Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
Add more filters










Publication year range
1.
Oncotarget ; 8(38): 63014-63025, 2017 Sep 08.
Article in English | MEDLINE | ID: mdl-28968967

ABSTRACT

Targeted therapeutic agents, such as inhibitors of epithelial growth factor receptor (EGFR), have transformed the management of non-small cell lung cancer (NSCLC) patients. MET-amplified NSCLC cells display resistance to EGFR-targeting agents, but are addicted to MET signaling for survival and proliferation and are sensitive to MET inhibition. However, responsive cancer cells invariably develop resistance to MET-targeted treatment. The tumor microenvironment plays a major role in resistance to anticancer therapy. We demonstrated that fibroblasts block the response of MET-amplified NSCLC cells to the MET kinase inhibitor, JNJ38877605 in an HGF-dependent manner. Thus, MET-amplified NSCLC cells become addicted to HGF upon pharmacological inhibition of MET. HGF restored phosphorylation of MET, EGFR and RON, and maintained pro-survival AKT and ERK signaling in MET-inhibited cells. We developed a small molecule inhibitor of pro-HGF activation, SRI31215, which acts as a triplex inhibitor of the pro-HGF activating proteases matriptase, hepsin and HGF activator (HGFA). SRI31215 blocked crosstalk between tumor cells and fibroblasts and overcame fibroblast-mediated resistance to MET inhibition by preventing fibroblast-mediated reactivation of AKT and ERK signaling. Structurally unrelated triplex inhibitors of matriptase, hepsin and HGFA that we developed in parallel showed similar biological activity. Our data suggest that simultaneous inhibition of HGF and MET is required to overcome resistance to MET inhibitors in MET-amplified NSCLC cells. This provides a rationale for the development of novel combination therapeutic strategies for the treatment of NSCLC patients with MET amplification.

2.
Oncotarget ; 7(20): 29492-506, 2016 May 17.
Article in English | MEDLINE | ID: mdl-27121052

ABSTRACT

The binding of hepatocyte growth factor (HGF) to its receptor MET activates a signaling cascade that promotes cell survival, proliferation, cell scattering, migration and invasion of malignant cells. HGF is secreted by cancer cells or by tumor-associated fibroblasts as pro-HGF, an inactive precursor. A key step in the regulation of HGF/MET signaling is proteolytic processing of pro-HGF to its active form by one of the three serine proteases, matriptase, hepsin or HGF activator (HGFA).We developed SRI 31215, a small molecule that acts as a triplex inhibitor of matriptase, hepsin and HGFA and mimics the activity of HAI-1/2, endogenous inhibitors of HGF activation. We demonstrated that SRI 31215 inhibits fibroblast-induced MET activation, epithelial-mesenchymal transition and migration of cancer cells. SRI 31215 overcomes primary resistance to cetuximab and gefitinib in HGF-producing colon cancer cells and prevents fibroblast-mediated resistance to EGFR inhibitors. Thus, SRI 31215 blocks signaling between cancer cells and fibroblasts and inhibits the tumor-promoting activity of cancer-associated fibroblasts.Aberrant HGF/MET signaling supports cell survival, proliferation, angiogenesis, invasion and metastatic spread of cancer cells, establishing HGF and MET as valid therapeutic targets. Our data demonstrate that inhibitors of HGF activation, such as SRI 31215, merit investigation as potential therapeutics in tumors that are addicted to HGF/MET signaling. The findings reported here also indicate that inhibitors of HGF activation overcome primary and acquired resistance to anti-EGFR therapy, providing a rationale for concurrent inhibition of EGFR and HGF to prevent therapeutic resistance and to improve the outcome of cancer patients.


Subject(s)
Antineoplastic Agents/pharmacology , Benzamidines/pharmacology , Hepatocyte Growth Factor/antagonists & inhibitors , Protein Precursors/antagonists & inhibitors , Proto-Oncogene Proteins c-met/antagonists & inhibitors , Pyrimidinones/pharmacology , Signal Transduction/drug effects , Cancer-Associated Fibroblasts/drug effects , Cell Line, Tumor , Cell Movement/drug effects , Drug Resistance, Neoplasm/drug effects , Drug Screening Assays, Antitumor , Epithelial-Mesenchymal Transition/drug effects , Humans , Protein Precursors/drug effects
3.
ACS Med Chem Lett ; 7(2): 177-81, 2016 Feb 11.
Article in English | MEDLINE | ID: mdl-26985294

ABSTRACT

In this letter we report first nonpeptide inhibitors of hepatocyte growth factor (HGF) activation. These compounds inhibit the three proteases (matriptase, hepsin, and HGF activator) required for HGF maturation. We show that 6, 8a, 8b, and 8d block activation of fibroblast-derived pro-HGF, thus preventing fibroblast-induced scattering of DU145 prostate cancer cells. Compound 6 (SRI 31215) is very soluble (91 µM) and has excellent microsome stability (human t 1/2 = 162 min; mouse t 1/2 = 296 min). In mouse 6 has an in vivo t 1/2 = 5.8 h following IV administration. The high solubility of 6 and IV t 1/2 make this compound a suitable prototype "triplex inhibitor" for the study of the inhibition of HGF activation in vivo.

4.
J Biol Chem ; 289(47): 32937-51, 2014 Nov 21.
Article in English | MEDLINE | ID: mdl-25228699

ABSTRACT

Pathogenic mutations in the LRRK2 gene can cause late-onset Parkinson disease. The most common mutation, G2019S, resides in the kinase domain and enhances activity. LRRK2 possesses the unique property of cis-autophosphorylation of its own GTPase domain. Because high-resolution structures of the human LRRK2 kinase domain are not available, we used novel high-throughput assays that measured both cis-autophosphorylation and trans-peptide phosphorylation to probe the ATP-binding pocket. We disclose hundreds of commercially available activity-selective LRRK2 kinase inhibitors. Some compounds inhibit cis-autophosphorylation more strongly than trans-peptide phosphorylation, and other compounds inhibit G2019S-LRRK2 more strongly than WT-LRRK2. Through exploitation of structure-activity relationships revealed through high-throughput analyses, we identified a useful probe inhibitor, SRI-29132 (11). SRI-29132 is exquisitely selective for LRRK2 kinase activity and is effective in attenuating proinflammatory responses in macrophages and rescuing neurite retraction phenotypes in neurons. Furthermore, the compound demonstrates excellent potency, is highly blood-brain barrier-permeant, but suffers from rapid first-pass metabolism. Despite the observed selectivity of SRI-29132, docking models highlighted critical interactions with residues conserved in many protein kinases, implying a unique structural configuration for the LRRK2 ATP-binding pocket. Although the human LRRK2 kinase domain is unstable and insoluble, we demonstrate that the LRRK2 homolog from ameba can be mutated to approximate some aspects of the human LRRK2 ATP-binding pocket. Our results provide a rich resource for LRRK2 small molecule inhibitor development. More broadly, our results provide a precedent for the functional interrogation of ATP-binding pockets when traditional approaches to ascertain structure prove difficult.


Subject(s)
Adenosine Triphosphate/chemistry , Protein Serine-Threonine Kinases/chemistry , Protein Serine-Threonine Kinases/metabolism , Protein Structure, Tertiary , Adenosine Triphosphate/metabolism , Amino Acid Sequence , Animals , Binding Sites/genetics , Biocatalysis/drug effects , Blotting, Western , Cell Line, Tumor , Cells, Cultured , Hep G2 Cells , Humans , Kinetics , Leucine-Rich Repeat Serine-Threonine Protein Kinase-2 , Mice , Models, Molecular , Molecular Sequence Data , Molecular Structure , Mutation , Phosphorylation/drug effects , Protein Binding , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/metabolism , Protein Kinase Inhibitors/pharmacology , Protein Serine-Threonine Kinases/genetics , Pyridazines/chemistry , Pyridazines/metabolism , Pyridazines/pharmacology , Sequence Homology, Amino Acid , Small Molecule Libraries/chemistry , Small Molecule Libraries/metabolism , Small Molecule Libraries/pharmacology , Structure-Activity Relationship , Triazoles/chemistry , Triazoles/metabolism , Triazoles/pharmacology
5.
Hum Mol Genet ; 22(24): 4988-5000, 2013 Dec 15.
Article in English | MEDLINE | ID: mdl-23886663

ABSTRACT

Mutations in the leucine-rich repeat kinase 2 (LRRK2) gene cause late-onset Parkinson's disease (PD). Emerging evidence suggests a role for LRRK2 in the endocytic pathway. Here, we show that LRRK2 is released in extracellular microvesicles (i.e. exosomes) from cells that natively express LRRK2. LRRK2 localizes to collecting duct epithelial cells in the kidney that actively secrete exosomes into urine. Purified urinary exosomes contain LRRK2 protein that is both dimerized and phosphorylated. We provide a quantitative proteomic profile of 1673 proteins in urinary exosomes and find that known LRRK2 interactors including 14-3-3 are some of the most abundant exosome proteins. Disruption of the 14-3-3 LRRK2 interaction with a 14-3-3 inhibitor or through acute LRRK2 kinase inhibition potently blocks LRRK2 release in exosomes, but familial mutations in LRRK2 had no effect on secretion. LRRK2 levels were overall comparable but highly variable in urinary exosomes derived from PD cases and age-matched controls, although very high LRRK2 levels were detected in some PD affected cases. We further characterized LRRK2 exosome release in neurons and macrophages in culture, and found that LRRK2-positive exosomes circulate in cerebral spinal fluid (CSF). Together, these results define a pathway for LRRK2 extracellular release, clarify one function of the LRRK2 14-3-3 interaction and provide a foundation for utilization of LRRK2 as a biomarker in clinical trials.


Subject(s)
14-3-3 Proteins/metabolism , Exosomes/metabolism , Protein Serine-Threonine Kinases/metabolism , Animals , Epithelial Cells/metabolism , Humans , Kidney Tubules, Collecting/metabolism , Leucine-Rich Repeat Serine-Threonine Protein Kinase-2 , Macrophages/metabolism , Male , Mice , Mice, Knockout , Models, Biological , Mutation , Neurons/metabolism , Protein Binding , Protein Serine-Threonine Kinases/cerebrospinal fluid , Protein Serine-Threonine Kinases/genetics , Protein Transport , Rats , Rats, Transgenic
6.
ChemMedChem ; 8(8): 1295-313, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23794260

ABSTRACT

Polo-like kinase-2 (Plk-2) has been implicated as the dominant kinase involved in the phosphorylation of α-synuclein in Lewy bodies, which are one of the hallmarks of Parkinson's disease neuropathology. Potent, selective, brain-penetrant inhibitors of Plk-2 were obtained from a structure-guided drug discovery approach driven by the first reported Plk-2-inhibitor complexes. The best of these compounds showed excellent isoform and kinome-wide selectivity, with physicochemical properties sufficient to interrogate the role of Plk-2 inhibition in vivo. One such compound significantly decreased phosphorylation of α-synuclein in rat brain upon oral administration and represents a useful probe for future studies of this therapeutic avenue toward the potential treatment of Parkinson's disease.


Subject(s)
Brain/metabolism , Protein Kinase Inhibitors/chemistry , Protein Serine-Threonine Kinases/antagonists & inhibitors , alpha-Synuclein/metabolism , Animals , Binding Sites , Blood-Brain Barrier/metabolism , Female , HEK293 Cells , Half-Life , Humans , Male , Mice , Molecular Dynamics Simulation , Phosphorylation/drug effects , Protein Isoforms/antagonists & inhibitors , Protein Isoforms/metabolism , Protein Kinase Inhibitors/chemical synthesis , Protein Kinase Inhibitors/pharmacokinetics , Protein Serine-Threonine Kinases/metabolism , Protein Structure, Tertiary , Rats , Rats, Sprague-Dawley
7.
Bioorg Med Chem Lett ; 23(9): 2743-9, 2013 May 01.
Article in English | MEDLINE | ID: mdl-23522834

ABSTRACT

Polo-like kinase-2 (Plk-2) is a potential therapeutic target for Parkinson's disease and this Letter describes the SAR of a series of dihydropteridinone based Plk-2 inhibitors. By optimizing both the N-8 substituent and the biaryl region of the inhibitors we obtained single digit nanomolar compounds such as 37 with excellent selectivity for Plk-2 over Plk-1. When dosed orally in rats, compound 37 demonstrated a 41-45% reduction of pS129-α-synuclein levels in the cerebral cortex.


Subject(s)
Drug Design , Protein Kinase Inhibitors/chemical synthesis , Protein Serine-Threonine Kinases/antagonists & inhibitors , Administration, Oral , Animals , Brain/metabolism , Cell Cycle Proteins/antagonists & inhibitors , Cell Cycle Proteins/metabolism , HEK293 Cells , Half-Life , Humans , Mice , Microsomes, Liver/metabolism , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/pharmacokinetics , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/antagonists & inhibitors , Proto-Oncogene Proteins/metabolism , Pteridines/chemical synthesis , Pteridines/chemistry , Pteridines/pharmacokinetics , Rats , Structure-Activity Relationship , Polo-Like Kinase 1
8.
J Pharm Sci ; 101(7): 2545-56, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22473491

ABSTRACT

Extensive first-pass metabolism can significantly limit a drug's oral exposure levels. In this work, we introduce an innovative approach for increasing the oral bioavailability of a drug that undergoes extensive reversible glucuronidation and enterohepatic recirculation through intraduodenal coadministration of the deconjugating enzyme ß-glucuronidase. Intraduodenal administration of JNJ-10198409 (10 mg/kg) with ß-glucuronidase (34,000-140,000 units/kg) to catheterized rats resulted in a significant increase (p < 0.005) in the mean area under the plasma concentration versus time curve (AUC; approx. threefold) and maximum plasma concentration (C(max); approx. twofold) of JNJ-10198409. The AUC and C(max) were 60 ± 18 ng h/mL and 76 ± 29 ng/mL, respectively, with no enzyme and 177 ± 55 ng h/mL and 129 ± 41 ng/mL, respectively, with ß-glucuronidase coadministered. Moreover, the AUC of the primary glucuronide metabolite increased approximately sevenfold from 1173 ± 361 (ng h)/mL with no enzyme coadministered to 8723 ± 2133 ng h/mL with coadministered enzyme. These pharmacokinetic data support the hypothesis that when the primary glucuronide is secreted into the duodenum via the bile duct, the glucuronide is converted by ß-glucuronidase back to the parent compound. The parent compound is then reabsorbed and reconjugated, resulting in elevated systemic exposures to both parent and glucuronide. Potential clinical and preclinical applications and considerations for this approach are discussed.


Subject(s)
Glucuronidase/administration & dosage , Glucuronides/metabolism , Indans/administration & dosage , Indans/pharmacokinetics , Pyrazoles/administration & dosage , Pyrazoles/pharmacokinetics , Receptors, Platelet-Derived Growth Factor/antagonists & inhibitors , Administration, Oral , Animals , Area Under Curve , Biological Availability , Glucuronidase/metabolism , Indans/metabolism , Intestinal Absorption , Male , Pyrazoles/metabolism , Rats , Rats, Sprague-Dawley
9.
Bioorg Med Chem Lett ; 21(18): 5521-7, 2011 Sep 15.
Article in English | MEDLINE | ID: mdl-21813278

ABSTRACT

The SAR of a series of brain penetrant, trisubstituted thiophene based JNK inhibitors with improved pharmacokinetic properties is described. These compounds were designed based on information derived from metabolite identification studies which led to compounds such as 42 with lower clearance, greater brain exposure and longer half life compared to earlier analogs.


Subject(s)
Brain/metabolism , Drug Design , Nerve Degeneration/prevention & control , Protein Kinase Inhibitors/chemical synthesis , Protein Kinase Inhibitors/pharmacology , Thiophenes/pharmacology , Thiophenes/pharmacokinetics , Animals , Chemistry Techniques, Synthetic , Crystallography, X-Ray , Dose-Response Relationship, Drug , Half-Life , JNK Mitogen-Activated Protein Kinases/antagonists & inhibitors , JNK Mitogen-Activated Protein Kinases/metabolism , Mice , Models, Molecular , Molecular Structure , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/pharmacokinetics , Stereoisomerism , Structure-Activity Relationship , Thiophenes/chemical synthesis , Thiophenes/chemistry
10.
Bioorg Med Chem Lett ; 21(6): 1838-43, 2011 Mar 15.
Article in English | MEDLINE | ID: mdl-21316234

ABSTRACT

The SAR of a series of tri-substituted thiophene JNK3 inhibitors is described. By optimizing both the N-aryl acetamide region of the inhibitor and the 4-position of the thiophene we obtained single digit nanomolar compounds, such as 47, which demonstrated an in vivo effect on JNK activity when dosed orally in our kainic acid mouse model as measured by phospho-c-jun reduction.


Subject(s)
Brain/metabolism , MAP Kinase Kinase 4/antagonists & inhibitors , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/pharmacokinetics , Administration, Oral , Drug Design , Hydrogen Bonding , Models, Molecular , Protein Kinase Inhibitors/chemical synthesis , Structure-Activity Relationship
11.
Bioorg Med Chem Lett ; 21(1): 315-9, 2011 Jan 01.
Article in English | MEDLINE | ID: mdl-21112785

ABSTRACT

In this Letter, we describe the discovery of selective JNK2 and JNK3 inhibitors, such as 10, that routinely exhibit >10-fold selectivity over JNK1 and >1000-fold selectivity over related MAPKs, p38α and ERK2. Substitution of the naphthalene ring affords an isoform selective JNK3 inhibitor, 30, with approximately 10-fold selectivity over both JNK1 and JNK2. A naphthalene ring penetrates deep into the selectivity pocket accounting for the differentiation amongst the kinases. Interestingly, the gatekeeper Met146 sulfide interacts with the naphthalene ring in a sulfur-π stacking interaction. Compound 38 ameliorates neurotoxicity induced by amyloid-ß in human cortical neurons. Lastly, we demonstrate how to install propitious in vitro CNS-like properties into these selective inhibitors.


Subject(s)
Aminopyridines/chemistry , Mitogen-Activated Protein Kinase 10/antagonists & inhibitors , Mitogen-Activated Protein Kinase 9/antagonists & inhibitors , Neurodegenerative Diseases/drug therapy , Neuroprotective Agents/chemistry , Protein Kinase Inhibitors/chemistry , Triazines/chemistry , Aminopyridines/pharmacokinetics , Aminopyridines/therapeutic use , Animals , Binding Sites , Central Nervous System/metabolism , Computer Simulation , Humans , Mice , Microsomes, Liver/metabolism , Mitogen-Activated Protein Kinase 10/metabolism , Mitogen-Activated Protein Kinase 9/metabolism , Neuroprotective Agents/pharmacokinetics , Neuroprotective Agents/therapeutic use , Protein Kinase Inhibitors/pharmacokinetics , Protein Kinase Inhibitors/therapeutic use , Structure-Activity Relationship , Triazines/pharmacokinetics , Triazines/therapeutic use
12.
J Pharmacol Exp Ther ; 324(3): 894-901, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18083913

ABSTRACT

The alpha(V) integrins are key receptors involved in mediating cell migration and angiogenesis. In age-related macular degeneration (AMD) and diabetic retinopathy, angiogenesis plays a critical role in the loss of vision. These ocular vasculopathies might be treatable with a suitable alpha(V) antagonist, and an oral drug would offer a distinct advantage over current therapies. (3,S,beta,S)-1,2,3,4-Tetrahydro-beta-[[1-[1-oxo-3-(1,5,6,7-tetrahydro-1,8-naphthyridin-2-yl)propyl]-4-piperidinyl]methyl]-3-quinolinepropanoic acid (JNJ-26076713) is a potent, orally bioavailable, nonpeptide alpha(V) antagonist derived from the arginine-glycine-asparagine binding motif in the matrix protein ligands (e.g., vitronectin). This compound inhibits alpha(V)beta(3) and alpha(V)beta(5) binding to vitronectin in the low nanomolar range, it has excellent selectivity over integrins alpha(IIb)beta(3) and alpha(5)beta(1), and it prevents adhesion to human, rat, and mouse endothelial cells. JNJ-26076713 blocks cell migration induced by vascular endothelial growth factor, fibroblast growth factor (FGF), and serum, and angiogenesis induced by FGF in the chick chorioallantoic membrane model. JNJ-26076713 is the first alpha(V) antagonist reported to inhibit retinal neovascularization in an oxygen-induced model of retinopathy of prematurity after oral administration. In diabetic rats, orally administered JNJ-26076713 markedly inhibits retinal vascular permeability, a key early event in diabetic macular edema and AMD. Given this profile, JNJ-26076713 represents a potential therapeutic candidate for the treatment of age-related macular degeneration, macular edema, and proliferative diabetic retinopathy.


Subject(s)
Capillary Permeability/physiology , Diabetes Mellitus, Experimental/metabolism , Diabetic Retinopathy/metabolism , Integrin alphaV/metabolism , Naphthyridines/administration & dosage , Naphthyridines/pharmacokinetics , Quinolines/administration & dosage , Quinolines/pharmacokinetics , Retinal Neovascularization/metabolism , Administration, Oral , Angiogenesis Inhibitors/administration & dosage , Angiogenesis Inhibitors/chemistry , Angiogenesis Inhibitors/pharmacokinetics , Animals , Biological Availability , Capillary Permeability/drug effects , Cell Line , Chick Embryo , Diabetes Mellitus, Experimental/drug therapy , Diabetic Retinopathy/drug therapy , Disease Models, Animal , Female , Humans , Male , Mice , Mice, Inbred C57BL , Naphthyridines/chemistry , Pregnancy , Quinolines/chemistry , Rats , Rats, Long-Evans , Rats, Sprague-Dawley , Retinal Neovascularization/drug therapy
13.
Bioorg Med Chem Lett ; 17(5): 1432-7, 2007 Mar 01.
Article in English | MEDLINE | ID: mdl-17174550

ABSTRACT

The synthesis, SAR, pharmacokinetic profile, and modeling studies of both monocyclic and fused pyrazoles containing substituted N-arylpiperidinyl P4 moieties that are potent and selective factor Xa inhibitors will be discussed. Fused pyrazole analog 16a, with a 2'-methylsulfonylphenyl piperidine P4 group, was shown to be the best compound in this series (FXa Ki = 0.35 nM) based on potency, selectivity, and pharmacokinetic profile.


Subject(s)
Factor Xa Inhibitors , Piperidines/chemical synthesis , Piperidines/pharmacokinetics , Pyrazoles/chemical synthesis , Pyrazoles/pharmacokinetics , Animals , Dogs , Models, Molecular , Piperidines/administration & dosage , Protein Binding , Pyrazoles/administration & dosage , Structure-Activity Relationship
14.
Bioorg Med Chem Lett ; 16(22): 5778-83, 2006 Nov 15.
Article in English | MEDLINE | ID: mdl-16949284

ABSTRACT

A new class of Aurora-A inhibitors have been identified based on the 2-amino-pyrrolo[2,3-d]pyrimidine scaffold. Here, we describe the synthesis and SAR of this novel series. We report compounds which exhibit nanomolar activity in the Aurora-A biochemical assay and are able to inhibit tumor cell proliferation. This study culminates in compound 30, an inhibitor with potent activity against Aurora A (IC50=0.008 microM), anti-proliferative activity against several tumor cell lines and induces polyploidy in H460 cells.


Subject(s)
Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/pharmacology , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/pharmacology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Pyrimidines/chemical synthesis , Pyrimidines/pharmacology , Pyrroles/chemical synthesis , Pyrroles/pharmacology , Aurora Kinases , Cell Line, Tumor , Drug Screening Assays, Antitumor , Humans , Inhibitory Concentration 50 , Polyploidy , Structure-Activity Relationship
15.
Bioorg Med Chem Lett ; 16(21): 5584-9, 2006 Nov 01.
Article in English | MEDLINE | ID: mdl-16963264

ABSTRACT

The bicyclic dihydropyrazolopyridinone scaffold allowed for incorporation of multiple P1 moieties with subnanomolar binding affinities for blood coagulation factor Xa. The compound 3-[6-(2'-dimethylaminomethyl-biphenyl-4-yl)-7-oxo-3-trifluoro-methyl-4,5,6,7-tetrahydro-pyrazolo[3,4-c]pyridine-l-yl]-benzamide 6d shows good fXa potency, selectivity, in vivo efficacy and oral bioavailability. Compound 6d was selected for further pre-clinical evaluations.


Subject(s)
Benzamides/chemistry , Benzamides/pharmacology , Factor Xa Inhibitors , Fibrinolytic Agents/chemistry , Fibrinolytic Agents/pharmacology , Administration, Oral , Benzamides/administration & dosage , Benzamides/chemical synthesis , Fibrinolytic Agents/administration & dosage , Fibrinolytic Agents/chemical synthesis , Humans , Treatment Outcome
16.
Bioorg Med Chem Lett ; 16(15): 4141-7, 2006 Aug 01.
Article in English | MEDLINE | ID: mdl-16730984

ABSTRACT

Attempts to further optimize the pyrazole factor Xa inhibitors centered on masking the aryl aniline P4 moiety. Scaffold optimization resulted in the identification of a novel bicyclic pyrazolo-pyridinone scaffold which retained fXa potency. The novel bicyclic scaffold preserved all binding interactions observed with the monocyclic counterpart and importantly the carboxamido moiety was integrated within the scaffold making it less susceptible to hydrolysis. These efforts led to the identification of 1-[3-aminobenzisoxazol-5'-yl]-3-trifluoromethyl-6-[2'-(3-(R)-hydroxy-N-pyrrolidinyl)methyl-[1,1']-biphen-4-yl]-1,4,5,6-tetrahydropyrazolo-[3,4-c]-pyridin-7-one 6f (BMS-740808), a highly potent (fXa Ki=30 pM) with a rapid onset of inhibition (2.7x10(7) M-1 s-1) in vitro, selective (>1000-fold over other proteases), efficacious in the AVShunt thrombosis model, and orally bioavailable inhibitor of blood coagulation factor Xa.


Subject(s)
Factor Xa Inhibitors , Pyrazoles/pharmacology , Pyridones/pharmacology , Serine Proteinase Inhibitors/pharmacology , Models, Molecular , Pyrazoles/administration & dosage , Pyrazoles/chemistry , Pyridones/administration & dosage , Pyridones/chemistry , Serine Proteinase Inhibitors/administration & dosage , Serine Proteinase Inhibitors/chemistry
17.
Biochemistry ; 45(17): 5678-85, 2006 May 02.
Article in English | MEDLINE | ID: mdl-16634649

ABSTRACT

Heat shock protein 90 (Hsp90) is critical for the maturation of numerous client proteins, many of which are involved in cellular transformation and oncogenesis. The ansamycins, geldanamycin (GA) and its derivative, 17-allylaminogeldanamycin (17-AAG), inhibit Hsp90. As such, the prototypical Hsp90 inhibitor, 17-AAG, has advanced into clinical oncology trials. GA and 17-AAG potently inhibit tumor cell proliferation and survival but have been reported to bind weakly to Hsp90 in vitro. Recent studies have suggested that the in vitro potency of ansamycins against Hsp90 may be enhanced in the presence of cochaperones. Here, we present evidence of an alternative explanation. Ansamycins reduced to their dihydroquinones in the presence of common reducing agents in vitro have approximately 40-fold greater affinity than the corresponding oxidized quinones. The dihydroquinone of 17-AAG is not generated in an aqueous environment in the absence of reducing agents but is produced in both tumor and normal quiescent epithelial cells. The reduced form of 17-AAG is differentiated from its oxidized form not only by the higher affinity for Hsp90 but also by a protracted K(off) rate. Therefore, the in vivo accumulation of the high-affinity dihydroquinone ansamycins in tumor cells contributes to the antitumor activity of these compounds and alters our understanding of the active species driving the efficacy of this class of compounds.


Subject(s)
HSP90 Heat-Shock Proteins/antagonists & inhibitors , Quinones/metabolism , Quinones/pharmacology , Antineoplastic Agents , Benzoquinones , Cell Line, Tumor/drug effects , Cells, Cultured , Drug Stability , Humans , Lactams, Macrocyclic , Protein Binding/drug effects , Reducing Agents/pharmacology , Rifabutin/analogs & derivatives , Rifabutin/metabolism , Rifabutin/pharmacology , Solubility
18.
J Med Chem ; 48(26): 8163-73, 2005 Dec 29.
Article in English | MEDLINE | ID: mdl-16366598

ABSTRACT

A series of (6,7-dimethoxy-2,4-dihydroindeno[1,2-c]pyrazol-3-yl)phenylamines has been optimized to preserve both potent kinase inhibition activity against the angiogenesis target, the receptor tyrosine kinase of Platelet-Derived Growth Factor-BB (PDGF-BB), and to improve the broad tumor cell antiproliferative activity of these compounds. This series culminates in the discovery of 17 (JNJ-10198409), a compound with anti-PDGFR-beta kinase activity (IC(50)=0.0042 microM) and potent antiproliferative activity in six of eight human tumor cell lines (IC(50) < 0.033 microM).


Subject(s)
Antineoplastic Agents/pharmacology , Indans/chemical synthesis , Protein-Tyrosine Kinases/antagonists & inhibitors , Pyrazoles/chemical synthesis , Receptors, Platelet-Derived Growth Factor/antagonists & inhibitors , Antineoplastic Agents/chemical synthesis , Becaplermin , Cell Line , Cell Line, Tumor , Endothelium, Vascular/drug effects , Humans , Indans/pharmacology , Inhibitory Concentration 50 , Platelet-Derived Growth Factor/antagonists & inhibitors , Proto-Oncogene Proteins c-sis , Pyrazoles/pharmacology
19.
Mol Cancer Ther ; 4(8): 1198-204, 2005 Aug.
Article in English | MEDLINE | ID: mdl-16093435

ABSTRACT

With the advent of agents directed against specific molecular targets in drug discovery, it has become imperative to show a compound's cellular impact on the intended biomolecule in vivo. The objective of the present study was to determine if we could develop an assay to validate the in vivo effects of a compound. Hence, we investigated the in vivo pharmacodynamic activity of JNJ-10198409, a relatively selective inhibitor of platelet-derived growth factor receptor tyrosine kinase (PDGF-RTK), in tumor tissues after administering the compound orally in a nude mouse xenograft model of human LoVo colon cancer. We developed a novel assay to quantify the in vivo anti-PDGF-RTK activity of the inhibitor in tumor tissue by determining the phosphorylation status of phospholipase Cgamma1 (PLCgamma1), a key downstream cellular molecule in the PDGF-RTK signaling cascade. We used two antibodies, one specific for the total (phosphorylated and unphosphorylated forms) PLCgamma1 (pan-PLCgamma1) and the other, specific for phosphorylated form of PLCgamma1 (ph-PLCgamma1) to immunohistochemically detect their expression in tumor tissues. Computer-assisted image analysis was then used to directly compare the ratio of ph-PLCgamma1 to pan-PLCgamma1 immunolabeling intensities in serial sections (5 mum) of tumors obtained from vehicle- and JNJ-10198409-treated tumor-bearing mice. Our data showed statistically significant, dose-dependent differences in the ph-PLC/pan-PLC ratio among the four treatment groups (vehicle, 25, 50, and 100 mg/kg b.i.d.). These results confirmed this compound's ability to suppress PDGF-RTK downstream signaling in tumor tissues in vivo. In addition to this specific application of this in vivo validation approach to those targets that use PLCgamma as a downstream signaling partner, these methods may also benefit other drug discovery targets.


Subject(s)
Antineoplastic Agents/pharmacology , Drug Screening Assays, Antitumor , Indans/pharmacology , Protein Kinase Inhibitors/pharmacology , Pyrazoles/pharmacology , Receptors, Platelet-Derived Growth Factor/antagonists & inhibitors , Animals , Antibodies, Phospho-Specific/immunology , Antineoplastic Agents/chemistry , Colonic Neoplasms/enzymology , Humans , Image Processing, Computer-Assisted , Immunohistochemistry , Indans/chemistry , Mice , Phospholipase C gamma , Phosphorylation , Protein Kinase Inhibitors/chemistry , Pyrazoles/chemistry , Type C Phospholipases/analysis , Type C Phospholipases/immunology , Xenograft Model Antitumor Assays
20.
J Org Chem ; 70(12): 4873-5, 2005 Jun 10.
Article in English | MEDLINE | ID: mdl-15932334

ABSTRACT

The addition of small amounts of solid KCN to solution and solid-phase esters in THF/MeOH/50% aqueous NH2OH increases the efficiency of their transformation to the corresponding hydroxamic acids.


Subject(s)
Hydroxamic Acids/chemical synthesis , Catalysis , Esters , Indicators and Reagents , Molecular Structure
SELECTION OF CITATIONS
SEARCH DETAIL
...