Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
1.
Proc Natl Acad Sci U S A ; 111(42): 15072-7, 2014 Oct 21.
Article in English | MEDLINE | ID: mdl-25288762

ABSTRACT

Necroptosis is considered to be complementary to the classical caspase-dependent programmed cell death pathway, apoptosis. The pseudokinase Mixed Lineage Kinase Domain-Like (MLKL) is an essential effector protein in the necroptotic cell death pathway downstream of the protein kinase Receptor Interacting Protein Kinase-3 (RIPK3). How MLKL causes cell death is unclear, however RIPK3-mediated phosphorylation of the activation loop in MLKL trips a molecular switch to induce necroptotic cell death. Here, we show that the MLKL pseudokinase domain acts as a latch to restrain the N-terminal four-helix bundle (4HB) domain and that unleashing this domain results in formation of a high-molecular-weight, membrane-localized complex and cell death. Using alanine-scanning mutagenesis, we identified two clusters of residues on opposing faces of the 4HB domain that were required for the 4HB domain to kill cells. The integrity of one cluster was essential for membrane localization, whereas MLKL mutations in the other cluster did not prevent membrane translocation but prevented killing; this demonstrates that membrane localization is necessary, but insufficient, to induce cell death. Finally, we identified a small molecule that binds the nucleotide binding site within the MLKL pseudokinase domain and retards MLKL translocation to membranes, thereby preventing necroptosis. This inhibitor provides a novel tool to investigate necroptosis and demonstrates the feasibility of using small molecules to target the nucleotide binding site of pseudokinases to modulate signal transduction.


Subject(s)
Apoptosis , Necrosis , Protein Kinases/metabolism , Adenosine Triphosphate/metabolism , Amino Acid Motifs , Amino Acid Sequence , Animals , Binding Sites , Cell Membrane/metabolism , Enzyme Activation , Inhibitory Concentration 50 , Mice , Mice, Transgenic , Molecular Sequence Data , Mutation , Phosphorylation , Protein Structure, Secondary , Protein Structure, Tertiary , Protein Transport , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Recombinant Proteins/metabolism , Sequence Homology, Amino Acid
2.
J Pharmacol Exp Ther ; 339(3): 799-806, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21917561

ABSTRACT

The orally active microtubule-disrupting agent (S)-1-ethyl-3-(2-methoxy-4-(5-methyl-4-((1-(pyridin-3-yl)butyl)amino)pyrimidin-2-yl)phenyl)urea (CYT997), reported previously by us (Bioorg Med Chem Lett 19:4639-4642, 2009; Mol Cancer Ther 8:3036-3045, 2009), is potently cytotoxic to a variety of cancer cell lines in vitro and shows antitumor activity in vivo. In addition to its cytotoxic activity, CYT997 possesses antivascular effects on tumor vasculature. To further characterize the vascular disrupting activity of CYT997 in terms of dose and temporal effects, we studied the activity of the compound on endothelial cells in vitro and on tumor blood flow in vivo by using a variety of techniques. In vitro, CYT997 is shown to potently inhibit the proliferation of vascular endothelial growth factor-stimulated human umbilical vein endothelial cells (IC(50) 3.7 Ā± 1.8 nM) and cause significant morphological changes at 100 nM, including membrane blebbing. Using the method of corrosion casting visualized with scanning electron microscopy, a single dose of CYT997 (7.5 mg/kg i.p.) in a metastatic cancer model was shown to cause destruction of tumor microvasculature in metastatic lesions. Furthermore, repeat dosing of CYT997 at 10 mg/kg and above (intraperitoneally, b.i.d.) was shown to effectively inhibit development of liver metastases. The time and dose dependence of the antivascular effects were studied in a DLD-1 colon adenocarcinoma xenograft model using the fluorescent dye Hoechst 33342. CYT997 demonstrated rapid and dose-dependent vascular shutdown, which persists for more than 24 h after a single oral dose. Together, the data demonstrate that CYT997 possesses potent antivascular activity and support continuing development of this promising compound.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Antineoplastic Agents/pharmacology , Colonic Neoplasms/blood supply , Neovascularization, Pathologic/drug therapy , Pyridines/pharmacology , Pyrimidines/pharmacology , Tubulin Modulators/pharmacology , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Animals , Cell Line, Tumor , Cell Proliferation/drug effects , Colonic Neoplasms/drug therapy , Colonic Neoplasms/pathology , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Human Umbilical Vein Endothelial Cells , Humans , Liver Neoplasms/drug therapy , Liver Neoplasms/pathology , Liver Neoplasms/secondary , Male , Mice , Mice, Nude , Time Factors , Xenograft Model Antitumor Assays
3.
J Leukoc Biol ; 85(2): 278-88, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19004987

ABSTRACT

M-CSF/CSF-1 supports the proliferation and differentiation of monocytes and macrophages. In mice, CSF-1 also promotes proinflammatory responses in vivo by regulating mature macrophage functions, but little is known about the acute effects of this growth factor on mature human macrophages. Here, we show that in contrast to its effects on mouse bone marrow-derived macrophages, CSF-1 did not induce expression of urokinase plasminogen activator mRNA, repress expression of apolipoprotein E mRNA, or prime LPS-induced TNF and IL-6 secretion in human monocyte-derived macrophages (HMDM) from several independent donors. Instead, we show by expression profiling that CSF-1 modulates the HMDM transcriptome to favor a proatherogenic environment. CSF-1 induced expression of the proatherogenic chemokines CXCL10/IFN-inducible protein 10, CCL2, and CCL7 but repressed expression of the antiatherogenic chemokine receptor CXCR4. CSF-1 also up-regulated genes encoding enzymes of the cholesterol biosynthetic pathway (HMGCR, MVD, IDI1, FDPS, SQLE, CYP51A1, EBP, NSDHL, DHCR7, and DHCR24), and expression of ABCG1, encoding a cholesterol efflux transporter, was repressed. Consistent with these effects, CSF-1 increased levels of free cholesterol in HMDM, and the selective CSF-1R kinase inhibitor GW2580 ablated this response. These data demonstrate that CSF-1 represents a further link between inflammation and cardiovascular disease and suggest two distinct mechanisms by which CSF-1, which is known to be present in atherosclerotic lesions, may contribute to plaque progression.


Subject(s)
Atherosclerosis/immunology , Macrophage Colony-Stimulating Factor/immunology , Macrophages/immunology , Animals , Autocrine Communication , Bone Marrow Cells/cytology , Chemokines/immunology , Cholesterol/biosynthesis , Down-Regulation , Humans , Lipid Metabolism , Macrophages/enzymology , Male , Mice , Monocytes/cytology , Receptors, CXCR4/genetics , Receptors, CXCR4/metabolism
6.
Bioorg Med Chem Lett ; 19(20): 5887-92, 2009 Oct 15.
Article in English | MEDLINE | ID: mdl-19762238

ABSTRACT

A series of phenylaminopyrimidines has been identified as inhibitors of Janus kinases (JAKs). Development of this initial series led to the potent JAK2/JAK1 inhibitor CYT387 (N-(cyanomethyl)-4-[2-[[4-(4-morpholinyl)phenyl]amino]-4-pyrimidinyl]-benzamide). Details of synthesis and SAR studies of these compounds are reported.


Subject(s)
Benzamides/chemistry , Janus Kinase 2/antagonists & inhibitors , Protein Kinase Inhibitors/chemistry , Pyrimidines/chemistry , Animals , Benzamides/chemical synthesis , Benzamides/pharmacology , Binding Sites , Caco-2 Cells , Cell Line, Tumor , Computer Simulation , Humans , Janus Kinase 2/metabolism , Male , Protein Isoforms/antagonists & inhibitors , Protein Isoforms/metabolism , Protein Kinase Inhibitors/chemical synthesis , Protein Kinase Inhibitors/pharmacokinetics , Pyrimidines/chemical synthesis , Pyrimidines/pharmacokinetics , Pyrimidines/pharmacology , Rats , Rats, Sprague-Dawley , Structure-Activity Relationship
7.
J Mol Biol ; 367(3): 839-47, 2007 Mar 30.
Article in English | MEDLINE | ID: mdl-17292918

ABSTRACT

c-Fms, a member of the Platelet-derived Growth Factor (PDGF) receptor family of receptor tyrosine kinases (RTKs), is the receptor for macrophage colony stimulating factor (CSF-1) that regulates proliferation, differentiation and survival of cells of the mononuclear phagocyte lineage. Abnormal expression of c-fms proto-oncogene is associated with a significant number of human pathologies, including a variety of cancers and rheumatoid arthritis. Accordingly, c-Fms represents an attractive therapeutic target. To further understand the regulation of c-Fms, we determined the 2.7 A resolution crystal structure of the cytosolic domain of c-Fms that comprised the kinase domain and the juxtamembrane domain. The structure reveals the crucial inhibitory role of the juxtamembrane domain (JM) that binds to a hydrophobic site immediately adjacent to the ATP binding pocket. This interaction prevents the activation loop from adopting an active conformation thereby locking the c-Fms kinase into an autoinhibited state. As observed for other members of the PDGF receptor family, namely c-Kit and Flt3, three JM-derived tyrosine residues primarily drive the mechanism for autoinhibition in c-Fms, therefore defining a common autoinhibitory mechanism within this family. Moreover the structure provides an understanding of c-Fms inhibition by Gleevec as well as providing a platform for the development of more selective inhibitors that target the inactive conformation of c-Fms kinase.


Subject(s)
Receptor, Macrophage Colony-Stimulating Factor/antagonists & inhibitors , Receptor, Macrophage Colony-Stimulating Factor/chemistry , Amino Acid Sequence , Benzamides , Crystallography, X-Ray , Humans , Imatinib Mesylate , In Vitro Techniques , Models, Molecular , Molecular Sequence Data , Piperazines/pharmacology , Protein Kinase Inhibitors/pharmacology , Protein Structure, Tertiary , Proto-Oncogene Mas , Pyrimidines/pharmacology , Receptor, Macrophage Colony-Stimulating Factor/genetics , Recombinant Proteins/antagonists & inhibitors , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Sequence Homology, Amino Acid
8.
J Cardiovasc Pharmacol ; 52(5): 403-12, 2008 Nov.
Article in English | MEDLINE | ID: mdl-19033819

ABSTRACT

Cardiovascular disease is a major cause of mortality with the underlying process being atherosclerosis. Modified proteoglycans bind low-density lipoproteins (LDL), a critical initial step in the atherosclerotic cascade, representing a potential therapeutic target. Platelet-derived growth factor (PDGF) stimulates proteoglycan synthesis and is strongly implicated in atherogenesis. In human vascular smooth muscle cells (VSMCs), CYC10424 (Cytopia Research Ltd), a pyrido-pyrimidine derivative, dose-dependently decreased PDGF-mediated radiolabel incorporation into proteoglycans associated with an increase in electrophoretic mobility by SDS-PAGE. PDGF stimulated increases in both chemically-cleaved and xyloside-associated glycosaminoglycan (GAG) chain size, which were inhibited in the presence of CYC10424 by size exclusion chromatography (Sepharose CL-6B). CYC10424 treatment inhibited the PDGF effect to increase the 6:4 position sulfation ratio of monosulfated disaccharides by fluorophore-assisted carbohydrate electrophoresis. Proteoglycans derived from cells treated with CYC10424 had a decreased binding affinity and capacity to human LDL by gel mobility shift assay. CYC10424 and related compounds are possible candidates as therapeutic agents for the prevention of lipid deposition as characteristic of diseases such as atherosclerosis.


Subject(s)
Glycosaminoglycans/antagonists & inhibitors , Lipoproteins, LDL/metabolism , Muscle, Smooth, Vascular/drug effects , Phenylurea Compounds/pharmacology , Proteoglycans/metabolism , Pyrimidines/pharmacology , Atherosclerosis/metabolism , Atherosclerosis/prevention & control , Cell Line , Electrophoresis, Polyacrylamide Gel , Electrophoretic Mobility Shift Assay , Glycosaminoglycans/biosynthesis , Humans , Molecular Structure , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/metabolism , Phenylurea Compounds/chemical synthesis , Phenylurea Compounds/chemistry , Platelet-Derived Growth Factor/metabolism , Protein Binding , Pyrimidines/chemical synthesis , Pyrimidines/chemistry
9.
FASEB J ; 20(11): 1921-3, 2006 Sep.
Article in English | MEDLINE | ID: mdl-16877523

ABSTRACT

CSF-1 regulates macrophage differentiation, survival, and function, and is an attractive therapeutic target for chronic inflammation and malignant diseases. Here we describe the effects of a potent and selective inhibitor of CSF-1R-CYC10268-on CSF-1R-dependent signaling. In in vitro kinase assays, CYC10268 was active in the low nanomolar range and showed selectivity over other kinases such as Abl and Kit. CYC10268 blocked survival mediated by CSF-1R in primary murine bone marrow-derived macrophages (BMM) and in the factor-dependent cell line Ba/F3, in which the CSF-1R was ectopically expressed. CYC10268 also inhibited CSF-1 regulated signaling (Akt, ERK-1/2), gene expression (urokinase plasminogen activator, toll-like receptor 9, and apolipoprotein E), and priming of LPS-inducible cytokine production in BMM. In thioglycollate-elicited peritoneal macrophages (TEPM), which survive in the absence of exogenous CSF-1, CYC10268 impaired LPS-induced cytokine production and regulated expression of known CSF-1 target genes. These observations support the conclusion that TEPM are CSF-1 autocrine and that CSF-1 plays a central role in macrophage effector functions during inflammation. CSF-1R inhibitors such as CYC10268 provide a powerful tool to dissect the role of the CSF-1/CSF-1R signaling system in a range of biological systems and have potential for a number of therapeutic applications.


Subject(s)
Inflammation/prevention & control , Macrophage Colony-Stimulating Factor/physiology , Macrophages/physiology , Receptor, Macrophage Colony-Stimulating Factor/antagonists & inhibitors , Animals , Cell Line , Cell Survival , Cloning, Molecular , Cytokines/antagonists & inhibitors , Humans , Inflammation/immunology , Macrophages/cytology , Macrophages/immunology , Mice , Mutant Chimeric Proteins/antagonists & inhibitors , Mutant Chimeric Proteins/metabolism , Plasmids , Receptor Protein-Tyrosine Kinases/metabolism , Receptor, Macrophage Colony-Stimulating Factor/genetics , Transfection
10.
Oncotarget ; 8(35): 57948-57963, 2017 Aug 29.
Article in English | MEDLINE | ID: mdl-28938529

ABSTRACT

Neutropenia represents one of the major dose-limiting toxicities of many current cancer therapies. To circumvent the off-target effects of cytotoxic chemotherapeutics, kinase inhibitors are increasingly being used as an adjunct therapy to target leukemia. In this study, we conducted a screen of leukemic cell lines in parallel with primary neutrophils to identify kinase inhibitors with the capacity to induce apoptosis of myeloid and lymphoid cell lines whilst sparing primary mouse and human neutrophils. We have utilized a high-throughput live cell imaging platform to demonstrate that cytotoxic drugs have limited effects on neutrophil viability but are toxic to hematopoietic progenitor cells, with the exception of the topoisomerase I inhibitor SN-38. The parallel screening of kinase inhibitors revealed that mouse and human neutrophil viability is dependent on cyclin-dependent kinase (CDK) activity but surprisingly only partially dependent on PI3 kinase and JAK/STAT signaling, revealing dominant pathways contributing to neutrophil viability. Mcl-1 haploinsufficiency sensitized neutrophils to CDK inhibition, demonstrating that Mcl-1 is a direct target for CDK inhibitors. This study reveals a therapeutic window for the kinase inhibitors BEZ235, BMS-3, AZD7762, and (R)-BI-2536 to induce apoptosis of leukemia cell lines whilst maintaining immunocompetence and hemostasis.

11.
ACS Med Chem Lett ; 8(12): 1298-1303, 2017 Dec 14.
Article in English | MEDLINE | ID: mdl-29259751

ABSTRACT

A number of diazepines are known to inhibit bromo- and extra-terminal domain (BET) proteins. Their BET inhibitory activity derives from the fusion of an acetyl-lysine mimetic heterocycle onto the diazepine framework. Herein we describe a straightforward, modular synthesis of novel 1,2,3-triazolobenzodiazepines and show that the 1,2,3-triazole acts as an effective acetyl-lysine mimetic heterocycle. Structure-based optimization of this series of compounds led to the development of potent BET bromodomain inhibitors with excellent activity against leukemic cells, concomitant with a reduction in c-MYC expression. These novel benzodiazepines therefore represent a promising class of therapeutic BET inhibitors.

12.
Methods Mol Biol ; 967: 99-113, 2013.
Article in English | MEDLINE | ID: mdl-23296724

ABSTRACT

The JAK family of protein tyrosine kinases are now recognized as important participants in a wide range of pathologies, from cancer to inflammatory diseases. In the last decade, the drive to develop drugs targeting members of this family has begun to deliver a panel of small molecule inhibitors of JAK family members, with a range of potencies and specificities. A number of these compounds have already found widespread use as biochemical tools in the elucidation of JAK activity in specific signaling and disease processes; however, many of the first generation compounds are poorly characterized with suboptimal potencies and selectivities.Herein, we present the data for those small molecule JAK inhibitors that have been described in the peer-reviewed literature and the benefits and potential issues that may be associated with the use of these tool compounds.


Subject(s)
Biology/methods , Chemistry/methods , Janus Kinases/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , Drug Evaluation, Preclinical , Humans , Protein Kinase Inhibitors/chemistry
13.
Future Med Chem ; 4(18): 2295-310, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23234552

ABSTRACT

There is an urgent need for the development of new antimalarial drugs with novel modes of actions. The malarial parasite, Plasmodium falciparum, has a relatively small kinome of <100 kinases, with many members exhibiting a high degree of structural divergence from their host counterparts. A number of Plasmodium kinases have recently been shown by reverse genetics to be essential for various parts of the complex parasitic life cycle, and are thus genetically validated as potential targets. Implementation of mass spectrometry-based phosphoproteomics approaches has informed on key phospho-signalling pathways in the parasite. In addition, global phenotypic screens have revealed a large number of putative protein kinase inhibitors with antimalarial potency. Taken together, these investigations point to the Plasmodium kinome as a rich source of potential new targets. In this review, we highlight recent progress made towards this goal.


Subject(s)
Antimalarials/chemistry , Antimalarials/pharmacology , Malaria, Falciparum/drug therapy , Plasmodium falciparum/drug effects , Plasmodium falciparum/enzymology , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/pharmacology , Animals , Antimalarials/therapeutic use , Drug Discovery/methods , Humans , Malaria, Falciparum/parasitology , Models, Molecular , Molecular Targeted Therapy/methods , Protein Kinase Inhibitors/therapeutic use , Protein Kinases/chemistry , Protein Kinases/metabolism , Proteomics/methods
14.
Expert Opin Ther Pat ; 21(2): 147-65, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21204725

ABSTRACT

INTRODUCTION: Macrophages are key drivers of both the innate and adaptive immune systems. The cellular receptor for CSF-1 and IL-34, c-FMS, is a key component of the mechanism(s) by which macrophages are regulated. Several drug discovery programs aimed at uncovering inhibitors of the tyrosine kinase activity of this receptor are now entering clinical phase, and the prospect of readjusting the behavior of macrophages in a number of pathological situations, such as inflammation and cancer, is now on us. AREAS COVERED: In this review, we evaluate the available patent literature on the topic of small molecule inhibitors of c-FMS. By way of background, we review the biology of c-FMS and make an analysis of the therapeutic opportunities that a small molecule c-FMS inhibitor might present. In order to place the pharmacology in perspective, we examine the literature concerning the role of the CSF-1-IL-34-c-FMS axis in macrophage function as well as cell types related to macrophages, such as the osteoclast, the dendritic cell and microglia, and provide a background to the understanding of the therapeutic opportunities for c-FMS inhibitors as well as potential obstacles that could limit their use. EXPERT OPINION: The c-FMS receptor is a hot target for the development of novel regulators of macrophage behavior. Some nice candidates have been developed by a number of groups, and their recent entry into clinical phase testing means that we are now on the cusp of a fuller understanding of the role of these important regulators of the innate and adaptive immune systems in the development of cancer and inflammatory diseases.


Subject(s)
Patents as Topic , Receptor, Macrophage Colony-Stimulating Factor/antagonists & inhibitors , Animals , Autoimmune Diseases/drug therapy , Cell Lineage , Dendritic Cells/physiology , Humans , Interleukins/physiology , Macrophage Colony-Stimulating Factor/physiology , Macrophages/physiology , Receptor, Macrophage Colony-Stimulating Factor/physiology
15.
J Mol Biol ; 387(1): 219-32, 2009 Mar 20.
Article in English | MEDLINE | ID: mdl-19361440

ABSTRACT

The Janus kinases (JAKs) are a pivotal family of protein tyrosine kinases (PTKs) that play prominent roles in numerous cytokine signaling pathways, with aberrant JAK activity associated with a variety of hematopoietic malignancies, cardiovascular diseases and immune-related disorders. Whereas the structures of the JAK2 and JAK3 PTK domains have been determined, the structure of the JAK1 PTK domain is unknown. Here, we report the high-resolution crystal structures of the "active form" of the JAK1 PTK domain in complex with two JAK inhibitors, a tetracyclic pyridone 2-t-butyl-9-fluoro-3,6-dihydro-7H-benz[h]-imidaz[4,5-f]isoquinoline-7-one (CMP6) and (3R,4R)-3-[4-methyl-3-[N-methyl-N-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)amino]piperidin-1-yl]-3-oxopropionitrile (CP-690,550), and compare them with the corresponding JAK2 PTK inhibitor complexes. Both inhibitors bound in a similar manner to JAK1, namely buried deep within a constricted ATP-binding site, thereby providing a basis for the potent inhibition of JAK1. As expected, the mode of inhibitor binding in JAK1 was very similar to that observed in JAK2, highlighting the challenges in developing JAK-specific inhibitors that target the ATP-binding site. Nevertheless, differences surrounding the JAK1 and JAK2 ATP-binding sites were apparent, thereby providing a platform for the rational design of JAK2- and JAK1-specific inhibitors.


Subject(s)
Janus Kinases/metabolism , Protein Kinase Inhibitors/chemistry , Crystallography, X-Ray , Humans , Janus Kinases/antagonists & inhibitors , Janus Kinases/chemistry , Models, Molecular , Protein Conformation , Protein Kinase Inhibitors/pharmacology , Substrate Specificity
16.
Mol Cancer Ther ; 8(11): 3036-45, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19887548

ABSTRACT

CYT997 is a wholly synthetic compound that possesses highly potent cytotoxic activity in vitro through inhibition of microtubule polymerization. CYT997 blocks the cell cycle at the G(2)-M boundary, and Western blot analysis indicates an increase in phosphorylated Bcl-2, along with increased expression of cyclin B1. Caspase-3 activation is also observed in cells treated with CYT997 along with the generation of poly(ADP-ribose) polymerase. The compound possesses favorable pharmacokinetic properties, is orally bioavailable, and is efficacious per os in a range of in vivo cancer models, including some refractory to paclitaxel treatment. CYT997 exhibits vascular disrupting activity as measured in vitro by effects on the permeability of human umbilical vein endothelial cell monolayers, and in vivo by effects on tumor blood flow. CYT997 possesses a useful combination of pharmacologic and pharmacokinetic properties and has considerable potential as a novel anticancer agent.


Subject(s)
Pyridines/pharmacology , Pyrimidines/pharmacology , Tubulin Modulators/pharmacology , Administration, Oral , Animals , Biological Availability , Cell Cycle/drug effects , Cell Growth Processes/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Drug Screening Assays, Antitumor , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Female , Humans , Lung Neoplasms/drug therapy , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Male , Mammary Neoplasms, Experimental/drug therapy , Mammary Neoplasms, Experimental/metabolism , Mammary Neoplasms, Experimental/pathology , Mice , Mice, Inbred BALB C , Mice, Nude , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Pyridines/pharmacokinetics , Pyrimidines/pharmacokinetics , Rats , Rats, Sprague-Dawley , Tubulin Modulators/pharmacokinetics , Xenograft Model Antitumor Assays
17.
Semin Cell Dev Biol ; 19(4): 319-28, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18721891

ABSTRACT

There are four members of the JAK family of protein tyrosine kinases (PTKs) in the human genome. Since their discovery in 1989, great strides have been made in the understanding of their role in normal intracellular signalling. Importantly, their roles in pathologies ranging from cancer to immune deficiencies have placed them front and centre as potential drug targets. The recent discovery of the role of activating mutations in the kinase-like domain (KLD) of JAK2 in the development of polycythemia rubra vera, and the elaboration of KLD mutation as a broader mechanism by which cells might become hyperproliferative has sparked enormous interest in the development of JAK selective drug candidates. I review herein the progress that has been made in the discovery of JAK-targeted inhibitors, and discuss the challenges that face the development of these drugs for use in the clinic.


Subject(s)
Drug Discovery , Isoenzymes/metabolism , Janus Kinases/metabolism , Amino Acid Sequence , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/metabolism , Humans , Isoenzymes/chemistry , Isoenzymes/classification , Isoenzymes/genetics , Janus Kinases/chemistry , Janus Kinases/classification , Janus Kinases/genetics , Models, Molecular , Molecular Sequence Data , Molecular Structure , Protein Conformation , Sequence Alignment , Signal Transduction/physiology
18.
Blood ; 107(1): 176-83, 2006 Jan 01.
Article in English | MEDLINE | ID: mdl-16174768

ABSTRACT

JAK2, a member of the Janus kinase (JAK) family of protein tyrosine kinases (PTKs), is an important intracellular mediator of cytokine signaling. Mutations of the JAK2 gene are associated with hematologic cancers, and aberrant JAK activity is also associated with a number of immune diseases, including rheumatoid arthritis. Accordingly, the development of JAK2-specific inhibitors has tremendous clinical relevance. Critical to the function of JAK2 is its PTK domain. We report the 2.0 A crystal structure of the active conformation of the JAK2 PTK domain in complex with a high-affinity, pan-JAK inhibitor that appears to bind via an induced fit mechanism. This inhibitor, the tetracyclic pyridone 2-tert-butyl-9-fluoro-3,6-dihydro-7H-benz[h]-imidaz[4,5-f]isoquinoline-7-1, was buried deep within a constricted ATP-binding site, in which extensive interactions, including residues that are unique to JAK2 and the JAK family, are made with the inhibitor. We present a structural basis of high-affinity JAK-specific inhibition that will undoubtedly provide an invaluable tool for the further design of novel, potent, and specific therapeutics against the JAK family.


Subject(s)
Enzyme Inhibitors/chemistry , Protein-Tyrosine Kinases/antagonists & inhibitors , Protein-Tyrosine Kinases/chemistry , Proto-Oncogene Proteins/antagonists & inhibitors , Proto-Oncogene Proteins/chemistry , Adenosine Triphosphate , Binding Sites , Crystallography, X-Ray , Humans , Janus Kinase 2 , Molecular Structure , Protein Binding , Protein Conformation
SELECTION OF CITATIONS
SEARCH DETAIL