Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 37
Filter
1.
N Engl J Med ; 344(14): 1031-7, 2001 Apr 05.
Article in English | MEDLINE | ID: mdl-11287972

ABSTRACT

BACKGROUND: BCR-ABL is a constitutively activated tyrosine kinase that causes chronic myeloid leukemia (CML). Since tyrosine kinase activity is essential to the transforming function of BCR-ABL, an inhibitor of the kinase could be an effective treatment for CML. METHODS: We conducted a phase 1, dose-escalating trial of STI571 (formerly known as CGP 57148B), a specific inhibitor of the BCR-ABL tyrosine kinase. STI571 was administered orally to 83 patients with CML in the chronic phase in whom treatment with interferon alfa had failed. Patients were successively assigned to 1 of 14 doses ranging from 25 to 1000 mg per day. RESULTS: Adverse effects of STI571 were minimal; the most common were nausea, myalgias, edema, and diarrhea. A maximal tolerated dose was not identified. Complete hematologic responses were observed in 53 of 54 patients treated with daily doses of 300 mg or more and typically occurred in the first four weeks of therapy. Of the 54 patients treated with doses of 300 mg or more, cytogenetic responses occurred in 29, including 17 (31 percent of the 54 patients who received this dose) with major responses (0 to 35 percent of cells in metaphase positive for the Philadelphia chromosome); 7 of these patients had complete cytogenetic remissions. CONCLUSIONS: STI571 is well tolerated and has significant antileukemic activity in patients with CML in whom treatment with interferon alfa had failed. Our results provide evidence of the essential role of BCR-ABL tyrosine kinase activity in CML and demonstrate the potential for the development of anticancer drugs based on the specific molecular abnormality present in a human cancer.


Subject(s)
Enzyme Inhibitors/administration & dosage , Fusion Proteins, bcr-abl/antagonists & inhibitors , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Piperazines/administration & dosage , Protein-Tyrosine Kinases/antagonists & inhibitors , Pyrimidines/administration & dosage , Adult , Aged , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/adverse effects , Antineoplastic Agents/pharmacokinetics , Benzamides , Blood Cell Count , Dose-Response Relationship, Drug , Enzyme Inhibitors/adverse effects , Enzyme Inhibitors/pharmacokinetics , Female , Fusion Proteins, bcr-abl/metabolism , Humans , Imatinib Mesylate , Interferon-alpha/therapeutic use , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/blood , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/enzymology , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , Male , Middle Aged , Phosphorylation , Piperazines/adverse effects , Piperazines/pharmacokinetics , Protein-Tyrosine Kinases/metabolism , Pyrimidines/adverse effects , Pyrimidines/pharmacokinetics , Recurrence , Remission Induction/methods
2.
J Pharmacol Exp Ther ; 295(1): 139-45, 2000 Oct.
Article in English | MEDLINE | ID: mdl-10991971

ABSTRACT

STI571 (formerly known as CGP 57148B) is a protein-tyrosine kinase inhibitor that is currently in clinical trials for the treatment of chronic myelogenous leukemia. STI571 selectively inhibits the Abl and platelet-derived growth factor (PDGF) receptor tyrosine kinases in vitro and blocks cellular proliferation and tumor growth of Bcr-abl- or v-abl-expressing cells. We have further investigated the profile of STI571 against related receptor tyrosine kinases. STI571 was found to potently inhibit the kinase activity of the alpha- and beta-PDGF receptors and the receptor for stem cell factor, but not the closely related c-Fms, Flt-3, Kdr, Flt-1, and Tek tyrosine kinases. Additionally, no inhibition of c-Met or nonreceptor tyrosine kinases such as Src and Jak-2 has been observed. In cell-based assays, STI571 selectively inhibited PDGF and stem cell factor-mediated cellular signaling, including ligand-stimulated receptor autophosphorylation, inositol phosphate formation, and mitogen-activated protein kinase activation and proliferation. These results expand the profile of STI571 and suggest that in addition to chronic myelogenous leukemia, STI571 may have clinical potential in the treatment of diseases that involve abnormal activation of c-Kit or PDGF receptor tyrosine kinases.


Subject(s)
Antineoplastic Agents/pharmacology , Enzyme Inhibitors/pharmacology , Piperazines/pharmacology , Proto-Oncogene Proteins c-abl/antagonists & inhibitors , Proto-Oncogene Proteins c-kit/drug effects , Pyrimidines/pharmacology , Receptors, Platelet-Derived Growth Factor/antagonists & inhibitors , Signal Transduction/drug effects , Animals , Benzamides , Cell Line , Imatinib Mesylate , Mice , Mitogen-Activated Protein Kinases/physiology , Proto-Oncogene Proteins c-kit/physiology , Receptors, Platelet-Derived Growth Factor/physiology , Stem Cell Factor/physiology
4.
Oncogene ; 18(33): 4647-53, 1999 Aug 19.
Article in English | MEDLINE | ID: mdl-10467411

ABSTRACT

Sam68 is a 68 kDa protein that associates with and is phosphorylated by the c-Src kinase at mitosis. It contains a KH domain implicated in RNA binding and several proline-rich motifs that resemble known SH3 binding sites. The SH3 domains of c-Src, phosphatidylinositol 3-OH kinase, phospholipase C-gamma and Grb2 protein (containing two SH3 domains), but not other SH3 domains tested, were capable of binding Sam68 in vitro. Synthetic peptides corresponding to the proline motifs of Sam68 inhibited with different efficiencies the binding of SH3 domains to Sam68 suggesting that the proline motifs of Sam68 function as specific SH3 domain binding sites. Mutation of Sam68 SH3 binding sites further indicated that the SRC SH3 domain mediates binding of Src to unphosphorylated Sam68. Phosphorylation of Sam68 by Src kinase was inhibited when the Src SH3 binding site of Sam68 was mutated or when corresponding peptides were added to in vitro kinase reactions indicating that binding of the Src SH3 domain to a specific site near the amino-terminus of Sam68 (including residues 38 - 45: PPLPHRSR) facilitates phosphorylation of Sam68 by the Src kinase domain. Sam68-based proline peptides had no effect on the phosphorylation of another in vitro substrate of Src, enolase. These results suggest that Src effectively mounts Sam68 through its SH3 domain, possibly as a mechanism to position the kinase domain close to substrate tyrosine residues in the carboxyl-half of the protein.


Subject(s)
Proto-Oncogene Proteins pp60(c-src)/metabolism , RNA-Binding Proteins/metabolism , src Homology Domains , Adaptor Proteins, Signal Transducing , Animals , Binding Sites , Cells, Cultured , DNA-Binding Proteins , Humans , Phosphorylation , Proline , Protein Binding , Rats
5.
Curr Med Chem ; 6(9): 775-805, 1999 Sep.
Article in English | MEDLINE | ID: mdl-10495352

ABSTRACT

The protein kinase family represents both a huge opportunity and a challenge for drug development. The conservation of structural features within the ATP binding cleft initially led to the belief that specificity would be difficult to achieve. This dogma has now been clearly dispelled with the discovery and clinical testing of a group of first generation compounds, which are characterized by a high degree of selectivity towards a variety of oncology targets. The structural basis for selectivity and potency has now been clarified with the crystallization of a number of such targets in complex with inhibitors. The protein kinase inhibitor field is now ripe for the structure based exploitation of additional highly validated targets from a variety of therapeutic areas.


Subject(s)
Adenosine Triphosphate/metabolism , Drug Design , Enzyme Inhibitors/pharmacology , Protein-Tyrosine Kinases/antagonists & inhibitors , Adenylyl Imidodiphosphate/chemistry , Anthraquinones/chemistry , Binding Sites/drug effects , Catalytic Domain/drug effects , Cyclin-Dependent Kinases/chemistry , Enzyme Inhibitors/chemistry , Humans , Kinetin , Piperazines/chemistry , Protein-Tyrosine Kinases/drug effects , Purines/chemistry , Pyridines/chemistry , Pyrimidines/chemistry , Pyrroles/chemistry , Quinazolines/chemistry , Receptors, Growth Factor/antagonists & inhibitors , Receptors, Growth Factor/drug effects , Roscovitine , Staurosporine/chemistry , Structure-Activity Relationship
6.
Curr Opin Drug Discov Devel ; 2(4): 274-85, 1999 Jul.
Article in English | MEDLINE | ID: mdl-19649955

ABSTRACT

Early efforts to discover and develop protein kinase inhibitors have focused largely on a small group of oncology targets such as the EGFR and PKC enzymes. More recently, hundreds of protein kinases have been identified at the genetic level, many of which are now being assigned functions in a variety of signaling pathways. Additionally, mutagenesis and X-ray crystallographic studies have further defined common structural features associated with binding of the ATP cofactor within a conserved ATP binding cleft. These studies have also demonstrated significant differences in the ATP binding cleft between individual kinases, providing a molecular basis for understanding and exploiting inhibitor specificity. The current review will focus on recent developments in the field of ATP site-directed inhibitors with particular emphasis on the major scaffolds being derivatized to take advantage of variable regions of the active site.

7.
Proc Natl Acad Sci U S A ; 95(20): 12069, 1998 Sep 29.
Article in English | MEDLINE | ID: mdl-9786782
8.
Clin Cancer Res ; 4(7): 1661-72, 1998 Jul.
Article in English | MEDLINE | ID: mdl-9676840

ABSTRACT

The excessive proliferation of the myeloid marrow compartment in Philadelphia chromosome (Ph)-positive acute and chronic leukemias has been largely attributed to a hyperactive and autonomously acting hybrid tyrosine kinase BCR-ABL, a product of the fusion between the second exon of the c-ABL proto-oncogene and 5' portions of the BCR gene on chromosome 22. This specific molecular event, amenable to attack with specifically designed inhibitors, has recently been successfully influenced by the drug CGP-57148 in mammalian cells transfected with full-length BCR-ABL gene and expressing full-length p210Bcr-Abl protein, as well as in primary human leukemic cells expressing p210Bcr-Abl fusion protein. In view of the heterogeneity of BCR-ABL transcripts associated with various phenotypes, we investigated the effect of CGP-57148 on p190Bcr-Abl- and p210Bcr-Abl-expressing, patient-derived cell lines and primary intact blast cells. In particular, we were interested in whether the variations in molecular events and/or the phenotype of Ph-positive cells would affect their susceptibility to the specific tyrosine kinase inhibitor CGP-57148. We have demonstrated that the sensitivity of human cells with lymphoblastic immunophenotype expressing p190Bcr-Abl protein is comparable to that for leukemic myeloid cells expressing p210Bcr-Abl protein. After documenting profound and phenotype-independent suppression of both autophosphorylation and cell growth, we explored the importance of time and dose of exposure on the manifestation and stability of the induced events. Although there were variations between target cells, in vitro exposure for 24-48 h induced extensive and apparently irreversible apoptosis in BCR-ABL-expressing but not other normal or BCR-ABL-negative leukemic cells. These findings support the potential use of CGP-57148 to purge Ph-positive cells from autologous bone marrow in vitro. Another important finding was the comparable suppressive effect of temporary CGP-57148 exposure on both clonogenic KBM-5 cells and the whole cell population. Exposure time and dose appeared to be important variables among various cell types. Moreover, effective doses appeared uniformly harmless to cells lacking BCR-ABL protein functioning as tyrosine kinase. Thus, the continuous exposure of target cells, at least during the initial period of 24-48 h, may prove to be an important variable in the design of in vitro and in vivo therapy using tyrosine kinase inhibitors.


Subject(s)
Enzyme Inhibitors/therapeutic use , Fusion Proteins, bcr-abl/drug effects , Philadelphia Chromosome , Piperazines/therapeutic use , Precursor Cell Lymphoblastic Leukemia-Lymphoma/therapy , Protein-Tyrosine Kinases/antagonists & inhibitors , Pyrimidines/therapeutic use , Apoptosis , Benzamides , Cell Division/drug effects , Cell Survival/drug effects , Drug Screening Assays, Antitumor , Fusion Proteins, bcr-abl/metabolism , Humans , Imatinib Mesylate , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , Phosphorylation , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , Precursor Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Precursor Cell Lymphoblastic Leukemia-Lymphoma/pathology , Proto-Oncogene Mas , Tumor Cells, Cultured/drug effects
9.
DNA Cell Biol ; 17(3): 265-74, 1998 Mar.
Article in English | MEDLINE | ID: mdl-9539106

ABSTRACT

The interruption of signaling cascades in intact cells through the introduction of nonpermeant compounds inferred by in vitro studies to specifically inhibit epidermal growth factor (EGF) receptor (EGF-R) function is described. Two nonpermeant [(alkylamino)methyl]acrylophenone derivatives, [(dimethylamino)methyl] acrylo-para-[(benzoylsulfonyl)-oxy]phenone and [(dimethylamino)-methyl]acrylo-para-[(hydroxy-benzoylsulfonyl++ +)-oxy]phenone, were introduced by in situ electroporation into mouse or rat fibroblasts growing on indium-tin oxide-coated glass. Cells were subsequently stimulated with growth factors and assessed for activation of a downstream target, the extracellular signal-regulated kinase (ERK1/2), by probing with specific antibodies. Electrodes and slides were configured to provide non-electroporated control cells side by side with the electroporated ones, both growing on the same type of indium-tin oxide-coated glass surface. Using this set-up, these compounds could inhibit EGF- but not platelet-derived growth factor (PDGF)-mediated ERK1/2 activation in vivo. These results demonstrate the potential of the in situ electroporation approach for the study of tyrosine kinase action using selective but nonpermeant inhibitors that would otherwise be ineffective in intact cells.


Subject(s)
Benzenesulfonates/pharmacology , Benzoates/pharmacology , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Electroporation/methods , Enzyme Inhibitors/pharmacology , Mitogen-Activated Protein Kinases , 3T3 Cells/drug effects , Animals , Antibodies , Calcium-Calmodulin-Dependent Protein Kinases/antagonists & inhibitors , Calcium-Calmodulin-Dependent Protein Kinases/immunology , Cell Division/drug effects , Cells, Cultured , Culture Media, Serum-Free , Enzyme Activation/drug effects , Epidermal Growth Factor/pharmacology , ErbB Receptors/metabolism , Fibroblasts/drug effects , Mice , Mitogen-Activated Protein Kinase 1 , Mitogen-Activated Protein Kinase 3 , Platelet-Derived Growth Factor/pharmacology , Rats , Receptors, Platelet-Derived Growth Factor/metabolism
10.
Int J Cancer ; 76(1): 154-63, 1998 Mar 30.
Article in English | MEDLINE | ID: mdl-9533776

ABSTRACT

A calculated 3-D model of the kinase domain of the epidermal growth factor receptor (EGF-R) protein-tyrosine kinase (PTK) was used to develop a pharmacophore model for ATP-competitive inhibitors and, subsequently, a new class of selective EGF-R kinase inhibitors. CGP 59326A, a highly selective and potent inhibitor of the EGF-R in vitro, inhibited the proliferation of EGF-R-expressing epithelial lines, while having little anti-proliferative activity against EGF-R-negative lines. In contrast to previously described inhibitors, CGP 59326A had potent and selective in vivo anti-tumor activity at well-tolerated doses against EGF-R-expressing tumors (e.g., ED50 of 0.78 to 1.5 mg/kg for inhibition of A431 tumor growth). CGP 59326A inhibited growth of human tumor xenografts expressing the EGF-R but showed little activity against EGF-R-negative xenografts. Combination of CGP 59326A with cytotoxic agents resulted in tumor regression and cures. The high selectivity and attractive biological profile of CGP 59326A suggest that it could have therapeutic value in the treatment of proliferative diseases which involve mitogenic signaling from the EGF-R.


Subject(s)
Neoplasms/metabolism , Protein-Tyrosine Kinases/antagonists & inhibitors , Pyrimidines/pharmacology , Pyrroles/pharmacology , 3T3 Cells , Animals , Cell Division/drug effects , Enzyme Inhibitors , Epithelial Cells , ErbB Receptors/metabolism , Female , Mice , Mice, Inbred BALB C , Neoplasm Transplantation , Neoplasms/pathology , Phosphorylation , Pyrimidines/chemistry , Pyrroles/chemistry , Time Factors
11.
Blood ; 90(12): 4947-52, 1997 Dec 15.
Article in English | MEDLINE | ID: mdl-9389713

ABSTRACT

CGP 57148 is a compound of the 2-phenylaminopyrimidine class that selectively inhibits the tyrosine kinase activity of the ABL and the platelet-derived growth factor receptor (PDGFR) protein tyrosine kinases. We previously showed that CGP 57148 selectively kills p210BCR-ABL-expressing cells. To extend these observations, we evaluated the ability of CGP 57148 to inhibit other activated ABL tyrosine kinases, including p185BCR-ABL and TEL-ABL. In cell-based assays of ABL tyrosine phosphorylation, inhibition of ABL kinase activity was observed at concentrations similar to that reported for p210BCR-ABL. Consistent with the in vitro profile of this compound, the growth of cells expressing activated ABL protein tyrosine kinases was inhibited in the absence of exogenous growth factor. Growth inhibition was also observed with a p185BCR-ABL-positive acute lymphocytic leukemia (ALL) cell line generated from a Philadelphia chromosome-positive ALL patient. As CGP 57148 inhibits the PDGFR kinase, we also showed that cells expressing an activated PDGFR tyrosine kinase, TEL-PDGFR, are sensitive to this compound. Thus, this compound may be useful for the treatment of a variety of BCR-ABL-positive leukemias and for treatment of the subset of chronic myelomonocytic leukemia patients with a TEL-PDGFR fusion protein.


Subject(s)
DNA-Binding Proteins/analysis , Enzyme Inhibitors/pharmacology , Fusion Proteins, bcr-abl/analysis , Oncogene Proteins v-abl/analysis , Piperazines/pharmacology , Protein-Tyrosine Kinases/antagonists & inhibitors , Pyrimidines/pharmacology , Receptors, Platelet-Derived Growth Factor/analysis , Repressor Proteins , Transcription Factors/analysis , Animals , Antineoplastic Agents/pharmacology , Benzamides , Cell Division/drug effects , Cell Line , Imatinib Mesylate , Mice , Proto-Oncogene Proteins c-ets , ETS Translocation Variant 6 Protein
12.
Structure ; 5(12): 1551-6, 1997 Dec 15.
Article in English | MEDLINE | ID: mdl-9438871

ABSTRACT

Protein kinases are involved in a wide variety of signal transduction processes; their deregulation has been implicated in a number of human diseases, including cancer and disorders of the immune system. The recently determined structures of two protein kinases, cAPK and CDK2, in complex with an inhibitor, staurosporine, provide a detailed account of inhibitor-kinase interactions and inhibitor selectivity.


Subject(s)
CDC2-CDC28 Kinases , Cyclic AMP-Dependent Protein Kinases/antagonists & inhibitors , Cyclic AMP-Dependent Protein Kinases/chemistry , Cyclin-Dependent Kinases/antagonists & inhibitors , Cyclin-Dependent Kinases/chemistry , Drug Design , Enzyme Inhibitors/chemical synthesis , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/chemistry , Staurosporine/chemistry , Binding Sites , Cyclic AMP-Dependent Protein Kinases/metabolism , Cyclin-Dependent Kinase 2 , Cyclin-Dependent Kinases/metabolism , Models, Molecular , Protein Serine-Threonine Kinases/metabolism , Staurosporine/metabolism , Structure-Activity Relationship
13.
Blood Cells Mol Dis ; 23(3): 380-94, 1997 Dec.
Article in English | MEDLINE | ID: mdl-9446752

ABSTRACT

The BCR/ABL fusion protein transforms myeloid stem cells. Both chronic myelogenous leukemias (CML) and a subset of acute lymphoblastic leukemias (ALL) are associated with the expression of BCR/ABL proteins. This knowledge has not yet been translated into any specific tool to control ABL driven neoplastic cells growth. CGP57148B is an ATP-competitive inhibitor of the ABL protein kinase; it has been shown to inhibit the kinase activity of ABL both in vitro and in vivo and to inhibit the growth of v-abl and bcr/abl transfectants, as well as the in vitro formation of bone marrow (BM)-derived colonies in the presence of growth factors in some CML patients. These studies were performed to investigate the activity of CGP57148B on the spontaneous proliferation of both fresh and cultured, leukemic and normal, BCR/ABL positive and negative cells, and to study its mechanism of action. Six cell lines derived from BCR/ABL+ leukemias (K562, BV173, KCL22, KU812, MC3, LAMA84), thirteen BCR/ABL negative lines, both neoplastic (KG1, SU-DHL-1, U937, Daudi, NB4, NB4.306) and derived from normal cells (PHA blasts, LAK, fibroblasts, LCL, renal epithelial cells, endothelial cells, CD34(+) cells), and 14 fresh leukemic samples were tested using a tritiated thymidine uptake assay. The in vivo phosphorylation of the BCR/ABL protein was evaluated by western blot, while apoptosis was detected by the annexin V/propidium binding test. The induction of differentiation was assayed by immunofluorescence using multiple antibodies. All six BCR/ABL+ lines showed a dose dependent inhibition of their spontaneous proliferative rate, which was not accompanied by differentiation. The treatment caused, within minutes, dephosphorylation of the BCR/ABL protein, followed in 16-24 hours by a decrease in cycling cells and induction of apoptosis. No significant inhibition of DNA synthesis was observed in any BCR/ABL negative normal or neoplastic line at concentrations

Subject(s)
Apoptosis , Fusion Proteins, bcr-abl/antagonists & inhibitors , Leukemia, Myeloid/enzymology , Leukemia, Myeloid/pathology , Protein-Tyrosine Kinases/antagonists & inhibitors , Apoptosis/drug effects , Benzamides , Cell Differentiation/drug effects , Cell Division/drug effects , Cell Line , Fusion Proteins, bcr-abl/classification , Fusion Proteins, bcr-abl/metabolism , Humans , Imatinib Mesylate , Phosphorylation/drug effects , Piperazines/pharmacology , Pyrimidines/pharmacology , Tumor Cells, Cultured
14.
Arch Pharm (Weinheim) ; 329(7): 371-6, 1996 Jul.
Article in English | MEDLINE | ID: mdl-8764886

ABSTRACT

Phenylamino-pyrimidines represent a novel class of inhibitors of the protein kinase C with a high degree of selectivity versus other serine/threonine and tyrosine kinases. Steady state kinetic analysis of N-(3-[1-imidazolyl]-phenyl-4-(3-pyridyl)-2-pyrimidinamine (5), which showed potent inhibitory activity, revealed competitive kinetics relative to ATP. The adjacent H-bond acceptor of the pyrimidine moiety next to an H-bond donor of the phenylamine was found to be crucial for inhibitory activity. N-(3-Nitro-phenyl)-4-(3-pyridyl)-2-pyrimidinamine (7) preferentially inhibited PKC-alpha (IC50 = 0.79 microM) and not the other subtypes tested. The inhibition constants of PKC-alpha and the antiproliferative effect on T24 human bladder carcinoma cells showed a qualitative correlation, although with some exceptions.


Subject(s)
Aniline Compounds/pharmacology , Enzyme Inhibitors/pharmacology , Isoenzymes/antagonists & inhibitors , Protein Kinase C/antagonists & inhibitors , Pyrimidines/pharmacology , Aniline Compounds/chemistry , Cell Division/drug effects , Enzyme Inhibitors/chemistry , Humans , Isoenzymes/metabolism , Protein Kinase C/metabolism , Pyrimidines/chemistry , Substrate Specificity , Tumor Cells, Cultured
15.
Int J Cancer ; 66(6): 753-9, 1996 Jun 11.
Article in English | MEDLINE | ID: mdl-8647645

ABSTRACT

A novel strategy for isolating potential anti-tumor drugs is presented. It is predicated on the idea that future anti-tumor drugs will be specific inhibitors of the signal-transduction pathways responsible for cell proliferation. Briefly, retroviral vectors are used to introduce focus-forming oncogenes into a test population of target cells, which are grown to confluence and treated with signal-transduction inhibitors. The inhibitors are screened for the ability to suppress the development of transformed foci without killing the confluent monolayer of non-transformed quiescent cells. For this work, a panel of inhibitors was first screened against the oncogene ras. The protein kinase C (PKC) inhibitor CGP 41251 and the protein tyrosine kinase (PTK) inhibitor CGP 45047 suppressed ras-induced focus formation and left a viable monolayer of quiescent cells. Focus inhibition was reversible; conversely, drug addition to developing foci retarded further expansion. CGP 41251 generally blocked proliferation of ras or control cells, suggesting that oncogenes cannot substitute for PKC. PTK inhibitors erbstatin and CGP 520 and phosphatase inhibitor okadaic acid failed to inhibit focus formation at concentrations toxic to the monolayer. Lavendustin A and CGP 47778A showed neither focus inhibition nor toxicity. In the complementary screen, a single inhibitor (CGP 41251) was tested against several oncogenes, including src, raf and polyomavirus middle T antigen. Focus formation by all oncogenes was suppressed. The strategy has several advantages over current drug-screening assays, and it can be adapted to large-scale screening with many drugs and many oncogenes.


Subject(s)
Drug Screening Assays, Antitumor/methods , Enzyme Inhibitors/pharmacology , Genetic Vectors/genetics , Oncogenes , Retroviridae/genetics , Signal Transduction/drug effects , Staurosporine/analogs & derivatives , 3T3 Cells , Alkaloids/pharmacology , Animals , Antigens, Polyomavirus Transforming/genetics , Benzamides/pharmacology , Cell Division/drug effects , Ethers, Cyclic/pharmacology , Genes, ras , Genes, src , Hydroquinones/pharmacology , Mice , Okadaic Acid , Phenols/pharmacology , Protein Kinase C/antagonists & inhibitors , Protein Serine-Threonine Kinases/genetics , Protein-Tyrosine Kinases/antagonists & inhibitors , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins c-raf , Styrenes/pharmacology , Sulfones/pharmacology , Thiazoles/pharmacology
16.
J Biol Chem ; 271(21): 12481-7, 1996 May 24.
Article in English | MEDLINE | ID: mdl-8647855

ABSTRACT

The Src family of kinases are held in an inactive state by interaction of their SH2 domain with a C-terminal phosphotyrosine. Dephosphorylation of this site can reactivate Src; however, recent evidence suggests that activation can also occur without dephosphorylation. In this study, platelet-derived growth factor receptor phosphorylation of Src on Tyr-213 specifically blocked binding of its SH2 domain to a phosphopeptide corresponding to the C-terminal regulatory sequence, while binding to other sequences, such as the platelet-derived growth factor receptor or a peptide from the epidermal growth factor receptor, was unaffected. Consequently, Src was activated over 50-fold. This is the first demonstration of regulation of a SH2 domain specificity by post-translational modification and is likely to be a general mechanism for regulation of all Src-like kinases.


Subject(s)
Oncogene Protein pp60(v-src)/metabolism , Protein-Tyrosine Kinases/metabolism , Tyrosine/metabolism , src Homology Domains , Amino Acid Sequence , Animals , Cell Line , Enzyme Activation , Mice , Mice, Inbred BALB C , Models, Molecular , Molecular Sequence Data , Oncogene Protein pp60(v-src)/chemistry , Peptide Mapping , Phosphopeptides/chemistry , Phosphorylation , Protein-Tyrosine Kinases/chemistry , Receptors, Platelet-Derived Growth Factor/metabolism , Spodoptera , Substrate Specificity
17.
Nat Med ; 2(5): 561-6, 1996 May.
Article in English | MEDLINE | ID: mdl-8616716

ABSTRACT

The bcr-abl oncogene, present in 95% of patients with chronic myelogenous leukemia (CML), has been implicated as the cause of this disease. A compound, designed to inhibit the Abl protein tyrosine kinase, was evaluated for its effects on cells containing the Bcr-Abl fusion protein. Cellular proliferation and tumor formation by Bcr-Abl-expressing cells were specifically inhibited by this compound. In colony-forming assays of peripheral blood or bone marrow from patients with CML, there was a 92-98% decrease in the number of bcr-abl colonies formed but no inhibition of normal colony formation. This compound may be useful in the treatment of bcr-abl-positive leukemias.


Subject(s)
Enzyme Inhibitors/pharmacology , Fusion Proteins, bcr-abl , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Piperazines/pharmacology , Protein-Tyrosine Kinases/antagonists & inhibitors , Proto-Oncogene Proteins c-abl/antagonists & inhibitors , Pyrimidines/pharmacology , Animals , Benzamides , Blood Cells/drug effects , Bone Marrow/drug effects , Dose-Response Relationship, Drug , Evaluation Studies as Topic , Humans , Imatinib Mesylate , Mice , Stem Cells/drug effects , Tumor Cells, Cultured
18.
Cancer Res ; 56(1): 100-4, 1996 Jan 01.
Article in English | MEDLINE | ID: mdl-8548747

ABSTRACT

Oncogenic activation of Abl proteins due to structural modifications can occur as a result of viral transduction or chromosomal translocation. The tyrosine protein kinase activity of oncogenic Abl proteins is known to be essential for their transforming activity. Therefore, we have attempted to identify selective inhibitors of the Abl tyrosine protein kinase. Herein we describe an inhibitor (CGP 57148) of the Abl and platelet-derived growth factor (PDGF) receptor protein-tyrosine kinases from the 2-phenylaminopyrimidine class, which is highly active in vitro and in vivo. Submicromolar concentrations of the compound inhibited both v-Abl and PDGF receptor autophosphorylation and PDGF-induced c-fos mRNA expression selectively in intact cells. In contrast, ligand-induced growth factor receptor autophosphorylation in response to epidermal growth factor (EGF), insulin-like growth factor-I, and insulin showed no or weak inhibition by high concentrations of CGP 57148. c-fos mRNA expression induced by EGF, fibroblast growth factor, or phorbol ester was also insensitive to inhibition by CGP 57148. In antiproliferative assays, the compound was more than 30-100-fold more potent in inhibiting growth of v-abl-transformed PB-3c cells and v-sis-transformed BALB/c 3T3 cells relative to inhibition of EGF-dependent BALB/MK cells, interleukin-3-dependent FDC-P1 cells, and the T24 bladder carcinoma line. Furthermore, anchorage-independent growth of v-abl- and v-sis-transformed BALB/c 3T3 cells was inhibited potently by CGP 57148. When tested in vivo, CGP 57148 showed antitumor activity at tolerated doses against tumorigenic v-abl- and v-sis-transformed BALB/c 3T3 cells. In contrast, CGP 57148 had no antitumor activity when tested using src-transformed BALB/c 3T3 cells. These findings suggest that CGP 57148 may have therapeutic potential for the treatment of diseases that involve abnormal cellular proliferation induced by Abl protein-tyrosine kinase deregulation or PDGF receptor activation.


Subject(s)
Oncogene Proteins v-abl/antagonists & inhibitors , Piperazines , Piperidines/pharmacology , Pyrimidines/pharmacology , Receptors, Platelet-Derived Growth Factor/antagonists & inhibitors , 3T3 Cells , Animals , Benzamides , Cell Line, Transformed , Growth Substances/pharmacology , Imatinib Mesylate , Mice , Mice, Inbred BALB C , Oncogene Proteins v-abl/metabolism , Proto-Oncogene Proteins c-fos/metabolism , Receptors, Platelet-Derived Growth Factor/metabolism , Tumor Cells, Cultured
19.
J Biol Chem ; 270(26): 15591-7, 1995 Jun 30.
Article in English | MEDLINE | ID: mdl-7797556

ABSTRACT

Following ligand binding, the epidermal growth factor receptor (EGF-R) autophosphorylates itself on tyrosine residues located in its carboxyl terminus; in vitro, three sites are highly phosphorylated, while two other sites are phosphorylated to lesser extents. In the presence of the Src protein-tyrosine kinase, in vitro phosphorylation of the minor autophosphorylation sites was increased, and four additional residues were phosphorylated. Following EGF stimulation, two (Tyr-891 and Tyr-920) were found to be phosphorylated in a colorectal cell line (DLD-1) and in a breast tumor cell line (MCF7). The remaining in vitro sites were not found to be highly phosphorylated in vivo. The sequences surrounding Tyr-891 and Tyr-920 match the reported consensus binding sequences for the SH2 domains of Src and the regulatory domain of phosphatidylinositol 3-kinase (p85 alpha), respectively. In vitro, both of these proteins were found to bind to Src-phosphorylated EGF-R with approximately 100-fold greater affinity than to autophosphorylated EGF-R, demonstrating that Src creates new sites for SH2 binding. Furthermore, Csk-inactivated Src was activated by interaction with Src-phosphorylated EGF-R but not by autophosphorylated EGF-R. Upon EGF treatment of MCF7 or three colorectal carcinoma cell lines (WiDr, DLD-1, and LS174T), the EGF-R coimmunoprecipitated with both p85 alpha and Src. Evidence is also presented that suggests that an EGF-R-related protein, ErbB2, may be involved in similar Src-mediated interactions. These data demonstrate that EGF-R is phosphorylated in vivo at non-autophosphorylation sites and that these novel sites can act as docking sites for Src, P85 alpha, and potentially other SH2-containing proteins. In addition, the data suggest a tyrosine phosphatase-independent mechanism for the elevation of Src activity in cells exposed to growth factors. Overexpression of Src, EGF-R, and/or ErbB2 in breast and colorectal tumor cells suggests the potential that such interactions may contribute to the transformed phenotype of these carcinomas.


Subject(s)
ErbB Receptors/metabolism , Oncogene Protein pp60(v-src)/metabolism , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Amino Acid Sequence , Humans , Molecular Sequence Data , Phosphatidylinositol 3-Kinases , Phosphorylation , Receptor, ErbB-2/metabolism , Tumor Cells, Cultured
20.
J Antibiot (Tokyo) ; 48(3): 191-8, 1995 Mar.
Article in English | MEDLINE | ID: mdl-7730151

ABSTRACT

Paeciloquinones A to F as well as versiconol have been isolated as inhibitors of protein tyrosine kinase from the culture broth of the fungus Paecilomyces carneus P-177. The novel anthraquinones inhibit epidermal growth factor receptor protein tyrosine kinase in the micromolar range. Two compounds, paeciloquinones A and C, are potent and selective inhibitors of the v-abl protein tyrosine kinase with an IC50 of 0.4 microM. Dependent on the fermentation conditions, partially different sets of paeciloquinones may be produced. An HPLC method allows separation of all major active components.


Subject(s)
Anthraquinones/isolation & purification , Paecilomyces/metabolism , Protein-Tyrosine Kinases/antagonists & inhibitors , Animals , Anthraquinones/pharmacology , Chromatography, High Pressure Liquid , ErbB Receptors/drug effects , Fermentation , Mice , Microbial Sensitivity Tests , Protein-Tyrosine Kinases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...