Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
1.
J Med Chem ; 65(8): 5886-5901, 2022 04 28.
Article in English | MEDLINE | ID: mdl-35439421

ABSTRACT

Covalent drugs have made a major impact on human health but until recently were shunned by the pharmaceutical industry over concerns about the potential for toxicity. A resurgence has occurred driven by the clinical success of targeted covalent inhibitors (TCIs), with eight drugs approved over the past decade. The opportunity to create unique drugs by exploiting the covalent mechanism of action has enabled clinically decisive target product profiles to be achieved. TCIs have revolutionized the treatment paradigm for non-small-cell lung cancer and chronic lymphocytic leukemia. This Perspective will highlight the clinical and financial success of this class of drugs and provide early insight into toxicity, a key factor that had hindered progress in the field. Further innovation in the TCI approach, including expanding beyond cysteine-directed electrophiles, kinases, and cancer, highlights the broad opportunity to deliver a new generation of breakthrough therapies.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , Carcinoma, Non-Small-Cell Lung/drug therapy , Humans , Phosphotransferases , Protein Kinase Inhibitors/pharmacology
2.
Sci Signal ; 11(553)2018 10 23.
Article in English | MEDLINE | ID: mdl-30352947

ABSTRACT

This Editorial discusses the state of research on drug resistance in the fields of cancer, infectious disease, and agriculture. Reaching across the aisle for a more cross-collaborative approach may lead to exciting breakthroughs toward tackling the challenges of drug resistance in each field.


Subject(s)
Drug Resistance, Microbial , Drug Resistance, Neoplasm , Neoplasms/drug therapy , Agriculture , Communicable Diseases/drug therapy , Humans , Interdisciplinary Communication , Mutation , Stochastic Processes
3.
Mol Cancer Ther ; 13(6): 1468-79, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24723450

ABSTRACT

Patients with non-small cell lung carcinoma (NSCLC) with activating mutations in epidermal growth factor receptor (EGFR) initially respond well to the EGFR inhibitors erlotinib and gefitinib. However, all patients relapse because of the emergence of drug-resistant mutations, with T790M mutations accounting for approximately 60% of all resistance. Second-generation irreversible EGFR inhibitors are effective against T790M mutations in vitro, but retain affinity for wild-type EGFR (EGFR(WT)). These inhibitors have not provided compelling clinical benefit in T790M-positive patients, apparently because of dose-limiting toxicities associated with inhibition of EGFR(WT). Thus, there is an urgent clinical need for therapeutics that overcome T790M drug resistance while sparing EGFR(WT). Here, we describe a lead optimization program that led to the discovery of four potent irreversible 2,4-diaminopyrimidine compounds that are EGFR mutant (EGFR(mut)) selective and have been designed to have low affinity for EGFR(WT). Pharmacokinetic and pharmacodynamic studies in H1975 tumor-bearing mice showed that exposure was dose proportional resulting in dose-dependent EGFR modulation. Importantly, evaluation of normal lung tissue from the same animals showed no inhibition of EGFR(WT). Of all the compounds tested, compound 3 displayed the best efficacy in EGFR(L858R/T790M)-driven tumors. Compound 3, now renamed CO-1686, is currently in a phase I/II clinical trial in patients with EGFR(mut)-advanced NSCLC that have received prior EGFR-directed therapy.


Subject(s)
4-Aminopyridine/analogs & derivatives , Carcinoma, Non-Small-Cell Lung/drug therapy , ErbB Receptors/metabolism , Neoplasm Recurrence, Local/drug therapy , 4-Aminopyridine/administration & dosage , Animals , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Clinical Trials as Topic , Drug Resistance, Neoplasm/genetics , ErbB Receptors/antagonists & inhibitors , Humans , In Vitro Techniques , Mice , Mutation , Neoplasm Recurrence, Local/metabolism , Neoplasm Recurrence, Local/pathology , Xenograft Model Antitumor Assays
4.
Cancer Discov ; 3(12): 1404-15, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24065731

ABSTRACT

UNLABELLED: Patients with non-small cell lung cancer (NSCLC) with activating EGF receptor (EGFR) mutations initially respond to first-generation reversible EGFR tyrosine kinase inhibitors. However, clinical efficacy is limited by acquired resistance, frequently driven by the EGFR(T790M) mutation. CO-1686 is a novel, irreversible, and orally delivered kinase inhibitor that specifically targets the mutant forms of EGFR, including T790M, while exhibiting minimal activity toward the wild-type (WT) receptor. Oral administration of CO-1686 as single agent induces tumor regression in EGFR-mutated NSCLC tumor xenograft and transgenic models. Minimal activity of CO-1686 against the WT EGFR receptor was observed. In NSCLC cells with acquired resistance to CO-1686 in vitro, there was no evidence of additional mutations or amplification of the EGFR gene, but resistant cells exhibited signs of epithelial-mesenchymal transition and demonstrated increased sensitivity to AKT inhibitors. These results suggest that CO-1686 may offer a novel therapeutic option for patients with mutant EGFR NSCLC. SIGNIFICANCE: We report the preclinical development of a novel covalent inhibitor, CO-1686, that irreversibly and selectively inhibits mutant EGFR, in particular the T790M drug-resistance mutation, in NSCLC models. CO-1686 is the fi rst drug of its class in clinical development for the treatment of T790M-positive NSCLC, potentially offering potent inhibition of mutant EGFR while avoiding the on-target toxicity observed with inhibition of the WT EGFR.


Subject(s)
Acrylamides/pharmacology , Antineoplastic Agents/pharmacology , Carcinoma, Non-Small-Cell Lung/genetics , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/genetics , Lung Neoplasms/genetics , Protein Kinase Inhibitors/pharmacology , Pyrimidines/pharmacology , Acrylamides/administration & dosage , Administration, Oral , Animals , Antineoplastic Agents/administration & dosage , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Drug Resistance, Neoplasm , Drug Screening Assays, Antitumor , Epithelial-Mesenchymal Transition/drug effects , ErbB Receptors/metabolism , Female , HEK293 Cells , Humans , Lung Neoplasms/drug therapy , Lung Neoplasms/pathology , Mice , Mice, Inbred BALB C , Mice, Nude , Mice, Transgenic , Molecular Targeted Therapy , Mutant Proteins/antagonists & inhibitors , Mutant Proteins/metabolism , Protein Kinase Inhibitors/administration & dosage , Pyrimidines/administration & dosage , Xenograft Model Antitumor Assays
5.
J Pharmacol Exp Ther ; 346(2): 219-28, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23709115

ABSTRACT

Targeted therapies that suppress B cell receptor (BCR) signaling have emerged as promising agents in autoimmune disease and B cell malignancies. Bruton's tyrosine kinase (Btk) plays a crucial role in B cell development and activation through the BCR signaling pathway and represents a new target for diseases characterized by inappropriate B cell activity. N-(3-(5-fluoro-2-(4-(2-methoxyethoxy)phenylamino)pyrimidin-4-ylamino)phenyl)acrylamide (CC-292) is a highly selective, covalent Btk inhibitor and a sensitive and quantitative assay that measures CC-292-Btk engagement has been developed. This translational pharmacodynamic assay has accompanied CC-292 through each step of drug discovery and development. These studies demonstrate the quantity of Btk bound by CC-292 correlates with the efficacy of CC-292 in vitro and in the collagen-induced arthritis model of autoimmune disease. Recently, CC-292 has entered human clinical trials with a trial design that has provided rapid insight into safety, pharmacokinetics, and pharmacodynamics. This first-in-human healthy volunteer trial has demonstrated that a single oral dose of 2 mg/kg CC-292 consistently engaged all circulating Btk protein and provides the basis for rational dose selection in future clinical trials. This targeted covalent drug design approach has enabled the discovery and early clinical development of CC-292 and has provided support for Btk as a valuable drug target for B-cell mediated disorders.


Subject(s)
Acrylamides/pharmacology , Protein-Tyrosine Kinases/antagonists & inhibitors , Pyrimidines/pharmacology , Acrylamides/pharmacokinetics , Acrylamides/therapeutic use , Agammaglobulinaemia Tyrosine Kinase , Animals , Arthritis, Experimental/drug therapy , Arthritis, Experimental/immunology , Arthritis, Rheumatoid/drug therapy , Arthritis, Rheumatoid/immunology , B-Lymphocytes/drug effects , B-Lymphocytes/metabolism , Double-Blind Method , Humans , Mice , Pyrimidines/pharmacokinetics , Pyrimidines/therapeutic use , Receptors, Antigen, B-Cell/metabolism , Signal Transduction
6.
J Med Chem ; 56(3): 712-21, 2013 Feb 14.
Article in English | MEDLINE | ID: mdl-23360348

ABSTRACT

PI3Kα has been identified as an oncogene in human tumors. By use of rational drug design, a targeted covalent inhibitor 3 (CNX-1351) was created that potently and specifically inhibits PI3Kα. We demonstrate, using mass spectrometry and X-ray crystallography, that the selective inhibitor covalently modifies PI3Kα on cysteine 862 (C862), an amino acid unique to the α isoform, and that PI3Kß, -γ, and -δ are not covalently modified. 3 is able to potently (EC(50) < 100 nM) and specifically inhibit signaling in PI3Kα-dependent cancer cell lines, and this leads to a potent antiproliferative effect (GI(50) < 100 nM). A covalent probe, 8 (CNX-1220), which selectively bonds to PI3Kα, was used to investigate the duration of occupancy of 3 with PI3Kα in vivo. This is the first report of a PI3Kα-selective inhibitor, and these data demonstrate the biological impact of selectively targeting PI3Kα.


Subject(s)
Drug Discovery , Isoenzymes/antagonists & inhibitors , Phosphoinositide-3 Kinase Inhibitors , Protein Kinase Inhibitors/pharmacology , Animals , Male , Mass Spectrometry , Mice , Mice, Inbred C57BL , Nuclear Magnetic Resonance, Biomolecular , Phosphatidylinositol 3-Kinases/metabolism , Protein Kinase Inhibitors/chemistry , Signal Transduction
7.
Virology ; 436(1): 235-43, 2013 Feb 05.
Article in English | MEDLINE | ID: mdl-23260110

ABSTRACT

Interleukin 2-inducible T cell kinase (ITK) influences T cell signaling by coordinating actin polymerization and polarization as well as recruitment of kinases and adapter proteins. ITK regulates multiple steps of HIV-1 replication, including virion assembly and release. Fluorescent microscopy was used to examine the functional interactions between ITK and HIV-1 Gag during viral particle release. ITK and Gag colocalized at the plasma membrane and were concentrated at sites of F-actin accumulation and membrane lipid rafts in HIV-1 infected T cells. There was polarized staining of ITK, Gag, and actin towards sites of T cell conjugates. Small molecule inhibitors of ITK disrupted F-actin capping, perturbed Gag-ITK colocalization, inhibited virus like particle release, and reduced HIV replication in primary human CD4+ T cells. These data provide insight as to how ITK influences HIV-1 replication and suggest that targeting host factors that regulate HIV-1 egress provides an innovative strategy for controlling HIV infection.


Subject(s)
Actins/metabolism , HIV-1/physiology , Protein-Tyrosine Kinases/metabolism , gag Gene Products, Human Immunodeficiency Virus/metabolism , CD4-Positive T-Lymphocytes/virology , Cell Line, Tumor , Cell Membrane/metabolism , Cell Membrane/virology , HIV Infections/virology , HIV-1/genetics , HIV-1/metabolism , Humans , Interleukin-2/metabolism , Jurkat Cells , Membrane Microdomains , Protein-Tyrosine Kinases/antagonists & inhibitors , Signal Transduction , Virus Assembly , Virus Release , Virus Replication
8.
Nat Rev Drug Discov ; 10(4): 307-17, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21455239

ABSTRACT

Covalent drugs have proved to be successful therapies for various indications, but largely owing to safety concerns, they are rarely considered when initiating a target-directed drug discovery project. There is a need to reassess this important class of drugs, and to reconcile the discordance between the historic success of covalent drugs and the reluctance of most drug discovery teams to include them in their armamentarium. This review surveys the prevalence and pharmacological advantages of covalent drugs, discusses how potential risks and challenges may be addressed through innovative design, and presents the broad opportunities provided by targeted covalent inhibitors.


Subject(s)
Drug Delivery Systems , Drug Design , Pharmaceutical Preparations/metabolism , Animals , Drug Discovery/methods , Drug-Related Side Effects and Adverse Reactions , Humans , Pharmaceutical Preparations/chemistry , Structure-Activity Relationship
9.
Nat Chem Biol ; 7(1): 22-4, 2011 Jan.
Article in English | MEDLINE | ID: mdl-21113170

ABSTRACT

Designing selective inhibitors of proteases has proven problematic, in part because pharmacophores that confer potency exploit the conserved catalytic apparatus. We developed a fundamentally different approach by designing irreversible inhibitors that target noncatalytic cysteines that are structurally unique to a target in a protein family. We have successfully applied this approach to the important therapeutic target HCV protease, which has broad implications for the design of other selective protease inhibitors.


Subject(s)
Cysteine Proteinase Inhibitors/therapeutic use , Cysteine/antagonists & inhibitors , Drug Design , Oligopeptides/therapeutic use , Biocatalysis , Biochemistry/methods , Crystallography, X-Ray , Cysteine/metabolism , Cysteine Proteinase Inhibitors/chemistry , Cysteine Proteinase Inhibitors/pharmacology , Hepacivirus/drug effects , Hepacivirus/enzymology , Hepacivirus/growth & development , Oligopeptides/chemistry , Oligopeptides/pharmacology , Virology/methods
10.
Curr Opin Chem Biol ; 14(4): 475-80, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20609616

ABSTRACT

In the past decade tremendous progress has been made toward a new class of therapeutics termed 'targeted covalent drugs', in which structure-based approaches are employed to create small molecules that inactivate their protein target through targeted covalent attachment to a specific cysteine. In the kinase field, this approach is demonstrating promise in overcoming the potency, selectivity, and efficacy challenges currently faced by reversible kinase inhibitors, with several advancing into late stage clinical testing. This design paradigm has been successfully applied to making drug candidates for epidermal growth factor receptor (EGFR), Her2, and Bruton's tyrosine kinase (Btk). Here we review recent pre-clinical and clinical advances with targeted covalent kinase inhibitors, and the potential for broader application of the approach.


Subject(s)
Drug Delivery Systems , Phosphotransferases/chemistry , Phosphotransferases/pharmacology , Computational Biology , Enzyme Inhibitors/administration & dosage , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Humans , Molecular Structure , Phosphotransferases/administration & dosage , Structure-Activity Relationship
11.
J Chem Inf Model ; 49(5): 1185-92, 2009 May.
Article in English | MEDLINE | ID: mdl-19415918

ABSTRACT

The desire to exploit structural information to aid structure based design and virtual screening led to the development of the interaction fingerprint for analyzing, mining, and filtering the binding patterns underlying the complex 3D data. In this paper we introduce a new approach, weighted SIFt (or w-SIFt), extending the concept of SIFt to capture the relative importance of different binding interactions. The methodology presented here for determining the weights in w-SIFt involves utilizing a dimensionality reduction technique for eliminating linear redundancies in the data followed by a stochastic optimization. We find that the relative weights of the fingerprint bits provide insight into what interactions are critical in determining inhibitor potency. Moreover, the weighted interaction fingerprint can serve as an interpretable position dependent scoring function for ligand protein interactions.


Subject(s)
Proteins/chemistry , Ligands , Models, Molecular , Quantitative Structure-Activity Relationship
12.
Arterioscler Thromb Vasc Biol ; 28(4): 665-71, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18202322

ABSTRACT

OBJECTIVE: TGF-beta plays a significant role in vascular injury-induced stenosis. This study evaluates the efficacy of a novel, small molecule inhibitor of ALK5/ALK4 kinase, in the rat carotid injury model of vascular fibrosis. METHODS AND RESULTS: The small molecule, SM16, was shown to bind with high affinity to ALK5 kinase ATP binding site using a competitive binding assay and biacore analysis. SM16 blocked TGF-beta and activin-induced Smad2/3 phosphorylation and TGF-beta-induced plasminogen activator inhibitor (PAI)-luciferase activity in cells. Good overall selectivity was demonstrated in a large panel of kinase assays, but SM16 also showed nanomolar inhibition of ALK4 and weak (micromolar) inhibition of Raf and p38. In the rat carotid injury model, SM16 dosed once daily orally at 15 or 30 mg/kg SM16 for 14 days caused significant inhibition of neointimal thickening and lumenal narrowing. SM16 also prevented induction of adventitial smooth muscle alpha-actin-positive myofibroblasts and the production of intimal collagen, but did not decrease the percentage of proliferative cells. CONCLUSIONS: These results are the first to demonstrate the efficacy of an orally active, small-molecule ALK5/ALK4 inhibitor in a vascular fibrosis model and suggest the potential therapeutic application of these inhibitors in vascular fibrosis.


Subject(s)
Azabicyclo Compounds/pharmacology , Carotid Artery Injuries/drug therapy , Protein Kinase Inhibitors/pharmacology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Receptors, Transforming Growth Factor beta/antagonists & inhibitors , Activin Receptors, Type I/antagonists & inhibitors , Adenosine Triphosphate/metabolism , Administration, Oral , Animals , Azabicyclo Compounds/administration & dosage , Azabicyclo Compounds/metabolism , Binding Sites , Carotid Artery Injuries/pathology , Carotid Artery Injuries/physiopathology , Cell Line , Fibroblasts/drug effects , Fibroblasts/pathology , Fibrosis , Humans , Male , Myoblasts, Smooth Muscle/drug effects , Myoblasts, Smooth Muscle/pathology , Protein Kinase Inhibitors/administration & dosage , Protein Kinase Inhibitors/metabolism , Rats , Rats, Sprague-Dawley , Receptor, Transforming Growth Factor-beta Type I , Transforming Growth Factor beta/physiology
13.
Cancer Res ; 67(5): 2351-9, 2007 Mar 01.
Article in English | MEDLINE | ID: mdl-17332368

ABSTRACT

Malignant mesothelioma is an aggressive and lethal pleural cancer that overexpresses transforming growth factor beta (TGFbeta). We investigated the efficacy of a novel small-molecule TGFbeta type I receptor (ALK5) kinase inhibitor, SM16, in the AB12 syngeneic model of malignant mesothelioma. SM16 inhibited TGFbeta signaling seen as decreased phosphorylated Smad2/3 levels in cultured AB12 cells (IC(50), approximately 200 nmol/L). SM16 penetrated tumor cells in vivo, suppressing tumor phosphorylated Smad2/3 levels for at least 3 h following treatment of tumor-bearing mice with a single i.p. bolus of 20 mg/kg SM16. The growth of established AB12 tumors was significantly inhibited by 5 mg/kg/d SM16 (P < 0.001) delivered via s.c. miniosmotic pumps over 28 days. The efficacy of SM16 was a result of a CD8+ antitumor response because (a) the antitumor effects were markedly diminished in severe combined immunodeficient mice and (b) CD8+ T cells isolated from spleens of mice treated with SM16 showed strong antitumor cytolytic effects whereas CD8+ T cells isolated from spleens of tumor-bearing mice treated with control vehicle showed minimal activity. Treatment of mice bearing large tumors with 5 mg/kg/d SM16 after debulking surgery reduced the extent of tumor recurrence from 80% to <20% (P < 0.05). SM16 was also highly effective in blocking and regressing tumors when given p.o. at doses of 0.45 or 0.65 g/kg in mouse chow. Thus, SM16 shows potent activity against established AB12 malignant mesothelioma tumors using an immune-mediated mechanism and can significantly prevent tumor recurrence after resection of bulky AB12 malignant mesothelioma tumors. These data suggest that ALK5 inhibitors, such as SM16, offer significant potential for the treatment of malignant mesothelioma and possibly other cancers.


Subject(s)
Activin Receptors, Type I/antagonists & inhibitors , Mesothelioma/drug therapy , Mesothelioma/surgery , Neoplasm Recurrence, Local/prevention & control , Protein Kinase Inhibitors/therapeutic use , Receptors, Transforming Growth Factor beta/antagonists & inhibitors , Animals , Cell Proliferation/drug effects , Disease-Free Survival , Drug Evaluation, Preclinical , Female , Humans , Immunity, Cellular/drug effects , Mesothelioma/pathology , Mice , Mice, Inbred BALB C , Mice, SCID , Neoplasm Transplantation , Protein Serine-Threonine Kinases , Receptor, Transforming Growth Factor-beta Type I , Tumor Cells, Cultured
14.
Chem Biol Drug Des ; 67(6): 385-94, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16882313

ABSTRACT

Lead identification by high-throughput screening of large compound libraries has been supplemented with virtual screening and focused compound libraries. To complement existing approaches for lead identification at Biogen Idec, a kinase-focused compound collection was designed, developed and validated. Two strategies were adopted to populate the compound collection: a ligand shape-based virtual screening and a receptor-based approach (structural interaction fingerprint). Compounds selected with the two approaches were cherry-picked from an existing high-throughput screening compound library, ordered from suppliers and supplemented with specific medicinal compounds from internal programs. Promising hits and leads have been generated from the kinase-focused compound collection against multiple kinase targets. The principle of the collection design and screening strategy was validated and the use of the kinase-focused compound collection for lead identification has been added to existing strategies.


Subject(s)
Drug Evaluation, Preclinical/methods , Phosphotransferases/metabolism , Combinatorial Chemistry Techniques , Databases, Protein , Inhibitory Concentration 50 , Ligands , Models, Molecular , Phosphotransferases/antagonists & inhibitors , Reproducibility of Results , Structure-Activity Relationship
15.
J Invest Dermatol ; 126(8): 1733-44, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16741519

ABSTRACT

Fibrosis, the hallmark of scleroderma, is characterized by excessive synthesis of collagen and extracellular matrix proteins and accumulation of myofibroblasts. Transforming growth factor-beta (TGF-beta), a potent inducer of collagen synthesis, cytokine production, and myofibroblast transdifferentiation, is implicated in fibrosis. Profibrotic TGF-beta responses are induced primarily via the type I activin-like receptor kinase 5 (ALK5) TGF-beta receptor coupled to Smad signal transducers. Here, we investigated the effect of blocking ALK5 function with SM305, a novel small-molecule kinase inhibitor, on fibrotic TGF-beta responses. In normal dermal fibroblasts, SM305 abrogated the ligand-induced phosphorylation, nuclear import, and DNA-binding activity of Smad2/3 and Smad4, and inhibited Smad2/3-dependent transcriptional responses. Furthermore, SM305 blocked TGF-beta-induced extracellular matrix gene expression, cytokine production, and myofibroblast transdifferentiation. In unstimulated scleroderma fibroblasts, SM305 caused a variable and modest reduction in type I collagen levels, and failed to abrogate constitutive nuclear accumulation of Smad2/3, or alter the proportion of smooth muscle actin stress fiber-positive fibroblasts. In vivo, SM305 prevented TGF-beta-induced Smad2/3 phosphorylation type I collagen (COL1)A2 promoter activation in dermal fibroblasts. Taken together, these results indicate that SM305 inhibits intracellular TGF-beta signaling through selective interference with ALK5-mediated Smad activation, resulting in marked suppression of profibrotic responses induced by TGF-beta in vivo and in vitro.


Subject(s)
Protein Kinase Inhibitors/pharmacology , Pyrazoles/pharmacology , Quinolines/pharmacology , Receptors, Transforming Growth Factor beta/antagonists & inhibitors , Scleroderma, Systemic/drug therapy , Scleroderma, Systemic/metabolism , Smad Proteins/metabolism , Animals , Biopsy , Cell Division/drug effects , Cells, Cultured , Dermis/cytology , Fibroblasts/cytology , Fibroblasts/metabolism , Humans , In Vitro Techniques , Mice , Mice, Transgenic , Receptors, Transforming Growth Factor beta/metabolism , Scleroderma, Systemic/pathology , Smad2 Protein/metabolism , Smad3 Protein/metabolism , Smad4 Protein/metabolism
16.
Chem Biol Drug Des ; 67(1): 5-12, 2006 Jan.
Article in English | MEDLINE | ID: mdl-16492144

ABSTRACT

The combination of advances in structure-based drug design efforts in the pharmaceutical industry in parallel with structural genomics initiatives in the public domain has led to an explosion in the number of structures of protein-small molecule complexes structures. This information has critical importance to both the understanding of the structural basis for molecular recognition in biological systems and the design of better drugs. A significant challenge exists in managing this vast amount of data and fully leveraging it. Here, we review our work to develop a simple, fast way to store, organize, mine, and analyze large numbers of protein-small molecule complexes. We illustrate the utility of the approach to the management of inhibitor complexes from the protein kinase family. Finally, we describe our recent efforts in applying this method to the design of target-focused chemical libraries.


Subject(s)
Enzyme Inhibitors/chemistry , Structure-Activity Relationship , Amino Acid Sequence , Animals , Binding Sites , Conserved Sequence , Drug Interactions , Enzyme Inhibitors/metabolism , Humans , Ligands , Models, Biological , Models, Molecular , Molecular Sequence Data , Protein Binding , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/metabolism , Protein Kinases/metabolism
17.
J Med Chem ; 49(2): 490-500, 2006 Jan 26.
Article in English | MEDLINE | ID: mdl-16420036

ABSTRACT

Here we present a new strategy for designing and filtering potentially massive combinatorial libraries using structural information of a binding site. We have developed a variation of the structural interaction fingerprint (SIFt) named r-SIFt, which incorporates the binding interactions of variable fragments in a combinatorial library. This method takes into account the 3D structure of the active site of the target molecule and translates desirable ligand-target binding interactions into library filtering constraints. We show using the MAP kinase p38 as a test case that we can efficiently analyze and classify compounds on the basis of their abilities to interact with the target in the desired binding mode. On the basis of these classifications, decision tree models were generated using the molecular descriptors of the compounds as predictor variables. Our results suggest that r-SIFt coupled with the classification models should be a valuable tool for structure-based focusing of combinatorial chemical libraries.


Subject(s)
Combinatorial Chemistry Techniques , Databases, Factual , Knowledge Bases , Ligands , Proteins/chemistry , Quantitative Structure-Activity Relationship , Binding Sites , Models, Molecular , Molecular Structure , Protein Binding
18.
J Med Chem ; 48(1): 121-33, 2005 Jan 13.
Article in English | MEDLINE | ID: mdl-15634006

ABSTRACT

A major challenge facing structure-based drug discovery efforts is how to leverage the massive amount of experimental (X-ray and NMR) and virtual structural information generated from drug discovery projects. Many important drug targets have large numbers of protein-inhibitor complexes, necessitating tools to compare and contrast their similarities and differences. This information would be valuable for understanding potency and selectivity of inhibitors and could be used to define target constraints to assist virtual screening. We describe a profile-based approach that enables us to capture the conservation of interactions between a set of protein-ligand receptor complexes. The use of profiles provides a sensitive means to compare multiple inhibitors binding to a drug target. We demonstrate the utility of profile-based analysis of small molecule complexes from the protein-kinase family to identify similarities and differences in binding of ATP, p38, and CDK2 compounds to kinases and how these profiles can be applied to differentiate the selectivity of these inhibitors. Importantly, our virtual screening results demonstrate superior enrichment of kinase inhibitors using profile-based methods relative to traditional scoring functions. Interaction-based analysis should provide a valuable tool for understanding inhibitor binding to other important drug targets.


Subject(s)
Drug Evaluation, Preclinical/methods , Protein Kinase Inhibitors/metabolism , Protein Kinases/metabolism , Adenosine Triphosphate/metabolism , Algorithms , CDC2-CDC28 Kinases/chemistry , CDC2-CDC28 Kinases/metabolism , Cyclin-Dependent Kinase 2 , Databases, Factual , Databases, Protein , Models, Molecular , Models, Theoretical , Protein Kinase Inhibitors/chemistry , Protein Kinases/chemistry , Sensitivity and Specificity , Structure-Activity Relationship , p38 Mitogen-Activated Protein Kinases/chemistry , p38 Mitogen-Activated Protein Kinases/metabolism
19.
Curr Top Med Chem ; 4(14): 1497-507, 2004.
Article in English | MEDLINE | ID: mdl-15544540

ABSTRACT

Asthma, a chronic inflammatory disease of the airways, is a significant burden on our healthcare system. There is high unmet need for treatments directed towards the underlying causes of the disease. The cell surface integrin VLA-4 (very late antigen-4; alpha4beta1; CD49d/CD29) plays an important role in the trafficking of white blood cells to sites of inflammation and represents an exciting target for the development of novel anti-inflammatory drugs for the treatment of asthma. Here, we review our efforts to use rational design to identify potent, selective inhibitors of VLA-4. We describe the discovery of a series of potent VLA-4 inhibitors through the addition of a novel N-terminal organic cap to a tetrapeptide VLA-4 binding motif 4-((N'-2-methylphenyl)uriedo)phenylacetyl-Leu-Asp-Val-Pro ; Kd = 70 pM), and rationalize their structure-activity relationships using 3D-QSAR. Also, we show our rational peptidomimetic design strategy using "template hopping" from the gpIIb/IIIa integrin antagonist field, and also a novel virtual screening strategy. Two series have been developed, one that has high selectivity for the activated over the non-activated state of the receptor, and the other which is non-selective inhibiting both activated and non-activated VLA-4. Both series are highly selective for VLA-4 versus against other integrin family members. These inhibitors show promise in the treatment of asthma, based upon efficacy in a sheep model of asthma, where they inhibit both the early and late-phase responses to asthma and also block hypersensitivity.


Subject(s)
Asthma/drug therapy , Integrin alpha4beta1/antagonists & inhibitors , Amides/chemistry , Amides/pharmacology , Amides/therapeutic use , Animals , Drug Design , Humans , Peptides/chemistry , Peptides/pharmacology , Peptides/therapeutic use , Structure-Activity Relationship
20.
Curr Opin Drug Discov Devel ; 7(4): 437-45, 2004 Jul.
Article in English | MEDLINE | ID: mdl-15338953

ABSTRACT

The pathological activation of the transforming growth factor beta (TGFbeta) pathway plays a critical role in the progression of fibrotic diseases and also enhances tumor invasiveness and metastasis. Due to its central role in TGFbeta signaling, the TGFbeta type I receptor (TbetaRI) is emerging as an exciting target for blockade of the TGFbeta pathway. In this review we will discuss how three independent drug discovery strategies, ie, target-hopping, high-throughput screening and virtual screening, have converged in the identification of inhibitors of TalphaRI kinase. Structural studies have provided insight into the potency and selectivity of these inhibitors and form the basis for structure-based design optimization strategies. These efforts have enabled the production of potent, selective inhibitors for dissecting the TGFalpha pathway and assessing the usefulness of TalphaRI blockade in the treatment of fibrotic diseases and cancer.


Subject(s)
Activin Receptors, Type I/therapeutic use , Receptors, Transforming Growth Factor beta/metabolism , Receptors, Transforming Growth Factor beta/therapeutic use , Transforming Growth Factor beta/metabolism , Activin Receptors, Type I/pharmacology , Amino Acid Sequence , Clinical Trials, Phase I as Topic , Clinical Trials, Phase II as Topic , Humans , Models, Biological , Molecular Sequence Data , Molecular Structure , Protein Isoforms , Protein Serine-Threonine Kinases , Receptor, Transforming Growth Factor-beta Type I , Receptors, Transforming Growth Factor beta/antagonists & inhibitors , Signal Transduction/physiology , Transforming Growth Factor beta/antagonists & inhibitors , Transforming Growth Factor beta/therapeutic use
SELECTION OF CITATIONS
SEARCH DETAIL
...