Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Biochim Biophys Acta ; 1804(3): 454-62, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20152788

ABSTRACT

The ATP-competitive inhibitors dasatinib and nilotinib, which bind to catalytically different conformations of the Abl kinase domain, have recently been approved for the treatment of imatinib-resistant CML. These two new drugs, albeit very efficient against most of the imatinib-resistant mutants of Bcr-Abl, fail to effectively suppress the Bcr-Abl activity of the T315I (or gatekeeper) mutation. Generating new ATP site-binding drugs that target the T315I in Abl has been hampered, amongst others, by target selectivity, which is frequently an issue when developing ATP-competitive inhibitors. Recently, using an unbiased cellular screening approach, GNF-2, a non-ATP-competitive inhibitor, has been identified that demonstrates cellular activity against Bcr-Abl transformed cells. The exquisite selectivity of GNF-2 is due to the finding that it targets the myristate binding site located near the C-terminus of the Abl kinase domain, as demonstrated by genetic approaches, solution NMR and X-ray crystallography. GNF-2, like myristate, is able to induce and/or stabilize the clamped inactive conformation of Abl analogous to the SH2-Y527 interaction of Src. The molecular mechanism for allosteric inhibition by the GNF-2 inhibitor class, and the combined effects with ATP-competitive inhibitors such as nilotinib and imatinib on wild-type Abl and imatinib-resistant mutants, in particular the T315I gatekeeper mutant, are reviewed.


Subject(s)
Adenosine Triphosphate/chemistry , Myristic Acid/chemistry , Protein Kinase Inhibitors/chemistry , Proto-Oncogene Proteins c-abl/antagonists & inhibitors , Proto-Oncogene Proteins c-abl/chemistry , Adenosine Triphosphate/metabolism , Allosteric Regulation/drug effects , Allosteric Regulation/genetics , Benzamides , Crystallography, X-Ray , Drug Resistance, Neoplasm/drug effects , Drug Resistance, Neoplasm/genetics , Humans , Imatinib Mesylate , Mutation, Missense , Myristic Acid/metabolism , Neoplasms/drug therapy , Neoplasms/enzymology , Neoplasms/genetics , Nuclear Magnetic Resonance, Biomolecular , Piperazines/chemistry , Piperazines/therapeutic use , Protein Kinase Inhibitors/therapeutic use , Protein Structure, Tertiary/genetics , Proto-Oncogene Proteins c-abl/genetics , Proto-Oncogene Proteins c-abl/metabolism , Pyrimidines/chemistry , Pyrimidines/therapeutic use
2.
Biochim Biophys Acta ; 1804(3): 445-53, 2010 Mar.
Article in English | MEDLINE | ID: mdl-19922818

ABSTRACT

As a drug used to treat imatinib-resistant and -intolerant, chronic and advanced phase chronic myelogenous leukaemia, nilotinib is well characterised as a potent inhibitor of the Abl tyrosine kinase activity of wild-type and imatinib-resistant mutant forms of BCR-Abl. Here we review the profile of nilotinib as a protein kinase inhibitor. Although an ATP-competitive inhibitor of Abl, nilotinib binds to a catalytically inactive conformation (DFG-out) of the activation loop. As a consequence of this, nilotinib exhibits time-dependent inhibition of Abl kinase in enzymatic assays, which can be extrapolated to other targets to explain differences between biochemical activity and cellular assays. Although these differences confound assessment of kinase selectivity, as assessed using a combination of protein binding and transphosphorylation assays, together with cellular autophosporylation and proliferation assays, well established kinase targets of nilotinib in rank order of inhibitory potency are DDR-1>DDR-2>BCR-Abl (Abl)>PDGFRalpha/beta>KIT>CSF-1R. In addition nilotinib has now been found to bind to both MAPK11 (p38beta) and MAPK12 (p38alpha), as well as with very high affinity to ZAK kinase. Although neither enzymatic nor cellular data are yet available to substantiate the drug as an inhibitor of ZAK phosphorylation, modeling predicts that it binds in an ATP-competitive fashion.


Subject(s)
Adenosine Triphosphate/antagonists & inhibitors , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/enzymology , Protein-Tyrosine Kinases/antagonists & inhibitors , Protein-Tyrosine Kinases/chemistry , Pyrimidines/chemistry , Adenosine Triphosphate/metabolism , Drug Resistance, Neoplasm/drug effects , Humans , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/therapeutic use , Protein-Tyrosine Kinases/metabolism , Pyrimidines/therapeutic use
3.
Protein Expr Purif ; 56(2): 167-76, 2007 Dec.
Article in English | MEDLINE | ID: mdl-17720535

ABSTRACT

As exemplified by three cases, we show that the addition of a small molecular weight inhibitor to the culture of Baculovirus-infected insect cells can dramatically improve the expression of a recombinant kinase. The expression of the tyrosine kinase KDR was sevenfold higher and mainly in a soluble form, when the KDR inhibitor PTK/ZK was added to the culture at the time of Baculovirus infection. The expression of the catalytic domain of the serine/threonine kinase PKCtheta, which is otherwise not possible with the Baculovirus expression system, was expressed mainly soluble at 120mg/L by the addition of the PKC inhibitor BIM XI to the culture of Baculovirus-infected insect cells. For Abl kinase, the expression could also be significantly increased by the addition of the Abl kinase inhibitor STI571 to the culture. For all three kinases, this method had previously been applied by us for the improved production of kinase/inhibitor complex protein, leading to the co-crystal structures. It is shown here at the cases KDR-PTK/ZK and PKCtheta-BIM XI, that the stimulatory effect of an inhibitor on kinase expression is applicable under many culture conditions. The presented method represents a valuable tool to obtain structural knowledge on kinase-inhibitor complexes.


Subject(s)
Baculoviridae/genetics , Protein Kinase Inhibitors/pharmacology , Protein Kinases/chemistry , Protein Kinases/genetics , Animals , Cell Line , Cells, Cultured , Models, Biological , Protein Kinases/metabolism , Proto-Oncogene Proteins c-abl/chemistry , Proto-Oncogene Proteins c-abl/genetics , Proto-Oncogene Proteins c-abl/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Spodoptera/cytology , Spodoptera/virology
4.
Acta Crystallogr D Biol Crystallogr ; 63(Pt 1): 80-93, 2007 Jan.
Article in English | MEDLINE | ID: mdl-17164530

ABSTRACT

Chronic myelogenous leukaemia (CML) results from the Bcr-Abl oncoprotein, which has a constitutively activated Abl tyrosine kinase domain. Although most chronic phase CML patients treated with imatinib as first-line therapy maintain excellent durable responses, patients who have progressed to advanced-stage CML frequently fail to respond or lose their response to therapy owing to the emergence of drug-resistant mutants of the protein. More than 40 such point mutations have been observed in imatinib-resistant patients. The crystal structures of wild-type and mutant Abl kinase in complex with imatinib and other small-molecule Abl inhibitors were determined, with the aim of understanding the molecular basis of resistance and to aid in the design and optimization of inhibitors active against the resistance mutants. These results are presented in a way which illustrates the approaches used to generate multiple structures, the type of information that can be gained and the way that this information is used to support drug discovery.


Subject(s)
Antineoplastic Agents/pharmacology , Chemistry, Pharmaceutical/methods , Drug Design , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Animals , Binding Sites , Crystallization , Crystallography, X-Ray , Drug Industry/methods , Humans , Models, Chemical , Models, Genetic , Models, Molecular , Molecular Conformation , Point Mutation
5.
Structure ; 13(6): 861-71, 2005 Jun.
Article in English | MEDLINE | ID: mdl-15939018

ABSTRACT

The regulation of the activity of Abl and Src family tyrosine kinases is mediated by intramolecular interactions between the SH3, SH2, and kinase (SH1) domains. We have determined the crystal structure of an unphosphorylated form of c-Src in which the SH2 domain is not bound to the C-terminal tail. This results in an open structure where the kinase domain adopts an active conformation and the C terminus binds within a hydrophobic pocket in the C-terminal lobe. NMR binding studies support the hypothesis that an N-terminal myristate could bind in this pocket, as observed for Abl, suggesting that c-Src may also be regulated by myristate binding. In addition, the structure contains a des-methyl analog of the antileukemia drug imatinib (STI571; Gleevec). This structure reveals why the drug shows a low affinity for active kinase conformations, contributing to its excellent kinase selectivity profile.


Subject(s)
Crystallography, X-Ray , Molecular Conformation , src-Family Kinases/chemistry , src-Family Kinases/metabolism , Amino Acid Sequence , Benzamides , Binding Sites , Conserved Sequence , Enzyme Activation , Humans , Hydrogen Bonding , Hydrophobic and Hydrophilic Interactions , Imatinib Mesylate , Inhibitory Concentration 50 , Leucine/metabolism , Models, Chemical , Molecular Sequence Data , Molecular Structure , Myristic Acid/chemistry , Myristic Acid/metabolism , Nuclear Magnetic Resonance, Biomolecular , Phosphorylation , Piperazines/chemistry , Protein Binding , Proto-Oncogene Proteins c-abl/chemistry , Proto-Oncogene Proteins c-abl/metabolism , Pyrimidines/chemistry , Sequence Homology, Amino Acid , Tyrosine/chemistry , src Homology Domains
6.
Cancer Cell ; 5(3): 231-9, 2004 Mar.
Article in English | MEDLINE | ID: mdl-15050915

ABSTRACT

IGF-IR-mediated signaling promotes survival, anchorage-independent growth, and oncogenic transformation, as well as tumor growth and metastasis formation in vivo. NVP-AEW541 is a pyrrolo[2,3-d]pyrimidine derivative small molecular weight kinase inhibitor of the IGF-IR, capable of distinguishing between the IGF-IR (IC50 = 0.086 microM) and the closely related InsR (IC50 = 2.3 microM) in cells. As expected for a specific IGF-IR kinase inhibitor, NVP-AEW541 abrogates IGF-I-mediated survival and colony formation in soft agar at concentrations that are consistent with inhibition of IGF-IR autophosphorylation. In vivo, this orally bioavailable compound inhibits IGF-IR signaling in tumor xenografts and significantly reduces the growth of IGF-IR-driven fibrosarcomas. Thus, NVP-AEW541 represents a class of selective, small molecule IGF-IR kinase inhibitors with proven in vivo antitumor activity and potential therapeutic application.


Subject(s)
Antineoplastic Agents/pharmacology , Receptor, IGF Type 1/metabolism , Signal Transduction/physiology , 3T3 Cells , Animals , Apoptosis/drug effects , Apoptosis/physiology , Cell Division , Enzyme Activation/drug effects , Enzyme Activation/physiology , Enzyme Inhibitors/pharmacology , Humans , MAP Kinase Signaling System/drug effects , MAP Kinase Signaling System/physiology , Mice , Phosphorylation/drug effects , Receptor, IGF Type 1/drug effects , Receptor, Insulin/drug effects , Receptor, Insulin/metabolism , Signal Transduction/drug effects , src-Family Kinases/drug effects , src-Family Kinases/metabolism
7.
Mini Rev Med Chem ; 4(3): 285-99, 2004 Mar.
Article in English | MEDLINE | ID: mdl-15032675

ABSTRACT

Following the paradigm set by STI571, protein tyrosine kinase inhibitors are emerging as a promising class of drugs, capable of modulating intracellular signaling and demonstrating therapeutic potential for the treatment of proliferative diseases. Although the majority of chronic phase CML patients treated with STI571 respond, some patients, especially those with more advanced disease, relapse. This article reviews the reasons for relapse and, in particular, analyses resistance resulting from Bcr-Abl tyrosine kinase domain mutations at the molecular level. Arguments are based upon the structure of the STI571-Abl complex, which is compared to the crystal structures of PD173955-Abl and PD180970-Abl, which bind to the kinase differently. Strategies to potentially circumvent or overcome resistance are discussed.


Subject(s)
Antineoplastic Agents/therapeutic use , Drug Resistance, Neoplasm/genetics , Enzyme Inhibitors/therapeutic use , Leukemia, Myelogenous, Chronic, BCR-ABL Positive , Piperazines/therapeutic use , Protein-Tyrosine Kinases , Pyrimidines/therapeutic use , Animals , Benzamides , Fusion Proteins, bcr-abl , Genes, abl/genetics , Humans , Imatinib Mesylate , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/enzymology , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics , Mutation , Protein-Tyrosine Kinases/antagonists & inhibitors , Protein-Tyrosine Kinases/genetics
8.
J Biomol NMR ; 26(4): 367-72, 2003 Aug.
Article in English | MEDLINE | ID: mdl-12815263

ABSTRACT

Culture conditions for successful amino-acid-type selective isotope labeling of proteins expressed in Baculovirus-infected insect cells are described. The method was applied to the selective labeling of the catalytic domain of c-Abl kinase with (15)N-phenylalanine, (15)N-glycine, (15)N-tyrosine or (15)N-valine. For the essential amino acids phenylalanine, tyrosine and valine high (15)N-label incorporation rates of >/=90% and approximately the expected number of resonances in the HSQC spectra were observed, which was not the case for the non-essential amino acid glycine. The method should be applicable to amino-acid-type selective isotope labeling of other recombinant proteins which have not been amenable to NMR analysis.


Subject(s)
Amino Acids/chemistry , Baculoviridae/genetics , Isotope Labeling/methods , Recombinant Fusion Proteins/chemistry , Amino Acid Sequence , Animals , Catalytic Domain , Cell Culture Techniques/methods , Cells, Cultured/metabolism , Cells, Cultured/virology , Cloning, Molecular , Genes, abl , Humans , Molecular Sequence Data , Nitrogen Isotopes , Proto-Oncogene Proteins c-abl/chemistry , Recombinant Fusion Proteins/biosynthesis , Recombinant Fusion Proteins/isolation & purification , Spodoptera/cytology
9.
Pharmacol Ther ; 93(2-3): 79-98, 2002.
Article in English | MEDLINE | ID: mdl-12191602

ABSTRACT

Many components of mitogenic signaling pathways in normal and neoplastic cells have been identified, including the large family of protein kinases, which function as components of signal transduction pathways, playing a central role in diverse biological processes, such as control of cell growth, metabolism, differentiation, and apoptosis. The development of selective protein kinase inhibitors that can block or modulate diseases caused by abnormalities in these signaling pathways is widely considered a promising approach for drug development. Because of their deregulation in human cancers, protein kinases, such as Bcr-Abl, those in the epidermal growth factor-receptor (HER) family, the cell cycle regulating kinases such as the cyclin-dependent kinases, as well as the vascular endothelial growth factor-receptor kinases involved in the neo-vascularization of tumors, are among the protein kinases considered as prime targets for the development of selective inhibitors. These drug-discovery efforts have generated inhibitors and low-molecular weight therapeutics directed against the ATP-binding site of various protein kinases that are in various stages of development (up to Phase II/III clinical trials). Three examples of inhibitors of protein kinases are reviewed, including low-molecular weight compounds targeting the cell cycle kinases; a potent and selective inhibitor of the HER1/HER2 receptor tyrosine kinase, the pyrollopyrimidine PKI166; and the 2-phenyl-aminopyrimidine STI571 (Glivec(R), Gleevec) a targeted drug therapy directed toward Bcr-Abl, the key player in chronic leukemia (CML). Some members of the HER family of receptor tyrosine kinases, in particular HER1 and HER2, have been found to be overexpressed in a variety of human tumors, suggesting that inhibition of HER signaling would be a viable antiproliferative strategy. The pyrrolo-pyrimidine PKI166 was developed as an HER1/HER2 inhibitor with potent in vitro antiproliferative and in vivo antitumor activity. Based upon its clear association with disease, the Bcr-Abl tyrosine kinase in CML represents the ideal target to validate the clinical utility of protein kinase inhibitors as therapeutic agents. In a preclinical model, STI571 (Glivec(R), Gleevec) showed potent in vitro and in vivo antitumor activity that was selective for Abl, c-Kit, and the platelet-derived growth factor-receptor. Phase I/II studies demonstrated that STI571 is well tolerated, and that it showed promising hematological and cytogenetic responses in CML and clinical responses in the c-Kit-driven gastrointestinal tumors.


Subject(s)
Antineoplastic Agents/pharmacology , Neoplasms/drug therapy , Protein Kinases , Signal Transduction/drug effects , Animals , Antineoplastic Agents/therapeutic use , Benzamides , Cell Cycle/drug effects , Clinical Trials as Topic , Humans , Imatinib Mesylate , Mice , Piperazines , Protein Kinase Inhibitors , Protein Kinases/metabolism , Protein Kinases/physiology , Pyrimidines/therapeutic use
SELECTION OF CITATIONS
SEARCH DETAIL
...