Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
J Med Chem ; 59(16): 7544-60, 2016 08 25.
Article in English | MEDLINE | ID: mdl-27502541

ABSTRACT

Cancer Osaka thyroid (COT) kinase is an important regulator of pro-inflammatory cytokines in macrophages. Thus, pharmacologic inhibition of COT should be a valid approach to therapeutically intervene in the pathogenesis of macrophage-driven inflammatory diseases such as rheumatoid arthritis. We report the discovery and chemical optimization of a novel series of COT kinase inhibitors, with unprecedented nanomolar potency for the inhibition of TNFα. Pharmacological profiling in vivo revealed a high metabolism of these compounds in rats which was demonstrated to be predominantly attributed to aldehyde oxidase. Due to the very low activity of hepatic AO in the dog, the selected candidate 32 displayed significant blood exposure in dogs which resulted in a clear prevention of inflammation-driven lameness. Taken together, the described compounds both potently and selectively inhibit COT kinase in primary human cells and ameliorate inflammatory pathologies in vivo, supporting the notion that COT is an appropriate therapeutic target for inflammatory diseases.


Subject(s)
Drug Discovery , Imidazoles/pharmacology , MAP Kinase Kinase Kinases/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins/antagonists & inhibitors , Quinolines/pharmacology , Animals , Crystallography, X-Ray , Dogs , Dose-Response Relationship, Drug , Humans , Imidazoles/chemical synthesis , Imidazoles/chemistry , MAP Kinase Kinase Kinases/metabolism , Male , Models, Molecular , Molecular Structure , Protein Kinase Inhibitors/chemical synthesis , Protein Kinase Inhibitors/chemistry , Proto-Oncogene Proteins/metabolism , Quinolines/chemical synthesis , Quinolines/chemistry , Rats , Rats, Sprague-Dawley , Structure-Activity Relationship , Tumor Necrosis Factor-alpha/antagonists & inhibitors
2.
J Biol Chem ; 290(24): 15210-8, 2015 Jun 12.
Article in English | MEDLINE | ID: mdl-25918157

ABSTRACT

Macrophages are important cellular effectors in innate immune responses and play a major role in autoimmune diseases such as rheumatoid arthritis. Cancer Osaka thyroid (COT) kinase, also known as mitogen-activated protein kinase kinase kinase 8 (MAP3K8) and tumor progression locus 2 (Tpl-2), is a serine-threonine (ST) kinase and is a key regulator in the production of pro-inflammatory cytokines in macrophages. Due to its pivotal role in immune biology, COT kinase has been identified as an attractive target for pharmaceutical research that is directed at the discovery of orally available, selective, and potent inhibitors for the treatment of autoimmune disorders and cancer. The production of monomeric, recombinant COT kinase has proven to be very difficult, and issues with solubility and stability of the enzyme have hampered the discovery and optimization of potent and selective inhibitors. We developed a protocol for the production of recombinant human COT kinase that yields pure and highly active enzyme in sufficient yields for biochemical and structural studies. The quality of the enzyme allowed us to establish a robust in vitro phosphorylation assay for the efficient biochemical characterization of COT kinase inhibitors and to determine the x-ray co-crystal structures of the COT kinase domain in complex with two ATP-binding site inhibitors. The structures presented in this study reveal two distinct ligand binding modes and a unique kinase domain architecture that has not been observed previously. The structurally versatile active site significantly impacts the design of potent, low molecular weight COT kinase inhibitors.


Subject(s)
MAP Kinase Kinase Kinases/chemistry , Protein Folding , Proto-Oncogene Proteins/chemistry , Crystallography, X-Ray , Humans , Models, Molecular , Protein Conformation , Recombinant Proteins/chemistry
3.
J Am Chem Soc ; 132(20): 7043-8, 2010 May 26.
Article in English | MEDLINE | ID: mdl-20450175

ABSTRACT

Allosteric inhibitors of Bcr-Abl have emerged as a novel therapeutic option for the treatment of CML. Using fragment-based screening, a search for novel Abl inhibitors that bind to the myristate pocket was carried out. Here we show that not all myristate ligands are functional inhibitors, but that the conformational state of C-terminal helix_I is a structural determinant for functional activity. We present an NMR-based conformational assay to monitor the conformation of this crucial helix_I and show that myristate ligands that bend helix_I are functional antagonists, whereas ligands that bind to the myristate pocket but do not induce this conformational change are kinase agonists. Activation of c-Abl by allosteric agonists has been confirmed in a biochemical assay.


Subject(s)
Drug Evaluation, Preclinical/methods , Protein Kinase Inhibitors/metabolism , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-abl/agonists , Proto-Oncogene Proteins c-abl/antagonists & inhibitors , Allosteric Regulation , Animals , Enzyme Activation/drug effects , Humans , Ligands , Magnetic Resonance Spectroscopy , Mice , Models, Molecular , Myristic Acid/metabolism , Protein Binding , Protein Kinase Inhibitors/chemistry , Protein Structure, Secondary , Proto-Oncogene Proteins c-abl/chemistry , Proto-Oncogene Proteins c-abl/metabolism
4.
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
5.
Nature ; 463(7280): 501-6, 2010 Jan 28.
Article in English | MEDLINE | ID: mdl-20072125

ABSTRACT

In an effort to find new pharmacological modalities to overcome resistance to ATP-binding-site inhibitors of Bcr-Abl, we recently reported the discovery of GNF-2, a selective allosteric Bcr-Abl inhibitor. Here, using solution NMR, X-ray crystallography, mutagenesis and hydrogen exchange mass spectrometry, we show that GNF-2 binds to the myristate-binding site of Abl, leading to changes in the structural dynamics of the ATP-binding site. GNF-5, an analogue of GNF-2 with improved pharmacokinetic properties, when used in combination with the ATP-competitive inhibitors imatinib or nilotinib, suppressed the emergence of resistance mutations in vitro, displayed additive inhibitory activity in biochemical and cellular assays against T315I mutant human Bcr-Abl and displayed in vivo efficacy against this recalcitrant mutant in a murine bone-marrow transplantation model. These results show that therapeutically relevant inhibition of Bcr-Abl activity can be achieved with inhibitors that bind to the myristate-binding site and that combining allosteric and ATP-competitive inhibitors can overcome resistance to either agent alone.


Subject(s)
Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Drug Resistance, Neoplasm/drug effects , Fusion Proteins, bcr-abl/chemistry , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism , Animals , Antineoplastic Agents/metabolism , Antineoplastic Combined Chemotherapy Protocols , Benzamides , Binding Sites , Bone Marrow Transplantation , Cell Line, Tumor , Crystallization , Disease Models, Animal , Female , Fusion Proteins, bcr-abl/genetics , Fusion Proteins, bcr-abl/metabolism , Humans , Imatinib Mesylate , Inhibitory Concentration 50 , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/enzymology , Male , Mass Spectrometry , Mice , Models, Molecular , Mutation/genetics , Piperazines/chemistry , Piperazines/pharmacology , Protein Structure, Tertiary , Pyrimidines/chemistry , Pyrimidines/metabolism , Pyrimidines/pharmacology , Transplantation, Heterologous
6.
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
7.
Protein Expr Purif ; 64(2): 185-93, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19038347

ABSTRACT

The recombinant expression of eukaryotic proteins in Escherichia coli often results in protein aggregation. Several articles report on improved solubility and increased purification yields of individual proteins upon over-expression of E. coli chaperones but this effect might potentially be protein-specific. To find out whether chaperone over-expression is a generally applicable strategy for the production of human protein kinases in E. coli, we analyzed 10 kinases, mainly as catalytic domain constructs. The kinases studied, namely c-Src, c-Abl, Hck, Lck, Igf1R, InsR, KDR, c-Met, b-Raf and Irak4, belong to the tyrosine and tyrosine kinase-like groups of kinases. Upon over-expression of the E. coli chaperones DnaK/DnaJ/GrpE and GroEL/GroES, the yields of 7 from 10 polyhistidine-tagged kinases were increased up to 5-fold after nickel-affinity purification (IMAC). Additive over-expression of the chaperones ClpB and/or trigger factor showed no further improvement. Co-purification of DnaJ and GroEL indicated incomplete kinase folding, therefore, the oligomerization state of the kinases was determined by size-exclusion chromatography. In our study, kinases behave in three different ways. Kinases where yields are not affected by E. coli chaperone over-expression e.g. c-Src elute in the monomeric fraction (category I). Although IMAC yields increase upon chaperone over-expression, InsR and b-Raf kinase are present as soluble aggregates (category II). Igf1R and c-Met kinase catalytic domains are partially complexed with E. coli chaperones upon over-expression; however, they show approximately 2-fold increased yields of monomer (category III). Together, our results suggest that the benefits of chaperone over-expression on the production of protein kinases in E. coli are indeed case-specific.


Subject(s)
Escherichia coli/metabolism , Molecular Chaperones/biosynthesis , Phosphotransferases/biosynthesis , Recombinant Proteins/biosynthesis , Chromatography, Gel , Escherichia coli/genetics , Mass Spectrometry , Molecular Chaperones/genetics , Molecular Chaperones/isolation & purification , Phosphotransferases/genetics , Phosphotransferases/isolation & purification , Protein Engineering , Protein Folding , Protein Structure, Tertiary , Recombinant Proteins/genetics , Recombinant Proteins/isolation & purification , Solubility
8.
J Biol Chem ; 283(26): 18292-302, 2008 Jun 27.
Article in English | MEDLINE | ID: mdl-18434310

ABSTRACT

Current structural understanding of kinases is largely based on x-ray crystallographic studies, whereas very little data exist on the conformations and dynamics that kinases adopt in the solution state. ABL kinase is an important drug target in the treatment of chronic myelogenous leukemia. Here, we present the first characterization of ABL kinase in complex with three clinical inhibitors (imatinib, nilotinib, and dasatinib) by modern solution NMR techniques. Structural and dynamical results were derived from complete backbone resonance assignments, experimental residual dipolar couplings, and (15)N relaxation data. Residual dipolar coupling data on the imatinib and nilotinib complexes show that the activation loop adopts the inactive conformation, whereas the dasatinib complex preserves the active conformation, which does not support contrary predictions based upon molecular modeling. Nanosecond as well as microsecond dynamics can be detected for certain residues in the activation loop in the inactive and active conformation complexes.


Subject(s)
Magnetic Resonance Spectroscopy/methods , Proto-Oncogene Proteins c-abl/chemistry , Amino Acid Sequence , Antineoplastic Agents/pharmacology , Benzamides , Chemistry, Pharmaceutical/methods , Dasatinib , Drug Design , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Humans , Imatinib Mesylate , Models, Molecular , Molecular Conformation , Molecular Sequence Data , Piperazines/pharmacology , Protein Conformation , Pyrimidines/pharmacology , Thiazoles/pharmacology
9.
Biomol NMR Assign ; 2(1): 41-2, 2008 Jun.
Article in English | MEDLINE | ID: mdl-19636920

ABSTRACT

Imatinib (Glivec or Gleevec) potently inhibits the tyrosine kinase activity of BCR-ABL, a constitutively activated kinase, which causes chronic myelogenous leukemia (CML). Here we report the first almost complete backbone assignment of c-ABL kinase domain in complex with imatinib.


Subject(s)
Fusion Proteins, bcr-abl/antagonists & inhibitors , Magnetic Resonance Spectroscopy/methods , Piperazines/chemistry , Pyrimidines/chemistry , Amino Acid Sequence , Benzamides , Binding Sites , Carbon Isotopes/chemistry , Imatinib Mesylate , Molecular Sequence Data , Molecular Weight , Nitrogen Isotopes/chemistry , Protein Binding , Protein Kinase Inhibitors/chemistry , Protons
10.
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
11.
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
12.
J Biomol NMR ; 31(4): 343-9, 2005 Apr.
Article in English | MEDLINE | ID: mdl-15929001

ABSTRACT

This report shows for the first time the efficient uniform isotope labeling of a recombinant protein expressed using Baculovirus-infected insect cells. The recent availability of suitable media for (15)N- and (13)C/(15)N-labeling in insect cells, the high expression of Abl kinase in these labeling media and a suitable labeling protocol made it possible to obtain a (1)H-(15)N-HSQC spectrum for the catalytic domain of Abl kinase of good quality and with label incorporation rates > 90%. The presented isotope labeling method should be applicable also to further proteins where successful expression is restricted to the Baculovirus expression system.


Subject(s)
Baculoviridae/metabolism , Nuclear Magnetic Resonance, Biomolecular/methods , Proto-Oncogene Proteins c-abl/chemistry , Animals , CHO Cells , Carbon Isotopes , Catalytic Domain , Cell Line , Cell Survival , Cricetinae , Electrophoresis, Polyacrylamide Gel , Humans , Insecta , Magnetic Resonance Spectroscopy , Nitrogen Isotopes , Recombinant Proteins/chemistry , Spectrometry, Mass, Electrospray Ionization
13.
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
14.
Cancer Cell ; 7(2): 129-41, 2005 Feb.
Article in English | MEDLINE | ID: mdl-15710326

ABSTRACT

The Bcr-Abl tyrosine kinase oncogene causes chronic myelogenous leukemia (CML) and Philadelphia chromosome-positive (Ph+) acute lymphoblastic leukemia (ALL). We describe a novel selective inhibitor of Bcr-Abl, AMN107 (IC50 <30 nM), which is significantly more potent than imatinib, and active against a number of imatinib-resistant Bcr-Abl mutants. Crystallographic analysis of Abl-AMN107 complexes provides a structural explanation for the differential activity of AMN107 and imatinib against imatinib-resistant Bcr-Abl. Consistent with its in vitro and pharmacokinetic profile, AMN107 prolonged survival of mice injected with Bcr-Abl-transformed hematopoietic cell lines or primary marrow cells, and prolonged survival in imatinib-resistant CML mouse models. AMN107 is a promising new inhibitor for the therapy of CML and Ph+ ALL.


Subject(s)
Antineoplastic Agents/pharmacology , Fusion Proteins, bcr-abl/antagonists & inhibitors , Fusion Proteins, bcr-abl/genetics , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Pyrimidines/chemistry , Pyrimidines/pharmacology , Animals , Benzamides , Bone Marrow Cells/cytology , Cell Line , Cell Line, Tumor , Cell Survival , Crystallography, X-Ray , Dose-Response Relationship, Drug , Drug Resistance, Neoplasm , Hematopoietic Stem Cells/cytology , Imatinib Mesylate , Inhibitory Concentration 50 , Mice , Models, Biological , Models, Chemical , Mutation , Mycoplasma/metabolism , Phosphorylation , Piperazines/pharmacology , Retroviridae/genetics , Time Factors
15.
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
16.
J Biomol NMR ; 30(2): 205-10, 2004 Oct.
Article in English | MEDLINE | ID: mdl-15702527

ABSTRACT

Selective isotopic labeling of larger proteins greatly simplifies protein NMR spectra and reduces signal overlap, but selectively labeled proteins cannot be easily assigned since the sequential assignment method is not applicable. Here we describe a strategy for resonance assignment in selectively labeled proteins. Our approach involves a spin-labeled analog of a ligand of which the three-dimensional structure in complex with the target protein is known. Other methods for introduction of the spin label are possible. The paramagnetic center causes faster relaxation of all neighboring nuclei in a distance-dependent manner. Measurement of this effect allows to deduce distances between isotopically labeled residues and the paramagnetic center which can be used for resonance assignment. The method is demonstrated for the catalytic domain of Abl kinase in complex with the inhibitor, STI571.


Subject(s)
Magnetics , Nuclear Magnetic Resonance, Biomolecular/methods , Proto-Oncogene Proteins c-abl/chemistry , Proto-Oncogene Proteins c-abl/metabolism , Pyrimidines/chemistry , Pyrimidines/metabolism , Benzamides , Catalytic Domain , Crystallography, X-Ray , Imatinib Mesylate , Ligands , Models, Molecular , Molecular Structure , Piperazines , Protein Structure, Tertiary , Spin Labels
17.
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
18.
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
...