Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 53
Filter
Add more filters










Publication year range
1.
Bioorg Med Chem Lett ; 10(9): 945-9, 2000 May 01.
Article in English | MEDLINE | ID: mdl-10853665

ABSTRACT

5,7-Diphenyl-pyrrolo[2,3d]pyrimidines represent a new class of highly potent inhibitors of the tyrosine kinase c-Src (IC50 < 50 nM) with specificity against a panel of different tyrosine kinases. The substitution pattern on the two phenyl rings determines potency and specificity and provides a means to modulate cellular activity.


Subject(s)
Enzyme Inhibitors/chemical synthesis , Protein-Tyrosine Kinases/antagonists & inhibitors , Pyrimidines/chemical synthesis , Pyrroles/chemical synthesis , Animals , Blotting, Western , CSK Tyrosine-Protein Kinase , Chickens , Enzyme Inhibitors/pharmacology , Models, Molecular , Phosphorylation , Pyrimidines/pharmacology , Pyrroles/pharmacology , Structure-Activity Relationship , Substrate Specificity , src-Family Kinases
2.
Cancer Res ; 60(8): 2178-89, 2000 Apr 15.
Article in English | MEDLINE | ID: mdl-10786682

ABSTRACT

PTK787/ZK 222584 (1-[4-chloroanilino]-4-[4-pyridylmethyl] phthalazine succinate) is a potent inhibitor of vascular endothelial growth factor (VEGF) receptor tyrosine kinases, active in the submicromolar range. It also inhibits other class III kinases, such as the platelet-derived growth factor (PDGF) receptor beta tyrosine kinase, c-Kit, and c-Fms, but at higher concentrations. It is not active against kinases from other receptor families, such as epidermal growth factor receptor, fibroblast growth factor receptor-1, c-Met, and Tie-2, or intracellular kinases such as c-Src, c-Abl, and protein kinase C-alpha. PTK787/ZK 222584 inhibits VEGF-induced autophosphorylation of kinase insert domain-containing receptor (KDR), endothelial cell proliferation, migration, and survival in the nanomolar range in cell-based assays. In concentrations up to 1 microM, PTK787/ZK 222584 does not have any cytotoxic or antiproliferative effect on cells that do not express VEGF receptors. After oral dosing (50 mg/kg) to mice, plasma concentrations of PTK787/ZK 222584 remain above 1 microM for more than 8 h. PTK787/ZK 222584 induces dose-dependent inhibition of VEGF and PDGF-induced angiogenesis in a growth factor implant model, as well as a tumor cell-driven angiogenesis model after once-daily oral dosing (25-100 mg/kg). In the same dose range, it also inhibits the growth of several human carcinomas, grown s.c. in nude mice, as well as a murine renal carcinoma and its metastases in a syngeneic, orthotopic model. Histological examination of tumors revealed inhibition of microvessel formation in the interior of the tumor. PTK787/ZK 222584 is very well tolerated and does not impair wound healing. It also does not have any significant effects on circulating blood cells or bone marrow leukocytes as a single agent or impair hematopoetic recovery after concomitant cytotoxic anti-cancer agent challenge. This novel compound has therapeutic potential for the treatment of solid tumors and other diseases where angiogenesis plays an important role.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Carcinoma/drug therapy , Carcinoma/pathology , Endothelial Growth Factors/antagonists & inhibitors , Lymphokines/antagonists & inhibitors , Neovascularization, Pathologic/drug therapy , Phthalazines , Pyridines , Receptor Protein-Tyrosine Kinases/antagonists & inhibitors , Receptors, Growth Factor/antagonists & inhibitors , Angiogenesis Inhibitors/adverse effects , Angiogenesis Inhibitors/blood , Angiogenesis Inhibitors/therapeutic use , Animals , Bone Marrow Cells/cytology , Bone Marrow Cells/drug effects , Carcinoma/blood supply , Cell Division/drug effects , Disease Models, Animal , Dose-Response Relationship, Drug , Endothelial Growth Factors/pharmacology , Endothelium, Vascular/cytology , Endothelium, Vascular/drug effects , Hematopoiesis/drug effects , Humans , Kidney Neoplasms/blood supply , Kidney Neoplasms/drug therapy , Kidney Neoplasms/pathology , Leukocytes/cytology , Leukocytes/drug effects , Lymphokines/pharmacology , Mice , Mice, Nude , Neoplasm Metastasis/drug therapy , Neoplasm Metastasis/pathology , Neoplasm Transplantation , Neovascularization, Pathologic/pathology , Phosphorylation/drug effects , Platelet-Derived Growth Factor/antagonists & inhibitors , Platelet-Derived Growth Factor/pharmacology , Receptor Protein-Tyrosine Kinases/metabolism , Receptors, Growth Factor/metabolism , Receptors, Vascular Endothelial Growth Factor , Tumor Cells, Cultured , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors , Wound Healing/drug effects
3.
Pharmacol Ther ; 82(2-3): 293-301, 1999.
Article in English | MEDLINE | ID: mdl-10454207

ABSTRACT

CGP 41251 was originally identified as an inhibitor of protein kinase C (PKC), inhibiting mainly the conventional PKC subtypes, and subsequently shown to inhibit the vascular endothelial growth factor (VEGF) receptor kinase insert domain-containing receptor, which is involved in angiogenesis. CGP 41251 inhibits reversibly intracellular PKC activity, induction of c-fos and the corresponding activation of the mitogen-activated protein kinase induced by either tumor promoting phorbol esters, platelet-derived growth factor, or basic fibroblast growth factor, but not by the epidermal growth factor. CGP 41251 inhibited the ligand-induced autophosphorylation of the receptors for platelet-derived growth factor, stem cell factor, and VEGF (kinase insert domain-containing receptor) that correlated with the inhibition of the mitogen-activated protein kinase activation, but did not affect the ligand-induced autophosphorylation of the receptors for insulin, insulin-like growth factor-I, or epidermal growth factor. CGP 41251 showed broad antiproliferative activity against various tumor and normal cell lines in vitro, and is able to reverse the p-glycoprotein-mediated multidrug resistance of tumor cells in vitro. CGP 41251 showed in vivo antitumor activity as single agent and inhibited angiogenesis in vivo. Thus, CGP 41251 may suppress tumor growth by inhibiting tumor angiogenesis (via its effects on the VEGF receptor tyrosine kinases) in addition to directly inhibiting tumor cell proliferation (via its effects on PKCs).


Subject(s)
Antineoplastic Agents/therapeutic use , Protein Kinase Inhibitors , Receptors, Growth Factor/drug effects , Receptors, Platelet-Derived Growth Factor/drug effects , Staurosporine/analogs & derivatives , Endothelial Growth Factors/physiology , Enzyme Inhibitors/therapeutic use , Forecasting , Humans , In Vitro Techniques , Protein Binding , Protein Kinases/classification , Staurosporine/pharmacology , Staurosporine/therapeutic use
4.
Int J Cancer ; 81(4): 669, 1999 May 17.
Article in English | MEDLINE | ID: mdl-10336342
5.
J Med Chem ; 42(6): 1018-26, 1999 Mar 25.
Article in English | MEDLINE | ID: mdl-10090785

ABSTRACT

Using a pharmacophore model for ATP-competitive inhibitors interacting with the active site of the EGFR protein tyrosine kinase together with published X-ray crystal data of quercetin (2) in complex with the Hck tyrosine kinase and of deschloroflavopiridol (3b) in complex with CDK2, a putative binding mode of the isoflavone genistein (1) was proposed. Then, based on literature data suggesting that a salicylic acid function, which is represented by the 5-hydroxy-4-keto motif in 1, could serve as a pharmacophore replacement of a pyrimidine ring, superposition of 1 onto the potent EGFR tyrosine kinase inhibitor 4-(3'-chlorophenylamino)-6, 7-dimethoxyquinazoline (4) led to 3'-chloro-5,7-dihydroxyisoflavone (6) as a target structure which in fact was 10 times more potent than 1. The putative binding mode of 6 suggests a sulfur-aromatic interaction of the m-chlorophenyl moiety with Cys 773 in the "sugar pocket" of the EGFR kinase model. Replacement of the oxygen in the chromenone ring of 6 by a nitrogen atom further improved the inhibitory activity against the EGFR kinase. With IC50 values of 38 and 8 nM, respectively, the quinolones 11 and 12 were the most potent compounds of the series. N-Alkylation of 11 did not further improve enzyme inhibitory activity but led to derivatives with cellular activity in the lower micromolar range.


Subject(s)
Enzyme Inhibitors/chemical synthesis , ErbB Receptors/antagonists & inhibitors , Isoflavones/chemical synthesis , Protein-Tyrosine Kinases/antagonists & inhibitors , Quinolones/chemical synthesis , Animals , Binding Sites , Cell Division/drug effects , Cell Line , Drug Design , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Genistein/metabolism , Isoflavones/chemistry , Isoflavones/pharmacology , Mice , Models, Molecular , Phosphorylation , Protein Binding , Protein-Tyrosine Kinases/metabolism , Quinolones/chemistry , Quinolones/pharmacology , Structure-Activity Relationship
7.
Proc Natl Acad Sci U S A ; 95(20): 12069, 1998 Sep 29.
Article in English | MEDLINE | ID: mdl-9786782
8.
Cancer Res ; 58(17): 3883-90, 1998 Sep 01.
Article in English | MEDLINE | ID: mdl-9731498

ABSTRACT

CGP-48664, an inhibitor of the polyamine biosynthetic enzyme S-adenosylmethionine decarboxylase (AdoMetDC), is presently undergoing Phase 1 clinical trials as an experimental anticancer agent. We have shown previously (D. L. Kramer et al., J. Biol. Chem., 270: 2124-2132, 1995) that Chinese hamster ovary (CHO) cells that are made resistant to the growth inhibitory effects of the drug overexpress AdoMetDC because of a stable gene amplification. Unexpectedly, these same cells (CHO/644) were found to be insensitive to the growth inhibitory effects of N1,N11-diethylnorspermine (DENSPM)-a polyamine analogue also undergoing Phase 1 clinical trials-despite accumulating approximately 5 times more analogue than parental cells. We now report that treatment of CHO/664 cells with DENSPM results in the formation of numerous large cytoplasmic vacuoles, which on the basis of electron microscopy and cytochemical staining seem to be lysosomal in origin. A series of newly established CHO cell lines made differentially resistant to 1, 3, 10, 30, and 100 microM CGP-48664 by chronic exposure were used to demonstrate that vacuole formation correlated with the accumulation of extremely high levels of DENSPM without increasing growth inhibition. These same cells were used to show that AdoMetDC gene overexpression as indicated by mRNA levels was unrelated to vacuole formation; cells resistant to 100 microM CGP-48664 displayed a 170-fold increase in AdoMetDC mRNA levels and formed vacuoles in response to DENSPM, whereas those resistant to 10 microM CGP-48664 displayed a 120-fold increase in AdoMetDC mRNA levels and failed to form vacuoles. Despite accumulating to high intracellular levels, DENSPM was much less effective than spermine at down-regulating ornithine decarboxylase and polyamine transport activities in highly resistant cells. Similarly, DENSPM was less able to induce spermidine/spermine N1-acetyltransferase activity in cells that formed vacuoles than in those that did not. Overall, natural polyamines failed to induce vacuoles and various analogues of DENSPM were used to probe the structural specificity of the effect. The data are consistent with the probability that DENSPM is sequestered to high concentrations in lysosomal vacuoles of CGP-48664-resistant cells and is, therefore, not available to interact with polyamine regulatory sites or to cytotoxically affect cell growth. In addition to implicating the lysosome as a potential new site of CGP-48664 drug action that could be involved in antitumor activity and/or host toxicities, the findings also suggest a potential mechanism of cell resistance to analogues such as DENSPM.


Subject(s)
Adenosylmethionine Decarboxylase/antagonists & inhibitors , Amidines/pharmacology , Antineoplastic Agents/pharmacology , Indans/pharmacology , Lysosomes/metabolism , Spermine/analogs & derivatives , Adenosylmethionine Decarboxylase/genetics , Animals , CHO Cells , Cricetinae , Drug Resistance, Neoplasm , Lysosomes/ultrastructure , RNA, Messenger/analysis , Spermine/metabolism , Spermine/pharmacology , Vacuoles/drug effects , Vacuoles/metabolism
9.
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
10.
Mol Biochem Parasitol ; 97(1-2): 13-9, 1998 Nov 30.
Article in English | MEDLINE | ID: mdl-9879883

ABSTRACT

Polyamines are essential for cell growth and differentiation and therefore, S-adenosylmethionine decarboxylase (SAMDC), a key regulatory enzyme of the polyamine biosynthesis, is considered as a potentially important target for chemotherapy of filarial infections. Recombinant Onchocerca volvulus SAMDC was expressed in Escherichia coli and characterised. The enzyme activity was found to be stimulated 15-fold by addition of 1 mM putrescine. The Km-value for S-adenosylmethionine was determined to be 36 microM. Furthermore, the efficiencies of SAMDC inhibitors were analysed: Berenil inhibits the enzyme activity competitively with a Ki-value of 0.1 microM. MDL 73811 acts as an irreversible inhibitor with a Ki-value of 1.4 microM. Recently synthesised aromatic methylglyoxal bis(guanylhydrazone) analogues demonstrated high efficacy as inhibitors of the SAMDCs. Some of these analogues exhibited Ki-values of 5 and 14 nM for the Onchocerca enzyme, a result which shows an up to 100-fold increase in specificity compared to the value of 0.47 microM for methylglyoxal bis(guanylhydrazone). These inhibitors might have potential as drug candidates against filarial worms.


Subject(s)
Adenosylmethionine Decarboxylase/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , Helminth Proteins/antagonists & inhibitors , Mitoguazone/pharmacology , Onchocerca volvulus/enzymology , Organic Chemicals , Adenosylmethionine Decarboxylase/genetics , Adenosylmethionine Decarboxylase/metabolism , Animals , Cloning, Molecular/methods , Deoxyadenosines/pharmacology , Diminazene/analogs & derivatives , Diminazene/pharmacology , Escherichia coli/genetics , Filaricides/pharmacology , Filarioidea/drug effects , Genetic Vectors , Helminth Proteins/genetics , Helminth Proteins/metabolism , Humans , Polyamines/metabolism , Recombinant Proteins/biosynthesis
11.
J Med Chem ; 40(22): 3601-16, 1997 Oct 24.
Article in English | MEDLINE | ID: mdl-9357527

ABSTRACT

In the course of the random screening of a pool of CIBA chemicals, the two pyrazolopyrimidines 1 and 2 have been identified as fairly potent inhibitors of the EGF-R tyrosine kinase. Using a pharmacophore model for ATP-competitive inhibitors interacting with the active site of the EGF-R protein tyrosine kinase (PTK), the class of the pyrazolo[3,4-d]pyrimidines was then optimized in an interactive process leading to a series of 4-(phenylamino)-1H-pyrazolo[3,4-d]-pyrimidines as highly potent inhibitors of the EGF-R tyrosine kinase. The most potent compounds 13, 14, 15, 17, 19, 22, 26, 28, and 30 of this series inhibited the EGF-R PTK with IC50 values below 10 nM. High selectivity toward a panel of nonreceptor tyrosine kinases (c-Src, v-Abl and serine/threonine kinases (PKC alpha, CDK1) was observed. In cells, EGF-stimulated cellular tyrosine phosphorylation was inhibited by compounds 13, 15, 19, 22, and 23 at IC50 values below 50 nM, whereas PDGF-induced tyrosine phosphorylation was not affected by concentrations up to 10 microM, thus indicating high selectivity for the inhibition of the ligand-activated EGF-R signal transduction pathway. Compounds 15 and 19 inhibited proliferation of the EGF-dependent MK cell line with IC50 values below 0.5 microM. In addition, two compounds, 9 and 11, showing satisfactory oral bioavailability in mice after oral administration, exhibited good in vivo efficacy at doses of 12.5 and 50 mg/kg in a nude mouse tumor model using xenografts of the EGF-R overexpressing A431 cell line. From SAR studies, a binding mode for 4-(phenylamino)-1H-pyrazolo[3,4-d]pyrimidines, especially for compound 15, at the ATP-binding site of the EGF-R tyrosine kinase is proposed. 4-(Phenylamino)-1H-pyrazolo[3,4-d]pyrimidines represent a new class of highly potent tyrosine kinase inhibitors which preferentially inhibit the EGF-mediated signal transduction pathway and have the potential for further evaluation as anticancer agents.


Subject(s)
Drug Design , Enzyme Inhibitors/chemical synthesis , ErbB Receptors/antagonists & inhibitors , Pyrimidines/chemical synthesis , 3T3 Cells , Animals , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/pharmacology , Biological Availability , Enzyme Inhibitors/pharmacokinetics , Enzyme Inhibitors/pharmacology , Female , Humans , Magnetic Resonance Spectroscopy , Mass Spectrometry/methods , Mice , Mice, Inbred BALB C , Models, Chemical , Pyrimidines/pharmacokinetics , Pyrimidines/pharmacology , Structure-Activity Relationship , Tumor Cells, Cultured
12.
Antimicrob Agents Chemother ; 41(9): 1922-5, 1997 Sep.
Article in English | MEDLINE | ID: mdl-9303385

ABSTRACT

Ornithine decarboxylase (ODC), the target enzyme of D,L-alpha-difluoromethylornithine (DFMO), was investigated in four DFMO-tolerant Trypanosoma brucei rhodesiense isolates from East Africa and two DFMO-susceptible T. b. gambiense isolates from West Africa. Neither drug uptake nor inhibition of ODC activity by DFMO in cellular extracts differed in the two trypanosome subspecies. However, the specific ODC activity of the cellular extracts was three times as high in T. b. rhodesiense isolates as in T. b. gambiense isolates. Furthermore, a significant difference in the turnover rate of ODC was observed. The time required to induce a 50% reduction of T. b. rhodesiense ODC activity under cycloheximide pressure (tentative half-life) was about 4.3 h, whereas that required for T. b. gambiense ODC was longer than 18 h. We concluded that the higher specific ODC activity and faster enzyme turnover contributed to a substantial degree to the DFMO tolerance observed in the East African T. b. rhodesiense isolates.


Subject(s)
Eflornithine/pharmacology , Enzyme Inhibitors/pharmacology , Ornithine Decarboxylase Inhibitors , Ornithine Decarboxylase/metabolism , Trypanocidal Agents/pharmacology , Trypanosoma brucei rhodesiense/drug effects , Trypanosoma brucei rhodesiense/enzymology , Animals , Blotting, Northern , Drug Resistance , Eflornithine/pharmacokinetics , Enzyme Inhibitors/pharmacokinetics , Half-Life , Putrescine/pharmacology , RNA, Messenger/metabolism , Trypanocidal Agents/pharmacokinetics , Trypanosoma brucei rhodesiense/metabolism
13.
J Antibiot (Tokyo) ; 50(5): 395-401, 1997 May.
Article in English | MEDLINE | ID: mdl-9207909

ABSTRACT

The known bisalkylated 2,5-dihydroxybenzoquinones didemethylasterriquinone D and isocochliodinol as well as the new metabolites semicochliodinol A and B have been isolated as inhibitors of HIV-1 protease from the culture broth of the fungus Chrysosporium merdarium P-5656. The structures were elucidated by spectroscopic methods. The NMR spectra of two compounds were completely assigned. The metabolites inhibit HIV-1 protease with an IC50 value as low as 0.17 microM and epidermal growth factor receptor protein tyrosine kinase at 15 to 60 microM and are therefore valuable lead compounds for these targets. Molecular modelling of the HIV-1-protease-inhibitor complexes showed hydrogen bonding between the dihydroxybenzoquinone moiety of didemethylasterriquinone D and isocochliodinol to both active-site aspartic acids (Asp25/Asp25') of the protease and the indole parts of the inhibitors filling the P2 and P2' pockets of the protease.


Subject(s)
Benzoquinones/chemistry , Benzoquinones/isolation & purification , HIV Protease Inhibitors/chemistry , HIV Protease Inhibitors/isolation & purification , Indoles/chemistry , Indoles/isolation & purification , Benzoquinones/pharmacology , Chromatography, High Pressure Liquid , Chrysosporium/metabolism , Fermentation , HIV Protease Inhibitors/pharmacology , Humans , Indoles/pharmacology , Models, Molecular , Structure-Activity Relationship
14.
J Pharm Belg ; 52(2): 88-96, 1997.
Article in English | MEDLINE | ID: mdl-9193132

ABSTRACT

One of the most promising targets for the rational design of anti-cancer drugs is the family of the EGF-receptor protein tyrosine kinases. Despite the high sequence homology within the ATP-binding region of protein tyrosine and/or serine threonine kinases, ATP-competitive compounds have the potential to be selective inhibitors of protein kinases. Dianilino-phthalimides CGP 52 411 and CGP 53,353 have been identified as potent and ATP-competitive inhibitors of the EGF-R tyrosine kinase with no or only minor activity against a panel of tyrosine and serine/threonine kinases. Using a calculated 3-D computer model of the catalytic domain of the EGF-R-tyrosine kinase together with CGP 52 411 as example of an ATP-competitive inhibitor, a pharmacophore model for ATP-competitive inhibitors in the active site of the EGF-R PTK was developed. With the help of this model, 4-phenylamino-7H-pyrrolo[2,3-d]pyrimidines were then identified as new potent EGF-R PTK inhibitors. In an interactive process, the class of the 4-phenylamino-pyrrolo-pyrimidines was optimized and structure-activity-relationship of a series of derivatives thereof are discussed. In vitro, the most active compounds (CGP 59 326, CGP 60 261, CGP 62 706) inhibited the EGF-R tyrosine kinase with IC50 value between 6-30 nM. High selectivity towards a panel of non-receptor tyrosine kinases (c-SRC, v-Abl) and serine/threonine kinases (PKC alpha, PKA) was observed. Kinetic analysis revealed competitive type kinetics relative to ATP. In cells, EGF-stimulated cellular tyrosine phosphorylation was inhibited by these compounds at IC50 values between 0.1-0.3 microM, whereas the ligand-induced receptor autophosphorylation of the PDGR-R was not effected by concentrations up to 100 microM. Furthermore, CGP 59 326, CGP 60 261, CGP 62 706 were able to selectively inhibit c-fos mRNA expression in EGF-dependent cell lines with (IC50) approx. 0.1-1 microM) but not in EGF-independent cell systems (IC50 > 100 microM). Proliferation of the EGF-dependent MK cell line was inhibited with similar IC50 values. In addition, CGP 59 326 and CGP 62 706 showed good in vivo efficacy at low doses after oral or subcutaneous administration in nude mice tumor models using xenografts of the EGF-dependent A431 cell lines. The ED50 values were between 1.5-2 mg/kg. Phenylamino-pyrrolo-pyrimidines therefore represent a new series of tyrosine kinase inhibitors which preferentially inhibit the EGF-mediated signal transduction pathway and have the characteristics for further evaluation as anticancer agents.


Subject(s)
Adenosine Triphosphate/metabolism , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/pharmacology , ErbB Receptors/chemistry , ErbB Receptors/drug effects , Protein-Tyrosine Kinases/antagonists & inhibitors , Animals , Binding Sites/drug effects , Drug Design , Humans , Models, Molecular
15.
Biochem J ; 322 ( Pt 1): 297-302, 1997 Feb 15.
Article in English | MEDLINE | ID: mdl-9078276

ABSTRACT

Mammalian S-adenosylmethionine decarboxylase (AdoMetDC) catalyses a regulatory important step in the biosynthesis of polyamines and is a potential target for therapeutic agents against various parasitic diseases and proliferative disorders. In the present study we examined the effects of a newly synthesized AdoMetDC inhibitor. 4-amidinoindan-1-one 2'-amidinohydrazone (CGP 48664), on polyamine metabolism in the mouse leukaemia cell line L1210. Treatment of the cells with 2 microM CGP 48664 led to a depletion of cellular spermidine and spermine. The putrescine content, in contrast, was markedly increased. Cells seeded in the presence of the inhibitor showed a significant decrease in growth rate, which was fully reversed by the addition of 2 microM spermidine or 1 microM spermine. The syntheses of ornithine decarboxylase and AdoMetDC were greatly increased in cells treated with CGP 48664. These increases were not correlated with similar changes in the mRNA levels, indicating the involvement of a translational mechanism. CGP 48664 was demonstrated to be a very poor competitor of spermidine uptake in the L1210 cells. L1210 cells deficient in polyamine transport were as sensitive to the antiproliferative effect of the inhibitor as were the parental cells, indicating that CGP 48664 did not enter the cells by the polyamine transport system. In addition to inhibiting AdoMetDC, CGP 48664 stabilized the enzyme against degradation. In the present study we also demonstrated that aminoguanidine (AMG), which is frequently used in cellular systems to inhibit any action of serum polyamine oxidase, apparently inhibits AdoMetDC by an irreversible mechanism that markedly stabilizes the enzyme against proteolytic degradation. CGP 48664 and the parental compound methylglyoxal bis(guanylhydrazone), which is also a potent inhibitor of AdoMetDC, contain one or two AMG-like moieties; the importance of these residues in the inhibition of AdoMetDC is discussed.


Subject(s)
Adenosylmethionine Decarboxylase/antagonists & inhibitors , Adenosylmethionine Decarboxylase/drug effects , Amidines/pharmacology , Indans/pharmacology , Adenosylmethionine Decarboxylase/metabolism , Animals , Cell Division/drug effects , Enzyme Activation/drug effects , Enzyme Stability/drug effects , Leukemia L1210/enzymology , Leukemia L1210/metabolism , Leukemia L1210/pathology , Mice , Ornithine Decarboxylase/metabolism , Polyamines/metabolism
16.
Anticancer Res ; 16(5A): 2517-23, 1996.
Article in English | MEDLINE | ID: mdl-8917345

ABSTRACT

Induction of the polyamine acetylating enzyme, spermidine/spermine N1-acetyltransferase (SSAT), is one of several biochemical effects associated with the antiproliferative action of polyamine analogs such as N1, N11 diethylnorspermine (DENSPM). Findings to date indicate that this complex and extremely potent gene response involves increased gene transcription, stabilization of mRNA, enhanced translation and protein stabilization. In this study, SSAT-directed antisense oligonucleotide analogs (AOs) were studied for their ability to prevent enzyme induction by DENSPM. Nine 18-mer fully phosphorothioate modified AOs targeting the start codon, exon 6, stop codon and polyadenylation regions of the human SSAT mRNA were synthesized and evaluated in MALME-3M human melanoma cells prior to and during a 6 hr treatment with 10 microM DENSPM. The most effective AOs were those targeting sequences in the stop codon region. Of these, AO-82 suppressed DENSPM induction of SSAT activity, enzyme protein and mRNA by 70-80%. The quantitative similarity of these effects suggests AO interference with mRNA stabilization, a property apparently mediated by sequences located in the stop codon region. Growth inhibition by DENSPM in the presence of the terminally phophorothioated analogs of AO-82 remained similar to that produced by DENSPM alone. While it is possible that SSAT induction may not be involved in analog-mediated antiproliferative activity, a more likely interpretation is that the approximately 50% suppression of the enzyme response achieved in growth studies is not sufficient to abrogate growth inhibition.


Subject(s)
Acetyltransferases/drug effects , Gene Expression Regulation, Enzymologic/drug effects , Oligonucleotides, Antisense/pharmacology , RNA, Messenger/drug effects , Acetyltransferases/genetics , Acetyltransferases/metabolism , Enzyme Induction/drug effects , Enzyme Induction/genetics , Humans , Oligonucleotides, Antisense/chemistry , RNA, Messenger/metabolism , Spermine/analogs & derivatives , Spermine/pharmacology , Transfection , Tumor Cells, Cultured
17.
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
18.
Antimicrob Agents Chemother ; 40(6): 1442-7, 1996 Jun.
Article in English | MEDLINE | ID: mdl-8726017

ABSTRACT

A series of novel aromatic derivatives based on the structure of methylglyoxal bis(guanylhydrazone) (MGBG) was examined for in vitro antitrypanosomal activities and cytotoxicities for human cells. One-third of the compounds tested showed trypanocidal activity at concentrations below 0.5 microM after an incubation period of 72 h. Structure-activity analysis revealed that bicyclic compounds with homocyclic rings and unmodified termini were the most active compounds. Results obtained in three laboratories employing different methods and trypanosome populations consistently ranked compound CGP 40215A highest. This compound had a 50% inhibitory concentration of 0.0045 microM for Trypanosoma brucei rhodesiense, was also active against other trypanosome species, including a multidrug-resistant Trypanosoma brucei brucei, and was significantly less toxic than other compounds tested for a human adenocarcinoma cell line, with a 50% inhibitory concentration of 1.14 mM. The effect of CGP 40215A was time and dose dependent, and low concentrations of the compound required exposure times of > 2 days to exert trypanocidal activity. Compounds were inactive against Leishmania donovani and Trypanosoma cruzi amastigotes in murine macrophages in vitro.


Subject(s)
Adenosylmethionine Decarboxylase/antagonists & inhibitors , Mitoguazone/analogs & derivatives , Trypanocidal Agents/pharmacology , Trypanosoma/drug effects , Adenocarcinoma/drug therapy , Animals , Humans , Structure-Activity Relationship , Time Factors , Tumor Cells, Cultured/drug effects
19.
J Med Chem ; 39(12): 2285-92, 1996 Jun 07.
Article in English | MEDLINE | ID: mdl-8691423

ABSTRACT

Using a pharmacophore model for ATP-competitive inhibitors interacting with the active site of the EGF-R protein tyrosine kinase (PTK), 4-(phenylamino)-7H-pyrrolo[2,3-d]pyrimidines have been identified as a novel class of potent EGF-R protein tyrosine kinase inhibitors. In an interactive process, this class of compounds was then optimized. 13, 14, 28, 36, 37, and 44, the most potent compounds of this series, inhibited the EGF-R PTK with IC50 values in the low nanomolar range. High selectivity toward a panel of nonreceptor tyrosine kinases (c-Src, v-Abl) and serine/threonine kinases (PKC alpha, PKA) was observed. Kinetic analysis revealed competitive type kinetics relative to ATP. In cells, EGF-stimulated cellular tyrosine phosphorylation was inhibited by compounds 13, 36, 37, and 44 at IC50 values between 0.1 and 0.4 microM, whereas PDGF-induced tyrosine phosphorylation was not affected by concentrations up to 10 microM. In addition, these compounds were able to selectively inhibit c-fos mRNA expression in EGF-dependent cell lines with IC50 values between 0.1 and 2 microM, but did not affect c-fos mRNA induction in response to PDGF or PMA (IC50 >100 microM). Proliferation of the EGF-dependent MK cell line was inhibited with similar IC50 values. From SAR studies, a binding mode for 4-(phenylamino)-7H-pyrrolo[2,3-d]pyrimidines as well as for the structurally related 4-(phenylamino)quinazolines at the ATP-binding site of the EGF-R tyrosine kinase is proposed. 4-(Phenylamino)7H-pyrrolo[2,3-d]pyrimidines therefore represent a new class of highly potent tyrosine kinase inhibitors which preferentially inhibit the EGF-mediated signal transduction pathway and have the potential for further evaluation as anticancer agents.


Subject(s)
Adenosine Triphosphate/metabolism , Antineoplastic Agents/chemistry , Enzyme Inhibitors/pharmacology , ErbB Receptors/antagonists & inhibitors , Growth Inhibitors/pharmacology , Pyrimidines/pharmacology , 3T3 Cells/drug effects , Animals , Binding Sites/drug effects , Computer-Aided Design , Drug Design , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Genes, fos , Growth Inhibitors/chemical synthesis , Growth Inhibitors/chemistry , Mice , Models, Molecular , Phosphorylation/drug effects , Protein Kinase Inhibitors , Protein Processing, Post-Translational/drug effects , Pyrimidines/chemistry , Signal Transduction/drug effects , Structure-Activity Relationship
SELECTION OF CITATIONS
SEARCH DETAIL
...