Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 84
Filtrar
1.
Gene Ther ; 21(11): 984-90, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25119379

RESUMO

Limited expression and distribution of nectin-1, the major herpes simplex virus (HSV) type-1 entry-receptor, within tumors has been proposed as an impediment to oncolytic HSV (oHSV) therapy. To determine whether resistance to oHSVs in malignant peripheral nerve sheath tumors (MPNSTs) was explained by this hypothesis, nectin-1 expression and oHSV viral yields were assessed in a panel of MPNST cell lines using γ134.5-attenuated (Δγ134.5) oHSVs and a γ134.5 wild-type (wt) virus for comparison. Although there was a correlation between nectin-1 levels and viral yields with the wt virus (R=0.75, P =0.03), there was no correlation for Δγ134.5 viruses (G207, R7020 or C101) and a modest trend for the second-generation oHSV C134 (R=0.62, P=0.10). Nectin-1 overexpression in resistant MPNST cell lines did not improve Δγ134.5 oHSV output. While multistep replication assays showed that nectin-1 overexpression improved Δγ134.5 oHSV cell-to-cell spread, it did not confer a sensitive phenotype to resistant cells. Finally, oHSV yields were not improved with increased nectin-1 in vivo. We conclude that nectin-1 expression is not the primary obstacle of productive infection for Δγ134.5 oHSVs in MPNST cell lines. In contrast, viruses that are competent in their ability to counter the antiviral response may derive benefit with higher nectin-1 expression.


Assuntos
Moléculas de Adesão Celular/metabolismo , Neoplasias de Bainha Neural/metabolismo , Vírus Oncolíticos/fisiologia , Receptores Virais/metabolismo , Simplexvirus/fisiologia , Animais , Linhagem Celular Tumoral , Chlorocebus aethiops , Cricetulus , Humanos , Camundongos , Nectinas , Neoplasias de Bainha Neural/virologia , Terapia Viral Oncolítica , Vírus Oncolíticos/metabolismo
2.
Cancer Gene Ther ; 14(1): 45-56, 2007 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-16990846

RESUMO

Lack of effective therapy of primary brain tumors has promoted the development of novel experimental approaches utilizing oncolytic viruses combined with gene therapy. Towards this end, we have assessed a conditionally replication-competent, gamma(1)34.5-deleted herpes simplex virus type 1 (HSV-1) expressing cytosine deaminase (CD) for treatment of malignant brain tumors. Our results are summarized as follows: (i) a recombinant HSV (M012) was constructed in which both copies of the gamma(1)34.5 gene were replaced with the bacterial CD gene, under the control of the cellular promoter Egr-1; (ii) M012-infected cells in vitro efficiently convert 5-fluorocytosine (5-FC) to 5-fluorouracil, thereby enhancing cytotoxicity of neighboring, uninfected cells; (iii) both direct and bystander cytotoxicity of murine neuroblastoma and human glioma cell lines after infection with M012 were demonstrated; (iv) direct intracerebral inoculation of A/J mice demonstrated lack of neurotoxicity at doses similar to G207, a gamma(1)34.5-deleted HSV with demonstrated safety in human patient trials and (v) intratumoral injection of M012 into Neuro-2a flank tumors in combination with 5-FC administration significantly reduced tumor growth versus tumors treated with R3659 combined with 5-FC, or treated with M012 alone. Thus, M012 is a promising new oncolytic HSV vector with an enhanced prodrug-mediated, antineoplastic effect that is safe for intracranial administration.


Assuntos
Bactérias/enzimologia , Neoplasias Encefálicas/terapia , Citosina Desaminase/genética , Simplexvirus/genética , Animais , Neoplasias Encefálicas/tratamento farmacológico , Chlorocebus aethiops , Feminino , Fluoruracila/uso terapêutico , Engenharia Genética , Humanos , Camundongos , Células Vero
3.
Antiviral Res ; 63 Suppl 1: S11-8, 2004 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-15450381

RESUMO

Surrogate markers are an important component in the process of investigating management and prevention strategies, and for increasing understanding of viral diseases. The importance of surrogate markers and applied statistical models is particularly true for HIV. For HIV infection, the development of such methods provides new approaches for evaluation of HIV therapies and vaccines, and for the study of HIV transmission and its pathogenesis. The complex natural history of hepatitis B infection demonstrates that viral load is not the only predictor of transmission of this virus; for hepatitis C infection, viral load per se is not a prognostic factor for disease progression, but cumulative viral load may affect the outcome, and therapy is aimed at eliminating active viral replication.


Assuntos
Infecções por HIV/transmissão , HIV/fisiologia , Hepacivirus/fisiologia , Vírus da Hepatite B/fisiologia , Hepatite B/transmissão , Hepatite C/transmissão , Biomarcadores , Contagem de Linfócito CD4 , Transmissão de Doença Infecciosa/prevenção & controle , Infecções por HIV/tratamento farmacológico , Infecções por HIV/epidemiologia , Infecções por HIV/virologia , Hepatite B/tratamento farmacológico , Hepatite B/epidemiologia , Hepatite B/virologia , Hepatite C/dietoterapia , Hepatite C/epidemiologia , Hepatite C/prevenção & controle , Hepatite C/virologia , Humanos , Transmissão Vertical de Doenças Infecciosas/prevenção & controle , Carga Viral , Replicação Viral
4.
Clin Microbiol Infect ; 8(9): 551-63, 2002 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-12427216

RESUMO

Herpes simplex virus-1 (HSV-1) is a relatively large double-stranded DNA virus encoding at least 89 proteins with well characterized disease pathology. An understanding of the functions of viral proteins together with the ability to genetically engineer specific viral mutants has led to the development of attenuated HSV-1 for gene therapy. This review highlights the progress in creating attenuated genetically engineered HSV-1 mutants that are either replication competent (viral non-essential gene deleted) or replication defective (viral essential gene deleted). The choice between a replication-competent or -defective virus is based on the end-goal of the therapeutic intervention. Replication-competent HSV-1 mutants have primarily been employed as antitumor oncolytic viruses, with the lytic nature of the virus harnessed to destroy tumor cells selectively. In replacement gene therapy, replication-defective viruses have been utilized as delivery vectors. The advantages of HSV-1 vectors are that they infect quiescent and dividing cells efficiently and can encode for relatively large transgenes.


Assuntos
Terapia Genética/métodos , Vetores Genéticos , Neoplasias/terapia , Simplexvirus/genética , Antineoplásicos/uso terapêutico , Ensaios Clínicos Fase I como Assunto , Terapia Combinada , Vírus Defeituosos/genética , Técnicas de Transferência de Genes , Humanos , Neoplasias/tratamento farmacológico , Neoplasias/radioterapia , Simplexvirus/enzimologia , Transcrição Gênica , Replicação Viral
5.
Gene Ther ; 9(1): 75-80, 2002 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-11850725

RESUMO

The herpes simplex virus (HSV) recombinant virus R7020 is an attenuated virus designed as a candidate for immunization against both HSV-1 and HSV-2 infections. It was extensively tested in an experimental animal system and in a healthy human adult population without significant untoward effects. We report on the use of R7020 with ionizing radiation as an oncolytic agent for hepatomas. Two hepatoma cell lines were studied, Hep3B and Huh7. R7020 replicated to higher titers in Hep3B cells than in Huh7 cells. Tissue culture studies correlated with hepatoma xenograft responses to R7020. R7020 was more effective in mediating Hep3B tumor xenograft regression compared with Huh7. Ionizing radiation combined with R7020 also showed differential results in antitumor efficacy between the two cell lines in tumor xenografts. Ionizing radiation enhanced the replication of R7020 in Hep3B xenografts. Moreover, the combination of ionizing radiation and virus caused a greater regression of xenograft volume than either R7020 or radiation alone. Ionizing radiation had no effect on the replication of R7020 virus in Huh7 xenografts. These results indicate that a regimen involving infection with an appropriate herpesvirus such as R7020 in combination with ionizing radiation can be highly effective in eradicating certain tumor xenografts.


Assuntos
Terapia Genética/métodos , Vacinas contra o Vírus do Herpes Simples/administração & dosagem , Herpesvirus Humano 1 , Herpesvirus Humano 2 , Neoplasias Hepáticas Experimentais/terapia , Animais , Terapia Combinada , Humanos , Neoplasias Hepáticas Experimentais/radioterapia , Camundongos , Camundongos Nus , Transplante de Neoplasias , Células Tumorais Cultivadas , Replicação Viral/efeitos da radiação
6.
Herpes ; 8(1): 17-22, 2001 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-11867012

RESUMO

Central nervous system malignancies--particularly glioblastoma multiforme--pose significant problems for the development of novel therapeutics. In the absence of advances with standard surgical and chemotherapeutic approaches, the utilization of genetically engineered viruses--both as direct oncolytic agents (virus therapy) and for the delivery of foreign proteins (gene therapy)--represents a significant advance in the experimental approach to the management of patients with incurable tumours. Among other viruses, herpes simplex virus (HSV) offers an opportunity to influence the replication of tumour cells directly within the central nervous system. The propensity for HSV to replicate in tumour cells, and its large coding capacity, provide an experimental model for the development of novel therapeutics. The status of these experimental approaches and Phase I studies are summarized.


Assuntos
Neoplasias do Sistema Nervoso Central/terapia , Terapia Genética , Glioma/terapia , Simplexvirus/genética , Animais , Neoplasias do Sistema Nervoso Central/virologia , Ensaios Clínicos como Assunto , Modelos Animais de Doenças , Engenharia Genética , Glioma/virologia , Humanos , Camundongos
7.
Rev Med Virol ; 10(1): 17-30, 2000.
Artigo em Inglês | MEDLINE | ID: mdl-10654002

RESUMO

Central nervous system malignancies, particularly glioblastoma multiforme, pose significant problems for the development of novel therapeutics. In the absence of advances with standard surgical and chemotherapeutic approaches, the utilisation of genetically engineered viruses, both as direct oncolytic agents as well as for the delivery of foreign proteins, represents a significant advance in the experimental approach to management of patients with these incurable tumours. Among other viruses, HSV offers an opportunity to directly influence the replication of tumour cells within the central nervous system. Because of its propensity to replicate in neuronal tissue as well as its large coding capacity, it provides an experimental model for the development of novel therapeutics. The status of these experimental approaches will be summarised in this review.


Assuntos
Neoplasias do Sistema Nervoso Central/terapia , Engenharia Genética , Glioma/terapia , Simplexvirus/genética , Animais , Neoplasias do Sistema Nervoso Central/virologia , Ensaios Clínicos como Assunto , Terapia Genética , Vetores Genéticos , Glioma/virologia , Humanos , Simplexvirus/fisiologia
8.
Proc Natl Acad Sci U S A ; 97(5): 2208-13, 2000 Feb 29.
Artigo em Inglês | MEDLINE | ID: mdl-10681459

RESUMO

Genetically engineered, neuroattenuated herpes simplex viruses (HSVs) expressing various cytokines can improve survival when used in the treatment of experimental brain tumors. These attenuated viruses have both copies of gamma(1)34.5 deleted. Recently, we demonstrated increased survival of C57BL/6 mice bearing syngeneic GL-261 gliomas when treated with an engineered HSV expressing IL-4, as compared with treatment with the parent construct (gamma(1)34. 5(-)) alone or with a virus expressing IL-10. Herein, we report construction of a conditionally replication-competent mutant expressing both subunits of mIL-12 (M002) and its evaluation in a syngeneic neuroblastoma murine model. IL-12 induces a helper T cell subset type 1 response, which may induce more durable antitumor effects. In vitro studies showed that, when infected with M002, both Vero cells and murine Neuro-2a neuroblastoma cells produced physiologically relevant levels of IL-12 heterodimers, as determined by ELISA. M002 was cytotoxic for Neuro-2a cells and human glioma cell lines U251MG and D54MG. Neurotoxicity studies, as defined by plaque-forming units/LD(50), performed in HSV-1-sensitive A/J strain mice found that M002 was not toxic even at high doses. When evaluated in an intracranial syngeneic neuroblastoma murine model, median survival of M002-treated animals was significantly longer than the median survival of animals treated with R3659, the parent gamma(1)34.5(-) mutant lacking any cytokine gene insert. Immunohistochemical analysis of M002-treated tumors identified a pronounced influx of CD4(+) T cells and macrophages as well as CD8(+) cells when compared with an analysis of R3659-treated tumors. We conclude that M002 produced a survival benefit via oncolytic effects combined with immunologic effects meditated by helper T cells of subset type 1.


Assuntos
Neoplasias Encefálicas/terapia , Vetores Genéticos , Glioma/terapia , Herpesvirus Humano 1 , Interleucina-12/genética , Neuroblastoma/terapia , Animais , Terapia Biológica , Neoplasias Encefálicas/imunologia , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD8-Positivos/imunologia , Chlorocebus aethiops , Modelos Animais de Doenças , Feminino , Expressão Gênica , Engenharia Genética , Vetores Genéticos/genética , Glioma/imunologia , Herpesvirus Humano 1/genética , Herpesvirus Humano 1/patogenicidade , Humanos , Interleucina-12/imunologia , Camundongos , Camundongos Endogâmicos A , Neuroblastoma/imunologia , Recombinação Genética , Células Tumorais Cultivadas , Células Vero , Virulência
9.
Clin Cancer Res ; 5(6): 1517-22, 1999 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-10389941

RESUMO

Malignant gliomas remain incurable with current interventions. Encouraging investigational approaches include the use of genetically modified herpes simplex-1 (HSV-1) viruses as direct cytotoxic agents. Combining attenuated HSV-1 with standard therapy, human U-87 malignant glioma xenografts grown in the hind limb or intracranially in athymic nude mice were exposed to ionizing radiation, inoculated with genetically modified HSV R3616, or received both virus and radiation. The combination of virus with fractionated ionizing radiation suggests a synergistic action and results in reduced tumor volumes and longer survivals when compared with treatment with either modality alone.


Assuntos
Neoplasias Encefálicas/terapia , Vacinas Anticâncer/virologia , Glioma/terapia , Herpesvirus Humano 1 , Animais , Neoplasias Encefálicas/mortalidade , Neoplasias Encefálicas/radioterapia , Neoplasias Encefálicas/virologia , Vacinas Anticâncer/uso terapêutico , Terapia Combinada , Feminino , Glioma/mortalidade , Glioma/radioterapia , Glioma/virologia , Humanos , Imuno-Histoquímica , Camundongos , Camundongos Nus , Transplante de Neoplasias , Distribuição Aleatória , Neoplasias de Tecidos Moles/mortalidade , Neoplasias de Tecidos Moles/radioterapia , Neoplasias de Tecidos Moles/terapia , Neoplasias de Tecidos Moles/virologia , Taxa de Sobrevida , Células Tumorais Cultivadas , Raios X
11.
Cancer Res ; 59(9): 2055-8, 1999 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-10232586

RESUMO

A genetically engineered, nonneurotropic herpes simplex virus (R7020) with a proven safety profile in both animals and humans was found effective in the treatment of large xenotransplanted tumors arising from a radiation- and chemotherapy-resistant human epidermoid carcinoma and a hormone-refractory prostate adenocarcinoma. R7020 replicated to high titer and caused rapid regression of the human tumor xenografts. Tumor destruction was accelerated in animals given both R7020 and fractionated ionizing radiation. Tumors arising from cells surviving one treatment with R7020 were fully susceptible to a second dose of virus. We conclude R7020 is an effective antitumor agent for non-central nervous system tumor xenografts with an excellent safety profile.


Assuntos
Adenocarcinoma/terapia , Carcinoma de Células Escamosas/terapia , Neoplasias da Próstata/terapia , Simplexvirus/fisiologia , Adenocarcinoma/genética , Adenocarcinoma/radioterapia , Animais , Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/radioterapia , Fracionamento da Dose de Radiação , Resistencia a Medicamentos Antineoplásicos , Regulação Viral da Expressão Gênica/efeitos da radiação , Genes p53 , Engenharia Genética , Humanos , Injeções Intralesionais , Masculino , Camundongos , Camundongos Nus , Neoplasias da Próstata/genética , Neoplasias da Próstata/radioterapia , Tolerância a Radiação , Simplexvirus/genética , Transplante Heterólogo , Replicação Viral
13.
J Clin Microbiol ; 36(8): 2229-34, 1998 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-9665997

RESUMO

The purpose of the present study was to determine if the quantity of herpes simplex virus (HSV) DNA in the cerebrospinal fluid (CSF) of patients with herpes encephalitis would be useful in establishing the prognosis of the disease and to determine the effect of antiviral therapy on the clearance of viral DNA from the CSF. Quantitation of HSV DNA was done by constructing an internal standard (IS) from the glycoprotein B amplicon which had a 25-bp deletion between primer annealing sites. Each CSF specimen was coamplified with the IS and the ratio of the amount of HSV/amount of IS was compared to the ratios on a standard curve constructed with the same IS plus known amounts of HSV DNA. CSF specimens were available from 16 patients who were treated with intravenous acyclovir, and the amount of HSV DNA ranged from < 25 to 18,000 copies per microliter in CSF obtained before or within 4 days of the initiation of acyclovir therapy. Patients with > 100 copies of HSV DNA per microliter were older, were found by computed tomography to have lesions, and had poorer outcomes than patients with < 100 copies. Follow-up CSF specimens were available from seven patients. In six of these seven patients, the HSV DNA levels decreased during therapy. One patient had a twofold increase in HSV DNA levels after 1 week of therapy and died on day 8. The application of this assay may be helpful in establishing the prognosis and in the monitoring of patients with herpes simplex encephalitis.


Assuntos
Líquido Cefalorraquidiano/virologia , DNA Viral/líquido cefalorraquidiano , Encefalite Viral/diagnóstico , Herpes Simples/diagnóstico , Herpesvirus Humano 1/isolamento & purificação , Reação em Cadeia da Polimerase/métodos , Aciclovir/uso terapêutico , Adolescente , Adulto , Antivirais/uso terapêutico , Sequência de Bases , Criança , Pré-Escolar , DNA Viral/análise , Encefalite Viral/líquido cefalorraquidiano , Encefalite Viral/tratamento farmacológico , Feminino , Herpes Simples/líquido cefalorraquidiano , Herpes Simples/tratamento farmacológico , Herpesvirus Humano 1/genética , Humanos , Lactente , Masculino , Pessoa de Meia-Idade , Dados de Sequência Molecular , Prognóstico , Fatores de Tempo , Resultado do Tratamento , Proteínas do Envelope Viral/genética
14.
Gene Ther ; 5(1): 121-30, 1998 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-9536273

RESUMO

This report describes a test of the hypothesis that the oncolytic effect of genetically engineered, replication competent herpes simplex viruses (HSV) depends both on cell destruction by the virus and an immune response to the tumor cells induced in an immunocompetent animal system. The oncolytic vector was a HSV recombinant virus in which both copies of the gamma 1 34.5 gene were replaced with the murine genes encoding the cytokine interleukin-4 (IL-4) or interleukin-10 (IL-10). The hypothesis predicted that if an immune response plays a role in survival following intratumoral treatment of tumor-bearing animals with HSV, expression of IL-4 should prolong survival whereas expression of IL-10 should reduce it. The results are that (1) these cytokines can be expressed by HSV in productively infected cells both in vitro and in vivo; (2) HSV-expressing IL-4 or IL-10 genes were able to infect and destroy glioma cells in vitro; (3) intracerebral inoculation of HSV expressing either IL-4 or IL-10 into syngeneic murine glioma GL-261 cells implanted in the brains of immunocompetent C57BL/6 mice produced dramatically opposite physiologic responses. The IL-4 HSV significantly prolonged survival of tumor bearers, whereas tumor-bearing mice that received the IL-10 HSV had a median survival that was identical to that of saline treated controls; (4) immunohistochemical analyses of mouse brains at 3 and 7 days after virus inoculation showed marked accumulation of inflammatory cells composed primarily of macrophages/microglia, with various proportions of CD8+ and CD4+ T cells, but few B lymphocytes. We conclude that the cytokines expressed from genes encoded in the viral genome influence HSV therapy of tumors and this is probably due to the host immune response. Thus, cytokine expression may be an important adjunct to tumor therapy utilizing genetically engineered HSV.


Assuntos
Neoplasias Encefálicas/terapia , Terapia Genética/métodos , Vetores Genéticos , Glioma/terapia , Interleucinas/genética , Simplexvirus , Animais , Expressão Gênica , Imuno-Histoquímica , Interleucina-10/genética , Interleucina-4/genética , Camundongos , Camundongos Endogâmicos C57BL
15.
Cancer Res ; 57(8): 1502-9, 1997 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-9108452

RESUMO

Earlier studies have shown that genetically engineered herpes simplex viruses (e.g., HSV-1) are effective in killing malignant tumor cells both in vitro and in various murine tumor models. This report focuses on a panel of five genetically engineered viral mutants of the gamma(1)34.5 gene, which was shown previously to cause reduction in viral replication and associated neurovirulence of HSV. These include R3616, which has both copies of gamma(1)34.5 deleted, R4009, which has a stop codon inserted after codon 28 in both copies of the gamma(1)34.5 gene, R849, which contains a lacZ gene inserted in place of the gamma(1)34.5, R908, which lacks 41 codons in frame after codon 72 of the gamma(1)34.5, and R939, which carries a stop codon precluding the translation of the COOH-terminal domain of the gamma(1)34.5 gene. We report the following: (a) all five mutant HSVs were avirulent in experimental animals but were cytotoxic for human tumor cells in vitro and in vivo; (b) the gamma(1)34.5- HSV replicated in human glioma cells almost as efficiently as wild-type HSV-1(F) based on replication assays, in situ hybridization for viral DNA, and expression of infected cell protein 27; (c) capacity of mutant HSVs to kill human cells derived from glioblastoma multiforme (CH-235MG, D-37MG, D-54MG, D-65MG, U-251MG, U-373MG, and SK-MG-1), anaplastic astrocytoma (Hs-683), anaplastic glioma (U-87MG and U-138MG), gliosarcoma (D-32GS), or normal human astrocytes demonstrated that glioma cells varied in their susceptibility to HSV-mediated cytotoxicity and that cultured astrocytes were two to three orders of magnitude less susceptible to killing than were malignant glia; and (d) scid mice, which received 0.5 or 5 x 10(6) plaque-forming units of R4009, either were coinoculated at the time of intracranial transplantation with 106 U251MG or D-54MG human glioma cells or received the cells intratumorally 5 days after tumor induction and experienced significant increases in median survivals, with no histopathological indication of an infectious encephalitic process. Genetically engineered gamma(1)34.5- HSV mutants appear to be a potentially safe biotherapeutic agent for experimental treatment of uniformly fatal malignant brain tumors.


Assuntos
Neoplasias Encefálicas/terapia , DNA Viral/biossíntese , Engenharia Genética , Glioma/terapia , Herpesvirus Humano 1/genética , Aciclovir/farmacologia , Animais , Antivirais/farmacologia , Astrócitos/patologia , Astrócitos/virologia , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/virologia , Chlorocebus aethiops , Efeito Citopatogênico Viral , Glioma/metabolismo , Glioma/virologia , Herpesvirus Humano 1/efeitos dos fármacos , Herpesvirus Humano 1/crescimento & desenvolvimento , Herpesvirus Humano 1/metabolismo , Humanos , Proteínas Imediatamente Precoces/metabolismo , Camundongos , Camundongos SCID , Transplante Heterólogo , Células Vero , Replicação Viral
16.
Proc Natl Acad Sci U S A ; 93(21): 11313-8, 1996 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-8876132

RESUMO

Due to lack of effective therapy, primary brain tumors are the focus of intense investigation of novel experimental approaches that use vectors and recombinant viruses. Therapeutic approaches have been both indirect, whereby vectors are used, or direct to allow for direct cell killing by the introduced virus. Genetically engineered herpes simplex viruses are currently being evaluated as an experimental approach to eradicate malignant human gliomas. Initial studies with gamma (1)34.5 mutants, R3616 (from which both copies of the gamma (1)34.5 gene have been deleted) and R4009 (a construct with two stop codons inserted into the gamma (1)34.5 gene), have been assessed. In a syngeneic scid mouse intracranial tumor model, recombinant herpes simplex virus can be experimentally used for the treatment of brain tumors. These viruses and additional engineered viruses were subsequently tested in human glioma cells both in vitro and in vivo. Using a xenogeneic scid mouse intracranial glioma model, R4009 therapy of established tumors significantly prolonged survival. Most importantly, long-term survival was achieved, with histologic evidence that R4009 eradicated intracranial tumors in this model. Furthermore, the opportunity to evaluate gamma (1)34.5 mutants that have enhanced oncolytic activity, e.g., R8309 where the carboxyl terminus of the gamma (1)34.5 gene has been replaced by the murine homologue, MyD116, are considered.


Assuntos
Neoplasias Encefálicas/terapia , Engenharia Genética , Terapia Genética/métodos , Vetores Genéticos , Glioma/terapia , Simplexvirus , Adenoviridae/genética , Animais , Neoplasias Encefálicas/mortalidade , Neoplasias Encefálicas/patologia , Sobrevivência Celular , Deleção de Genes , Genes Virais , Glioma/mortalidade , Glioma/patologia , Humanos , Camundongos , Camundongos SCID , Mutagênese Insercional , Retroviridae/genética , Simplexvirus/genética , Taxa de Sobrevida , Transplante Heterólogo , Transplante Isogênico
17.
Ann Pharmacother ; 30(9): 967-71, 1996 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-8876858

RESUMO

OBJECTIVE: To review the first generation of antiviral agents (e.g., idoxuridine, amantadine, vidarabine) that paralleled discovery of antineoplastic agents. DATA SOURCES: A MEDLINE search (1962 to 1996) of the English-language literature pertaining to antiviral agents was performed. DATA EXTRACTION: All articles were considered for this review. Pertinent references on antiviral therapy, as judged by the author, were selected. DATA SYNTHESIS: Acyclovir, the first second-generation antiviral agent, has a known selective mechanism of action and provides the model for development of future antiviral therapies. Despite the safety and clinical value of acyclovir, therapy does not prevent establishment of latency or decrease frequency of occurrences, resistance has been documented, and outcome is frequently poor. With the emergence of the HIV/AIDS epidemic, several antiretroviral agents have been developed and approved. However, none of the four available nucleoside analogs provides a cure. CONCLUSIONS: Viral resistance has emerged as an important component of antiviral therapy. Improved therapies for cytomegalovirus are needed. Several new therapies for herpes zoster, including prodrugs, are licensed or in Phase III clinical trials. Future directions include the use of molecular biologic techniques to identify enzymes unique to viral replication and to accelerate diagnosis of viral diseases.


Assuntos
Aciclovir/uso terapêutico , Antivirais/uso terapêutico , Infecções por Herpesviridae/tratamento farmacológico , Aciclovir/farmacologia , Antivirais/história , Antivirais/farmacologia , Avaliação de Medicamentos , Previsões , Infecções por HIV/tratamento farmacológico , História do Século XX , Humanos
19.
Clin Infect Dis ; 21 Suppl 1: S114-20, 1995 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-8547499

RESUMO

Herpesviruses are among the most common causes of infections of humans. Viruses in this family share the unique biological property of being able to establish latency and to recur. Furthermore, chronic excretion of virus is not uncommon. In the immunocompromised host, including persons with human immunodeficiency virus (HIV) infection, herpesvirus disease can be particularly severe, resulting in chronic, persistent, active infection and, in some cases, life-threatening disease. The most pathogenic of the herpesviruses in patients with AIDS include herpes simplex viruses, human cytomegalovirus, and varicella-zoster virus. Disease caused by Epstein-Barr virus, particularly opportunistic malignancies, has been recognized. A new herpesvirus that is associated with Kaposi's sarcoma was recently described. On the other hand, disease caused by human herpesviruses 6 and 7 in persons infected with HIV remains to be unequivocally recognized. Prevention of exposure to herpesviruses, disease, and recurrence requires different measures than those for some of the other opportunistic infections in HIV-infected patients; this is because herpesvirus disease develops in most of these individuals as a result of reactivation rather than primary infection. Thus, approaches to the prevention and control of herpesvirus infections must be individualized according to both the type of virus as well as the type of infection (i.e., primary or recurrent). We discuss recommended measures for the prevention and control of these infections.


Assuntos
Infecções por HIV/complicações , Infecções por Herpesviridae/epidemiologia , Herpes Simples , Infecções por Herpesviridae/prevenção & controle , Humanos , Incidência , Recidiva , Fatores de Risco , Estados Unidos/epidemiologia
20.
J Infect Dis ; 171(4): 857-63, 1995 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-7706811

RESUMO

Isolation of herpes simplex virus (HSV) from brain tissue after biopsy has been considered the reference standard for the diagnosis of herpes simplex encephalitis (HSE). During the evaluation of antiviral treatment of HSE, cerebrospinal fluid (CSF) was obtained from patients with clinical disease indicative of HSE who underwent diagnostic brain biopsy. HSV DNA was detected by polymerase chain reaction (PCR) in CSF of 53 (98%) of 54 patients with biopsy-proven HSE and was detected in all 18 CSF specimens obtained before brain biopsy from patients with proven HSE. Four of 19 CSF specimens were positive after 2 weeks of antiviral therapy. Positive results were found in 3 (6%) of 47 patients whose brain tissue was culture-negative. Detection of HSV DNA in the CSF correlated significantly with age and focal radiographic findings. Thus, PCR detection of HSV DNA should be the standard for diagnosis of HSE.


Assuntos
DNA Viral/líquido cefalorraquidiano , Encefalite Viral/diagnóstico , Herpes Simples/diagnóstico , Reação em Cadeia da Polimerase/métodos , Simplexvirus/isolamento & purificação , Sequência de Bases , Encéfalo/virologia , DNA Polimerase Dirigida por DNA/genética , Encefalite Viral/líquido cefalorraquidiano , Encefalite Viral/tratamento farmacológico , Herpes Simples/líquido cefalorraquidiano , Herpes Simples/tratamento farmacológico , Humanos , Dados de Sequência Molecular , Valor Preditivo dos Testes , Sensibilidade e Especificidade , Simplexvirus/genética , Vidarabina/uso terapêutico , Proteínas do Envelope Viral/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA