Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 36
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Semin Oncol ; 28(5 Suppl 16): 80-5, 2001 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-11706399

RESUMEN

Overexpression of the erbB family of receptor tyrosine kinases has been implicated in a variety of tumors including breast, lung, prostate, and brain. Most solid tumors express one or more of these receptors, which can often be related to tumor aggressiveness and poor patient prognosis. CI-1033, a pan-erbB tyrosine kinase inhibitor, is a clinically promising agent that is active against all four members of the erbB receptor tyrosine kinase family. In vitro studies of human cancer cell lines indicate that CI-1033 results in prompt, potent, and sustained inhibition of tyrosine kinase activity. This inhibition is highly selective for erbB1 (epidermal growth factor receptor), erbB2, erbB3, and erbB4 without inhibiting tyrosine kinase activity of receptors such as platelet-derived growth factor receptor, fibroblast growth factor receptor, and insulin receptor, even at high concentrations. Treatment of athymic nude mice bearing xenografts of human A431 epidermoid carcinoma, H125 non-small cell lung carcinoma, and SF-767 glioblastoma results in highly significant suppression of tumor growth. The major toxicity in animals is diarrhea, which is more severe at higher doses. In animal models, all side effects are reversible on cessation of treatment. Thus, CI-1033, which is currently undergoing phase I clinical trials, holds significant potential for use in a broad range of solid tumors.


Asunto(s)
Antineoplásicos/farmacología , Inhibidores Enzimáticos/farmacología , Morfolinas/farmacología , Proteínas Tirosina Quinasas/antagonistas & inhibidores , Receptor ErbB-2/antagonistas & inhibidores , Animales , Antineoplásicos/uso terapéutico , Ensayos de Selección de Medicamentos Antitumorales , Inhibidores Enzimáticos/uso terapéutico , Humanos , Morfolinas/uso terapéutico
2.
J Med Chem ; 44(3): 429-40, 2001 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-11462982

RESUMEN

4-Anilinoquinazoline- and 4-anilinopyrido[3,4-d]pyrimidine-6-acrylamides are potent pan-erbB tyrosine kinase inactivators, and one example (CI-1033) is in clinical trial. A series of analogues with a variety of Michael acceptor units at the 6-position were prepared to define the structural requirements for irreversible inhibition. A particular goal was to determine whether additional functions to increase solubility could be appended to the Michael acceptor. Substituted acrylamides were prepared by direct acylation of the corresponding 6-amines with the requisite acid or acid chloride. Vinylsulfonamide derivatives were obtained by acylation of the amines with chloroethylsulfonyl chloride followed by base-promoted elimination. Vinylsulfone and vinylsulfine derivatives were prepared by oxidation and base elimination of a hydroxyethylthio intermediate. The compounds were evaluated for their inhibition of phosphorylation of the isolated EGFR enzyme and for inhibition of EGF-stimulated autophosphorylation of EGFR in A431 cells and of heregulin-stimulated autophosphorylation of erbB2 in MDA-MB 453 cells. Substitution at the nitrogen of the acrylamide was tolerated only with a methyl group; larger substituents were dystherapeutic, and no substitution at all was tolerated at the acrylamide alpha-carbon. In contrast, while electron-donating groups at the acrylamide beta-carbon were not useful, even quite large electron-withdrawing groups (which increase its electrophilicity) were tolerated. A series of derivatives with solubility-enhancing substituents linked to the acrylamide beta-carbon via amides were potent irreversible inhibitors of isolated EGFR (IC50s = 0.4-1.1 nM), with weakly basic morpholine and imidazole derivatives being the best. Vinylsulfonamides were also potent and irreversible inhibitors, but vinylsulfones and vinylsulfines were reversible and only poorly active. Two compounds were evaluated against A431, H125, and MCF-7 xenografts in nude mice but were inferior in these assays to the clinical trial compound CI-1033.


Asunto(s)
Antineoplásicos/síntesis química , Inhibidores Enzimáticos/síntesis química , Receptores ErbB/antagonistas & inhibidores , Pirimidinas/síntesis química , Quinazolinas/síntesis química , Animales , Antineoplásicos/química , Antineoplásicos/farmacología , Ensayos de Selección de Medicamentos Antitumorales , Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/farmacología , Receptores ErbB/metabolismo , Concentración 50 Inhibidora , Ratones , Fosforilación , Pirimidinas/química , Pirimidinas/farmacología , Quinazolinas/química , Quinazolinas/farmacología , Solubilidad , Relación Estructura-Actividad , Trasplante Heterólogo , Células Tumorales Cultivadas
3.
J Med Chem ; 44(12): 1915-26, 2001 Jun 07.
Artículo en Inglés | MEDLINE | ID: mdl-11384237

RESUMEN

In continuing our search for medicinal agents to treat proliferative diseases, we have discovered 2-substituted aminopyrido[2,3-d]pyrimidin-7-yl ureas as a novel class of soluble, potent, broadly active tyrosine kinase (TK) inhibitors. An efficient route was developed that enabled the synthesis of a wide variety of analogues with substitution on several positions of the template. From the lead structure 1, several series of analogues were made that examined the C-6 aryl substituent, a variety of water solublizing substitutents at the C-2 position, and urea or other acyl functionality at the N-7 position. Compounds of this series were competitive with ATP and displayed submicromolar to low nanomolar potency against a panel of TKs, including receptor (platelet-derived growth factor, PDGFr; fibroblast growth factor, FGFr;) and nonreceptor (c-Src) classes. Several of the most potent compounds displayed submicromolar inhibition of PDGF-mediated receptor autophosphorylation in rat aortic vascular smooth muscle cells and low micromolar inhibition of cellular growth in five human tumor cell lines. One of the more thoroughly evaluated members, 32, with IC50 values of 0.21 microM (PDGFr), 0.049 microM (bFGFr), and 0.018 microM (c-Src), was evaluated in in vivo studies against a panel of five human tumor xenografts, with known and/or inferred dependence on the EGFr, PDGFr, and c-Src TKs. Compound 32 produced a tumor growth delay of 14 days against the Colo-205 colon xenograft model.


Asunto(s)
Antineoplásicos/síntesis química , Inhibidores Enzimáticos/síntesis química , Proteínas Tirosina Quinasas/antagonistas & inhibidores , Pirimidinas/síntesis química , Urea/análogos & derivados , Urea/síntesis química , Células 3T3 , Animales , Antineoplásicos/química , Antineoplásicos/farmacología , Proteína Tirosina Quinasa CSK , División Celular/efectos de los fármacos , Neoplasias del Colon , Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/farmacología , Receptores ErbB/antagonistas & inhibidores , Glioma , Humanos , Indicadores y Reactivos , Cinética , Ratones , Conformación Molecular , Estructura Molecular , Fosforilación , Pirimidinas/química , Pirimidinas/farmacología , Ratas , Receptores de Factores de Crecimiento de Fibroblastos/antagonistas & inhibidores , Receptores del Factor de Crecimiento Derivado de Plaquetas/antagonistas & inhibidores , Proteínas Recombinantes/antagonistas & inhibidores , Relación Estructura-Actividad , Transfección , Células Tumorales Cultivadas , Urea/química , Urea/farmacología , Familia-src Quinasas
4.
J Med Chem ; 43(22): 4200-11, 2000 Nov 02.
Artículo en Inglés | MEDLINE | ID: mdl-11063616

RESUMEN

A series of 3-aryl-1,6-naphthyridine-2,7-diamines and related 2-ureas were prepared and evaluated as inhibitors of the FGF receptor-1 tyrosine kinase. Condensation of 4,6-diaminonicotinaldehyde and substituted phenylacetonitriles gave intermediate naphthyridine-2,7-diamines, and direct reaction of the monoanion of these (NaH/DMF) with alkyl or aryl isocyanates selectively gave the 2-ureas in varying yields (23-93%). For the preparation of more soluble 7-alkylamino-2-ureas, a number of protecting groups for the 2-amine were evaluated (phthaloyl, 4-methoxybenzyl) following selective blocking of the 7-amine (trityl), but these were not superior to the (required) 2-tert-Bu-urea group itself. Direct alkylation of the anion of the (unprotected) 7-amino group with excess 4-(3-chloropropyl)morpholine in DMF gave low (10%) yields of the desired product, but alkylation of the 7-acetamido anion, followed by mild alkaline hydrolysis, raised this to 64%. 3-Phenyl analogues were nonspecific inhibitors of isolated c-Src, FGFR, and PDGFR tyrosine kinases, whereas 3-(2,6-dichlorophenyl) analogues were most effective against c-Src and FGFR, and 3-(3,5-dimethoxyphenyl) derivatives showed high selectivity for FGFR alone. A water-soluble (7-morpholinylpropylamino) analogue retained high FGFR potency (IC(50) 31 nM) and selectivity. Pairwise comparison of the 1, 6-naphthyridines and the corresponding known pyrido[2,3-d]pyrimidine analogues showed little differences in potency or patterns of selectivity, suggesting that the 1-aza atom of the latter is not important for activity. A 7-acetamide derivative inhibited the growth of FGFR-expressing tumor cell lines and was particularly potent against HUVECs (IC(50) 4 nM). This compound was also a very potent inhibitor of HUVEC microcapillary formation (IC(50) 0.01 nM) and Matrigel invasion (IC(50) 7 nM) and showed significant in vivo antitumor effects in a highly vascularized mammary adenocarcinoma 16/c model at nontoxic doses. The compounds are worthy of further evaluation as antiangiogenesis agents.


Asunto(s)
Antineoplásicos/síntesis química , Inhibidores Enzimáticos/síntesis química , Naftiridinas/síntesis química , Proteínas Tirosina Quinasas Receptoras/antagonistas & inhibidores , Receptores de Factores de Crecimiento de Fibroblastos/antagonistas & inhibidores , Urea/análogos & derivados , Urea/síntesis química , Animales , Antineoplásicos/química , Antineoplásicos/farmacología , Ensayos de Selección de Medicamentos Antitumorales , Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/farmacología , Humanos , Ratones , Ratones Endogámicos , Naftiridinas/química , Naftiridinas/farmacología , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos , Relación Estructura-Actividad , Células Tumorales Cultivadas , Urea/química , Urea/farmacología , Ensayos Antitumor por Modelo de Xenoinjerto
5.
Cancer Chemother Pharmacol ; 45(3): 231-8, 2000.
Artículo en Inglés | MEDLINE | ID: mdl-10663641

RESUMEN

PURPOSE: The involvement of the EGF receptor (EGFr) family of receptors in cancers suggests that a selective inhibitor of the tyrosine kinase activity of the EGFr family could have a therapeutic effect. PD 0169414, an anilinoquinazoline, is a potent irreversible inhibitor of the EGFr family tyrosine kinase activity with IC(50) values of 0.42 nM against the isolated EGF receptor, and 4.7 nM and 22 nM against EGF- and heregulin-mediated receptor phosphorylation in A431 and MDA-MB-453 cells, respectively. METHODS AND RESULTS: Oral administration of 260 mg/kg per day PD 0169414 for 15 days to animals bearing advanced-stage A431 epidermoid carcinoma produced a 28.2-day delay in tumor growth and resulted in three complete and three partial tumor regressions in six animals. Toxicity at this dose level was limited to <6% loss of initial body weight. Doses of 160 and 100 mg/kg per day produced tumor growth delays of 29.5 and 20.9 days and two and one complete regressions in six animals, respectively. Subcutaneous, intraperitoneal, and oral routes of administration have also shown in vivo antitumor activity of PD 0169414 in a panel of human tumor xenografts. Responsive tumor lines include A431 (human epidermoid carcinoma), H125 (NSCL carcinoma), MCF-7 and UISO-BCA1 (human breast carcinoma), and SK-OV-03 (human ovarian carcinoma). The therapeutic effect ranged from delayed tumor growth (6.4 days delayed tumor growth for 14 days of treatment) to tumor regressions (32.2 days delayed tumor growth and five partial regressions in six animals) in these model systems. CONCLUSION: PD 0169414 is a specific, irreversible inhibitor of EGFr family tyrosine kinases with significant in vivo activity against a variety of relevant human tumor xenografts.


Asunto(s)
Antineoplásicos/farmacocinética , Inhibidores Enzimáticos/farmacocinética , Receptores ErbB/efectos de los fármacos , Neoplasias Experimentales/tratamiento farmacológico , Quinazolinas/farmacología , Quinazolinas/farmacocinética , Proteínas Tirosina Quinasas Receptoras/antagonistas & inhibidores , Células 3T3 , Animales , Antineoplásicos/efectos adversos , Antineoplásicos/farmacología , Área Bajo la Curva , Vías de Administración de Medicamentos , Esquema de Medicación , Inhibidores Enzimáticos/efectos adversos , Inhibidores Enzimáticos/farmacología , Receptores ErbB/metabolismo , Humanos , Bombas de Infusión Implantables , Hígado/efectos de los fármacos , Hígado/patología , Ratones , Ratones Endogámicos ICR , Ratones Desnudos , Ratones SCID , Trasplante de Neoplasias , Neoplasias Experimentales/patología , Fosforilación/efectos de los fármacos , Quinazolinas/sangre , Quinazolinas/uso terapéutico , Proteínas Tirosina Quinasas Receptoras/metabolismo , Trasplante Heterólogo , Resultado del Tratamiento , Células Tumorales Cultivadas
6.
J Med Chem ; 43(7): 1380-97, 2000 Apr 06.
Artículo en Inglés | MEDLINE | ID: mdl-10753475

RESUMEN

4-Anilinoquinazoline- and 4-anilinopyrido[3,2-d]pyrimidine-6-acrylamides substituted with solubilizing 7-alkylamine or 7-alkoxyamine side chains were prepared by reaction of the corresponding 6-amines with acrylic acid or acrylic acid anhydrides. In the pyrido[3,2-d]pyrimidine series, the intermediate 6-amino-7-alkylamines were prepared from 7-bromo-6-fluoropyrido[3,2-d]pyrimidine via Stille coupling with the appropriate stannane under palladium(0) catalysis. This proved a versatile method for the introduction of cationic solubilizing side chains. The compounds were evaluated for their inhibition of phosphorylation of the isolated EGFR enzyme and for inhibition of EGF-stimulated autophosphorylation of EGFR in A431 cells and of heregulin-stimulated autophosphorylation of erbB2 in MDA-MB 453 cells. Quinazoline analogues with 7-alkoxyamine solubilizing groups were potent irreversible inhibitors of the isolated EGFR enzyme, with IC(50[app]) values from 2 to 4 nM, and potently inhibited both EGFR and erbB2 autophosphorylation in cells. 7-Alkylamino- and 7-alkoxyaminopyrido[3,2-d]pyrimidines were also irreversible inhibitors with equal or superior potency against the isolated enzyme but were less effective in the cellular autophosphorylation assays. Both quinazoline- and pyrido[3,2-d]pyrimidine-6-acrylamides bound at the ATP site alkylating cysteine 773, as shown by electrospray ionization mass spectrometry, and had similar rates of absorptive and secretory transport in Caco-2 cells. A comparison of two 7-propoxymorpholide analogues showed that the pyrido[3,2-d]pyrimidine-6-acrylamide had greater amide instability and higher acrylamide reactivity, being converted to glutathione adducts in cells more rapidly than the corresponding quinazoline. This difference may contribute to the observed lower cellular potency of the pyrido[3,2-d]pyrimidine-6-acrylamides. Selected compounds showed high in vivo activity against A431 xenografts on oral dosing, with the quinazolines being superior to the pyrido[3,2-d]pyrimidines. Overall, the quinazolines proved superior to previous analogues in terms of aqueous solubility, potency, and in vivo antitumor activity, and one example (CI 1033) has been selected for clinical evaluation.


Asunto(s)
Acrilamidas/síntesis química , Inhibidores Enzimáticos/síntesis química , Receptores ErbB/antagonistas & inhibidores , Morfolinas/síntesis química , Pirimidinas/síntesis química , Quinazolinas/síntesis química , Acrilamidas/química , Acrilamidas/farmacología , Animales , Antineoplásicos/síntesis química , Antineoplásicos/química , Antineoplásicos/farmacología , Transporte Biológico/efectos de los fármacos , Células CACO-2 , Ensayos de Selección de Medicamentos Antitumorales , Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/farmacología , Receptores ErbB/metabolismo , Humanos , Ratones , Ratones Desnudos , Morfolinas/química , Morfolinas/farmacología , Trasplante de Neoplasias , Fosforilación , Pirimidinas/química , Pirimidinas/farmacología , Quinazolinas/química , Quinazolinas/farmacología , Receptor ErbB-2/metabolismo , Solubilidad , Espectrometría de Masa por Ionización de Electrospray , Trasplante Heterólogo , Células Tumorales Cultivadas
7.
J Med Chem ; 41(22): 4365-77, 1998 Oct 22.
Artículo en Inglés | MEDLINE | ID: mdl-9784112

RESUMEN

Screening of a compound library led to the identification of 2-amino-6-(2,6-dichlorophenyl)-8-methylpyrido[2,3-d]pyrimidine (1) as a inhibitor of the platelet-derived growth factor receptor (PDGFr), fibroblast growth factor receptor (FGFr), and c-src tyrosine kinases (TKs). Replacement of the primary amino group at C-2 of 1 with a 4-(N,N-diethylaminoethoxy)phenylamino group yielded 2a, which had greatly increased activity against all three TKs. In the present work, variation of the aromatic group at C-6 and of the alkyl group at N-8 of the pyrido[2,3-d]pyrimidine core provided several analogues that retained potency, including derivatives that were biased toward inhibition of the TK activity of PDGFr. Analogues of 2a with a 3-thiophene or an unsubstituted phenyl group at C-6 were the most potent inhibitors. Compound 54, which had IC50 values of 31, 88, and 31 nM against PDGFr, FGFr, and c-src TK activity, respectively, was active in a variety of PDGF-dependent cellular assays and blocked the in vivo growth of three PDGF-dependent tumor lines.


Asunto(s)
Inhibidores Enzimáticos/síntesis química , Proteínas Tirosina Quinasas/antagonistas & inhibidores , Piridonas/síntesis química , Pirimidinas/síntesis química , Receptores del Factor de Crecimiento Derivado de Plaquetas/antagonistas & inhibidores , Células 3T3 , Animales , Disponibilidad Biológica , Proteína Tirosina Quinasa CSK , Ensayos de Selección de Medicamentos Antitumorales , Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/farmacocinética , Inhibidores Enzimáticos/farmacología , Humanos , Masculino , Ratones , Ratones Desnudos , Músculo Liso Vascular/citología , Músculo Liso Vascular/efectos de los fármacos , Fosforilación , Piridonas/química , Piridonas/farmacocinética , Piridonas/farmacología , Pirimidinas/química , Pirimidinas/farmacocinética , Pirimidinas/farmacología , Ratas , Ratas Wistar , Receptores de Factores de Crecimiento de Fibroblastos/antagonistas & inhibidores , Receptores del Factor de Crecimiento Derivado de Plaquetas/metabolismo , Relación Estructura-Actividad , Trasplante Heterólogo , Células Tumorales Cultivadas , Familia-src Quinasas
8.
J Med Chem ; 41(17): 3276-92, 1998 Aug 13.
Artículo en Inglés | MEDLINE | ID: mdl-9703473

RESUMEN

While engaged in therapeutic intervention against a number of proliferative diseases, we have discovered the 2-aminopyrido[2, 3-d]pyrimidin-7(8H)-ones as a novel class of potent, broadly active tyrosine kinase (TK) inhibitors. An efficient route was developed that enabled the synthesis of a wide variety of analogues with substitution on several positions of the template. From the lead structure 2, a series of analogues bearing variable substituents at the C-2 position and methyl or ethyl at N-8 was made. Compounds of this series were competitive with ATP and displayed submicromolar to low nanomolar potency against a panel of TKs, including receptor (platelet-derived growth factor, PDGFr; fibroblast growth factor, FGFr; epidermal growth factor, EGFr) and nonreceptor (c-Src) classes. One of the more thoroughly evaluated members was 63 with IC50 values of 0.079 microM (PDGFr), 0.043 microM (bFGFr), 0.044 microM (EGFr), and 0.009 microM (c-Src). In cellular studies, 63 inhibited PDGF-mediated receptor autophosphorylation in a number of cell lines at IC50 values of 0.026-0.002 microM and proliferation of two PDGF-dependent lines at 0.3 microM. It also caused inhibition of soft agar colony formation in three cell lines that overexpress the c-Src TK, with IC50 values of 0.33-1.8 microM. In in vivo studies against a panel of seven xenograft tumor models with known and/or inferred dependence on the EGFr, PDGFr, and c-Src TKs, compound 63 produced a tumor growth delay of 10.6 days against the relatively refractory SK-OV-3 ovarian xenograft and also displayed activity against the HT-29 tumor. In rat oral bioavailability studies, compound 63 plasma concentrations declined in a biexponential manner, and systemic plasma clearance was high relative to liver blood flow. Finally, in rat metabolism studies, HPLC chromatography identified two metabolites of 63, which were proved by mass spectrometry and synthesis to be the primary amine (58) and N-oxide (66). Because of the excellent potency of 63 against selected TKs, in vitro and in vivo studies are underway for this compound in additional tumor models dependent upon PDGFr, FGFr, and c-Src to assess its potential for advancement to clinical trials.


Asunto(s)
Antineoplásicos/síntesis química , Inhibidores Enzimáticos/síntesis química , Neoplasias Ováricas/tratamiento farmacológico , Proteínas Tirosina Quinasas/antagonistas & inhibidores , Pirimidinonas/síntesis química , Proteínas Tirosina Quinasas Receptoras/antagonistas & inhibidores , Células 3T3 , Animales , Antineoplásicos/química , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Biotransformación , División Celular/efectos de los fármacos , Cisplatino/uso terapéutico , Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/farmacocinética , Inhibidores Enzimáticos/farmacología , Femenino , Humanos , Masculino , Ratones , Ratones Desnudos , Conformación Molecular , Estructura Molecular , Factor de Crecimiento Derivado de Plaquetas/farmacología , Pirimidinonas/química , Pirimidinonas/farmacocinética , Pirimidinonas/farmacología , Ratas , Relación Estructura-Actividad , Trasplante Heterólogo , Células Tumorales Cultivadas
9.
J Med Chem ; 41(5): 742-51, 1998 Feb 26.
Artículo en Inglés | MEDLINE | ID: mdl-9513602

RESUMEN

The 4-[(3-bromophenyl)amino]pyrido[3,4-d]pyrimidine PD 158780 is a very potent in vitro inhibitor of the tyrosine kinase activity of the epidermal growth factor receptor (EGFR) (IC50 0.08 nM), and other members of the erbB family, by competitive binding at the ATP site of these signal transduction enzymes. A series of analogues of PD 158780 bearing solubilizing functions off the 6-methylamino substituent were prepared by reaction of the 6-fluoro derivatives with appropriate amine nucleophiles. These were evaluated for their ability to inhibit the tyrosine phosphorylating action of EGF-stimulated full-length EGFR enzyme and for inhibition of autophosphorylation of the EGFR in A431 human epidermoid carcinoma cells in culture. The most effective analogues were those bearing weakly basic substituents through a secondary amine linkage, which proved water-soluble (> 10 mM) and potent (IC50S generally < 1 nM). No clear SAR could be discerned for these compounds with respect to amine base strength or the distance of the cationic center from the chromophore, suggesting that 6-substituents are in a favorable area of bulk tolerance in the enzyme binding site. More distinct SAR emerged for the ability of the compounds to inhibit EGFR autophosphorylation in A431 cells, where analogues bearing lipophilic weak bases were preferred. Representative analogues were evaluated for antitumor effectiveness against four in vivo tumor models. Significant in vivo activity was observed in estrogen-dependent MCF-7 breast and A431 epidermoid tumors. Marginal activity was seen in an EGFR-transfected tumor model, suggesting that while this cell line requires EGF for clone formation in soft agar, other growth factors may be able to replace EGF in vivo. Also, no activity was seen against the SK-OV-3 ovarian cancer model, which is known to express other EGF receptor family members (although it is not clear whether these are absolutely required for growth in vivo). While substantial growth delays were seen in A431 and MCF-7 tumor models, the treated tumors remained approximately the same size throughout therapy, suggesting that the compounds are cytostatic rather than cytotoxic under these test conditions. It remains to be determined if more prolonged therapy has cytotoxic effects in vivo, resulting in net tumor cell kill.


Asunto(s)
Antineoplásicos/síntesis química , Inhibidores Enzimáticos/síntesis química , Receptores ErbB/antagonistas & inhibidores , Proteínas Tirosina Quinasas/antagonistas & inhibidores , Pirimidinas/química , Proteínas Tirosina Quinasas Receptoras/antagonistas & inhibidores , Células 3T3 , Animales , Antineoplásicos/uso terapéutico , Neoplasias de la Mama/tratamiento farmacológico , Carcinoma de Células Escamosas/tratamiento farmacológico , Carcinoma de Células Escamosas/metabolismo , Inhibidores Enzimáticos/farmacología , Receptores ErbB/genética , Femenino , Humanos , Ratones , Ratones Desnudos , Estructura Molecular , Neoplasias Ováricas/tratamiento farmacológico , Fosforilación , Pirimidinas/farmacología , Relación Estructura-Actividad , Transfección , Células Tumorales Cultivadas
10.
J Med Chem ; 40(24): 3915-25, 1997 Nov 21.
Artículo en Inglés | MEDLINE | ID: mdl-9397172

RESUMEN

The general class of 4-(phenylamino)quinazolines are potent (some members with IC50 values << 1 nM) and selective inhibitors of the tyrosine kinase activity of the epidermal growth factor receptor (EGFR), via competitive binding at the ATP site of the enzyme, but many of the early analogues had poor aqueous solubility (<< 1 mM). A series of 7-substituted 4-[(3-bromophenyl)-amino]pyrido[4,3-d]pyrimidines, together with selected (3-methylphenyl)amino analogues, were prepared by reaction of the analogous 7-fluoro derivatives with appropriate amine nucleophiles in 2-BuOH or aqueous 1-PrOH. All of the compounds were evaluated for their ability to inhibit the tyrosine-phosphorylating action of EGF-stimulated full-length EGFR enzyme. Selected analogues were also evaluated for their inhibition of autophosphorylation of the EGF receptor in A431 human epidermoid carcinoma cells in culture and against A431 tumor xenografts in mice. Analogues bearing a wide variety of polyol, cationic, and anionic solubilizing substituents retained activity, but the most effective in terms of both increased aqueous solubility (> 40 mM) and retention of overall inhibitory activity (IC50's of 0.5-10 nM against isolated enzyme and 8-40 nM for inhibition of EGFR autophosphorylation in A431 cells) were weakly basic amine derivatives. These results are broadly consistent with a proposed model for the binding of these compounds to EGFR, in which the 6- and 7-positions of the pyridopyrimidine ring are in a largely hydrophobic binding region of considerable steric freedom, at the entrance of the adenine binding cleft. The most active cationic analogues have a weakly basic side chain where the amine moiety is three or more carbon atoms away from the nucleus. Two of the compounds (bearing weakly basic morpholinopropyl and strongly basic (dimethylamino)butyl solubilizing groups) produced in vivo tumor growth delays of 13-21 days against advanced stage A431 epidermoid xenografts in nude mice, when administered i.p. twice per day on days 7-21 posttumor implant. Treated tumors did not increase in size during therapy and resumed growth at the termination of therapy, indicating an apparent cytostatic effect for these compounds under these treatment conditions. The data suggest that continuous long-term therapy with these compounds may result in substantial tumor growth inhibition.


Asunto(s)
Inhibidores Enzimáticos/síntesis química , Inhibidores Enzimáticos/farmacología , Receptores ErbB/antagonistas & inhibidores , Proteínas Tirosina Quinasas/antagonistas & inhibidores , Pirimidinas/síntesis química , Pirimidinas/farmacología , Animales , Carcinoma de Células Escamosas/tratamiento farmacológico , Receptores ErbB/metabolismo , Humanos , Ratones , Ratones Desnudos , Proteínas Tirosina Quinasas/metabolismo , Solubilidad , Relación Estructura-Actividad , Trasplante Heterólogo , Células Tumorales Cultivadas
11.
J Med Chem ; 40(4): 413-26, 1997 Feb 14.
Artículo en Inglés | MEDLINE | ID: mdl-9046331

RESUMEN

A small series of 2,2'-diselenobis(1H-indoles) was synthesized as redox-modified congeners of our earlier reported 2,2'-dithiobis(1H-indole) series. Utilizing chemistry similar to that developed earlier for the disulfur series, compounds were made from 2-halogeno-3-indolecarboxylic acid precursors bearing various polar functionality at the C-3 position and small alkyl substituents at the N-1 position of the indole nucleus. Additional compounds were derived from (R)- or (S)-tryptophan via a novel application of diselenium dichloride as an electrophilic source of diselenium, and a much improved process to a 2,2'-dithiobis(1H-indole) congener was developed utilizing disulfur dichloride as a source of disulfur. Against isolated epidermal growth factor receptor (EGFr), platelet-derived growth factor receptor (PDGFr), and v-src tyrosine kinases, compounds in this series displayed broad inhibitory activity with IC50 = 0.9 to > 100 microM vs EGFr, 3.4 to > 50 microM vs PDGFr, and 0.4-6.7 microM vs v-src. In general, compounds derived from tryptophan displayed the greatest potency against EGFr and those from 2-halogeno-3-indolecarboxylic acids greater potency against PDGFr and v-src. Enzyme kinetics studies showed that both classes of compounds display primarily noncompetitive inhibition with respect to either ATP or peptide substrate. The sulfhydryl reducing agent dithiothreitol (DTT) caused a general decrease in inhibition of the EGFr and v-src tyrosine kinases by both the diselenium and disulfur series with the reversal of enzyme inhibition occurring less readily within the diselenium series. In whole cell studies, compounds of this class were growth inhibitory against Swiss 3T3 mouse fibroblasts with IC50 values from 0.5 to 19.5 microM, and the observed SAR was different from that of the 2,2'-dithiobis(1H-indoles). A comparative study in the same cell line on the effects of the 2,2'-diselenobis(1H-indole) derived from (R)-tryptophan vs its disulfur congener on growth factor mediated tyrosine phosphorylation showed that this compound significantly inhibited EGFr and PDGFr (in response to its ligand) autophosphorylation with complete suppression at 25 and 5 microM, respectively. Tyrosine phosphorylation of an 85 kDa protein typically phosphorylated in response to bFGF was also exquisitely sensitive to this compound, and it displayed inhibitory effects on DNA, RNA, and protein synthesis at submicromolar concentrations. The disulfur congener exhibited a qualitatively similar pattern; however, its potency was 10-fold less. This same diselenium/disulfur pair was evaluated in vivo against the B16 melanoma, colon carcinoma 26, and M5076 sarcoma murine tumors, and the A431 epidermoid, and C6 glioma human tumor xenografts. At maximum tolerated doses (1.8 and 5.0 mg/kg/injection, respectively), neither the diselenium nor disulfur congener was effective against the C6 glioma when administered intraperitoneally on a d1-9 schedule. Studies were also carried out against the A431 epidermoid xenograft to evaluate the same pair of compounds via continuous subcutaneous infusion from Alzet miniosmotic pumps. The maximum dose that could be administered daily was limited by compound solubility. Neither compound produced an antitumor effect in a 7-day continuous infusion study. In the 27-day study, the disulfur compound was inactive whereas the diselenium compound produced a 10.8-day growth delay without appreciable treatment related weight loss. The in vitro and in vivo findings offer a mechanistic rationale as to why the 2,2'-diselenobis(1H-indoles) are more potent inhibitors than their disulfur congeners.


Asunto(s)
Inhibidores Enzimáticos/farmacología , Indoles/química , Compuestos de Organoselenio/química , Proteínas Tirosina Quinasas/antagonistas & inhibidores , Azufre , Células 3T3 , Animales , Neoplasias del Colon/tratamiento farmacológico , Ditiotreitol/farmacología , Inhibidores Enzimáticos/química , Receptores ErbB/antagonistas & inhibidores , Humanos , Cinética , Melanoma/tratamiento farmacológico , Ratones , Fosforilación , Sarcoma Experimental/tratamiento farmacológico , Relación Estructura-Actividad , Células Tumorales Cultivadas
12.
Invest New Drugs ; 13(4): 295-302, 1996.
Artículo en Inglés | MEDLINE | ID: mdl-8824347

RESUMEN

PD153035 is a potent (Ki = 6 pm) and specific inhibitor of the epidermal growth factor (EGF) receptor tyrosine kinase that suppresses tyrosine phosphorylation of the EGF receptor in A431 cells at nanomolar concentrations in cell culture. We have examined the pharmacokinetics of this compound and its ability to rapidly suppress phosphorylation of the EGF receptor in A431 human epidermoid tumors grown as xenografts in immunodeficient nude mice. Following a single i.p. dose of 80 mg/kg, the drug levels in the plasma and tumor rose to 50 and 22 microM within 15 minutes. While the plasma levels of PD153035 fell below 1 microM by 3 hours, in the tumors it remained at micromolar concentrations for at least 12 hours. The tyrosine phosphorylation of the EGF receptor was rapidly suppressed by 80-90% in the tumors. However receptor phosphorylation returned to control levels after 3 hours despite the continued presence of the drug at concentrations which, based on previous in vitro results, were predicted to maintain inhibition. EGF-stimulated tyrosine kinase activity in tumor extracts was decreased and recovered in parallel with the effects of PD153035 on receptor phosphorylation though the activity had reached only about half of the control activity after three hours. These results demonstrate the potential for using small molecule inhibitors to inhibit the EGF receptor tyrosine kinase in vivo, though a fair evaluation of their potential anti-cancer activity will have to wait for solutions to problems with sustained delivery which may allow us to maintain suppression of EGF receptor phosphorylation.


Asunto(s)
Carcinoma de Células Escamosas/tratamiento farmacológico , Carcinoma de Células Escamosas/enzimología , Inhibidores Enzimáticos/farmacología , Receptores ErbB/antagonistas & inhibidores , Neoplasias/tratamiento farmacológico , Neoplasias/enzimología , Quinazolinas/farmacología , Animales , División Celular/efectos de los fármacos , Inhibidores Enzimáticos/farmacocinética , Humanos , Ratones , Ratones Desnudos , Neoplasias/metabolismo , Quinazolinas/farmacocinética , Trasplante Heterólogo , Células Tumorales Cultivadas
13.
Cancer Res ; 54(16): 4412-8, 1994 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-8044790

RESUMEN

[SP-4-3-(R)]-[1,1-cyclobutanedicarboxylato(2-)](2-methyl-1,4- butane-diamine-N,N')platinum (CI-973) is a cisplatin analogue that is currently in clinical trial. Preclinically, CI-973 retained activity against L1210, P388, K562, and human ovarian carcinoma sublines resistant to cisplatin in vitro. CI-973 also retained substantial activity [ratio of median life span of treated to control groups x 100% (%T/C) > 190] against cisplatin resistant L1210 and P388 sublines in vivo. Good activity (stasis or tumor burden reduction) was also obtained against five murine solid tumors including breast, colon, and sarcoma. Binary combination therapy with CI-973 and seven clinically utilized anticancer agents was evaluated against murine tumors in vivo for the ability of each combination to produce a superior therapeutic response compared to optimal single agent therapy alone at tolerated doses. The seven agents combined with CI-973 were mitomycin C, cyclophosphamide, doxorubicin, vinblastine, etoposide, ifosfamide, and methotrexate. Of the combination regimens evaluated, only the combination of CI-973 and methotrexate was therapeutically superior to single agent therapy. Against i.v. inoculated P388 leukemia, combination therapy with CI-973 at 32 mg/kg/injection and methotrexate at 43 mg/kg/injection produced 5.3 logs greater cell kill (%T/C = 284, with one cell surviving therapy) than either methotrexate therapy (%T/C = 208, with 2.5 x 10(5) cells surviving therapy) or CI-973 therapy (%T/C = 193 with 2.5 x 10(6) cells surviving therapy) alone. The combination toxicity index of 1.0 indicated additive normal host tissue toxicity with the same target organs for dose limiting toxicity. To better understand the enhanced cell kill in vivo, the combination of CI-973 and methotrexate was evaluated against P388 leukemia in vitro. Clonogenic survival studies showed that the combination of CI-973 and methotrexate was additive rather than synergistic with respect to cell killing in vitro. The lack of greater than additive cell kill in vitro suggested that the mechanism responsible for synergistic kill in vivo was not operative in vitro. The in vivo results suggest that the combination of CI-973 and methotrexate may be useful in the clinic.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Carboplatino/análogos & derivados , Animales , Carboplatino/uso terapéutico , Neoplasias del Colon/tratamiento farmacológico , Ciclofosfamida/administración & dosificación , Doxorrubicina/administración & dosificación , Ensayos de Selección de Medicamentos Antitumorales , Etopósido/administración & dosificación , Ifosfamida/administración & dosificación , Leucemia P388/tratamiento farmacológico , Neoplasias Mamarias Animales/tratamiento farmacológico , Melanoma Experimental/tratamiento farmacológico , Metotrexato/administración & dosificación , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Endogámicos DBA , Mitomicina/administración & dosificación , Sarcoma Experimental/tratamiento farmacológico , Vinblastina/administración & dosificación
14.
J Med Chem ; 37(17): 2630-6, 1994 Aug 19.
Artículo en Inglés | MEDLINE | ID: mdl-8064793

RESUMEN

The first two analogs 5a,b of a new class of neutral large-ring square-planar Pt(II) chelate complexes of the generic structure [Pt(cis-1,4-dach)X2] were synthesized via a refined technique, structurally characterized by NMR (1H, 13C, 195Pt), FAB mass spectrometry, and X-ray crystallography, and evaluated for antitumor activity in vitro and in vivo in sensitive and Pt-resistant murine leukemia cell systems. An X-ray crystal structure analysis confirmed that [Pt(cis-1,4-dach)malonate] 5b is monomeric and that the cis-1,4-diaminocyclohexane (dach) ligand is incorporated in a unique and previously unknown locked boat conformation. Complex 5b crystallized as colorless rectangular plates in the orthorhombic space group Pcmn with Z = 4 and the lattice parameters a = 6.239(1) A, b = 9.965(2) A, and c = 18.437(4) A. Important structural parameters are Pt-O = 2.024(5) A, Pt-N = 2.021(6) A, N-Pt-N = 100 degrees, and N-Pt-O = 85 degrees; R = 0.0515, Rw = 0.0635. Antitumor results in murine tumor models show that the parent molecule 5a (X2 = 2 Cl) (a) is more dose potent than cisplatin against the leukemias and solid tumors examined, (b) possesses significant activity against cisplatin-resistant leukemias, but exhibits partial cross-resistance with cisplatin, and (c) may possess a spectrum of activity different from that of cisplatin. Antitumor test results in vitro indicate that (a) 5a is at least equivalent to cisplatin in dose potency and effectiveness in the leukemia cell systems studied except in the [Pt(1,2-dach)Cl2]-resistant L1210 cell line, (b) the cisplatin-resistant leukemia cell systems exhibit partial cross-resistance to 5a, (c) 5a possesses either comparable or greater cytotoxicity than the reference complexes, CI-973 (3) and bis(platinum) complex 4, and (d) 5a is more effective (approximately 18-fold) than [Pt(1R,2R-dach)Cl2] 2 in inhibiting growth in the Pt(1,2-dach)-resistant L1210 cell line, suggesting that [Pt(cis-1,4-dach)Cl2] is either not recognized as or is not acting as a "typical" Pt(dach) complex. The encouraging antitumor activity of 5a, coupled with a 10-fold higher aqueous solubility compared to [Pt(1R,2R-dach)-Cl2] 2 warrants the following future studies: synthesis of selected analogs, elucidating the nature of Pt-DNA binding sites, the mechanism of action, and the mechanistic basis for the lack of cross-resistance of [Pt(cis-1,4-dach)Cl2] against the [Pt(1,2-dach)Cl2]-resistant L1210 cell line.


Asunto(s)
Antineoplásicos/síntesis química , Ciclohexilaminas , Compuestos Organoplatinos/síntesis química , Animales , Antineoplásicos/química , Antineoplásicos/uso terapéutico , Cristalografía por Rayos X , Leucemia L1210/tratamiento farmacológico , Leucemia Experimental/tratamiento farmacológico , Ligandos , Espectroscopía de Resonancia Magnética , Melanoma Experimental/tratamiento farmacológico , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C3H , Ratones Endogámicos C57BL , Ratones Endogámicos DBA , Conformación Molecular , Estructura Molecular , Compuestos Organoplatinos/química , Compuestos Organoplatinos/uso terapéutico , Sarcoma Experimental/tratamiento farmacológico , Relación Estructura-Actividad
15.
Anticancer Drug Des ; 9(4): 331-51, 1994 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-7916900

RESUMEN

We present a general drug discovery strategy to find novel inhibitors of tyrosine kinases. This scheme includes the production of initial protein targets for primary screening, an evaluation of enzyme potency and specificity, biochemical and biological effects on various cellular processes in whole-cell models, and strategies for in vivo evaluation of antitumor activity. These aspects are illustrated using a newly discovered class of tyrosine kinase inhibitors, the 2-thioindoles. Certain members of this class of compounds inhibit specific tyrosine kinases at low micromolar concentrations and exhibit a distinct structure-activity relationship, as well as enzyme specificity depending on the chemical substitution. These compounds suppress growth factor-mediated tyrosine phosphorylation and mitogenesis in viable cells and, in some cases, exhibit marked specificity for these effects depending on the substituents on the indole ring system. The issue is stressed that since inhibitors of signal transduction pathways represent an entirely new class of potential antitumor agents, distinct from past and currently used cancer therapies, alternative in vivo tumor models may be needed as well as different requirements for dose levels, scheduling and endpoint evaluation. These inherent difficulties emphasize a need for more basic research at the in vivo stage of drug evaluation to enhance model development and provide a better understanding of the pharmacology for these newer classes of drugs.


Asunto(s)
Antineoplásicos/farmacología , Evaluación Preclínica de Medicamentos/métodos , Proteínas Tirosina Quinasas/antagonistas & inhibidores , Células 3T3 , Animales , Antineoplásicos/síntesis química , División Celular , Ensayos de Selección de Medicamentos Antitumorales , Factor 2 de Crecimiento de Fibroblastos/fisiología , Humanos , Indoles/química , Indoles/farmacología , Ratones , Ratones Desnudos , Fosforilación , Factor de Crecimiento Derivado de Plaquetas/fisiología , Relación Estructura-Actividad , Células Tumorales Cultivadas , Tirosina/metabolismo
16.
J Med Chem ; 35(24): 4526-32, 1992 Nov 27.
Artículo en Inglés | MEDLINE | ID: mdl-1335074

RESUMEN

The biological activity of a series of dinuclear bis(platinum) complexes of formula [(cis-PtX2-(NH3)]2(NH2(CH2)nNH2)] (X = Cl, n = 4-9, compounds 6-11; X2 = malonate, n = 5 or 6, compounds 12 and 13) is described in selected murine leukemia, murine solid tumor, and human tumor cell lines and in murine leukemia cell lines rendered resistant to cisplatin (cis-[Pt(NH3)2Cl2]). The bis(platinum) compounds showed greater activity in vitro against murine tumor cell lines resistant to either cisplatin or DACH ([Pt(DACH)Cl2]). The resistance factor is dependent on chain length of the diamine, and the structural feature of a dinuclear complex is of general use in reducing cross-resistance with cisplatin. In vivo [(cis-PtCl2(NH3)]2(NH2(CH2)5NH2)] (7) showed a % T/C of 204 against murine L1210 leukemia resistant to cisplatin compared to a % T/C of 104 for cisplatin itself at optimal doses. The complex [(Pt(mal)(NH3)]2(NH2(CH2)6NH2)] (13) was highly active in the colon 26 tumor line with 3/10 tumor-free survivors (dose of 186 mg/kg, ip D1,5,9); however, 13 was subject to substantial cross-resistance in the cisplatin resistant L1210 leukemia (% T/C 139 versus % T/C of 223 in the sensitive line). In four selected human tumor lines in vitro, compounds 6-11 were uniformly more potent than cisplatin. In the corresponding xenografts, compound 7 showed greater activity in the HCT-8 (coloadenocarcinoma) and H23 (nonsmall cell lung), but diminished potency in AH125 and H520 (both nonsmall cell lung) lines in comparison to cisplatin. Retention of activity against cisplatin-resistant cell lines and a different spectrum of activity compared to cisplatin in some human tumor cell lines suggest that this class of complexes is mechanistically different from mononuclear complexes and worthy of further development toward clinical trials.


Asunto(s)
Antineoplásicos/uso terapéutico , Diaminas/química , Platino (Metal)/uso terapéutico , Adenocarcinoma/tratamiento farmacológico , Animales , Antineoplásicos/síntesis química , Antineoplásicos/farmacología , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Cisplatino/uso terapéutico , Neoplasias del Colon/tratamiento farmacológico , Resistencia a Medicamentos , Humanos , Leucemia L1210/tratamiento farmacológico , Leucemia Experimental/tratamiento farmacológico , Neoplasias Pulmonares/tratamiento farmacológico , Malonatos , Neoplasias Mamarias Experimentales/tratamiento farmacológico , Melanoma Experimental/tratamiento farmacológico , Ratones , Ratones Desnudos , Estructura Molecular , Platino (Metal)/química , Relación Estructura-Actividad , Células Tumorales Cultivadas
17.
Int J Radiat Oncol Biol Phys ; 22(3): 549-51, 1992.
Artículo en Inglés | MEDLINE | ID: mdl-1531213

RESUMEN

RB 6145, the ring-opened analog of RSU 1069, and PD 130908, the desoxy ring-opened analog of RSU 1069, were compared to RSU 1069 for their emetic potential in dogs. When RB 6145 and PD 130908 were administered intravenously at doses ranging from 20% to 50% of the mouse equivalent maximum tolerated dose (MTD), both analogs were less emetic than RSU 1069 on a molar basis. Furthermore, the 5HT3 antagonist ondansetron prevented emesis at doses as high as 75% of the MTD. The reactivity, and hence the emetic liability of these compounds, is thought to be mediated by formation of the corresponding aziridine intermediate. In mouse plasma, both analogs rapidly converted to two products, the reactive aziridine and a stable oxiazolidinone species formed upon reaction with bicarbonate in the blood. A positive correlation exists between the amounts of aziridine formed by these analogs and their emetic potential.


Asunto(s)
Antieméticos/uso terapéutico , Misonidazol/análogos & derivados , Nitroimidazoles/toxicidad , Fármacos Sensibilizantes a Radiaciones/toxicidad , Vómitos/inducido químicamente , Animales , Perros , Evaluación de Medicamentos , Imidazoles/uso terapéutico , Ratones , Misonidazol/sangre , Misonidazol/farmacocinética , Misonidazol/toxicidad , Nitroimidazoles/sangre , Nitroimidazoles/farmacocinética , Ondansetrón , Fármacos Sensibilizantes a Radiaciones/farmacocinética , Vómitos/prevención & control
18.
Cancer Res ; 51(17): 4581-7, 1991 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-1873802

RESUMEN

Carmethizole, a novel bis-carbamate alkylating agent, was evaluated in vitro for potential mechanisms of interaction with DNA and in vivo for spectrum and degree of antitumor activity. In vitro, the concentration of carmethizole required to produce a 50% reduction in clonogenic cell survival was identical in O6-alkylguanine DNA alkyltransferase-positive and -negative human cell lines. The CHO cell line UV4, hypersensitive to mono- and bifunctional alkylating agents, was 37-fold more sensitive to carmethizole than normal cells. The UV5 cell line, which is not hypersensitive to cross-linkers, was 13-fold more sensitive to carmethizole than normal cells. Alkaline elution studies in L1210 cells exposed to carmethizole showed the presence of DNA-protein and DNA-DNA cross-links but not DNA strand breaks. These data suggested that the interaction of carmethizole with DNA produces monoadducts, DNA-protein, and DNA-DNA interstrand cross-links at several sites. In vivo, carmethizole was not cross-resistant with 1,3-bis(2-chloroethyl)-1-nitrosourea or Cytoxan as determined by testing against P388 leukemias resistant to the latter 2 agents. Carmethizole activity was similar to that of melphalan across the murine solid tumor panel, which consisted of B16 melanoma; colon adenocarcinomas 11a, 26, and 36; and the KHT sarcoma. Carmethizole, Cytoxan, and melphalan were all active and had comparable activity against the HCT-8 and MX-1 human tumor xenografts. The in vivo spectrum of activity and efficacy of carmethizole was similar to that of melphalan.


Asunto(s)
Antineoplásicos/farmacología , ADN de Neoplasias/efectos de los fármacos , Imidazoles/farmacología , Leucemia L1210/tratamiento farmacológico , Leucemia P388/tratamiento farmacológico , Animales , Carmustina/farmacología , Reparación del ADN , Ensayos de Selección de Medicamentos Antitumorales , Leucemia L1210/genética , Leucemia P388/genética , Melfalán/farmacología , Células Tumorales Cultivadas/efectos de los fármacos
19.
Invest New Drugs ; 9(1): 1-7, 1991 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-2026477

RESUMEN

The antitumor activity of [SP-4-3-(R)]-[1,1-cyclobutanedicarboxylato-(2-)](2 methyl-1,4-butanediamine-N,N')platinum (CI-973) was characterized in a number of preclinical model systems. CI-973 retained substantial activity in cisplatin resistant murine leukemia cell lines, in vitro and in vivo; in L1210 leukemia resistant to cisplatin in vivo, CI-973 retained as much activity as was found in animals bearing sensitive L1210 leukemia. When compared in five solid murine tumors in vivo, both CI-973 and cisplatin were approximately equivalent in activity. In one human colon tumor and one human non-small cell lung carcinoma tested as xenografts in immunodeficient mice, cisplatin and CI-973 were ineffective. In two other human non-small cell lung carcinomas tested in the same fashion, cisplatin did possess activity. CI-973 has entered phase I clinical trials.


Asunto(s)
Antineoplásicos/farmacología , Carboplatino/análogos & derivados , Animales , Carboplatino/farmacología , División Celular/efectos de los fármacos , Cisplatino/farmacología , Resistencia a Medicamentos , Ensayos de Selección de Medicamentos Antitumorales , Humanos , Leucemia Experimental/tratamiento farmacológico , Leucemia Experimental/patología , Ratones , Ratones Desnudos , Neoplasias Experimentales/tratamiento farmacológico , Neoplasias Experimentales/patología , Trasplante Heterólogo , Células Tumorales Cultivadas
20.
Cancer Res ; 50(11): 3239-44, 1990 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-2334919

RESUMEN

Ethyl 5-amino-1,2-dihydro-2-methyl-3-phenylpyrido[3,4-b]pyrazin- 7-ylcarbamate, 2-hydroxyethanesulfonate, hydrate (NSC 370147) was evaluated for antitumor activity against a spectrum of tumor systems in culture and in mice. NSC 370147 was cytotoxic to a variety of mouse and human cell lines at nanomolar concentrations. The compound exhibited good in vivo antitumor activity against several murine tumors (P388 and L1210 leukemia, colon 11/A and 36, mammary 16/C, and M5076 sarcoma). Activity was largely independent of route of administration but favored a prolonged treatment schedule. NSC 370147 was as active against murine leukemia sublines resistant to Adriamycin, amsacrine, vincristine, melphalan, cisplatin, methotrexate, and CI-920 (a topoisomerase II inhibitor) as against the corresponding parental lines. Only the 1-beta-D-arabinofuranosylcytosine-resistant P388 subline exhibited any cross-resistance to NSC 370147. NSC 370147 has a spectrum of activity similar to that of vincristine and, unlike vincristine, is active against multidrug-resistant cell lines. Therefore, NSC 370147 is a candidate for clinical trial because of its favorable activity compared to vincristine, its effectiveness against multidrug-resistant cells, and its retention of activity for p.o. administration.


Asunto(s)
Neoplasias/tratamiento farmacológico , Pirazinas/farmacología , Animales , Antineoplásicos/uso terapéutico , Esquema de Medicación , Resistencia a Medicamentos , Ensayos de Selección de Medicamentos Antitumorales , Humanos , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos DBA , Pirazinas/administración & dosificación , Células Tumorales Cultivadas/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...