Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 72
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
J Thromb Haemost ; 16(9): 1873-1886, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-29956444

RESUMEN

Essentials Endothelial activation initiates multiple processes, including hemostasis and inflammation. The molecules that contribute to these processes are co-stored in secretory granules. How can the cells control release of granule content to allow differentiated responses? Selected agonists recruit an exocytosis-linked actin ring to boost release of a subset of cargo. SUMMARY: Background Endothelial cells harbor specialized storage organelles, Weibel-Palade bodies (WPBs). Exocytosis of WPB content into the vascular lumen initiates primary hemostasis, mediated by von Willebrand factor (VWF), and inflammation, mediated by several proteins including P-selectin. During full fusion, secretion of this large hemostatic protein and smaller pro-inflammatory proteins are thought to be inextricably linked. Objective To determine if secretagogue-dependent differential release of WPB cargo occurs, and whether this is mediated by the formation of an actomyosin ring during exocytosis. Methods We used VWF string analysis, leukocyte rolling assays, ELISA, spinning disk confocal microscopy, high-throughput confocal microscopy and inhibitor and siRNA treatments to demonstrate the existence of cellular machinery that allows differential release of WPB cargo proteins. Results Inhibition of the actomyosin ring differentially effects two processes regulated by WPB exocytosis; it perturbs VWF string formation but has no effect on leukocyte rolling. The efficiency of ring recruitment correlates with VWF release; the ratio of release of VWF to small cargoes decreases when ring recruitment is inhibited. The recruitment of the actin ring is time dependent (fusion events occurring directly after stimulation are less likely to initiate hemostasis than later events) and is activated by protein kinase C (PKC) isoforms. Conclusions Secretagogues differentially recruit the actomyosin ring, thus demonstrating one mechanism by which the prothrombotic effect of endothelial activation can be modulated. This potentially limits thrombosis whilst permitting a normal inflammatory response. These results have implications for the assessment of WPB fusion, cargo-content release and the treatment of patients with von Willebrand disease.


Asunto(s)
Actomiosina/fisiología , Células Endoteliales/metabolismo , Exocitosis/efectos de los fármacos , Hemostasis/fisiología , Inflamación/fisiopatología , Cuerpos de Weibel-Palade/metabolismo , 1-Metil-3-Isobutilxantina/farmacología , Actomiosina/antagonistas & inhibidores , Actomiosina/química , Citocalasinas/farmacología , Células Endoteliales/efectos de los fármacos , Epinefrina/farmacología , Compuestos Heterocíclicos de 4 o más Anillos/farmacología , Histamina/farmacología , Células Endoteliales de la Vena Umbilical Humana , Humanos , Rodamiento de Leucocito/fisiología , Selectina-P/genética , Selectina-P/fisiología , Conformación Proteica , Interferencia de ARN , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/farmacología , Acetato de Tetradecanoilforbol/farmacología , Cuerpos de Weibel-Palade/efectos de los fármacos , Factor de von Willebrand/fisiología
2.
Sci Rep ; 7(1): 9249, 2017 08 23.
Artículo en Inglés | MEDLINE | ID: mdl-28835688

RESUMEN

Levels of active Rac1 at epithelial junctions are partially modulated via interaction with Ajuba, an actin binding and scaffolding protein. Here we demonstrate that Ajuba interacts with the Cdc42 GTPase activating protein CdGAP, a GAP for Rac1 and Cdc42, at cell-cell contacts. CdGAP recruitment to junctions does not require Ajuba; rather Ajuba seems to control CdGAP residence at sites of cell-cell adhesion. CdGAP expression potently perturbs junctions and Ajuba binding inhibits CdGAP activity. Ajuba interacts with Rac1 and CdGAP via distinct domains and can potentially bring them in close proximity at junctions to facilitate activity regulation. Functionally, CdGAP-Ajuba interaction maintains junctional integrity in homeostasis and diseases: (i) gain-of-function CdGAP mutants found in Adams-Oliver Syndrome patients strongly destabilize cell-cell contacts and (ii) CdGAP mRNA levels are inversely correlated with E-cadherin protein expression in different cancers. We present conceptual insights on how Ajuba can integrate CdGAP binding and inactivation with the spatio-temporal regulation of Rac1 activity at junctions. Ajuba provides a novel mechanism due to its ability to bind to CdGAP and Rac1 via distinct domains and influence the activation status of both proteins. This functional interplay may contribute towards conserving the epithelial tissue architecture at steady-state and in different pathologies.


Asunto(s)
Comunicación Celular , Epitelio/metabolismo , Proteínas Activadoras de GTPasa/antagonistas & inhibidores , Proteínas con Dominio LIM/metabolismo , Secuencia de Aminoácidos , Animales , Línea Celular , Técnica del Anticuerpo Fluorescente , Proteínas Activadoras de GTPasa/química , Proteínas Activadoras de GTPasa/metabolismo , Humanos , Uniones Intercelulares/metabolismo , Queratinocitos/metabolismo , Proteínas con Dominio LIM/química , Ratones , Modelos Biológicos , Unión Proteica , Dominios y Motivos de Interacción de Proteínas , Transporte de Proteínas
3.
Cancer Res ; 48(16): 4529-34, 1988 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-3396005

RESUMEN

Experiments were designed to measure the effect of folic acid deficiency on a major determinant of cancer lethality, the propensity to form metastases. Murine B16 melanoma cells (F10 strain) were grown in folate-deficient and -supplemented media. After 3 days, cells in the deficient medium had restricted proliferative capacity, low folate levels by bioassay, increased cell volume, abnormal deoxyuridine suppression tests, accumulation of cells in S phase by flow cytometry, and increased numbers of DNA strand breaks. These folate-deficient cells consistently initiated more pulmonary metastases than control cells when injected into host mice. Cell size did not appear to be a major factor in pulmonary metastasis formation. In vitro growth rates and cloning efficiencies were comparable for cells in both types of medium as was subcutaneous growth of tumors. We conclude that folate deficiency increases the metastatic potential of cultured melanoma cells.


Asunto(s)
Deficiencia de Ácido Fólico/patología , Metástasis de la Neoplasia/patología , Animales , Ciclo Celular , Aberraciones Cromosómicas , ADN/biosíntesis , Daño del ADN , Melanoma Experimental/patología , Ratones
4.
Cancer Res ; 45(10): 4748-53, 1985 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-4040806

RESUMEN

Dichloro-1,2-diaminocyclohexane (DACH):platinum(II), the prototype DACH:platinum complex, had good antitumor activity, was not cross-resistant with cis-dichlorodiammineplatinum(II) (DDP), but was, unfortunately, virtually insoluble in water and was, therefore, not evaluated clinically. This paper summarizes some of the chemical and biological attributes of a series of cis-bisascorbato-DACH:platinum(II) complexes (DAP). Although the primary emphasis has been placed on the DAP complex consisting of the isomeric mixture DACH, a series of complexes using the isomers of either DACH or ascorbic acid have also been synthesized. The synthetic procedure entailed reacting the water-soluble sulfato-DACH:platinum(II) with barium ascorbate, and the water-soluble product DAP was removed from the BaSO4 precipitate by filtration. Based upon elemental analysis, all the complexes had stoichiometric composition of one DACH:one platinum and two ascorbate monoanions. High-pressure liquid chromatography of cis-bisascorbato (mixed-isomer DACH):platinum revealed a series of platinum-containing, ultraviolet-absorbing peaks. All the DAP complexes had significant in vitro cytotoxicity against L1210 leukemia cells (L1210/0) with 50%-inhibitory dose values ranging from 2 to 5 micrograms/ml. None of the complexes was cross-resistant with DDP when tested in vitro against L1210 cells 50-fold resistant to DDP (L1210/DDP). The cis-bisascorbato (mixed-isomer DACH):platinum (DAP-1) was administered i.p. to C57BL X DBA/2 F1 mice inoculated i.p. with 10(6) L1210/0 cells. When administered on Days 1, 5, and 9, the DAP-1 complex consistently produced treated:control values in excess of 200% with several long-term survivors (alive 60 days after tumor inoculation). Further, the DAP-1 complex was totally non-cross-resistant with DDP when tested in vivo against a DDP-resistant L1210 line. Toxicological investigations revealed that DAP-1 was relatively nonnephrotoxic but did cause the expected bone marrow and gastrointestinal toxicity. In summary, the DAP complexes are highly water-soluble, nonnephrotoxic platinum complexes with sufficient antitumor activity to warrant further pharmacological, biochemical, and chemical investigations.


Asunto(s)
Antineoplásicos/farmacología , Compuestos Organoplatinos/farmacología , Animales , Antineoplásicos/toxicidad , Fenómenos Químicos , Química , Cisplatino/farmacología , Resistencia a Medicamentos , Masculino , Ratones , Ratones Endogámicos , Compuestos Organoplatinos/toxicidad , Solubilidad
5.
Cancer Res ; 46(12 Pt 1): 6250-4, 1986 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-3779645

RESUMEN

A series of water-soluble N-substituted iminodiacetato(1,2-diaminocyclohexane)-platinum(II) complexes (IDP) were synthesized and tested for chemical stability, antitumor activity, and toxicity. The results obtained suggest that these complexes are relatively stable for more than 48 h when dissolved in water or phosphate buffer. All complexes had good in vitro cytotoxicity and were not cross-resistant with cis-dichloro-diammineplatinum(II) (DDP) in a DDP-resistant cell line in vitro. When the complexes were administered as a single i.p. injection to C57BL/6 X DBA/2F1 (hereafter called B6D2F1) mice inoculated with L1210 leukemia cells, a significant increase in mean survival time was observed, but there were few long-term survivors. When the complexes were administered on Days 1, 5, and 9 after tumor inoculation, however, cure rates of 50 to 85% were obtained. The oncolytic activity of the IDP complexes against L1210 ascites appeared much greater than that of DDP. The IDP complexes also had good antitumor activity when administered i.p. on Days 1, 5, and 9 following i.p. inoculation of B16 melanoma to B6D2F1 mice. Five of the six IDP complexes had no significant nephrotoxicity (as evidenced by lack of elevated blood urea nitrogen levels). N-Benzyl-iminodiacetato(1,2-diaminocyclohexane)-platinum(II) resolved into three distinct peaks of UV-absorbing material that corresponded with three distinct peaks of platinum-containing material. The exact chemical identity of the active component of this mixture is currently under investigation. The results obtained to date, however, suggest that the N-substituted iminodiacetato(1,2-diaminocyclohexane)-platinum(II) complexes are good candidates for further developmental studies.


Asunto(s)
Antineoplásicos/farmacología , Compuestos Organoplatinos/farmacología , Animales , Antineoplásicos/toxicidad , Fenómenos Químicos , Química , Cisplatino/farmacología , Dosificación Letal Mediana , Leucemia L1210/patología , Masculino , Ratones , Ratones Endogámicos , Solubilidad , Relación Estructura-Actividad
6.
Cancer Res ; 48(17): 5029-35, 1988 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-2970294

RESUMEN

Trimetrexate (TMQ; NSC 352122) is a potent inhibitor of dihydrofolate reductase with good activity against murine i.p.-implanted B16 melanoma and colon 26 tumors. Preclinical antineoplastic activity, demonstrated schedule dependency, and data suggesting effectiveness against methotrexate-resistant cells prompted a Phase I clinical and pharmacokinetic study of trimetrexate using an i.v. daily x5 schedule. Forty-three good performance status patients were treated with 12 dose levels using daily doses varying from 0.5 to 15 mg/m2/d. Plasma and urine samples were obtained for pharmacokinetic analysis using a high-performance liquid chromatographic method. Myelosuppression was dose limiting and 15 mg/m2/d x5 was the maximum tolerated dose. White blood cell (WBC) and platelet toxicity were noted at doses of 1.6 mg/m2 and above. Median WBC and platelet nadirs occurred on approximately Days 11-12 with recovery by Days 15-18. Nonhematological toxicity included mucositis, nausea and vomiting, stomatitis, diarrhea, and rash. Evidence for antitumor activity was seen in seven patients. Trimetrexate elimination from plasma could be represented as either a bi- or triexponential process. Terminal elimination half-lives were in the range of 5-14 h in patients represented by a triexponential model. Approximately 10-20% of the dose administered was excreted in urine over a 24-h period. The recommended starting dose for patients in Phase II trials using the d x5 i.v. schedule is 8.0 mg/m2/d repeated every 21 days. Dose escalations may be possible depending on the extent of prior therapy and individual tolerance of the drug.


Asunto(s)
Antineoplásicos/farmacocinética , Neoplasias/tratamiento farmacológico , Quinazolinas/farmacocinética , Adolescente , Adulto , Anciano , Antineoplásicos/administración & dosificación , Antineoplásicos/efectos adversos , Médula Ósea/efectos de los fármacos , Evaluación de Medicamentos , Femenino , Humanos , Masculino , Persona de Mediana Edad , Mucosa Bucal/efectos de los fármacos , Quinazolinas/administración & dosificación , Quinazolinas/efectos adversos , Piel/efectos de los fármacos , Trimetrexato
7.
J Med Chem ; 28(8): 1124-6, 1985 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-4020835

RESUMEN

Several 5,8-bis[(aminoalkyl)amino]-1-azaanthracene-9,10-diones have been synthesized and evaluated for antitumor activity against L1210 leukemia both in vitro and in vivo. Comparisons are made to the corresponding carbocyclic analogues. One of the aza analogues showed modest in vivo activity.


Asunto(s)
Antracenos/farmacología , Antineoplásicos/síntesis química , Animales , Antracenos/síntesis química , División Celular/efectos de los fármacos , Leucemia L1210/tratamiento farmacológico , Ratones , Relación Estructura-Actividad
8.
J Med Chem ; 29(8): 1374-80, 1986 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-3735306

RESUMEN

One of the problems encountered in the use of tetrahydrouridine (THU, 2) and saturated 2-oxo-1,3-diazepine nucleosides as orally administered cytidine deaminase (CDA) inhibitors is their acid instability. Under acid conditions these compounds are rapidly converted into inactive ribopyranoside forms. A solution this problem was sought by functionalizing the acid-stable but less potent CDA inhibitor 1-beta-D-ribofuranosyl-2(1H)-pyrimidinone (1) with the hope of increasing its potency to the level achieved with THU. The selection of the hydroxymethyl substituent at C-4, which led to the synthesis of 4-(hydroxymethyl)-1-beta-D-ribofuranosyl-2(1H)-pyrimidinone (10), 3,4-dihydro-4-(hydroxymethyl)-1-beta-D-ribofuranosyl-2(1H)-pyrimidinone (7), and 3,4,5,6-tetrahydro-4-(dihydroxymethyl)-1-beta-D-ribofuranosyl-2(1H)-p yrimidinone (28) was based on the transition-state (TS) concept. The key intermediate precursor, 4-[(benzoyloxy)methyl]-1-(2,3,5-tri-O-benzoyl-beta-D-ribofuranosyl)-2(H) -pyrimidinone (24), was obtained via the classical Hilbert-Johnson reaction between 2-methoxy-4-[(benzoyloxy)methyl]pyrimidine (20) and 2,3,5-tri-O-benzoyl-1-D-ribofuranosyl bromide (21). Deprotection of 24 afforded compound 10, while its sodium borohydride reduction products afforded compounds 7 and 28 after removal of the blocking groups. Syntheses of 3,4-dihydro-1-beta-D-ribofuranosyl-2(1H)-pyrimidinone (9) and 3,6-dihydro-1-beta-D-ribofuranosyl-2(1H)-pyrimidinone (8), which lack the hydroxymethyl substituent, was accomplished in a similar fashion. The new compounds bearing the hydroxymethyl substituent were more acid stable than THU, and their CDA inhibitory potency, expressed in terms of Ki values, spanned from 10(-4) to 10(-7) M in a manner consistent with the TS theory. Compound 7, in particular, was superior to its parent 1 and equipotent to THU (Ki = 4 X 10(-7) M) when examined against mouse kidney CDA. The superior acid stability of this compound coupled to its potent inhibitory properties against CDA should provide a means of testing oral combinations of rapidly deaminated drugs, viz. ara-C, without the complications associated with the acid instability of THU.


Asunto(s)
Citidina Desaminasa/antagonistas & inhibidores , Nucleósido Desaminasas/antagonistas & inhibidores , Nucleósidos de Pirimidina/síntesis química , Pirimidinonas/síntesis química , Inhibidores de la Adenosina Desaminasa , Animales , Riñón/enzimología , Espectroscopía de Resonancia Magnética , Ratones , Espectrofotometría Ultravioleta , Relación Estructura-Actividad , Tetrahidrouridina/farmacología
9.
J Med Chem ; 29(11): 2351-8, 1986 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-3783592

RESUMEN

Tetrahydrouridine (THU, 2) and other fully reduced cyclic urea ribofuranosyl nucleosides undergo a rapid, acid-catalyzed isomerization to their more stable ribopyranosyl form. This isomerization is characterized by a change in spectral properties and by a greater than 10-fold decrease in potency for those nucleosides that act as potent inhibitors of cytidine deaminase in their ribofuranose form. 1-(beta-D-Ribopyranosyl)hexahydropyrimidin-2-one (7) was synthesized and used in conjunction with its furanose isomer 6 as a model compound for more extensive 1H and 13C NMR, mass spectral, and kinetic studies of this isomerization. The 0.4 delta upfield shift and 4-Hz increase in the J1',2' coupling constant for the pyranose anomeric proton in the 1H NMR spectrum is indicative of a pyranose beta-CI conformation in which the aglycon and C-2' and C-4' hydroxyls are equatorial. The mass spectra of trimethylsilylated pyranose nucleosides also show a characteristic large shift in the m/z 204-217 abundance and the appearance of two new rearrangement ions at M-133 and M-206. For furanose 6 the rate of isomerization is pH and temperature dependent with pyranose 7 predominating by a factor of 6-9 equilibrium. At pH 1 and 37 degrees C, furanose 6 has an initial half-life of less than 12 min. Accordingly, this isomerization may explain the observed lack of enhanced ara-C levels in studies evaluating the oral administration of an ara-C and THU combination to species with an acidic stomach content.


Asunto(s)
Citidina Desaminasa/antagonistas & inhibidores , Nucleósido Desaminasas/antagonistas & inhibidores , Nucleósidos/farmacología , Tetrahidrouridina/farmacología , Uridina/análogos & derivados , Citarabina/metabolismo , Cromatografía de Gases y Espectrometría de Masas , Determinación de la Acidez Gástrica , Concentración de Iones de Hidrógeno , Cinética , Espectroscopía de Resonancia Magnética
10.
J Med Chem ; 41(14): 2572-8, 1998 Jul 02.
Artículo en Inglés | MEDLINE | ID: mdl-9651161

RESUMEN

Three carbocylic analogues of the potent cytidine deaminase inhibitor (CDA) zebularine [1-(beta-D-ribofuranosyl)-1, 2-dihydropyrimidin-2-one, 1a] were synthesized. The selected pseudosugar templates correspond, respectively, to the cyclopentenyl moiety of neplanocin A (compound 4), the cyclopentyl moiety of aristeromycin (compound 5), and a newly designed, rigid bicyclo[3.1. 0]hexane moiety (compound 6). These three carba-nucleoside versions of zebularine were fashioned to overcome the inherent instability of the parent drug. Each target compound was approached differently using either convergent or linear approaches. The immediate precursor to the cyclopentenyl analogue 4 was obtained by a Mitsunobu coupling of pseudosugar 7 with 2-hydroxypyrimidine. The cyclopentyl analogue 5 was linearly constructed from carbocyclic amine 17, and the final target 6 was similarly constructed from the carbobicyclic amine 27. Of the three target compounds, only 5 showed a significant level of inhibition against human CDA, but it was 16 times less potent than zebularine (Ki = 38 microM vs Ki(apparent) = 2.3 microM). Although these carbocyclic analogues appeared to be more stable than zebularine, replacement of the electronegative CO4' oxygen for the less electronegative carbon in 4-6 presumably reduces the capacity of the pyrimidin-2(1H)-one ring to form a covalent hydrate, a step considered crucial for the compound to function as a transition-state inhibitor of the enzyme.


Asunto(s)
Citidina Desaminasa/antagonistas & inhibidores , Inhibidores Enzimáticos , Nucleósidos de Pirimidina , Animales , Citidina/análogos & derivados , Inhibidores Enzimáticos/síntesis química , Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/farmacología , Humanos , Ratones , Nucleósidos de Pirimidina/síntesis química , Nucleósidos de Pirimidina/química , Nucleósidos de Pirimidina/farmacología , Relación Estructura-Actividad
11.
J Med Chem ; 20(9): 1215-8, 1977 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-411935

RESUMEN

A number of homologous 2,4-diaminocycloalka[g]pteridines varying in ring size from 5 to 15 were prepared by (a) condensation of aminomalononitrile tosylate with alpha-oximinocycloalkanones, deoxygenation of the resulting 2-amino-3-cyanocycloalka[b]pyrazine 1-oxides, and guanidine cyclization; (b) guanidine cyclization of the above pyrazine 1-oxides to give 2,4-diaminocycloalka[g]pteridine 8-oxides, followed by deoxygenation; or (c) condensation of 2,4,5,6-tetraaminopyrimidine with a cycloalka-1,2-dione (for the cyclohepta- and cycloocta[g]pteridines only). These compounds were examined for their activity as dihydrofolate reductase inhibitors against Lactobacillus casei, rat liver, L1210, and Trypanosoma cruzi. Activity was found to depend upon ring size, with the greatest activity exhibited by the cyclododeca derivatives 31.


Asunto(s)
Antagonistas del Ácido Fólico , Pteridinas , Animales , Lacticaseibacillus casei/enzimología , Leucemia L1210/enzimología , Hígado/enzimología , Pteridinas/síntesis química , Pteridinas/farmacología , Ratas , Relación Estructura-Actividad
12.
J Med Chem ; 24(6): 662-6, 1981 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-7252974

RESUMEN

Five beta-D-ribofuranosyl cyclic urea nucleosides (14-18), ranging in ring size from five to eight membered, were synthesized and evaluated as cytidine deaminase (CDA) inhibitors. The precursor protected nucleosides (9-13) were prepared by a condensation procedure utilizing persilylated ureas with a halo sugar under the specific catalytic activity of a HgO/HgBr2 mixture which provided exclusively the beta-anomers. Catalytic hydrogenation of known 1-(2,3,5-tri-O-benzoyl-beta-ribofuranosyl)-1,2-dihydropyrimidin-2-one (19) afforded nucleoside 10 identical with that obtained by the mercury-catalyzed condensation procedure. CDA activity varies significantly with the ring size of the urea aglycon the reaches its maximum level for the seven-membered analogues 16 and 17. The unexpected high potency of nucleoside 17 (Ki = 2.5 X 10(-8) M, human liver enzyme) is reported. This compound represents the most potent inhibitor of human liver CDA yet discovered.


Asunto(s)
Azepinas/farmacología , Citidina Desaminasa/antagonistas & inhibidores , Nucleósido Desaminasas/antagonistas & inhibidores , Ribonucleósidos/farmacología , Urea/análogos & derivados , Animales , Azepinas/síntesis química , Humanos , Técnicas In Vitro , Riñón/enzimología , Hígado/enzimología , Ratones , Ribonucleósidos/síntesis química , Relación Estructura-Actividad , Urea/síntesis química , Urea/farmacología
13.
J Med Chem ; 36(21): 3103-12, 1993 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-8230096

RESUMEN

A series of eight previously undescribed 2,4-diaminothieno[2,3-d]pyrimidine analogues of the potent dihydrofolate reductase (DHFR) inhibitors trimetrexate (TMQ) and piritrexim (PTX) were synthesized as potential drugs against Pneumocystis carinii and Toxoplasma gondii, which are major causes of severe opportunistic infections in AIDS patients. 2,4-Diamino-5-methyl-6-(aryl/aralkyl)thieno[2,3-d]pyrimidines with 3,4,5-trimethoxy or 2,5-dimethoxy substitution in the aryl/aralkyl moiety and 2,4-diamino-5-(aryl/aralkyl)thieno[2,3-d]pyrimidines with 2,5-dimethoxy substitution in the aryl/aralkyl moiety were obtained by reaction of the corresponding 2-amino-3-cyanothiophenes with chloroformamidine hydrochloride. The aryl group in the 5,6-disubstituted analogues was either attached directly to the hetero ring or was separated from it by one or two carbons, whereas the aryl group in the 5-monosubstituted analogues was separated from the hetero ring by two or three carbons. 2-Amino-3-cyano-5-methyl-6-(aryl/alkyl)thiophene intermediates for the preparation of the 5,6-disubstituted analogues were prepared from omega-aryl-2-alkylidene-malononitriles and sulfur in the presence of a secondary amine, and 2-amino-3-cyano-4-(aryl/aralkyl)thiophene intermediates for the preparation of the 5-monosubstituted analogues were obtained from omega-aryl-1-chloro-2-alkylidenemalononitriles and sodium hydrosulfide. Synthetic routes to the heretofore unknown ylidenemalononitriles, and the ketone precursors thereof, were developed. The final products were tested in vitro as inhibitors of DHFR from Pneumocystis carinii, Toxoplasma gondii, rat liver, beef liver, and Lactobacillus casei. A selected number of previously known 2,4-diaminothieno[2,3-d]pyrimidines lacking the 3,4,5-trimethoxyphenyl and 2,5-dimethoxyphenyl substitution pattern of TMQ and PTX, respectively, were also tested for comparison. None of the compounds was as potent as TMQ or PTX, and while some of them showed some selectivity in their binding to Pneumocystis carinii and Toxoplasma gondii versus rat liver DHFR, this effect was not deemed large enough to warrant further preclinical evaluation.


Asunto(s)
Antineoplásicos/síntesis química , Antagonistas del Ácido Fólico , Pneumocystis/enzimología , Pirimidinas/síntesis química , Pirimidinas/farmacología , Toxoplasma/enzimología , Trimetrexato/análogos & derivados , Animales , Antineoplásicos/química , Antineoplásicos/farmacología , Humanos , Hígado/efectos de los fármacos , Hígado/enzimología , Pneumocystis/efectos de los fármacos , Pneumocystis/metabolismo , Pirimidinas/química , Ratas , Relación Estructura-Actividad , Toxoplasma/efectos de los fármacos , Toxoplasma/metabolismo , Trimetrexato/farmacología
14.
J Med Chem ; 29(8): 1370-3, 1986 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-3735305

RESUMEN

A number of unsymmetrically substituted 1,4-bis[(aminoalkyl)amino]anthracene-9,10-diones have been synthesized and evaluated for their antitumor activity against L1210 in vitro and in vivo. The high activity of several compounds observed in vitro was not paralleled by comparable activity in vivo. The activities of the substituted 1,4-bis[(aminoalkyl)amino]anthracene-9,10-diones as inhibitors of cell growth were generally much higher than those of the related 1-[(aminoalkyl)amino]-4-methoxyanthracene-9,10-diones, and this correlated with the relative abilities of compounds of the two types to interact with calf thymus DNA.


Asunto(s)
Antracenos/síntesis química , Antineoplásicos/uso terapéutico , Animales , Antineoplásicos/síntesis química , Bovinos , División Celular/efectos de los fármacos , ADN/metabolismo , Leucemia L1210/tratamiento farmacológico , Ratones
15.
J Med Chem ; 25(8): 952-6, 1982 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-7120284

RESUMEN

A new type of antitumor platinum complex has been prepared and examined for antitumor activity against L1210 leukemia both in vitro and in vivo. The coordination environment of platinum in these complexes consists of three anionic chloride ions and a positively charged amine. The positive charge is introduced by monoprotonation or monoalkylation of a diamine. Platinum(IV) derivatives have been prepared for several of the complexes, and a water-soluble sulfate derivative has been prepared for one of them. Several of these complexes exhibit significant in vitro activity, and trichloro(3-aminoquinuclidinium)platinum(II) (QTP) exhibits significant in vivo activity as well. An increase in life span of approximately 40% has been observed using QTP. QTP is toxic at doses slightly in excess of effective doses.


Asunto(s)
Antineoplásicos/síntesis química , Compuestos Organoplatinos/síntesis química , Animales , Línea Celular , Fenómenos Químicos , Química , Leucemia L1210/tratamiento farmacológico , Masculino , Ratones , Compuestos Organoplatinos/farmacología
16.
J Med Chem ; 41(13): 2184-93, 1998 Jun 18.
Artículo en Inglés | MEDLINE | ID: mdl-9632350

RESUMEN

Select chemokine receptors act as coreceptors for HIV-1 entry into human cells and represent targets for antiviral therapy. In this report we describe a distamycin analogue, 2,2'-[4, 4'-[[aminocarbonyl]amino]bis[N,4'-di[pryrrole-2-carboxamide- 1, 1'-dimethyl]]-6,8-naphthalenedisulfonic acid]hexasodium salt (NSC 651016), that selectively inhibited chemokine binding to CCR5, CCR3, CCR1, and CXCR4, but not to CXCR2 or CCR2b, and blocked chemokine-induced calcium flux. Inhibition was not due to nonspecific charge interactions at the cell surface, but was based on a specific competition for the ligand receptor interaction sites since the inhibitory effect was specific for some but not all chemoattractant receptors. NSC 651016 inhibited in vitro replication of a wide range of HIV-1 isolates, as well as HIV-2 and SIV, and exhibited in vivo anti-HIV-1 activity in a murine model. In contrast, a distamycin analogue with similar structure and charge and the monomeric form of NSC 651016 demonstrated no inhibitory effects. These data demonstrate that molecules which interfere with HIV-1 entry into cells by targeting specific chemokine coreceptors can provide a viable approach to anti-HIV-1 therapy. NSC 651016 represents an attractive candidate for the chemotherapeutic treatment of HIV-1 infection and as a microbicide to prevent the sexual transmisssion of HIV-1. Moreover, NSC 651016 can serve as a template for medicinal chemical modifications leading to more effective antivirals.


Asunto(s)
Fármacos Anti-VIH/farmacología , VIH-1/efectos de los fármacos , Naftalenosulfonatos/farmacología , Receptores de Quimiocina/antagonistas & inhibidores , Replicación Viral/efectos de los fármacos , Administración Cutánea , Animales , Fármacos Anti-VIH/administración & dosificación , Fármacos Anti-VIH/química , Fármacos Anti-VIH/farmacocinética , Anticuerpos Monoclonales/inmunología , Anticuerpos Monoclonales/metabolismo , Unión Competitiva , Calcio/metabolismo , Línea Celular , Dimerización , Farmacorresistencia Microbiana , VIH-1/aislamiento & purificación , VIH-1/metabolismo , VIH-1/fisiología , VIH-2/efectos de los fármacos , VIH-2/aislamiento & purificación , Humanos , Inyecciones Intraperitoneales , Inyecciones Intravenosas , Ligandos , Fusión de Membrana/efectos de los fármacos , Ratones , Monocitos/efectos de los fármacos , Monocitos/inmunología , Monocitos/metabolismo , Naftalenosulfonatos/administración & dosificación , Naftalenosulfonatos/química , Naftalenosulfonatos/farmacocinética , Receptores CXCR4/antagonistas & inhibidores , Receptores CXCR4/inmunología , Inhibidores de la Transcriptasa Inversa/farmacología , Transducción de Señal/efectos de los fármacos , Virus de la Inmunodeficiencia de los Simios/efectos de los fármacos , Virus de la Inmunodeficiencia de los Simios/aislamiento & purificación
17.
Cancer Lett ; 29(1): 65-71, 1985 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-2933139

RESUMEN

The metabolism and biliary excretion of trimetrexate (TMTX), a lipid soluble antifolate, were examined using a recirculating isolated perfused rat liver system. Elimination of TMTX into perfusate was biphasic and dose-independent, with distribution and elimination half-lives of 2 and 13 min. Two metabolites, M1 and M2, both known to inhibit dihydrofolate reductase activity, were present in perfusate only in small concentrations. However, of the total TMTX dose, approximately 50% was excreted in bile as M1, and 20% as M2. Up to 75% of the total dose was accounted for as TMTX, M1, or M2 in perfusate and bile.


Asunto(s)
Bilis/metabolismo , Antagonistas del Ácido Fólico/metabolismo , Hígado/metabolismo , Quinazolinas/metabolismo , Animales , Cromatografía Líquida de Alta Presión , Semivida , Cinética , Masculino , Perfusión , Ratas , Ratas Endogámicas , Trimetrexato
18.
Biochem Pharmacol ; 37(23): 4557-64, 1988 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-3264501

RESUMEN

Chloroquinoxaline sulfonamide (CQS), a chlorinated derivative of sulfaquinoxaline (SQ), inhibited proliferation of murine B16 melanoma cells, but only when relatively high drug concentrations (1 mM) were used. The inhibition of cell growth by CQS was at least partially reversible by incubation in drug-free medium. Incubation of melanoma cells with CQS was associated with an arrest of the cell cycle in G0/G1 as measured by flow cytometry. The drug slightly decreased uptake of radiolabeled deoxyuridine and thymidine after 24- and 48-hr incubation periods but increased nucleoside incorporation at 72 hr. No evidence of intercalation with DNA was found. Because SQ previously was reported to inhibit an aspect of folate metabolism, we investigated the possibility that CQS limits tumor cell growth by altering folate homeostasis. This appears unlikely, however, in view of the following observations: (1) the cytotoxic effects of CQS could not be reversed by folinic acid; (2) deoxyuridine suppression of thymidine incorporation was not affected by CQS treatment; (3) CQS did not inhibit dihydrofolate reductase from mammalian or bacterial sources; and (4) CQS toxicity in mice was not reduced by folinic acid. Experiments performed with analogues modified in the quinoxaline and para-amino phenyl functions indicated that tumor cell inhibition did not require preservation of the conventional sulfonamide structure.


Asunto(s)
Melanoma/metabolismo , Quinoxalinas/farmacología , Sulfanilamidas/farmacología , Animales , División Celular/efectos de los fármacos , Línea Celular/efectos de los fármacos , Cromatografía Líquida de Alta Presión , ADN/metabolismo , Desoxiuridina/farmacocinética , Citometría de Flujo , Leucovorina/farmacología , Ratones , Sulfaquinoxalina/farmacología , Tetrahidrofolato Deshidrogenasa/metabolismo , Timidina/farmacocinética
19.
Biochem Pharmacol ; 38(20): 3521-6, 1989 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-2818644

RESUMEN

Chloroquinoxaline sulfonamide (CQS), a sulfanilamide derivative with antitumor activity, was found to be toxic to lymphoid tissue during preclinical studies. The mechanism of this toxicity appears to involve profound inhibition of lymphocyte activation. Incubation of human peripheral blood mononuclear cells (PBMNCs) with CQS decreased cellular incorporation of thymidine and deoxyuridine in a dose-dependent manner. Analysis of cell cycle distribution by flow cytometry indicated that CQS blocked movement out of the G0/G1 phase. Drug-treated cells were smaller and expressed fewer receptors for interleukin-2 (IL-2) and transferrin than untreated mitogen-stimulated lymphocytes. These observations support the notion that CQS has cell cycle specificity in regulating lymphocyte proliferation. As little as 10 microM CQS markedly inhibited both human lymphocyte and murine CTLL cell replication in response to IL-2 containing growth factors. However, CQS did not block secretion of IL-2 into culture supernatant fractions by human PBMNCs. Finally, CQS inhibited in vitro production of immunoglobulins G and M by mitogen-stimulated lymphocytes, primarily by causing cytotoxicity. In all of these drug effects, CQS was approximately one to two logs more potent than the parent compound, sulfaquinoxaline (SQ). These studies indicate that CQS inhibits essential basic processes in human lymphocytes. This agent may find use as an immunosuppressive drug.


Asunto(s)
Inmunosupresores/farmacología , Quinoxalinas/farmacología , Sulfanilamidas/farmacología , Animales , Femenino , Humanos , Inmunoglobulinas/biosíntesis , Interleucina-2/biosíntesis , Interleucina-2/farmacología , Interfase/efectos de los fármacos , Activación de Linfocitos/efectos de los fármacos , Ratas , Ratas Endogámicas , Receptores de Interleucina-2/análisis , Sulfaquinoxalina/farmacología
20.
Biochem Pharmacol ; 36(10): 1605-12, 1987 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-2954554

RESUMEN

Trimetrexate is a lipid soluble dihydrofolate reductase inhibitor which, unlike methotrexate, does not depend upon the membrane folate transport system for cell entry. We investigated the possibility that trimetrexate (but not methotrexate) might permeate intermitotic lymphocytes and, following stimulation, impair only the responding cells, rather than all dividing cells, as is the case with methotrexate. Peripheral blood mononuclear cells from normal individuals were incubated for 1 hr in three moderate to high concentrations (1, 10 and 100 microM) of methotrexate or trimetrexate, washed, and incubated with phytohemagglutinin. Intracellular folate activity, as assessed by the deoxyuridine suppression test, was abnormal at all three concentrations of trimetrexate but only at the highest concentration of methotrexate. Similarly, incorporation of [3H]deoxyuridine was depressed profoundly in trimetrexate-treated cells (2% of control) but unaffected by methotrexate. Analysis of cell cycle distribution by flow cytometry confirmed G0 + G1 arrest in trimetrexate but not methotrexate-treated cells. Neither drug altered morphologic transformation, Tac antigen expression, or incorporation of [3H]thymidine by the "salvage" pathway. Therefore, brief exposure to methotrexate has little effect on intermitotic lymphocytes, whereas trimetrexate very specifically inhibits the conversion of deoxyuridine to thymidine in these cells and leads to the arrest of DNA synthesis in the G0 + G1 phase. This metabolic abnormality markedly reduces in vitro antibody synthesis: a 1-hr treatment of lymphocytes with 10 or 100 microM trimetrexate prior to incubation with pokeweed mitogen on four occasions completely inhibited both IgG and IgM secretion. Similar treatment with methotrexate had no effect until the highest concentration (100 microM) was used. We conclude that brief exposure of peripheral blood mononuclear cells to the nonclassical dihydrofolate reductase inhibitor, trimetrexate, results in inhibition of nucleic acid synthesis and impairment of antibody production. This drug effect may permit more incisive modulation of immune responses.


Asunto(s)
Formación de Anticuerpos/efectos de los fármacos , Linfocitos/metabolismo , Ácidos Nucleicos/sangre , Quinazolinas/farmacología , ADN/biosíntesis , Desoxiuridina/sangre , Humanos , Interfase/efectos de los fármacos , Linfocitos/efectos de los fármacos , Linfocitos/inmunología , Metotrexato/farmacología , Timidina/sangre , Trimetrexato
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA