyl amino acids, ⁇ -alanine ( ⁇ -Ala), norvaline (Nva), norleucine (NIe), 4-aminobutyric acid ( ⁇ -Abu), 2-aminoisobutyric acid (Aib), 6-aminohexanoic acid ( ⁇ -Ahx), ornithine (orn), hydroxyproline (Hyp), sarcosine, citrulline, cysteic acid, cyclohexylalanine, ⁇ -amino isobutyric acid, t-butylglycine, t-butylalanine, 3-amino
  • D- or L-2- indole(alkyl)alanines and D- or L-alkylalanines wherein alkyl is substituted or unsubstituted methyl, ethyl, propyl, hexyl, butyl, pentyl, isopropyl, iso-butyl, or iso-pentyl, and phosphono- or sulfated (e.g., -SO3H) non-carboxylate amino acids.
  • non-naturally occurring amino acids include 3-(2-chlorophenyl)- alanine, 3-chloro-phenylalanine, 4-chloro-phenylalanine, 2-fluoro-phenylalanine, 3-fluoro- phenylalanine, 4-fluoro-phenylalanine, 2-bromo-phenylalanine, 3-bromo-phenylalanine, 4- bromo-phenylalanine, homophenylalanine, 2-methyl-phenylalanine, 3-methyl- phenylalanine, 4-methyl-phenylalanine, 2,4-dimethyl-phenylalanine, 2-nitro-phenylalanine, 3-nitro-phenylalanine, 4-nitro-phenylalanine, 2,4-dinitro-phenylalanine, 1,2,3,4- Tetrahydroisoquinoline-3-carboxylic acid, l,2,3,4-tetrahydronorharman-3-carboxylic acid, 1-naphthyl
  • peptide derivative refers to a peptide comprising additional chemical or biochemical moieties not normally a part of a naturally occurring peptide.
  • Peptide derivatives include peptides in which the amino-terminus and/or the carboxy-terminus and/or one or more amino acid side chain has been derivatised with a suitable chemical substituent group, as well as cyclic peptides, dual peptides, multimers of the peptides, peptides fused to other proteins or carriers, glycosylated peptides, phosphorylated peptides, peptides conjugated to lipophilic moieties (for example, caproyl, lauryl, stearoyl moieties) and peptides conjugated to an antibody or other biological ligand.
  • Examples of chemical substituent groups that may be used to derivatise a peptide include, but are not limited to, alkyl, cycloalkyl and aryl groups; acyl groups, including alkanoyl and aroyl groups; esters; amides; halogens; hydroxyls; carbamyls, and the like.
  • the substituent group may also be a blocking group such as Fmoc (fluorenylmethyl-O-CO-), carbobenzoxy (benzyl-O-CO-), monomethoxysuccinyl, naphthyl-NH-CO-, acetylamino- caproyl and adamantyl-NH-CO-.
  • C-terminal hydroxymethyl derivatives include C-terminal hydroxymethyl derivatives, O-modif ⁇ ed derivatives (for example, C-terminal hydroxymethyl benzyl ether) and //-terminally modified derivatives including substituted amides such as alkylamides and hydrazides.
  • the substituent group may be a "protecting group" as detailed herein.
  • protecting group means substituents which protect the reactive functional group from undesirable chemical reactions.
  • protecting groups include esters of carboxylic acids and boronic acids, ethers of alcohols, and acetals and ketals of aldehydes and ketones.
  • N-terminal protecting group or amino-protecting group refers to various amino- protecting groups which can be employed to protect the N-terminus of an amino acid or peptide against undesirable reactions during synthetic procedures.
  • acyl protecting groups such as, to illustrate, formyl, dansyl, acetyl, benzoyl, trifluoroacetyl, succinyl, and methoxysuccinyl
  • aromatic urethane protecting groups as, for example, benzyloxycarbonyl (Cbz)
  • aliphatic urethane protecting groups such as t- butoxycarbonyl (Boc) or 9-Fluorenylmethoxycarbonyl (Fmoc).
  • amino-protecting group or 'W-terminal protecting group refers to those groups intended to protect the ⁇ -iV-terminal of an amino acid or peptide or to otherwise protect the amino group of an amino acid or peptide against undesirable reactions during synthetic procedures.
  • JV-protecting groups are disclosed in Greene, Protective Groups In Organic Synthesis, (John Wiley & Sons, New York (1981)), which is hereby incorporated by reference. Additionally, protecting groups may be used as pro- drugs which are readily cleaved in vivo, for example, by enzymatic hydrolysis, to release the biologically active parent.
  • ⁇ -iV-Protecting groups comprise lower alkanoyl groups such as formyl, acetyl ("Ac"), propionyl, pivaloyl, t-butylacetyl and the like; other acyl groups include 2-chloroacetyl, 2-bromoacetyl, trifluoroacetyl, trichloroacetyl, phthalyl, o- nitrophenoxyacetyl, -chlorobutyryl, benzoyl, 4-chlorobenzoyl, 4-bromobenzoyl, 4- nitrobenzoyl and the like; sulfonyl groups such as benzenesulfonyl, p-toluenesulfonyl and the like; carbamate forming groups such as benzyloxycarbonyl, p- chlorobenzyloxycarbonyl, p-methoxybenzyloxycarbonyl, p-nitrobenzyloxycarbony
  • Still other examples include theyl, succinyl, methoxysuccinyl, subery, adipyl, azelayl, dansyl, benzyloxycarbonyl, methoxyazelaly, methoxyadipyl, methoxysuberyl, and 2,4- dinitrophenyl.
  • carboxy protecting group or "C-terminal protecting group” refers to a carboxylic acid protecting ester or amide group employed to block or protect the carboxylic acid functionality while the reactions involving other functional sites of the compound are performed.
  • Carboxy protecting groups are disclosed in Greene, Protective Groups in Organic Synthesis pp. 152-186 (1981), which is hereby incorporated by reference. Additionally, a carboxy protecting group may be used as a pro-drug whereby the carboxy protecting group can be readily cleaved in vivo, for example by enzymatic hydrolysis, to release the biologically active parent.
  • carboxy protecting groups are well known to those skilled in the art, having been extensively used in the protection of carboxyl groups in the penicillin and cephalosporin fields as described in U.S. Pat. Nos. 3,840,556 and 3,719,667, the disclosures of which are hereby incorporated herein by reference.
  • carboxy protecting groups are Ci -Cs loweralkyl (e.g., methyl, ethyl or t- butyl and the like); arylalkyl such as phenethyl or benzyl and substituted derivatives thereof such as alkoxybenzyl or nitrobenzyl groups and the like; arylalkenyl such as phenylethenyl and the like; aryl and substituted derivatives thereofsuch as 5-indanyl and the like; dialkylaminoalkyl such as dimethylaminoethyl and the like); alkanoyloxyalkyl groups such as acetoxymethyl, butyryloxymethyl, valeryloxymethyl, isobutyryloxymethyl, isovaleryloxymethyl, l-(propionyloxy)-l -ethyl, l-(pivaloyloxyl)-l -ethyl, 1 -methyl- 1- (propionyloxy)-l -ethyl,
  • amide carboxy protecting groups are aminocarbonyl and loweralkylaminocarbonyl groups.
  • aspartic acid may be protected at the ⁇ -C-terminal by an acid labile group (e.g., t-butyl) and protected at the ⁇ -C-terminal by a hydrogenation labile group (e.g., benzyl) then deprotected selectively during synthesis.
  • an acid labile group e.g., t-butyl
  • a hydrogenation labile group e.g., benzyl
  • the protected carboxy group may also be a loweralkyl, cycloalkyl or arylalkyl ester, for example, methyl ester, ethyl ester, propyl ester, isopropyl ester, butyl ester, sec-butyl ester, isobutyl ester, amyl ester, isoamyl ester, octyl ester, cyclohexyl ester, phenylethyl ester and the like or an alkanoyloxyalkyl, cycloalkanoyloxyalkyl, aroyloxyalkyl or an arylalkylcarbonyloxyalkyl ester.
  • certain compounds of the present invention may exist in particular geometric or stereoisomeric forms.
  • the present invention contemplates all such compounds, including cis- and trans-isomers, R- and S-enantiomers, diastereomers, (D)- isomers, (L)-isomers, the racemic mixtures thereof, and other mixtures thereof, as falling within the scope of the invention.
  • Additional asymmetric carbon atoms may be present in a substituent such as an alkyl group. All such isomers, as well as mixtures thereof, are intended to be included in this invention.
  • a particular enantiomer of a compound of the present invention may be prepared by asymmetric synthesis or by derivation with a chiral auxiliary, where the resulting diastereomeric mixture is separated and the auxiliary group cleaved to provide the pure desired enantiomer.
  • the molecule contains a basic functional group, such as amino, or an acidic functional group, such as carboxyl, diastereomeric salts are formed with an appropriate optically-active acid or base, followed by resolution of the diastereomers thus formed by fractional crystallization or chromatographic means well known in the art, and subsequent recovery of the pure enantiomer.
  • a compound is said to have an "insulinotropic activity" if it is able to stimulate, or cause the stimulation of, the synthesis or expression of the hormone insulin.
  • compositions of dipeptidylpeptidase inhibitors particularly inhibitor(s) and their uses in treating and/or preventing disorders which can be improved by altering the homeostasis of peptide hormones.
  • the inhibitors have hypoglycemic and antidiabetic activities, and may be used in the treatment of disorders marked by aberrant glucose metabolism (including storage).
  • the compositions of the subject methods are useful as insulinotropic agents, or to potentiate the insulinotropic effects of such molecules as GLP-I .
  • the present method can be useful for the treatment and/or prophylaxis of a variety of disorders, including one or more of: hyperlipemia, hyperglycemia, obesity, glucose tolerance insufficiency, insulin resistance, and diabetic complications.
  • the method involves administration of an inhibitor(s), preferably at a predetermined interval(s) during a 24-hour period, in an amount effective to improve one or more aberrant indices associated with glucose metabolism disorders (e.g., glucose intolerance, insulin resistance, hyperglycemia, hyperinsulinemia, and Type II diabetes).
  • the effective amount of the inhibitor may be about 0.01, 0.1, 1, 10, 30, 50, 70, 100, 150, 200, 500, or 1000 mg/kg of the subject. ///.
  • Exemplary Embodiments (i). Compounds preferably at a predetermined interval(s) during a 24-hour period, in an amount effective to improve one or more aberrant indices associated with glucose metabolism disorders (e.g., glucose intolerance, insulin resistance, hyperglycemia, hyperinsulinemia, and Type II diabetes).
  • the effective amount of the inhibitor may be about 0.01, 0.1, 1, 10, 30, 50, 70, 100, 150, 200, 500, or 1000 mg/kg of the subject. ///.
  • variables are defined specifically for each individual formula.
  • a definition of a variable for one formula should not be used to vary a definition provided for another formula, although a variable that has not been defined for one formula may be interpreted by analogy with a definition elsewhere for a similar formula.
  • a protease inhibitor of the invention is represented by
  • R 4 represents H, alkyl, alkenyl, alkynyl, -C(X 1 )(X 2 )(X 3 ), -(CH 2 )Hi-R 43 , -(CH 2 )Ii-OH, -(CH 2 )n-O-alkyl, -(CH 2 )n-O-alkenyl, -(CH 2 )n-O-alkynyl, -(CH 2 )H-O-(CH 2 )Hi-R 43 , -(CH 2 )n-SH, -(CH 2 )n-S-alkyl, -(CH 2 )n-S-alkenyl, -(CH 2 )n-S-alkynyl, -(CH 2 )H-S-(CH 2 )Hi-R 43 , -C(O)C(O)NH 2 , and -C(O)C(O)OR 44 ;
  • R 43 represents independently for each occurrence aryl, aralkyl, or non-aromatic hetereocyclyl
  • R 44 represents independently for each occurrence H, alkyl, alkenyl, aryl, aralkyl, or non-aromatic heterocyclyl;
  • Yi and Y 2 independently represent OH, or a group capable of being hydro lyzed to a hydroxyl group; or taken together with the boron to which they are bonded form a 5- membered to 8-membered ring comprising said boron and two oxygen atoms bonded to said boron;
  • R 40 is O or S;
  • R41 is selected from the group consisting of N 3 , SH, NH 2 , NO 2 , and -OK ⁇ ;
  • R 42 is selected from the group consisting of hydrogen, lower alkyl, amine, and -OR 44 , or a pharmaceutically acceptable salt; or R 41 and R 42 taken together with the phosphorous atom to which they are attached complete a heterocyclic ring having from 5 to 8 atoms in the ring structure;
  • X 1 is halogen;
  • X 2 and X 3 is hydrogen or halogen; m is zero or an integer in the range of 1 to 8; and n is an integer in the range of 1 to 8.
  • Ri is selected from a C-terminally linked amino acid residue or amino acid analog, a C-terminally linked peptide or peptide analog, an amino-protecting group, OO O u
  • R 2 is absent or represents one or more substituents attached at positions 3, 4, or 5 of the dehydroproline ring, each of which is independently selected from the group consisting of hydrogen, halogen, hydroxyl, lower alkyl, lower alkenyl, lower alkynyl, lower alkoxy, lower hydroxyalkyl, lower alkoxyalkyl, carbonyl, carboxyl, ester, formate, ketone, thiocarbonyl, thioester, thioacetate, thioformate, amino, acylamino, amido, nitro, sulfate, sulfonate, sulfonamido, -(CH 2 X n -R 51 , -(CH 2 X n -OH, -(CH 2 ) m -O-lower alkyl, -(CH 2 X n -O- lower alkenyl, -(CH 2 ) n -O-(CH 2 ) m
  • R 3 is selected from the group consisting of hydrogen, halogen, lower alkyl, lower alkenyl, lower alkynyl, carbonyl, thiocarbonyl, amino, acylamino, amido, nitro, sulfate, sulfonate, a sulfonamido, -(CH 2 X n -R 51 , -(CH 2 X n -OH, -(CH 2 ) m -O-lower alkyl, -(CH 2 X n -O- lower alkenyl, -(CH 2 ) n -O-(CH 2 ) m -R 51 , -(CH 2 X n -SH, -(CH 2 ) m -S-lower alkyl, -(CH 2 X n -S- lower alkenyl, or -(CH 2 ) n -S-(CH 2 ) m -R 51 , azi
  • R 5 is selected from the group consisting of hydrogen, halogen, alkyl, alkenyl, alkynyl, aryl, -(CH 2 ) m -R 51 , -(CH 2 ) m -0H, -(CH 2 ) m -O-alkyl, -(CH 2 ) m -O-alkenyl, -(CH 2 ) m -O-alkynyl, -(CH 2 ) m -O-(CH 2 ) m -R 51 , -(CH 2 ) m -SH, -(CH 2 ) m -S-alkyl, -(CH 2 ) m -S- alkenyl, -(CH 2 ) m -S-alkynyl, -(CH 2 ) m -S-(CH 2 ) m -R 51 ,
  • R51 represents for each occurrence aryl, aralkyl, or non-aromatic heterocyclyl;
  • R 52 and R 53 taken together with the N atom to which they are attached complete a heterocyclic ring having from 4 to 8 atoms in the ring structure; m is zero or an integer in the range of 1 to 8; and n is an integer in the range of 1 to 8.
  • W is a group of formula -B(Yi)(Y 2 ).
  • Ri is a C-terminally linked amino acid residue or amino acid analog or a C-terminally linked peptide or peptide analog.
  • R 3 is hydrogen or a lower alkyl group.
  • the compound is represented by:
  • Another aspect of the invention provides a protease inhibitor having a structure represented by Formula (II):
  • R 2 is absent or represents one or more substituents attached at positions 3, 4, or 5 of the dehydroproline ring, each of which is independently selected from the group consisting of hydrogen, halogen, hydroxyl, lower alkyl, lower alkenyl, lower alkynyl, lower alkoxy, lower hydroxyalkyl, lower alkoxyalkyl, carbonyl, carboxyl, ester, formate, ketone, thiocarbonyl, thioester, thioacetate, thioformate, amino, acylamino, amido, nitro, sulfate, sulfonate, sulfonamido, -(CH 2 ) m -R 43 , -(CH 2 ) m -OH, -(CH 2 ) m -O-lower alkyl, -(CH 2 ) m -O- lower alkenyl, -(CH 2 ) n -O-(CH 2 ) m
  • Rioi is selected from the group consisting of H, alkyl, alkoxy, alkenyl, alkynyl, amino, alkylamino, acylamino, cyano, sulfonylamino, acyloxy, aryl, non-aromatic heterocyclyl, heteroaryl, and a polypeptide chain of 1 to 8 amino acid residues;
  • Rio2 is selected from the group consisting of H, lower alkyl, and aralkyl
  • Rio3 is selected from the group consisting of H, halogen, and alkyl, or R102 and R103 together with the atoms to which they are attached, form a 3- to 6-membered heterocyclic ring;
  • R105 is selected from the group consisting of H, halogen, lower alkyl, and aralkyl;
  • R 4 represents H, alkyl, alkenyl, alkynyl, -C(X 1 )(X 2 )(X 3 ), -(CH 2 )Di-R 43 , -(CH 2 )Ii-OH, -(CH 2 )n-O-alkyl, -(CH 2 )n-O-alkenyl, -(CH 2 )n-O-alkynyl, -(CH 2 )n-O-(CH 2 )m-R43, -(CH 2 )n-SH, -(CH 2 )n-S-alkyl, -(CH 2 )n-S-alkenyl,
  • R43 represents independently for each occurrence aryl, aralkyl, or non-aromatic hetereocyclyl
  • R 44 independently for each occurrence is selected from the group consisting of H, alkyl, alkenyl, aryl, aralkyl, and non-aromatic heterocyclyl;
  • Yi and Y 2 independently represent OH, or a group capable of being hydro lyzed to a hydroxyl group; or taken together with the boron to which they are bonded form a 5- membered to 8-membered ring comprising said boron and two oxygen atoms bonded to said boron; R 40 is O or S;
  • R41 is selected from the group consisting of N 3 , SH, NH 2 , NO 2 and -OR ⁇ ;
  • R 42 is selected from the group consisting of H, lower alkyl, amine, and -OR 44 , or a pharmaceutically acceptable salt; or R 41 and R 42 taken together with the phosphorous atom to which they are attached complete a heterocyclic ring having from 5 to 8 atoms in the ring structure;
  • Xi is halogen;
  • X 2 and X 3 is hydrogen or halogen; and n is an integer in the range of 2 to 6;
  • R 107 is selected from the group consisting of H, aryl, alkyl, aralkyl, non-aromatic heterocyclyl, heteroaryl, heteroaralkyl, and polypeptide chains of 1 to 8 amino acid residues;
  • L is absent or is selected from the group consisting of alkyl, alkenyl, alkynyl, -(CH 2 ) m O(CH 2 ) m -, -(CH 2 ) m NR 2 (CH 2 ) m -, and -(CH 2 ) m S(CH 2 ) m -;
  • Rio ⁇ is a group of formula -B(Yi)(Y 2 ).
  • one or more of L, X and Y is absent.
  • two or more of L, X and Y are absent.
  • each of L, X and Y is absent.
  • R 1 O 1 is generally -H.
  • one or both of Rio 3 and R105 are -H in compound represented by Formula (II). More typically, both R i03 and R 105 are -H.
  • Exemplary compounds represented by Formula (II) are: or pharmaceutically acceptable salts thereof.
  • Ri is a polypeptide chain of 2 to 8 amino acid residues, where proline is the residue that is directly attached to the leftmost residue of Formula (II). In certain such embodiments, Ri is a polypeptide chain of 2 amino acid residues, where proline is the residue that is directly attached to the leftmost nitrogen of Formula (II).
  • the dehydroproline ring in Formulas (I) and (II) can be substituted with one or more groups R 2 .
  • R 2 is selected from hydroxyl, lower alkyl (e.g., methyl), lower alkenyl, lower alkynyl, lower alkoxy, lower hydroxyalkyl (e.g., hydroxymethyl), and lower alkoxyalkyl.
  • the dehydroproline substituent group is selected from lower alkyl, lower hydroxyalkyl and lower alkoxyalkyl.
  • R 2 is located at the 5 -position of the ring, particularly when the substituent is hydroxyl.
  • the subject inhibitors are DPIV inhibitors with a K 1 for
  • the inhibitors of the subject method are small molecules, e.g., with molecular weights less than 7500 amu, less than 5000 amu, less than 2000 amu, and less than 1000 amu.
  • the inhibitors are orally active.
  • compositions of the dipeptidylpeptidase inhibitors disclosed herein and their uses in treating and/or preventing (inhibiting) disorders which can be improved by altering the homeostasis of peptide hormones.
  • the inhibitors have hypoglycemic and antidiabetic activities, and may be used in the treatment of disorders marked by aberrant glucose metabolism (including storage).
  • the compositions of the subject methods are useful as insulinotropic agents, or to potentiate the insulinotropic effects of such molecules as GLP-I .
  • the present method may be useful for the treatment and/or prophylaxis of a variety of disorders, including: hyperlipemia, hyperglycemia, obesity, glucose tolerance insufficiency, insulin resistance, and diabetic complications.
  • the method involves administration of an inhibitor(s), preferably at a predetermined interval(s) during a 24-hour period, in an amount effective to improve one or more aberrant indices associated with glucose metabolism disorders (e.g., glucose intolerance, insulin resistance, hyperglycemia, hyperinsulinemia, and Type II diabetes).
  • the effective amount of the inhibitor may be about 0.01, 0.1, 1, 10, 30, 50, 70, 100, 150, 200, 500, or 1000 mg/kg of the subject.
  • the compounds useful in the subject methods possess, in certain embodiments, the ability to lower blood glucose levels, to relieve obesity, to alleviate impaired glucose tolerance, to inhibit hepatic glucose neogenesis, and to lower blood lipid levels and to inhibit aldose reductase. They are thus useful for the prevention and/or therapy of hyperglycemia, obesity, hyperlipidemia, diabetic complications (including retinopathy, nephropathy, neuropathy, cataracts, coronary artery disease and arteriosclerosis), and furthermore for obesity-related hypertension and osteoporosis.
  • Diabetes mellitus is a disease characterized by hyperglycemia caused by from a decrease in insulin secretion, decreased insulin sensitivity, and/or insulin resistance. The morbidity and mortality of this disease result from vascular, renal, and neurological complications.
  • An oral glucose tolerance test is a clinical test used to diagnose diabetes. In an oral glucose tolerance test, a patient's physiological response to a glucose load or challenge is evaluated. After ingesting the glucose, the patient's physiological response to the glucose challenge is evaluated. Generally, this is accomplished by determining the patient's blood glucose levels (the concentration of glucose in the patient's plasma, serum, or whole blood) at several predetermined points in time.
  • the present invention provides a method for agonizing the action of GLP-I. It has been determined that isoforms of GLP-I (GLP- 1(7-37) and GLP- 1(7-36)), which are derived from preproglucagon in the intestine and the hind brain, have insulinotropic activity; i.e., they modulate glucose metabolism. DPIV cleaves the isoforms to inactive peptides. Thus, in certain embodiments, compound(s) of the present invention can agonize insulinotropic activity by interfering with the degradation of bioactive GLP-I peptides.
  • the subject agents may be used to agonize (e.g., mimic or potentiate) the activity of peptide hormones, e.g., GLP-2, GIP and NPY.
  • the present invention provides a method for agonizing the action of GLP-2. It has been determined that GLP-2 acts as a trophic agent, to promote growth of gastrointestinal tissue. The effect of GLP-2 is marked particularly by increased growth of the small bowel, and is therefore herein referred to as an "intestinotrophic" effect.
  • DPIV is known to cleave GLP-2 into a biologically inactive peptide. Thus, in one embodiment, inhibition of DPIV interferes with the degradation of GLP-2, and thereby increases the plasma half- life of that hormone.
  • the subject method may be used to increase the half- life of other proglucagon-derived peptides, such as glicentin, oxyntomodulin, glicentin-related pancreatic polypeptide (GRPP), and/or intervening peptide -2 (IP-2).
  • glicentin has been demonstrated to cause proliferation of intestinal mucosa and also inhibits a peristalsis of the stomach, and has thus been elucidated as useful as a therapeutic agent for digestive tract diseases.
  • the present invention relates to therapeutic and related uses of compound(s) for promoting the growth and proliferation of gastrointestinal tissue, particularly small bowel tissue.
  • the subject method may be used as part of a regimen for treating injury, inflammation, or resection of intestinal tissue, e.g., where enhanced growth and repair of the intestinal mucosal epithelial is desired.
  • small bowel tissue With respect to small bowel tissue, such growth is measured conveniently as an increase in small bowel mass and length, relative to an untreated control.
  • the effect of compounds on small bowel tissue also manifests as an increase in the height of the crypt plus villus axis.
  • Such activity is referred to herein as an "intestinotrophic" activity.
  • the efficacy of the subject method may also be detectable as an increase in crypt cell proliferation and/or a decrease in small bowel epithelium apoptosis.
  • a compound is considered to have "intestinotrophic effect" if a test animal exhibits increased small bowel weight, increased height of the crypt plus villus axis or increased crypt cell proliferation, or decreased small bowel epithelium apoptosis when treated with the compound (or genetically engineered to express it themselves).
  • a model suitable for determining such gastrointestinal growth is described by US Patent 5,834,428 (incorporated by reference). In general, patients who would benefit from either increased small intestinal mass and consequent increased small bowel mucosal function are candidates for treatment by the subject method.
  • Particular conditions that may be treated include the various forms of sprue, including celiac sprue which results from a toxic reaction to ⁇ -gliadin from wheat, and is marked by a tremendous loss of villae of the bowel; tropical sprue which results from infection and is marked by partial flattening of the villae; hypogammaglobulinemic sprue which is observed in patients with common variable immunodeficiency or hypogammaglobulinemia and is marked by significant decrease in villus height.
  • the therapeutic efficacy of the treatment may be monitored by enteric biopsy to examine the villus morphology, by biochemical assessment of nutrient absorption, by patient weight gain, or by amelioration of the symptoms associated with these conditions.
  • Other conditions that may be treated by the subject method, or for which the subject method may be useful prophylactically, include radiation enteritis, infectious or post-infectious enteritis, regional enteritis (Crohn's disease), small intestinal damage due to toxic or other chemotherapeutic agents, and patients with short bowel syndrome.
  • the present invention provides a therapeutic method for treating digestive tract diseases.
  • the term "digestive tract” as used herein means a organismic tube through which food passes, including stomach and intestine.
  • the term “digestive tract diseases” as used herein means diseases accompanied by a qualitative or quantitative abnormality in the digestive tract mucosa, which include ulceric or inflammatory disease; congenital or acquired digestion and absorption disorder including malabsorption syndrome; disease caused by loss of a mucosal barrier function of the gut; and protein- losing gastroenteropathy.
  • the ulceric disease includes gastric ulcer, duodenal ulcer, small intestinal ulcer, colonic ulcer, and rectal ulcer.
  • the inflammatory disease includes esophagitis, gastritis, duodenitis, enteritis, colitis, Crohn's disease, proctitis, gastrointestinal Behcet, radiation enteritis, radiation colitis, radiation proctitis, enteritis, and medicamentosa.
  • the malabsorption syndrome includes the essential malabsorption syndrome, such as disaccharide-decomposing enzyme deficiency, glucose-galactose malabsorption, fructose malabsorption; secondary malabsorption syndrome, e.g., the disorder caused by a mucosal atrophy in the digestive tract through the intravenous or parenteral nutrition or elemental diet, the disease caused by the resection and shunt of the small intestine such as short gut syndrome, cul-de-sac syndrome; and indigestible malabsorption syndrome, such as the disease caused by resection of the stomach, e.g., dumping syndrome.
  • essential malabsorption syndrome such as disaccharide-decomposing enzyme deficiency, glucose-galactose malabsorption, fructose malabsorption
  • secondary malabsorption syndrome e.g., the disorder caused by a mucosal atrophy in the digestive tract through the intravenous or parenteral nutrition or elemental diet, the disease caused by the resection and shunt of
  • therapeutic agent for digestive tract diseases means the agents for the prevention and treatment of the digestive tract diseases, which include the therapeutic agent for digestive tract ulcer, the therapeutic agent for inflammatory digestive tract disease, the therapeutic agent for mucosal atrophy in the digestive tract, the therapeutic agent for a digestive tract wound, the amelioration agent for the function of the digestive tract including the agent for recovery of the mucosal barrier function, and the amelioration agent for digestive and absorptive function. Ulcers include digestive ulcers and erosions, and acute ulcers, namely acute mucosal lesions.
  • the subject method because of promoting proliferation of intestinal mucosa, may be used in the treatment and prevention of pathologic conditions of insufficiency in digestion and absorption, that is, treatment and prevention of mucosal atrophy, or treatment of hypoplasia of the digestive tract tissues and decrease in these tissues by surgical removal as well as improvement of digestion and absorption. Further, the subject method may be used in the treatment of pathologic mucosal conditions due to inflammatory diseases, such as enteritis, Crohn's disease, and ulceric colitis and also in the treatment of reduction in function of the digestive tract after operation, for example, in damping syndrome as well as in the treatment of duodenal ulcer in conjunction with the inhibition of peristalsis of the stomach and rapid migration of food from the stomach to the jejunum.
  • pathologic mucosal conditions due to inflammatory diseases, such as enteritis, Crohn's disease, and ulceric colitis
  • inflammatory diseases such as enteritis, Crohn's disease, and ulceric colitis
  • glicentin can effectively be used in promoting cure of surgical invasion as well as in improving functions of the digestive tract.
  • the present invention also provides a therapeutic agent for atrophy of the digestive tract mucosa, a therapeutic agent for wounds in the digestive tract and a drug for improving functions of the digestive tract which comprise glicentin as active ingredients.
  • the compound(s) of the subject invention may be used to alter the plasma half-life of secretin, VIP, PHI, PACAP, GIP, and/or helodermin. Additionally, the subject method may be used to alter the pharmacokinetics of Peptide YY and neuropeptide Y, both members of the pancreatic polypeptide family, as DPIV has been implicated in the processing of those peptides in a manner which alters receptor selectivity.
  • Neuropeptide Y is believed to act in the regulation vascular smooth muscle tone, as well as regulation of blood pressure. NPY also decreases cardiac contractility. NPY is also the most powerful appetite stimulant known (Wilding et al., J. Endocrinology 1992, 132, 299-302). The centrally evoked food intake (appetite stimulation) effect is predominantly mediated by NPY Yl receptors and causes increase in body fat stores and obesity (Stanley et al., Physiology and Behavior 1989, 46, 173-177).
  • a method for treatment of anorexia comprises administering to a subject in need thereof a therapeutically effective amount of a compound(s) of the present invention, to stimulate the appetite and increase body fat stores.
  • a method for treatment of hypotension comprises administering to a subject in need thereof a therapeutically effective amount of a compound(s) of the present invention to mediate vasoconstriction and increase blood pressure.
  • DPIV has also been implicated in the metabolism and inactivation of growth hormone-releasing factor (GHRF).
  • GHRF is a member of the family of homologous peptides that includes glucagon, secretin, vasoactive intestinal peptide (VIP), peptide histidine isoleucine (PHI), pituitary adenylate cyclase activating peptide (PACAP), gastric inhibitory peptide (GIP) and helodermin (Kubiak et al. Peptide Res. 1994, 7, 153).
  • GHRF is secreted by the hypothalamus, and stimulates the release of growth hormone (GH) from the anterior pituitary.
  • the subject method may be used to improve clinical therapy for certain growth hormone deficient children, and in clinical therapy of adults to improve nutrition and to alter body composition (muscle vs. fat).
  • the subject method can also be used in veterinary practice, for example, to develop higher yield milk production and higher yield, leaner livestock.
  • the insulinotropic property of a compound may be determined by providing that compound to animal cells, or injecting that compound into animals and monitoring the release of immunoreactive insulin (IRI) into the media or circulatory system of the animal, respectively.
  • IRI immunoreactive insulin
  • the presence of IRI can be detected through the use of a radioimmunoassay which can specifically detect insulin.
  • the db/db mouse is a genetically obese and diabetic strain of mouse.
  • the db/db mouse develops hyperglycemia and hyperinsulinemia concomitant with its development of obesity and thus serves as a model of obese type 2 diabetes (NIDDM).
  • the db/db mice can be purchased from, for example, The Jackson Laboratories (Bar Harbor, Me.).
  • sub-orbital sinus blood samples are taken before and at some time (e.g., 60 min) after dosing of each animal.
  • Blood glucose measurements can be made by any of several conventional techniques, such as using a glucose meter. The blood glucose levels of the control and compound(s) dosed animals are compared.
  • the metabolic fate of exogenous GLP-I can also be followed in both nondiabetic and type II diabetic subjects, and the effect of a candidate compound(s) determined.
  • a combination of high-pressure liquid chromatography (HPLC), specific radioimmunoassays (RIAs), and an enzyme-linked immunosorbent assay (ELISA) can be used, whereby intact biologically active GLP-I and its metabolites can be detected. See, for example, Deacon et al. Diabetes, 1995, 44, 1126-1131.
  • the intact peptide can be measured using an NH 2 -terminally directed RIA or ELISA, while the difference in concentration between these assays and a COOH-terminal- specif ⁇ c RIA allowed determination of NH2-terminally truncated metabolites.
  • subcutaneous GLP-I is rapidly degraded in a time-dependent manner, forming a metabolite which co-elutes on HPLC with GLP- 1(9-36) amide and has the same immunoreactive profile.
  • Another aspect of the invention provides a conjoint therapy wherein one or more other therapeutic agents are administered with the compound.
  • Such conjoint treatment may be achieved by way of the simultaneous, sequential, or separate dosing of the individual components of the treatment.
  • a compound(s) of the present invention is conjointly administered with insulin or other insulinotropic agents, such as GLP-I, peptide hormones, such as GLP-2, GIP, or NPY, or a gene therapy vector which causes the ectopic expression of said agents and peptide hormones.
  • insulin or other insulinotropic agents such as GLP-I, peptide hormones, such as GLP-2, GIP, or NPY, or a gene therapy vector which causes the ectopic expression of said agents and peptide hormones.
  • said agents or peptide hormones may be variants or derivatives of a naturally occurring or synthetic peptide hormone, wherein one or more amino acids have been added, deleted, or substituted.
  • the compounds of the present invention can be conjointly administered with an Ml receptor antagonist.
  • Cholinergic agents are potent modulators of insulin release that act via muscarinic receptors. Moreover, the use of such agents can have the added benefit of decreasing cholesterol levels, while increasing HDL levels.
  • Suitable muscarinic receptor antagonists include substances that directly or indirectly block activation of muscarinic cholinergic receptors. Preferably, such substances are selective (or are used in amounts that promote such selectivity) for the Ml receptor.
  • Non-limiting examples include quaternary amines (such as methantheline, ipratropium, and propantheline), tertiary amines (e.g., dicyclomine and scopolamine), and tricyclic amines (e.g., telenzepine).
  • Suitable muscarinic receptor antagonists include benztropine (commercially available as COGENTIN from Merck), hexahydro-sila-difenidol hydrochloride (HHSID hydrochloride disclosed in Lambrecht et al. Trends in Pharmacol. Sci.
  • the compound(s) of the present invention may also act synergistically with prolactin inhibitors such as d2 dopamine agonists (e.g., bromocriptine).
  • prolactin inhibitors such as d2 dopamine agonists (e.g., bromocriptine).
  • the subject method can include the conjoint administration of such prolactin inhibitors as prolactin-inhibiting ergo alkaloids and prolactin-inhibiting dopamine agonists.
  • suitable compounds include 2-bromo-alpha-ergocriptine, 6-methyl-8-beta- carbobenzyloxyaminoethyl-10-alpha-ergoline, 8-acylaminoergo lines, 6-methyl-8-alpha-(N- acyl)amino-9-ergoline, 6-methyl-8-alpha-(N-phenylacetyl)amino-9-ergoline, ergocornine, 9,10-dihydroergocornine, D-2-halo-6-alkyl-8-substituted ergo lines, D-2-bromo-6-methyl-8- cyanomethylergoline, carbidopa, benserazide, and other dopadecarboxylase inhibitors, L- dopa, dopamine, and non toxic salts thereof.
  • the compound(s) of the present invention used according to the invention can also be used conjointly with agents acting on the ATP-dependent potassium channel of the ⁇ - cells, such as glibenclamide, glipizide, gliclazide, and AG-EE 623 ZW.
  • agents acting on the ATP-dependent potassium channel of the ⁇ - cells such as glibenclamide, glipizide, gliclazide, and AG-EE 623 ZW.
  • the compound(s) may also advantageously be applied in combination with other oral agents, such as metformin and related compounds or glucosidase inhibitors, such as acarbose.
  • a compound of the present invention While it is possible for a compound of the present invention to be administered alone, in certain cases it is preferable to administer the compound as a pharmaceutical formulation (composition).
  • Protease inhibitors according to the invention may be formulated for administration in any convenient way for use in human or veterinary medicine.
  • the compound included in the pharmaceutical preparation may be active itself, or may be a prodrug, e.g., capable of being converted to an active compound in a physiological setting.
  • Compounds prepared as described herein can be administered in various forms, depending on the disorder to be treated and the age, condition, and body weight of the patient, as is well known in the art.
  • the compounds may be formulated as tablets, capsules, granules, powders, or syrups; or for parenteral administration, they may be formulated as injections (intravenous, intramuscular, or subcutaneous), drop infusion preparations, or suppositories.
  • injections intravenous, intramuscular, or subcutaneous
  • drop infusion preparations or suppositories.
  • suppositories For application by the ophthalmic mucous membrane route, they may be formulated as eye drops or eye ointments.
  • formulations can be prepared by conventional means, and, if desired, the active ingredient may be mixed with any conventional additive, such as an excipient, a binder, a disintegrating agent, a lubricant, a corrigent, a solubilizing agent, a suspension aid, an emulsifying agent, or a coating agent.
  • an excipient such as an excipient, a binder, a disintegrating agent, a lubricant, a corrigent, a solubilizing agent, a suspension aid, an emulsifying agent, or a coating agent.
  • a daily dosage of from 0.01 to 2000 mg of the compound is recommended for an adult human patient, and this may be administered in a single dose or in divided doses.
  • the precise time of administration and/or amount of the compound that will yield the most effective results in terms of efficacy of treatment in a given patient will depend upon the activity, pharmacokinetics, and bioavailability of a particular compound, physiological condition of the patient (including age, sex, disease type and stage, general physical condition, responsiveness to a given dosage, and type of medication), route of administration, etc.
  • physiological condition of the patient including age, sex, disease type and stage, general physical condition, responsiveness to a given dosage, and type of medication
  • route of administration etc.
  • the above guidelines may be used as the basis for fine-tuning the treatment, e.g., determining the optimum time and/or amount of administration, which will require no more than routine experimentation consisting of monitoring the subject and adjusting the dosage and/or timing.
  • phrases "pharmaceutically acceptable” is employed herein to refer to those ligands, materials, compositions, and/or dosage forms which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of human beings and animals without excessive toxicity, irritation, allergic response, or other problem or complication, commensurate with a reasonable benefit/risk ratio.
  • pharmaceutically acceptable carrier means a pharmaceutically acceptable material, composition or vehicle, such as a liquid or solid filler, diluent, excipient, solvent or encapsulating material. Each carrier must be “acceptable” in the sense of being compatible with the other ingredients of the formulation and not injurious to the patient.
  • materials which can serve as pharmaceutically acceptable carriers include: (1) sugars, such as lactose, glucose, and sucrose; (2) starches, such as corn starch and potato starch; (3) cellulose, and its derivatives, such as sodium carboxymethyl cellulose, ethyl cellulose, and cellulose acetate; (4) powdered tragacanth; (5) malt; (6) gelatin; (7) talc; (8) excipients, such as cocoa butter and suppository waxes; (9) oils, such as peanut oil, cottonseed oil, safflower oil, sesame oil, olive oil, corn oil, and soybean oil; (10) glycols, such as propylene glycol; (11) polyols, such as glycerin, sorbitol, mannitol, and polyethylene glycol; (12) esters, such as ethyl oleate and ethyl laurate; (13) agar; (14) buffering agents, such as magnesium hydroxide and aluminum
  • compositions of the present invention are non-pyrogenic, i.e., do not induce significant temperature elevations when administered to a patient.
  • pharmaceutically acceptable salts refers to the relatively non-toxic, inorganic and organic acid addition salts of the compound(s). These salts can be prepared in situ during the final isolation and purification of the compound(s), or by separately reacting a purified compound(s) in its free base form with a suitable organic or inorganic acid, and isolating the salt thus formed.
  • Representative salts include the hydrobromide, hydrochloride, sulfate, bisulfate, phosphate, nitrate, acetate, valerate, oleate, palmitate, stearate, laurate, benzoate, lactate, phosphate, tosylate, citrate, maleate, fumarate, succinate, tartrate, naphthylate, mesylate, glucoheptonate, lactobionate, and laurylsulphonate salts, and the like. (See, e.g., Berge et al. J. Pharm. Sci. 1977, 66, 1-19)
  • the compounds useful in the methods of the present invention may contain one or more acidic functional groups and, thus, are capable of forming pharmaceutically acceptable salts with pharmaceutically acceptable bases.
  • pharmaceutically acceptable salts refers to the relatively non-toxic inorganic and organic base addition salts of an compound(s). These salts can likewise be prepared in situ during the final isolation and purification of the compound(s), or by separately reacting the purified compound(s) in its free acid form with a suitable base, such as the hydroxide, carbonate, or bicarbonate of a pharmaceutically acceptable metal cation, with ammonia, or with a pharmaceutically acceptable organic primary, secondary, or tertiary amine.
  • Representative alkali or alkaline earth salts include the lithium, sodium, potassium, calcium, magnesium, and aluminum salts, and the like.
  • Representative organic amines useful for the formation of base addition salts include ethylamine, diethylamine, ethylenediamine, ethanolamine, diethanolamine, piperazine, and the like (see, e.g., Berge et al., supra).
  • Wetting agents, emulsif ⁇ ers, and lubricants, such as sodium lauryl sulfate and magnesium stearate, as well as coloring agents, release agents, coating agents, sweetening, flavoring, and perfuming agents, preservatives and antioxidants can also be present in the compositions.
  • antioxidants examples include: (1) water soluble antioxidants, such as ascorbic acid, cysteine hydrochloride, sodium bisulfate, sodium metabisulfite, sodium sulfite, and the like; (2) oil-soluble antioxidants, such as ascorbyl palmitate, butylated hydroxyanisole (BHA), butylated hydroxytoluene (BHT), lecithin, propyl gallate, alpha-tocopherol, and the like; and (3) metal chelating agents, such as citric acid, ethylenediamine tetraacetic acid (EDTA), sorbitol, tartaric acid, phosphoric acid, and the like.
  • water soluble antioxidants such as ascorbic acid, cysteine hydrochloride, sodium bisulfate, sodium metabisulfite, sodium sulfite, and the like
  • oil-soluble antioxidants such as ascorbyl palmitate, butylated hydroxyanisole (BHA), butylated hydroxytoluene (BHT
  • Formulations useful in the methods of the present invention include those suitable for oral, nasal, topical (including buccal and sublingual), rectal, vaginal, aerosol, and/or parenteral administration.
  • the formulations may conveniently be presented in unit dosage form and may be prepared by any methods well known in the art of pharmacy.
  • the amount of active ingredient which can be combined with a carrier material to produce a single dosage form will vary depending upon the host being treated and the particular mode of administration.
  • the amount of active ingredient which can be combined with a carrier material to produce a single dosage form will generally be that amount of the compound which produces a therapeutic effect. Generally, out of 100%, this amount will range from about 1% to about 99% of active ingredient, preferably from about 5% to about 70%, most preferably from about 10% to about 30%.
  • Methods of preparing these formulations or compositions include the step of bringing into association a compound(s) with the carrier and, optionally, one or more accessory ingredients.
  • the formulations are prepared by uniformly and intimately bringing into association a ligand with liquid carriers, or finely divided solid carriers, or both, and then, if necessary, shaping the product.
  • Formulations suitable for oral administration may be in the form of capsules, cachets, pills, tablets, lozenges (using a flavored basis, usually sucrose and acacia or tragacanth), powders, granules, or as a solution or a suspension in an aqueous or nonaqueous liquid, or as an oil-in-water or water-in-oil liquid emulsion, or as an elixir or syrup, or as pastilles (using an inert base, such as gelatin and glycerin, or sucrose and acacia) and/or as mouthwashes, and the like, each containing a predetermined amount of a compound(s) as an active ingredient.
  • a compound may also be administered as a bolus, electuary or paste.
  • the active ingredient is mixed with one or more pharmaceutically acceptable carriers, such as sodium citrate or dicalcium phosphate, and/or any of the following: (1) fillers or extenders, such as starches, lactose, sucrose, glucose, mannitol, and/or silicic acid; (2) binders, such as, for example, carboxymethylcellulose, alginates, gelatin, polyvinyl pyrrolidone, sucrose, and/or acacia; (3) humectants, such as glycerol; (4) disintegrating agents, such as agar-agar, calcium carbonate, potato or tapioca starch, alginic acid, certain silicates, and sodium carbonate; (5) solution retarding agents, such as paraffin; (6) absorption accelerators, such as quaternary ammonium compounds; (7) wetting agents, such as, for example, acet
  • compositions may also comprise buffering agents.
  • Solid compositions of a similar type may also be employed as fillers in soft and hard- filled gelatin capsules using such excipients as lactose or milk sugars, as well as high molecular weight polyethylene glycols, and the like.
  • a tablet may be made by compression or molding, optionally with one or more accessory ingredients.
  • Compressed tablets may be prepared using binder (for example, gelatin or hydroxypropylmethyl cellulose), lubricant, inert diluent, preservative, disintegrant (for example, sodium starch glycolate or cross-linked sodium carboxymethyl cellulose), surface-active or dispersing agent.
  • Molded tablets may be made by molding in a suitable machine a mixture of the powdered peptide or peptidomimetic moistened with an inert liquid diluent.
  • Tablets, and other solid dosage forms may optionally be scored or prepared with coatings and shells, such as enteric coatings and other coatings well known in the pharmaceutical-formulating art. They may also be formulated so as to provide slow or controlled release of the active ingredient therein using, for example, hydroxypropylmethyl cellulose in varying proportions to provide the desired release profile, other polymer matrices, liposomes, and/or microspheres. They may be sterilized by, for example, filtration through a bacteria-retaining filter, or by incorporating sterilizing agents in the form of sterile solid compositions which can be dissolved in sterile water, or some other sterile injectable medium immediately before use.
  • compositions may also optionally contain opacifying agents and may be of a composition that they release the active ingredient(s) only, or preferentially, in a certain portion of the gastrointestinal tract, optionally, in a delayed manner.
  • opacifying agents include polymeric substances and waxes.
  • the active ingredient can also be in micro-encapsulated form, if appropriate, with one or more of the above-described excipients.
  • Liquid dosage forms for oral administration include pharmaceutically acceptable emulsions, microemulsions, solutions, suspensions, syrups, and elixirs.
  • the liquid dosage forms may contain inert diluents commonly used in the art, such as, for example, water or other solvents, solubilizing agents, and emulsif ⁇ ers such as ethyl alcohol, isopropyl alcohol, ethyl carbonate, ethyl acetate, benzyl alcohol, benzyl benzoate, propylene glycol, 1,3-butylene glycol, oils (in particular, cottonseed, groundnut, corn, germ, olive, castor, and sesame oils), glycerol, tetrahydrofuryl alcohol, polyethylene glycols, and fatty acid esters of sorbitan, and mixtures thereof.
  • inert diluents commonly used in the art, such as, for example, water or other solvents, solubilizing agents, and
  • the oral compositions can also include adjuvants such as wetting agents, emulsifying and suspending agents, sweetening, flavoring, coloring, perfuming, and preservative agents.
  • adjuvants such as wetting agents, emulsifying and suspending agents, sweetening, flavoring, coloring, perfuming, and preservative agents.
  • Suspensions in addition to the active compound(s) may contain suspending agents as, for example, ethoxylated isostearyl alcohols, polyoxyethylene sorbitol and sorbitan esters, microcrystalline cellulose, aluminum metahydroxide, bentonite, agar-agar and tragacanth, and mixtures thereof.
  • suspending agents as, for example, ethoxylated isostearyl alcohols, polyoxyethylene sorbitol and sorbitan esters, microcrystalline cellulose, aluminum metahydroxide, bentonite, agar-agar and tragacanth, and mixtures thereof.
  • Formulations for rectal or vaginal administration may be presented as a suppository, which may be prepared by mixing one or more compound(s) with one or more suitable nonirritating excipients or carriers comprising, for example, cocoa butter, polyethylene glycol, a suppository wax or a salicylate, which is solid at room temperature, but liquid at body temperature and, therefore, will melt in the rectum or vaginal cavity and release the active agent.
  • suitable nonirritating excipients or carriers comprising, for example, cocoa butter, polyethylene glycol, a suppository wax or a salicylate, which is solid at room temperature, but liquid at body temperature and, therefore, will melt in the rectum or vaginal cavity and release the active agent.
  • Formulations which are suitable for vaginal administration also include pessaries, tampons, creams, gels, pastes, foams, or spray formulations containing such carriers as are known in the art to be appropriate.
  • Dosage forms for the topical or transdermal administration of a compound(s) include powders, sprays, ointments, pastes, creams, lotions, gels, solutions, patches, and inhalants.
  • the active component may be mixed under sterile conditions with a pharmaceutically acceptable carrier, and with any preservatives, buffers, or propellants which may be required.
  • the ointments, pastes, creams, and gels may contain, in addition to compound(s), excipients, such as animal and vegetable fats, oils, waxes, paraffins, starch, tragacanth, cellulose derivatives, polyethylene glycols, silicones, bentonites, silicic acid, talc, and zinc oxide, or mixtures thereof.
  • excipients such as animal and vegetable fats, oils, waxes, paraffins, starch, tragacanth, cellulose derivatives, polyethylene glycols, silicones, bentonites, silicic acid, talc, and zinc oxide, or mixtures thereof.
  • Powders and sprays can contain, in addition to a compound(s), excipients such as lactose, talc, silicic acid, aluminum hydroxide, calcium silicates, and polyamide powder, or mixtures of these substances.
  • Sprays can additionally contain customary propellants, such as chlorofluorohydrocarbons and volatile unsubstituted hydrocarbons, such as butane and propane.
  • the compound(s) can be alternatively administered by aerosol. This is accomplished by preparing an aqueous aerosol, liposomal preparation, or solid particles containing the compound. A non-aqueous (e.g., fluorocarbon propellant) suspension could be used.
  • a non-aqueous (e.g., fluorocarbon propellant) suspension could be used.
  • an aqueous aerosol is made by formulating an aqueous solution or suspension of the agent together with conventional pharmaceutically acceptable carriers and stabilizers.
  • the carriers and stabilizers vary with the requirements of the particular compound, but typically include nonionic surfactants (Tweens, Pluronics, or polyethylene glycol), innocuous proteins like serum albumin, sorbitan esters, oleic acid, lecithin, amino acids such as glycine, buffers, salts, sugars, or sugar alcohols.
  • Aerosols generally are prepared from isotonic solutions.
  • Medicaments which may be administered in inhalant or aerosol formulations according to the invention include protease inhibitor prodrugs useful in inhalation therapy which may be presented in a form which is soluble or substantially soluble in the selected propellant system.
  • the particle size of the particulate medicament should be such as to permit inhalation of substantially all of the medicament into the lungs upon administration of the aerosol formulation and will thus desirably be less than 20 microns, preferably in the range 1 to 10 microns, e.g., 1 to 5 microns.
  • the particle size of the medicament may be reduced by conventional means, for example by milling or micronisation.
  • Administration of medicament may be indicated for the treatment of mild, moderate or severe acute or chronic symptoms or for prophylactic treatment. It will be appreciated that the precise dose administered will depend on the age and condition of the patient, the particular particulate medicament used and the frequency of administration and will ultimately be at the discretion of the attendant physician. When combinations of medicaments are employed the dose of each component of the combination will in general be that employed for each component when used alone. Typically, administration may be one or more times, for example from 1 to 8 times per day, giving for example 1, 2, 3 or 4 puffs each time. Preferably, administration may be one time per day.
  • the drug is suitably inhaled from a nebulizer, from a pressurized metered dose inhaler, or as a dry powder from a dry powder inhaler (e.g., sold as TURBUHALER ® ) or from a dry powder inhaler utilizing gelatin, plastic or other capsules, cartridges or blister packs.
  • a dry powder inhaler e.g., sold as TURBUHALER ®
  • a diluent or carrier generally non-toxic and chemically inert to the medicament; e.g., lactose, dextran, mannitol, glucose or any additives that will give the medicament a desired taste, can be added to the powdered medicament.
  • the micronized mixture may be suspended or dissolved in a liquid propellant mixture which is kept in a container that is sealed with a metering valve and fitted into a plastic actuator.
  • the propellants used may be halocarbons of different chemical formulae. The most frequently used halocarbon propellants are trichlorofluoromethane, dichlorodifluoromethane, dichlorotetrafluoroethane, tetrafluoroethane, and 1,1-difluoroethane.
  • Low concentrations of a surfactant such as sorbitan trioleate, lecithin, disodium dioctylsulphosuccinate, or oleic acid may also be used to improve the physical stability.
  • Transdermal patches have the added advantage of providing controlled delivery of a compound(s) to the body.
  • dosage forms can be made by dissolving or dispersing the agent in the proper medium.
  • Absorption enhancers can also be used to increase the flux of the compound(s) across the skin. The rate of such flux can be controlled by either providing a rate controlling membrane or dispersing the peptidomimetic in a polymer matrix or gel.
  • compositions of this invention suitable for parenteral administration comprise one or more compounds(s) in combination with one or more pharmaceutically acceptable sterile isotonic aqueous or non-aqueous solutions, dispersions, suspensions or emulsions, or sterile powders which may be reconstituted into sterile injectable solutions or dispersions just prior to use, which may contain antioxidants, buffers, bacteriostats, solutes which render the formulation isotonic with the blood of the intended recipient or suspending or thickening agents.
  • aqueous and non-aqueous carriers examples include water, ethanol, polyols (such as glycerol, propylene glycol, polyethylene glycol, and the like), and suitable mixtures thereof, vegetable oils, such as olive oil, and injectable organic esters, such as ethyl oleate.
  • polyols such as glycerol, propylene glycol, polyethylene glycol, and the like
  • vegetable oils such as olive oil
  • injectable organic esters such as ethyl oleate.
  • Proper fluidity can be maintained, for example, by the use of coating materials, such as lecithin, by the maintenance of the required particle size in the case of dispersions, and by the use of surfactants.
  • compositions may also contain adjuvants such as preservatives, wetting agents, emulsifying agents, and dispersing agents. Prevention of the action of microorganisms may be ensured by the inclusion of various antibacterial and antifungal agents, for example, paraben, chlorobutanol, phenol sorbic acid, and the like. It may also be desirable to include isotonic agents, such as sugars, sodium chloride, and the like into the compositions. In addition, prolonged absorption of the injectable pharmaceutical form may be brought about by the inclusion of agents which delay absorption such as aluminum monostearate and gelatin.
  • adjuvants such as preservatives, wetting agents, emulsifying agents, and dispersing agents.
  • Prevention of the action of microorganisms may be ensured by the inclusion of various antibacterial and antifungal agents, for example, paraben, chlorobutanol, phenol sorbic acid, and the like. It may also be desirable to include isotonic agents, such as sugars, sodium
  • Injectable depot forms are made by forming microencapsule matrices of compound(s) in biodegradable polymers such as polylactide-polyglycolide.
  • the rate of drug release can be controlled.
  • biodegradable polymers include poly(orthoesters) and poly(anhydrides).
  • Depot injectable formulations are also prepared by entrapping the drug in liposomes or microemulsions which are compatible with body tissue.
  • the compounds(s) of the present invention are administered as pharmaceuticals to humans and animals, they can be given per se or as a pharmaceutical composition containing, for example, 0.1 to 99.5% (more preferably, 0.5 to 90%) of active ingredient in combination with a pharmaceutically acceptable carrier.
  • agents may be given orally, parenterally, topically, or rectally . They are of course given by forms suitable for each administration route. For example, they are administered in tablets or capsule form, by injection, inhalation, eye lotion, ointment, suppository, infusion; topically by lotion or ointment; and rectally by suppositories.
  • parenteral administration and “administered parenterally” as used herein means modes of administration other than enteral and topical administration, usually by injection, and includes, without limitation, intravenous, intramuscular, intraarterial, intrathecal, intracapsular, intraorbital, intracardiac, intradermal, intraperitoneal, transtracheal, subcutaneous, subcuticular, intraarticular, subcapsular, subarachnoid, intraspinal and intrasternal injection, and infusion.
  • systemic administration means the administration of a ligand, drug, or other material other than directly into the central nervous system, such that it enters the patient's system and thus, is subject to metabolism and other like processes, for example, subcutaneous administration.
  • These compounds(s) may be administered to humans and other animals for therapy by any suitable route of administration, including orally, nasally, as by, for example, a spray, rectally, intravaginally, parenterally, intracisternally, and topically, as by powders, ointments or drops, including buccally and sublingually.
  • the addition of the active compound of the invention to animal feed is preferably accomplished by preparing an appropriate feed premix containing the active compound in an effective amount and incorporating the premix into the complete ration.
  • an intermediate concentrate or feed supplement containing the active ingredient can be blended into the feed.
  • feed premixes and complete rations can be prepared and administered are described in reference books (such as Applied Animal Nutrition; San Francisco: Freedman, 1969; ox Livestock Feeds and Feeding; Corvallis: O & B Books, 1977).
  • the compound(s), which may be used in a suitable hydrated form, and/or the pharmaceutical compositions of the present invention, are formulated into pharmaceutically acceptable dosage forms by conventional methods known to those of skill in the art.
  • Actual dosage levels of the active ingredients in the pharmaceutical compositions of this invention may be varied so as to obtain an amount of the active ingredient which is effective to achieve the desired therapeutic response for a particular patient, composition, and mode of administration, without being toxic to the patient.
  • Combinatorial Libraries The compounds of the present invention, particularly libraries of variants having various representative classes of substituents, are amenable to combinatorial chemistry and other parallel synthesis schemes (see, for example, PCT WO 94/08051).
  • a combinatorial library for the purposes of the present invention is a mixture of chemically related compounds which may be screened together for a desired property.
  • the preparation of many related compounds in a single reaction greatly reduces and simplifies the number of screening processes which need to be carried out. Screening for the appropriate physical properties can be done by conventional methods.
  • the substrate aryl groups used in the combinatorial reactions can be diverse in terms of the core aryl moiety, e.g., a variegation in terms of the ring structure, and/or can be varied with respect to the other substituents.
  • a library of candidate protease inhibitor diversomers can be synthesized utilizing a scheme adapted to the techniques described in the Still et al. PCT publication WO 94/08051 , e.g., being linked to a polymer bead by a hydrolyzable or photo lyzable group, optionally located at one of the positions of the candidate agonists or a substituent of a synthetic intermediate.
  • the library is synthesized on a set of beads, each bead including a set of tags identifying the particular diversomer on that bead.
  • the bead library can then be "plated" with proteases for which an inhibitor is sought.
  • the diversomers can be released from the bead, e.g., by hydrolysis.
  • the structures of the compounds useful in the present invention lend themselves readily to efficient synthesis.
  • the nature of the structures of the subject compounds, as generally set forth above, allows the rapid combinatorial assembly of such compounds.
  • an activated aryl group such as an aryl triflate or bromide
  • an activated aryl group attached to a bead or other solid support
  • an activated aryl group such as an aryl triflate or bromide
  • an aryl stannane or an aryl boronic acid can be linked to another aryl group by performing a Stille or Suzuki coupling with an aryl stannane or an aryl boronic acid.
  • an aryl group is functionalized with an aldehyde, an amine substituent can be added through a reductive amination.
  • the second aryl group could be functionalized with a leaving group, such as a triflate, tosylate, or halide, capable of being displaced by an amine.
  • the second aryl group may be functionalized with an amine group capable of undergoing reductive amination with an amine, e.g., CyKNH 2 .
  • Other possible coupling techniques include transition metal-mediated amine arylation reactions.
  • the resultant secondary amine can then be further functionalized by an acylation, alkylation, or arylation to generate a tertiary amine or amide which can then be cleaved from the resin or support.
  • the inhibitor solution is prepared by dissolving 3 - 5 mg of inhibitor in pH 2 solution (0.01 N HCl), such that the concentration of the solution is equal to 1 mg/10 ⁇ L. A 10 ⁇ L sample of this solution is then added to 990 ⁇ L of pH 8 buffer (0.1 M HEPES, 0.14
  • the enzyme solution is prepared by diluting 20 ⁇ L of DPIV (concentration 2.5 ⁇ M) into 40 mL of pH 8 buffer.
  • the substrate solution is prepared by dissolving 2.0 mg of L-alanyl-L-proline-/? ⁇ ra- nitroanilide into 20 mL of pH 8 buffer.
  • the solution in #D5 is then mixed well before transferring 10 ⁇ L of this solution from #D5 to #E5.
  • the solution in #E5 is then mixed well before transferring 10 ⁇ L of this solution from #E5 to #F5.
  • the solution in #F5 is then mixed well before transferring 10 ⁇ L of this solution from #F5 to #G5.
  • the solution in #G5 is then mixed well before transferring 10 ⁇ L of this solution from #G5 to #H5.
  • a 30 ⁇ L aliquot is then transferred from #H5 to #H3 for row H, and the contents are mixed well.
  • the analogous procedure is repeated for rows G, F, E, D, C, B, and A sequentially.
  • the plate is then shaken on a plate shaker for 5 minutes before allowing the plate to incubate at room temperature for an additional 5 minutes.
  • Example 1 The assay described in Example 1 is used to determine the IC 50 values for several compounds of the invention.
  • the assay is conducted for DPIV and DP8 or
  • IC50 values were measured at the same pH throughout the assay.
  • Certain compounds of the invention inhibit DPIV at least 10 times, preferably at least 100 times, more strongly than they inhibit DP8 and/or DP9, i.e., have an IC 50 at least 10 (or 100) times lower against DPIV than against DP8 and/or DP9.

Abstract

One aspect of the present invention relates to inhibitors of post-pro line protease enzymes, such as inhibitors of dipeptidyl peptidase IV, and pharmaceutical compositions thereof. Another aspect of the present invention relates to methods of using the inhibitors to inhibit a dipeptidyl peptidase IV. Yet another aspect of the present invention relates to methods for using the inhibitors in the manufacture of the medicament for inhibiting a post- pro line enzyme in vivo.

Description

3,4-Dehydro-Proline-Containing Inhibitors of Dipeptidylpeptidase IV
Related Applications This application claims the benefit of priority to United States Provisional Patent
Application serial number 60/908,267, filed March 27, 2007.
Background of the Invention
Proteases are enzymes that cleave proteins at specific peptide bonds. Proteases can be classified into four generic classes: serine; thiol or cysteinyl; acid or aspartyl; and metalloproteases (Cuypers et al, J. Biol. Chem. 1982, 257, 7086. Proteases are essential to a variety of biological activities, such as digestion, formation and dissolution of blood clots, reproduction, and immune reaction to foreign cells and organisms. However, aberrant proteolysis is associated with a number of diseases in humans and other mammals. Accordingly, it is often beneficial to disrupt the function of one or more proteolytic enzymes in the course of treating a patient.
The binding site for a peptide substrate consists of a series of "specificity subsites" across the surface of the enzyme. The term "specificity subsite" refers to a pocket or other site on the enzyme capable of interacting with a portion of a substrate for the enzyme. In discussing the interactions of peptides with proteases, e.g., serine and cysteine proteases, the present application utilizes the nomenclature of Schechter and Berger (Biochem. Biophys. Res. Commun. 1967, 27, 157-162). The individual amino acid residues of a substrate or inhibitor are designated Pl, P2, etc. and the corresponding subsites of the enzyme are designated Sl, S2, etc., starting with the carboxy terminal residue produced in the cleavage reaction. The scissile bond of the substrate is the amide bond between Pl-Pl ' of the substrate. Thus, for a peptide Xaal-Xaa2-Xaa3-Xaa4, which is cleaved between the Xaa3 and Xaa4 residues, the Xaa3 residue is referred to as the Pl residue and binds to the Sl subsite of the enzyme, Xaa2 is referred to as the P2 residue and binds to the S2 subsite, and so forth.
Dipeptidyl peptidase IV (DPIV or DPPIV) is a serine protease that cleaves N- terminal dipeptides from a peptide chain containing, preferably, a pro line residue in the penultimate position, i.e., in the Pl position. DPIV belongs to a group of cell-membrane- associated peptidases and, like the majority of cell-surface peptidases, is a type II integral membrane protein, being anchored to the plasma membrane by its signal sequence. DPIV is found in a variety of differentiated mammalian epithelia, endothelia and hematopoetic cells and tissues, including those of lymphoid origin where it is found specifically on the surface of CD4+ T cells. DPIV has been identified as leukocyte differentiation marker CD26.
Summary of the Invention
One aspect of the present invention relates to inhibitors of post-pro line protease enzymes, such as inhibitors of dipeptidyl peptidase IV (DPIV), as well as pharmaceutical compositions thereof, and methods for using such inhibitors. In certain embodiments, the inhibitors of the present invention include a dehydroproline residue having a functional group that covalently reacts with the active site of DPIV.
One aspect of the invention is a protease inhibitor represented by Formula (I):
Figure imgf000003_0001
or a pharmaceutically acceptable salt thereof; wherein:
W is selected from the group consisting of -CN, -CH=NR4,
Figure imgf000003_0002
; wherein
R4 represents H, alkyl, alkenyl, alkynyl, -C(X1)(X2)(X3), -(CH2)Di-R43, -(CH2)Ii-OH, -(CH2)n-O-alkyl, -(CH2)n-O-alkenyl, -(CH2)n-O-alkynyl, -(CH2)U-O-(CH2)In-R43, -(CH2)n-SH, -(CH2)n-S-alkyl, -(CH2)n-S-alkenyl,
-(CH2)n-S-alkynyl, -(CH2)U-S-(CH2)In-R43, -C(O)C(O)NH2, and -C(O)C(O)OR44;
R43 represents independently for each occurrence aryl, aralkyl, or non-aromatic hetereocyclyl; R-44 represents independently for each occurrence H, alkyl, alkenyl, aryl, aralkyl, or non-aromatic heterocyclyl;
Yi and Y2 independently represent OH, or a group capable of being hydro lyzed to a hydroxyl group; or taken together with the boron to which they are bonded form a 5- membered to 8-membered ring comprising said boron and two oxygen atoms bonded to said boron;
Figure imgf000004_0001
R41 is selected from the group consisting of N3, SH, NH2, NO2, and -ORg';
R42 is selected from the group consisting of hydrogen, lower alkyl, amine, and -OR44, or a pharmaceutically acceptable salt; or R41 and R42 taken together with the phosphorous atom to which they are attached complete a heterocyclic ring having from 5 to 8 atoms in the ring structure; Xl is halogen;
X2 and X3 is hydrogen or halogen; Ri is selected from a C-terminally linked amino acid residue or amino acid analog, a
C-terminally linked peptide or peptide analog, an amino-protecting group,
Figure imgf000004_0002
R2 is absent or represents one or more substituents attached at positions 3, 4, or 5 of the dehydroproline ring, each of which is independently selected from the group consisting of hydrogen, halogen, hydroxyl, lower alkyl, lower alkenyl, lower alkynyl, lower alkoxy, lower hydroxyalkyl, lower alkoxyalkyl, carbonyl, carboxyl, ester, formate, ketone, thiocarbonyl, thioester, thioacetate, thioformate, amino, acylamino, amido, nitro, sulfate, sulfonate, sulfonamido, -(CH2Xn-R51, -(CH2Xn-OH, -(CH2)m-O-lower alkyl, -(CH2Xn-O- lower alkenyl, -(CH2)n-O-(CH2)m-R51, -(CH2Xn-SH, -(CH2)m-S-lower alkyl, -(CH2)m-S-lower alkenyl, or -(CH2X1-S-(CH2Xn-R51' azido, cyano, isocyanato, thiocyanato,
isothiocyanato, cyanato, Nz^=C, and C C ^22;
R3 is selected from the group consisting of hydrogen, halogen, lower alkyl, lower alkenyl, lower alkynyl, carbonyl, thiocarbonyl, amino, acylamino, amido, nitro, sulfate, sulfonate, a sulfonamido, -(CH2Xn-R51, -(CH2Xn-OH, -(CH2)m-O-lower alkyl, -(CH2Xn-O- lower alkenyl, -(CH2)n-O-(CH2)m-R5 { , -(CH2Xn-SH, -(CH2)m-S-lower alkyl, -(CH2Xn-S- lower alkenyl, or -(CH2)n-S-(CH2)m-R5i, azido, cyano, isocyanato, thiocyanato,
isothiocyanato, cyanato,
Figure imgf000005_0001
and CzzzzzzzzC R22;
R5 is selected from the group consisting of hydrogen, halogen, alkyl, alkenyl, alkynyl, aryl, -(CH2)m-R51, -(CH2)m-OH, -(CH2)m-O-alkyl, -(CH2)m-O-alkenyl, -(CH2)m-O-alkynyl, -(CH2)m-O-(CH2)m-R51, -(CH2)m-SH, -(CH2)m-S-alkyl, -(CH2)m-S- alkenyl, -(CH2)m-S-alkynyl, -(CH2)m-S-(CH2)m-R5i,
° ,R52 NH2 0 (CH2)m— N^ (CH2)n— C-N^ (CH2)n— NH2-C-NH2 , —(CH2)n— C-O-R51 ,
R53 ' R53 '
0 0 0 O
Il Il Il Il
— (CH2)n— C— alkyl , — (CH2)n— C— alkenyl , — (CH2)n— C— alkynyl , and — (CH2)n-C-(CH2)m-R51; wherein
R51 represents for each occurrence aryl, aralkyl, or non-aromatic heterocyclyl; R52 and R53 independently represent hydrogen, alkyl, alkenyl, -(CH2)m-R5i,
-C(=O)-alkyl, -C(=O)-alkenyl, -C(=O)-alkynyl, or -C(=O)-(CH2)m-R5i; or R52 and R53 taken together with the N atom to which they are attached complete a heterocyclic ring having from 4 to 8 atoms in the ring structure; m is zero or an integer in the range of 1 to 8; and n is an integer in the range of 1 to 8.
In certain embodiments, W is a group of formula -B(Yi)(Y2). In certain embodiments, such as when W has one of the values described above, Ri is a C-terminally linked amino acid residue or amino acid analog or a C-terminally linked peptide or peptide analog. In certain embodiments, such as when W and Ri have the values described above,
R3 is hydrogen or a lower alkyl group.
In certain embodiments, the compound is represented by:
Figure imgf000005_0002
or pharmaceutically acceptable salts thereof. Another aspect of the invention provides a protease inhibitor having a structure represented by Formula (II):
Figure imgf000006_0001
(H) or a pharmaceutically acceptable salt thereof, wherein:
R2 is absent or represents one or more substituents attached at positions 3, 4, or 5 of the dehydroproline ring, each of which is independently selected from the group consisting of hydrogen, halogen, hydroxyl, lower alkyl, lower alkenyl, lower alkynyl, lower alkoxy, lower hydroxyalkyl, lower alkoxyalkyl, carbonyl, carboxyl, ester, formate, ketone, thiocarbonyl, thioester, thioacetate, thioformate, amino, acylamino, amido, nitro, sulfate, sulfonate, sulfonamido, -(CH2Xn-R43, -(CH2Xn-OH, -(CH2)m-O-lower alkyl, -(CH2Xn-O- lower alkenyl, -(CH2X1-O-(CH2Xn-R43, -(CH2Xn-SH, -(CH2)m-S-lower alkyl, -(CH2)m-S-lower alkenyl, or -(CH2)n-S-(CH2)m-R43' azido, cyano, isocyanato, thiocyanato,
isothiocyanato, cyanato,
Figure imgf000006_0002
and CzzzzzzzzC R22;
Rioi is selected from the group consisting of H, alkyl, alkoxy, alkenyl, alkynyl, amino, alkylamino, acylamino, cyano, sulfonylamino, acyloxy, aryl, non-aromatic heterocyclyl, heteroaryl, and a polypeptide chain of 1 to 8 amino acid residues;
Rio2 is selected from the group consisting of H, lower alkyl, and aralkyl; Rio3 is selected from the group consisting of H, halogen, and alkyl, or R102 and R103 together with the atoms to which they are attached, form a 3- to 6-membered heterocyclic ring;
R105 is selected from the group consisting of H, halogen, lower alkyl, and aralkyl;
R106 is selected from the group consisting of -CN, -CH=NR4,
Figure imgf000007_0001
; wherein
R4 represents H, alkyl, alkenyl, alkynyl, -C(X1)(X2)(X3), -(CH2)Hi-R43, -(CH2)Ii-OH, -(CH2)n-O-alkyl, -(CH2)n-O-alkenyl, -(CH2)n-O-alkynyl, -(CH2)H-O-(CH2)Hi-R43, -(CH2)n-SH, -(CH2)n-S-alkyl, -(CH2)n-S-alkenyl, -(CH2)n-S-alkynyl, -(CH2)H-S-(CH2)Hi-R43, -C(O)C(O)NH2, or -C(O)C(O)OR44;
R43 represents independently for each occurrence aryl, aralkyl, or non-aromatic hetereocyclyl;
R44 independently for each occurrence is selected from the group consisting of H, alkyl, alkenyl, aryl, aralkyl, and non-aromatic heterocyclyl; Yi and Y2 independently represent OH, or a group capable of being hydro lyzed to a hydroxyl group; or taken together with the boron to which they are bonded form a 5- membered to 8-membered ring comprising said boron and two oxygen atoms bonded to said boron;
R40 is O or S; R41 is selected from the group consisting of N3, SH, NH2, NO2 and -OR^;
R42 is selected from the group consisting of H, lower alkyl, amine, and -OR44, or a pharmaceutically acceptable salt; or R41 and R42 taken together with the phosphorous atom to which they are attached complete a heterocyclic ring having from 5 to 8 atoms in the ring structure; X1 is halogen;
X2 and X3 is hydrogen or halogen; and n is an integer in the range of 2 to 6;
R107 is selected from the group consisting of H, aryl, alkyl, aralkyl, non-aromatic heterocyclyl, heteroaryl, heteroaralkyl, and polypeptide chains of 1 to 8 amino acid residues;
L is absent or is selected from the group consisting of alkyl, alkenyl, alkynyl, -(CH2)mO(CH2)m-, -(CH2)mNR2(CH2)m-, and -(CH2)mS(CH2)m-; X is absent or is selected from the group consisting of -N(R107)-, -O-, and -S-;
Y is absent or is selected from the group consisting of -C(=O)-, -C(=S)-, and -SO2-; and m is, independently for each occurrence, an integer from 0 to 10.
In particular embodiments, Rioβ is a group of formula -B(Yi)(Y2).
In certain embodiments, such as when Rioβ has one of the values described above, Rio3 is selected from -H, -CH3, -CH(CH3)2, -CH2CH(CH3)2, -C(CH3)(CH2CH3), -CH2CH2SCH3, -CH2-phenyl, -CH2-(3-indolyl), -CH2OH, -CH(OH)CH3, -CH2CONH2, -CH2CH2CONH2, -CH2-(4-hydroxyphenyl), -CH2SH, -(CH2)4NH2, -(CH2)3NH-C(=NH)NH2, -CH-(5-imidazolyl), -CH2COOH, and -CH2CH2COOH, particularly -H, -CH3 and -(CH2)3NH-C(=NH)NH2.
In certain embodiments, such as when Rioβ and/or Rio3 have the values described above, one or more of L, X and Y is absent. Preferably, two or more of L, X and Y are absent. In other embodiments, each of L, X and Y is absent. When one or more of L, X and Y is absent, Rioi is generally -H.
In many embodiments, one or both of Rio3 and R105 are -H in compound represented by Formula (II). More typically, both Ri03 and R105 are -H.
Exemplary compounds represented by Formula (II) are:
Figure imgf000008_0001
or pharmaceutically acceptable salts thereof.
In certain above embodiments, the protease inhibitor inhibits DPIV with a K1 of 50 nm or less. In other embodiments, the protease inhibitor inhibits dipeptidyl peptidase VIII with a K1 of 100 μM or greater, such as 200 μM or greater, 500 μM or greater or even 1 mM or greater.
In certain embodiments, the inhibitor is orally active.
In certain embodiments, the inhibitor has a therapeutic index in humans of at least 2, at least 5, at least 10, or at least 100; e.g., such as a therapeutic index for regulating glucose metabolism. Another aspect of the invention provides a pharmaceutical composition, comprising a pharmaceutically acceptable carrier; and a compound of the present invention or a pharmaceutically acceptable salt thereof.
Another aspect of the invention provides for use of one or more of the compounds of the present invention in the manufacture of a medicament for inhibiting a post-pro line protease enzyme in vivo. For example, the subject inhibitors may be used to manufacture medicaments for increasing plasma concentrations of one or more peptide hormones processed by post-proline protease enzymes (e.g., DP-IV and the like). Exemplary medicaments are useful in increasing plasma concentrations of such hormones as glucagons-like peptide, NPY, PPY, secretin, GLP-I, GLP-2, and GIP. In certain embodiments, the subject inhibitors may be used to manufacture medicaments for regulating glucose metabolism, such as for use in treating patients suffering from Type II diabetes, insulin resistance, glucose intolerance, hyperglycemia, hypoglycemia, hyperinsulinemia, obesity, hyperlipidemia, or hyperlipoproteinemia.
Yet another aspect of the invention provides a packaged pharmaceutical, comprising a preparation of a compound of the present invention; optionally a pharmaceutically acceptable carrier; and written and/or pictorial instructions describing the use of the preparation for inhibiting a post-proline protease enzyme in vivo, such as for regulating glucose metabolism.
The packaged pharmaceutical can also include, e.g., as a co-formulation with the protease inhibitor or simply co-packaged with the protease inhibitor, insulin and/or an insulinotropic agent.
The packaged pharmaceutical can also include, e.g., as a co-formulation with the protease inhibitor or simply co-packaged with the protease inhibitor, an Ml receptor antagonist, a prolactin inhibitor, agents acting on the ATP-dependent potassium channel of β-cells, metformin, and/or glucosidase inhibitors.
The present invention also relates to improved methods for the long-term reduction and abatement of at least one of the foregoing disorders based on a therapeutic regimen administered over the short-term.
The present invention further provides a method for regulating and altering on a long-term basis the glucose and lipogenic responses of vertebrate animals, including humans.
In particular, the compounds of the invention may produce long-lasting beneficial changes in one or more of the following: the sensitivity of the cellular response of a species to insulin (reduction of insulin resistance), blood insulin levels, hyperinsulinemia, blood glucose levels, the amount of body fat stores, and blood lipoprotein levels, thus providing effective treatments for diabetes, obesity and/or atherosclerosis.
Detailed Description of the Invention /. Overview
The present invention relates to inhibitors of post-pro line protease enzymes, such as inhibitors of dipeptidyl peptidase IV, as well as pharmaceutical compositions thereof, and methods for using such inhibitors.
Advantageous features for compounds of the present invention may include: better therapeutic indices, owing in part to reduced toxicity and/or improved specificity for the targeted protease; better oral availability; increased shelf-life; and/or increased duration of action (such as single oral dosage formulations which are effective for more than 4 hours, and even more preferably for more than 8, 12, or 16 hours).
The compounds of the present invention may be used as part of treatments for a variety of disorders/conditions, such as those which are mediated by DPIV. For instance, the subject inhibitors may be used to up-regulate GIP and GLP-I activities, e.g., by increasing the half- life of those hormones, as part of a treatment for regulating glucose levels and/or metabolism, e.g., to reduce insulin resistance, treat hyperglycemia, hyperinsulinemia, obesity, hyperlipidemia, hyperlipoproteinemia (such as chylomicrons, VLDL, and LDL), and to regulate body fat and more generally lipid stores, and, more generally, for the improvement of metabolism disorders, especially those associated with diabetes, obesity and/or atherosclerosis. Compounds which inhibit DPIV are, correlatively, able to improve glucose tolerance, though not necessarily through mechanisms involving DPIV inhibition per se.
Indeed, similar compounds have been shown to be effective in mice lacking a GLP-I receptor suggesting that the subject method may not include a mechanism of action directly implicating GLP-I itself, though it has not been ruled out that GLP-I may have other receptors. However, in light of the correlation with DPIV inhibition, in certain embodiments, the subject method utilizes an agent with a K1 for DPIV inhibition of 50.0 nM or less, 10.0 nM or less, or 1.0, 0.1, or even 0.01 nM or less. Indeed, inhibitors with K1 values in the picomolar and even femtomolar range are contemplated. Thus, while the active agents are described herein, for convenience, as "DPIV inhibitors", it will be understood that such nomenclature is not intending to limit the subject invention to a particular mechanism of action.
Certain of the subject compounds may extended duration. Accordingly, in certain embodiments, the inhibitor(s) is selected, and the amount of inhibitor formulated, to provide a dosage which inhibits serum post-pro line protease enzymes (e.g., DPIV) levels by at least 50 % for at least 4 hours after a single dose, for at least 8 hours, or for at least 12 or
16 hours after a single dose.
For instance, in certain embodiments, the method involves administration of a DPIV inhibitor, preferably at a predetermined time(s) during a 24-hour period, in an amount effective to improve one or more aberrant indices associated with glucose metabolism disorders (e.g., glucose intolerance, insulin resistance, hyperglycemia, hyperinsulinemia, and Type I and II diabetes).
In other embodiments, the method involves administration of a DPIV inhibitor in an amount effective to improve aberrant indices associated with obesity. Fat cells release the hormone leptin, which travels in the bloodstream to the brain and, through leptin receptors there, stimulates production of GLP-I. GLP-I, in turn, produces the sensation of being full.
The leading theory is that the fat cells of most obese people probably produce enough leptin, but the leptin may not be able to engage properly the leptin receptors in the brain, and so does not stimulate production of GLP-I. Accordingly, a great deal of research has been focused on utilizing preparations of GLP-I as an appetite suppressant. The subject method provides a means for increasing the half- life of both endogenous and ectopically added GLP-I in the treatment of disorders associated with obesity. In a more general sense, the present invention provides methods and compositions for altering the pharmacokinetics of a variety of different polypeptide hormones by inhibiting the proteolysis by DPIV or some other proteolytic activity of one or more peptide hormones. Post-secretory metabolism is an important element in the overall homeostasis of regulatory peptides, and the other enzymes involved in these processes may be suitable targets for pharmacological intervention by the subject compounds and methods.
In certain instances, the subject method may be used to increase the half-life of other proglucagon-derived peptides, such as glicentin (corresponding to PG 1-69), oxyntomodulin (PG 33-69), glicentin-related pancreatic polypeptide (GRPP, PG 1-30), intervening peptide-2 (IP-2, PG 111-122 amide), and glucagon-like peptide-2 (GLP-2, PG 126-158). GLP-2, for example, has been identified as a factor responsible for inducing proliferation of intestinal epithelium. See, e.g., Drucker et al. Proc. Natl. Acad. Sci. USA 1996, 93, 7911. The DPP IV inhibitors can also be used as part of a regimen for treating injury, inflammation or resection of intestinal tissue, e.g., where enhanced growth and repair of the intestinal mucosal epithelial is desired, such as in the treatment of Crohn's disease or Inflammatory Bowel Disease (IBD).
Another aspect of the invention relates to a method of treating growth hormone deficient children or improving nutrition or altering body composition (muscle vs. fat) in adults. DPP IV has been implicated in the metabolism and inactivation of growth hormone- releasing factor (GHRF). GHRF is a member of the family of homologous peptides that includes glucagon, secretin, vasoactive intestinal peptide (VIP), peptide histidine isoleucine (PHI), pituitary adenylate cyclase activating peptide (PACAP), gastric inhibitory peptide (GIP) and helodermin. Kubiak et al., Peptide Res. 1994, 7, 153. GHRF is secreted by the hypothalamus, and stimulates the release of growth hormone (GH) from the anterior pituitary. The subject method can also be used in veterinary practice, for example, to develop higher yield milk production and higher yield, leaner livestock.
The DPP IV inhibitors of the invention may be used to alter the plasma half-life of secretin, VIP, PHI, PACAP, GIP and/or helodermin. In certain instances, the inhibitors may also be used to alter the pharmacokinetics of Peptide YY and neuropeptide Y, both members of the pancreatic polypeptide family, because DPP IV has been implicated in the processing of those peptides in a manner which alters receptor selectivity. In other embodiments, the DPP IV inhibitors may be used to stimulate hematopoiesis. In still other embodiments, the DPP IV inhibitors may be used to inhibit growth or vascularization of transformed cells/tissues, e.g., to inhibit cell proliferation such as that associated with tumor growth and metastasis, and for inhibiting angiogenesis in an abnormal proliferative cell mass. In yet other embodiments, the subject DPP IV inhibitors may be used as an immunosuppressant. In yet other examples, the DPP IV inhibitors according to the present invention may 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 may be used to treat disorders having a more peripheral nature, including multiple sclerosis and diabetic neuropathy.
Another aspect of the present invention provides a method for stimulating hematopoietic cells in culture or in vivo. In certain embodiments, the subject DPP IV pro- inhibitors include an address moiety that is a substrate for a protease that is expressed in bone marrow. The DPP IV inhibitors of the invention may be used to restore or prevent a deficiency in hematopoietic cell number in a subject. Such deficiencies can arise, for example, from genetic abnormalities, disease, stress, chemotherapy, and radiation treatment.
In certain embodiments, the present invention provides a method of reducing the rate of degradation of tumor suppressors. In other embodiments, compounds of the present invention inhibit the growth of cancer cells. In yet other embodiments, the compounds of the present invention may be formulated in topical form for treatment of skin disorders. Such inhibitors are contemplated as possessing important practical application in treating cell proliferative diseases, such as cancer, restenosis, and psoriasis.
In other embodiments, the subject inhibitors may be used to inhibit growth or vascularization of transformed cells/tissues, e.g., to inhibit cell proliferation, such as that associated with tumor growth and metastasis, and for inhibiting angiogenesis in an abnormal proliferative cell mass.
In yet other embodiments, the subject inhibitors may be used as an immunosuppressant. Another aspect of the present invention relates to pharmaceutical compositions of the subject post-proline protease enzyme inhibitors, particularly DPIV inhibitors, and their uses in treating and/or preventing disorders which can be improved by altering the homeostasis of peptide hormones. In a certain embodiments, the inhibitors have hypoglycemic and antidiabetic activities, and may be used in the treatment of disorders marked by aberrant glucose metabolism (including storage). In particular embodiments, the compositions of the subject methods are useful as insulinotropic agents, or to potentiate the insulinotropic effects of such molecules as GLP-I. In this regard, certain embodiments of the present compositions may be useful for the treatment and/or prophylaxis of a variety of disorders, including one or more of: hyperlipidemia, hyperglycemia, obesity, glucose tolerance insufficiency, insulin resistance, and diabetic complications.
In general, the inhibitors of the subject method are small molecules, e.g., with molecular weights less than 7500 amu, less than 5000 amu, less than 2000, or less than 1000 amu. In certain embodiments, the inhibitors are orally active. //. Definitions
The term "high affinity" as used herein means strong binding affinity between molecules with a dissociation constant KD of no greater than 1 μM. In a certain case, the KD is less than 100 nM, 10 nM, 1 nM, 100 pM, or even 10 pM or less. In another embodiment, the two molecules can be covalently linked (KD is essentially 0).
The term "boro-Ala" refers to the analog of alanine in which the carboxylate group (COOH) is replaced with a boronyl group (B(OH)2). Likewise, the term "boro-Pro" refers to the analog of pro line in which the carboxylate group (COOH) is replaced with a boronyl group (B(OH)2). More generally, the term "boro-Xaa", where Xaa is an amino acid residue, refers to the analog of an amino acid in which the carboxylate group (COOH) is replaced with a boronyl group (B(OH)2).
A "patient" or "subject" to be treated by the subject method can mean either a human or non-human subject.
The term "ED50" means the dose of a drug that, in 50% of patients, will provide a clinically relevant improvement or change in a physiological measurement, such as glucose responsiveness, increase in hematocrit, decrease in tumor volume, etc.
The term "IC50" means the dose of a drug that inhibits a biological activity by 50%, e.g., the amount of inhibitor required to inhibit at least 50% of DPIV (or other PPCE) activity in vivo. As used herein, the term "inhibitor" is meant to describe a compound that blocks or reduces an activity of an enzyme (for example, inhibition of proteolytic cleavage of standard fluorogenic peptide substrates such as suc-LLVY-AMC, Box-LLR-AMC and Z- LLE-AMC, inhibition of various catalytic activities of the 2OS proteasome). An inhibitor can act with competitive, uncompetitive, or noncompetitive inhibition. An inhibitor can bind reversibly or irreversibly, and therefore the term includes compounds that are suicide substrates of an enzyme. An inhibitor can modify one or more sites on or near the active site of the enzyme, or it can cause a conformational change elsewhere on the enzyme. A compound is said to have an "insulinotropic activity" if it is able to stimulate, or cause the stimulation of, the synthesis or expression of the hormone insulin.
The term "interact" as used herein is meant to include all interactions (e.g., biochemical, chemical, or biophysical interactions) between molecules, such as protein- protein, protein-nucleic acid, nucleic acid-nucleic acid, protein-small molecule, nucleic acid-small molecule, or small molecule-small molecule interactions.
The term "LD50" means the dose of a drug that is lethal in 50% of test subjects.
The term "prophylactic or therapeutic" treatment is art-recognized and includes administration to the host of one or more of the subject compositions. If it is administered prior to clinical manifestation of the unwanted condition (e.g., disease or other unwanted state of the host animal) then the treatment is prophylactic, (i.e., it protects the host against developing the unwanted condition), whereas if it is administered after manifestation of the unwanted condition, the treatment is therapeutic, (i.e., it is intended to diminish, ameliorate, or stabilize the existing unwanted condition or side effects thereof).
The term "preventing" is art-recognized, and when used in relation to a condition, such as a local recurrence (e.g., pain), a disease such as cancer, a syndrome complex such as heart failure or any other medical condition, is well understood in the art, and includes administration of a composition which reduces the frequency of, or delays the onset of, symptoms of a medical condition in a subject relative to a subject which does not receive the composition. Thus, prevention of cancer includes, for example, reducing the number of detectable cancerous growths in a population of patients receiving a prophylactic treatment relative to an untreated control population, and/or delaying the appearance of detectable cancerous growths in a treated population versus an untreated control population, e.g., by a statistically and/or clinically significant amount. Prevention of an infection includes, for example, reducing the number of diagnoses of the infection in a treated population versus an untreated control population, and/or delaying the onset of symptoms of the infection in a treated population versus an untreated control population. Prevention of pain includes, for example, reducing the magnitude of, or alternatively delaying, pain sensations experienced by subjects in a treated population versus an untreated control population. The term "therapeutic index" refers to the therapeutic index of a drug defined as LD50/ED50.
A "therapeutically effective amount" of a compound, e.g., such as a DPIV inhibitor of the present invention, with respect to the subject method of treatment, refers to an amount of the compound(s) in a preparation which, when administered as part of a desired dosage regimen (to a mammal, preferably a human) alleviates a symptom, ameliorates a condition, or slows the onset of disease conditions according to clinically acceptable standards for the disorder or condition to be treated or the cosmetic purpose, e.g., at a reasonable benefit/risk ratio applicable to any medical treatment. A "single oral dosage formulation" is a dosage which provides an amount of drug to produce a serum concentration at least as great as the EC50 for that drug, but less than the LD50. Another measure for a single oral dosage formulation is that it provides an amount of drug necessary to produce a serum concentration at least as great as the IC50 for that drug, but less than the LD50. By either measure, a single oral dosage formulation is preferably an amount of drug which produces a serum concentration at least 10 percent less than the
LD50, and even more preferably at least 50 percent, 75 percent, or even 90 percent less than the drug's the LD50.
As used herein, the term "inhibitor" is meant to describe a compound that blocks or reduces an activity of an enzyme (for example, inhibition of proteolytic cleavage of standard fluorogenic peptide substrates such as suc-LLVY-AMC, Box-LLR-AMC and Z- LLE-AMC, inhibition of various catalytic activities of the 2OS proteasome). An inhibitor can act with competitive, uncompetitive, or noncompetitive inhibition. An inhibitor can bind reversibly or irreversibly, and therefore the term includes compounds that are suicide substrates of an enzyme. An inhibitor can modify one or more sites on or near the active site of the enzyme, or it can cause a conformational change elsewhere on the enzyme.
The term "Cx_yalkyl" refers to substituted or unsubstituted saturated hydrocarbon groups, including straight-chain alkyl and branched-chain alkyl groups that contain from x to y carbons in the chain, including haloalkyl groups, such as trifluoromethyl and 2,2,2- tirfluoroethyl, etc. Co alkyl indicates a hydrogen where the group is in a terminal position, a bond if internal. The terms "C2-yalkenyl" and "C2_yalkynyl" refer to substituted or unsubstituted unsaturated aliphatic groups analogous in length and possible substitution to the alkyls described above, but that contain at least one double or triple bond respectively. Unless the number of carbons is otherwise specified, "lower alkyl", as used herein, means an alkyl group, as defined above, but having from one to ten carbons, more preferably from one to six carbon atoms in its backbone structure such as methyl, ethyl, n- propyl, isopropyl, n-butyl, isobutyl, sec-butyl, and tert-butyl. Likewise, "lower alkenyl" and "lower alkynyl" have similar chain lengths. In certain embodiments, a substituent designated herein as alkyl is a lower alkyl.
The terms "alkoxyl" or "alkoxy" as used herein refers to an alkyl group, as defined below, having an oxygen moiety attached thereto. Representative alkoxyl groups include methoxy, ethoxy, propoxy, tert-butoxy, and the like. An "ether" is two hydrocarbons covalently linked by an oxygen. Accordingly, the substituent of an alkyl that renders that alkyl an ether is or resembles an alkoxyl, such as can be represented by one of -O-alkyl, -O- alkenyl, -O-alkynyl, -O-(CH2)m-Ri, where m and Ri are described below.
The terms "amine" and "amino" are art-recognized and refer to both unsubstituted and substituted amines, e.g., a moiety that can be represented by the general formulae:
Figure imgf000017_0001
wherein R35 Rs, and R6 each independently represent a hydrogen, an alkyl, an alkenyl, -(CH2)m-Ri, or R3 and R5 taken together with the N atom to which they are attached complete a heterocycle having from 4 to 8 atoms in the ring structure; Ri represents an alkenyl, aryl, cycloalkyl, a cycloalkenyl, a heterocyclyl, or a polycyclyl; and m is zero or an integer in the range of 1 to 8. In certain embodiments, only one of R3 or R5 can be a carbonyl, e.g., R3, R5, and the nitrogen together do not form an imide. In even more certain embodiments, R3 and R5 (and optionally R6) each independently represent a hydrogen, an alkyl, an alkenyl, or -(CH2)m-Ri. Thus, the term "alkylamine" as used herein means an amine group, as defined above, having a substituted or unsubstituted alkyl attached thereto, i.e., at least one of R3 and R5 is an alkyl group. In certain embodiments, an amino group or an alkylamine is basic, meaning it has a conjugate acid with a pKa >7.00, i.e., the protonated forms of these functional groups have pKas relative to water above about 7.00..
The term "carbonyl" is art-recognized and includes such moieties as can be represented by the general formula:
Figure imgf000017_0002
wherein X is a bond or represents an oxygen or a sulfur, and R7 represents a hydrogen, an alkyl, an alkenyl, -(CH2)m-Ri or a pharmaceutically acceptable salt, R8 represents a hydrogen, an alkyl, an alkenyl or -(CH2)m-Ri, where m and Ri are as defined above. Where X is an oxygen and R7 or R8 is not hydrogen, the formula represents an "ester." Where X is an oxygen, and R7 is as defined above, the moiety is referred to herein as a carboxyl group, and particularly when R7 is a hydrogen, the formula represents a "carboxylic acid." Where X is an oxygen, and R8 is a hydrogen, the formula represents a "formate." In general, where the oxygen atom of the above formula is replaced by a sulfur, the formula represents a "thiocarbonyl" group. Where X is a sulfur and R7 or R8 is not hydrogen, the formula represents a "thioester" group. Where X is a sulfur and R7 is a hydrogen, the formula represents a "thiocarboxylic acid" group. Where X is a sulfur and R8 is a hydrogen, the formula represents a "thioformate" group. On the other hand, where X is a bond, and R7 is not hydrogen, the above formula represents a "ketone" group. Where X is a bond, and R7 is a hydrogen, the above formula represents an "aldehyde" group. The terms "heterocyclyl" or "heterocyclic group" refer to substituted or unsubstituted non-aromatic 3- to 10-membered ring structures, more preferably 3- to 7- membered rings, whose ring structures include one to four heteroatoms. The term terms "heterocyclyl" or "heterocyclic group" also include polycyclic ring systems having two or more cyclic rings in which two or more carbons are common to two adjoining rings wherein at least one of the rings is heterocyclic, e.g., the other cyclic rings can be cycloalkyls, cycloalkenyls, cycloalkynyls, aryls, heteroaryls, and/or heterocyclyls. Heterocyclyl groups include, for example, piperidine, piperazine, pyrrolidine, morpholine, lactones, lactams, and the like.
The term "substituted" refers to moieties having substituents replacing a hydrogen on one or more carbons of the backbone. It will be understood that "substitution" or
"substituted with" includes the implicit proviso that such substitution is in accordance with permitted valence of the substituted atom and the substituent, and that the substitution results in a stable compound, e.g., which does not spontaneously undergo transformation such as by rearrangement, cyclization, elimination, etc. As used herein, the term "substituted" is contemplated to include all permissible substituents of organic compounds. In a broad aspect, the permissible substituents include acyclic and cyclic, branched and unbranched, carbocyclic and heterocyclic, aromatic and non-aromatic substituents of organic compounds. The permissible substituents can be one or more and the same or different for appropriate organic compounds. For purposes of this invention, the heteroatoms such as nitrogen may have hydrogen substituents and/or any permissible substituents of organic compounds described herein which satisfy the valences of the heteroatoms. Substituents can include, for example, a halogen, a hydroxyl, a carbonyl (such as a carboxyl, an alkoxycarbonyl, a formyl, or an acyl), a thiocarbonyl (such as a thioester, a thioacetate, or a thioformate), an alkoxyl, a phosphoryl, a phosphate, a phosphonate, a phosphinate, an amino, an amido, an amidine, an imine, a cyano, a nitro, an azido, a sulfhydryl, an alkylthio, a sulfate, a sulfonate, a sulfamoyl, a sulfonamido, a sulfonyl, a heterocyclyl, an aralkyl, or an aromatic or heteroaromatic moiety. It will be understood by those skilled in the art that the moieties substituted on the hydrocarbon chain can themselves be substituted, if appropriate.
As used herein, the definition of each expression, e.g., alkyl, m, n, etc., when it occurs more than once in any structure, is intended to be independent of its definition elsewhere in the same structure. The terms "amino acid residue" and "peptide residue" mean an amino acid or peptide molecule without the -OH of its carboxyl group. In general the abbreviations used herein for designating the amino acids and the protective groups are based on recommendations of the IUPAC-IUB Commission on Biochemical Nomenclature (see Biochemistry 1972, 11, 1726-1732). For instance, Met, He, Leu, Ala, and GIy represent "residues" of methionine, isoleucine, leucine, alanine, and glycine, respectively. Residue means a moiety derived from the corresponding α-amino acid by eliminating the OH portion of the carboxyl group and the H portion of the α-amino group. The term "amino acid side chain" is that part of an amino acid exclusive of the -CH(NH2)COOH portion, as defined by K. D. Kopple, Peptides and Amino Acids; Benjamin: New York, 1966; pp. 2 and 33; examples of such side chains of the common amino acids are -CH2CH2SCH3 (the side chain of methionine), -CH2(CH3)-CH2CH3 (the side chain of isoleucine), -CH2CH(CH3)2 (the side chain of leucine) or H-(the side chain of glycine).
In certain embodiments, the amino acids used in the present invention are those naturally occurring amino acids found in native proteins, or the naturally occurring anabolic or catabolic products of such amino acids which contain amino and carboxyl groups.
Particularly suitable amino acid side chains include side chains selected from those of the following amino acids: glycine, alanine, valine, cysteine, leucine, isoleucine, serine, threonine, methionine, glutamic acid, aspartic acid, glutamine, asparagine, lysine, arginine, proline, histidine, phenylalanine, tyrosine, and tryptophan, and those amino acids and amino acid analogs which have been identified as constituents of peptidylglycan bacterial cell walls.
The term amino acid residue further includes analogs, derivatives and congeners of any specific amino acid referred to herein, as well as C-terminal or N-terminal protected amino acid derivatives (e.g., modified with an N-terminal or C-terminal protecting group). For example, the present invention contemplates the use of amino acid analogs wherein a side chain is lengthened or shortened while still providing a carboxyl, amino or other reactive precursor functional group for cyclization, as well as amino acid analogs having variant side chains with appropriate functional groups). For instance, the subject compound can include an amino acid analog such as, for example, cyanoalanine, canavanine, djenkolic acid, norleucine, 3-phosphoserine, homoserine, dihydroxy-phenylalanine, 5- hydroxytryptophan, 1-methylhistidine, 3-methylhistidine, diaminopimelic acid, ornithine, or diaminobutyric acid. Other naturally occurring amino acid metabolites or precursors having side chains which are suitable herein will be recognized by those skilled in the art and are included in the scope of the present invention.
Also included are the (D) and (L) stereoisomers of such amino acids when the structure of the amino acid admits of stereoisomeric forms. The configuration of the amino acids and amino acid residues herein are designated by the appropriate symbols (D), (L) or (DL), furthermore when the configuration is not designated, the amino acid or residue can have the configuration (D), (L), or (DL). It will be noted that the structure of some of the compounds of this invention includes asymmetric carbon atoms. It is to be understood accordingly that the isomers arising from such asymmetry are included within the scope of this invention. Such isomers can be obtained in substantially pure form by classical separation techniques and by sterically controlled synthesis. For the purposes of this application, unless expressly noted to the contrary, a named amino acid shall be construed to include both the (D) and (L) stereoisomers.
The term "peptide analogue," as used herein, refers to a peptide comprising one or more non-naturally occurring amino acid. Examples of non-naturally occurring amino acids include, but are not limited to, D-amino acids (i.e., an amino acid of an opposite chirality to the naturally occurring form), JV-α-methyl amino acids, C-α-methyl amino acids, β-methyl amino acids, β-alanine (β-Ala), norvaline (Nva), norleucine (NIe), 4-aminobutyric acid (γ-Abu), 2-aminoisobutyric acid (Aib), 6-aminohexanoic acid (ε-Ahx), ornithine (orn), hydroxyproline (Hyp), sarcosine, citrulline, cysteic acid, cyclohexylalanine, α-amino isobutyric acid, t-butylglycine, t-butylalanine, 3-aminopropionic acid, 2,3-diaminopropionic acid (2,3-diaP), D- or L-phenylglycine, D- or L-2-naphthylalanine (2 -NaI), 1,2,3,4- tetrahydroisoquinoline-3-carboxylic acid (Tic), D- or L-2-thienylalanine (Thi), D- or L-3- thienylalanine, D- or L-I-, 2-, 3- or 4-pyrenylalanine, D- or L-(2-pyridinyl)-alanine, D- or L- (3-pyridinyl)-alanine, D- or L-(2-pyrazinyl)-alanine, D- or L-(4-isopropyl)-phenylglycine, D- (trifluoromethyl)-phenylglycine, D-(trifluoromethyl)-phenylalanine, D-p- fluorophenylalanine, D- or L-p-biphenylalanine, D- or L-p-methoxybiphenylalanine, methionine sulphoxide (MSO) and homoarginine (Har). Other examples include D- or L-2- indole(alkyl)alanines and D- or L-alkylalanines, wherein alkyl is substituted or unsubstituted methyl, ethyl, propyl, hexyl, butyl, pentyl, isopropyl, iso-butyl, or iso-pentyl, and phosphono- or sulfated (e.g., -SO3H) non-carboxylate amino acids.
Other examples of non-naturally occurring amino acids include 3-(2-chlorophenyl)- alanine, 3-chloro-phenylalanine, 4-chloro-phenylalanine, 2-fluoro-phenylalanine, 3-fluoro- phenylalanine, 4-fluoro-phenylalanine, 2-bromo-phenylalanine, 3-bromo-phenylalanine, 4- bromo-phenylalanine, homophenylalanine, 2-methyl-phenylalanine, 3-methyl- phenylalanine, 4-methyl-phenylalanine, 2,4-dimethyl-phenylalanine, 2-nitro-phenylalanine, 3-nitro-phenylalanine, 4-nitro-phenylalanine, 2,4-dinitro-phenylalanine, 1,2,3,4- Tetrahydroisoquinoline-3-carboxylic acid, l,2,3,4-tetrahydronorharman-3-carboxylic acid, 1-naphthylalanine, 2-naphthylalanine, pentafluorophenylalanine, 2,4-dichloro- phenylalanine, 3,4-dichloro-phenylalanine, 3,4-difluoro-phenylalanine, 3,5-difluoro- phenylalanine, 2,4,5-trifluoro-phenylalanine, 2-trifluoromethyl-phenylalanine, 3- trifluoromethyl-phenylalanine, 4-trifluoromethyl-phenylalanine, 2-cyano-phenyalanine, 3- cyano-phenyalanine, 4-cyano-phenyalanine, 2-iodo-phenyalanine, 3-iodo-phenyalanine, 4- iodo-phenyalanine, 4-methoxyphenylalanine, 2-aminomethyl-phenylalanine, 3- aminomethyl-phenylalanine, 4-aminomethyl-phenylalanine, 2-carbamoyl-phenylalanine, 3- carbamoyl-phenylalanine, 4-carbamoyl-phenylalanine, m-tyrosine, 4-amino-phenylalanine, styrylalanine, 2-amino-5-phenyl-pentanoic acid, 9-anthrylalanine, 4-tert-butyl- phenylalanine, 3,3-diphenylalanine, 4,4'-diphenylalanine, benzoylphenylalanine, α-methyl- phenylalanine, α-methyl-4-fluoro-phenylalanine, 4-thiazolylalanine, 3-benzothienylalanine, 2-thienylalanine, 2-(5-bromothienyl)-alanine, 3 -thienylalanine, 2-furylalanine, 2- pyridylalanine, 3-pyridylalanine, 4-pyridylalanine, 2,3-diaminopropionic acid, 2,4- diaminobutyric acid, allylglycine, 2-amino-4-bromo-4-pentenoic acid, propargylglycine, 4- aminocyclopent-2-enecarboxylic acid, 3-aminocyclopentanecarboxylic acid, 7-amino- heptanoic acid, dipropylglycine, pipecolic acid, azetidine-3-carboxylic acid, cyclopropylglycine, cyclopropylalanine, 2-methoxy-phenylglycine, 2-thienylglycine, 3- thienylglycine, α-benzyl-proline, α-(2-fluoro-benzyl)-proline, α-(3-fluoro-benzyl)-proline, α-(4-fluoro-benzyl)-proline, α-(2-chloro-benzyl)-proline, α-(3-chloro-benzyl)-proline, α-(4- chloro-benzyl)-proline, α-(2-bromo-benzyl)-proline, α-(3-bromo-benzyl)-proline, α-(4- bromo-benzyl)-proline, α-phenethyl-proline, α-(2-methyl-benzyl)-proline, α-(3-methyl- benzyl)-proline, α-(4-methyl-benzyl)-proline, α-(2-nitro-benzyl)-proline, α-(3-nitro- benzyl)-proline, α-(4-nitro-benzyl)-proline, α-(l-naphthalenylmethyl)-proline, α-(2- naphthalenylmethyl)-proline, α-(2,4-dichloro-benzyl)-proline, α-(3,4-dichloro-benzyl)- proline, α-(3,4-difluoro-benzyl)-proline, α-(2-trifluoromethyl-benzyl)-proline, α-(3- trifluoromethyl-benzyl)-proline, α-(4-trifluoromethyl-benzyl)-proline, α-(2-cyano-benzyl)- proline, α-(3-cyano-benzyl)-proline, α-(4-cyano-benzyl)-proline, α-(2-iodo-benzyl)-proline, α-(3-iodo-benzyl)-proline, α-(4-iodo-benzyl)-proline, α-(3-phenyl-allyl)-proline, α-(3- phenyl-propyl)-proline, α-(4-tert-butyl-benzyl)-proline, α-benzhydryl-proline, α-(4- biphenylmethyl)-proline, α-(4-thiazolylmethyl)-proline, α-(3-benzo[b]thiophenylmethyl)- proline, α-(2-thiophenylmethyl)-proline, α-(5-bromo-2-thiophenylmethyl)-proline, α-(3- thiophenylmethyl)-proline, α-(2-furanylmethyl)-proline, α-(2-pyridinylmethyl)-proline, α- (3-pyridinylmethyl)-proline, α-(4-pyridinylmethyl)-proline, α-allyl-proline, α-propynyl- proline, γ-benzyl-proline, γ-(2-fluoro-benzyl)-proline, γ-(3-fluoro-benzyl)-proline, γ-(4- fluoro-benzyl)-proline, γ-(2-chloro-benzyl)-proline, γ-(3-chloro-benzyl)-proline, γ-(4- chloro-benzyl)-proline, γ-(2-bromo-benzyl)-proline, γ-(3-bromo-benzyl)-proline, γ-(4- bromo-benzyl)-proline, γ-(2-methyl-benzyl)-proline, γ-(3-methyl-benzyl)-proline, γ-(4- methyl-benzyl)-proline, γ-(2-nitro-benzyl)-proline, γ-(3-nitro-benzyl)-proline, γ-(4-nitro- benzyl)-proline, γ-(l-naphthalenylmethyl)-proline, γ-(2-naphthalenylmethyl)-proline, γ- (2,4-dichloro-benzyl)-proline, γ-(3 ,4-dichloro-benzyl)-proline, γ-(3 ,4-difluoro-benzyl)- proline, γ-(2-trifluoromethyl-benzyl)-proline, γ-(3-trifluoromethyl-benzyl)-proline, γ-(4- trifluoromethyl-benzyl)-proline, γ-(2-cyano-benzyl)-proline, γ-(3-cyano-benzyl)-proline, γ- (4-cyano-benzyl)-proline, γ-(2-iodo-benzyl)-proline, γ-(3-iodo-benzyl)-proline, γ-(4-iodo- benzyl)-proline, γ-(3-phenyl-allyl-benzyl)-proline, γ-(3-phenyl-propyl-benzyl)-proline, γ- (4-tert-butyl-benzyl)-proline, γ-benzhydryl-proline, γ-(4-biphenylmethyl)-proline, γ-(4- thiazolylmethyl)-proline, γ-(3 -benzothioienylmethyl)-proline, γ-(2-thienylmethyl)-proline, γ-(3-thienylmethyl)-proline, γ-(2-furanylmethyl)-proline, γ-(2-pyridinylmethyl)-proline, γ- (3-pyridinylmethyl)-proline, γ-(4-pyridinylmethyl)-proline, γ-allyl-proline, γ-propynyl- proline, trans-4-phenyl-pyrrolidine-3-carboxylic acid, trans-4-(2-fluoro-phenyl)- pyrrolidine-3-carboxylic acid, trans-4-(3-fluoro-phenyl)-pyrrolidine-3-carboxylic acid, trans-4-(4-fluoro-phenyl)-pyrrolidine-3-carboxylic acid, trans-4-(2-chloro-phenyl)- pyrrolidine-3-carboxylic acid, trans-4-(3-chloro-phenyl)-pyrrolidine-3-carboxylic acid, trans-4-(4-chloro-phenyl)-pyrrolidine-3-carboxylic acid, trans-4-(2-bromo-phenyl)- pyrrolidine-3-carboxylic acid, trans-4-(3-bromo-phenyl)-pyrrolidine-3-carboxylic acid, trans-4-(4-bromo-phenyl)-pyrrolidine-3-carboxylic acid, trans-4-(2-methyl-phenyl)- pyrrolidine-3-carboxylic acid, trans-4-(3-methyl-phenyl)-pyrrolidine-3-carboxylic acid, trans-4-(4-methyl-phenyl)-pyrrolidine-3-carboxylic acid, trans-4-(2-nitro-phenyl)- pyrrolidine-3-carboxylic acid, trans-4-(3-nitro-phenyl)-pyrrolidine-3-carboxylic acid, trans- 4-(4-nitro-phenyl)-pyrrolidine-3-carboxylic acid, trans-4-(l-naphthyl)-pyrrolidine-3- carboxylic acid, trans-4-(2-naphthyl)-pyrrolidine-3-carboxylic acid, trans-4-(2,5-dichloro- phenyl)-pyrrolidine-3-carboxylic acid, trans-4-(2,3-dichloro-phenyl)-pyrrolidine-3- carboxylic acid, trans-4-(2-trifluoromethyl-phenyl)-pyrrolidine-3-carboxylic acid, trans-4- (3 -trifluoromethyl-phenyl)-pyrrolidine-3 -carboxylic acid, trans-4-(4-trifluoromethyl- phenyl)-pyrrolidine-3 -carboxylic acid, trans-4-(2-cyano-phenyl)-pyrrolidine-3 -carboxylic acid, trans-4-(3-cyano-phenyl)-pyrrolidine-3 -carboxylic acid, trans-4-(4-cyano-phenyl)- pyrrolidine-3 -carboxylic acid, trans-4-(2-methoxy-phenyl)-pyrrolidine-3 -carboxylic acid, trans-4-(3-methoxy-phenyl)-pyrrolidine-3-carboxylic acid, trans-4-(4-methoxy-phenyl)- pyrrolidine-3 -carboxylic acid, trans-4-(2-hydroxy-phenyl)-pyrrolidine-3 -carboxylic acid, trans-4-(3 -hydroxy-phenyl)-pyrrolidine-3 -carboxylic acid, trans-4-(4-hydroxy-phenyl)- pyrrolidine-3 -carboxylic acid, trans-4-(2,3-dimethoxy-phenyl)-pyrrolidine-3-carboxylic acid, trans-4-(3,4-dimethoxy-phenyl)-pyrrolidine-3 -carboxylic acid, trans-4-(3,5- dimethoxy-phenyl)-pyrrolidine-3 -carboxylic acid, trans-4-(2-pyridinyl)-pyrrolidine-3- carboxylic acid, trans-4-(3-pyridinyl)-pyrrolidine-3 -carboxylic acid, trans-4-(6-methoxy-3- pyridinyl)-pyrrolidine-3 -carboxylic acid, trans-4-(4-pyridinyl)-pyrrolidine-3 -carboxylic acid, trans-4-(2-thienyl)-pyrrolidine-3 -carboxylic acid, trans-4-(3-thienyl)-pyrrolidine-3- carboxylic acid, trans-4-(2-furanyl)-pyrrolidine-3 -carboxylic acid, trans-4-isopropyl- pyrrolidine-3-carboxylic acid, 4-phosphonomethyl-phenylalanine, benzyl- phospho threonine, (1 '-amino-2-phenyl-ethyl)oxirane, (1 '-amino-2-cyclohexyl- ethyl)oxirane, (1 '-amino-2-[3-bromo-phenyl]ethyl)oxirane, (1 '-amino-2-[4- (benzyloxy)phenyl]ethyl)oxirane, (1 '-amino-2-[3,5-difluoro-phenyl]ethyl)oxirane, (1 '- amino-2-[4-carbamoyl-phenyl]ethyl)oxirane, (1 '-amino-2-[benzyloxy-ethyl])oxirane, (1 '- amino-2-[4-nitro-phenyl]ethyl)oxirane, (1 '-amino-3-phenyl-propyl)oxirane, (1 '-amino-3- phenyl-propyl)oxirane, and/or salts and/or protecting group variants thereof.
The term "peptide derivative," as used herein, refers to a peptide comprising additional chemical or biochemical moieties not normally a part of a naturally occurring peptide. Peptide derivatives include peptides in which the amino-terminus and/or the carboxy-terminus and/or one or more amino acid side chain has been derivatised with a suitable chemical substituent group, as well as cyclic peptides, dual peptides, multimers of the peptides, peptides fused to other proteins or carriers, glycosylated peptides, phosphorylated peptides, peptides conjugated to lipophilic moieties (for example, caproyl, lauryl, stearoyl moieties) and peptides conjugated to an antibody or other biological ligand. Examples of chemical substituent groups that may be used to derivatise a peptide include, but are not limited to, alkyl, cycloalkyl and aryl groups; acyl groups, including alkanoyl and aroyl groups; esters; amides; halogens; hydroxyls; carbamyls, and the like. The substituent group may also be a blocking group such as Fmoc (fluorenylmethyl-O-CO-), carbobenzoxy (benzyl-O-CO-), monomethoxysuccinyl, naphthyl-NH-CO-, acetylamino- caproyl and adamantyl-NH-CO-. Other derivatives include C-terminal hydroxymethyl derivatives, O-modifϊed derivatives (for example, C-terminal hydroxymethyl benzyl ether) and //-terminally modified derivatives including substituted amides such as alkylamides and hydrazides. The substituent group may be a "protecting group" as detailed herein.
The phrase "protecting group" as used herein means substituents which protect the reactive functional group from undesirable chemical reactions. Examples of such protecting groups include esters of carboxylic acids and boronic acids, ethers of alcohols, and acetals and ketals of aldehydes and ketones. For instance, the phrase "N-terminal protecting group" or "amino-protecting group" as used herein refers to various amino- protecting groups which can be employed to protect the N-terminus of an amino acid or peptide against undesirable reactions during synthetic procedures. Examples of suitable groups include acyl protecting groups such as, to illustrate, formyl, dansyl, acetyl, benzoyl, trifluoroacetyl, succinyl, and methoxysuccinyl; aromatic urethane protecting groups as, for example, benzyloxycarbonyl (Cbz); and aliphatic urethane protecting groups such as t- butoxycarbonyl (Boc) or 9-Fluorenylmethoxycarbonyl (Fmoc).
The term "amino-protecting group" or 'W-terminal protecting group" refers to those groups intended to protect the α-iV-terminal of an amino acid or peptide or to otherwise protect the amino group of an amino acid or peptide against undesirable reactions during synthetic procedures. Commonly used JV-protecting groups are disclosed in Greene, Protective Groups In Organic Synthesis, (John Wiley & Sons, New York (1981)), which is hereby incorporated by reference. Additionally, protecting groups may be used as pro- drugs which are readily cleaved in vivo, for example, by enzymatic hydrolysis, to release the biologically active parent. α-iV-Protecting groups comprise lower alkanoyl groups such as formyl, acetyl ("Ac"), propionyl, pivaloyl, t-butylacetyl and the like; other acyl groups include 2-chloroacetyl, 2-bromoacetyl, trifluoroacetyl, trichloroacetyl, phthalyl, o- nitrophenoxyacetyl, -chlorobutyryl, benzoyl, 4-chlorobenzoyl, 4-bromobenzoyl, 4- nitrobenzoyl and the like; sulfonyl groups such as benzenesulfonyl, p-toluenesulfonyl and the like; carbamate forming groups such as benzyloxycarbonyl, p- chlorobenzyloxycarbonyl, p-methoxybenzyloxycarbonyl, p-nitrobenzyloxycarbonyl, 2- nitrobenzyloxycarbonyl, p-bromobenzyloxycarbonyl, 3 ,4-dimethoxybenzyloxycarbonyl, 3,5-dimethoxybenzyloxycarbonyl, 2,4-dimethoxybenzyloxycarbonyl, 4- ethoxybenzyloxycarbonyl, 2-nitro-4,5-dimethoxybenzyloxycarbonyl, 3,4,5- trimethoxybenzyloxycarbonyl, 1 -(p-biphenylyl)- 1 -methylethoxycarbonyl, α,α-dimethyl- 3,5-dimethoxybenzyloxycarbonyl, benzhydryloxycarbonyl, t-butyoxycarbonyl, diisopropylmethoxycarbonyl, isopropyloxycarbonyl, ethoxycarbonyl, methoxycarbonyl, allyloxycarbonyl, 2,2,2, -trichloroethoxycarbonyl, phenoxycarbonyl, 4- nitrophenoxycarbonyl, fluorenyl-9-methoxycarbonyl, cyclopentyloxycarbonyl, adamantyloxycarbonyl, cyclohexyloxycarbonyl, phenylthiocarbonyl and the like; arylalkyl groups such as benzyl, triphenylmethyl, benzyloxymethyl, 9-fluorenylmethyloxycarbonyl (Fmoc) and the like and silyl groups such as trimethylsilyl and the like. Still other examples include theyl, succinyl, methoxysuccinyl, subery, adipyl, azelayl, dansyl, benzyloxycarbonyl, methoxyazelaly, methoxyadipyl, methoxysuberyl, and 2,4- dinitrophenyl.
The term "carboxy protecting group" or "C-terminal protecting group" refers to a carboxylic acid protecting ester or amide group employed to block or protect the carboxylic acid functionality while the reactions involving other functional sites of the compound are performed. Carboxy protecting groups are disclosed in Greene, Protective Groups in Organic Synthesis pp. 152-186 (1981), which is hereby incorporated by reference. Additionally, a carboxy protecting group may be used as a pro-drug whereby the carboxy protecting group can be readily cleaved in vivo, for example by enzymatic hydrolysis, to release the biologically active parent. Such carboxy protecting groups are well known to those skilled in the art, having been extensively used in the protection of carboxyl groups in the penicillin and cephalosporin fields as described in U.S. Pat. Nos. 3,840,556 and 3,719,667, the disclosures of which are hereby incorporated herein by reference. Representative carboxy protecting groups are Ci -Cs loweralkyl (e.g., methyl, ethyl or t- butyl and the like); arylalkyl such as phenethyl or benzyl and substituted derivatives thereof such as alkoxybenzyl or nitrobenzyl groups and the like; arylalkenyl such as phenylethenyl and the like; aryl and substituted derivatives thereofsuch as 5-indanyl and the like; dialkylaminoalkyl such as dimethylaminoethyl and the like); alkanoyloxyalkyl groups such as acetoxymethyl, butyryloxymethyl, valeryloxymethyl, isobutyryloxymethyl, isovaleryloxymethyl, l-(propionyloxy)-l -ethyl, l-(pivaloyloxyl)-l -ethyl, 1 -methyl- 1- (propionyloxy)-l -ethyl, pivaloyloxymethyl, propionyloxymethyl and the like; cycloalkanoyloxyalkyl groups such as cyclopropylcarbonyloxymethyl, cyclobutylcarbonyloxymethyl, cyclopentylcarbonyloxymethyl, cyclohexylcarbonyloxymethyl and the like; aroyloxyalkyl such as benzoyloxymethyl, benzoyloxyethyl and the like; arylalkylcarbonyloxyalkyl such as benzylcarbonyloxymethyl, 2-benzylcarbonyloxyethyl and the like; alkoxycarbonylalkyl or cycloalkyloxycarbonylalkyl such as methoxycarbonylmethyl, cyclohexyloxycarbonylmethyl, 1-methoxycarbonyl-l- ethyl and the like; alkoxycarbonyloxyalkyl or cycloalkyloxycarbonyloxyalkyl such as methoxycarbonyloxymethyl, t-butyloxycarbonyloxymethyl, 1-ethoxycarbonyloxy-l -ethyl, 1-cyclohexyloxycarbonyloxy-l -ethyl and the like; aryloxycarbonyloxyalkyl such as 2- (phenoxycarbonyloxy)ethyl, 2-(5-indanyloxycarbonyloxy)ethyl and the like; alkoxyalkylcarbonyloxyalkyl such as 2-(l-methoxy-2-methylpropan-2-oyloxy)ethyl and like; arylalkyloxycarbonyloxyalkyl such as 2-(benzyloxycarbonyloxy)ethyl and the like; arylalkenyloxycarbonyloxyalkyl such as 2-(3-phenylpropen-2-yloxycarbonyloxy)ethyl and the like; alkoxycarbonylaminoalkyl such as t-butyloxycarbonylaminomethyl and the like; alkylaminocarbonylaminoalkyl such as methylaminocarbonylaminomethyl and the like; alkanoylaminoalkyl such as acetylaminomethyl and the like; heterocycliccarbonyloxyalkyl such as 4-methylpiperazinylcarbonyloxymethyl and the like; dialkylaminocarbonylalkyl such as dimethylaminocarbonylmethyl, diethylaminocarbonylmethyl and the like; (5- (loweralkyl)-2-oxo-l,3-dioxolen-4-yl)alkyl such as (5-t-butyl-2-oxo-l,3-dioxolen-4- yl)methyl and the like; and (5-phenyl-2-oxo-l,3-dioxolen-4-yl)alkyl such as (5-phenyl-2- oxo-l,3-dioxolen-4-yl)methyl and the like. Representative amide carboxy protecting groups are aminocarbonyl and loweralkylaminocarbonyl groups. For example, aspartic acid may be protected at the α-C-terminal by an acid labile group (e.g., t-butyl) and protected at the β-C-terminal by a hydrogenation labile group (e.g., benzyl) then deprotected selectively during synthesis. As mentioned above, the protected carboxy group may also be a loweralkyl, cycloalkyl or arylalkyl ester, for example, methyl ester, ethyl ester, propyl ester, isopropyl ester, butyl ester, sec-butyl ester, isobutyl ester, amyl ester, isoamyl ester, octyl ester, cyclohexyl ester, phenylethyl ester and the like or an alkanoyloxyalkyl, cycloalkanoyloxyalkyl, aroyloxyalkyl or an arylalkylcarbonyloxyalkyl ester. As noted above, certain compounds of the present invention may exist in particular geometric or stereoisomeric forms. The present invention contemplates all such compounds, including cis- and trans-isomers, R- and S-enantiomers, diastereomers, (D)- isomers, (L)-isomers, the racemic mixtures thereof, and other mixtures thereof, as falling within the scope of the invention. Additional asymmetric carbon atoms may be present in a substituent such as an alkyl group. All such isomers, as well as mixtures thereof, are intended to be included in this invention.
If, for instance, a particular enantiomer of a compound of the present invention is desired, it may be prepared by asymmetric synthesis or by derivation with a chiral auxiliary, where the resulting diastereomeric mixture is separated and the auxiliary group cleaved to provide the pure desired enantiomer. Alternatively, where the molecule contains a basic functional group, such as amino, or an acidic functional group, such as carboxyl, diastereomeric salts are formed with an appropriate optically-active acid or base, followed by resolution of the diastereomers thus formed by fractional crystallization or chromatographic means well known in the art, and subsequent recovery of the pure enantiomer.
For purposes of this invention, the chemical elements are identified in accordance with the Periodic Table of the Elements, CAS version, Handbook of Chemistry and Physics, 67th ed., 1986-87, inside cover.
A compound is said to have an "insulinotropic activity" if it is able to stimulate, or cause the stimulation of, the synthesis or expression of the hormone insulin.
Another aspect of the present invention relates to pharmaceutical compositions of dipeptidylpeptidase inhibitors, particularly inhibitor(s) and their uses in treating and/or preventing disorders which can be improved by altering the homeostasis of peptide hormones. In an embodiment, the inhibitors have hypoglycemic and antidiabetic activities, and may be used in the treatment of disorders marked by aberrant glucose metabolism (including storage). In particular embodiments, the compositions of the subject methods are useful as insulinotropic agents, or to potentiate the insulinotropic effects of such molecules as GLP-I . In this regard, the present method can be useful for the treatment and/or prophylaxis of a variety of disorders, including one or more of: hyperlipemia, hyperglycemia, obesity, glucose tolerance insufficiency, insulin resistance, and diabetic complications.
For instance, in certain embodiments the method involves administration of an inhibitor(s), preferably at a predetermined interval(s) during a 24-hour period, in an amount effective to improve one or more aberrant indices associated with glucose metabolism disorders (e.g., glucose intolerance, insulin resistance, hyperglycemia, hyperinsulinemia, and Type II diabetes). The effective amount of the inhibitor may be about 0.01, 0.1, 1, 10, 30, 50, 70, 100, 150, 200, 500, or 1000 mg/kg of the subject. ///. Exemplary Embodiments (i). Compounds
Exemplary compounds are described below using various formulas. In each case, the variables are defined specifically for each individual formula. A definition of a variable for one formula should not be used to vary a definition provided for another formula, although a variable that has not been defined for one formula may be interpreted by analogy with a definition elsewhere for a similar formula.
In certain embodiments, a protease inhibitor of the invention is represented by
Formula (I):
Figure imgf000028_0001
or a pharmaceutically acceptable salt thereof; wherein:
W is selected from the group consisting of -CN, -CH=NR^ ,
Figure imgf000029_0001
; wherein
R4 represents H, alkyl, alkenyl, alkynyl, -C(X1)(X2)(X3), -(CH2)Hi-R43, -(CH2)Ii-OH, -(CH2)n-O-alkyl, -(CH2)n-O-alkenyl, -(CH2)n-O-alkynyl, -(CH2)H-O-(CH2)Hi-R43, -(CH2)n-SH, -(CH2)n-S-alkyl, -(CH2)n-S-alkenyl, -(CH2)n-S-alkynyl, -(CH2)H-S-(CH2)Hi-R43, -C(O)C(O)NH2, and -C(O)C(O)OR44;
R43 represents independently for each occurrence aryl, aralkyl, or non-aromatic hetereocyclyl;
R44 represents independently for each occurrence H, alkyl, alkenyl, aryl, aralkyl, or non-aromatic heterocyclyl; Yi and Y2 independently represent OH, or a group capable of being hydro lyzed to a hydroxyl group; or taken together with the boron to which they are bonded form a 5- membered to 8-membered ring comprising said boron and two oxygen atoms bonded to said boron;
R40 is O or S; R41 is selected from the group consisting of N3, SH, NH2, NO2, and -OK^;
R42 is selected from the group consisting of hydrogen, lower alkyl, amine, and -OR44, or a pharmaceutically acceptable salt; or R41 and R42 taken together with the phosphorous atom to which they are attached complete a heterocyclic ring having from 5 to 8 atoms in the ring structure; X1 is halogen;
X2 and X3 is hydrogen or halogen; m is zero or an integer in the range of 1 to 8; and n is an integer in the range of 1 to 8.
Ri is selected from a C-terminally linked amino acid residue or amino acid analog, a C-terminally linked peptide or peptide analog, an amino-protecting group, OO O u
R 51 A ^ r 1 R κ55A/ 5- , and R5 0-OIh/
R2 is absent or represents one or more substituents attached at positions 3, 4, or 5 of the dehydroproline ring, each of which is independently selected from the group consisting of hydrogen, halogen, hydroxyl, lower alkyl, lower alkenyl, lower alkynyl, lower alkoxy, lower hydroxyalkyl, lower alkoxyalkyl, carbonyl, carboxyl, ester, formate, ketone, thiocarbonyl, thioester, thioacetate, thioformate, amino, acylamino, amido, nitro, sulfate, sulfonate, sulfonamido, -(CH2Xn-R51, -(CH2Xn-OH, -(CH2)m-O-lower alkyl, -(CH2Xn-O- lower alkenyl, -(CH2)n-O-(CH2)m-R51, -(CH2Xn-SH, -(CH2)m-S-lower alkyl, -(CH2)m-S-lower alkenyl, or -(CH2X1-S-(CH2Xn-R51' azido, cyano, isocyanato, thiocyanato,
isothiocyanato, cyanato, Nz^=C, and C===C R22;
R3 is selected from the group consisting of hydrogen, halogen, lower alkyl, lower alkenyl, lower alkynyl, carbonyl, thiocarbonyl, amino, acylamino, amido, nitro, sulfate, sulfonate, a sulfonamido, -(CH2Xn-R51, -(CH2Xn-OH, -(CH2)m-O-lower alkyl, -(CH2Xn-O- lower alkenyl, -(CH2)n-O-(CH2)m-R51, -(CH2Xn-SH, -(CH2)m-S-lower alkyl, -(CH2Xn-S- lower alkenyl, or -(CH2)n-S-(CH2)m-R51, azido, cyano, isocyanato, thiocyanato,
isothiocyanato, cyanato, Nmzz^C, and C C ^22;
R5 is selected from the group consisting of hydrogen, halogen, alkyl, alkenyl, alkynyl, aryl, -(CH2)m-R51, -(CH2)m-0H, -(CH2)m-O-alkyl, -(CH2)m-O-alkenyl, -(CH2)m-O-alkynyl, -(CH2)m-O-(CH2)m-R51, -(CH2)m-SH, -(CH2)m-S-alkyl, -(CH2)m-S- alkenyl, -(CH2)m-S-alkynyl, -(CH2)m-S-(CH2)m-R51,
(CH2)m— O-R51 ,
Figure imgf000030_0001
0 0 0 O
Il Il Il Il
— (CH2)n— C— alkyl , — (CH2)n— C— alkenyl , — (CH2)n— C— alkynyl , and — (CH2)n— C-(CH2)m-R51; wherein
R51 represents for each occurrence aryl, aralkyl, or non-aromatic heterocyclyl; R52 and R53 independently represent hydrogen, alkyl, alkenyl, -(CH2)m-R5i, -C(=O)-alkyl, -C(=O)-alkenyl, -C(=O)-alkynyl, or -C(=O)-(CH2)m-R5i; or
R52 and R53 taken together with the N atom to which they are attached complete a heterocyclic ring having from 4 to 8 atoms in the ring structure; m is zero or an integer in the range of 1 to 8; and n is an integer in the range of 1 to 8.
In certain embodiments, W is a group of formula -B(Yi)(Y2).
In certain embodiments, such as when W has one of the values described above, Ri is a C-terminally linked amino acid residue or amino acid analog or a C-terminally linked peptide or peptide analog.
In certain embodiments, such as when W and Ri have the values described above, R3 is hydrogen or a lower alkyl group.
In certain embodiments, the compound is represented by:
Figure imgf000031_0001
or pharmaceutically acceptable salts thereof.
Another aspect of the invention provides a protease inhibitor having a structure represented by Formula (II):
Figure imgf000031_0002
(II) or a pharmaceutically acceptable salt thereof; wherein:
R2 is absent or represents one or more substituents attached at positions 3, 4, or 5 of the dehydroproline ring, each of which is independently selected from the group consisting of hydrogen, halogen, hydroxyl, lower alkyl, lower alkenyl, lower alkynyl, lower alkoxy, lower hydroxyalkyl, lower alkoxyalkyl, carbonyl, carboxyl, ester, formate, ketone, thiocarbonyl, thioester, thioacetate, thioformate, amino, acylamino, amido, nitro, sulfate, sulfonate, sulfonamido, -(CH2)m-R43, -(CH2)m-OH, -(CH2)m-O-lower alkyl, -(CH2)m-O- lower alkenyl, -(CH2)n-O-(CH2)m-R43, -(CH2)m-SH, -(CH2)m-S-lower alkyl, -(CH2)m-S-lower alkenyl, or -(CH2)n-S-(CH2)m-R43' azido, cyano, isocyanato, thiocyanato,
isothiocyanato, cyanato, Nz^=C, and C===C R22;
Rioi is selected from the group consisting of H, alkyl, alkoxy, alkenyl, alkynyl, amino, alkylamino, acylamino, cyano, sulfonylamino, acyloxy, aryl, non-aromatic heterocyclyl, heteroaryl, and a polypeptide chain of 1 to 8 amino acid residues;
Rio2 is selected from the group consisting of H, lower alkyl, and aralkyl;
Rio3 is selected from the group consisting of H, halogen, and alkyl, or R102 and R103 together with the atoms to which they are attached, form a 3- to 6-membered heterocyclic ring; R105 is selected from the group consisting of H, halogen, lower alkyl, and aralkyl;
R106 is selected from the group consisting of -CN, -CH=NR4,
Figure imgf000032_0001
; wherein
R4 represents H, alkyl, alkenyl, alkynyl, -C(X1)(X2)(X3), -(CH2)Di-R43, -(CH2)Ii-OH, -(CH2)n-O-alkyl, -(CH2)n-O-alkenyl, -(CH2)n-O-alkynyl, -(CH2)n-O-(CH2)m-R43, -(CH2)n-SH, -(CH2)n-S-alkyl, -(CH2)n-S-alkenyl,
-(CH2)n-S-alkynyl, -(CH2)n-S-(CH2)m-R43, -C(O)C(O)NH2, or -C(O)C(O)OR44;
R43 represents independently for each occurrence aryl, aralkyl, or non-aromatic hetereocyclyl;
R44 independently for each occurrence is selected from the group consisting of H, alkyl, alkenyl, aryl, aralkyl, and non-aromatic heterocyclyl;
Yi and Y2 independently represent OH, or a group capable of being hydro lyzed to a hydroxyl group; or taken together with the boron to which they are bonded form a 5- membered to 8-membered ring comprising said boron and two oxygen atoms bonded to said boron; R40 is O or S;
R41 is selected from the group consisting of N3, SH, NH2, NO2 and -OR^; R42 is selected from the group consisting of H, lower alkyl, amine, and -OR44, or a pharmaceutically acceptable salt; or R41 and R42 taken together with the phosphorous atom to which they are attached complete a heterocyclic ring having from 5 to 8 atoms in the ring structure; Xi is halogen;
X2 and X3 is hydrogen or halogen; and n is an integer in the range of 2 to 6;
R107 is selected from the group consisting of H, aryl, alkyl, aralkyl, non-aromatic heterocyclyl, heteroaryl, heteroaralkyl, and polypeptide chains of 1 to 8 amino acid residues;
L is absent or is selected from the group consisting of alkyl, alkenyl, alkynyl, -(CH2)mO(CH2)m-, -(CH2)mNR2(CH2)m-, and -(CH2)mS(CH2)m-;
X is absent or is selected from the group consisting of -N(R107)-, -O-, and -S-; Y is absent or is selected from the group consisting of -C(=O)-, -C(=S)-, and -SO2-; and m is, independently for each occurrence, an integer from 0 to 10. In particular embodiments, Rioβ is a group of formula -B(Yi)(Y2). In certain embodiments, such as when Rioβ has one of the values described above, R103 is selected from -H, -CH3, -CH(CH3)2, -CH2CH(CH3)2, -C(CH3)(CH2CH3), -CH2CH2SCH3, -CH2-phenyl, -CH2-(3-indolyl), -CH2OH, -CH(OH)CH3, -CH2CONH2, -CH2CH2CONH2, -CH2-(4-hydroxyphenyl), -CH2SH, -(CH2)4NH2, -(CH2)3NH-C(=NH)NH2, -CH-(5-imidazolyl), -CH2COOH, and -CH2CH2COOH, particularly -H, -CH3 and -(CH2)3NH-C(=NH)NH2.
In certain embodiments, such as when Rioβ and/or Rio3 have the values described above, one or more of L, X and Y is absent. Preferably, two or more of L, X and Y are absent. In other embodiments, each of L, X and Y is absent. When one or more of L, X and Y is absent, R1O1 is generally -H.
In many embodiments, one or both of Rio3 and R105 are -H in compound represented by Formula (II). More typically, both Ri03 and R105 are -H. Exemplary compounds represented by Formula (II) are:
Figure imgf000034_0001
or pharmaceutically acceptable salts thereof.
More exemplary compounds represented by Formula (II) are:
Figure imgf000034_0002
or pharmaceutically acceptable salts thereof.
In certain embodiments where X, Y, and L are absent, Ri is a polypeptide chain of 2 to 8 amino acid residues, where proline is the residue that is directly attached to the leftmost residue of Formula (II). In certain such embodiments, Ri is a polypeptide chain of 2 amino acid residues, where proline is the residue that is directly attached to the leftmost nitrogen of Formula (II).
The dehydroproline ring in Formulas (I) and (II) can be substituted with one or more groups R2. In certain embodiments, R2 is selected from hydroxyl, lower alkyl (e.g., methyl), lower alkenyl, lower alkynyl, lower alkoxy, lower hydroxyalkyl (e.g., hydroxymethyl), and lower alkoxyalkyl. Preferably, the dehydroproline substituent group is selected from lower alkyl, lower hydroxyalkyl and lower alkoxyalkyl. In certain embodiments, R2 is located at the 5 -position of the ring, particularly when the substituent is hydroxyl. In certain embodiments, the subject inhibitors are DPIV inhibitors with a K1 for
DPIV inhibition of 10 nM or less, 1.0 nM or less, 0.1 or 0.01 nM or less. Indeed, inhibitors with K1 values in the picomolar and even femtomolar range are contemplated.
In general, the inhibitors of the subject method are small molecules, e.g., with molecular weights less than 7500 amu, less than 5000 amu, less than 2000 amu, and less than 1000 amu. In certain embodiments, the inhibitors are orally active.
Another aspect of the present invention relates to pharmaceutical compositions of the dipeptidylpeptidase inhibitors disclosed herein, and their uses in treating and/or preventing (inhibiting) disorders which can be improved by altering the homeostasis of peptide hormones. In an embodiment, the inhibitors have hypoglycemic and antidiabetic activities, and may be used in the treatment of disorders marked by aberrant glucose metabolism (including storage). In particular embodiments, the compositions of the subject methods are useful as insulinotropic agents, or to potentiate the insulinotropic effects of such molecules as GLP-I . In this regard, the present method may be useful for the treatment and/or prophylaxis of a variety of disorders, including: hyperlipemia, hyperglycemia, obesity, glucose tolerance insufficiency, insulin resistance, and diabetic complications.
For instance, in certain embodiments the method involves administration of an inhibitor(s), preferably at a predetermined interval(s) during a 24-hour period, in an amount effective to improve one or more aberrant indices associated with glucose metabolism disorders (e.g., glucose intolerance, insulin resistance, hyperglycemia, hyperinsulinemia, and Type II diabetes). The effective amount of the inhibitor may be about 0.01, 0.1, 1, 10, 30, 50, 70, 100, 150, 200, 500, or 1000 mg/kg of the subject. (U) Agonism of GLP-I effects The compounds useful in the subject methods possess, in certain embodiments, the ability to lower blood glucose levels, to relieve obesity, to alleviate impaired glucose tolerance, to inhibit hepatic glucose neogenesis, and to lower blood lipid levels and to inhibit aldose reductase. They are thus useful for the prevention and/or therapy of hyperglycemia, obesity, hyperlipidemia, diabetic complications (including retinopathy, nephropathy, neuropathy, cataracts, coronary artery disease and arteriosclerosis), and furthermore for obesity-related hypertension and osteoporosis.
Diabetes mellitus is a disease characterized by hyperglycemia caused by from a decrease in insulin secretion, decreased insulin sensitivity, and/or insulin resistance. The morbidity and mortality of this disease result from vascular, renal, and neurological complications. An oral glucose tolerance test is a clinical test used to diagnose diabetes. In an oral glucose tolerance test, a patient's physiological response to a glucose load or challenge is evaluated. After ingesting the glucose, the patient's physiological response to the glucose challenge is evaluated. Generally, this is accomplished by determining the patient's blood glucose levels (the concentration of glucose in the patient's plasma, serum, or whole blood) at several predetermined points in time.
In one embodiment, the present invention provides a method for agonizing the action of GLP-I. It has been determined that isoforms of GLP-I (GLP- 1(7-37) and GLP- 1(7-36)), which are derived from preproglucagon in the intestine and the hind brain, have insulinotropic activity; i.e., they modulate glucose metabolism. DPIV cleaves the isoforms to inactive peptides. Thus, in certain embodiments, compound(s) of the present invention can agonize insulinotropic activity by interfering with the degradation of bioactive GLP-I peptides. (Hi) Agonism of the effects of other peptide hormones In another embodiment, the subject agents may be used to agonize (e.g., mimic or potentiate) the activity of peptide hormones, e.g., GLP-2, GIP and NPY.
To illustrate further, the present invention provides a method for agonizing the action of GLP-2. It has been determined that GLP-2 acts as a trophic agent, to promote growth of gastrointestinal tissue. The effect of GLP-2 is marked particularly by increased growth of the small bowel, and is therefore herein referred to as an "intestinotrophic" effect. DPIV is known to cleave GLP-2 into a biologically inactive peptide. Thus, in one embodiment, inhibition of DPIV interferes with the degradation of GLP-2, and thereby increases the plasma half- life of that hormone. In still other embodiments, the subject method may be used to increase the half- life of other proglucagon-derived peptides, such as glicentin, oxyntomodulin, glicentin-related pancreatic polypeptide (GRPP), and/or intervening peptide -2 (IP-2). For example, glicentin has been demonstrated to cause proliferation of intestinal mucosa and also inhibits a peristalsis of the stomach, and has thus been elucidated as useful as a therapeutic agent for digestive tract diseases.
Thus, in one aspect, the present invention relates to therapeutic and related uses of compound(s) for promoting the growth and proliferation of gastrointestinal tissue, particularly small bowel tissue. For instance, the subject method may be used as part of a regimen for treating injury, inflammation, or resection of intestinal tissue, e.g., where enhanced growth and repair of the intestinal mucosal epithelial is desired.
With respect to small bowel tissue, such growth is measured conveniently as an increase in small bowel mass and length, relative to an untreated control. The effect of compounds on small bowel tissue also manifests as an increase in the height of the crypt plus villus axis. Such activity is referred to herein as an "intestinotrophic" activity. The efficacy of the subject method may also be detectable as an increase in crypt cell proliferation and/or a decrease in small bowel epithelium apoptosis. These cellular effects may be noted most significantly in relation to the jejunum, including the distal jejunum and particularly the proximal jejunum, and also in the distal ileum. A compound is considered to have "intestinotrophic effect" if a test animal exhibits increased small bowel weight, increased height of the crypt plus villus axis or increased crypt cell proliferation, or decreased small bowel epithelium apoptosis when treated with the compound (or genetically engineered to express it themselves). A model suitable for determining such gastrointestinal growth is described by US Patent 5,834,428 (incorporated by reference). In general, patients who would benefit from either increased small intestinal mass and consequent increased small bowel mucosal function are candidates for treatment by the subject method. Particular conditions that may be treated include the various forms of sprue, including celiac sprue which results from a toxic reaction to α-gliadin from wheat, and is marked by a tremendous loss of villae of the bowel; tropical sprue which results from infection and is marked by partial flattening of the villae; hypogammaglobulinemic sprue which is observed in patients with common variable immunodeficiency or hypogammaglobulinemia and is marked by significant decrease in villus height. The therapeutic efficacy of the treatment may be monitored by enteric biopsy to examine the villus morphology, by biochemical assessment of nutrient absorption, by patient weight gain, or by amelioration of the symptoms associated with these conditions. Other conditions that may be treated by the subject method, or for which the subject method may be useful prophylactically, include radiation enteritis, infectious or post-infectious enteritis, regional enteritis (Crohn's disease), small intestinal damage due to toxic or other chemotherapeutic agents, and patients with short bowel syndrome.
More generally, the present invention provides a therapeutic method for treating digestive tract diseases. The term "digestive tract" as used herein means a organismic tube through which food passes, including stomach and intestine. The term "digestive tract diseases" as used herein means diseases accompanied by a qualitative or quantitative abnormality in the digestive tract mucosa, which include ulceric or inflammatory disease; congenital or acquired digestion and absorption disorder including malabsorption syndrome; disease caused by loss of a mucosal barrier function of the gut; and protein- losing gastroenteropathy. The ulceric disease includes gastric ulcer, duodenal ulcer, small intestinal ulcer, colonic ulcer, and rectal ulcer. The inflammatory disease includes esophagitis, gastritis, duodenitis, enteritis, colitis, Crohn's disease, proctitis, gastrointestinal Behcet, radiation enteritis, radiation colitis, radiation proctitis, enteritis, and medicamentosa. The malabsorption syndrome includes the essential malabsorption syndrome, such as disaccharide-decomposing enzyme deficiency, glucose-galactose malabsorption, fructose malabsorption; secondary malabsorption syndrome, e.g., the disorder caused by a mucosal atrophy in the digestive tract through the intravenous or parenteral nutrition or elemental diet, the disease caused by the resection and shunt of the small intestine such as short gut syndrome, cul-de-sac syndrome; and indigestible malabsorption syndrome, such as the disease caused by resection of the stomach, e.g., dumping syndrome.
The term "therapeutic agent for digestive tract diseases" as used herein means the agents for the prevention and treatment of the digestive tract diseases, which include the therapeutic agent for digestive tract ulcer, the therapeutic agent for inflammatory digestive tract disease, the therapeutic agent for mucosal atrophy in the digestive tract, the therapeutic agent for a digestive tract wound, the amelioration agent for the function of the digestive tract including the agent for recovery of the mucosal barrier function, and the amelioration agent for digestive and absorptive function. Ulcers include digestive ulcers and erosions, and acute ulcers, namely acute mucosal lesions. The subject method, because of promoting proliferation of intestinal mucosa, may be used in the treatment and prevention of pathologic conditions of insufficiency in digestion and absorption, that is, treatment and prevention of mucosal atrophy, or treatment of hypoplasia of the digestive tract tissues and decrease in these tissues by surgical removal as well as improvement of digestion and absorption. Further, the subject method may be used in the treatment of pathologic mucosal conditions due to inflammatory diseases, such as enteritis, Crohn's disease, and ulceric colitis and also in the treatment of reduction in function of the digestive tract after operation, for example, in damping syndrome as well as in the treatment of duodenal ulcer in conjunction with the inhibition of peristalsis of the stomach and rapid migration of food from the stomach to the jejunum. Furthermore, glicentin can effectively be used in promoting cure of surgical invasion as well as in improving functions of the digestive tract. Thus, the present invention also provides a therapeutic agent for atrophy of the digestive tract mucosa, a therapeutic agent for wounds in the digestive tract and a drug for improving functions of the digestive tract which comprise glicentin as active ingredients.
Likewise, the compound(s) of the subject invention may be used to alter the plasma half-life of secretin, VIP, PHI, PACAP, GIP, and/or helodermin. Additionally, the subject method may be used to alter the pharmacokinetics of Peptide YY and neuropeptide Y, both members of the pancreatic polypeptide family, as DPIV has been implicated in the processing of those peptides in a manner which alters receptor selectivity.
Neuropeptide Y (NPY) is believed to act in the regulation vascular smooth muscle tone, as well as regulation of blood pressure. NPY also decreases cardiac contractility. NPY is also the most powerful appetite stimulant known (Wilding et al., J. Endocrinology 1992, 132, 299-302). The centrally evoked food intake (appetite stimulation) effect is predominantly mediated by NPY Yl receptors and causes increase in body fat stores and obesity (Stanley et al., Physiology and Behavior 1989, 46, 173-177).
According to the present invention, a method for treatment of anorexia comprises administering to a subject in need thereof a therapeutically effective amount of a compound(s) of the present invention, to stimulate the appetite and increase body fat stores. A method for treatment of hypotension comprises administering to a subject in need thereof a therapeutically effective amount of a compound(s) of the present invention to mediate vasoconstriction and increase blood pressure. DPIV has also been implicated in the metabolism and inactivation of growth hormone-releasing factor (GHRF). GHRF is a member of the family of homologous peptides that includes glucagon, secretin, vasoactive intestinal peptide (VIP), peptide histidine isoleucine (PHI), pituitary adenylate cyclase activating peptide (PACAP), gastric inhibitory peptide (GIP) and helodermin (Kubiak et al. Peptide Res. 1994, 7, 153). GHRF is secreted by the hypothalamus, and stimulates the release of growth hormone (GH) from the anterior pituitary. Thus, the subject method may be used to improve clinical therapy for certain growth hormone deficient children, and in clinical therapy of adults to improve nutrition and to alter body composition (muscle vs. fat). The subject method can also be used in veterinary practice, for example, to develop higher yield milk production and higher yield, leaner livestock. (iv) Assays of Insulinotropic Activity
In selecting a compound suitable for use in the subject method, it is noted that the insulinotropic property of a compound may be determined by providing that compound to animal cells, or injecting that compound into animals and monitoring the release of immunoreactive insulin (IRI) into the media or circulatory system of the animal, respectively. The presence of IRI can be detected through the use of a radioimmunoassay which can specifically detect insulin.
The db/db mouse is a genetically obese and diabetic strain of mouse. The db/db mouse develops hyperglycemia and hyperinsulinemia concomitant with its development of obesity and thus serves as a model of obese type 2 diabetes (NIDDM). The db/db mice can be purchased from, for example, The Jackson Laboratories (Bar Harbor, Me.). In an exemplary embodiment, for treatment of the mice with a regimen including a compound(s) or control, sub-orbital sinus blood samples are taken before and at some time (e.g., 60 min) after dosing of each animal. Blood glucose measurements can be made by any of several conventional techniques, such as using a glucose meter. The blood glucose levels of the control and compound(s) dosed animals are compared.
The metabolic fate of exogenous GLP-I can also be followed in both nondiabetic and type II diabetic subjects, and the effect of a candidate compound(s) determined. For instance, a combination of high-pressure liquid chromatography (HPLC), specific radioimmunoassays (RIAs), and an enzyme-linked immunosorbent assay (ELISA), can be used, whereby intact biologically active GLP-I and its metabolites can be detected. See, for example, Deacon et al. Diabetes, 1995, 44, 1126-1131. To illustrate, after GLP-I administration, the intact peptide can be measured using an NH2-terminally directed RIA or ELISA, while the difference in concentration between these assays and a COOH-terminal- specifϊc RIA allowed determination of NH2-terminally truncated metabolites. Without compound, subcutaneous GLP-I is rapidly degraded in a time-dependent manner, forming a metabolite which co-elutes on HPLC with GLP- 1(9-36) amide and has the same immunoreactive profile. For instance, 30 min after subcutaneous GLP-I administration to diabetic patients (n = 8), the metabolite accounted for 88.5 + 1.9% of the increase in plasma immunoreactivity determined by the COOH-terminal RIA, which was higher than the levels measured in healthy subjects (78.4 + 3.2%; n = 8; P < 0.05). See Deacon et al., supra. Intravenously infused GLP-I was also extensively degraded. (v) Conjoint administration
Another aspect of the invention provides a conjoint therapy wherein one or more other therapeutic agents are administered with the compound. Such conjoint treatment may be achieved by way of the simultaneous, sequential, or separate dosing of the individual components of the treatment.
In one embodiment, a compound(s) of the present invention is conjointly administered with insulin or other insulinotropic agents, such as GLP-I, peptide hormones, such as GLP-2, GIP, or NPY, or a gene therapy vector which causes the ectopic expression of said agents and peptide hormones. In certain embodiments, said agents or peptide hormones may be variants or derivatives of a naturally occurring or synthetic peptide hormone, wherein one or more amino acids have been added, deleted, or substituted.
In another illustrative embodiment, the compounds of the present invention can be conjointly administered with an Ml receptor antagonist. Cholinergic agents are potent modulators of insulin release that act via muscarinic receptors. Moreover, the use of such agents can have the added benefit of decreasing cholesterol levels, while increasing HDL levels. Suitable muscarinic receptor antagonists include substances that directly or indirectly block activation of muscarinic cholinergic receptors. Preferably, such substances are selective (or are used in amounts that promote such selectivity) for the Ml receptor. Non-limiting examples include quaternary amines (such as methantheline, ipratropium, and propantheline), tertiary amines (e.g., dicyclomine and scopolamine), and tricyclic amines (e.g., telenzepine). Other suitable muscarinic receptor antagonists include benztropine (commercially available as COGENTIN from Merck), hexahydro-sila-difenidol hydrochloride (HHSID hydrochloride disclosed in Lambrecht et al. Trends in Pharmacol. Sci. 1989, iO(Suppl), 60; (+/-)-3-quinuclidinyl xanthene-9-carboxylate hemioxalate (QNX- hemioxalate; Birdsall et al., Trends in Pharmacol. Sci. 1983, 4, 459; telenzepine dihydrochloride (Coruzzi et al. Arch. Int. Pharmacodyn. Ther. 1989, 302, 232; and Kawashima et al. Gen. Pharmacol. 1990, 21, 17), and atropine. The dosages of such muscarinic receptor antagonists will be generally subject to optimization as outlined above. In the case of lipid metabolism disorders, dosage optimization may be necessary independent of whether administration is timed by reference to the lipid metabolism responsiveness window or not.
In terms of regulating insulin and lipid metabolism and reducing the foregoing disorders, the compound(s) of the present invention may also act synergistically with prolactin inhibitors such as d2 dopamine agonists (e.g., bromocriptine). Accordingly, the subject method can include the conjoint administration of such prolactin inhibitors as prolactin-inhibiting ergo alkaloids and prolactin-inhibiting dopamine agonists. Examples of suitable compounds include 2-bromo-alpha-ergocriptine, 6-methyl-8-beta- carbobenzyloxyaminoethyl-10-alpha-ergoline, 8-acylaminoergo lines, 6-methyl-8-alpha-(N- acyl)amino-9-ergoline, 6-methyl-8-alpha-(N-phenylacetyl)amino-9-ergoline, ergocornine, 9,10-dihydroergocornine, D-2-halo-6-alkyl-8-substituted ergo lines, D-2-bromo-6-methyl-8- cyanomethylergoline, carbidopa, benserazide, and other dopadecarboxylase inhibitors, L- dopa, dopamine, and non toxic salts thereof. The compound(s) of the present invention used according to the invention can also be used conjointly with agents acting on the ATP-dependent potassium channel of the β- cells, such as glibenclamide, glipizide, gliclazide, and AG-EE 623 ZW. The compound(s) may also advantageously be applied in combination with other oral agents, such as metformin and related compounds or glucosidase inhibitors, such as acarbose. (vi) Pharmaceutical Compositions
While it is possible for a compound of the present invention to be administered alone, in certain cases it is preferable to administer the compound as a pharmaceutical formulation (composition). Protease inhibitors according to the invention may be formulated for administration in any convenient way for use in human or veterinary medicine. In certain embodiments, the compound included in the pharmaceutical preparation may be active itself, or may be a prodrug, e.g., capable of being converted to an active compound in a physiological setting. Compounds prepared as described herein can be administered in various forms, depending on the disorder to be treated and the age, condition, and body weight of the patient, as is well known in the art. For example, where the compounds are to be administered orally, they may be formulated as tablets, capsules, granules, powders, or syrups; or for parenteral administration, they may be formulated as injections (intravenous, intramuscular, or subcutaneous), drop infusion preparations, or suppositories. For application by the ophthalmic mucous membrane route, they may be formulated as eye drops or eye ointments. These formulations can be prepared by conventional means, and, if desired, the active ingredient may be mixed with any conventional additive, such as an excipient, a binder, a disintegrating agent, a lubricant, a corrigent, a solubilizing agent, a suspension aid, an emulsifying agent, or a coating agent. Although the dosage will vary depending on the symptoms, age and body weight of the patient, the nature and severity of the disorder to be treated or prevented, the route of administration and the form of the drug, in general, a daily dosage of from 0.01 to 2000 mg of the compound is recommended for an adult human patient, and this may be administered in a single dose or in divided doses.
The precise time of administration and/or amount of the compound that will yield the most effective results in terms of efficacy of treatment in a given patient will depend upon the activity, pharmacokinetics, and bioavailability of a particular compound, physiological condition of the patient (including age, sex, disease type and stage, general physical condition, responsiveness to a given dosage, and type of medication), route of administration, etc. However, the above guidelines may be used as the basis for fine-tuning the treatment, e.g., determining the optimum time and/or amount of administration, which will require no more than routine experimentation consisting of monitoring the subject and adjusting the dosage and/or timing. The phrase "pharmaceutically acceptable" is employed herein to refer to those ligands, materials, compositions, and/or dosage forms which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of human beings and animals without excessive toxicity, irritation, allergic response, or other problem or complication, commensurate with a reasonable benefit/risk ratio. The phrase "pharmaceutically acceptable carrier" as used herein means a pharmaceutically acceptable material, composition or vehicle, such as a liquid or solid filler, diluent, excipient, solvent or encapsulating material. Each carrier must be "acceptable" in the sense of being compatible with the other ingredients of the formulation and not injurious to the patient. Some examples of materials which can serve as pharmaceutically acceptable carriers include: (1) sugars, such as lactose, glucose, and sucrose; (2) starches, such as corn starch and potato starch; (3) cellulose, and its derivatives, such as sodium carboxymethyl cellulose, ethyl cellulose, and cellulose acetate; (4) powdered tragacanth; (5) malt; (6) gelatin; (7) talc; (8) excipients, such as cocoa butter and suppository waxes; (9) oils, such as peanut oil, cottonseed oil, safflower oil, sesame oil, olive oil, corn oil, and soybean oil; (10) glycols, such as propylene glycol; (11) polyols, such as glycerin, sorbitol, mannitol, and polyethylene glycol; (12) esters, such as ethyl oleate and ethyl laurate; (13) agar; (14) buffering agents, such as magnesium hydroxide and aluminum hydroxide; (15) alginic acid; (16) pyrogen-free water; (17) isotonic saline; (18) Ringer's solution; (19) ethyl alcohol; (20) phosphate buffer solutions; and (21) other nontoxic compatible substances employed in pharmaceutical formulations. In certain embodiments, pharmaceutical compositions of the present invention are non-pyrogenic, i.e., do not induce significant temperature elevations when administered to a patient. The term "pharmaceutically acceptable salts" refers to the relatively non-toxic, inorganic and organic acid addition salts of the compound(s). These salts can be prepared in situ during the final isolation and purification of the compound(s), or by separately reacting a purified compound(s) in its free base form with a suitable organic or inorganic acid, and isolating the salt thus formed. Representative salts include the hydrobromide, hydrochloride, sulfate, bisulfate, phosphate, nitrate, acetate, valerate, oleate, palmitate, stearate, laurate, benzoate, lactate, phosphate, tosylate, citrate, maleate, fumarate, succinate, tartrate, naphthylate, mesylate, glucoheptonate, lactobionate, and laurylsulphonate salts, and the like. (See, e.g., Berge et al. J. Pharm. Sci. 1977, 66, 1-19)
In other cases, the compounds useful in the methods of the present invention may contain one or more acidic functional groups and, thus, are capable of forming pharmaceutically acceptable salts with pharmaceutically acceptable bases. The term "pharmaceutically acceptable salts" in these instances refers to the relatively non-toxic inorganic and organic base addition salts of an compound(s). These salts can likewise be prepared in situ during the final isolation and purification of the compound(s), or by separately reacting the purified compound(s) in its free acid form with a suitable base, such as the hydroxide, carbonate, or bicarbonate of a pharmaceutically acceptable metal cation, with ammonia, or with a pharmaceutically acceptable organic primary, secondary, or tertiary amine. Representative alkali or alkaline earth salts include the lithium, sodium, potassium, calcium, magnesium, and aluminum salts, and the like. Representative organic amines useful for the formation of base addition salts include ethylamine, diethylamine, ethylenediamine, ethanolamine, diethanolamine, piperazine, and the like (see, e.g., Berge et al., supra). Wetting agents, emulsifϊers, and lubricants, such as sodium lauryl sulfate and magnesium stearate, as well as coloring agents, release agents, coating agents, sweetening, flavoring, and perfuming agents, preservatives and antioxidants can also be present in the compositions.
Examples of pharmaceutically acceptable antioxidants include: (1) water soluble antioxidants, such as ascorbic acid, cysteine hydrochloride, sodium bisulfate, sodium metabisulfite, sodium sulfite, and the like; (2) oil-soluble antioxidants, such as ascorbyl palmitate, butylated hydroxyanisole (BHA), butylated hydroxytoluene (BHT), lecithin, propyl gallate, alpha-tocopherol, and the like; and (3) metal chelating agents, such as citric acid, ethylenediamine tetraacetic acid (EDTA), sorbitol, tartaric acid, phosphoric acid, and the like.
Formulations useful in the methods of the present invention include those suitable for oral, nasal, topical (including buccal and sublingual), rectal, vaginal, aerosol, and/or parenteral administration. The formulations may conveniently be presented in unit dosage form and may be prepared by any methods well known in the art of pharmacy. The amount of active ingredient which can be combined with a carrier material to produce a single dosage form will vary depending upon the host being treated and the particular mode of administration. The amount of active ingredient which can be combined with a carrier material to produce a single dosage form will generally be that amount of the compound which produces a therapeutic effect. Generally, out of 100%, this amount will range from about 1% to about 99% of active ingredient, preferably from about 5% to about 70%, most preferably from about 10% to about 30%.
Methods of preparing these formulations or compositions include the step of bringing into association a compound(s) with the carrier and, optionally, one or more accessory ingredients. In general, the formulations are prepared by uniformly and intimately bringing into association a ligand with liquid carriers, or finely divided solid carriers, or both, and then, if necessary, shaping the product.
Formulations suitable for oral administration may be in the form of capsules, cachets, pills, tablets, lozenges (using a flavored basis, usually sucrose and acacia or tragacanth), powders, granules, or as a solution or a suspension in an aqueous or nonaqueous liquid, or as an oil-in-water or water-in-oil liquid emulsion, or as an elixir or syrup, or as pastilles (using an inert base, such as gelatin and glycerin, or sucrose and acacia) and/or as mouthwashes, and the like, each containing a predetermined amount of a compound(s) as an active ingredient. A compound may also be administered as a bolus, electuary or paste.
In solid dosage forms for oral administration (capsules, tablets, pills, dragees, powders, granules, and the like), the active ingredient is mixed with one or more pharmaceutically acceptable carriers, such as sodium citrate or dicalcium phosphate, and/or any of the following: (1) fillers or extenders, such as starches, lactose, sucrose, glucose, mannitol, and/or silicic acid; (2) binders, such as, for example, carboxymethylcellulose, alginates, gelatin, polyvinyl pyrrolidone, sucrose, and/or acacia; (3) humectants, such as glycerol; (4) disintegrating agents, such as agar-agar, calcium carbonate, potato or tapioca starch, alginic acid, certain silicates, and sodium carbonate; (5) solution retarding agents, such as paraffin; (6) absorption accelerators, such as quaternary ammonium compounds; (7) wetting agents, such as, for example, acetyl alcohol and glycerol monostearate; (8) absorbents, such as kaolin and bentonite clay; (9) lubricants, such a talc, calcium stearate, magnesium stearate, solid polyethylene glycols, sodium lauryl sulfate, and mixtures thereof; and (10) coloring agents. In the case of capsules, tablets, and pills, the pharmaceutical compositions may also comprise buffering agents. Solid compositions of a similar type may also be employed as fillers in soft and hard- filled gelatin capsules using such excipients as lactose or milk sugars, as well as high molecular weight polyethylene glycols, and the like.
A tablet may be made by compression or molding, optionally with one or more accessory ingredients. Compressed tablets may be prepared using binder (for example, gelatin or hydroxypropylmethyl cellulose), lubricant, inert diluent, preservative, disintegrant (for example, sodium starch glycolate or cross-linked sodium carboxymethyl cellulose), surface-active or dispersing agent. Molded tablets may be made by molding in a suitable machine a mixture of the powdered peptide or peptidomimetic moistened with an inert liquid diluent.
Tablets, and other solid dosage forms, such as dragees, capsules, pills, and granules, may optionally be scored or prepared with coatings and shells, such as enteric coatings and other coatings well known in the pharmaceutical-formulating art. They may also be formulated so as to provide slow or controlled release of the active ingredient therein using, for example, hydroxypropylmethyl cellulose in varying proportions to provide the desired release profile, other polymer matrices, liposomes, and/or microspheres. They may be sterilized by, for example, filtration through a bacteria-retaining filter, or by incorporating sterilizing agents in the form of sterile solid compositions which can be dissolved in sterile water, or some other sterile injectable medium immediately before use. These compositions may also optionally contain opacifying agents and may be of a composition that they release the active ingredient(s) only, or preferentially, in a certain portion of the gastrointestinal tract, optionally, in a delayed manner. Examples of embedding compositions which may be used include polymeric substances and waxes. The active ingredient can also be in micro-encapsulated form, if appropriate, with one or more of the above-described excipients.
Liquid dosage forms for oral administration include pharmaceutically acceptable emulsions, microemulsions, solutions, suspensions, syrups, and elixirs. In addition to the active ingredient, the liquid dosage forms may contain inert diluents commonly used in the art, such as, for example, water or other solvents, solubilizing agents, and emulsifϊers such as ethyl alcohol, isopropyl alcohol, ethyl carbonate, ethyl acetate, benzyl alcohol, benzyl benzoate, propylene glycol, 1,3-butylene glycol, oils (in particular, cottonseed, groundnut, corn, germ, olive, castor, and sesame oils), glycerol, tetrahydrofuryl alcohol, polyethylene glycols, and fatty acid esters of sorbitan, and mixtures thereof.
Besides inert diluents, the oral compositions can also include adjuvants such as wetting agents, emulsifying and suspending agents, sweetening, flavoring, coloring, perfuming, and preservative agents.
Suspensions, in addition to the active compound(s) may contain suspending agents as, for example, ethoxylated isostearyl alcohols, polyoxyethylene sorbitol and sorbitan esters, microcrystalline cellulose, aluminum metahydroxide, bentonite, agar-agar and tragacanth, and mixtures thereof.
Formulations for rectal or vaginal administration may be presented as a suppository, which may be prepared by mixing one or more compound(s) with one or more suitable nonirritating excipients or carriers comprising, for example, cocoa butter, polyethylene glycol, a suppository wax or a salicylate, which is solid at room temperature, but liquid at body temperature and, therefore, will melt in the rectum or vaginal cavity and release the active agent. Formulations which are suitable for vaginal administration also include pessaries, tampons, creams, gels, pastes, foams, or spray formulations containing such carriers as are known in the art to be appropriate.
Dosage forms for the topical or transdermal administration of a compound(s) include powders, sprays, ointments, pastes, creams, lotions, gels, solutions, patches, and inhalants. The active component may be mixed under sterile conditions with a pharmaceutically acceptable carrier, and with any preservatives, buffers, or propellants which may be required.
The ointments, pastes, creams, and gels may contain, in addition to compound(s), excipients, such as animal and vegetable fats, oils, waxes, paraffins, starch, tragacanth, cellulose derivatives, polyethylene glycols, silicones, bentonites, silicic acid, talc, and zinc oxide, or mixtures thereof.
Powders and sprays can contain, in addition to a compound(s), excipients such as lactose, talc, silicic acid, aluminum hydroxide, calcium silicates, and polyamide powder, or mixtures of these substances. Sprays can additionally contain customary propellants, such as chlorofluorohydrocarbons and volatile unsubstituted hydrocarbons, such as butane and propane.
The compound(s) can be alternatively administered by aerosol. This is accomplished by preparing an aqueous aerosol, liposomal preparation, or solid particles containing the compound. A non-aqueous (e.g., fluorocarbon propellant) suspension could be used.
Ordinarily, an aqueous aerosol is made by formulating an aqueous solution or suspension of the agent together with conventional pharmaceutically acceptable carriers and stabilizers. The carriers and stabilizers vary with the requirements of the particular compound, but typically include nonionic surfactants (Tweens, Pluronics, or polyethylene glycol), innocuous proteins like serum albumin, sorbitan esters, oleic acid, lecithin, amino acids such as glycine, buffers, salts, sugars, or sugar alcohols. Aerosols generally are prepared from isotonic solutions.
Medicaments which may be administered in inhalant or aerosol formulations according to the invention include protease inhibitor prodrugs useful in inhalation therapy which may be presented in a form which is soluble or substantially soluble in the selected propellant system. The particle size of the particulate medicament should be such as to permit inhalation of substantially all of the medicament into the lungs upon administration of the aerosol formulation and will thus desirably be less than 20 microns, preferably in the range 1 to 10 microns, e.g., 1 to 5 microns. The particle size of the medicament may be reduced by conventional means, for example by milling or micronisation.
Administration of medicament may be indicated for the treatment of mild, moderate or severe acute or chronic symptoms or for prophylactic treatment. It will be appreciated that the precise dose administered will depend on the age and condition of the patient, the particular particulate medicament used and the frequency of administration and will ultimately be at the discretion of the attendant physician. When combinations of medicaments are employed the dose of each component of the combination will in general be that employed for each component when used alone. Typically, administration may be one or more times, for example from 1 to 8 times per day, giving for example 1, 2, 3 or 4 puffs each time. Preferably, administration may be one time per day. For administration, the drug is suitably inhaled from a nebulizer, from a pressurized metered dose inhaler, or as a dry powder from a dry powder inhaler (e.g., sold as TURBUHALER®) or from a dry powder inhaler utilizing gelatin, plastic or other capsules, cartridges or blister packs.
A diluent or carrier, generally non-toxic and chemically inert to the medicament; e.g., lactose, dextran, mannitol, glucose or any additives that will give the medicament a desired taste, can be added to the powdered medicament.
The micronized mixture may be suspended or dissolved in a liquid propellant mixture which is kept in a container that is sealed with a metering valve and fitted into a plastic actuator. The propellants used may be halocarbons of different chemical formulae. The most frequently used halocarbon propellants are trichlorofluoromethane, dichlorodifluoromethane, dichlorotetrafluoroethane, tetrafluoroethane, and 1,1-difluoroethane. Low concentrations of a surfactant such as sorbitan trioleate, lecithin, disodium dioctylsulphosuccinate, or oleic acid may also be used to improve the physical stability. Transdermal patches have the added advantage of providing controlled delivery of a compound(s) to the body. Such dosage forms can be made by dissolving or dispersing the agent in the proper medium. Absorption enhancers can also be used to increase the flux of the compound(s) across the skin. The rate of such flux can be controlled by either providing a rate controlling membrane or dispersing the peptidomimetic in a polymer matrix or gel.
Ophthalmic formulations, eye ointments, powders, solutions, and the like, are also contemplated as being within the scope of this invention. Pharmaceutical compositions of this invention suitable for parenteral administration comprise one or more compounds(s) in combination with one or more pharmaceutically acceptable sterile isotonic aqueous or non-aqueous solutions, dispersions, suspensions or emulsions, or sterile powders which may be reconstituted into sterile injectable solutions or dispersions just prior to use, which may contain antioxidants, buffers, bacteriostats, solutes which render the formulation isotonic with the blood of the intended recipient or suspending or thickening agents.
Examples of suitable aqueous and non-aqueous carriers which may be employed in the pharmaceutical compositions of the invention include water, ethanol, polyols (such as glycerol, propylene glycol, polyethylene glycol, and the like), and suitable mixtures thereof, vegetable oils, such as olive oil, and injectable organic esters, such as ethyl oleate. Proper fluidity can be maintained, for example, by the use of coating materials, such as lecithin, by the maintenance of the required particle size in the case of dispersions, and by the use of surfactants.
These compositions may also contain adjuvants such as preservatives, wetting agents, emulsifying agents, and dispersing agents. Prevention of the action of microorganisms may be ensured by the inclusion of various antibacterial and antifungal agents, for example, paraben, chlorobutanol, phenol sorbic acid, and the like. It may also be desirable to include isotonic agents, such as sugars, sodium chloride, and the like into the compositions. In addition, prolonged absorption of the injectable pharmaceutical form may be brought about by the inclusion of agents which delay absorption such as aluminum monostearate and gelatin.
In some cases, in order to prolong the effect of a drug, it is desirable to slow the absorption of the drug from subcutaneous or intramuscular injection. This may be accomplished by the use of a liquid suspension of crystalline or amorphous material having poor water solubility. The rate of absorption of the drug then depends upon its rate of dissolution which, in turn, may depend upon crystal size and crystalline form. Alternatively, delayed absorption of a parenterally administered drug form is accomplished by dissolving or suspending the drug in an oil vehicle. Injectable depot forms are made by forming microencapsule matrices of compound(s) in biodegradable polymers such as polylactide-polyglycolide. Depending on the ratio of drug to polymer, and the nature of the particular polymer employed, the rate of drug release can be controlled. Examples of other biodegradable polymers include poly(orthoesters) and poly(anhydrides). Depot injectable formulations are also prepared by entrapping the drug in liposomes or microemulsions which are compatible with body tissue.
When the compounds(s) of the present invention are administered as pharmaceuticals to humans and animals, they can be given per se or as a pharmaceutical composition containing, for example, 0.1 to 99.5% (more preferably, 0.5 to 90%) of active ingredient in combination with a pharmaceutically acceptable carrier.
The preparations of agents may be given orally, parenterally, topically, or rectally . They are of course given by forms suitable for each administration route. For example, they are administered in tablets or capsule form, by injection, inhalation, eye lotion, ointment, suppository, infusion; topically by lotion or ointment; and rectally by suppositories.
The phrases "parenteral administration" and "administered parenterally" as used herein means modes of administration other than enteral and topical administration, usually by injection, and includes, without limitation, intravenous, intramuscular, intraarterial, intrathecal, intracapsular, intraorbital, intracardiac, intradermal, intraperitoneal, transtracheal, subcutaneous, subcuticular, intraarticular, subcapsular, subarachnoid, intraspinal and intrasternal injection, and infusion.
The phrases "systemic administration," "administered systemically," "peripheral administration" and "administered peripherally" as used herein mean the administration of a ligand, drug, or other material other than directly into the central nervous system, such that it enters the patient's system and thus, is subject to metabolism and other like processes, for example, subcutaneous administration.
These compounds(s) may be administered to humans and other animals for therapy by any suitable route of administration, including orally, nasally, as by, for example, a spray, rectally, intravaginally, parenterally, intracisternally, and topically, as by powders, ointments or drops, including buccally and sublingually.
The addition of the active compound of the invention to animal feed is preferably accomplished by preparing an appropriate feed premix containing the active compound in an effective amount and incorporating the premix into the complete ration. Alternatively, an intermediate concentrate or feed supplement containing the active ingredient can be blended into the feed. The way in which such feed premixes and complete rations can be prepared and administered are described in reference books (such as Applied Animal Nutrition; San Francisco: Freedman, 1969; ox Livestock Feeds and Feeding; Corvallis: O & B Books, 1977).
Regardless of the route of administration selected, the compound(s), which may be used in a suitable hydrated form, and/or the pharmaceutical compositions of the present invention, are formulated into pharmaceutically acceptable dosage forms by conventional methods known to those of skill in the art. Actual dosage levels of the active ingredients in the pharmaceutical compositions of this invention may be varied so as to obtain an amount of the active ingredient which is effective to achieve the desired therapeutic response for a particular patient, composition, and mode of administration, without being toxic to the patient. (vii) Combinatorial Libraries The compounds of the present invention, particularly libraries of variants having various representative classes of substituents, are amenable to combinatorial chemistry and other parallel synthesis schemes (see, for example, PCT WO 94/08051). The result is that large libraries of related compounds, e.g., a variegated library of compounds represented above, can be screened rapidly in high throughput assays in order to identify potential protease inhibitor lead compounds, as well as to refine the specificity, toxicity, and/or cytotoxic-kinetic profile of a lead compound.
Simply for illustration, a combinatorial library for the purposes of the present invention is a mixture of chemically related compounds which may be screened together for a desired property. The preparation of many related compounds in a single reaction greatly reduces and simplifies the number of screening processes which need to be carried out. Screening for the appropriate physical properties can be done by conventional methods.
Diversity in the library can be created at a variety of different levels. For instance, the substrate aryl groups used in the combinatorial reactions can be diverse in terms of the core aryl moiety, e.g., a variegation in terms of the ring structure, and/or can be varied with respect to the other substituents.
A variety of techniques are available in the art for generating combinatorial libraries of small organic molecules such as the subject protease inhibitors. See, for example, Blondelle et al. Trends Anal. Chem. 1995, 14, 83; the Affymax U.S. Patents 5,359,115 and 5,362,899: the Ellman U.S. Patent 5,288,514: the Still et al. PCT publication WO 94/08051; the ArQuIe U.S. Patents 5,736,412 and 5,712,171; Chen et al. J. Am. Chem. Soc. 1994, 116, 2661 : Kerr et al. J. Am. Chem. Soc. 1993, 115, 252; PCT publications WO92/10092, WO93/09668 and WO91/07087; and the Lerner et al. PCT publication WO93/20242). Accordingly, a variety of libraries on the order of about 100 to 1,000,000 or more diversomers of the subject protease inhibitors can be synthesized and screened for particular activity or property.
In an exemplary embodiment, a library of candidate protease inhibitor diversomers can be synthesized utilizing a scheme adapted to the techniques described in the Still et al. PCT publication WO 94/08051 , e.g., being linked to a polymer bead by a hydrolyzable or photo lyzable group, optionally located at one of the positions of the candidate agonists or a substituent of a synthetic intermediate. According to the Still et al. technique, the library is synthesized on a set of beads, each bead including a set of tags identifying the particular diversomer on that bead. The bead library can then be "plated" with proteases for which an inhibitor is sought. The diversomers can be released from the bead, e.g., by hydrolysis. The structures of the compounds useful in the present invention lend themselves readily to efficient synthesis. The nature of the structures of the subject compounds, as generally set forth above, allows the rapid combinatorial assembly of such compounds. For example, as in the scheme set forth below, an activated aryl group, such as an aryl triflate or bromide, attached to a bead or other solid support can be linked to another aryl group by performing a Stille or Suzuki coupling with an aryl stannane or an aryl boronic acid. If the second aryl group is functionalized with an aldehyde, an amine substituent can be added through a reductive amination. Alternatively, the second aryl group could be functionalized with a leaving group, such as a triflate, tosylate, or halide, capable of being displaced by an amine. Or, the second aryl group may be functionalized with an amine group capable of undergoing reductive amination with an amine, e.g., CyKNH2. Other possible coupling techniques include transition metal-mediated amine arylation reactions. The resultant secondary amine can then be further functionalized by an acylation, alkylation, or arylation to generate a tertiary amine or amide which can then be cleaved from the resin or support. These reactions generally are quite mild and have been successfully applied in combinatorial solid-phase synthesis schemes. Furthermore, the wide range of substrates and coupling partners suitable and available for these reactions permits the rapid assembly of large, diverse libraries of compounds for testing in assays as set forth herein. For certain schemes, and for certain substitutions on the various substituents of the subject compounds, one of skill in the art will recognize the need for masking certain functional groups with a suitable protecting group. Such techniques are well known in the art and are easily applied to combinatorial synthesis schemes.
Figure imgf000054_0001
Cy-K-NH 2 NaBH4
Figure imgf000054_0002
Many variations on the above and related pathways permit the synthesis of widely diverse libraries of compounds which may be tested as protease inhibitors.
Variations, modifications, and other implementations of what is described herein will occur to those of ordinary skill without departing from the spirit and the scope of the invention. Accordingly, the invention is not to be limited only to the preceding illustrative description. For additional illustrative features that may be used with the invention, including the embodiments described here, refer to the documents listed herein above and incorporated by reference in their entirety. All operative combinations between the above described illustrative embodiments and those features described below are considered to be potentially patentable embodiments of the invention. IV. Exemplification
The invention now being generally described, it will be more readily understood by reference to the following examples which are included merely for purposes of illustration of certain aspects and embodiments of the present invention, and are not intended to limit the invention. Example 1 DPIV Inhibition Assay
The inhibitor solution is prepared by dissolving 3 - 5 mg of inhibitor in pH 2 solution (0.01 N HCl), such that the concentration of the solution is equal to 1 mg/10 μL. A 10 μL sample of this solution is then added to 990 μL of pH 8 buffer (0.1 M HEPES, 0.14
M NaCl), and the solution is allowed to stand at room temperature overnight.
The enzyme solution is prepared by diluting 20 μL of DPIV (concentration 2.5 μM) into 40 mL of pH 8 buffer. The substrate solution is prepared by dissolving 2.0 mg of L-alanyl-L-proline-/?αra- nitroanilide into 20 mL of pH 8 buffer.
250 μL of enzyme solution is added to well #B1 to #H1, #A2 to #H2, and #A3 to
#H3 of a 96 well plate, while well #A1 receives 250 μL of pH 8 buffer instead of enzyme solution. 90μL of pH 8 buffer is then added to column 5 (from well #A5 to #H5). A 1 : 10 dilution is then performed by adding inhibitor solution to #A5 and the solution is mixed well before transferring 10 μL of this solution from #A5 to #B5. The solution in #B5 is then mixed well before transferring 10 μL of this solution from #B5 to
#C5. The solution in #C5 is then mixed well before transferring 10 μL of this solution from
#C5 to #D5. The solution in #D5 is then mixed well before transferring 10 μL of this solution from #D5 to #E5. The solution in #E5 is then mixed well before transferring 10 μL of this solution from #E5 to #F5. The solution in #F5 is then mixed well before transferring 10 μL of this solution from #F5 to #G5. The solution in #G5 is then mixed well before transferring 10 μL of this solution from #G5 to #H5.
A 30 μL aliquot is then transferred from #H5 to #H3 for row H, and the contents are mixed well. The analogous procedure is repeated for rows G, F, E, D, C, B, and A sequentially. The plate is then shaken on a plate shaker for 5 minutes before allowing the plate to incubate at room temperature for an additional 5 minutes.
Once the plate has been allowed to incubate, 30 μL of substrate is added to each well except well #A1. The plate is then placed on a plate shaker for 5 minutes before allowing the plate to incubate at room temperature for 25 minutes. The absorbance is then immediately read at a wavelength of 410 nm.
Example 2
Selectivity for Dipeptidyl Peptidase Isoforms
The assay described in Example 1 is used to determine the IC50 values for several compounds of the invention. In this example, the assay is conducted for DPIV and DP8 or
DP9. The ratio of IC50 values for each tested compound is calculated in order to determine the selectivity for the DPIV isoform. IC50 values were measured at the same pH throughout the assay.
Certain compounds of the invention inhibit DPIV at least 10 times, preferably at least 100 times, more strongly than they inhibit DP8 and/or DP9, i.e., have an IC50 at least 10 (or 100) times lower against DPIV than against DP8 and/or DP9.
IV. Equivalents
Those skilled in the art will recognize, or be able to ascertain using no more than routine experimentation, many equivalents to the specific embodiments of the invention described herein. Such equivalents are intended to be encompassed by the following claims.
Incorporation by Reference
All of the U.S. patents and U.S. patent application publications cited herein are hereby incorporated by reference.

Claims

We claim:
1. A compound represented by the formula:
Figure imgf000057_0001
or a pharmaceutically acceptable salt thereof; wherein:
W is selected from the group consisting of -CN, -CH=NR4,
Figure imgf000057_0002
; wherein
R4 represents H, alkyl, alkenyl, alkynyl, -C(X1)(X2)(X3), -(CH2)Di-R43, -(CH2)Ii-OH, -(CH2)n-O-alkyl, -(CH2)n-O-alkenyl, -(CH2)n-O-alkynyl, -(CH2)n-O-(CH2)m-R43, -(CH2)n-SH, -(CH2)n-S-alkyl, -(CH2)n-S-alkenyl,
-(CH2)n-S-alkynyl, -(CH2)U-S-(CH2)In-R43, -C(O)C(O)NH2, and -C(O)C(O)OR44;
R43 represents independently for each occurrence aryl, aralkyl, or non-aromatic hetereocyclyl;
R44 represents independently for each occurrence H, alkyl, alkenyl, aryl, aralkyl, or non-aromatic heterocyc IyI;
Yi and Y2 independently represent OH, or a group capable of being hydro lyzed to a hydroxyl group; or taken together with the boron to which they are bonded form a 5- membered to 8-membered ring comprising said boron and two oxygen atoms bonded to said boron; R40 is O or S;
R41 is selected from the group consisting of N3, SH, NH2, NO2, and -OR^S
R42 is selected from the group consisting of hydrogen, lower alkyl, amine, and -OR44, or a pharmaceutically acceptable salt; or R41 and R42 taken together with the phosphorous atom to which they are attached complete a heterocyclic ring having from 5 to 8 atoms in the ring structure; X1 is halogen;
X2 and X3 is hydrogen or halogen;
Ri is selected from a C-terminally linked amino acid residue or amino acid analog, a C-terminally linked peptide or peptide analog, an amino-protecting group,
Figure imgf000058_0001
R2 is absent or represents one or more substituents attached at positions 3, 4, or 5 of the dehydroproline ring, each of which is independently selected from the group consisting of hydrogen, halogen, hydroxyl, lower alkyl, lower alkenyl, lower alkynyl, lower alkoxy, lower hydroxyalkyl, lower alkoxyalkyl, carbonyl, carboxyl, ester, formate, ketone, thiocarbonyl, thioester, thioacetate, thioformate, amino, acylamino, amido, nitro, sulfate, sulfonate, sulfonamido, -(CH2Xn-R51, -(CH2)m-OH, -(CH2)m-O-lower alkyl, -(CH2X11-O- lower alkenyl, -(CH2)n-O-(CH2)m-R51, -(CH2Xn-SH, -(CH2)m-S-lower alkyl, -(CH2)m-S-lower alkenyl, or -(CH2)J1-S-(CH2)J11-R51' azido, cyano, isocyanato, thiocyanato,
isothiocyanato, cyanato, NzzzzzzzzC, and C C ^22;
R3 is selected from the group consisting of hydrogen, halogen, lower alkyl, lower alkenyl, lower alkynyl, carbonyl, thiocarbonyl, amino, acylamino, amido, nitro, sulfate, sulfonate, a sulfonamido, -(CH2Xn-R51, -(CH2Xn-OH, -(CH2)m-O-lower alkyl, -(CH2Xn-O- lower alkenyl, -(CH2)n-O-(CH2)m-R5 { , -(CH2Xn-SH, -(CH2)m-S-lower alkyl, -(CH2Xn-S- lower alkenyl, or -(CH2)J1-S-(CH2)J11-R51, azido, cyano, isocyanato, thiocyanato,
© θ isothiocyanato, cyanato, NzzzzzzzzC, and C-Zn=C R22;
R5 is selected from the group consisting of hydrogen, halogen, alkyl, alkenyl, alkynyl, aryl, -(CH2)m-R51, -(CH2)m-OH, -(CH2)m-O-alkyl, -(CH2)m-O-alkenyl, -(CH2)m-O-alkynyl, -(CH2)m-O-(CH2)m-R51, -(CH2)m-SH, -(CH2)m-S-alkyl, -(CH2)m-S- alkenyl, -(CH2)m-S-alkynyl, -(CH2)m-S-(CH2)m-R51,
° ,R52 NH2 0 (CH2)m— NN (CH2)n— C-N^ (CH2)n— NH2-C-NH2 , -(CH2Jn-C-O-R51 ,
R53 ' R53 '
0 0 0 O
Il Il Il Il
— (CH2)n— C— alkyl , — (CH2)n— C— alkenyl , — (CH2)n— C— alkynyl , and — (CH2)n-C-(CH2)m-R51- wherein
R51 represents for each occurrence aryl, aralkyl, or non-aromatic heterocyclyl;
R52 and R53 independently represent hydrogen, alkyl, alkenyl, -(CH2)m-R5i, -C(=O)-alkyl, -C(=O)-alkenyl, -C(=O)-alkynyl, or -C(=O)-(CH2)m-R5i; or R52 and R53 taken together with the N atom to which they are attached complete a heterocyclic ring having from 4 to 8 atoms in the ring structure; m is zero or an integer in the range of 1 to 8; and n is an integer in the range of 1 to 8.
2. The compound of claim 1, wherein W is -B(Yi)(Y2).
3. The compound of claim 1, wherein Ri is a C-terminally linked amino acid residue or amino acid analog or a C-terminally linked peptide or peptide analog.
4. The compound of any one of claims 1-3, wherein R3 is hydrogen or lower alkyl.
5. The compound of any one of claims 1-3, wherein R2 is lower alkyl, lower hydroxyalkyl, lower alkoxyalkyl, or hydroxyl.
6. The compound of any one of claims 1-3, wherein R2 occurs once and is located at the 5-position of said dehydroproline ring.
7. The compound of claim 6, wherein R2 is hydroxyl.
8. The compound of claim 1 , wherein said compound is represented by:
Figure imgf000059_0001
or a pharmaceutically acceptable salt thereof.
9. The compound of claim 1, wherein said compound is represented by:
Figure imgf000059_0002
or a pharmaceutically acceptable salt thereof.
10. The compound of claim 1, wherein said compound is represented by:
Figure imgf000060_0001
or a pharmaceutically acceptable salt thereof.
11. The compound of any one of claims 8-10, wherein W is -B(Yi)(Y2).
12. A compound of any one of claims 8-10, wherein Ri is a C-terminally linked amino acid residue, amino acid analog, or C-terminally linked peptide or peptide analog.
13. A compound of claim 11 , wherein R3 is hydrogen or lower alkyl.
14. A compound as in any one of claims 10-13, wherein R2 is lower alkyl, lower hydroxyalkyl, lower alkoxyalkyl, or hydroxyl.
15. A compound of any one of claims 10-13, wherein R2 is hydroxyl.
16. A compound represented by the formula:
Figure imgf000060_0002
or a pharmaceutically acceptable salt thereof; wherein:
R2 is absent or represents one or more substituents attached at positions 3, 4, or 5 of the dehydroproline ring, each of which is independently selected from the group consisting of hydrogen, halogen, hydroxyl, lower alkyl, lower alkenyl, lower alkynyl, lower alkoxy, lower hydroxyalkyl, lower alkoxyalkyl, carbonyl, carboxyl, ester, formate, ketone, thiocarbonyl, thioester, thioacetate, thioformate, amino, acylamino, amido, nitro, sulfate, sulfonate, sulfonamido, -(CH2)m-R43, -(CH2Xn-OH, -(CH2)m-O-lower alkyl, -(CH2Xn-O- lower alkenyl, -(CH2X1-O-(CH2Xn-R43, -(CH2Xn-SH, -(CH2)m-S-lower alkyl,
-(CH2)m-S-lower alkenyl, or -(CH2)n-S-(CH2)m-R43> azido, cyano, isocyanato, thiocyanato,
isothiocyanato, cyanato, Nz^=C, and C C R22;
Rioi is selected from the group consisting of H, alkyl, alkoxy, alkenyl, alkynyl, amino, alkylamino, acylamino, cyano, sulfonylamino, acyloxy, aryl, non-aromatic heterocyclyl, heteroaryl, and a polypeptide chain of 1 to 8 amino acid residues; Rio2 is selected from the group consisting of H, lower alkyl, and aralkyl;
Rio3 is selected from the group consisting of H, halogen, and alkyl, or R102 and R103 together with the atoms to which they are attached, form a 3- to 6-membered heterocyclic ring;
R105 is selected from the group consisting of H, halogen, lower alkyl, and aralkyl;
R106 is selected from the group consisting of -CN, -CH=NR4,
Figure imgf000061_0001
; wherein
R4 represents H, alkyl, alkenyl, alkynyl, -C(X1)(X2)(X3), -(CH2)Hi-R43, -(CH2)n-OH, -(CH2)n-O-alkyl, -(CH2)n-O-alkenyl, -(CH2)n-O-alkynyl, -(CH2)n-O-(CH2)m-R43, -(CH2)n-SH, -(CH2)n-S-alkyl, -(CH2)n-S-alkenyl,
-(CH2)n-S-alkynyl, -(CH2)n-S-(CH2)m-R43, -C(O)C(O)NH2, or -C(O)C(O)OR44;
R43 represents independently for each occurrence aryl, aralkyl, or non-aromatic hetereocyclyl;
R44 independently for each occurrence is selected from the group consisting of H, alkyl, alkenyl, aryl, aralkyl, and non-aromatic heterocyclyl;
Yi and Y2 independently represent OH, or a group capable of being hydro lyzed to a hydroxyl group; or taken together with the boron to which they are bonded form a 5- membered to 8-membered ring comprising said boron and two oxygen atoms bonded to said boron; R40 is O or S;
R41 is selected from the group consisting OfN3, SH, NH2, NO2 and -ORgS
R42 is selected from the group consisting of H, lower alkyl, amine, and -OR44, or a pharmaceutically acceptable salt; or R41 and R42 taken together with the phosphorous atom to which they are attached complete a heterocyclic ring having from 5 to 8 atoms in the ring structure;
X1 is halogen;
X2 and X3 is hydrogen or halogen; and n is an integer in the range of 2 to 6;
Rio7 is selected from the group consisting of H, aryl, alkyl, aralkyl, non-aromatic heterocyclyl, heteroaryl, heteroaralkyl, and polypeptide chains of 1 to 8 amino acid residues;
L is absent or is selected from the group consisting of alkyl, alkenyl, alkynyl, -(CH2)mO(CH2)m-, -(CH2)mNR2(CH2)m-, and -(CH2)mS(CH2)m-;
X is absent or is selected from the group consisting of -N(R107)-, -O-, and -S-;
Y is absent or is selected from the group consisting of -C(=O)-, -C(=S)-, and -SO2-; and m is, independently for each occurrence, an integer from 0 to 10.
17. The compound of claim 16, wherein Ri06 is -B(Yi)(Y2).
18. The compound of claim 16, wherein R103 is selected from the group consisting of -CH3, -CH(CHs)2, -CH2CH(CHs)2, -C(CH3)(CH2CH3), -CH2CH2SCH3, -CH2-phenyl, -CH2-(3-indolyl), -CH2OH, -CH(OH)CH3, -CH2CONH2, -CH2CH2CONH2, -CH2-(4-hydroxyphenyl), -CH2SH, -(CH2)4NH2, -(CH2)3NH-C(=NH)NH2, -CH-(5-imidazolyl), -CH2COOH, and -CH2CH2COOH.
19. The compound of claim 16, wherein Ri03 is selected from the group consisting of H, -CH3 and -(CH2)3NH-C(=NH)NH2.
20. The compound of claim 16, wherein L, X and Y are absent.
21. The compound of claim 20, wherein R101 is hydrogen.
22. The compound of claim 16, wherein Ri03 and R105 are hydrogen.
23. The compound of claim 16, wherein the compound is represented by a structural formula selected from the group consisting of:
Figure imgf000062_0001
24. The compound of any one of claims 16-23, wherein R2 is lower alkyl, lower hydroxyalkyl, lower alkoxyalkyl, or hydroxyl.
25. The compound of any one of claims 16-24, wherein R2 occurs once and is located at the 5-position of said dehydroproline ring.
26. The compound of any one of claims 16-25, wherein R2 is hydroxyl.
27. The compound of claim 16, wherein said compound is represented by:
Figure imgf000063_0001
or a pharmaceutically acceptable salt thereof.
28. The compound of claim 16, wherein said compound is represented by:
Figure imgf000063_0002
or a pharmaceutically acceptable salt thereof.
29. The compound of claim 16, wherein said compound is represented by:
Figure imgf000063_0003
or a pharmaceutically acceptable salt thereof.
30. The compound of any one of claims 27-29, wherein Rioβ is -B(Yi)(Y2).
31. The compound of any one of claims 27-30, wherein R103 is selected from the group consisting of H, -CH3, -CH(CH3)2, -CH2CH(CH3)2, -C(CH3)(CH2CH3), -CH2CH2SCH3, -CH2-phenyl, -CH2-(3-indolyl), -CH2OH, -CH(OH)CH3, -CH2CONH2, -CH2CH2CONH2, -CH2-(4-hydroxyphenyl), -CH2SH, -(CH2)4NH2, -(CH2)3NH-C(=NH)NH2, -CH-(5-imidazolyl), -CH2COOH, and -CH2CH2COOH.
32. The compound of any one of claims 27-30, wherein R103 is selected from hydrogen, -CH3 and -(CH2)3NH-C(=NH)NH2.
33. The compound of any one of claims 27-32, wherein L, X and Y are absent.
34. The compound of claim 33, wherein R101 is hydrogen.
35. The compound of any one of claims 27-34, wherein R103 and R105 are hydrogen.
36. A compound selected from the group consisting of:
Figure imgf000064_0001
37. The compound of claim 28, wherein said compound is selected from the group consisting of:
Figure imgf000064_0002
38. The compound of claim 29, wherein said compound is selected from the group consisting of:
Figure imgf000065_0001
39. The compound of claim 37 or 38, wherein R2 is lower alkyl, lower hydroxyalkyl, lower alkoxyalkyl, or hydroxyl.
40. The compound of claim 37 or 38, wherein said R2 is hydroxyl.
41. The compound of any one of claims 1 -40, wherein the compound is a protease inhibitor.
42. The compound of claim 41, wherein the compound inhibits dipeptidyl peptidase IV with a K1 of less than 50 nM.
43. The compound of claim 42, wherein the compound inhibits dipeptidyl peptidase VIII or dipeptidyl peptidase IX with a K1 of greater than 100 μM.
44. The compound of any one of claims 1-40, wherein the compound is effective when administered orally.
45. A pharmaceutical composition, comprising a pharmaceutically acceptable carrier; and a compound of any one of claims 1 -40.
46. A method for inhibiting the proteolytic activity of a post-pro line protease enzyme in a patient, comprising administering to a patient in need thereof a therapeutically effective amount of a compound of any one of claims 1-40.
47. The method of claim 46, wherein the compound increases the patient's plasma concentrations of a peptide hormone selected from the group consisting of glucagon- like peptide, NPY, PPY, secretin, GLP-I, GLP-2, and GIP.
48. A method of regulating glucose metabolism in a patient, comprising administering to a patient in need thereof a therapeutically effective amount of a compound of any one of claims 1-40.
49. The method of claim 48, wherein the patient is suffering from Type II diabetes, insulin resistance, glucose intolerance, hyperglycemia, hypoglycemia, hyperinsulinemia, obesity, hyperlipidemia, or hyperlipoproteinemia.
50. The method of claim 48 or 49, further comprising administering a therapeutically effective amount of insulin or an insulinotropic agent.
51. The method of claim 48 or 49, further comprising administering a therapeutically effective amount of Ml receptor antagonist, a prolactin inhibitor, an agent acting on the ATP-dependent potassium channel of β-cells, metformin, or a glucosidase inhibitor.
52. A packaged pharmaceutical, comprising a compound of any one of claims 1-40, and instructions describing the use for inhibiting a post-pro line protease enzyme.
53. A packaged pharmaceutical, comprising a compound of any one of claims 1-40, and instructions describing the use for regulating glucose metabolism.
54. The packaged pharmaceutical of claim 53, wherein the compound is co-formulated with or co-packaged with insulin or an insulinotropic agent.
55. The packaged pharmaceutical of claim 53, wherein the compound is co-formulated with or co-packaged with an Ml receptor antagonist, a prolactin inhibitor, an agent acting on the ATP-dependent potassium channel of β-cells, metformin, or a glucosidase inhibitor.
PCT/US2008/058378 2007-03-27 2008-03-27 3,4-dehydro-proline-containing inhibitors of dipeptidylpeptidase iv WO2008119005A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US90826707P 2007-03-27 2007-03-27
US60/908,267 2007-03-27

Publications (1)

Publication Number Publication Date
WO2008119005A1 true WO2008119005A1 (en) 2008-10-02

Family

ID=39789032

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2008/058378 WO2008119005A1 (en) 2007-03-27 2008-03-27 3,4-dehydro-proline-containing inhibitors of dipeptidylpeptidase iv

Country Status (1)

Country Link
WO (1) WO2008119005A1 (en)

Cited By (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2011107494A1 (en) 2010-03-03 2011-09-09 Sanofi Novel aromatic glycoside derivatives, medicaments containing said compounds, and the use thereof
WO2011157827A1 (en) 2010-06-18 2011-12-22 Sanofi Azolopyridin-3-one derivatives as inhibitors of lipases and phospholipases
WO2011161030A1 (en) 2010-06-21 2011-12-29 Sanofi Heterocyclic substituted methoxyphenyl derivatives having an oxo group, method for producing same, and use thereof as gpr40 receptor modulators
WO2012004270A1 (en) 2010-07-05 2012-01-12 Sanofi Spirocyclically substituted 1,3-propane dioxide derivatives, methods for the production thereof and use of the same as medicament
WO2012004269A1 (en) 2010-07-05 2012-01-12 Sanofi (2-aryloxy-acetylamino)-phenyl-propionic acid derivatives, method for producing same and use thereof as pharmaceuticals
WO2012010413A1 (en) 2010-07-05 2012-01-26 Sanofi Aryloxy-alkylene substituted hydroxyphenyl hexynoic acids, methods for the production thereof and use of the same as medicament
WO2013037390A1 (en) 2011-09-12 2013-03-21 Sanofi 6-(4-hydroxy-phenyl)-3-styryl-1h-pyrazolo[3,4-b]pyridine-4-carboxylic acid amide derivatives as kinase inhibitors
WO2013045413A1 (en) 2011-09-27 2013-04-04 Sanofi 6-(4-hydroxy-phenyl)-3-alkyl-1h-pyrazolo[3,4-b]pyridine-4-carboxylic acid amide derivatives as kinase inhibitors
WO2014064215A1 (en) 2012-10-24 2014-05-01 INSERM (Institut National de la Santé et de la Recherche Médicale) TPL2 KINASE INHIBITORS FOR PREVENTING OR TREATING DIABETES AND FOR PROMOTING β-CELL SURVIVAL
WO2016151018A1 (en) 2015-03-24 2016-09-29 INSERM (Institut National de la Santé et de la Recherche Médicale) Method and pharmaceutical composition for use in the treatment of diabetes

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5385889A (en) * 1991-04-19 1995-01-31 Scios Nova Inc. Bradykinin antagonist peptides
US5543396A (en) * 1994-04-28 1996-08-06 Georgia Tech Research Corp. Proline phosphonate derivatives
US6949514B2 (en) * 1999-05-25 2005-09-27 Point Therapeutics, Inc. Anti-tumor agents

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5385889A (en) * 1991-04-19 1995-01-31 Scios Nova Inc. Bradykinin antagonist peptides
US5543396A (en) * 1994-04-28 1996-08-06 Georgia Tech Research Corp. Proline phosphonate derivatives
US6949514B2 (en) * 1999-05-25 2005-09-27 Point Therapeutics, Inc. Anti-tumor agents

Cited By (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2011107494A1 (en) 2010-03-03 2011-09-09 Sanofi Novel aromatic glycoside derivatives, medicaments containing said compounds, and the use thereof
WO2011157827A1 (en) 2010-06-18 2011-12-22 Sanofi Azolopyridin-3-one derivatives as inhibitors of lipases and phospholipases
WO2011161030A1 (en) 2010-06-21 2011-12-29 Sanofi Heterocyclic substituted methoxyphenyl derivatives having an oxo group, method for producing same, and use thereof as gpr40 receptor modulators
WO2012004270A1 (en) 2010-07-05 2012-01-12 Sanofi Spirocyclically substituted 1,3-propane dioxide derivatives, methods for the production thereof and use of the same as medicament
WO2012004269A1 (en) 2010-07-05 2012-01-12 Sanofi (2-aryloxy-acetylamino)-phenyl-propionic acid derivatives, method for producing same and use thereof as pharmaceuticals
WO2012010413A1 (en) 2010-07-05 2012-01-26 Sanofi Aryloxy-alkylene substituted hydroxyphenyl hexynoic acids, methods for the production thereof and use of the same as medicament
WO2013037390A1 (en) 2011-09-12 2013-03-21 Sanofi 6-(4-hydroxy-phenyl)-3-styryl-1h-pyrazolo[3,4-b]pyridine-4-carboxylic acid amide derivatives as kinase inhibitors
WO2013045413A1 (en) 2011-09-27 2013-04-04 Sanofi 6-(4-hydroxy-phenyl)-3-alkyl-1h-pyrazolo[3,4-b]pyridine-4-carboxylic acid amide derivatives as kinase inhibitors
WO2014064215A1 (en) 2012-10-24 2014-05-01 INSERM (Institut National de la Santé et de la Recherche Médicale) TPL2 KINASE INHIBITORS FOR PREVENTING OR TREATING DIABETES AND FOR PROMOTING β-CELL SURVIVAL
WO2016151018A1 (en) 2015-03-24 2016-09-29 INSERM (Institut National de la Santé et de la Recherche Médicale) Method and pharmaceutical composition for use in the treatment of diabetes

Similar Documents

Publication Publication Date Title
US20100105629A1 (en) N-Substituted Peptidomimetic Inhibitors of Dipeptidylpeptidase IV
US9757400B2 (en) Peptidomimetic inhibitors of post-proline cleaving enzymes
WO2008119005A1 (en) 3,4-dehydro-proline-containing inhibitors of dipeptidylpeptidase iv
AU2005216970B2 (en) Inhibitors of dipeptidylpeptidase IV for regulating glucose metabolism
AU2005217642B2 (en) Lactams as conformationally constrained peptidomimetic inhibitors
AU2003228793B2 (en) Smart Pro-Drugs of Serine Protease Inhibitors
US8513187B2 (en) Soft protease inhibitors, and pro-soft forms thereof
AU2011203039B2 (en) Inhibitors of dipeptidylpeptidase IV for regulating glucose metabolism

Legal Events

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

Ref document number: 08732907

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 08732907

Country of ref document: EP

Kind code of ref document: A1