Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 120
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Science ; 250(4985): 1262-6, 1990 Nov 30.
Artículo en Inglés | MEDLINE | ID: mdl-2173860

RESUMEN

The gene designated gamma 134.5 maps in the inverted repeats flanking the long unique sequence of herpes simplex virus-1 (HSV-1) DNA, and therefore it is present in two copies per genome. This gene is not essential for viral growth in cell culture. Four recombinant viruses were genetically engineered to test the function of this gene. These were (i) a virus from which both copies of the gene were deleted, (ii) a virus containing a stop codon in both copies of the gene, (iii) a virus containing after the first codon an insert encoding a 16-amino acid epitope known to react with a specific monoclonal antibody, and (iv) a virus in which the deleted sequences were restored. The viruses from which the gene was deleted or which carried stop codons were avirulent on intracerebral inoculation of mice. The virus with the gene tagged by the sequence encoding the epitope was moderately virulent, whereas the restored virus reacquired the phenotype of the parent virus. Significant amounts of virus were recovered only from brains of animals inoculated with virulent viruses. Inasmuch as the product of the gamma 134.5 gene extended the host range of the virus by enabling it to replicate and destroy brain cells, it is a viral neurovirulence factor.


Asunto(s)
Mapeo Cromosómico , Encefalitis/microbiología , Genes Virales , Herpes Simple/microbiología , Proteínas Inmediatas-Precoces , Simplexvirus/genética , Proteínas Virales/genética , Secuencia de Aminoácidos , Animales , Anticuerpos Monoclonales , Antígenos Virales/genética , Antígenos Virales/inmunología , Secuencia de Bases , Deleción Cromosómica , Codón , ADN Viral/genética , Humanos , Datos de Secuencia Molecular , Conejos , Secuencias Repetitivas de Ácidos Nucleicos , Simplexvirus/crecimiento & desarrollo , Simplexvirus/patogenicidad , Timidina Quinasa/genética , Transfección , Proteínas Reguladoras y Accesorias Virales/genética , Proteínas Reguladoras y Accesorias Virales/inmunología
2.
J Clin Invest ; 91(6): 2837-43, 1993 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-8390490

RESUMEN

Previous studies have shown that a gene mapping in the inverted repeats of the L component of herpes simplex virus, type 1 DNA, designated as gamma (1) 34.5, was dispensable for growth in cells in culture but that the deletion mutant (R3616) and a mutant containing a stop codon (R4009) in each copy of the gene were incapable of replicating in the central nervous systems (CNS) of mice. Restoration of the deleted sequences restored the wild type virus phenotype. We report here that the gamma (1) 34.5 mutant viruses (R3616 and R4009) replicated in the vaginal tract of two different strains of mice and guinea pig, although both viruses were shed at lower titer and for fewer days than the wild type and restored viruses. Both R3616 and R4009 failed to replicate or cause significant pathology in the cornea of Balb/C mice or following intranasal inoculation of Swiss Webster mice. Analyses of sensory trigeminal and dorsal root ganglia innervating the site of inoculation indicated that the incidence of establishment of latency or reactivation from latency by R3616 and R4009 viruses was significantly lower than that determined for mice infected with wild type or restored virus. Thus, selective deletion of gamma (1) 34.5 gene abolished the capacity of the virus to spread from peripheral mucosal sites to the CNS or replicate in the CNS, and diminished the capacity of the virus to replicate at mucosal sites and, subsequently, establish latency, or be able to be reactivated ex vivo. The results of our studies may have direct implications for the development of genetically engineered herpes simplex virus vaccines.


Asunto(s)
Genes Virales/genética , Simplexvirus/patogenicidad , Activación Viral/genética , Animales , Secuencia de Bases , Femenino , Ganglios Espinales/microbiología , Herpes Simple/fisiopatología , Ratones , Ratones Endogámicos BALB C , Datos de Secuencia Molecular , Cavidad Nasal/microbiología , Reacción en Cadena de la Polimerasa , Simplexvirus/genética , Simplexvirus/crecimiento & desarrollo , Nervio Trigémino/microbiología , Vagina/microbiología , Virulencia , Replicación Viral , Esparcimiento de Virus
3.
J Clin Invest ; 102(7): 1431-43, 1998 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-9769336

RESUMEN

The role of Fas- and TNF-receptor 1 (TNF-R1)-mediated apoptosis in the clearance of virally infected cells and in the regulation of the immune response was analyzed after murine cytomegalovirus (MCMV) infection of C57BL/6 (B6)-+/+ mice, Fas-mutant B6-lpr/lpr mice, TNF-R1 knockout B6-tnfr0/0 mice, and double-deficient B6-tnfr0/0 lpr/lpr mice. There was approximately equivalent clearance of MCMV in B6-+/+, B6-tnfr0/0, and B6-lpr/lpr mice, and by day 28 no infectious virus could be detected in the liver, kidney, lung, or peritoneal exudate. However, delayed virus clearance was observed in B6-tnfr0/0 lpr/lpr mice. An acute inflammatory response occurred in the liver, lung, and kidney of all mice, which was most severe 7 d after MCMV infection, but resolved by day 28 in B6-+/+ and B6-tnfr0/0 mice, but not in B6-lpr/lpr or B6-tnfr0/0 lpr/lpr mice. These results indicate that apoptosis mediated by either Fas or TNF-R1 is sufficient for rapid clearance of the virus. However, apoptosis induced by Fas, but not TNF-R1, is required for the downmodulation of the immune response to the virus and prevention of a chronic inflammatory reaction.


Asunto(s)
Antígenos CD/fisiología , Apoptosis/fisiología , Infecciones por Citomegalovirus/prevención & control , Receptores del Factor de Necrosis Tumoral/fisiología , Receptor fas/fisiología , Animales , Antígenos CD/genética , Antígenos CD/metabolismo , Enfermedad Crónica , Infecciones por Citomegalovirus/patología , Infecciones por Citomegalovirus/fisiopatología , Femenino , Etiquetado Corte-Fin in Situ/métodos , Riñón/patología , Hígado/patología , Pulmón/patología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Modelos Biológicos , Muromegalovirus/fisiología , Receptores del Factor de Necrosis Tumoral/genética , Receptores del Factor de Necrosis Tumoral/metabolismo , Receptores Tipo I de Factores de Necrosis Tumoral
4.
J Clin Invest ; 105(6): 813-21, 2000 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-10727450

RESUMEN

We assessed the effect of modified antigen presenting cells (APCs) expressing high levels of Fas ligand (APC-FasL) on post-viral chronic inflammatory disease. FasL-deficient B6-gld/gld mice infected with murine cytomegalovirus (MCMV) cleared the virus from their lungs, kidneys, and livers within 2 weeks of infection. However, inflammation persisted in these organs for more than 8 weeks, with a chronically increased T-cell response to MCMV-infected APCs and production of autoantibodies. Administration of APC-AdFasL at 4 weeks suppressed this inflammation and diminished the T-cell response and autoantibody production. APC-AdFasL that had been transfected with ultraviolet-irradiated MCMV were more effective than uninfected APC-AdFasL in ameliorating the chronic inflammation. APC-AdFasL migrated preferentially to the spleen, where they triggered apoptosis of lymphocytes in the marginal zone of the spleen. These results confirm that Fas-mediated apoptosis is not required for clearance of virus, but is required for down-modulation of the virally induced chronic inflammatory response. This organwide effect of APC-AdFasL appears to be mediated by elimination of activated T lymphocytes in the spleen before their emigration to the target organs.


Asunto(s)
Células Presentadoras de Antígenos/inmunología , Apoptosis , Infecciones por Citomegalovirus/inmunología , Glicoproteínas de Membrana/fisiología , Adenoviridae/genética , Animales , Células Presentadoras de Antígenos/trasplante , Autoanticuerpos/biosíntesis , Infecciones por Citomegalovirus/patología , Infecciones por Citomegalovirus/terapia , Proteína Ligando Fas , Femenino , Vectores Genéticos/genética , Hepatitis Viral Animal/etiología , Hepatitis Viral Animal/inmunología , Hepatitis Viral Animal/patología , Inflamación , Riñón/patología , Hígado/patología , Pulmón/patología , Enfermedades Pulmonares Intersticiales/etiología , Enfermedades Pulmonares Intersticiales/inmunología , Enfermedades Pulmonares Intersticiales/patología , Macrófagos/inmunología , Glicoproteínas de Membrana/deficiencia , Glicoproteínas de Membrana/genética , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos MRL lpr , Ratones Mutantes , Nefritis/etiología , Nefritis/inmunología , Nefritis/patología , Organismos Libres de Patógenos Específicos , Bazo/inmunología , Bazo/patología , Subgrupos de Linfocitos T/inmunología , Transfección , Receptor fas/fisiología
5.
J Natl Cancer Inst ; 67(1): 207-12, 1981 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-6166780

RESUMEN

The iv inoculation of a suspension of osteogenic sarcoma cells induced metastatic tumor nodules in the lungs of C57BL/6 mice. The administration of 50,000-100,000 U of interferon daily for 7 days strikingly reduced the tumor mass in the lung and the number of tumor nodules present in histopathologic sections when the interferon treatment was initiated immediately after tumor cell inoculation. In some animals the development of any detectable metastatic lesion was completely prevented. Extending the therapy form 7 days to 21 days failed to improve the protective effect. Interferon therapy delayed until 7 days after tumor cell inoculation had no effect. These findings indicate the effectiveness of exogenous interferon in this murine osteogenic sarcoma model when interferon treatment is initiated within 1 hour of tumor cell inoculation, but not when it is delayed until tumor nodules are established in the lungs.


Asunto(s)
Interferones/uso terapéutico , Neoplasias Pulmonares/terapia , Osteosarcoma/terapia , Animales , Femenino , Neoplasias Pulmonares/patología , Neoplasias Pulmonares/secundario , Ratones , Ratones Endogámicos C57BL , Metástasis de la Neoplasia , Osteosarcoma/secundario , Sarcoma Experimental/terapia , Factores de Tiempo
6.
J Natl Cancer Inst ; 60(3): 659-66, 1978 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-272470

RESUMEN

Murine interferon inhibited the growth of a continuous line of osteogenic sarcoma (OGS) cells in tissue culture. Inhibition of tumor cell growth by interferon was demonstrated by: a) decreased colony formation in soft agar, b) suppression of clone formation in liquid medium, and c) reduction of tumor cell counts in monolayer cultures. This inhibition of cell growth was further documented by suppression of [3H]thymidine uptake by OGS cells exposed to interferon, which suggested inhibition of DNA synthesis of tumor cells. Exposure of tumor cells for 4 hours, 24 hours, and 2,3,4,6, and 8 days demonstrated greater activity with prolonged exposure to interferon. Inhibition of cell growth was significantly greater for OGS cells than for normal mouse embryo fibroblasts. Finally, the antitumor activity of the interferon preparation could be reversed by anti-interferon antibody.


Asunto(s)
Interferones/farmacología , Sarcoma Experimental/tratamiento farmacológico , Animales , Anticuerpos/administración & dosificación , División Celular/efectos de los fármacos , Células Cultivadas , Interferones/inmunología , Osteosarcoma/tratamiento farmacológico , Sarcoma Experimental/metabolismo , Timidina/metabolismo
7.
J Natl Cancer Inst ; 61(3): 871-4, 1978 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-278865

RESUMEN

Interferon was used to treat C57BL/6 female mice inoculated with a continuous line of murine osteogenic sarcoma cells. A short 7-day course of 30,000--60,000 U/day of tpe I interferon either completely inhibited or delayed the appearance of tumors in experimental animals. The therapeutic efficacy of type I interferon was compared with murine serum that contained type II interferon as well as other lymphokine activity. Tumor development was strikingly inhibited in animals treated for 7 days with serum containing only 600 U of type II interferon. Inhibition of tumor development was thus achieved with 100-fold less interferon than that required with type I preparation.


Asunto(s)
Interferones/uso terapéutico , Sarcoma Experimental/tratamiento farmacológico , Animales , Femenino , Interferones/administración & dosificación , Interferones/aislamiento & purificación , Ratones , Ratones Endogámicos C57BL , Osteosarcoma/tratamiento farmacológico , Neoplasias de los Tejidos Blandos/tratamiento farmacológico
8.
J Natl Cancer Inst ; 72(5): 1137-40, 1984 May.
Artículo en Inglés | MEDLINE | ID: mdl-6201642

RESUMEN

The injection of murine interferon three times weekly in dose levels of 1,250, 5,000, or 20,000 U produced no significant antitumor effect against primary 239Pu-induced osteosarcomas in C57BL/6J mice. The interferon treatment was begun 94 days after the plutonium injection, which is well before the radiographic or microscopic appearance of neoplasia, and was continued until the moribund state or death. The average radiation dose accumulated by the skeleton at the time of first treatment was approximately 300 rad. The largest dose of interferon studied, 20,000 U/injection, was approximately 3 X 10(6) U/m2 of body surface, or 10(6) U/kg body weight.


Asunto(s)
Neoplasias Óseas/prevención & control , Interferones/uso terapéutico , Neoplasias Inducidas por Radiación/prevención & control , Osteosarcoma/prevención & control , Plutonio/toxicidad , Animales , Neoplasias Óseas/etiología , Ratones , Ratones Endogámicos C57BL , Osteosarcoma/etiología
9.
Neurology ; 37(7): 1189-93, 1987 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-3037437

RESUMEN

Ross River virus (RRV), an alpha togavirus, causes an inflammatory myopathy in mice, which probably results from direct lytic effects of virus or viral products on myofibers. Administration of recombinant hybrid human leukocyte interferon-alpha A/D (rIFN-alpha A/D) ameliorates clinical illness and reduces mortality from 86 to 42%. Peak concentrations of virus are reduced by 1,000-fold in serum and by 30-fold in muscle, but anti-RRV antibody production is not altered. Treatment with rIFN-alpha A/D dramatically reduces inflammation and necrosis in muscle. Beneficial effects of rIFN-alpha A/D on experimental, RRV-induced polymyositis result in part from inhibition of viral replication and spread, though immunomodulation might also play an important role.


Asunto(s)
Interferón Tipo I/uso terapéutico , Miositis/terapia , Infecciones por Togaviridae/terapia , Animales , Anticuerpos Antivirales/análisis , Ratones , Músculos/microbiología , Músculos/patología , Miositis/microbiología , Miositis/patología , Virus del Río Ross/inmunología
10.
Am J Med ; 73(1A): 100-8, 1982 Jul 20.
Artículo en Inglés | MEDLINE | ID: mdl-6285699

RESUMEN

Genital herpes simplex virus type 2 (HSV-2) infections in mice and guinea pigs were used to determine the effectiveness of acyclovir administered topically, orally, or parenterally. Topical treatment with 1 percent or 5 percent acyclovir begun six or 24 hours and 5 percent acyclovir begun 48 or 72 hours after intravaginal inoculation of mice with HSV-2 significantly inhibited viral replication in the genital tract. Oral administration of acyclovir resulted in a significant reduction in vaginal virus titers when therapy was begun as late as 72 hours after infection. Topical treatment with 5 percent acyclovir initiated 24, 48, or 96 hours after intravaginal inoculation of guinea pigs with HSV-2 significantly altered lesion severity and virus titers but not vaginal HSV titers. Oral acyclovir therapy begun 24 or 48 hours after infection also significantly reduced the mean lesion score and prevented their development in some animals. Lesion and vaginal virus titers were only partially reduced. Intramuscular administration of acyclovir begun on day 2 or day 4 of infection significantly reduced the mean lesion scores but failed to alter lesion or vaginal virus titers.


Asunto(s)
Antivirales/uso terapéutico , Guanina/análogos & derivados , Herpes Genital/tratamiento farmacológico , Aciclovir , Administración Oral , Administración Tópica , Animales , Antivirales/administración & dosificación , Evaluación Preclínica de Medicamentos , Femenino , Guanina/administración & dosificación , Guanina/uso terapéutico , Inyecciones Intramusculares , Masculino , Ratones , Simplexvirus/efectos de los fármacos
11.
Am J Med ; 73(1A): 132-7, 1982 Jul 20.
Artículo en Inglés | MEDLINE | ID: mdl-6285702

RESUMEN

Murine cytomegalovirus (MCMV) is inhibited in vitro by 1 to 2 microM acyclovir. Therapy of a systemic MCMV infection in weanling mice with acyclovir was only minimally effective when drug was administered intraperitoneally, while oral administration by addition of acyclovir to the drinking water was highly efficacious in mice with disseminated MCMV. Effective therapy was characterized by reduction of virus titers in lung, liver, spleen, and kidney. In mice chronically infected with MCMV, treatment for 30 days with oral acyclovir eliminated or reduced virus titers in all target organs except the salivary gland. Therapeutic efficacy in this model infection using oral administration of acyclovir could be correlated with the achievement of acyclovir levels in the plasma of experimental animals two to 10 times greater than the mean inhibitory concentration for MCMV in vitro throughout treatment. The lack of efficacy observed when drug was administered intraperitoneally was associated with acyclovir levels exceeding 1 microM for one to three hours after each dose.


Asunto(s)
Antivirales/uso terapéutico , Infecciones por Citomegalovirus/tratamiento farmacológico , Citomegalovirus/efectos de los fármacos , Guanina/análogos & derivados , Enfermedad Aguda , Aciclovir , Administración Oral , Animales , Antivirales/sangre , Enfermedad Crónica , Femenino , Guanina/sangre , Guanina/uso terapéutico , Ratones
12.
Am J Med ; 73(1A): 125-31, 1982 Jul 20.
Artículo en Inglés | MEDLINE | ID: mdl-7102699

RESUMEN

The effect of oral or intraperitoneal administration of acyclovir was evaluated in four experimental models of herpes simplex virus (HSV) encephalitis in mice. Mice were inoculated with HSV-1 or HSV-2 intracerebrally or with HSV-2 intranasally, intraperitoneally, or intravaginally. With all four routes of inoculation, oral acyclovir therapy significantly reduced mortality when started as late as 72 to 96 hours after viral challenge. Intraperitoneal acyclovir was not effective in protecting mice inoculated intravaginally, but was effective if given 24 to 48 hours after intracerebral or intranasal challenge and as late as 96 hours after intraperitoneal infection. Oral acyclovir was more active than intraperitoneal treatment in all four model infections. Levels of acyclovir inhibitory for HSV in cell culture were maintained in plasma and brain tissue throughout oral treatment but lasted only three to six hours after each intraperitoneal treatment. These results suggest that acyclovir may be useful in treating serious HSV infections in humans.


Asunto(s)
Antivirales/administración & dosificación , Encefalitis/tratamiento farmacológico , Guanina/análogos & derivados , Herpes Simple/tratamiento farmacológico , Aciclovir , Administración Oral , Animales , Antivirales/metabolismo , Encéfalo/metabolismo , Femenino , Guanina/administración & dosificación , Guanina/metabolismo , Inyecciones Intraperitoneales , Ratones , Factores de Tiempo , Distribución Tisular
13.
J Med Chem ; 44(23): 4019-22, 2001 Nov 08.
Artículo en Inglés | MEDLINE | ID: mdl-11689090

RESUMEN

Alkylation-elimination of adenine and 2-amino-6-chloropurine with gem-difluorocyclopropane dibromide 10 gave E- and Z-methylene-gem-difluorocyclopropanes 11a, 11b, 12a, and 12b and gem-difluorocyclopropenes 13a and 13b. Debenzylation of intermediates 11a, 11b, 12a, and 12b afforded E- and Z-methylenecyclopropanes 4a, 4b, 5a, and 5b. Hydrolysis of 2-amino-6-chloropurine derivatives 4b and 5b afforded guanine analogues 4c and 5c. Composition of products (except 14b) obtained from alkylation-elimination reflects thermodynamically controlled cyclopropene-methylenecyclopropene rearrangement. The E-isomer 4a was moderately active against human cytomegalovirus and along with the Z-isomer 5a was active against leukemia L1210 and solid tumors in vitro.


Asunto(s)
Antineoplásicos/síntesis química , Antivirales/síntesis química , Ciclopropanos/química , Nucleósidos/síntesis química , Animales , Fármacos Anti-VIH/síntesis química , Fármacos Anti-VIH/química , Fármacos Anti-VIH/farmacología , Antineoplásicos/química , Antineoplásicos/farmacología , Antivirales/química , Antivirales/farmacología , Línea Celular , Ciclopropanos/síntesis química , Ciclopropanos/farmacología , Ensayos de Selección de Medicamentos Antitumorales , Humanos , Espectroscopía de Resonancia Magnética , Ratones , Nucleósidos/química , Nucleósidos/farmacología , Estereoisomerismo , Relación Estructura-Actividad
14.
J Med Chem ; 33(12): 3160-9, 1990 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-2175356

RESUMEN

Protection of the 3'- and 5'-hydroxyl groups of the nucleoside antibiotic toyocamycin (1) with 1,3-dichloro-1,1,3,3-tetraisopropyldisiloxane was followed by (trifluoromethyl)sulfonylation of the 2'-hydroxyl group. A displacement of the resulting triflate ester moiety with lithium chloride, lithium bromide, sodium iodide, and lithium azide in hexamethylphosphoramide was followed by a removal of the disilyl moiety with tetra-n-butylammonium fluoride to afford the appropriate (2'-deoxy-2'-substituted-arabinofuranosyl)toyocamycin analogues 6a-d. Hydrolysis of the carbonitrile moieties of 6a-d with hydrogen peroxide gave the corresponding sangivamycin analogues (7a-d). A reduction of the azido moiety of 6a and 7a with 1,3-propanedithiol furnished the corresponding amino derivatives (6e and 7e). The antiproliferative activity of 6a-e and 7a-e was evaluated in L1210 cell cultures. None of these compounds caused significant inhibition of cell growth. Evaluation of these compounds for antiviral activity showed that all the toyocamycin analogues were active against human CMV, but of the sangivamycin analogues, only (2'-deoxy-2'-azidoarabinosyl)sangivamycin (7a) was active against this virus. None of the compounds were active against HSV-1 or HSV-2. (2'-Deoxy-2'-aminoarabinofuranosyl)toyocamycin (6e) was studied more extensively and showed some separation between antiviral activity and cytotoxicity as measured by effects on DNA synthesis, cell growth, and cell-plating efficiency. Although 6e also was active against murine CMV in vitro, it was not active against this virus in infected mice. We conclude that arabinosylpyrrolopyrimidines have potential as antivirals, but no members of the current series are potent enough to show significant activity in vivo.


Asunto(s)
Antivirales/farmacología , Arabinonucleósidos/farmacología , Infecciones por Citomegalovirus/tratamiento farmacológico , Citomegalovirus/efectos de los fármacos , Nucleósidos de Pirimidina/farmacología , Toyocamicina/análogos & derivados , Animales , Antivirales/síntesis química , Antivirales/uso terapéutico , Arabinonucleósidos/síntesis química , Arabinonucleósidos/uso terapéutico , División Celular/efectos de los fármacos , Fenómenos Químicos , Química , Humanos , Leucemia L1210/patología , Ratones , Estructura Molecular , Nucleósidos de Pirimidina/síntesis química , Nucleósidos de Pirimidina/uso terapéutico , Simplexvirus/efectos de los fármacos , Relación Estructura-Actividad , Toyocamicina/síntesis química , Toyocamicina/farmacología , Toyocamicina/uso terapéutico , Células Tumorales Cultivadas
15.
J Med Chem ; 33(7): 1984-92, 1990 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-2163454

RESUMEN

The sodium salts of 4-chloro- and several 4-chloro-5-substituted-7H-pyrrolo[2,3-d]pyrimidines were treated with [1,3-bis(benzyloxy)-2-propoxy]methyl chloride (6) to provide the corresponding 4-chloro- and 4-chloro-5-substituted-7-[[1,3-bis(benzyloxy)-2-propoxy]methyl]pyrrolo [2,3-d]pyrimidines (7-11). Debenzylation with boron trichloride at -78 degrees C furnished 4-chloro- and several 4-chloro-5-substituted-7-[(1,3-dihydroxy-2-propoxy)methyl]pyrrolo[2,3- d]pyrimidines (12.16). Subsequent amination of 12-16 yielded the 4-amino-5-substituted-7-[(1,3-dihydroxy-2-propoxy)methyl]pyrrolo[2,3- d]pyrimidines (17-21). Treatment of 14 with methylamine and 13 and 14 with ethylamine yielded the 4-(alkylamino)-5-halo-7-[(1,3-dihydroxy-2- propoxy)methyl]pyrrolo[2,3-d]pyrimidines (22-24). Treatment of 12-15 with hydroxylamine in refluxing 2-propanol yielded the 5-substituted-4-(hydroxyamino)-7-[(1,3-dihydroxy-2-propoxy)methyl]pyrrol o [2,3-d]pyrimidines (25-28). Treatment of compound 12 with Pd/C under a hydrogen atmosphere has furnished the nebularine analogue 31. The antiproliferative activity of compounds 17-28 and 31 was studied using L1210 cells in vitro. The 4-amino- and 4-(hydroxyamino)-5-halogenated derivatives (compounds 18-20, 26-28) inhibited cell growth. Although the effect of compounds 18-20 and 27 on final growth rate was pronounced (IC50 = 2.3, 0.7, 2.8, and 3.7 microM, respectively), cells underwent at least one doubling before cell division stopped. The remaining compounds were less cytotoxic, with IC50's greater than 30 microM for 21, 23, 26, and 28, whereas no inhibition of L1210 cell growth was observed with compounds 17, 22, 24, 25, and 31 at 100 microM. The antiviral activity of these compounds also was tested. Compounds 18-20 and 26-28 were active against human cytomegalovirus and herpes simplex type 1. The 4-amino derivatives (18-20) were more active than the 4-hydroxyamino derivatives (26-28), the 4-amino-5-bromo and 4-amino-5-iodo derivatives produced more than five log reductions in virus titer at concentrations of 10-100 microM. Although some cytotoxicity was observed at these concentrations, compound 19 was active against murine cytomegalovirus in vivo. At 5.6 mg/kg, 14/15 animals survived compared to 10/15 treated with 5.6 mg/kg of ganciclovir or 1/15 treated with placebo.


Asunto(s)
Antineoplásicos/síntesis química , Antivirales/síntesis química , Citomegalovirus/efectos de los fármacos , Pirimidinas/síntesis química , Pirroles/síntesis química , Simplexvirus/efectos de los fármacos , Animales , División Celular/efectos de los fármacos , Línea Celular , Supervivencia Celular/efectos de los fármacos , Citomegalovirus/crecimiento & desarrollo , Infecciones por Citomegalovirus/tratamiento farmacológico , Femenino , Humanos , Indicadores y Reactivos , Células KB , Leucemia L1210 , Espectroscopía de Resonancia Magnética , Ratones , Pruebas de Sensibilidad Microbiana , Estructura Molecular , Pirimidinas/farmacología , Pirimidinas/uso terapéutico , Pirroles/farmacología , Pirroles/uso terapéutico , Simplexvirus/crecimiento & desarrollo , Relación Estructura-Actividad , Células Tumorales Cultivadas/citología , Células Tumorales Cultivadas/efectos de los fármacos
16.
J Med Chem ; 38(20): 4106-14, 1995 Sep 29.
Artículo en Inglés | MEDLINE | ID: mdl-7562946

RESUMEN

The glycosylation of 3,4-dicyano-2-[(ethoxymethylene)amino]pyrrole (7) with 2-deoxy-2-fluoro-alpha-D-erythro-pentofuranosyl bromide (2) furnished an anomeric mixture of nucleosides (8a,b). This mixture was separated, and the individual anomers were treated with methanolic ammonia to effect a concomitant deblocking and ring closure. This furnished both anomers of 2'-deoxy-2'-fluoro-ara-toyocamycin (9a,b). The cyano moiety of 9b was converted to the carboxamide moiety to furnish 2'-deoxy-2'-fluoro-ara-sangivamycin (10) and to the thiocarboxamide moiety to furnish 2'-deoxy-2'-fluoro-ara-thiosangivamycin (11). The target compounds 10 and 11 showed similar antiproliferative activity against L1210 cells in vitro, with IC50's of 3 and 5 microM. Antiviral evaluation revealed a somewhat different pattern of activity. All analogs, both alpha and beta anomers, were active against human cytomegalovirus (HCMV), albeit the beta anomers were most active. The beta anomers also were active against herpes simplex virus type 1 (HSV-1) and human immunodeficiency virus (HIV). Compound 10 was most active in the series, ca. 10-fold more potent than 11; IC50's for 10 ranged from 4 to 25 nM for HCMV, HIV, and varicella zoster virus (VZV) and from 30 to 500 nM for HSV-1. Even though compound 10 was cytotoxic, which will probably preclude its use as an antiviral drug (IC50's = 0.2-5.5 microM), the difference between cytotoxicity and activity against HCMV, HIV, and VZV was sufficient to indicate specific activity against a viral target.


Asunto(s)
Antibióticos Antineoplásicos/síntesis química , Antivirales/síntesis química , Arabinonucleósidos/síntesis química , Nucleósidos de Pirimidina/síntesis química , Toyocamicina/síntesis química , Arabinonucleósidos/farmacología , Citomegalovirus/efectos de los fármacos , VIH/efectos de los fármacos , Herpesvirus Humano 1/efectos de los fármacos , Humanos , Células KB , Nucleósidos de Pirimidina/farmacología , Toyocamicina/farmacología
17.
J Med Chem ; 41(1): 10-23, 1998 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-9438017

RESUMEN

New nucleoside analogues 14-17 based on a methylenecyclopropane structure were synthesized and evaluated for antiviral activity. Reaction of 2,3-dibromopropene (19) with adenine (18) led to bromoalkene 20, which was benzoylated to give N6,N6-dibenzoyl derivative 23. Attempts to convert 20 or 23 to bromocyclopropanes 21 and 22 by reaction with ethyl diazoacetate catalyzed by Rh2(OAc)4 were futile. By contrast, 2,3-dibromopropene (19) afforded smoothly (E)- and (Z)-dibromocyclopropane carboxylic esters 24 + 25. Alkylation of adenine (18) with 24 + 25 gave (E)- and (Z)-bromo derivatives 21 + 22. Base-catalyzed elimination of HBr resulted in the formation of (Z)- and (E)-methylenecyclopropanecarboxylic esters 26 + 27. More convenient one-pot alkylation-elimination of adenine (18) or 2-amino-6-chloropurine (30) with 24 + 25 afforded (Z)- and (E)-methylenecyclopropane derivatives 26 + 27 and 31 + 32. The Z-isomers were always predominant in these mixtures (Z/E approximately 2/1). Reduction of 26 + 27 and 31 + 32 with DIBALH afforded (Z)- and (E)-methylenecyclopropane alcohols 14 + 16 and 33 + 34. The latter were resolved directly by chromatography. Compounds 14 + 16 were converted to N6-(dimethylamino)methylene derivatives 28 and 29 which were separated and deprotected to give 14 and 16. Reaction of 33 and 34 with HCO2H led to guanine analogues 15 and 17. The 1H NMR spectra of the Z-analogues 14 and 15 are consistent with an anti-like conformation of the nucleobases. By contrast, 1H NMR and IR spectra of bromo ester 21 are indicative of syn-conformation of adenine. Several Z-(hydroxymethyl)methylenecyclopropanes exhibited in vitro antiviral activity in micromolar or submicromolar range against human and murine cytomegalovirus (HCMV and MCMV), Epstein-Barr virus (EBV), human herpes virus 6 (HHV-6), varicella zoster virus (VZV), and hepatitis B virus (HBV). Analogues 14, 15, and 33 were the most effective agents against HCMV (IC50 1-2.1, 0.04-2.1, and 0.8-5.6 microM), MCMV (IC50 2.1, 0.3, and 0.3 microM) and EBV in H-1 (IC50 0.2, 0.3, and 0.7 microM) and Daudi cells (IC50 3.2, 5.6, and 1.2 microM). Adenine analogue 14 was active against HBV (IC50 2 microM), VZV (IC50 2.5 microM), and HHV-6 (IC50 14 microM). Synadenol (14) and the E-isomer (16) were substrates of moderate efficiency for adenosine deaminase from calf intestine. The E-isomer 16 was more reactive than Z-isomer 14. The deamination of 14 effectively stopped at 50% conversion. Synadenol (14) was also deaminated by AMP deaminase from aspergillus sp.


Asunto(s)
Adenosina/análogos & derivados , Adenosina/síntesis química , Fármacos Anti-VIH/síntesis química , Antivirales/síntesis química , Ciclopropanos/síntesis química , Guanosina/análogos & derivados , Guanosina/síntesis química , Replicación Viral/efectos de los fármacos , Adenosina/química , Adenosina/farmacología , Animales , Fármacos Anti-VIH/química , Fármacos Anti-VIH/farmacología , Antivirales/química , Antivirales/farmacología , Línea Celular , Supervivencia Celular/efectos de los fármacos , Chlorocebus aethiops , Ciclopropanos/química , Ciclopropanos/farmacología , Guanosina/química , Guanosina/farmacología , VIH-1/efectos de los fármacos , VIH-1/fisiología , Virus de la Hepatitis B/efectos de los fármacos , Virus de la Hepatitis B/fisiología , Herpesvirus Humano 1/efectos de los fármacos , Herpesvirus Humano 1/fisiología , Herpesvirus Humano 2/efectos de los fármacos , Herpesvirus Humano 2/fisiología , Humanos , Ratones , Pruebas de Sensibilidad Microbiana , Estructura Molecular , Resonancia Magnética Nuclear Biomolecular , Espectrofotometría Infrarroja , Células Vero
18.
Viral Immunol ; 12(3): 263-75, 1999.
Artículo en Inglés | MEDLINE | ID: mdl-10532654

RESUMEN

Freshly isolated macrophages from mature mice are poorly or nonpermissive for infections with HSV. However, despite lack of significant viral replication, HSV infection has been demonstrated to induce substantial cell death among macrophages. To determine if HSV-induced cytotoxicity of macrophages is due to apoptosis, peritoneal macrophages were obtained from C57BL/6 (B6) mice, and apoptosis was analyzed following HSV-2 infection in vitro. Macrophages underwent apoptosis upon HSV-2 infection indicated by annexin V staining, labeling of DNA strand breaks and electronmicroscopy. Apoptosis was associated with macrophage activation demonstrated by upregulation of MHC class II and Mac-1 surface expression. Though there was also an upregulation of Fas (Apo-1/CD95) and tumor necrosis factor (TNF)-receptor 1 (TNF-R1) pathways, inhibition of Fas by soluble Fas and blocking of TNF-alpha using a TNF-binding protein did not prevent HSV-induced apoptosis. Moreover, apoptosis was not impaired in HSV-2 infected macrophages from Fas-deficient B6-lpr/lpr mice suggesting involvement of other apoptosis pathways, or activation of Fas or TNF-R pathways downstream of the receptor level. The present results demonstrate that HSV-2 infection leads to activation and subsequent apoptosis in peritoneal macrophages independent of Fas or TNF-R1 signaling.


Asunto(s)
Antígenos CD/metabolismo , Apoptosis , Herpesvirus Humano 2/fisiología , Macrófagos Peritoneales/virología , Receptores del Factor de Necrosis Tumoral/metabolismo , Transducción de Señal , Receptor fas/metabolismo , Animales , Células Cultivadas , Femenino , Fibroblastos/citología , Fibroblastos/virología , Expresión Génica , Regulación Viral de la Expresión Génica , Genes Virales , Humanos , Riñón/citología , Activación de Macrófagos , Macrófagos Peritoneales/citología , Macrófagos Peritoneales/inmunología , Ratones , Ratones Endogámicos C57BL , Conejos , Receptores Tipo I de Factores de Necrosis Tumoral , Transcripción Genética , Replicación Viral
19.
Environ Health Perspect ; 43: 71-9, 1982 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-6174323

RESUMEN

Viral infections in animal models appear to be ideal systems for determining toxicity to the immune system by environmental substances. Since many viral infections that are utilized in animals produce systemic disease, these models provide an opportunity to evaluate the interaction between virus and components of host resistance. In these infections it is possible to delineate the role of antibody, interferon, cell-mediated immunity, neutrophils and macrophages in response to infection. A change in any of these components responsible for resistance to a particular virus may be correlated with an alteration of mortality an pathogenesis of the viral infection. Three experimental viral infections in mice that are potential candidates for use in determining immunotoxicity are discussed in terms of the response of individual components of resistance to infection and how changes in there components result in alterations of viral pathogenesis. The resistance to encephalomyocarditis virus infection in mice appears to be primarily mediated by antibody and interferon while with herpes simplex virus, infections are mainly controlled through cell-mediated immunity, macrophages, and possible interferon. Cellular immunity also appears to be primarily responsible for resistance to cytomegalovirus infections. Therefore, it is important in the use of these systems for evaluating immunotoxicity to define the pathogenesis of the viral infection and the specific host responses to these infections and to be able to correlate a change in host resistance with an alteration of the viral infection.


Asunto(s)
Modelos Animales de Enfermedad , Virosis/inmunología , Animales , Infecciones por Citomegalovirus/inmunología , Virus de la Encefalomiocarditis , Infecciones por Enterovirus/inmunología , Contaminantes Ambientales/toxicidad , Herpes Simple/inmunología , Inmunidad/efectos de los fármacos , Interferones/farmacología , Macrófagos/inmunología , Ratones , Poli I-C/farmacología
20.
Antiviral Res ; 29(1): 57-9, 1996 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-8721546

RESUMEN

Although experimental viral infections in animals have been used extensively in the development of antiviral drugs used as monotherapy, they have not been utilized widely for evaluation of combination chemotherapy. One of the major reasons for the lack of use of animal models is that for the diseases that are the main target for combination therapy, AIDS and hepatitis B and C infections, there is a lack of suitable models for these diseases. In contrast, most combination studies in animal models have been directed against herpes simplex virus infections but there are relatively few patients available who would benefit from combination therapy over single agent therapy. In between those two extremes are the cytomegalovirus infections. While there are animal models available that have been predictive of efficacy in humans and there are sufficient patients available, the use of antiviral combinations in animal models and in humans have begun only recently. At the present time there is not enough information available to establish the predictability for any of the animal models for efficacy of combinations of antiviral agents.


Asunto(s)
Antivirales/uso terapéutico , Modelos Animales de Enfermedad , Síndrome de Inmunodeficiencia Adquirida/tratamiento farmacológico , Animales , Antivirales/administración & dosificación , Gatos , Infecciones por Citomegalovirus/tratamiento farmacológico , Quimioterapia Combinada , Patos , Cobayas , Haplorrinos , Hepatitis B/tratamiento farmacológico , Hepatitis C/tratamiento farmacológico , Herpes Simple/tratamiento farmacológico , Marmota , Ratones , Conejos , Ratas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA