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










Publication year range
1.
Inflammopharmacology ; 28(2): 499-511, 2020 Apr.
Article in English | MEDLINE | ID: mdl-31549280

ABSTRACT

INTRODUCTION: Retinoic Acid Related Orphan Nuclear Receptor gamma T (RORγT) is a lineage specifying transcription factor for IL-17 expressing cells, which may contribute to the pathogenesis of Inflammatory Bowel Disease (IBD). VPR-254 is a selective in vitro inhibitor of RORγT. AIMS: The main goals of our study were twofold: (1) To determine if ex vivo treatment with VPR-254 reduced relevant cytokine (IL-17 and IL-21) secretion from colonic strips of mice with colitis; (2) To determine if treatment of mice with VPR-254 attenuated parameters of colitis, using three murine IBD models. METHODS: VPR-254 was evaluated ex vivo in a colonic strip assay, using tissue from mice with Dextran sulfate sodium (DSS)-induced colitis. In vivo, VPR-254 was evaluated for efficacy in DSS, Trintirobenzenesulfonic acid (TNBS) and Anti-CD40 antibody-induced murine models of colitis. RESULTS: VPR-254 reduced the production of key pro-inflammatory cytokines (e.g., IL-17) in ex vivo and in vivo models of colitis. This small molecule inhibitor of RORγT also improved various morphometric and histological parameters associated with three diverse murine models of IBD. CONCLUSION: Our results support the concept that an inhibitor of ROR-gamma T may have potential utility for the treatment of IBD.


Subject(s)
Cytokines/metabolism , Inflammatory Bowel Diseases/drug therapy , Nuclear Receptor Subfamily 1, Group F, Member 3/antagonists & inhibitors , Adult , Animals , CHO Cells , Cricetulus , Dextran Sulfate , Disease Models, Animal , Female , Humans , Inflammatory Bowel Diseases/physiopathology , Interleukin-17/metabolism , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, SCID
3.
J Med Chem ; 54(24): 8490-500, 2011 Dec 22.
Article in English | MEDLINE | ID: mdl-22040023

ABSTRACT

Analogues substituted with various amines at the 6-position of the pyrazine ring on (4-amino-7-isopropyl-7H-pyrrolo[2,3-d]pyrimidin-5-yl)pyrazin-2-ylmethanone were discovered as potent and selective inhibitors of PDK1 with potential as anticancer agents. An early lead with 2-pyridine-3-ylethylamine as the pyrazine substituent showed moderate potency and selectivity. Structure-based drug design led to improved potency and selectivity against PI3Kα through a combination of cyclizing the ethylene spacer into a saturated, five-membered ring and substituting on the 4-position of the aryl ring with a fluorine. ADME properties were improved by lowering the lipophilicity with heteroatom replacements in the saturated, five-membered ring. The optimized analogues have a PDK1 Ki of 1 nM and >100-fold selectivity against PI3K/AKT-pathway kinases. The cellular potency of these analogues was assessed by the inhibition of AKT phosphorylation (T308) and by their antiproliferation activity against a number of tumor cell lines.


Subject(s)
Antineoplastic Agents/chemical synthesis , Protein Serine-Threonine Kinases/antagonists & inhibitors , Pyridines/chemical synthesis , Pyrroles/chemical synthesis , 3-Phosphoinositide-Dependent Protein Kinases , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Crystallography, X-Ray , Drug Screening Assays, Antitumor , Ethylamines/chemical synthesis , Ethylamines/chemistry , Ethylamines/pharmacology , Humans , Models, Molecular , Phosphorylation , Protein Conformation , Proto-Oncogene Proteins c-akt/metabolism , Pyridines/chemistry , Pyridines/pharmacology , Pyrroles/chemistry , Pyrroles/pharmacology , Signal Transduction , Structure-Activity Relationship
4.
J Med Chem ; 54(18): 6342-63, 2011 Sep 22.
Article in English | MEDLINE | ID: mdl-21812414

ABSTRACT

Because of the critical roles of aberrant signaling in cancer, both c-MET and ALK receptor tyrosine kinases are attractive oncology targets for therapeutic intervention. The cocrystal structure of 3 (PHA-665752), bound to c-MET kinase domain, revealed a novel ATP site environment, which served as the target to guide parallel, multiattribute drug design. A novel 2-amino-5-aryl-3-benzyloxypyridine series was created to more effectively make the key interactions achieved with 3. In the novel series, the 2-aminopyridine core allowed a 3-benzyloxy group to reach into the same pocket as the 2,6-dichlorophenyl group of 3 via a more direct vector and thus with a better ligand efficiency (LE). Further optimization of the lead series generated the clinical candidate crizotinib (PF-02341066), which demonstrated potent in vitro and in vivo c-MET kinase and ALK inhibition, effective tumor growth inhibition, and good pharmaceutical properties.


Subject(s)
Proto-Oncogene Proteins c-met/antagonists & inhibitors , Pyrazoles/chemical synthesis , Pyridines/chemical synthesis , Receptor Protein-Tyrosine Kinases/antagonists & inhibitors , Anaplastic Lymphoma Kinase , Cell Line, Tumor , Cell Proliferation/drug effects , Crizotinib , Crystallography, X-Ray , Drug Design , Drug Screening Assays, Antitumor , Epithelial-Mesenchymal Transition , Humans , Indoles/chemical synthesis , Indoles/chemistry , Indoles/pharmacology , Models, Molecular , Molecular Conformation , Pyrazoles/chemistry , Pyrazoles/pharmacology , Pyridines/chemistry , Pyridines/pharmacology , Stereoisomerism , Structure-Activity Relationship
5.
J Comput Aided Mol Des ; 25(7): 689-98, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21779981

ABSTRACT

Phosphoinositide-dependent kinase-1 (PDK1) is a critical enzyme in the PI3K/AKT pathway and to the activation of AGC family protein kinases, including S6K, SGK, and PKC. Dysregulation of this pathway plays a key role in cancer cell growth, survival and tumor angiogenesis. As such, inhibitors of PDK1 offer the promise of a new therapeutic modality for cancer treatment. Fragment based drug screening has recently become a viable entry point for hit identification. In this work, NMR spectroscopy fragment screening of PDK1 afforded novel chemotypes as orthogonal starting points from HTS screening hits. Compounds identified as hits by NMR spectroscopy were tested in a biochemical assay, and fragments with activity in both assays were clustered. The Pfizer compound file was mined via substructure and 2D similarity search, and the chemotypes were prioritized by ligand efficiency (LE), SAR mining, chemical attractiveness, and chemical enablement of promising vectors. From this effort, an isoquinolone fragment hit, 5 (IC(50) 870 µM, LE = 0.39), was identified as a novel, ligand efficient inhibitor of PDK1 and a suitable scaffold for further optimization. Initially in the absence of crystallographic data, a fragment growing approach efficiently explored four vectors of the isoquinolone scaffold via parallel synthesis to afford a compound with crystallographic data, 16 (IC(50) 41.4 µM, LE = 0.33). Subsequent lead optimization efforts provided 24 (IC(50) 1.8 µM, LE = 0.42), with greater than fivefold selectivity against other key pathway kinases.


Subject(s)
Drug Discovery/methods , Drug Evaluation, Preclinical/methods , Protein Serine-Threonine Kinases/antagonists & inhibitors , Binding Sites , Crystallography, X-Ray , High-Throughput Screening Assays , Humans , Hydrogen Bonding , Ligands , Magnetic Resonance Imaging , Peptide Fragments/chemistry , Protein Binding , Protein Serine-Threonine Kinases/chemistry , Pyruvate Dehydrogenase Acetyl-Transferring Kinase
6.
Anal Biochem ; 414(2): 179-86, 2011 Jul 15.
Article in English | MEDLINE | ID: mdl-21402045

ABSTRACT

The PI3K/AKT signaling pathway has an important regulatory role in cancer cell growth and tumorigenesis. Signal transduction through this pathway requires the assembly and activation of PDK1 and AKT at the plasma membrane. On activation of the pathway, PDK1 and AKT1/2 translocate to the membrane and bind to phosphatidylinositol-(3,4,5)-trisphosphate (PIP(3)) through interaction with their pleckstrin-homology domains. A biochemical method was developed to measure the kinase activity of PDK1 and AKT1/2, utilizing nickel-chelating coated lipid vesicles as a way to mimic the membrane environment. The presence of these vesicles in the reaction buffer enhanced the specific activity of the His-tagged PDK1 (full-length, and the truncated kinase domain) and the activity of the full-length His-tagged AKT1 and AKT2 when assayed in a cascade-type reaction. This enhanced biochemical assay is also suitable for measuring the inhibition of PDK1 by several selective compounds from the carbonyl-4-amino-pyrrolopyrimidine (CAP) series. One of these inhibitors, PF-5168899, was further evaluated using a high content cell-based assay in the presence of CHO cells engineered with GFP-PDK1.


Subject(s)
Adenine/analogs & derivatives , Enzyme Assays/methods , Protein Kinase Inhibitors/pharmacology , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Pyrazines/pharmacology , Signal Transduction , 3-Phosphoinositide-Dependent Protein Kinases , Adenine/chemistry , Adenine/pharmacology , Animals , CHO Cells , Cricetinae , Cricetulus , Histidine/genetics , Histidine/metabolism , Humans , Kinetics , Oligopeptides/genetics , Oligopeptides/metabolism , Phosphorylation , Protein Kinase Inhibitors/chemistry , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/genetics , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Proto-Oncogene Proteins c-akt/genetics , Pyrazines/chemistry , Recombinant Proteins/antagonists & inhibitors , Recombinant Proteins/genetics , Recombinant Proteins/metabolism
7.
Bioorg Med Chem Lett ; 21(2): 849-52, 2011 Jan 15.
Article in English | MEDLINE | ID: mdl-21185721

ABSTRACT

S6K1 (p70 S6 kinase-1) is thought to play a critical role in the development of obesity and insulin resistance, thus making it an attractive target in developing medicines for the treatment of these disorders. We describe a novel thiophene urea class of S6K inhibitors. The lead matter for the development of these inhibitors came from mining the literature for reports of weak off-target S6K activity. These optimized inhibitors exhibit good potency and excellent selectivity for S6K over a panel of 43 kinases.


Subject(s)
Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/pharmacology , Ribosomal Protein S6 Kinases, 70-kDa/antagonists & inhibitors , Thiophenes/chemistry , Thiophenes/pharmacology , Humans , Microsomes, Liver/metabolism , Protein Kinase Inhibitors/metabolism , Ribosomal Protein S6 Kinases, 70-kDa/metabolism , Structure-Activity Relationship , Thiophenes/metabolism , Urea/chemistry , Urea/metabolism , Urea/pharmacology
8.
Bioorg Med Chem Lett ; 21(1): 584-7, 2011 Jan 01.
Article in English | MEDLINE | ID: mdl-21084194

ABSTRACT

A novel series of pyrrolopyrazole-based protein kinase C ß II inhibitors has been identified from high-throughput screening. Herein, we report our initial structure-activity relationship studies with a focus on optimizing compound ligand efficiency and physicochemical properties, which has led to potent inhibitors with good cell permeability.


Subject(s)
Protein Kinase C/antagonists & inhibitors , Protein Kinase Inhibitors/chemistry , Pyrazoles/chemistry , High-Throughput Screening Assays , Protein Kinase C/metabolism , Protein Kinase C beta , Protein Kinase Inhibitors/chemical synthesis , Protein Kinase Inhibitors/pharmacology , Pyrazoles/chemical synthesis , Pyrazoles/pharmacology , Structure-Activity Relationship
9.
Biochem J ; 431(2): 245-55, 2010 Oct 15.
Article in English | MEDLINE | ID: mdl-20704563

ABSTRACT

S6K1 (p70 ribosomal S6 kinase 1) is activated by insulin and growth factors via the PI3K (phosphoinositide 3-kinase) and mTOR (mammalian target of rapamycin) signalling pathways. S6K1 regulates numerous processes, such as protein synthesis, growth, proliferation and longevity, and its inhibition has been proposed as a strategy for the treatment of cancer and insulin resistance. In the present paper we describe a novel cell-permeable inhibitor of S6K1, PF-4708671, which specifically inhibits the S6K1 isoform with a Ki of 20 nM and IC50 of 160 nM. PF-4708671 prevents the S6K1-mediated phosphorylation of S6 protein in response to IGF-1 (insulin-like growth factor 1), while having no effect upon the PMA-induced phosphorylation of substrates of the highly related RSK (p90 ribosomal S6 kinase) and MSK (mitogen- and stress-activated kinase) kinases. PF-4708671 was also found to induce phosphorylation of the T-loop and hydrophobic motif of S6K1, an effect that is dependent upon mTORC1 (mTOR complex 1). PF-4708671 is the first S6K1-specific inhibitor to be reported and will be a useful tool for delineating S6K1-specific roles downstream of mTOR.


Subject(s)
Imidazoles/pharmacology , Piperazines/pharmacology , Protein Kinase Inhibitors/pharmacology , Ribosomal Protein S6 Kinases, 70-kDa/antagonists & inhibitors , Cell Line , Humans , Imidazoles/chemistry , Mechanistic Target of Rapamycin Complex 1 , Multiprotein Complexes , Phosphorylation/drug effects , Phosphothreonine/metabolism , Piperazines/chemistry , Protein Kinase Inhibitors/chemistry , Proteins , Ribosomal Protein S6 Kinases, 70-kDa/metabolism , Ribosomal Protein S6 Kinases, 90-kDa/metabolism , Substrate Specificity/drug effects , TOR Serine-Threonine Kinases , Transcription Factors/metabolism
10.
Invest Ophthalmol Vis Sci ; 51(4): 2158-64, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20071676

ABSTRACT

Purpose. p38 mitogen-activated protein kinase (MAPK) is known to play a regulatory role in inflammatory processes in disease. Inflammation has been linked also to the development of diabetic retinopathy in rodents. This study was conducted to evaluate the effect of a p38 MAPK inhibitor on the development of early stages of diabetic retinopathy in rats. Methods. Streptozotocin-diabetic rats were assigned to two groups-treated with the p38 MAPK inhibitor PHA666859 (Pfizer, New York, NY) and untreated-and compared with age-matched nondiabetic control animals. Results. At 2 months of diabetes, insulin-deficient diabetic control rats exhibited significant increases in retinal superoxide, nitric oxide (NO), cyclooxygenase (COX)-2, and leukostasis within retinal microvessels. All these abnormalities were significantly inhibited by the p38 MAPK inhibitor (25 mg/kgBW/d). At 10 months of diabetes, significant increases in the number of degenerate (acellular) capillaries and pericyte ghosts were measured in control diabetic rats versus those in nondiabetic control animals, and pharmacologic inhibition of p38 MAPK significantly inhibited all these abnormalities (all P < 0.05). This therapy also had beneficial effects outside the eye in diabetes, as evidenced by the inhibition of a diabetes-induced hypersensitivity of peripheral nerves to light touch (tactile allodynia). Conclusions. p38 MAPK plays an important role in diabetes-induced inflammation in the retina, and inhibition of p38 MAPK offers a novel therapeutic approach to inhibiting the development of early stages of diabetic retinopathy and other complications of diabetes.


Subject(s)
Diabetic Retinopathy/prevention & control , Enzyme Inhibitors/pharmacology , Hyperalgesia/drug therapy , Pyrazoles/pharmacology , Pyrimidines/pharmacology , Sensory Receptor Cells/drug effects , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors , Animals , Blotting, Western , Cyclooxygenase 2 , Diabetes Mellitus, Experimental/enzymology , Diabetes Mellitus, Experimental/prevention & control , Diabetic Retinopathy/enzymology , Endothelium, Vascular/drug effects , Endothelium, Vascular/metabolism , Leukostasis/drug therapy , Male , Nitric Oxide/metabolism , Pericytes/drug effects , Pericytes/metabolism , Rats , Rats, Inbred Lew , Retinal Vessels/pathology , Superoxides/metabolism , Touch
11.
Biochemistry ; 48(41): 9823-30, 2009 Oct 20.
Article in English | MEDLINE | ID: mdl-19743875

ABSTRACT

Checkpoint kinase 1 (CHK1) is a key element in the DNA damage response pathway and plays a crucial role in the S-G(2)-phase checkpoint. Inhibiting CHK1 is a therapeutic strategy involving abrogation of the G2/M mitotic checkpoint defense of tumor cells toward lethal damage induced by DNA-directed chemotherapeutic agents. To date, most CHK1 inhibition approaches have involved targeting the ATP site of this kinase. In this study, we provide crystallographic and kinetic characterization of two small molecule inhibitors that bind to an allosteric site in the proximity of the CHK1 substrate site. Analysis of kinetic and biophysical data has led to the conclusion that these small molecule allosteric site inhibitors of CHK1 are reversible and are neither ATP- nor peptide substrate-competitive. K(i) values of 1.89 and 0.15 microM, respectively, have been determined for these compounds using a mixed inhibitor kinetic analysis. Cocrystal structures of the inhibitors bound to CHK1 reveal an allosteric site, unique to CHK1, located in the C-terminal domain and consisting of a shallow groove linked to a small hydrophobic pocket. The pocket displays induced fit characteristics in the presence of the two inhibitors. These findings establish the potential for the development of highly selective CHK1 inhibitors.


Subject(s)
Protein Kinases/metabolism , Allosteric Regulation , Binding Sites , Catalytic Domain , Checkpoint Kinase 1 , Cloning, Molecular , Crystallography, X-Ray , Enzyme Inhibitors/pharmacology , Humans , Kinetics , Models, Molecular , Peptide Fragments/chemistry , Protein Conformation , Protein Kinases/chemistry , Protein Kinases/genetics , Surface Plasmon Resonance
12.
Biochemistry ; 48(23): 5339-49, 2009 Jun 16.
Article in English | MEDLINE | ID: mdl-19459657

ABSTRACT

The c-Met receptor tyrosine kinase (RTK) is a key regulator in cancer, in part, through oncogenic mutations. Eight clinically relevant mutants were characterized by biochemical, biophysical, and cellular methods. The c-Met catalytic domain was highly active in the unphosphorylated state (k(cat) = 1.0 s(-1)) and achieved 160-fold enhanced catalytic efficiency (k(cat)/K(m)) upon activation to 425000 s(-1) M(-1). c-Met mutants had 2-10-fold higher basal enzymatic activity (k(cat)) but achieved maximal activities similar to those of wild-type c-Met, except for Y1235D, which underwent a reduction in maximal activity. Small enhancements of basal activity were shown to have profound effects on the acquisition of full enzymatic activity achieved through accelerating rates of autophosphorylation. Biophysical analysis of c-Met mutants revealed minimal melting temperature differences indicating that the mutations did not alter protein stability. A model of RTK activation is proposed to describe how a RTK response may be matched to a biological context through enzymatic properties. Two c-Met clinical candidates from aminopyridine and triazolopyrazine chemical series (PF-02341066 and PF-04217903) were studied. Biochemically, each series produced molecules that are highly selective against a large panel of kinases, with PF-04217903 (>1000-fold selective relative to 208 kinases) being more selective than PF-02341066. Although these prototype inhibitors have similar potencies against wild-type c-Met (K(i) = 6-7 nM), significant differences in potency were observed for clinically relevant mutations evaluated in both biochemical and cellular contexts. In particular, PF-02341066 was 180-fold more active against the Y1230C mutant c-Met than PF-04217903. These highly optimized inhibitors indicate that for kinases susceptible to active site mutations, inhibitor design may need to balance overall kinase selectivity with the ability to inhibit multiple mutant forms of the kinase (penetrance).


Subject(s)
Aminopyridines/chemistry , Mutation , Protein Kinase Inhibitors/chemistry , Proto-Oncogene Proteins c-met/chemistry , Pyrazines/chemistry , Aminopyridines/pharmacology , Binding Sites , Catalysis , Humans , Kinetics , Phosphorylation , Protein Conformation , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-met/antagonists & inhibitors , Proto-Oncogene Proteins c-met/genetics , Pyrazines/pharmacology
13.
Expert Opin Drug Discov ; 3(6): 595-605, 2008 Jun.
Article in English | MEDLINE | ID: mdl-23506143

ABSTRACT

BACKGROUND: The number of drugs in active clinical development or on the market that target the unactivated conformational states of protein kinases is growing and represents a significant portion of kinase research at biopharmaceutical companies. These non-classical kinase inhibitors have a mode of action which may overcome some of the liabilities of classical ATP-site inhibitors that substantially overlap the space that ATP occupies in the activated kinase. OBJECTIVE: This review will discuss state-of-the-art methods of inhibiting protein kinases by targeting the unactivated conformations of the enzyme with small molecules directed to the ATP binding region. METHODS: Biochemical and structural biology publications and public domain crystal structures were evaluated to identify key concepts in drug discovery for unactivated protein kinase inhibitors that target the ATP binding region. CONCLUSION: The potential for enhanced selectivity, potency and duration of pharmacological action may allow non-classical kinase therapeutics to be used for chronic dosing in non-life-threatening indications. Moreover, by targeting additional conformational space on the kinase protein it is possible that new chemical matter will be discovered such that current intellectual property limitations on traditional ATP-site chemical scaffolds may be circumvented.

14.
Eur J Med Chem ; 43(6): 1321-9, 2008 Jun.
Article in English | MEDLINE | ID: mdl-17964000

ABSTRACT

A series of quinoline-containing c-Met inhibitors were prepared and studied. Chemistry was developed to introduce a pyridyl moiety onto the 2-aryl ring present in a lead molecule which mitigated the potential for quinone formation relative to the original compound. The study also assessed the importance of an acylthiourea moiety present in the lead structure for effective binding to the c-Met protein ATP site.


Subject(s)
Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-met/antagonists & inhibitors , Quinolines/pharmacology , Magnetic Resonance Spectroscopy , Mass Spectrometry , Structure-Activity Relationship
15.
Mol Cancer Ther ; 6(12 Pt 1): 3314-22, 2007 Dec.
Article in English | MEDLINE | ID: mdl-18089725

ABSTRACT

A t(2;5) chromosomal translocation resulting in expression of an oncogenic kinase fusion protein known as nucleophosmin-anaplastic lymphoma kinase (NPM-ALK) has been implicated in the pathogenesis of anaplastic large-cell lymphoma (ALCL). PF-2341066 was recently identified as a p.o. bioavailable, small-molecule inhibitor of the catalytic activity of c-Met kinase and the NPM-ALK fusion protein. PF-2341066 also potently inhibited NPM-ALK phosphorylation in Karpas299 or SU-DHL-1 ALCL cells (mean IC(50) value, 24 nmol/L). In biochemical and cellular screens, PF-2341066 was shown to be selective for c-Met and ALK at pharmacologically relevant concentrations across a panel of >120 diverse kinases. PF-2341066 potently inhibited cell proliferation, which was associated with G(1)-S-phase cell cycle arrest and induction of apoptosis in ALK-positive ALCL cells (IC(50) values, approximately 30 nmol/L) but not ALK-negative lymphoma cells. The induction of apoptosis was confirmed using terminal deoxyribonucleotide transferase-mediated nick-end labeling and Annexin V staining (IC(50) values, 25-50 nmol/L). P.o. administration of PF-2341066 to severe combined immunodeficient-Beige mice bearing Karpas299 ALCL tumor xenografts resulted in dose-dependent antitumor efficacy with complete regression of all tumors at the 100 mg/kg/d dose within 15 days of initial compound administration. A strong correlation was observed between antitumor response and inhibition of NPM-ALK phosphorylation and induction of apoptosis in tumor tissue. In addition, inhibition of key NPM-ALK signaling mediators, including phospholipase C-gamma, signal transducers and activators of transcription 3, extracellular signal-regulated kinases, and Akt by PF-2341066 were observed at concentrations or dose levels, which correlated with inhibition of NPM-ALK phosphorylation and function. Collectively, these data illustrate the potential clinical utility of inhibitors of NPM-ALK in treatment of patients with ALK-positive ALCL.


Subject(s)
Antineoplastic Agents/pharmacology , Lymphoma, Large-Cell, Anaplastic/pathology , Piperidines/pharmacology , Protein Kinase Inhibitors/pharmacology , Protein-Tyrosine Kinases/antagonists & inhibitors , Proto-Oncogene Proteins c-met/antagonists & inhibitors , Pyridines/pharmacology , Anaplastic Lymphoma Kinase , Animals , Apoptosis/drug effects , Cell Line, Tumor , Crizotinib , Drug Screening Assays, Antitumor , Enzyme-Linked Immunosorbent Assay , Female , Humans , Mice , Mice, SCID , Phosphorylation , Protein-Tyrosine Kinases/metabolism , Pyrazoles , Receptor Protein-Tyrosine Kinases
16.
Cancer Res ; 67(9): 4408-17, 2007 May 01.
Article in English | MEDLINE | ID: mdl-17483355

ABSTRACT

The c-Met receptor tyrosine kinase and its ligand, hepatocyte growth factor (HGF), have been implicated in the progression of several human cancers and are attractive therapeutic targets. PF-2341066 was identified as a potent, orally bioavailable, ATP-competitive small-molecule inhibitor of the catalytic activity of c-Met kinase. PF-2341066 was selective for c-Met (and anaplastic lymphoma kinase) compared with a panel of >120 diverse tyrosine and serine-threonine kinases. PF-2341066 potently inhibited c-Met phosphorylation and c-Met-dependent proliferation, migration, or invasion of human tumor cells in vitro (IC(50) values, 5-20 nmol/L). In addition, PF-2341066 potently inhibited HGF-stimulated endothelial cell survival or invasion and serum-stimulated tubulogenesis in vitro, suggesting that this agent also exhibits antiangiogenic properties. PF-2341066 showed efficacy at well-tolerated doses, including marked cytoreductive antitumor activity, in several tumor models that expressed activated c-Met. The antitumor efficacy of PF-2341066 was dose dependent and showed a strong correlation to inhibition of c-Met phosphorylation in vivo. Near-maximal inhibition of c-Met activity for the full dosing interval was necessary to maximize the efficacy of PF-2341066. Additional mechanism-of-action studies showed dose-dependent inhibition of c-Met-dependent signal transduction, tumor cell proliferation (Ki67), induction of apoptosis (caspase-3), and reduction of microvessel density (CD31). These results indicated that the antitumor activity of PF-2341066 may be mediated by direct effects on tumor cell growth or survival as well as antiangiogenic mechanisms. Collectively, these results show the therapeutic potential of targeting c-Met with selective small-molecule inhibitors for the treatment of human cancers.


Subject(s)
Breast Neoplasms/drug therapy , Piperidines/pharmacology , Pyridines/pharmacology , Receptor Protein-Tyrosine Kinases/antagonists & inhibitors , Stomach Neoplasms/drug therapy , Angiogenesis Inhibitors/pharmacology , Animals , Breast Neoplasms/blood supply , Breast Neoplasms/enzymology , Breast Neoplasms/pathology , Cell Growth Processes/drug effects , Crizotinib , Dogs , Dose-Response Relationship, Drug , Endothelial Cells/drug effects , Female , Humans , Male , Mice , Mice, Nude , Neovascularization, Pathologic/drug therapy , Phosphorylation/drug effects , Protein Kinase Inhibitors/pharmacology , Pyrazoles , Receptor Protein-Tyrosine Kinases/metabolism , Signal Transduction/drug effects , Stomach Neoplasms/blood supply , Stomach Neoplasms/enzymology , Stomach Neoplasms/pathology , Xenograft Model Antitumor Assays
17.
Biochem Biophys Res Commun ; 357(2): 561-6, 2007 Jun 01.
Article in English | MEDLINE | ID: mdl-17434447

ABSTRACT

Assay conditions for the 11beta-hydroxysteroid dehydrogenase have been optimized by adding phospholipids in the media buffer to increase and stabilize the enzymatic activity. The presence of phospholipids greatly facilitates the study of the binding of cortisone and NADPH at the enzyme catalytic site. Kinetic analyses conducted with the human and rabbit enzyme isoforms suggest that both enzymes behave according to an ordered sequential bi-bi mechanism where the NADPH is the first to bind at the active site followed by cortisone. The equilibrium dissociation constant, K(i)a as well as the apparent Michaelis-Menten constants K(m)a, K(m)b, k(cat)a, and k(cat)b for NADPH and cortisone, have been determined to be 147.5 microM, 14.4 microM, 43.8 nM, 0.21 min(-1), and 0.27 min(-1), respectively, for the human enzyme and 41.1 microM, 3.1 microM, 161.7 nM, 0.49 min(-1), and 0.52min(-1), respectively, for the rabbit enzyme.


Subject(s)
11-beta-Hydroxysteroid Dehydrogenase Type 1/chemistry , Animals , Enzyme Activation , Enzyme Stability , Humans , Isoenzymes/chemistry , Kinetics , Rabbits , Species Specificity
18.
Mol Cell Biol ; 23(5): 1633-46, 2003 Mar.
Article in English | MEDLINE | ID: mdl-12588983

ABSTRACT

Estrogen receptors (ER) have been localized to the cell plasma membrane (PM), where signal transduction mediates some estradiol (E2) actions. However, the precise structural features of ER that result in membrane localization have not been determined. We obtained a partial tryptic peptide/mass spectrometry analysis of membrane mouse ERalpha protein. Based on this, we substituted alanine for the determined serine at amino acid 522 within the E domain of wild-type (wt) ERalpha. Upon transfection in CHO cells, the S522A mutant ERalpha resulted in a 62% decrease in membrane receptor number and reduced colocalization with caveolin 1 relative to those with expression of wt ERalpha. E2 was significantly less effective in stimulating multiple rapid signals from the membranes of CHO cells expressing ERalpha S522A than from those of CHO cells expressing wt ERalpha. In contrast, nuclear receptor expression and transcriptional function were very similar. The S522A mutant was also 60% less effective than wt ERalpha in binding caveolin 1, which facilitates ER transport to the PM. All functions of ERalpha mutants with other S-to-A substitutions were comparable to those of wt ER, and deletion of the A/B or C domain had little consequence for membrane localization or function. Transfection of ERalpha S522A into breast cancer cells that express native ER downregulated E2 binding at the membrane, signaling to ERK, and G1/S cell cycle events and progression. However, there was no effect on the E2 transactivation of an ERE-luciferase reporter. In summary, serine 522 is necessary for the efficient translocation and function of ERalpha at the PM. The S522A mutant also serves as a dominant-negative construct, identifying important functions of E2 that originate from activating PM ER.


Subject(s)
Cell Membrane/metabolism , Receptors, Estrogen/metabolism , Animals , Binding, Competitive , Breast Neoplasms/metabolism , CHO Cells , Caveolin 1 , Caveolins/metabolism , Cell Cycle , Cell Nucleus/metabolism , Cricetinae , Cyclin D1/metabolism , Dimerization , Dose-Response Relationship, Drug , Endoplasmic Reticulum/metabolism , Estrogen Receptor alpha , G1 Phase , Genes, Dominant , Kinetics , Mice , Microscopy, Fluorescence , Mitogen-Activated Protein Kinases/metabolism , Mutagenesis, Site-Directed , Mutation , Myristic Acid/metabolism , Palmitic Acid/metabolism , Protein Binding , Protein Structure, Tertiary , S Phase , Signal Transduction , Transcription, Genetic , Transcriptional Activation , Transfection , Tumor Cells, Cultured , Type C Phospholipases/metabolism
19.
Expert Opin Biol Ther ; 2(6): 621-32, 2002 Aug.
Article in English | MEDLINE | ID: mdl-12171506

ABSTRACT

Altered gene expression contributes to the aetiology of inflammatory disease by modulation of the concentration of disease-related proteins. The expression of inflammatory genes is controlled through the concerted actions of specific transcription factors. Signal transduction networks positively or negatively regulate the activity of these transcription factors. Key components of these networks are protein kinases, which phosphorylate substrates on tyrosine, threonine or serine residues. During the disease process, pro-inflammatory signalling at the cell surface leads to a cascade of kinase activation, which ultimately culminates in modulation of the activity of transcription factors. Thus, pharmacological inhibition of protein kinases is a potential therapeutic strategy to treat inflammation. There are approximately 500 protein kinases in the human genome. Targeted small molecule inhibitors of these kinases should allow for tissue- and disease-specific therapies of unprecedented selectivity. Heralding this new era in molecular medicine is imatinib (Gleevec, Norvartis) a recently marketed tyrosine kinase inhibitor. This review focuses on kinase inhibitors that are currently in development for inflammatory diseases and the transcription factors that are involved.


Subject(s)
Enzyme Inhibitors/pharmacology , Inflammation/therapy , Phosphotransferases/antagonists & inhibitors , Transcription, Genetic , Animals , Drug Design , Gene Expression Regulation , Humans , Models, Biological , Models, Chemical , Phosphorylation , Signal Transduction
20.
Glycobiology ; 12(7): 435-42, 2002 Jul.
Article in English | MEDLINE | ID: mdl-12122025

ABSTRACT

Phosphomannose isomerase (PMI) interconverts fructose-6-P (Fru-6-P) and mannose-6-P (Man-6-P), linking energy metabolism to protein glycosylation. We have cloned the mouse Mpi cDNA, analyzed its genomic organization, and studied the expression in different tissues. The Mpi gene has eight exons covering 7.2 kb. The structure and intron-exon boundaries are essentially the same as its human ortholog with 85% amino acid identity. Mpi is alternatively spliced at the 3' end, resulting in three messages with different 3'-untranslated regions. Mpi expression is regulated at both the transcription and translation levels, with the highest expression level in testis. Rabbit antibodies prepared against mouse PMI expressed in E. coli recognize a single 47-kDa band. Immunohistochemistry of mouse tissues shows general cytosolic staining in all cells. In testis, staining is intense in round spermatids and residual bodies, moderate in pachytene spermatocytes, and weak in spermatogonia and spermatozoa. In contrast, northern blot analysis shows comparable transcripts of 1.8 and 1.6 kb in pachytene spermatocytes and round spermatids, suggesting delayed translation of PMI. The stage-specific expression of PMI in testis may be important for KDN synthesis, which requires Man-6-P, or it may be needed to ensure sufficient glycosylation precursors in cells that do not utilize glucose and instead rely on lactate and pyruvate.


Subject(s)
Mannose-6-Phosphate Isomerase/genetics , Amino Acid Sequence , Animals , Base Sequence , Cloning, Molecular , DNA, Complementary , Exons , Immunohistochemistry , Introns , Male , Mice , Mice, Inbred C57BL , Molecular Sequence Data , Sequence Homology, Amino Acid , Spermatozoa/enzymology
SELECTION OF CITATIONS
SEARCH DETAIL
...