Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 93
Filtrar
1.
J Virol ; 86(9): 5304-13, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22379082

RESUMEN

Herpes simplex virus 1 (HSV-1) mutants that lack the γ(1)34.5 gene are unable to replicate in the central nervous system but maintain replication competence in dividing cell populations, such as those found in brain tumors. We have previously demonstrated that a γ(1)34.5-deleted HSV-1 expressing murine interleukin-12 (IL-12; M002) prolonged survival of immunocompetent mice in intracranial models of brain tumors. We hypothesized that M002 would be suitable for use in clinical trials for patients with malignant glioma. To test this hypothesis, we (i) compared the efficacy of M002 to three other HSV-1 mutants, R3659, R8306, and G207, in murine models of brain tumors, (ii) examined the safety and biodistribution of M002 in the HSV-1-sensitive primate Aotus nancymae following intracerebral inoculation, and (iii) determined whether murine IL-12 produced by M002 was capable of activating primate lymphocytes. Results are summarized as follows: (i) M002 demonstrated superior antitumor activity in two different murine brain tumor models compared to three other genetically engineered HSV-1 mutants; (ii) no significant clinical or magnetic resonance imaging evidence of toxicity was observed following direct inoculation of M002 into the right frontal lobes of A. nancymae; (iii) there was no histopathologic evidence of disease in A. nancymae 1 month or 5.5 years following direct inoculation; and (iv) murine IL-12 produced by M002 activates A. nancymae lymphocytes in vitro. We conclude that the safety and preclinical efficacy of M002 warrants the advancement of a Δγ(1)34.5 virus expressing IL-12 to phase I clinical trials for patients with recurrent malignant glioma.


Asunto(s)
Vectores Genéticos/genética , Interleucina-12/genética , Simplexvirus/genética , Aciclovir/farmacología , Animales , Antivirales/farmacología , Aotidae , Encéfalo/patología , Línea Celular , Chlorocebus aethiops , Evaluación Preclínica de Medicamentos , Femenino , Expresión Génica , Terapia Genética , Vectores Genéticos/administración & dosificación , Vectores Genéticos/efectos adversos , Glioma/genética , Glioma/mortalidad , Glioma/terapia , Humanos , Interleucina-12/metabolismo , Imagen por Resonancia Magnética , Masculino , Ratones , Ratones SCID , Simplexvirus/efectos de los fármacos , Análisis de Supervivencia , Replicación Viral/efectos de los fármacos , Replicación Viral/genética , Ensayos Antitumor por Modelo de Xenoinjerto
2.
Antiviral Res ; 94(2): 111-25, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22406470

RESUMEN

Investments in the development of new drugs for orthopoxvirus infections have fostered new avenues of research, provided an improved understanding of orthopoxvirus biology and yielded new therapies that are currently progressing through clinical trials. These broad-based efforts have also resulted in the identification of new inhibitors of orthopoxvirus replication that target many different stages of viral replication cycle. This review will discuss progress in the development of new anti-poxvirus drugs and the identification of new molecular targets that can be exploited for the development of new inhibitors. The prototype of the orthopoxvirus group is vaccinia virus and its replication cycle will be discussed in detail noting specific viral functions and their associated gene products that have the potential to serve as new targets for drug development. Progress that has been achieved in recent years should yield new drugs for the treatment of these infections and might also reveal new approaches for antiviral drug development with other viruses.


Asunto(s)
Antivirales/farmacología , Descubrimiento de Drogas/métodos , Orthopoxvirus/efectos de los fármacos , Replicación Viral/efectos de los fármacos , Animales , Humanos , Orthopoxvirus/fisiología
3.
Antimicrob Agents Chemother ; 55(10): 4682-91, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21788463

RESUMEN

Cyclopropavir (CPV) is active against human cytomegalovirus (CMV), as well as both variants of human herpesvirus 6 and human herpesvirus 8. The mechanism of action of CPV against CMV is similar to that of ganciclovir (GCV) in that it is phosphorylated initially by the CMV UL97 kinase, resulting in inhibition of viral DNA synthesis. Resistance to CPV maps to the UL97 kinase but is associated primarily with H520Q mutations and thus retains good antiviral activity against most GCV-resistant isolates. An examination of CMV-infected cultures treated with CPV revealed unusual cell morphology typically associated with the absence of UL97 kinase activity. A surrogate assay for UL97 kinase activity confirmed that CPV inhibited the activity of this enzyme and that its action was similar to the inhibition seen with maribavir (MBV) in this assay. Combination studies using real-time PCR indicated that, like MBV, CPV also antagonized the efficacy of GCV and were consistent with the observed inhibition of the UL97 kinase. Deep sequencing of CPV-resistant laboratory isolates identified a frameshift mutation in UL27, presumably to compensate for a loss of UL97 enzymatic activity. We conclude that the mechanism of action of CPV against CMV is complex and involves both the inhibition of DNA synthesis and the inhibition of the normal activity of the UL97 kinase.


Asunto(s)
Antivirales/farmacología , Ciclopropanos/farmacología , Citomegalovirus/efectos de los fármacos , Citomegalovirus/enzimología , ADN Viral , Guanina/análogos & derivados , Fosfotransferasas (Aceptor de Grupo Alcohol)/antagonistas & inhibidores , Animales , Secuencia de Bases , Bencimidazoles/farmacología , Células COS , Línea Celular , Chlorocebus aethiops , Citomegalovirus/genética , Citomegalovirus/aislamiento & purificación , ADN Viral/biosíntesis , Farmacorresistencia Viral/genética , Mutación del Sistema de Lectura , Ganciclovir/farmacología , Guanina/farmacología , Herpesvirus Humano 6/efectos de los fármacos , Herpesvirus Humano 8/efectos de los fármacos , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Fosfotransferasas (Aceptor de Grupo Alcohol)/metabolismo , Ribonucleósidos/farmacología , Análisis de Secuencia de ADN
4.
Antimicrob Agents Chemother ; 55(10): 4728-34, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21788472

RESUMEN

Although acyclovir (ACV) has proven to be of value in the therapy of certain herpes simplex virus (HSV) infections, there is a need for more effective therapies, particularly for serious infections in neonates and immunocompromised individuals, where resistance to this drug can be problematic. CMX001 is an orally bioavailable lipid conjugate of cidofovir that is substantially less nephrotoxic than the parent drug and has excellent antiviral activity against all the human herpesviruses. This compound retains full antiviral activity against ACV-resistant laboratory and clinical isolates. The combined efficacy of CMX001 and ACV was evaluated in a new real-time PCR combination assay, which demonstrated that the combination synergistically inhibited the replication of HSV in cell culture. This was also confirmed in murine models of HSV infection, where the combined therapy with these two drugs synergistically reduced mortality. These results suggest that CMX001 may be effective in the treatment of ACV-resistant HSV infections and as an adjunct therapy in individuals with suboptimal responses to ACV.


Asunto(s)
Aciclovir/farmacología , Antivirales/farmacología , Citosina/análogos & derivados , Herpes Simple/tratamiento farmacológico , Herpesvirus Humano 1/efectos de los fármacos , Herpesvirus Humano 2/efectos de los fármacos , Organofosfonatos/farmacología , Aciclovir/uso terapéutico , Aciclovir/toxicidad , Animales , Antivirales/uso terapéutico , Antivirales/toxicidad , Células Cultivadas , Citosina/farmacología , Citosina/uso terapéutico , Citosina/toxicidad , Farmacorresistencia Viral , Sinergismo Farmacológico , Quimioterapia Combinada , Femenino , Herpes Simple/virología , Humanos , Ratones , Ratones Endogámicos BALB C , Organofosfonatos/uso terapéutico , Organofosfonatos/toxicidad
5.
Antimicrob Agents Chemother ; 55(5): 2442-5, 2011 May.
Artículo en Inglés | MEDLINE | ID: mdl-21300829

RESUMEN

Several benzimidazole nucleoside analogs, including 1H-ß-D-ribofuranosyl-2-bromo-5,6-dichlorobenzimidazole (BDCRB) and 1H-ß-L-ribofuranosyl-2-isopropylamino-5,6-dichlorobenzimidazole (maribavir [MBV]), inhibit the replication of human cytomegalovirus. Neither analog inhibited the related betaherpesvirus human herpesvirus 6 (HHV-6). Additional analogs of these compounds were evaluated against both variants of HHV-6, and two L-analogs of BDCRB had good antiviral activity against HHV-6A, as well as more modest inhibition of HHV-6B replication.


Asunto(s)
Antivirales/farmacología , Bencimidazoles/farmacología , Herpesvirus Humano 6/efectos de los fármacos , Antivirales/química , Bencimidazoles/química , Citomegalovirus/efectos de los fármacos , Humanos , Replicación Viral/efectos de los fármacos
6.
Virus Res ; 157(2): 212-21, 2011 May.
Artículo en Inglés | MEDLINE | ID: mdl-21095209

RESUMEN

Ganciclovir (GCV), the therapy of choice for human cytomegalovirus (CMV) infections and foscarnet, a drug used to treat GCV-resistant CMV infections was approved more than twenty years ago. Although cidofovir and a prodrug of GCV have since been added to the armamentarium, a highly effective drug without significant toxicities has yet to be approved. Such a therapeutic agent is required for treatment of immunocompromised hosts and infants, which bear the greatest burden of disease. The modest antiviral activity of existing drugs is insufficient to completely suppress viral replication, which results in the selection of drug-resistant variants that remain pathogenic, continue to replicate, and contribute to disease. Sustained efforts, largely in the biotech industry and academia, have identified highly active lead compounds that have progressed into clinical studies with varying levels of success. A few of these compounds inhibit new molecular targets, remain effective against isolates that have developed resistance to existing therapies, and promise to augment existing therapies. Some of the more promising drugs will be discussed with an emphasis on those progressing to clinical studies. Their antiviral activity both in vitro and in vivo, spectrum of antiviral activity, and mechanism of action will be reviewed to provide an update on the progress of potential new therapies for CMV infections.


Asunto(s)
Antivirales/uso terapéutico , Infecciones por Citomegalovirus/tratamiento farmacológico , Citomegalovirus/efectos de los fármacos , Cidofovir , Ciclopropanos/uso terapéutico , Citomegalovirus/fisiología , Citosina/análogos & derivados , Citosina/uso terapéutico , Foscarnet/uso terapéutico , Ganciclovir/análogos & derivados , Ganciclovir/uso terapéutico , Guanina/análogos & derivados , Guanina/uso terapéutico , Humanos , Organofosfonatos/uso terapéutico , Valganciclovir , Replicación Viral/efectos de los fármacos
7.
J Infect Dis ; 202(10): 1492-9, 2010 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-20923374

RESUMEN

CMX001, an orally active lipid conjugate of cidofovir, is 50 times more active in vitro against herpes simplex virus (HSV) replication than acyclovir or cidofovir. These studies compared the efficacy of CMX001 to acyclovir in BALB/c mice inoculated intranasally with HSV types 1 or 2. CMX001 was effective in reducing mortality using doses of 5 to 1.25 mg/kg administered orally once daily, even when treatments were delayed 48-72 h post viral inoculation. Organ samples obtained from mice treated with CMX001 had titers 3-5 log(10) plaque-forming units per gram of tissue lower than samples obtained from mice treated with acyclovir, including 5 different regions of the brain. Detectable concentrations of drug-related radioactivity were documented in the central nervous system of mice after oral administration of (14)C-CMX001. These studies indicate that CMX001 penetrates the blood-brain barrier, is a potent inhibitor of HSV replication in disseminated infections and central nervous system infections, and is superior to acyclovir.


Asunto(s)
Antivirales/administración & dosificación , Antivirales/farmacocinética , Citosina/análogos & derivados , Herpes Simple/tratamiento farmacológico , Herpesvirus Humano 1 , Herpesvirus Humano 2 , Organofosfonatos/administración & dosificación , Organofosfonatos/farmacocinética , Administración Oral , Animales , Citosina/administración & dosificación , Citosina/farmacocinética , Modelos Animales de Enfermedad , Evaluación Preclínica de Medicamentos , Femenino , Ratones , Ratones Endogámicos BALB C , Distribución Tisular
8.
Viruses ; 2(12): 2681-95, 2010 12.
Artículo en Inglés | MEDLINE | ID: mdl-21994637

RESUMEN

Although a large number of compounds have been identified with antiviral activity against orthopoxviruses in tissue culture systems, it is highly preferred that these compounds have activity in vivo before they can be seriously considered for further development. One of the most commonly used animal models for the confirmation of this activity has been the use of mice infected with either vaccinia or cowpox viruses. These model systems have the advantage that they are relatively inexpensive, readily available and do not require any special containment facilities; therefore, relatively large numbers of compounds can be evaluated in vivo for their activity. The two antiviral agents that have progressed from preclinical studies to human safety trials for the treatment of orthopoxvirus infections are the cidofovir analog, CMX001, and an inhibitor of extracellular virus formation, ST-246. These compounds are the ones most likely to be used in the event of a bioterror attack. The purpose of this communication is to review the advantages and disadvantages of using mice infected with vaccinia and cowpox virus as surrogate models for human orthopoxvirus infections and to summarize the activity of CMX001 and ST-246 in these model infections.

9.
Viruses ; 2(9): 1968-1983, 2010 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21994716

RESUMEN

The search for effective therapies for orthopoxvirus infections has identified diverse classes of molecules with antiviral activity. Pyrimidine analogs, such as 5-iodo-2'-deoxyuridine (idoxuridine, IDU) were among the first compounds identified with antiviral activity against a number of orthopoxviruses and have been reported to be active both in vitro and in animal models of infection. More recently, additional analogs have been reported to have improved antiviral activity against orthopoxviruses including several derivatives of deoxyuridine with large substituents in the 5 position, as well as analogs with modifications in the deoxyribose moiety including (north)-methanocarbathymidine, and 5-iodo-4'-thio-2'-deoxyuridine (4'-thioIDU). The latter molecule has proven to have good antiviral activity against the orthopoxviruses both in vitro and in vivo and has the potential to be an effective therapy in humans.

10.
Antimicrob Agents Chemother ; 53(12): 5251-8, 2009 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19770274

RESUMEN

A series of 4'-thionucleosides were synthesized and evaluated for activities against orthopoxviruses and herpesviruses. We reported previously that one analog, 5-iodo-4'-thio-2'-deoxyuridine (4'-thioIDU), exhibits good activity both in vitro and in vivo against two orthopoxviruses. This compound also has good activity in cell culture against many of the herpesviruses. It inhibited the replication of herpes simplex virus type 1 (HSV-1), HSV-2, and varicella-zoster virus with 50% effective concentrations (EC(50)s) of 0.1, 0.5, and 2 microM, respectively. It also inhibited the replication of human cytomegalovirus (HCMV) with an EC(50) of 5.9 microM but did not selectively inhibit Epstein-Barr virus, human herpesvirus 6, or human herpesvirus 8. While acyclovir-resistant strains of HSV-1 and HSV-2 were comparatively resistant to 4'-thioIDU, it retained modest activity (EC(50)s of 4 to 12 microM) against these strains. Some ganciclovir-resistant strains of HCMV also exhibited reduced susceptibilities to the compound, which appeared to be related to the specific mutations in the DNA polymerase, consistent with the observed incorporation of the compound into viral DNA. The activity of 4'-thioIDU was also evaluated using mice infected intranasally with the MS strain of HSV-2. Although there was no decrease in final mortality rates, the mean length of survival after inoculation increased significantly (P < 0.05) for all animals receiving 4'-thioIDU. The findings from the studies presented here suggest that 4'-thioIDU is a good inhibitor of some herpesviruses, as well as orthopoxviruses, and this class of compounds warrants further study as a therapy for infections with these viruses.


Asunto(s)
Antivirales/farmacología , Antivirales/uso terapéutico , Infecciones por Herpesviridae/tratamiento farmacológico , Herpesviridae/efectos de los fármacos , Nucleósidos de Pirimidina/farmacología , Nucleósidos de Pirimidina/uso terapéutico , Replicación Viral/efectos de los fármacos , Animales , Antivirales/efectos adversos , Antivirales/química , Línea Celular , Proliferación Celular/efectos de los fármacos , Células Cultivadas , ADN Polimerasa Dirigida por ADN/genética , ADN Polimerasa Dirigida por ADN/fisiología , Farmacorresistencia Viral/efectos de los fármacos , Farmacorresistencia Viral/genética , Técnica del Anticuerpo Fluorescente Indirecta , Herpesviridae/genética , Infecciones por Herpesviridae/virología , Herpesvirus Humano 1/efectos de los fármacos , Herpesvirus Humano 1/genética , Herpesvirus Humano 2/efectos de los fármacos , Herpesvirus Humano 2/genética , Herpesvirus Humano 6/efectos de los fármacos , Herpesvirus Humano 6/genética , Herpesvirus Humano 8/efectos de los fármacos , Herpesvirus Humano 8/genética , Humanos , Ratones , Ratones Endogámicos BALB C , Microscopía Fluorescente , Estructura Molecular , Nucleósidos de Pirimidina/síntesis química , Nucleósidos de Pirimidina/química , Proteínas Virales/genética , Proteínas Virales/fisiología
11.
Antiviral Res ; 83(3): 282-9, 2009 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19576248

RESUMEN

Herpes simplex virus types 1 and 2 (HSV-1, HSV-2) infections are common, but can cause serious infections in neonates and the immunocompromised. Drugs currently used to treat cutaneous or genital HSV infections are effective in limiting disease, but the emergence of drug resistant viruses in immunocompromised individuals can be problematic. While the prophylactic oral treatment with antiviral drugs can reduce virus shedding and transmission, there is a need for topical microbicides that have the potential to limit sexual transmission of the virus. Previous reports demonstrated the antiviral activity of complex sulfated polysaccharides extracted from various species of marine algae and suggested that they interfered with the attachment of virions to host cells. Here, we evaluated the antiviral activity of extracts from Undaria pinnatifida, Splachnidium rugosum, Gigartina atropurpurea, and Plocamium cartilagineum against HSV-1 and HSV-2. These extracts exhibited good activity when added during the first hour of viral infection, but were ineffective if added later. Plaque reduction assays, when the extracts were added prior to viral inoculation, yielded EC(50) values that ranged from 2.5-3.6 microg/ml for HSV-1 and 0.7-6.6 microg/ml for HSV-2. None of the extracts exhibited significant toxicity in a neutral red uptake assay (IC(50) >100 microg/ml). Subsequent assays showed that the compounds had potent virucidal activity and were active at very low concentrations. We conclude that these extracts are nontoxic and effective virucidal agents that warrant further investigation to examine their potential role in the prevention of HSV infections of humans.


Asunto(s)
Antivirales/aislamiento & purificación , Antivirales/farmacología , Eucariontes/química , Herpesvirus Humano 1/efectos de los fármacos , Herpesvirus Humano 2/efectos de los fármacos , Polisacáridos/aislamiento & purificación , Polisacáridos/farmacología , Células Cultivadas , Fibroblastos/virología , Humanos , Concentración 50 Inhibidora , Pruebas de Sensibilidad Microbiana , Ensayo de Placa Viral , Acoplamiento Viral/efectos de los fármacos
14.
Antimicrob Agents Chemother ; 53(2): 572-9, 2009 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19029322

RESUMEN

As part of a program to identify new compounds that have activity against orthopoxviruses, a number of 4'-thionucleosides were synthesized and evaluated for their efficacies against vaccinia and cowpox viruses. Seven compounds that were active at about 1 microM against both viruses in human cells but that did not have significant toxicity were identified. The 5-iodo analog, 1-(2-deoxy-4-thio-beta-d-ribofuranosyl)-5-iodouracil (4'-thioIDU), was selected as a representative molecule; and this compound also inhibited viral DNA synthesis at less than 1 microM but only partially inhibited the replication of a recombinant vaccinia virus that lacked a thymidine kinase. This compound retained complete activity against cidofovir- and ST-246-resistant mutants. To determine if this analog had activity in an animal model, mice were infected intranasally with vaccinia or cowpox virus and treatment with 4'-thioIDU was given intraperitoneally or orally twice daily at 50, 15, 5, or 1.5 mg/kg of body weight beginning at 24 to 120 h postinfection and was continued for 5 days. Almost complete protection (87%) was observed when treatment with 1.5 mg/kg was begun at 72 h postinfection, and significant protection (73%) was still obtained when treatment with 5 mg/kg was initiated at 96 h. Virus titers in the liver, spleen, and kidney were reduced by about 4 log(10) units and about 2 log(10) units in mice infected with vaccinia virus and cowpox virus, respectively. These results indicate that 4'-thioIDU is a potent, nontoxic inhibitor of orthopoxvirus replication in cell culture and experimental animal infections and suggest that it may have potential for use in the treatment of orthopoxvirus infections in animals and humans.


Asunto(s)
Antivirales/uso terapéutico , Nucleósidos/uso terapéutico , Orthopoxvirus , Infecciones por Poxviridae/tratamiento farmacológico , Animales , Línea Celular , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Viruela Vacuna/tratamiento farmacológico , Efecto Citopatogénico Viral/efectos de los fármacos , ADN Viral/biosíntesis , ADN Viral/genética , Farmacorresistencia Viral , Humanos , Ratones , Ratones Endogámicos BALB C , Infecciones por Poxviridae/virología , Relación Estructura-Actividad , Vaccinia/tratamiento farmacológico , beta-Galactosidasa/metabolismo
15.
Antimicrob Agents Chemother ; 52(12): 4326-30, 2008 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-18852272

RESUMEN

Patients infected with human immunodeficiency virus (HIV) often suffer from herpesvirus infections as a result of immunosuppression. These infections can occur while patients are receiving antiretroviral therapy, and additional drugs required to treat their infection can adversely affect compliance. It would be useful to have antivirals with a broader spectrum of activity that included both HIV and the herpesviruses. We reported previously that alkoxyalkyl ester prodrugs of cidofovir are up to 3 orders of magnitude more active against herpesvirus replication and may be less toxic than the unmodified drug. To determine if this strategy would be effective for certain phosphonomethoxyethyl nucleoside phosphonates which are also active against HIV infections, the hexadecyloxypropyl (HDP) esters of 1-(phosphonomethoxyethyl)-cytosine, 1-(phosphonomethoxyethyl)-5-bromo-cytosine (PME-5BrC), 1-(phosphonomethoxyethyl)-5-fluoro-cytosine, 9-(phosphonomethoxyethyl)-2,6-diaminopurine (PME-DAP), and 9-(phosphonomethoxyethyl)-2-amino-6-cyclopropylaminopurine (PME-cPrDAP) were evaluated for activity against herpesvirus replication. The HDP esters were substantially more active than the unmodified acyclic nucleoside phosphonates, indicating that esterification with alkoxyalkyl groups increases the antiviral activity of many acyclic nucleoside phosphonates. The most interesting compounds included HDP-PME-cPrDAP and HDP-PME-DAP, which were 12- to 43-fold more active than the parent nucleoside phosphonates against herpes simplex virus and cytomegalovirus, and HDP-PME-cPrDAP and HDP-PME-5BrC which were especially active against Epstein-Barr virus. The results presented here indicate that HDP-esterified acyclic nucleoside phosphonates with antiviral activity against HIV also inhibit the replication of some herpesviruses and can extend the spectrum of activity for these compounds.


Asunto(s)
Antivirales/farmacología , Ésteres/farmacología , Herpesviridae/efectos de los fármacos , Organofosfonatos/farmacología , Nucleósidos de Purina/farmacología , Nucleósidos de Pirimidina/farmacología , Replicación Viral/efectos de los fármacos , Antivirales/química , Línea Celular , Ésteres/química , Herpesviridae/clasificación , Herpesviridae/fisiología , Humanos , Organofosfonatos/química , Nucleósidos de Purina/química , Nucleósidos de Pirimidina/química , Ensayo de Placa Viral
16.
Antiviral Res ; 80(2): 223-4, 2008 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-18573279

RESUMEN

These studies were performed to determine the effect of AD-472, an attenuated human herpes simplex virus (HSV) type 2 or HSV-2 glycoprotein D (gD) when combined with an adjuvant, GPI-0100, a semi-synthetic Quillaja Saponin analog in a genital HSV-2 infection in guinea pigs. While animals immunized with either vaccine had reduced clinical disease, GPI-0100 only improved the efficacy of gD and did not affect the efficacy of the live vaccine. Neither vaccine had any therapeutic effect if administered 24 h after viral infection.


Asunto(s)
Adyuvantes Inmunológicos , Herpes Genital/prevención & control , Vacunas contra el Virus del Herpes Simple/inmunología , Herpesvirus Humano 2/inmunología , Saponinas/inmunología , Vacunación , Vacunas Atenuadas/inmunología , Proteínas del Envoltorio Viral/inmunología , Animales , Femenino , Cobayas , Herpes Genital/inmunología , Humanos
17.
Virol J ; 5: 58, 2008 May 14.
Artículo en Inglés | MEDLINE | ID: mdl-18479513

RESUMEN

BACKGROUND: The emergence of drug resistant viruses, together with the possibility of increased virulence, is an important concern in the development of new antiviral compounds. Cidofovir (CDV) is a phosphonate nucleotide that is approved for use against cytomegalovirus retinitis and for the emergency treatment of smallpox or complications following vaccination. One mode of action for CDV has been demonstrated to be the inhibition of the viral DNA polymerase. RESULTS: We have isolated several CDV resistant (CDVR) vaccinia viruses through a one step process, two of which have unique single mutations within the DNA polymerase. An additional resistant virus isolate provides evidence of a second site mutation within the genome involved in CDV resistance. The CDVR viruses were 3-7 fold more resistant to the drug than the parental viruses. The virulence of the CDVR viruses was tested in mice inoculated intranasally and all were found to be attenuated. CONCLUSION: Resistance to CDV in vaccinia virus can be conferred individually by at least two different mutations within the DNA polymerase gene. Additional genes may be involved. This one step approach for isolating resistant viruses without serial passage and in the presence of low doses of drug minimizes unintended secondary mutations and is applicable to other potential antiviral agents.


Asunto(s)
Citosina/análogos & derivados , Farmacorresistencia Viral , Organofosfonatos/farmacología , Virus Vaccinia/efectos de los fármacos , Virus Vaccinia/aislamiento & purificación , Animales , Antivirales/farmacología , Línea Celular , Chlorocebus aethiops , Cidofovir , Citosina/farmacología , ADN Polimerasa Dirigida por ADN/química , ADN Polimerasa Dirigida por ADN/genética , ADN Polimerasa Dirigida por ADN/metabolismo , Femenino , Humanos , Ratones , Modelos Moleculares , Mutación , Vaccinia/mortalidad , Vaccinia/virología , Virus Vaccinia/patogenicidad , Virus Vaccinia/fisiología , Células Vero , Ensayo de Placa Viral , Virulencia
18.
Antimicrob Agents Chemother ; 52(8): 2727-33, 2008 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-18505857

RESUMEN

Phosphorothioated oligonucleotides have a sequence-independent antiviral activity as amphipathic polymers (APs). The activity of these agents against herpesvirus infections in vitro and in vivo was investigated. The previously established sequence-independent, phosphorothioation-dependent antiviral activity of APs was confirmed in vitro by showing that a variety of equivalently sized homo- and heteropolymeric AP sequences were similarly active against herpes simplex virus type 1 (HSV-1) infection in vitro compared to the 40mer degenerate parent compound (REP 9), while the absence of phosphorothioation resulted in the loss of antiviral activity. In addition, REP 9 demonstrated in vitro activity against a broad spectrum of other herpesviruses: HSV-2 (50% effective concentration [EC(50)], 0.02 to 0.06 microM), human cytomegalovirus (EC(50), 0.02 to 0.13 microM), varicella zoster virus (EC(50), <0.02 microM), Epstein-Barr virus (EC(50), 14.7 microM) and human herpesvirus types 6A/B (EC(50), 2.9 to 10.2 microM). The murine microbicide model of genital HSV-2 was then used to evaluate in vivo activity. REP 9 (275 mg/ml) protected 75% of animals from disease and infection when provided 5 or 30 min prior to vaginal challenge. When an acid-stable analog (REP 9C) was used, 75% of mice were protected when treated with 240 mg/ml 5 min prior to infection (P < 0.001), while a lower dose (100 mg/ml) protected 100% of the mice (P < 0.001). The acid stable REP 9C formulation also provided protection at 30 min (83%, P < 0.001) and 60 min (50%, P = 0.07) against disease. These observations suggest that APs may have microbicidal activity and potential as broad-spectrum antiherpetic agents and represent a novel class of agents that should be studied further.


Asunto(s)
Antivirales/farmacología , ADN/farmacología , Herpes Genital/prevención & control , Animales , Antivirales/síntesis química , Antivirales/química , Chlorocebus aethiops , Citomegalovirus/efectos de los fármacos , ADN/síntesis química , ADN/química , Estabilidad de Medicamentos , Femenino , Herpes Genital/virología , Herpesvirus Humano 1/efectos de los fármacos , Herpesvirus Humano 2/efectos de los fármacos , Humanos , Concentración de Iones de Hidrógeno , Ratones , Pruebas de Sensibilidad Microbiana , Estructura Molecular , Células Vero
19.
Virol J ; 5: 39, 2008 Mar 05.
Artículo en Inglés | MEDLINE | ID: mdl-18321387

RESUMEN

BACKGROUND: The vaccinia virus (VV) F2L gene encodes a functional deoxyuridine triphosphatase (dUTPase) that catalyzes the conversion of dUTP to dUMP and is thought to minimize the incorporation of deoxyuridine residues into the viral genome. Previous studies with with a complex, multigene deletion in this virus suggested that the gene was not required for viral replication, but the impact of deleting this gene alone has not been determined in vitro or in vivo. Although the crystal structure for this enzyme has been determined, its potential as a target for antiviral therapy is unclear. RESULTS: The F2L gene was replaced with GFP in the WR strain of VV to assess its effect on viral replication. The resulting virus replicated well in cell culture and its replication kinetics were almost indistinguishable from those of the wt virus and attained similar titers. The virus also appeared to be as pathogenic as the WR strain suggesting that it also replicated well in mice. Cells infected with the dUTPase mutant would be predicted to affect pyrimidine deoxynucleotide pools and might be expected to exhibit altered susceptibility to pyrimidine analogs. The antiviral activity of cidofovir and four thymidine analogs were evaluated both in the mutant and the parent strain of this virus. The dUTPase knockout remained fully susceptible to cidofovir and idoxuridine, but was hypersensitive to the drug (N)-methanocarbathymidine, suggesting that pyrimidine metabolism was altered in cells infected with the mutant virus. The absence of dUTPase should reduce cellular dUMP pools and may result in a reduced conversion to dTMP by thymidylate synthetase or an increased reliance on the salvage of thymidine by the viral thymidine kinase. CONCLUSION: We confirmed that F2L was not required for replication in cell culture and determined that it does not play a significant role on virulence of the virus in intranasally infected mice. The recombinant virus is hypersensitive to (N)-methanocarbathymidine and may reflect metabolic differences in the mutant virus.


Asunto(s)
Antivirales/farmacología , Pirofosfatasas/metabolismo , Timidina/análogos & derivados , Virus Vaccinia/fisiología , Vaccinia/virología , Replicación Viral/efectos de los fármacos , Animales , Femenino , Proteínas Fluorescentes Verdes/metabolismo , Humanos , Ratones , Ratones Endogámicos BALB C , Pirimidinas/metabolismo , Pirofosfatasas/genética , Proteínas Recombinantes de Fusión/metabolismo , Eliminación de Secuencia , Timidina/farmacología , Virus Vaccinia/efectos de los fármacos , Virus Vaccinia/enzimología , Virus Vaccinia/patogenicidad , Ensayo de Placa Viral , Virulencia
20.
J Virol ; 82(10): 5054-67, 2008 May.
Artículo en Inglés | MEDLINE | ID: mdl-18321963

RESUMEN

Cells infected with human cytomegalovirus in the absence of UL97 kinase activity produce large nuclear aggregates that sequester considerable quantities of viral proteins. A transient expression assay suggested that pp71 and IE1 were also involved in this process, and this suggestion was significant, since both proteins have been reported to interact with components of promyelocytic leukemia (PML) bodies (ND10) and also interact functionally with retinoblastoma pocket proteins (RB). PML bodies have been linked to the formation of nuclear aggresomes, and colocalization studies suggested that viral proteins were recruited to these structures and that UL97 kinase activity inhibited their formation. Proteins associated with PML bodies were examined by Western blot analysis, and pUL97 appeared to specifically affect the phosphorylation of RB in a kinase-dependent manner. Three consensus RB binding motifs were identified in the UL97 kinase, and recombinant viruses were constructed in which each was mutated to assess a potential role in the phosphorylation of RB and the inhibition of nuclear aggresome formation. The mutation of either the conserved LxCxE RB binding motif or the lysine required for kinase activity impaired the ability of the virus to stabilize and phosphorylate RB. We concluded from these studies that both UL97 kinase activity and the LxCxE RB binding motif are required for the phosphorylation and stabilization of RB in infected cells and that this effect can be antagonized by the antiviral drug maribavir. These data also suggest a potential link between RB function and the formation of aggresomes.


Asunto(s)
Citomegalovirus/fisiología , Fosfotransferasas (Aceptor de Grupo Alcohol)/metabolismo , Proteína de Retinoblastoma/metabolismo , Secuencia de Aminoácidos , Animales , Sitios de Unión , Línea Celular , Núcleo Celular/química , Chlorocebus aethiops , Cromatografía Liquida , Secuencia Conservada , Citomegalovirus/genética , Citoplasma/química , Humanos , Espectrometría de Masas , Microscopía Fluorescente , Datos de Secuencia Molecular , Fosforilación , Fosfotransferasas (Aceptor de Grupo Alcohol)/genética , Unión Proteica , Proteínas/aislamiento & purificación , Alineación de Secuencia
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...