Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
1.
Proc Natl Acad Sci U S A ; 111(41): 14882-7, 2014 Oct 14.
Article in English | MEDLINE | ID: mdl-25267635

ABSTRACT

Efforts to develop unbiased screens for identifying novel function-blocking monoclonal antibodies (mAbs) in human carcinomatous states have been hampered by the limited ability to design in vitro models that recapitulate tumor cell behavior in vivo. Given that only invasive carcinoma cells gain permanent access to type I collagen-rich interstitial tissues, an experimental platform was established in which human breast cancer cells were embedded in 3D aldimine cross-linked collagen matrices and used as an immunogen to generate mAb libraries. In turn, cancer-cell-reactive antibodies were screened for their ability to block carcinoma cell proliferation within collagen hydrogels that mimic the in vivo environment. As a proof of principle, a single function-blocking mAb out of 15 identified was selected for further analysis and found to be capable of halting carcinoma cell proliferation, inducing apoptosis, and exerting global changes in gene expression in vitro. The ability of this mAb to block carcinoma cell proliferation and metastatic activity was confirmed in vivo, and the target antigen was identified by mass spectroscopy as the α2 subunit of the α2ß1 integrin, one of the major type I collagen-binding receptors in mammalian cells. Validating the ability of the in vitro model to predict patterns of antigen expression in the disease setting, immunohistochemical analyses of tissues from patients with breast cancer verified markedly increased expression of the α2 subunit in vivo. These results not only highlight the utility of this discovery platform for rapidly selecting and characterizing function-blocking, anticancer mAbs in an unbiased fashion, but also identify α2ß1 as a potential target in human carcinomatous states.


Subject(s)
Antibodies, Monoclonal/immunology , Breast Neoplasms/immunology , Immunoassay/methods , Animals , Antigens, Neoplasm/metabolism , Bone Neoplasms/pathology , Bone Neoplasms/secondary , Breast Neoplasms/genetics , Cell Line, Tumor , Cell Proliferation , Chickens , Extravasation of Diagnostic and Therapeutic Materials , Female , Humans , Integrin alpha2/metabolism , Mice, Nude , Transcriptome/genetics , Xenograft Model Antitumor Assays
2.
J Pharmacol Exp Ther ; 338(1): 114-24, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21505060

ABSTRACT

The endogenous cannabinoid (endocannabinoid) anandamide is principally degraded by the integral membrane enzyme fatty acid amide hydrolase (FAAH). Pharmacological blockade of FAAH has emerged as a potentially attractive strategy for augmenting endocannabinoid signaling and retaining the beneficial effects of cannabinoid receptor activation, while avoiding the undesirable side effects, such as weight gain and impairments in cognition and motor control, observed with direct cannabinoid receptor 1 agonists. Here, we report the detailed mechanistic and pharmacological characterization of N-pyridazin-3-yl-4-(3-{[5-(trifluoromethyl)pyridin-2-yl]oxy}benzylidene)piperidine-1-carboxamide (PF-04457845), a highly efficacious and selective FAAH inhibitor. Mechanistic studies confirm that PF-04457845 is a time-dependent, covalent FAAH inhibitor that carbamylates FAAH's catalytic serine nucleophile. PF-04457845 inhibits human FAAH with high potency (k(inact)/K(i) = 40,300 M(-1)s(-1); IC(50) = 7.2 nM) and is exquisitely selective in vivo as determined by activity-based protein profiling. Oral administration of PF-04457845 produced potent antinociceptive effects in both inflammatory [complete Freund's adjuvant (CFA)] and noninflammatory (monosodium iodoacetate) pain models in rats, with a minimum effective dose of 0.1 mg/kg (CFA model). PF-04457845 displayed a long duration of action as a single oral administration at 1 mg/kg showed in vivo efficacy for 24 h with a concomitant near-complete inhibition of FAAH activity and maximal sustained elevation of anandamide in brain. Significantly, PF-04457845-treated mice at 10 mg/kg elicited no effect in motility, catalepsy, and body temperature. Based on its exceptional selectivity and in vivo efficacy, combined with long duration of action and optimal pharmacokinetic properties, PF-04457845 is a clinical candidate for the treatment of pain and other nervous system disorders.


Subject(s)
Amidohydrolases/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/therapeutic use , Pain/drug therapy , Pain/enzymology , Pyridazines/pharmacology , Pyridazines/therapeutic use , Urea/analogs & derivatives , Amidohydrolases/metabolism , Animals , Enzyme Inhibitors/chemistry , Humans , Inflammation/drug therapy , Inflammation/enzymology , Inflammation/pathology , Male , Mice , Mice, Inbred C57BL , Piperidines/chemistry , Piperidines/pharmacology , Piperidines/therapeutic use , Pyridazines/chemistry , Rats , Rats, Sprague-Dawley , Urea/chemistry , Urea/pharmacology , Urea/therapeutic use
3.
Nat Struct Mol Biol ; 11(12): 1192-7, 2004 Dec.
Article in English | MEDLINE | ID: mdl-15543157

ABSTRACT

MEK1 and MEK2 are closely related, dual-specificity tyrosine/threonine protein kinases found in the Ras/Raf/MEK/ERK mitogen-activated protein kinase (MAPK) signaling pathway. Approximately 30% of all human cancers have a constitutively activated MAPK pathway, and constitutive activation of MEK1 results in cellular transformation. Here we present the X-ray structures of human MEK1 and MEK2, each determined as a ternary complex with MgATP and an inhibitor to a resolution of 2.4 A and 3.2 A, respectively. The structures reveal that MEK1 and MEK2 each have a unique inhibitor-binding pocket adjacent to the MgATP-binding site. The presence of the potent inhibitor induces several conformational changes in the unphosphorylated MEK1 and MEK2 enzymes that lock them into a closed but catalytically inactive species. Thus, the structures reported here reveal a novel, noncompetitive mechanism for protein kinase inhibition.


Subject(s)
Enzyme Inhibitors/pharmacology , MAP Kinase Kinase 1/chemistry , MAP Kinase Kinase 1/metabolism , MAP Kinase Kinase 2/chemistry , MAP Kinase Kinase 2/metabolism , Binding Sites , Conserved Sequence , Dimerization , Enzyme Inhibitors/chemistry , Humans , MAP Kinase Kinase 1/antagonists & inhibitors , MAP Kinase Kinase 2/antagonists & inhibitors , Models, Molecular , Molecular Structure , Protein Structure, Quaternary , Structural Homology, Protein
4.
Bioorg Med Chem Lett ; 18(24): 6501-4, 2008 Dec 15.
Article in English | MEDLINE | ID: mdl-18952427

ABSTRACT

A novel series of benzhydroxamate esters derived from their precursor anthranilic acids have been prepared and have been identified as potent MEK inhibitors. 2-(2-Chloro-4-iodo-phenylamino)-N-cyclopropylmethoxy-3,4-difluoro-benzamide, CI-1040, was the first MEK inhibitor to demonstrate in vivo activity in preclinical animal models and subsequently became the first MEK inhibitor to enter clinical trial. CI-1040 suffered however from poor exposure due to its poor solubility and rapid clearance, and as a result, development of the compound was terminated. Optimization of the diphenylamine core and modification of the hydroxamate side chain for cell potency, solubility, and exposure with oral delivery resulted in the discovery of the clinical candidate N-(2,3-dihydroxy-propoxy)-3,4-difluoro-2-(2-fluoro-4-iodo-phenylamino)-benzamide PD 0325901.


Subject(s)
Benzamides/chemical synthesis , Diphenylamine/analogs & derivatives , Enzyme Inhibitors/chemical synthesis , MAP Kinase Kinase Kinase 1/antagonists & inhibitors , Animals , Benzamides/pharmacology , Benzoates/chemistry , Cell Line, Tumor , Chemistry, Pharmaceutical/methods , Diphenylamine/chemical synthesis , Diphenylamine/pharmacology , Drug Design , Enzyme Inhibitors/pharmacology , Hydroxamic Acids/chemistry , Inhibitory Concentration 50 , Mice , Neoplasm Transplantation , Solubility , ortho-Aminobenzoates/chemistry
5.
Mol Cell Biol ; 22(21): 7593-602, 2002 Nov.
Article in English | MEDLINE | ID: mdl-12370306

ABSTRACT

Utilizing a genetic screen in the yeast Saccharomyces cerevisiae, we identified a novel autoactivation region in mammalian MEK1 that is involved in binding the specific MEK inhibitor, PD 184352. The genetic screen is possible due to the homology between components of the yeast pheromone response pathway and the eukaryotic Raf-MEK-ERK signaling cascade. Using the FUS1::HIS3 reporter as a functional readout for activation of a reconstituted Raf-MEK-ERK signaling cascade, randomly mutagenized MEK variants that were insensitive to PD 184352 were obtained. Seven single-base-change mutations were identified, five of which mapped to kinase subdomains III and IV of MEK. Of the seven variants, only one, a leucine-to-proline substitution at amino acid 115 (Leu115Pro), was completely insensitive to PD 184352 in vitro (50% inhibitory concentration >10 micro M). However, all seven mutants displayed strikingly high basal activity compared to wild-type MEK. Overexpression of the MEK variants in HEK293T cells resulted in an increase in mitogen-activated protein (MAP) kinase phosphorylation, a finding consistent with the elevated basal activity of these constructs. Further, treatment with PD 184352 failed to inhibit Leu115Pro-stimulated MAP kinase activation in HEK293T cells, whereas all other variants had some reduction in phospho-MAP kinase levels. By using cyclic AMP-dependent protein kinase (1CDK) as a template, an MEK homology model was generated, with five of the seven identified residues clustered together, forming a potential hydrophobic binding pocket for PD 184352. Additionally, the model allowed identification of other potential residues that would interact with the inhibitor. Directed mutation of these residues supported this region's involvement with inhibitor binding.


Subject(s)
Benzamides/pharmacology , Mitogen-Activated Protein Kinase Kinases/chemistry , Mitogen-Activated Protein Kinase Kinases/metabolism , Signal Transduction , Amino Acid Sequence , Blotting, Western , CDC2 Protein Kinase/metabolism , Cell Line , Dose-Response Relationship, Drug , Enzyme Activation , Enzyme Inhibitors/pharmacology , Gene Library , Humans , Inhibitory Concentration 50 , MAP Kinase Signaling System , Models, Genetic , Models, Molecular , Molecular Sequence Data , Mutagenesis , Mutation , Plasmids/metabolism , Protein Binding , Protein Structure, Tertiary , Recombinant Proteins/metabolism , Saccharomyces cerevisiae/metabolism
6.
Pharmacol Biochem Behav ; 99(4): 718-25, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21740924

ABSTRACT

Fatty acid amide hydrolase (FAAH) is the primary degradative enzyme of the endocannabinoid anandamide (N-arachidonoylethanolamine), which activates cannabinoid CB(1) and CB(2) receptors. FAAH disruption reduces nociception in a variety of acute rodent models of inflammatory pain. The present study investigated whether these actions extend to the chronic, collagen-induced arthritis (CIA) model. We investigated the anti-arthritic and anti-hyperalgesic effects of genetic deletion or pharmacological inhibition of FAAH in the CIA model. FAAH (-/-) mice, and FAAH-NS mice that express FAAH exclusively in nervous tissue, displayed decreased severity of CIA and associated hyperalgesia. These phenotypic anti-arthritic effects were prevented by repeated daily injections of the CB(2) receptor antagonist, SR144528, but not the CB(1) receptor antagonist rimonabant. Similarly, repeated administration of the FAAH inhibitor URB597 reduced CIA severity, and acute administration of rimonabant, but not SR144528, blocked the anti-hyperalgesic effects of prolonged FAAH inhibition, suggesting that prolonged CB(2) receptor activation reduces the severity of CIA, whereas acute CB(1) receptor activation reduces CIA-induced hyperalgesia. In contrast, acute administration of URB597 elicited a CB(1) receptor-dependent anti-hyperalgesic effect. The observed anti-arthritic and anti-hyperalgesic properties of FAAH inhibition, coupled with a lack of apparent behavioral alterations, suggest that endocannabinoid modulating enzymes offer a promising therapeutic target for the development of novel pharmacological approaches to treat rheumatoid arthritis and associated hyperalgesia.


Subject(s)
Amidohydrolases/antagonists & inhibitors , Arthritis, Experimental/prevention & control , Hot Temperature , Hyperalgesia/drug therapy , Amidohydrolases/genetics , Amidohydrolases/physiology , Animals , Arthritis, Experimental/genetics , Arthritis, Experimental/pathology , Benzamides/pharmacology , Camphanes/pharmacology , Carbamates/pharmacology , Foot/pathology , Genotype , Male , Mice , Mice, Inbred DBA , Mice, Knockout , Pain Measurement/drug effects , Phenotype , Pyrazoles/pharmacology , Receptor, Cannabinoid, CB1/drug effects , Receptor, Cannabinoid, CB2/drug effects
7.
ACS Med Chem Lett ; 2(2): 91-96, 2011 Feb 10.
Article in English | MEDLINE | ID: mdl-21666860

ABSTRACT

Fatty acid amide hydrolase (FAAH) is an integral membrane serine hydrolase that degrades the fatty acid amide family of signaling lipids, including the endocannabinoid anandamide. Genetic or pharmacological inactivation of FAAH leads to analgesic and anti-inflammatory phenotypes in rodents without showing the undesirable side effects observed with direct cannabinoid receptor agonists, indicating that FAAH may represent an attractive therapeutic target for the treatment of inflammatory pain and other nervous system disorders. Herein, we report the discovery and characterization of a highly efficacious and selective FAAH inhibitor PF-04457845 (23). Compound 23 inhibits FAAH by a covalent, irreversible mechanism involving carbamylation of the active-site serine nucleophile of FAAH with high in vitro potency (k(inact)/K(i) and IC(50) values of 40300 M(-1) s(-1) and 7.2 nM, respectively, for human FAAH). Compound 23 has exquisite selectivity for FAAH relative to other members of the serine hydrolase superfamily as demonstrated by competitive activity-based protein profiling. Oral administration of 23 at 0.1 mg/kg results in efficacy comparable to that of naproxen at 10 mg/kg in a rat model of inflammatory pain. Compound 23 is being evaluated in human clinical trials.

8.
Bioorg Med Chem Lett ; 17(3): 756-60, 2007 Feb 01.
Article in English | MEDLINE | ID: mdl-17095227

ABSTRACT

The Type 1 PI3Kinases comprise a family of enzymes, which primarily phosphorylate PIP2 to give the second messenger PIP3, a key player in many intracellular signaling processes [Science, 2002, 296, 1655; Trends Pharmacol. Sci.2003, 24, 366]. Of the four type 1 PI3Ks, the gamma-isoform, which is expressed almost exclusively in leukocytes [Curr. Biol., 1997, 7, R470], is of particular interest with respect to its role in inflammatory diseases such as rheumatoid arthritis (RA) and chronic obstructive pulmonary disease (COPD) [Mol. Med. Today, 2000, 6, 347]. Investigation of a series of 4,6-disubstituted-4H-benzo[1,4]oxazin-3-ones has led to the identification of single-digit nanomolar inhibitors of PI3Kgamma, several of which had good cell based activity and were shown to be active in vivo in an aspectic peritonitis model of inflammatory cell migration.


Subject(s)
Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/pharmacology , Oxazines/chemical synthesis , Oxazines/pharmacology , Phosphoinositide-3 Kinase Inhibitors , Animals , Baculoviridae/drug effects , Baculoviridae/enzymology , Class Ib Phosphatidylinositol 3-Kinase , Drug Design , Drug Evaluation, Preclinical , Escherichia coli/drug effects , Indicators and Reagents , Isoenzymes/antagonists & inhibitors , Isoenzymes/genetics , Magnetic Resonance Spectroscopy , Mice , Mice, Knockout , Phosphatidylinositol 3-Kinases/genetics
9.
Arthritis Rheum ; 56(10): 3347-57, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17907188

ABSTRACT

OBJECTIVE: To evaluate the role of the MEK/ERK MAP kinase pathway in murine collagen-induced arthritis (CIA) using the selective MEK inhibitor PD184352. We examined the effects of the inhibitor in cytokine-stimulated synovial fibroblasts and in cytokine-induced arthritis in rabbits to investigate its antiinflammatory mechanisms. METHODS: Murine CIA was used to assess the effects of the selective MEK inhibitor on paw edema, clinical scores, weight loss, histopathologic features, and joint levels of p-ERK. Western blotting and immunohistochemistry techniques were used to assess p-ERK in human and rabbit synovial fibroblasts and synovial tissue from rheumatoid arthritis (RA) patients. Interleukin-1alpha (IL-1alpha)-stimulated stromelysin production in rabbit synovial fibroblasts was assessed by enzyme-linked immunosorbent assay. A rabbit IL-1alpha-induced arthritis model was used to assess the effects of the inhibitor on IL-1alpha-induced MEK activity, stromelysin production, and cartilage degradation. RESULTS: In the CIA model, PD184352 inhibited paw edema and clinical arthritis scores in a dose-dependent manner. Disease-induced weight loss and histopathologic changes were also significantly improved by treatment. Inhibition of disease-induced p-ERK levels in the joints was seen with the inhibitor. Levels of p-ERK in the synovium were higher in RA patients than in normal individuals. PD184352 reduced IL-1alpha-induced p-ERK levels in human RA synovial fibroblasts. The production of p-ERK and stromelysin was also inhibited in IL-1alpha-stimulated rabbit synovial fibroblasts. We observed IL-1alpha-induced p-ERK in the synovial lining, subsynovial vasculature, and articular chondrocytes. IL-1alpha-induced stromelysin production and proteoglycan loss from the articular cartilage were reduced by PD184352. CONCLUSION: These data demonstrate the inhibition of murine CIA by PD184352, support the hypothesis that antiinflammatory activity contributes to the mechanism of action of the inhibitor, and suggest that a selective inhibitor may effectively treat RA and other inflammatory disorders.


Subject(s)
Arthritis, Rheumatoid/enzymology , Benzamides/pharmacology , Enzyme Inhibitors/pharmacology , Extracellular Signal-Regulated MAP Kinases/drug effects , MAP Kinase Kinase Kinases/drug effects , Animals , Arthritis, Rheumatoid/drug therapy , Blotting, Western , Cartilage, Articular/drug effects , Cartilage, Articular/enzymology , Disease Models, Animal , Extracellular Signal-Regulated MAP Kinases/metabolism , Female , Fibroblasts/drug effects , Fibroblasts/enzymology , Immunohistochemistry , In Vitro Techniques , MAP Kinase Kinase Kinases/metabolism , Male , Mice , Mice, Inbred DBA , Rabbits
SELECTION OF CITATIONS
SEARCH DETAIL