Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
J Med Chem ; 64(4): 2024-2045, 2021 02 25.
Article in English | MEDLINE | ID: mdl-33538587

ABSTRACT

We identified a set of thiosemicarbazone (TSC) metal ion chelators that reactivate specific zinc-deficient p53 mutants using a mechanism called zinc metallochaperones (ZMCs) that restore zinc binding by shuttling zinc into cells. We defined biophysical and cellular assays necessary for structure-activity relationship studies using this mechanism. We investigated an alternative class of zinc scaffolds that differ from TSCs by substitution of the thiocarbamoyl moiety with benzothiazolyl, benzoxazolyl, and benzimidazolyl hydrazones. Members of this series bound zinc with similar affinity and functioned to reactivate mutant p53 comparable to the TSCs. Acute toxicity and efficacy assays in rodents demonstrated C1 to be significantly less toxic than the TSCs while demonstrating equivalent growth inhibition. We identified C85 as a ZMC with diminished copper binding that functions as a chemotherapy and radiation sensitizer. We conclude that the benzothiazolyl, benzoxazolyl, and benzimidazolyl hydrazones can function as ZMCs to reactivate mutant p53 in vitro and in vivo.


Subject(s)
Benzothiazoles/therapeutic use , Benzoxazoles/therapeutic use , Chelating Agents/therapeutic use , Hydrazones/therapeutic use , Tumor Suppressor Protein p53/metabolism , Zinc/metabolism , Animals , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Benzothiazoles/chemical synthesis , Benzothiazoles/pharmacology , Benzoxazoles/chemical synthesis , Benzoxazoles/pharmacology , Cell Line, Tumor , Chelating Agents/chemical synthesis , Chelating Agents/pharmacology , Humans , Hydrazones/chemical synthesis , Hydrazones/pharmacology , Mice, Nude , Molecular Structure , Neoplasms/drug therapy , Photosensitizing Agents/chemical synthesis , Photosensitizing Agents/pharmacology , Photosensitizing Agents/therapeutic use , Structure-Activity Relationship , Tumor Suppressor Protein p53/drug effects , Xenograft Model Antitumor Assays
3.
Oncogene ; 37(27): 3672-3685, 2018 07.
Article in English | MEDLINE | ID: mdl-29622797

ABSTRACT

BMP receptor inhibitors induce death of cancer cells through the downregulation of antiapoptotic proteins XIAP, pTAK1, and Id1-Id3. However, the current most potent BMP receptor inhibitor, DMH2, does not downregulate BMP signaling in vivo because of metabolic instability and poor pharmacokinetics. Here we identified the site of metabolic instability of DMH2 and designed a novel BMP receptor inhibitor, JL5. We show that JL5 has a greater volume of distribution and suppresses the expression of Id1 and pTak1 in tumor xenografts. Moreover, we demonstrate JL5-induced tumor cell death and tumor regression in xenograft mouse models without immune cells and humanized with adoptively transferred human immune cells. In humanized mice, JL5 additionally induces the infiltration of immune cells within the tumor microenvironment. Our studies show that the BMP signaling pathway is targetable in vivo and BMP receptor inhibitors can be developed as a therapeutic to treat cancer patients.


Subject(s)
Antineoplastic Agents/pharmacology , Bone Morphogenetic Protein Receptors/antagonists & inhibitors , Carcinoma, Non-Small-Cell Lung/drug therapy , Lung Neoplasms/drug therapy , Protein Kinase Inhibitors/pharmacology , Pyrazoles/pharmacology , Pyrimidines/pharmacology , Quinolones/pharmacology , Tumor Microenvironment/drug effects , A549 Cells , Adoptive Transfer , Animals , Antineoplastic Agents/chemistry , Carcinoma, Non-Small-Cell Lung/pathology , Cell Line, Tumor , Cell Survival/drug effects , Humans , Inhibitor of Differentiation Protein 1/biosynthesis , Lung Neoplasms/pathology , MAP Kinase Kinase Kinases/biosynthesis , Mice , Mice, Inbred NOD , Mice, SCID , Protein Kinase Inhibitors/chemistry , Pyrazoles/chemistry , Pyrazoles/therapeutic use , Pyrimidines/chemistry , Pyrimidines/therapeutic use , Quinolones/chemistry , Quinolones/therapeutic use , Signal Transduction/drug effects , Xenograft Model Antitumor Assays
4.
Target Oncol ; 12(4): 449-462, 2017 08.
Article in English | MEDLINE | ID: mdl-28589491

ABSTRACT

BACKGROUND: Hepatocellular carcinoma (HCC) represents one of the most lethal cancers worldwide due to therapy resistance and disease recurrence. Tumor relapse following treatment could be driven by the persistence of liver cancer stem-like cells (CSCs). The protein BMI1 is a member of the polycomb epigenetic factors governing cellular self-renewal, proliferation, and stemness maintenance. BMI1 expression also correlates with poor patient survival in various cancer types. OBJECTIVE: We aimed to elucidate the extent to which BMI1 can be used as a potential therapeutic target for CSC eradication in HCC. METHODS: We have recently participated in characterizing the first known pharmacological small molecule inhibitor of BMI1. Here, we synthesized a panel of novel BMI1 inhibitors and examined their ability to alter cellular growth and eliminate cancer progenitor/stem-like cells in HCC with different p53 backgrounds. RESULTS: Among various molecules examined, RU-A1 particularly downregulated BMI1 expression, impaired cell viability, reduced cell migration, and sensitized HCC cells to 5-fluorouracil (5-FU) in vitro. Notably, long-term analysis of HCC survival showed that, unlike chemotherapy, RU-A1 effectively reduced CSC content, even as monotherapy. BMI1 inhibition with RU-A1 diminished the number of stem-like cells in vitro more efficiently than the model compound C-209, as demonstrated by clonogenic assays and impairment of CSC marker expression. Furthermore, xenograft assays in zebrafish showed that RU-A1 abrogated tumor growth in vivo. CONCLUSIONS: This study demonstrates the ability to identify agents with the propensity for targeting CSCs in HCC that could be explored as novel treatments in the clinical setting.


Subject(s)
Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/pharmacology , Carcinoma, Hepatocellular/drug therapy , Liver Neoplasms/drug therapy , Polycomb Repressive Complex 1/antagonists & inhibitors , Small Molecule Libraries/chemical synthesis , Small Molecule Libraries/pharmacology , Animals , Antineoplastic Agents/chemistry , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , HEK293 Cells , Hep G2 Cells , Humans , Immunohistochemistry , Liver Neoplasms/genetics , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Polycomb Repressive Complex 1/biosynthesis , Polycomb Repressive Complex 1/genetics , Small Molecule Libraries/chemistry , Xenograft Model Antitumor Assays , Zebrafish
5.
Mol Pharmacol ; 87(5): 825-31, 2015 May.
Article in English | MEDLINE | ID: mdl-25710967

ABSTRACT

p53 is a Zn(2+)-dependent tumor suppressor inactivated in >50% of human cancers. The most common mutation, R175H, inactivates p53 by reducing its affinity for the essential zinc ion, leaving the mutant protein unable to bind the metal in the low [Zn(2+)]free environment of the cell. The exploratory cancer drug zinc metallochaperone-1 (ZMC1) was previously demonstrated to reactivate this and other Zn(2+)-binding mutants by binding Zn(2+) and buffering it to a level such that Zn(2+) can repopulate the defective binding site, but how it accomplishes this in the context of living cells and organisms is unclear. In this study, we demonstrated that ZMC1 increases intracellular [Zn(2+)]free by functioning as a Zn(2+) ionophore, binding Zn(2+) in the extracellular environment, diffusing across the plasma membrane, and releasing it intracellularly. It raises intracellular [Zn(2+)]free in cancer (TOV112D) and noncancer human embryonic kidney cell line 293 to 15.8 and 18.1 nM, respectively, with half-times of 2-3 minutes. These [Zn(2+)]free levels are predicted to result in ∼90% saturation of p53-R175H, thus accounting for its observed reactivation. This mechanism is supported by the X-ray crystal structure of the [Zn(ZMC1)2] complex, which demonstrates structural and chemical features consistent with those of known metal ionophores. These findings provide a physical mechanism linking zinc metallochaperone-1 in both in vitro and in vivo activities and define the remaining critical parameter necessary for developing synthetic metallochaperones for clinical use.


Subject(s)
Biological Transport/physiology , Carrier Proteins/metabolism , Ionophores/metabolism , Metallochaperones/metabolism , Tumor Suppressor Protein p53/metabolism , Zinc/metabolism , Binding Sites , Cell Line , Cell Membrane/metabolism , HEK293 Cells , Humans , Mutation/genetics , Protein Conformation , Tumor Suppressor Protein p53/genetics
6.
PLoS One ; 9(5): e98151, 2014.
Article in English | MEDLINE | ID: mdl-24852423

ABSTRACT

Mammalian sterile 20-like kinase 1 (Mst1) is a MAPK kinase kinase kinase which is involved in a wide range of cellular responses, including apoptosis, lymphocyte adhesion and trafficking. The contribution of Mst1 to Ag-specific immune responses and autoimmunity has not been well defined. In this study, we provide evidence for the essential role of Mst1 in T cell differentiation and autoimmunity, using both genetic and pharmacologic approaches. Absence of Mst1 in mice reduced T cell proliferation and IL-2 production in vitro, blocked cell cycle progression, and elevated activation-induced cell death in Th1 cells. Mst1 deficiency led to a CD4+ T cell development path that was biased toward Th2 and immunoregulatory cytokine production with suppressed Th1 responses. In addition, Mst1-/- B cells showed decreased stimulation to B cell mitogens in vitro and deficient Ag-specific Ig production in vivo. Consistent with altered lymphocyte function, deletion of Mst1 reduced the severity of experimental autoimmune encephalomyelitis (EAE) and protected against collagen-induced arthritis development. Mst1-/- CD4+ T cells displayed an intrinsic defect in their ability to respond to encephalitogenic antigens and deletion of Mst1 in the CD4+ T cell compartment was sufficient to alleviate CNS inflammation during EAE. These findings have prompted the discovery of novel compounds that are potent inhibitors of Mst1 and exhibit desirable pharmacokinetic properties. In conclusion, this report implicates Mst1 as a critical regulator of adaptive immune responses, Th1/Th2-dependent cytokine production, and as a potential therapeutic target for immune disorders.


Subject(s)
Autoimmunity , Hepatocyte Growth Factor/genetics , Proto-Oncogene Proteins/genetics , T-Lymphocytes/immunology , Animals , Arthritis, Rheumatoid/immunology , Base Sequence , DNA Primers , Lymphocyte Activation , Lymphocyte Culture Test, Mixed , Mice , Mice, Inbred BALB C , Reverse Transcriptase Polymerase Chain Reaction
7.
J Med Chem ; 53(24): 8650-62, 2010 Dec 23.
Article in English | MEDLINE | ID: mdl-21090716

ABSTRACT

Sphingosine 1-phosphate lyase (S1PL) has been characterized as a novel target for the treatment of autoimmune disorders using genetic and pharmacological methods. Medicinal chemistry efforts targeting S1PL by direct in vivo evaluation of synthetic analogues of 2-acetyl-4(5)-(1(R),2(S),3(R),4-tetrahydroxybutyl)-imidazole (THI, 1) led to the discovery of 2 (LX2931) and 4 (LX2932). The immunological phenotypes observed in S1PL deficient mice were recapitulated by oral administration of 2 or 4. Oral dosing of 2 or 4 yielded a dose-dependent decrease in circulating lymphocyte numbers in multiple species and showed a therapeutic effect in rodent models of rheumatoid arthritis (RA). Phase I clinical trials indicated that 2, the first clinically studied inhibitor of S1PL, produced a dose-dependent and reversible reduction of circulating lymphocytes and was well tolerated at dose levels of up to 180 mg daily. Phase II evaluation of 2 in patients with active rheumatoid arthritis is currently underway.


Subject(s)
Aldehyde-Lyases/antagonists & inhibitors , Antirheumatic Agents/chemical synthesis , Imidazoles/chemical synthesis , Isoxazoles/chemical synthesis , Oximes/chemical synthesis , Aldehyde-Lyases/genetics , Animals , Antirheumatic Agents/pharmacokinetics , Antirheumatic Agents/pharmacology , Arthritis, Experimental/drug therapy , Arthritis, Experimental/immunology , Arthritis, Experimental/pathology , Blood Pressure/drug effects , Cell Movement , Dogs , Heart Rate/drug effects , Imidazoles/pharmacokinetics , Imidazoles/pharmacology , Isoxazoles/pharmacokinetics , Isoxazoles/pharmacology , Lymphocytes/drug effects , Lymphocytes/physiology , Macaca fascicularis , Male , Mice , Mice, Inbred C57BL , Mice, Inbred DBA , Oximes/pharmacokinetics , Oximes/pharmacology , Rats , Rats, Sprague-Dawley , Stereoisomerism , Structure-Activity Relationship
8.
Bioorg Med Chem Lett ; 19(23): 6780-3, 2009 Dec 01.
Article in English | MEDLINE | ID: mdl-19836229

ABSTRACT

A series of potent piperidine-linked cytosine derivatives were prepared as inhibitors of deoxycytidine kinase (dCK). Compound 9h was discovered to be a potent inhibitor of dCK and shows a good combination of cellular potency and pharmacokinetic parameters. Compound 9h blocks the incorporation of radiolabeled cytosine into mouse T-cells in vitro, as well as in vivo in mice following a T-cell challenge.


Subject(s)
Deoxycytidine Kinase/antagonists & inhibitors , Flucytosine/pharmacology , Protein Kinase Inhibitors/pharmacology , Animals , Drug Design , Flucytosine/chemical synthesis , Flucytosine/chemistry , Humans , Mice , Molecular Structure , Protein Kinase Inhibitors/chemical synthesis , Protein Kinase Inhibitors/chemistry , Stereoisomerism , Structure-Activity Relationship
9.
Bioorg Med Chem Lett ; 19(23): 6784-7, 2009 Dec 01.
Article in English | MEDLINE | ID: mdl-19836232

ABSTRACT

A series of deoxycytidine kinase inhibitors was simultaneously optimized for potency and PK properties. A co-crystal structure then allowed merging this series with a high throughput screening hit to afford a highly potent, selective and orally bioavailable inhibitor, compound 10. This compound showed dose dependent inhibition of deoxycytidine kinase in vivo.


Subject(s)
Deoxycytidine Kinase/antagonists & inhibitors , Deoxycytidine/analogs & derivatives , Drug Design , Protein Kinase Inhibitors/pharmacology , Deoxycytidine/chemical synthesis , Deoxycytidine/chemistry , Deoxycytidine/pharmacology , Dose-Response Relationship, Drug , Models, Molecular , Molecular Structure , Protein Kinase Inhibitors/chemical synthesis , Protein Kinase Inhibitors/chemistry , Stereoisomerism , Structure-Activity Relationship
10.
J Med Chem ; 52(13): 3941-53, 2009 Jul 09.
Article in English | MEDLINE | ID: mdl-19489538

ABSTRACT

During nearly a decade of research dedicated to the study of sphingosine signaling pathways, we identified sphingosine-1-phosphate lyase (S1PL) as a drug target for the treatment of autoimmune disorders. S1PL catalyzes the irreversible decomposition of sphingosine-1-phosphate (S1P) by a retro-aldol fragmentation that yields hexadecanaldehyde and phosphoethanolamine. Genetic models demonstrated that mice expressing reduced S1PL activity had decreased numbers of circulating lymphocytes due to altered lymphocyte trafficking, which prevented disease development in multiple models of autoimmune disease. Mechanistic studies of lymphoid tissue following oral administration of 2-acetyl-4(5)-(1(R),2(S),3(R),4-tetrahydroxybutyl)-imidazole (THI) 3 showed a clear relationship between reduced lyase activity, elevated S1P levels, and lower levels of circulating lymphocytes. Our internal medicinal chemistry efforts discovered potent analogues of 3 bearing heterocycles as chemical equivalents of the pendant carbonyl present in the parent structure. Reduction of S1PL activity by oral administration of these analogues recapitulated the phenotype of mice with genetically reduced S1PL expression.


Subject(s)
Aldehyde-Lyases/antagonists & inhibitors , Autoimmune Diseases/drug therapy , Imidazoles/pharmacology , Administration, Oral , Animals , Enzyme Inhibitors/administration & dosage , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/therapeutic use , Imidazoles/administration & dosage , Imidazoles/therapeutic use , Lymphocyte Count , Mice , Structure-Activity Relationship
11.
Bioorg Med Chem Lett ; 17(23): 6476-80, 2007 Dec 01.
Article in English | MEDLINE | ID: mdl-17937986

ABSTRACT

The synthesis and structure-activity relationships of novel dipeptidyl peptidase IV inhibitors replacing the classical cyanopyrrolidine P1 group with other small nitrogen heterocycles are described. A unique potency enhancement was achieved with beta-branched natural and unnatural amino acids, particularly adamantylglycines, linked to a (2S,3R)-2,3-methanopyrrolidine based scaffold.


Subject(s)
Dipeptides/chemistry , Dipeptidyl-Peptidase IV Inhibitors , Dipeptidyl-Peptidase IV Inhibitors/chemistry , Dipeptides/pharmacology , Dipeptidyl-Peptidase IV Inhibitors/pharmacology , Drug Evaluation, Preclinical , Humans , Nitriles/chemistry , Nitriles/pharmacology , Structure-Activity Relationship
12.
Bioorg Med Chem Lett ; 17(7): 1860-4, 2007 Apr 01.
Article in English | MEDLINE | ID: mdl-17292608

ABSTRACT

Pharmacokinetic studies in cynomolgus monkeys with a novel prototype selective androgen receptor modulator revealed trace amounts of an aniline fragment released through hydrolytic metabolism. This aniline fragment was determined to be mutagenic in an Ames assay. Subsequent concurrent optimization for target activity and avoidance of mutagenicity led to the identification of a pharmacologically superior clinical candidate without mutagenic potential.


Subject(s)
Androgen Antagonists/chemistry , Androgen Antagonists/chemical synthesis , Chemistry, Pharmaceutical/methods , Hydantoins/chemistry , Hydantoins/chemical synthesis , Receptors, Androgen/metabolism , Androgen Antagonists/pharmacology , Animals , Drug Design , Escherichia coli/metabolism , Genes, Reporter , Kinetics , Macaca fascicularis , Models, Chemical , Molecular Conformation , Mutagenesis , Mutagens , Structure-Activity Relationship
13.
J Med Chem ; 49(2): 656-63, 2006 Jan 26.
Article in English | MEDLINE | ID: mdl-16420051

ABSTRACT

The antiapoptotic proteins Bcl-x(L) and Bcl-2 play key roles in the maintenance of normal cellular homeostasis. However, their overexpression can lead to oncogenic transformation and is responsible for drug resistance in certain types of cancer. This makes Bcl-x(L) and Bcl-2 attractive targets for the development of potential anticancer agents. Here we describe the structure-based discovery of a potent Bcl-x(L) inhibitor directed at a hydrophobic groove on the surface of the protein. This groove represents the binding site for BH3 peptides from proapoptotic Bcl-2 family members such as Bak and Bad. Application of NMR-based screening yielded an initial biaryl acid with an affinity (K(d)) of approximately 300 microM for the protein. Following the classical "SAR by NMR" approach, a second-site ligand was identified that bound proximal to the first-site ligand in the hydrophobic groove. From NMR-based structural studies and parallel synthesis, a potent ligand was obtained, which binds to Bcl-x(L) with an inhibition constant (K(i)) of 36 +/- 2 nM.


Subject(s)
Aniline Compounds/chemical synthesis , Models, Molecular , Sulfonamides/chemical synthesis , bcl-X Protein/antagonists & inhibitors , Aniline Compounds/chemistry , Binding Sites , Hydrophobic and Hydrophilic Interactions , Ligands , Magnetic Resonance Spectroscopy , Protein Binding , Solubility , Structure-Activity Relationship , Sulfonamides/chemistry , bcl-X Protein/chemistry
14.
J Med Chem ; 48(15): 5025-37, 2005 Jul 28.
Article in English | MEDLINE | ID: mdl-16033281

ABSTRACT

Efforts to further elucidate structure-activity relationships (SAR) within our previously disclosed series of beta-quaternary amino acid linked l-cis-4,5-methanoprolinenitrile dipeptidyl peptidase IV (DPP-IV) inhibitors led to the investigation of vinyl substitution at the beta-position of alpha-cycloalkyl-substituted glycines. Despite poor systemic exposure, vinyl-substituted compounds showed extended duration of action in acute rat ex vivo plasma DPP-IV inhibition models. Oxygenated putative metabolites were prepared and were shown to exhibit the potency and extended duration of action of their precursors in efficacy models measuring glucose clearance in Zucker(fa/fa) rats. Extension of this approach to adamantylglycine-derived inhibitors led to the discovery of highly potent inhibitors, including hydroxyadamantyl compound BMS-477118 (saxagliptin), a highly efficacious, stable, and long-acting DPP-IV inhibitor, which is currently undergoing clinical trials for treatment of type 2 diabetes.


Subject(s)
Adamantane/analogs & derivatives , Adamantane/chemical synthesis , Diabetes Mellitus, Type 2/drug therapy , Dipeptides/chemical synthesis , Dipeptidyl Peptidase 4/metabolism , Glycine/analogs & derivatives , Glycine/chemical synthesis , Hypoglycemic Agents/chemical synthesis , Protease Inhibitors/chemical synthesis , Adamantane/pharmacology , Animals , Biological Availability , Blood Glucose/analysis , Diabetes Mellitus, Type 2/physiopathology , Dipeptides/pharmacology , Glucose Tolerance Test , Glycine/pharmacology , Humans , Hypoglycemic Agents/pharmacology , In Vitro Techniques , Insulin/blood , Male , Mice , Mice, Obese , Microsomes, Liver/metabolism , Nitriles/chemical synthesis , Nitriles/pharmacology , Proline/analogs & derivatives , Proline/chemical synthesis , Proline/pharmacology , Protease Inhibitors/pharmacology , Rats , Rats, Zucker , Stereoisomerism , Structure-Activity Relationship
15.
Nature ; 435(7042): 677-81, 2005 Jun 02.
Article in English | MEDLINE | ID: mdl-15902208

ABSTRACT

Proteins in the Bcl-2 family are central regulators of programmed cell death, and members that inhibit apoptosis, such as Bcl-X(L) and Bcl-2, are overexpressed in many cancers and contribute to tumour initiation, progression and resistance to therapy. Bcl-X(L) expression correlates with chemo-resistance of tumour cell lines, and reductions in Bcl-2 increase sensitivity to anticancer drugs and enhance in vivo survival. The development of inhibitors of these proteins as potential anti-cancer therapeutics has been previously explored, but obtaining potent small-molecule inhibitors has proved difficult owing to the necessity of targeting a protein-protein interaction. Here, using nuclear magnetic resonance (NMR)-based screening, parallel synthesis and structure-based design, we have discovered ABT-737, a small-molecule inhibitor of the anti-apoptotic proteins Bcl-2, Bcl-X(L) and Bcl-w, with an affinity two to three orders of magnitude more potent than previously reported compounds. Mechanistic studies reveal that ABT-737 does not directly initiate the apoptotic process, but enhances the effects of death signals, displaying synergistic cytotoxicity with chemotherapeutics and radiation. ABT-737 exhibits single-agent-mechanism-based killing of cells from lymphoma and small-cell lung carcinoma lines, as well as primary patient-derived cells, and in animal models, ABT-737 improves survival, causes regression of established tumours, and produces cures in a high percentage of the mice.


Subject(s)
Antineoplastic Agents/therapeutic use , Biphenyl Compounds/pharmacology , Biphenyl Compounds/therapeutic use , Neoplasms/drug therapy , Neoplasms/pathology , Proto-Oncogene Proteins c-bcl-2/antagonists & inhibitors , Proto-Oncogene Proteins c-bcl-2/classification , Animals , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Biphenyl Compounds/chemical synthesis , Biphenyl Compounds/chemistry , Carcinoma, Small Cell/drug therapy , Carcinoma, Small Cell/pathology , Cell Line, Tumor , Cytochromes c/metabolism , Disease Models, Animal , Drug Synergism , Humans , Lymphoma/drug therapy , Lymphoma/pathology , Magnetic Resonance Spectroscopy , Mice , Mitochondria/drug effects , Mitochondria/metabolism , Models, Molecular , Nitrophenols , Paclitaxel/pharmacology , Piperazines , Proto-Oncogene Proteins c-bcl-2/metabolism , Structure-Activity Relationship , Sulfonamides , Survival Rate
16.
J Med Chem ; 47(10): 2587-98, 2004 May 06.
Article in English | MEDLINE | ID: mdl-15115400

ABSTRACT

A series of methanoprolinenitrile-containing dipeptide mimetics were synthesized and assayed as inhibitors of the N-terminal sequence-specific serine protease dipeptidyl peptidase IV (DPP-IV). The catalytic action of DPP-IV is the principle means of degradation of glucagon-like peptide-1, a key mediator of glucose-stimulated insulin secretion, and DPP-IV inhibition shows clinical benefit as a novel mechanism for treatment of type 2 diabetes. However, many of the reversible inhibitors to date suffer from chemical instability stemming from an amine to nitrile intramolecular cyclization. Installation of a cyclopropyl moiety at either the 3,4- or 4,5-position of traditional 2-cyanopyrrolidide proline mimetics led to compounds with potent inhibitory activity against the enzyme. Additionally, cis-4,5-methanoprolinenitriles with beta-branching in the N-terminal amino acid provided enhanced chemical stability and high inhibitory potency. This class of inhibitors also exhibited the ability to suppress prandial glucose elevations after an oral glucose challenge in male Zucker rats.


Subject(s)
Cyclopropanes/chemical synthesis , Dipeptidyl Peptidase 4/metabolism , Enzyme Inhibitors/chemical synthesis , Nitriles/chemical synthesis , Proline/analogs & derivatives , Proline/chemical synthesis , Animals , Computer Simulation , Crystallography, X-Ray , Cyclopropanes/chemistry , Cyclopropanes/pharmacology , Dipeptides/chemistry , Dipeptidyl Peptidase 4/chemistry , Drug Stability , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Hypoglycemic Agents/chemical synthesis , Hypoglycemic Agents/chemistry , Hypoglycemic Agents/pharmacology , Male , Models, Molecular , Molecular Conformation , Molecular Mimicry , Nitriles/chemistry , Nitriles/pharmacology , Proline/chemistry , Proline/pharmacology , Rats , Rats, Zucker , Solutions
17.
J Med Chem ; 45(26): 5628-39, 2002 Dec 19.
Article in English | MEDLINE | ID: mdl-12477346

ABSTRACT

The NMR-based discovery of biaryl hydroxamate inhibitors of the matrix metalloproteinase stromelysin (MMP-3) has been previously described (Hajduk et al. J. Am. Chem. Soc. 1997, 119, 5818-5827). While potent in vitro, these inhibitors exhibited no in vivo activity due, at least in part, to the poor pharmacokinetic properties of the alkylhydroxamate moiety. To circumvent this liability, NMR-based screening was implemented to identify alternative zinc-chelating groups. Using this technique, 1-naphthyl hydroxamate was found to bind tightly to the protein (K(D) = 50 microM) and was identified as a candidate for incorporation into the lead series. On the basis of NMR-derived structural information, the naphthyl hydroxamate and biaryl fragments were linked together to yield inhibitors of this enzyme that exhibited improved bioavailability. These studies demonstrate that the NMR-based screening of fragments can be effectively applied to improve the physicochemical or pharmacokinetic profile of lead compounds.


Subject(s)
Hydroxamic Acids/chemical synthesis , Matrix Metalloproteinase Inhibitors , Protease Inhibitors/chemical synthesis , Animals , Biological Availability , Catalytic Domain , Hydroxamic Acids/chemistry , Hydroxamic Acids/pharmacology , Magnetic Resonance Spectroscopy , Matrix Metalloproteinase 3/chemistry , Models, Molecular , Naphthalenes/chemical synthesis , Naphthalenes/chemistry , Naphthalenes/pharmacology , Protease Inhibitors/chemistry , Protease Inhibitors/pharmacokinetics , Protein Binding , Rats , Structure-Activity Relationship
SELECTION OF CITATIONS
SEARCH DETAIL