Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 54
Filtrar
1.
Transplant Proc ; 50(10): 4038-4041, 2018 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-30577310

RESUMEN

Intraoperative extracorporeal membrane oxygenation (ECMO) support, both venoarterial and venovenous (VV), have been used sparingly and with limited success in the setting of liver transplantation. Here, we report the successful use of VV-ECMO in the resuscitation and pulmonary bridging support after severe systemic inflammatory response in a combined liver and kidney transplant recipient who suffered primary nonfunction of both allografts. Where conventional ventilator maneuvers may prove ineffective, the implementation of VV-ECMO should be considered as a therapeutic option in limited, short-lived acute pulmonary injury.


Asunto(s)
Oxigenación por Membrana Extracorpórea/métodos , Trasplante de Hígado/efectos adversos , Síndrome de Dificultad Respiratoria/etiología , Síndrome de Dificultad Respiratoria/terapia , Lesión Pulmonar Aguda/etiología , Lesión Pulmonar Aguda/terapia , Humanos , Trasplante de Riñón/efectos adversos , Masculino , Persona de Mediana Edad , Complicaciones Posoperatorias/etiología , Complicaciones Posoperatorias/terapia , Reoperación
2.
Oncogene ; 36(35): 4963-4974, 2017 08 31.
Artículo en Inglés | MEDLINE | ID: mdl-28459464

RESUMEN

Bone morphogenetic protein (BMP) signaling exerts antitumor activities in glioblastoma; however, its precise mechanisms remain to be elucidated. Here, we demonstrated that the BMP type I receptor ALK-2 (encoded by the ACVR1 gene) has crucial roles in apoptosis induction of patient-derived glioma-initiating cells (GICs), TGS-01 and TGS-04. We also characterized a BMP target gene, Distal-less homeobox 2 (DLX2), and found that DLX2 promoted apoptosis and neural differentiation of GICs. The tumor-suppressive effects of ALK-2 and DLX2 were further confirmed in a mouse orthotopic transplantation model. Interestingly, valproic acid (VPA), an anti-epileptic compound, induced BMP2, BMP4, ACVR1 and DLX2 mRNA expression with a concomitant increase in phosphorylation of Smad1/5. Consistently, we showed that treatment with VPA induced apoptosis of GICs, whereas silencing of ALK-2 or DLX2 expression partially suppressed it. Our study thus reveals BMP-mediated inhibitory mechanisms for glioblastoma, which explains, at least in part, the therapeutic effects of VPA.


Asunto(s)
Receptores de Activinas Tipo I/metabolismo , Proteínas Morfogenéticas Óseas/metabolismo , Neoplasias Encefálicas/metabolismo , Glioma/metabolismo , Proteínas de Homeodominio/metabolismo , Células Madre Neoplásicas/metabolismo , Factores de Transcripción/metabolismo , Ácido Valproico/farmacología , Animales , Apoptosis/efectos de los fármacos , Apoptosis/fisiología , Neoplasias Encefálicas/tratamiento farmacológico , Neoplasias Encefálicas/patología , Diferenciación Celular/fisiología , Femenino , Glioma/tratamiento farmacológico , Glioma/patología , Células HEK293 , Xenoinjertos , Humanos , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Células Madre Neoplásicas/efectos de los fármacos , Células Madre Neoplásicas/patología , Fosforilación , Transducción de Señal/efectos de los fármacos , Transfección
3.
Oncogene ; 33(17): 2236-44, 2014 Apr 24.
Artículo en Inglés | MEDLINE | ID: mdl-23686309

RESUMEN

Increasing evidence suggests that brain tumors arise from the transformation of neural stem/precursor/progenitor cells. Much current research on human brain tumors is focused on the stem-like properties of glioblastoma. Here we show that anaplastic lymphoma kinase (ALK) and its ligand pleiotrophin are required for the self-renewal and tumorigenicity of glioblastoma stem cells (GSCs). Furthermore, we demonstrate that pleiotrophin is transactivated directly by SOX2, a transcription factor essential for the maintenance of both neural stem cells and GSCs. We speculate that the pleiotrophin-ALK axis may be a promising target for the therapy of glioblastoma.


Asunto(s)
Neoplasias Encefálicas/metabolismo , Carcinogénesis/metabolismo , Proteínas Portadoras/genética , Citocinas/genética , Glioblastoma/metabolismo , Células Madre Neoplásicas/metabolismo , Proteínas Tirosina Quinasas Receptoras/metabolismo , Quinasa de Linfoma Anaplásico , Animales , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/patología , Proteínas Portadoras/metabolismo , Proliferación Celular , Citocinas/metabolismo , Regulación Neoplásica de la Expresión Génica , Glioblastoma/genética , Glioblastoma/patología , Células HEK293 , Humanos , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Trasplante de Neoplasias , Células Madre Neoplásicas/patología , Células Madre Neoplásicas/fisiología , Factores de Transcripción SOXB1/metabolismo , Activación Transcripcional , Células Tumorales Cultivadas
4.
Oncogene ; 32(33): 3840-5, 2013 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-22964630

RESUMEN

Cancer stem cells are believed to be responsible for tumor initiation and development. Much current research on human brain tumors is focused on the stem-like properties of glioblastoma stem cells (GSCs). However, little is known about the molecular mechanisms of cell cycle regulation that discriminate between GSCs and differentiated glioblastoma cells. Here we show that cyclin D2 is the cyclin that is predominantly expressed in GSCs and suppression of its expression by RNA interference causes G1 arrest in vitro and growth retardation of GSCs xenografted into immunocompromised mice in vivo. We also demonstrate that the expression of cyclin D2 is suppressed upon serum-induced differentiation similar to what was observed for the cancer stem cell marker CD133. Taken together, our results demonstrate that cyclin D2 has a critical role in cell cycle progression and the tumorigenicity of GSCs.


Asunto(s)
Ciclo Celular/fisiología , Ciclina D2/metabolismo , Glioblastoma/metabolismo , Células Madre Neoplásicas/metabolismo , Animales , Línea Celular Tumoral , Citometría de Flujo , Glioblastoma/patología , Humanos , Immunoblotting , Inmunohistoquímica , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Células Madre Neoplásicas/patología , Análisis de Secuencia por Matrices de Oligonucleótidos , Interferencia de ARN , ARN Interferente Pequeño , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Trasplante Heterólogo
5.
Cell Death Dis ; 3: e395, 2012 Oct 04.
Artículo en Inglés | MEDLINE | ID: mdl-23034330

RESUMEN

In this study, the roles of p53 in impaired spermatogenic male germ cells of p53-deficient medaka were investigated by analyzing histological changes, and gene expressions of 42Sp50, Oct 4 and vitellogenin (VTG2) by RT-PCR or in situ hybridization in the testes. We found that a small number of oocyte-like cells (testis-ova) differentiated spontaneously in the cysts of type A and early type B spermatogonia in the p53-deficient testes, in contrast to the wild-type (wt) testes in which testis-ova were never found. Furthermore, ionizing radiation (IR) irradiation increased the number of testis-ova in p53-deficient testes, increased testis-ova size and proceeded up to the zygotene or pachytene stages of premature meiosis within 14 days after irradiation. However, 28 days after irradiation, almost all the testis-ova were eliminated presumably by p53-independent apoptosis, and spermatogenesis was restored completely. In the wt testis, IR never induced testis-ova differentiation. This is the first study to demonstrate the pivotal role of the p53 gene in the elimination of spontaneous testis-ova in testes, and that p53 is not indispensable for the restoration of spermatogenesis in the impaired testes in which cell cycle regulation is disturbed by IR irradiation.


Asunto(s)
Rayos gamma , Meiosis/efectos de la radiación , Espermatogonias/citología , Proteína p53 Supresora de Tumor/metabolismo , Animales , Diferenciación Celular/efectos de la radiación , Masculino , Factor 3 de Transcripción de Unión a Octámeros/metabolismo , Oryzias/crecimiento & desarrollo , Factores de Elongación de Péptidos/metabolismo , Espermatogénesis/efectos de la radiación , Espermatogonias/metabolismo , Espermatogonias/efectos de la radiación , Testículo/citología , Testículo/metabolismo , Testículo/patología , Proteína p53 Supresora de Tumor/deficiencia , Proteína p53 Supresora de Tumor/genética , Vitelogeninas/metabolismo , Proteínas de Xenopus/metabolismo
6.
Ann Oncol ; 23(11): 2914-2919, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22776705

RESUMEN

BACKGROUND: Epidermal growth factor receptor (EGFR) mutation is predictive for the efficacy of EGFR tyrosine kinase inhibitors in advanced non-small-cell lung cancer (NSCLC) treatment. We evaluated the performance, sensitivity, and concordance between five EGFR tests. MATERIALS AND METHODS: DNA admixtures (n = 34; 1%-50% mutant plasmid DNA) and samples from NSCLC patients [116 formalin-fixed paraffin-embedded (FFPE) tissue, 29 matched bronchofiberscopic brushing (BB) cytology, and 20 additional pleural effusion (PE) cytology samples] were analyzed. EGFR mutation tests were PCR-Invader, peptide nucleic acid-locked nucleic acid PCR clamp, direct sequencing, Cycleave, and Scorpion Amplification Refractory Mutation System (ARMS). Analysis success, mutation status, and concordance rates were assessed. RESULTS: All tests except direct sequencing detected four mutation types at ≥1% mutant DNA. Analysis success rates were 91.4%-100% (FFPE) and 100% (BB and PE cytology), respectively. Inter-assay concordance rates of successfully analyzed samples were 94.3%-100% (FFPE; kappa coefficients: 0.88-1.00), 93.1%-100% (BB cytology; 0.86-1.00), and 85.0%-100% (PE cytology; 0.70-1.00), and 93.1%-96.6% (0.86-0.93) between BB cytology and matched FFPE. CONCLUSIONS: All EGFR assays carried out comparably in the analysis of FFPE and cytology samples. Cytology-derived DNA is a viable alternative to FFPE samples for analyzing EGFR mutations.


Asunto(s)
Adenocarcinoma/diagnóstico , Carcinoma de Pulmón de Células no Pequeñas/diagnóstico , Análisis Mutacional de ADN/métodos , Receptores ErbB/genética , Neoplasias Pulmonares/diagnóstico , Adenocarcinoma/tratamiento farmacológico , Adenocarcinoma/genética , Antineoplásicos/uso terapéutico , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Carcinoma de Pulmón de Células no Pequeñas/genética , Receptores ErbB/antagonistas & inhibidores , Receptores ErbB/metabolismo , Humanos , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/genética , Mutación , Análisis de Secuencia de ADN
7.
Neoplasia ; 3(5): 451-6, 2001.
Artículo en Inglés | MEDLINE | ID: mdl-11687957

RESUMEN

Viral gene therapy against malignant tumors holds great promise for tumors that are susceptible to the oncolytic activity of viruses. One advantage of oncolytic viral therapy is that it can potentially be combined with other therapies, such as radiotherapy, to obtain an enhanced tumor response. In the case of prostate cancer, herpes simplex virus-mediated therapies have been shown to be highly effective in animal models; however, studies of the efficacy of combined viral and radiation therapy have not yet been reported. In this study, we have combined G207, a multimutated HSV type 1 vector, with external beam radiation therapy of prostate tumors grown subcutaneously in mice. We examined both the human LNCaP tumor in athymic mice and the mouse transgenic TRAMP tumor in either athymic mice or its syngeneic host, C57BL/6 mice. Virus was delivered either intravenously, in the case of LNCaP, or intratumorally, in the case of TRAMP. We found that individually, either G207 or radiation was effective in delaying tumor growth in these models. However, delivering the treatments simultaneously did not produce an enhanced effect.


Asunto(s)
Terapia Genética/métodos , Vectores Genéticos/efectos de la radiación , Herpesvirus Humano 1/fisiología , Neoplasias de la Próstata/terapia , Neoplasias Cutáneas/terapia , Animales , División Celular , Terapia Combinada , Herpesvirus Humano 1/genética , Humanos , Inyecciones Subcutáneas , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Desnudos , Trasplante de Neoplasias , Neoplasias de la Próstata/patología , Dosis de Radiación , Radiación Ionizante , Transfección , Células Tumorales Cultivadas , Replicación Viral
8.
Hum Gene Ther ; 12(8): 999-1010, 2001 May 20.
Artículo en Inglés | MEDLINE | ID: mdl-11387063

RESUMEN

G207, a replication-competent herpes simplex virus type 1 (HSV-1) virus, has been previously shown to be effective against human prostate cancer xenografts in mice. This study assesses its safety in the prostate of two animal models known for their sensitivity to HSV-1. BALB/c mice were injected intraprostatically with either HSV-1 G207 or strain F and observed for 5 months. None of the G207-injected animals exhibited any clinical signs of disease or died. However, 50% of strain F-injected mice displayed sluggish, hunched behavior and died by day 13. Histopathologically, the G207-injected prostates were normal whereas strain F-injected prostates showed epithelial flattening, sloughing, and stromal edema. Four Aotus nancymae monkeys were also injected with G207 intraprostatically and observed short term (up to 21 days) and long term (56 days). Safety was assessed on the basis of clinical observations, viral biodistribution, virus shedding, and histopathology. None of the injected monkeys displayed evidence of clinical disease, shedding of infectious virus, or spread of the virus into other organs. Except for minor histological changes unrelated to the study, no significant abnormalities were observed. These results demonstrate that G207 can be safely inoculated into the prostate and should be considered for human trials for the treatment of prostate cancer.


Asunto(s)
Técnicas de Transferencia de Gen , Terapia Genética/efectos adversos , Vectores Genéticos , Herpesvirus Humano 1/genética , Animales , Aotus trivirgatus , Haplorrinos , Masculino , Ratones , Ratones Endogámicos BALB C , Reacción en Cadena de la Polimerasa , Primates , Próstata/metabolismo , Próstata/patología , Factores de Tiempo , Células Tumorales Cultivadas , Esparcimiento de Virus
9.
Proc Natl Acad Sci U S A ; 98(11): 6396-401, 2001 May 22.
Artículo en Inglés | MEDLINE | ID: mdl-11353831

RESUMEN

Oncolytic herpes simplex virus type 1 (HSV-1) vectors are promising therapeutic agents for cancer. Their efficacy depends on the extent of both intratumoral viral replication and induction of a host antitumor immune response. To enhance these properties while employing ample safeguards, two conditionally replicating HSV-1 vectors, termed G47Delta and R47Delta, have been constructed by deleting the alpha47 gene and the promoter region of US11 from gamma34.5-deficient HSV-1 vectors, G207 and R3616, respectively. Because the alpha47 gene product is responsible for inhibiting the transporter associated with antigen presentation (TAP), its absence led to increased MHC class I expression in infected human cells. Moreover, some G47Delta-infected human melanoma cells exhibited enhanced stimulation of matched antitumor T cell activity. The deletion also places the late US11 gene under control of the immediate-early alpha47 promoter, which suppresses the reduced growth properties of gamma34.5-deficient mutants. G47Delta and R47Delta showed enhanced viral growth in a variety of cell lines, leading to higher virus yields and enhanced cytopathic effect in tumor cells. G47Delta was significantly more efficacious in vivo than its parent G207 at inhibiting tumor growth in both immune-competent and immune-deficient animal models. Yet, when inoculated into the brains of HSV-1-sensitive A/J mice at 2 x 10(6) plaque forming units, G47Delta was as safe as G207. These results suggest that G47Delta may have enhanced antitumor activity in humans.


Asunto(s)
Presentación de Antígeno/inmunología , Vectores Genéticos/inmunología , Glioma/prevención & control , Herpesvirus Humano 1/inmunología , Antígenos de Histocompatibilidad Clase I/inmunología , Neuroblastoma/prevención & control , Animales , Chlorocebus aethiops , Seguridad de Productos para el Consumidor , Efecto Citopatogénico Viral , Femenino , Vectores Genéticos/genética , Vectores Genéticos/fisiología , Glioma/inmunología , Herpesvirus Humano 1/genética , Herpesvirus Humano 1/fisiología , Antígenos de Histocompatibilidad Clase I/biosíntesis , Humanos , Proteínas Inmediatas-Precoces/genética , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Trasplante de Neoplasias , Neuroblastoma/inmunología , Proteínas de Unión al ARN/genética , Linfocitos T/inmunología , Células Tumorales Cultivadas , Células Vero , Proteínas Virales/genética , Replicación Viral
10.
Mol Ther ; 3(4): 431-7, 2001 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-11319903

RESUMEN

Gallbladder cancer is an extremely difficult disease to cure once metastases occur. In this paper, we explored the potential of G207, an oncolytic, replication-competent herpes simplex virus type 1 mutant, as a new therapeutic means for gallbladder cancer. Gallbladder carcinoma cell lines (four human and one hamster) showed nearly total cell killing within 72 h of G207 infection at a m.o.i. of 0.25 to 2.5 in vitro. The susceptibility to G207 cytopathic activity correlated with the infection efficiency demonstrated by lacZ expression. Intraneoplastic inoculation of G207 (1 x 10(7) pfu) in immunocompetent hamsters bearing established subcutaneous KIGB-5 tumors caused a significant inhibition of tumor growth and prolongation of survival. Repeated inoculations (three times with 4-day intervals) were significantly more efficacious than a single inoculation. In hamsters with bilateral subcutaneous KIGB-5 tumors, inoculation of one tumor alone with G207 caused regression or growth reduction of uninoculated tumors as well as inoculated tumors. In athymic mice, however, the anti-tumor effect was largely reduced in inoculated tumors and completely abolished in remote tumors, suggesting large contribution of T-cell-mediated immune responses to both local and systemic anti-tumor effect of G207. These results indicate that G207 may be useful as a new strategy for gallbladder cancer treatment.


Asunto(s)
Carcinoma/terapia , Neoplasias de la Vesícula Biliar/terapia , Terapia Genética/métodos , Herpesvirus Humano 1/genética , Mutación , Animales , Línea Celular , Cricetinae , Femenino , Vectores Genéticos/genética , Mesocricetus , Ratones , Ratones Desnudos , Factores de Tiempo , Células Tumorales Cultivadas
11.
Cancer Res ; 61(1): 153-61, 2001 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-11196154

RESUMEN

In vivo delivery of immunomodulatory genes is a promising strategy for solid tumor vaccination. A drawback is that it necessitates induction of a large effect from transgene expression in a small percentage of tumor cells. Although the B7 family is known to be the most potent of the costimulatory molecules, gene transduction of B7 alone has not been effective in inducing antitumor immunity in nonimmunogenic tumors by ex vivo methods, much less in vivo. We have developed a novel approach where a gene encoding soluble B7-1, a fusion protein of the extracellular domain of murine B7-1 and the Fc portion of human IgG1, is delivered to tumor cells in vivo in the context of an oncolytic replication-competent herpes simplex virus, and the gene product is secreted by tumor cells rather than expressed on the cell surface. Defective herpes simplex virus vectors containing the B7-1-immunoglobulin (B7-1-Ig) fusion transgene (dvB7Ig) were generated using G207 as a helper virus and tested in the poorly immunogenic murine neuroblastoma, Neuro2a, in syngeneic A/J mice. Intraneoplastic inoculation of dvB7Ig/G207 at a low titer successfully inhibited the growth of established s.c. tumors, despite the expression of B7-1-Ig being detected in only 1% or fewer of tumor cells at the inoculation site, and prolonged the survival of mice bearing intracerebral tumors. Immunohistochemistry of dvB7Ig/G207-inoculated tumors revealed a significant increase in CD4+ and CD8+ T-cell infiltration compared with control tumors inoculated with defective vector expressing alkaline phosphatase (dvAP/G207). The antitumor effect of dvB7Ig/G207 was not manifested in athymic mice. In vivo depletion of immune cell subsets in A/J mice further revealed that CD8+ T cells, but not CD4+ T cells, were required. Animals cured of their tumors by dvB7Ig/G207 treatment were protected against rechallenge with a lethal dose of Neuro2a cells but not SaI/N cells. The results demonstrate that the use of soluble B7-1 for immune gene therapy is a potent and clinically applicable means of in situ cancer vaccination.


Asunto(s)
Antígeno B7-1/genética , Antígeno B7-1/inmunología , Inmunoglobulina G/genética , Inmunoglobulina G/inmunología , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/inmunología , Animales , Antígenos de Diferenciación/inmunología , Antígeno B7-1/biosíntesis , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/inmunología , Neoplasias Encefálicas/terapia , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Vacunas contra el Cáncer/genética , Vacunas contra el Cáncer/inmunología , División Celular/inmunología , Chlorocebus aethiops , Femenino , Terapia Genética/métodos , Vectores Genéticos/genética , Herpesvirus Humano 1/genética , Inmunoconjugados/genética , Inmunoconjugados/inmunología , Fragmentos Fc de Inmunoglobulinas/biosíntesis , Fragmentos Fc de Inmunoglobulinas/genética , Fragmentos Fc de Inmunoglobulinas/inmunología , Inmunoglobulina G/biosíntesis , Inmunoterapia Activa/métodos , Macrófagos/inmunología , Ratones , Ratones Endogámicos A , Ratones Endogámicos BALB C , Ratones Desnudos , Neuroblastoma/inmunología , Neuroblastoma/patología , Neuroblastoma/terapia , Proteínas Recombinantes de Fusión/biosíntesis , Neoplasias Cutáneas/inmunología , Neoplasias Cutáneas/patología , Neoplasias Cutáneas/terapia , Solubilidad , Transgenes , Células Vero
12.
Mol Ther ; 2(6): 588-95, 2000 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-11124059

RESUMEN

G207 is a multimutated, conditionally replicating herpes simplex virus type 1 (HSV-1) that is currently in clinical trial for patients with malignant glioma. G207 exhibits an efficient oncolytic activity in tumor cells, yet minimal toxicity in normal tissue when injected into the brains of HSV-susceptible mice or nonhuman primates. In this study, we evaluated the shedding and biodistribution of clinical-grade G207 after intracerebral inoculation (3 x 10(7) pfu) in four New World owl monkeys (Aotus nancymae). Using PCR analyses and viral cultures, neither infectious virus nor viral DNA was detected from tear, saliva, or vaginal secretion samples at any time point up to 1 month postinoculation. Analyses of tissues obtained at necropsy at 1 month from two of the four monkeys, plus one monkey inoculated with laboratory-grade G207 (10(9) pfu) 2 years earlier, showed the distribution of G207 DNA restricted to the brain, although infectious virus was not isolated. Histopathology revealed normal brain tissues including the sites of inoculation. A measurable increase of serum anti-HSV antibody titer was observed in all monkeys, as early as 21 days postinoculation. The results ascertain the safety of G207 in the brain and indicate that strict biohazard management may not be required for G207-treated patients.


Asunto(s)
Encéfalo , Herpesvirus Humano 1/fisiología , Mutación , Replicación Viral , Esparcimiento de Virus , Animales , Anticuerpos Antivirales/análisis , Aotidae , Secuencia de Bases , Cartilla de ADN , ADN Viral/análisis , Femenino , Terapia Genética , Herpesvirus Humano 1/genética , Herpesvirus Humano 1/inmunología , Herpesvirus Humano 1/aislamiento & purificación , Masculino , Reacción en Cadena de la Polimerasa , Saliva/virología , Lágrimas/virología , Vagina/metabolismo , Vagina/virología
13.
Cancer Gene Ther ; 7(6): 939-46, 2000 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-10880026

RESUMEN

G207 is a multimutated, conditionally replicating herpes simplex virus type 1 (HSV-1) that retains an intact viral thymidine kinase (HSV-tk) gene. The virus exhibits oncolytic activity in various tumors and is being evaluated in patients with recurrent malignant glioma. In view of the potential for ganciclovir (GCV) to either enhance or inhibit the antitumoral activity of HSV-tk-retaining HSV-1 vectors, we evaluated the effect of GCV administration on the antitumoral activity of G207. In culture, addition of GCV either had no effect or inhibited the cytocidal action of G207 at replication-permissive temperatures, while it significantly increased the cell killing in three of the four cell lines studied when virus replication was inhibited at nonpermissive temperatures. Using a G207-permissive immunocompetent mouse tumor model, subcutaneous N18 neuroblastoma in syngeneic A/J mice, we found that GCV treatment did not affect G207-mediated tumor growth inhibition at a variety of viral doses (10(5), 10(7), and 10(7) x 2 plaque-forming units). In A/J mice harboring intracerebral N18 tumors, GCV administration had no significant effect on the prolongation of survival by G207 inoculation. These findings suggest that GCV administration may not be beneficial to the efficacy of G207 tumor therapy under conditions that favor active viral replication, because the potential HSV-tk/GCV-mediated enhancement of G207 oncolytic activity may be balanced out by the inhibitory action of GCV on viral replication.


Asunto(s)
Antivirales/farmacología , Neoplasias Encefálicas/terapia , Ganciclovir/farmacología , Herpesvirus Humano 1/efectos de los fármacos , Mutación , Neuroblastoma/terapia , Células Tumorales Cultivadas/efectos de los fármacos , Animales , Neoplasias Encefálicas/patología , Neoplasias Encefálicas/virología , Terapia Combinada , Efecto Citopatogénico Viral/efectos de los fármacos , Femenino , Herpesvirus Humano 1/genética , Herpesvirus Humano 1/fisiología , Inyecciones Intraperitoneales , Ratones , Neuroblastoma/patología , Neuroblastoma/virología , Neoplasias Cutáneas/patología , Neoplasias Cutáneas/terapia , Neoplasias Cutáneas/virología , Análisis de Supervivencia , Temperatura , Replicación Viral/efectos de los fármacos
14.
Gene Ther ; 7(10): 867-74, 2000 May.
Artículo en Inglés | MEDLINE | ID: mdl-10845725

RESUMEN

G207 is a conditionally replicating derivative of herpes simplex virus (HSV) type-1 strain F engineered with deletions of both gamma(1)34.5 loci and a lacZ insertion disabling the UL39 gene. We have demonstrated the efficacy of G207 in treating malignant glial tumors in athymic mice, as well as the safety of intracerebral G207 inoculation in mice and in Aotus nancymai. We sought to determine the safety of G207 inoculation into cerebral malignant glial tumors in humans. Criteria for inclusion into this dose-escalation study were the diagnosis of histologically proven malignant glioma, Karnofsky score > or = 70, recurrence despite surgery and radiation therapy, and an enhancing lesion greater than 1 cm in diameter. Serial magnetic resonance images were obtained for volumetric analysis. The trial commenced at a dose of 10(6) plaque forming units (p.f.u.) inoculated at a single enhancing site and was completed when the 21st patient was inoculated with 3x10(9) p.f.u. at five sites. While adverse events were noted in some patients, no toxicity or serious adverse events could unequivocally be ascribed to G207. No patient developed HSV encephalitis. We found radiographic and neuropathologic evidence suggestive of anti-tumor activity and long-term presence of viral DNA in some cases.


Asunto(s)
Neoplasias Encefálicas/terapia , Terapia Genética/métodos , Glioblastoma/terapia , Herpesvirus Humano 1/crecimiento & desarrollo , Recurrencia Local de Neoplasia/terapia , Replicación Viral , Adulto , Anciano , Anticuerpos Antivirales/sangre , Neoplasias Encefálicas/patología , Neoplasias Encefálicas/virología , Progresión de la Enfermedad , Femenino , Estudios de Seguimiento , Glioblastoma/patología , Glioblastoma/virología , Herpesvirus Humano 1/genética , Herpesvirus Humano 1/inmunología , Humanos , Imagen por Resonancia Magnética , Masculino , Persona de Mediana Edad , Recurrencia Local de Neoplasia/patología , Recurrencia Local de Neoplasia/virología , Tasa de Supervivencia , Resultado del Tratamiento
15.
Cancer Res ; 60(6): 1729-35, 2000 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-10749146

RESUMEN

The role of UV light-induced photoproducts in initiating base substitution mutation in human cells was examined by determining the frequency and spectrum of mutation in a supF tRNA gene in a shuttle vector plasmid transfected into DNA repair deficient cells (xeroderma pigmentosum complementation group A). To compare the role of two major UV-induced photoproducts, cis-syn cyclobutane-type pyrimidine dimers (CPDs) and pyrimidine (6-4) pyrimidone photoproducts (6-4PPs), each photoproduct was removed from UV-irradiated plasmid by photoreactivation before transfection. Removal of either CPDs or 6-4PPs by in vitro photoreactivation reduced the mutation frequency while keeping the mutation distribution and the predominance of G:C-A:T transitions as UV-irradiated plasmid without photoreactivation, indicating that both cytosine-containing CPDs and 6-4PPs were premutagenic lesions for G:C-A:T transitions. On the other hand, A:T-G:C transitions were not recovered from plasmids after the removal of 6-4PPs, whereas this type of mutation occurred at a significant level (11%) after the removal of CPDs. Thus, the premutagenic lesions for the A:T-G:C transition are 6-4PPs. Removal of both CPDs and 6-4PPs resulted in the disappearance of mutational hot spots and random distribution of mutation as observed in unirradiated control plasmids. However, the mutational spectrum of photoreactivated plasmids differed significantly from that of unirradiated plasmids. A characteristic feature is a high portion of A:T-T:A transversions (11%) in the photoreactivated plasmid. This mutation is due to nondipyrimidinic "minor" photoproducts, and the mutation spectrum suggests that TA*, the major photoproduct of thymidylyl-(3'-5')-deoxyadenosine, is the premutagenic lesion for this mutation. This is the first report revealing the distinct mutagenic roles of the major UV photoproducts and "minor" photoproducts by the use of (6-4)photolyase.


Asunto(s)
Reparación del ADN , ADN/efectos de la radiación , Dímeros de Pirimidina/metabolismo , Rayos Ultravioleta , Secuencia de Bases , Línea Celular Transformada , ADN/química , ADN/genética , Análisis Mutacional de ADN , Ensayo de Inmunoadsorción Enzimática , Genes Supresores/genética , Humanos , Datos de Secuencia Molecular , Mutagénesis , Mutación , Plásmidos/química , Plásmidos/genética , Plásmidos/efectos de la radiación , Mutación Puntual , Dímeros de Pirimidina/fisiología , ARN de Transferencia/genética , Homología de Secuencia de Ácido Nucleico , Xerodermia Pigmentosa/genética , Xerodermia Pigmentosa/patología
16.
Gen Comp Endocrinol ; 118(1): 123-33, 2000 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-10753574

RESUMEN

As a first step in investigating the mechanism underlying the steroidogenic shift from the production of ovarian androgens (vitellogenic stage) to that of 17alpha-hydroxylated progestins (maturational stage) in Japanese eel during induced oogenesis, a cDNA encoding Japanese eel (Anguilla japonica) ovarian P450c17 (CYP17: steroid 17alpha-hydroxylase/C(17-20) lyase) was cloned and sequenced. This cDNA contained the complete coding region representing 510 amino acid residues, which showed high sequence homology to those of rainbow trout (74%) and mammals (45-55%). The protein encoded by this cDNA possessed high enzymatic activities of 17alpha-hydroxylase and C(17-20) lyase, thus quickly converting pregnenolone and progesterone to their respective delta(4) and delta(5) C19 products. P450c17 produced a single transcript of 2.4 kb in length, as assessed by Northern blot. Transcript levels of this enzyme significantly increased throughout artificially induced ovarian development. Considering this together with the previous data showing that C(17-20) lyase activity decreased from the vitellogenic to the maturational stage, whereas 17alpha-hydroxylase activity increased, the present data suggest that changes in C(17-20) lyase activity (the production of androgens) do not depend on transcriptional changes of the P450c17 gene.


Asunto(s)
ADN Complementario/biosíntesis , Anguilas/fisiología , Ovario/enzimología , Esteroide 17-alfa-Hidroxilasa/biosíntesis , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Northern Blotting , Células COS , Clonación Molecular , ADN Complementario/análisis , ADN Complementario/genética , Femenino , Biblioteca de Genes , Datos de Secuencia Molecular , Poli A/biosíntesis , Poli A/genética , Pregnenolona/biosíntesis , Progesterona/biosíntesis , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Especificidad de la Especie , Esteroide 17-alfa-Hidroxilasa/genética
17.
Mutat Res ; 435(3): 255-62, 1999 Dec 07.
Artículo en Inglés | MEDLINE | ID: mdl-10606816

RESUMEN

Photoreactivation is one of the DNA repair mechanisms to remove UV lesions from cellular DNA with a function of the DNA photolyase and visible light. Two types of photolyase specific for cyclobutane pyrimidine dimers (CPD) and for pyrimidine (6-4) pyrimidones (6-4PD) are found in nature, but neither is present in cells from placental mammals. To investigate the effect of the CPD-specific photolyase on killing and mutations induced by UV, we expressed a marsupial DNA photolyase in DNA repair-deficient group A xeroderma pigmentosum (XP-A) cells. Expression of the photolyase and visible light irradiation removed CPD from cellular DNA and elevated survival of the UV-irradiated XP-A cells, and also reduced mutation frequencies of UV-irradiated shuttle vector plasmids replicating in XP-A cells. The survival of UV-irradiated cells and mutation frequencies of UV-irradiated plasmids were not completely restored to the unirradiated levels by the removal of CPD. These results suggest that both CPD and other UV damage, probably 6-4PD, can lead to cell killing and mutations.


Asunto(s)
Reparación del ADN/efectos de la radiación , Desoxirribodipirimidina Fotoliasa/biosíntesis , Vectores Genéticos/genética , Luz , Mutación/genética , Rayos Ultravioleta , Xerodermia Pigmentosa/enzimología , Xerodermia Pigmentosa/genética , Secuencia de Bases , Supervivencia Celular/genética , Supervivencia Celular/efectos de la radiación , Células Cultivadas , Análisis Mutacional de ADN , ADN Complementario/genética , Desoxirribodipirimidina Fotoliasa/genética , Desoxirribodipirimidina Fotoliasa/metabolismo , Humanos , Datos de Secuencia Molecular , Mutación/efectos de la radiación , Plásmidos/efectos de la radiación , ARN Mensajero/metabolismo , Transfección
18.
Hum Gene Ther ; 10(17): 2741-55, 1999 Nov 20.
Artículo en Inglés | MEDLINE | ID: mdl-10584921

RESUMEN

Replication-competent, attenuated herpes simplex virus (HSV) vectors have been developed for viral oncolytic therapy of primary and metastatic malignant brain tumors. However, the role of the host immune responses in the brain has not been elucidated. N18 neuroblastoma cells were used as a tumor model in syngeneic A/J mice to test the therapeutic efficacy of G207, a conditionally replicating HSV vector, in an immunocompetent condition. G207 inoculated intraneoplastically exhibited a prominent oncolytic antitumor effect in mice harboring N18 tumors in the brain or subcutaneously, and, in addition, elicited a systemic antitumor immune response. Subcutaneous tumor therapy with G207 caused regression of a remote, established tumor in the brain or in the periphery, which was potentially mediated by the systemic antitumor immune response, and provided persistent tumor-specific protection against N18 tumor rechallenge in the brain as well as in the periphery. Antitumor immunity was associated with an elevation of specific CTL activity against N18 tumor cells that persisted for at least 13 months. The results suggest that the oncolytic antitumor action of replication-competent HSV may be augmented by induction of specific and systemic antitumor immunity effective both in the periphery and in the brain.


Asunto(s)
Neoplasias Encefálicas/terapia , Vacunas contra el Cáncer/uso terapéutico , Vectores Genéticos , Neuroblastoma/terapia , Simplexvirus/genética , Linfocitos T Citotóxicos/inmunología , Animales , Neoplasias Encefálicas/inmunología , Neoplasias Encefálicas/patología , Vacunas contra el Cáncer/genética , Vacunas contra el Cáncer/inmunología , Femenino , Inmunoterapia , Ratones , Neuroblastoma/inmunología , Neuroblastoma/patología , Simplexvirus/inmunología , Simplexvirus/fisiología , Neoplasias Cutáneas/patología , Neoplasias Cutáneas/terapia , Replicación Viral
19.
Hum Gene Ther ; 10(17): 2869-78, 1999 Nov 20.
Artículo en Inglés | MEDLINE | ID: mdl-10584932

RESUMEN

A multimutated, conditionally replicating herpes simplex virus type 1, G207, has been developed as an effective means of treating human malignant brain tumors. We have shown that intraneoplastic inoculation of G207 induces a specific and systemic antitumor immune response that plays an important role in the antitumor activity, in addition to the direct oncolytic action of G207. Since a large number of malignant brain tumor patients are treated with corticosteroids, it is important to evaluate whether the therapeutic efficacy of G207 is affected by corticosteroid-induced immunosuppression. For a tumor model, we used G207-permissive N18 murine neuroblastoma cells implanted subcutaneously in syngeneic A/J mice. Intraneoplastic inoculation of G207 (10(7) PFU) induced significant suppression of tumor growth whether or not dexamethasone (5 mg/kg) was given. When dexamethasone was given for an extensive time (16 days starting on day -2), all G207-treated mice showed tumor growth despite initial shrinkage, whereas in the saline group, four of eight of the G207-treated mice were cured. Dexamethasone administration significantly reduced serum neutralizing antibodies against G207 at 14 and 21 days after intraneoplastic G207 inoculation. However, there was no difference between the dexamethasone and saline groups in terms of the amount of infectious G207 isolated from tumors. Dexamethasone administration completely abolished G207-induced cytotoxic T lymphocyte activity against N18 cells. These results indicate that the oncolytic activity of G207 is retained under corticosteroid administration. However, intensive immunosuppression may diminish the long-term efficacy of G207 owing to suppression of tumor-specific cytotoxic T lymphocyte induction.


Asunto(s)
Antineoplásicos Hormonales/farmacología , Terapia Biológica , Dexametasona/farmacología , Neuroblastoma/terapia , Simplexvirus/efectos de los fármacos , Animales , Antineoplásicos Hormonales/uso terapéutico , Terapia Combinada , Efecto Citopatogénico Viral/efectos de los fármacos , Dexametasona/uso terapéutico , Modelos Animales de Enfermedad , Femenino , Humanos , Ratones , Trasplante de Neoplasias , Neuroblastoma/inmunología , Simplexvirus/genética , Simplexvirus/fisiología , Linfocitos T Citotóxicos/inmunología , Células Tumorales Cultivadas , Replicación Viral
20.
Gene Ther ; 6(10): 1751-8, 1999 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-10516725

RESUMEN

Replication-competent, attenuated mutants of herpes simplex virus type 1 (HSV-1) have been shown to be efficacious for tumor therapy. However, these studies did not address the consequences of prior exposure to HSV, as will be the case with many patients likely to receive this therapy. Two strains of mice, A/J and BALB/c, were infected with wild-type HSV-1 by intraperitoneal injection and the immune response was determined by plaque reduction assay for neutralizing antibody and ELISA for IgG and IgM. Syngeneic tumors, N18 neuroblastoma and CT26 colon carcinoma, were implanted subcutaneously in HSV-1 seropositive and naive A/J and BALB/c mice, respectively. Established tumors were subsequently treated intratumorally with a multi-mutated HSV-1, G207. G207 inhibited tumor growth to a similar extent whether the mice were seropositive or not. We next examined the effect of multiple intratumoral inoculations of a 10-fold lower dose of G207 on tumor growth. In the multiple treatment group (biweekly for 3 weeks), 75% of tumors were cured, whereas no cures were seen in the single treatment group. We conclude that HSV seropositivity should not deleteriously affect the efficacy of G207 tumor therapy, and multiple inoculations of virus should be considered for clinical evaluation.


Asunto(s)
Terapia Genética/métodos , Vectores Genéticos/administración & dosificación , Herpes Simple/inmunología , Herpesvirus Humano 1/genética , Neoplasias Experimentales/terapia , Animales , Anticuerpos Antivirales/sangre , Femenino , Inmunización , Inmunoglobulina G/análisis , Inmunoglobulina M/análisis , Ratones , Ratones Endogámicos A , Ratones Endogámicos BALB C , Trasplante de Neoplasias , Neoplasias Experimentales/inmunología , Distribución Aleatoria , Células Tumorales Cultivadas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA