Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 348
Filtrar
1.
Oncogene ; 36(5): 585-592, 2017 02 02.
Artículo en Inglés | MEDLINE | ID: mdl-27425593

RESUMEN

In advanced cancer, current conventional therapies or immunotherapies cannot eradicate all tumor cells for most patients. Integration of these two treatments for synergistic effects could eradicate more tumor cells and increase the overall survival rates. However, since how conventional treatments impact on immune system remains unclear, proper integration is still a challenge. Intensive chemo/radiotherapy may impair ongoing immune responses, while lower intensity of therapy might not kill enough tumor cells, both leading to tumor relapse. Current understanding of mechanisms of resistance to conventional and targeted cancer therapies has focused on cell intrinsic pathways that trigger DNA damage/repair or signaling pathways related to cell growth. Recent reports show that host T cells properly primed against tumor-specific antigens after conventional treatment, which can integrate with direct cytotoxic effects induced by radiation or chemotherapy to profoundly control tumors. Following cytotoxic anticancer treatment, tumor-derived DAMPs (damage-associated molecular patterns) can be sensed by innate cells, which drives type I interferon production for cross-priming of CD8+ T cells. Some types and protocols of chemotherapy or radiation can increase tumor-infiltrating lymphocytes that overcome resistance to immunotherapy. As such, a deeper understanding of the immune mechanisms of conventional and targeted cancer therapies will lead toward novel combinatorial anticancer strategies with improved clinical benefit.


Asunto(s)
Inmunoterapia/métodos , Neoplasias/terapia , Animales , Anticuerpos/inmunología , Terapia Combinada , Humanos , Terapia Molecular Dirigida , Neoplasias/inmunología , Neoplasias/patología
2.
Sci Rep ; 6: 35854, 2016 10 24.
Artículo en Inglés | MEDLINE | ID: mdl-27775025

RESUMEN

Strategies to identify tumors at highest risk for treatment failure are currently under investigation for patients with bladder cancer. We demonstrate that flow cytometric detection of poorly differentiated basal tumor cells (BTCs), as defined by the co-expression of CD90, CD44 and CD49f, directly from patients with early stage tumors (T1-T2 and N0) and patient-derived xenograft (PDX) engraftment in locally advanced tumors (T3-T4 or N+) predict poor prognosis in patients with bladder cancer. Comparative transcriptomic analysis of bladder tumor cells isolated from PDXs indicates unique patterns of gene expression during bladder tumor cell differentiation. We found cell division cycle 25C (CDC25C) overexpression in poorly differentiated BTCs and determined that CDC25C expression predicts adverse survival independent of standard clinical and pathologic features in bladder cancer patients. Taken together, our findings support the utility of BTCs and bladder cancer PDX models in the discovery of novel molecular targets and predictive biomarkers for personalizing oncology care for patients.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Neoplasias de la Vejiga Urinaria/mortalidad , Neoplasias de la Vejiga Urinaria/patología , Ensayos Antitumor por Modelo de Xenoinjerto/métodos , Anciano , Animales , Biomarcadores de Tumor/genética , Diferenciación Celular/genética , Femenino , Citometría de Flujo , Regulación Neoplásica de la Expresión Génica , Humanos , Masculino , Ratones SCID , Persona de Mediana Edad , Pronóstico , Estudios Prospectivos , Neoplasias de la Vejiga Urinaria/genética , Neoplasias de la Vejiga Urinaria/cirugía , Fosfatasas cdc25/genética
3.
Br J Cancer ; 113(2): 327-35, 2015 Jul 14.
Artículo en Inglés | MEDLINE | ID: mdl-26042934

RESUMEN

BACKGROUND: The CXCL10/CXCR3 signalling mediates paracrine interactions between tumour and stromal cells that govern leukocyte trafficking and angiogenesis. Emerging data implicate noncanonical CXCL10/CXCR3 signalling in tumourigenesis and metastasis. However, little is known regarding the role for autocrine CXCL10/CXCR3 signalling in regulating the metastatic potential of individual tumour clones. METHODS: We performed transcriptomic and cytokine profiling to characterise the functions of CXCL10 and CXCR3 in tumour cells with different metastatic abilities. We modulated the expression of the CXCL10/CXCR3 pathway using shRNA-mediated silencing in both in vitro and in vivo models of B16F1 melanoma. In addition, we examined the expression of CXCL10 and CXCR3 and their associations with clinical outcomes in clinical data sets derived from over 670 patients with melanoma and colon and renal cell carcinomas. RESULTS: We identified a critical role for autocrine CXCL10/CXCR3 signalling in promoting tumour cell growth, motility and metastasis. Analysis of publicly available clinical data sets demonstrated that coexpression of CXCL10 and CXCR3 predicted an increased metastatic potential and was associated with early metastatic disease progression and poor overall survival. CONCLUSION: These findings support the potential for CXCL10/CXCR3 coexpression as a predictor of metastatic recurrence and point towards a role for targeting of this oncogenic axis in the treatment of metastatic disease.


Asunto(s)
Quimiocina CXCL10/fisiología , Transducción de Señal/fisiología , Animales , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular , Ratones , Ratones Endogámicos C57BL , Metástasis de la Neoplasia , Receptores CXCR3/fisiología
4.
Oncogene ; 34(21): 2807-13, 2015 May 21.
Artículo en Inglés | MEDLINE | ID: mdl-25043302

RESUMEN

NF-κB proteins play a central and subunit-specific role in the response to DNA damage. Previous work identified p50/NF-κB1 as being necessary for cytotoxicity in response to DNA alkylation damage. Given the importance of damage-induced cell death for the maintenance of genomic stability, we examined whether Nfkb1 acts as a tumor suppressor in the setting of alkylation damage. Hprt mutation analysis demonstrates that Nfkb1(-/-) cells accumulate more alkylator-induced, but not ionizing radiation (IR)-induced, mutations than similarly treated wild-type cells. Subsequent in vivo tumor induction studies reveal that following alkylator treatment, but not IR, Nfkb1(-/-) mice develop more lymphomas than similarly treated Nfkb1(+/+) animals. Heterozygous mice develop lymphomas at an intermediate rate and retain functional p50 in their tumors, indicating that Nfkb1 acts in a haploinsufficient manner. Analysis of human cancers, including therapy-related myeloid neoplasms, demonstrates that NFKB1 mRNA expression is downregulated compared with control samples in multiple hematological malignancies. These data indicate that Nfkb1 is a haploinsufficient, pathway-specific tumor suppressor that prevents the development of hematologic malignancy in the setting of alkylation damage.


Asunto(s)
Daño del ADN/genética , Haploinsuficiencia/genética , Subunidad p50 de NF-kappa B/genética , Proteínas Supresoras de Tumor/genética , Alquilación/genética , Animales , Muerte Celular/genética , Regulación hacia Abajo/genética , Femenino , Heterocigoto , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , ARN Mensajero/genética , Radiación Ionizante , Células Tumorales Cultivadas
6.
Ann Oncol ; 24(3): 769-76, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23104721

RESUMEN

BACKGROUND: AdGV.EGR.TNF.11D (TNFerade™ Biologic) is a replication-deficient adenoviral vector expressing human tumor necrosis factor alpha (TNF-α) under the control of the chemoradiation-inducible EGR-1 promoter. TNF-α has been shown to function as a radiation sensitizer. We conducted a phase I dose escalation study to determine the maximum tolerated dose (MTD) and dose-limiting toxicity (DLT) of TNFerade™ Biologic, when added to chemoradiotherapy in poor prognosis patients with recurrent, previously irradiated head and neck cancer (HNC). METHODS: TNFerade™ Biologic was injected intratumorally on day 1 of each 14-day cycle and dose-escalated in log increments from 4 × 10(9) to 4 × 10(11) PU. Daily radiation, infusional 5-fluorouracil (5-FU), and hydroxyurea were given on days 1-5 for seven cycles (FHX). Tumor biopsies were obtained before, during, and after treatment. RESULTS: Fourteen patients were treated. DLT was reached at a dose level of 3 (4 × 10(11) PU) with three thrombotic events. The response rate was 83.3%. The median survival was 9.6 months. One patient (7.1%) remained alive 3 years after treatment. Biopsies were obtained in 90% of patients. Nearly all tumors expressed adenovirus receptors, TNF-α, and TNF-α receptors. Adenoviral DNA was detected in three biopsies from one patient. CONCLUSIONS: TNFerade™ Biologic can be safely integrated with FHX chemoradiotherapy at an MTD of 4 × 10(10) PU. Monitoring for thrombotic events is indicated.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Carcinoma de Células Escamosas/terapia , ADN/administración & dosificación , Neoplasias de Cabeza y Cuello/terapia , Fármacos Sensibilizantes a Radiaciones/administración & dosificación , Adulto , Anciano , Carcinoma de Células Escamosas/mortalidad , Quimioradioterapia , ADN/genética , Fraccionamiento de la Dosis de Radiación , Femenino , Fluorouracilo/administración & dosificación , Terapia Genética , Neoplasias de Cabeza y Cuello/mortalidad , Humanos , Hidroxiurea/administración & dosificación , Estimación de Kaplan-Meier , Masculino , Dosis Máxima Tolerada , Persona de Mediana Edad , Planificación de la Radioterapia Asistida por Computador , Radioterapia de Intensidad Modulada , Retratamiento , Carcinoma de Células Escamosas de Cabeza y Cuello , Resultado del Tratamiento
7.
Free Radic Biol Med ; 51(10): 1918-25, 2011 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-21945096

RESUMEN

Manganese superoxide dismutase (SOD2)-mediated adaptive processes that protect against radiation-induced micronucleus formation can be induced in cells after a 2-Gy exposure by previously exposing them to either low-dose ionizing radiation (10cGy) or WR1065 (40µM), the active thiol form of amifostine. Although both adaptive processes culminate in elevated levels of SOD2 enzymatic activity, the underlying pathways differ in complexity, with the tumor necrosis factor α (TNFα) signaling pathway implicated in the low-dose radiation-induced response, but not in the thiol-induced pathway. The goal of this study was the characterization of the effects of TNFα receptors 1 and 2 (TNFR1, TNFR2) on the adaptive responses induced by low-dose irradiation or thiol exposure using micronucleus formation as an endpoint. BFS-1 wild-type cells with functional TNFR1 and 2 were exposed 24h before a 2-Gy dose of ionizing radiation to either 10cGy or a 40µM dose of WR1065. BFS2C-SH02 cells, defective in TNFR1, and BFS2C-SH22 cells, defective in both TNFR1 and TNFR2 and generated from BFS2C-SH02 cells by transfection with a murine TNFR2-targeting vector and confirmed to be TNFR2 defective by quantitative PCR, were also exposed under similar conditions for comparison. A 10-cGy dose of radiation induced a significant elevation in SOD2 activity in BFS-1 (P<0.001) and BFS2C-SH02 (P=0.005) but not BFS2C-SH22 cells (P=0.433), compared to their respective untreated controls. In contrast, WR1065 significantly induced elevations in SOD2 activity in all three cell lines (P=0.001, P=0.007, P=0.020, respectively). A significant reduction in the frequency of radiation-induced micronuclei was observed in each cell line when exposure to a 2-Gy challenge dose of radiation occurred during the period of maximal elevation in SOD2 activity. However, this adaptive effect was completely inhibited if the cells were transfected 24h before low-dose radiation or thiol exposure with SOD2 siRNA. Under the conditions tested, TNFR1 and 2 inhibition negatively affected the low-dose radiation-induced but not the thiol-induced adaptive responses observed to be mediated by elevations in SOD2 activity.


Asunto(s)
Mercaptoetilaminas/farmacología , Superóxido Dismutasa/metabolismo , Factor de Necrosis Tumoral alfa/farmacología , Amifostina/análogos & derivados , Amifostina/química , Animales , Línea Celular Tumoral , Activación Enzimática/genética , Activación Enzimática/efectos de la radiación , Mercaptoetilaminas/química , Ratones , Micronúcleos con Defecto Cromosómico/efectos de los fármacos , Micronúcleos con Defecto Cromosómico/efectos de la radiación , Pruebas de Micronúcleos , ARN Interferente Pequeño/genética , Radiación Ionizante , Receptores Tipo I de Factores de Necrosis Tumoral/genética , Receptores Tipo I de Factores de Necrosis Tumoral/metabolismo , Receptores Tipo II del Factor de Necrosis Tumoral/genética , Receptores Tipo II del Factor de Necrosis Tumoral/metabolismo , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Transducción de Señal/efectos de la radiación , Superóxido Dismutasa/genética
8.
Gene Ther ; 18(11): 1098-102, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21544094

RESUMEN

Oncolytic viruses have been combined with standard cancer therapies to increase therapeutic efficacy. Given the sequential activation of herpes viral genes (herpes simplex virus-1, HSV-1) and the temporal cellular changes induced by ionizing radiation, we hypothesized an optimal temporal sequence existed in combining oncolytic HSV-1 with ionizing radiation. Murine U-87 glioma xenografts were injected with luciferase encoding HSV-1, and ionizing radiation (IR) was given at times before or after viral injection. HSV-1 replication and tumor-volume response were followed. Radiation given 6-9 h after HSV-1 injection resulted in maximal viral luciferase expression and infectious viral production in tumor xenografts. The greatest xenograft regression was also seen with radiation given 6 h after viral injection. We then tested if HSV-1 replication had a dose response to ionizing radiation. HSV-1 luciferase expression exhibited a dose response as xenografts were irradiated from 0 to 5 Gy. There was no difference in viral luciferase expression as IR dose increased from 5 Gy up to 20 Gy. These results suggest that the interaction of IR with the HSV-1 lytic cycle can be manipulated for therapeutic gain by delivering IR at a specific time within viral replicative cycle.


Asunto(s)
Neoplasias Encefálicas/terapia , Glioma/terapia , Herpesvirus Humano 1/crecimiento & desarrollo , Viroterapia Oncolítica/métodos , Radiación Ionizante , Replicación Viral/efectos de la radiación , Animales , Terapia Combinada , Relación Dosis-Respuesta en la Radiación , Herpesvirus Humano 1/efectos de la radiación , Ratones , Ratones Desnudos , Virus Oncolíticos/crecimiento & desarrollo , Virus Oncolíticos/efectos de la radiación , Replicación Viral/genética
9.
Cancer Gene Ther ; 16(8): 609-19, 2009 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-19444302

RESUMEN

Radiotherapy is a widely used treatment for localized malignancies that is often delivered in combination with cytotoxic chemotherapeutic agents. The concept that treatment of localized tumors can be improved with a radio- and chemo-inducible gene therapy strategy has been investigated in the laboratory and now translated to the clinic. The TNFerade (Ad.Egr-TNF11D) adenoviral vector was engineered by inserting radio- and chemo-inducible elements from the Egr-1 promoter upstream to a cDNA encoding tumor necrosis factor-alpha (TNF-alpha). Transduction of tumor cells with TNFerade and then treatment with radiation or chemotherapy is associated with spatial and temporal control of TNF-alpha secretion and enhanced antitumor activity. TNFerade has been evaluated in trials for patients with sarcomas, melanomas and cancers of the pancreas, esophagus, rectum and head and neck. If the ongoing phase III trial for pancreatic cancer is successful, TNFerade will likely become the first gene therapy approved for cancer in the United States.


Asunto(s)
Antineoplásicos/uso terapéutico , Terapia Genética/métodos , Neoplasias/terapia , Radioterapia , Factor de Necrosis Tumoral alfa/biosíntesis , Sistemas de Liberación de Medicamentos/métodos , Vectores Genéticos/metabolismo , Humanos , Neoplasias/metabolismo , Radiación Ionizante , Factor de Necrosis Tumoral alfa/administración & dosificación
10.
Technol Cancer Res Treat ; 8(3): 201-6, 2009 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-19445537

RESUMEN

Intensity modulated radiation therapy (IMRT) has achieved widespread use for prostate cancer; however, in relation to this use, outcomes studies are still relatively sparse. We report a single-institutional experience in outcomes analysis with the use of IMRT for the primary management of prostate cancer. One hundred thirty consecutive patients with adenocarcinoma of the prostate were treated at a single institution using IMRT with curative intent. Thirty-six (28%) patients were classified as low-risk, 69 (53%) as intermediate-risk, and 25 (19%) as high-risk. The median dose prescription was 76 Gy to the planning target volume. Sixty-five (50%) patients received androgen deprivation therapy (ADT) for a median 4 months, starting 2 months prior to IMRT. Biochemical failure was defined as PSA < post-treatment nadir+2. Gastrointestinal (GI) and Genitourinary (GU) toxicity were defined by RTOG criteria. Median follow-up was 53 months. By NCCN risk category, 4-year biochemical control was 97%, 94%, and 87% for low, intermediate, and high-risk patients, respectively. Among disease factors, multivariable analysis demonstrated the strongest association between biochemical control and Gleason score < or =6 (p=0.0371). Therapy was well tolerated with no Grade 4 toxicity and limited grade 3 GI or GU toxicity. Acute Grade 3+ GI and GU toxicity rates were 0% and 2%, and maximal late Grade 3+ GI and GU toxicity rates were 5% and 6%, respectively. Late rectal toxicity was associated with higher volumes of RT to the rectum. By last follow-up late Grade 3+ toxicity was 2% for both GI and GU systems. In conclusion, patients treated with IMRT for prostate cancer have excellent rates of biochemical control and low rates of severe toxicity of treatment.


Asunto(s)
Neoplasias de la Próstata/radioterapia , Radioterapia de Intensidad Modulada/efectos adversos , Anciano , Anciano de 80 o más Años , Tracto Gastrointestinal/efectos de la radiación , Humanos , Masculino , Persona de Mediana Edad , Análisis Multivariante , Sistema Urogenital/efectos de la radiación
11.
Cancer Gene Ther ; 16(4): 373-81, 2009 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-18974777

RESUMEN

TNFerade is a radioinducible adenoviral vector expressing tumor necrosis factor-alpha (TNF-alpha) (Ad.Egr-TNF) currently in a phase III trial for inoperable pancreatic cancer. We studied B16-F1 melanoma tumors in TNF receptor wild-type (C57BL/6) and deficient (TNFR1,2-/- and TNFR1-/-) mice. Ad.Egr-TNF+IR inhibited tumor growth compared with IR in C57BL/6 but not in receptor-deficient mice. Tumors resistant to TNF-alpha were also sensitive to Ad.Egr-TNF+IR in C57BL/6 mice. Ad.Egr-TNF+IR produced an increase in tumor-associated endothelial cell apoptosis not observed in receptor-deficient animals. Also, B16-F1 tumors in mice with germline deletions of TNFR1,2, TNFR1 or TNF-alpha, or in mice receiving anti-TNF-alpha exhibited radiosensitivity. These results show that tumor-associated endothelium is the principal target for Ad.Egr-TNF radiosensitization and implicate TNF-alpha signaling in tumor radiosensitivity.


Asunto(s)
Terapia Genética/métodos , Melanoma Experimental/terapia , Fármacos Sensibilizantes a Radiaciones , Factor de Necrosis Tumoral alfa/metabolismo , Terapia por Rayos X , Animales , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Células Endoteliales/efectos de los fármacos , Células Endoteliales/fisiología , Etanercept , Humanos , Inmunoglobulina G/farmacología , Inmunosupresores/farmacología , Ratones , Trasplante de Neoplasias , Receptores del Factor de Necrosis Tumoral , Receptores Tipo I de Factores de Necrosis Tumoral/deficiencia , Receptores Tipo II del Factor de Necrosis Tumoral/deficiencia , Factor de Necrosis Tumoral alfa/antagonistas & inhibidores
12.
Cancer Gene Ther ; 15(3): 133-9, 2008 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-18157146

RESUMEN

We report the anticarcinogenic, anti-aging polyphenol resveratrol activates the radio- and chemo-inducible cancer gene therapy vector Ad.Egr.TNF, a replication-deficient adenovirus that expresses human tumor necrosis factor alpha (TNF-alpha) under control of the Egr-1 promoter. Like ionizing radiation or chemotherapeutic agents previously shown to activate Ad.Egr.TNF, resveratrol also induces Egr-1 expression from its chromosomal locus with a possible role for Egr-1 promoter CC(A+T)richGG sequences in the expression of TNF-alpha. Resveratrol induction of TNF-alpha in Ad.Egr.TNF-infected tumor xenografts demonstrated antitumor response in human and rat tumor models comparable to that of radio- or chemotherapy-induced TNF-alpha. Although sirtuins are known targets of resveratrol, in vitro inhibition of SIRT1 activity did not abrogate resveratrol induction of Egr-1 expression. This suggests that SIRT1 is not essential to mediate resveratrol induction of Egr-1. Nevertheless, control of transgene expression via resveratrol activation of Egr-1 may extend use of Ad.Egr.TNF to patients intolerant of radiation or cytotoxic therapy and offer a novel tool for development of other inducible gene therapies.


Asunto(s)
Adenoviridae/genética , Terapia Genética/métodos , Estilbenos/farmacología , Factor de Necrosis Tumoral alfa/genética , Ensayos Antitumor por Modelo de Xenoinjerto/métodos , Acetilación/efectos de los fármacos , Animales , Western Blotting , Línea Celular Tumoral , Proteína 1 de la Respuesta de Crecimiento Precoz/genética , Proteína 1 de la Respuesta de Crecimiento Precoz/metabolismo , Ensayo de Inmunoadsorción Enzimática , Etopósido/farmacología , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Ratones , Ratones Desnudos , Ratas , Resveratrol , Sirtuinas/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Proteína p53 Supresora de Tumor/metabolismo
13.
Curr Cancer Drug Targets ; 6(3): 197-205, 2006 May.
Artículo en Inglés | MEDLINE | ID: mdl-16712457

RESUMEN

Exposure to ionizing radiation (IR) results in the formation of DNA double strand breaks, resulting in the activation of phosphatidylinositol 3'-kinase-like kinases ATM, ATR and DNK-PKcs. A physiologically important downstream target is the minor histone H2A variant, H2AX, which is rapidly phosphorylated on Ser 139 of the carboxyl tail after IR. Recent work suggests that phosphorylated H2AX (gamma-H2AX) plays an important role in the recruitment and/or retention of DNA repair and checkpoint proteins such as BRCA1, MRE11/RAD50/NBS1 complex, MDC1 and 53BP1. H2AX-/- mouse embryonic fibroblasts are radiation sensitive and demonstrate deficits in repairing DNA damage compared to their wildtype counterparts. Cells treated with peptide inhibitors of gamma-H2AX demonstrate increased radiosensitivity following radiation compared with untreated irradiated cells. Analysis of the kinetics of gamma-H2AX clearance after IR or other DNA damaging agents reveals a correlation between increased gamma-H2AX persistence and unrepaired DNA damage and cell death. These data highlight the potential of post-translational modifications of chromatin as a therapeutic target for enhancing the efficacy of radiotherapy. Therapies that either block gamma-H2AX foci formation by inhibiting upstream kinase activity or that directly inhibit H2AX function may interfere with DNA damage repair processes and warrant further investigation as potential radiosensitizing agents. Agents that increase persistence of gamma-H2AX after IR are likely to increase unrepaired DNA damage.


Asunto(s)
Histonas/efectos de la radiación , Neoplasias/genética , Neoplasias/radioterapia , Radioterapia , Antineoplásicos/farmacología , Biomarcadores , Daño del ADN/genética , ADN de Neoplasias/efectos de los fármacos , ADN de Neoplasias/efectos de la radiación , Humanos
14.
Cancer Gene Ther ; 13(1): 1-6, 2006 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-16082378

RESUMEN

Gene therapy of cancer represents a promising but challenging area of therapeutic research. The discovery of radiation-inducible genes led to the concept and development of radiation-targeted gene therapy. In this approach, promoters of radiation-inducible genes are used to drive transcription of transgenes in the response to radiation. Constructs in which the radiation-inducible promoter elements activate a transgene encoding a cytotoxic protein are delivered to tumors by adenoviral vectors. The tumoricidal effects are then localized temporally and spatially by X-rays. We review the conceptual development of TNFerade, an adenoviral vector containing radiation-inducible elements of the early growth response-1 promoter upstream of a cDNA encoding human tumor necrosis factor-alpha. We also summarize the preclinical work and clinical trials utilizing this vector as a treatment for diverse solid tumors.


Asunto(s)
Regulación Neoplásica de la Expresión Génica/efectos de la radiación , Terapia Genética/métodos , Neoplasias/terapia , Adenoviridae/genética , Adenoviridae/metabolismo , Ensayos Clínicos como Asunto , Proteína 1 de la Respuesta de Crecimiento Precoz/genética , Proteína 1 de la Respuesta de Crecimiento Precoz/metabolismo , Vectores Genéticos/genética , Vectores Genéticos/efectos de la radiación , Humanos , Modelos Biológicos , Regiones Promotoras Genéticas , Radiación Ionizante , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Factores de Tiempo , Factor de Necrosis Tumoral alfa/efectos de la radiación , Factor de Necrosis Tumoral alfa/uso terapéutico
15.
Ann Oncol ; 14(4): 564-9, 2003 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-12649102

RESUMEN

BACKGROUND: Since 1990, we have treated patients with advanced nasopharyngeal cancer with induction chemotherapy and concomitant chemoradiotherapy. We herein report the results of our experience. PATIENTS AND METHODS: From 1990 to 1999, 27 patients with locoregionally advanced nasopharyngeal cancer were treated with induction chemotherapy followed by concomitant chemoradiotherapy. Using the American Joint Committee on Cancer's 1992 stage classification, all patients were stage III (11%) or IV (89%). By histology, 63% were poorly differentiated carcinoma and 37% squamous cell carcinoma. The median age was 42 years. Three cycles of induction chemotherapy consisting of cisplatin, 5-fluorouracil, leucovorin and interferon-alpha2b were administered, followed by concomitant chemoradiotherapy consisting of seven cycles of 5-fluorouracil, hydroxyurea and once-daily radiotherapy (FHX) on a week-on week-off schedule. The median radiotherapy dose was 70 Gy. RESULTS: Clinical response to induction chemotherapy was 100%, 54.2% complete response (CR) and 45.8% partial response. Clinical and/or pathological (37% of all patients had post-treatment biopsy with or without neck dissection) CR after FHX was 100%. At a median follow-up of 52 months, three failures were observed. Two patients have died of disease, one of local failure and one of distant metastases. One patient is alive with an isolated rib metastasis. At 5 years, actuarial locoregional control is 93% and actuarial distant control 92%. The overall survival at 3 and 5 years is 88% and 77%, respectively. Four patients died of unrelated illnesses and had no evidence of disease with respect to their nasopharyngeal cancer. The progression-free survival at 3 and 5 years is 92% and 86%, respectively. Thirty-three per cent of patients required a reduction in the chemotherapy dose due to acute toxicity. Chronic toxicity was not observed, with all patients able to eat orally without dietary restrictions. CONCLUSIONS: Treatment of locoregionally advanced nasopharyngeal cancer with induction chemotherapy followed by concomitant chemoradiotherapy resulted in excellent overall survival with acceptable toxicity. These results are encouraging and warrant further investigation of intensified approaches.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Carcinoma de Células Escamosas/tratamiento farmacológico , Carcinoma de Células Escamosas/radioterapia , Carcinoma/tratamiento farmacológico , Carcinoma/radioterapia , Neoplasias Nasofaríngeas/tratamiento farmacológico , Neoplasias Nasofaríngeas/radioterapia , Adulto , Anciano , Protocolos de Quimioterapia Combinada Antineoplásica/efectos adversos , Carcinoma/patología , Carcinoma de Células Escamosas/patología , Cisplatino/administración & dosificación , Terapia Combinada , Supervivencia sin Enfermedad , Femenino , Fluorouracilo/administración & dosificación , Humanos , Hidroxiurea/administración & dosificación , Interferón-alfa/administración & dosificación , Leucovorina/administración & dosificación , Masculino , Persona de Mediana Edad , Neoplasias Nasofaríngeas/patología , Terapia Neoadyuvante , Estadificación de Neoplasias , Resultado del Tratamiento
16.
Gene Ther ; 10(3): 206-12, 2003 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-12571627

RESUMEN

The efficacy of replication-deficient adenoviral vectors in gene therapy is confined to the number of tumor cells the vector infects. To focus and enhance the therapeutic efficacy, we employed a conditionally replication-competent adenoviral vector with a tissue-specific promoter, DF3/MUC1, in a human esophageal adenocarcinoma model. Our results demonstrate that Ad.DF3.E1A.CMV.TNF (Ad.DF3.TNF) specifically replicates in Bic-1 (DF3-producing cells) and mediates an enhanced biologic effect due to increased TNF-alpha in the same DF3-producing cells. We also show that the increased TNF-alpha interacts with ionizing radiation to produce greater tumor regression and a greater delay in tumor regrowth in Bic-1 (DF3-producing cells) compared to Seg-1 (DF3 non-producers). Tumor cell targeting using conditionally replication-competent adenoviral vectors with tumor-specific promoters to drive viral replication and deliver TNF-alpha provides a novel approach to enhancing tumor radiosensitivity.


Asunto(s)
Adenocarcinoma/terapia , Neoplasias Esofágicas/terapia , Terapia Genética/métodos , Mucina-1/genética , Fragmentos de Péptidos/genética , Adenocarcinoma/inmunología , Adenocarcinoma/radioterapia , Adenoviridae/genética , Animales , Terapia Combinada , Neoplasias Esofágicas/inmunología , Neoplasias Esofágicas/radioterapia , Vectores Genéticos/administración & dosificación , Proteínas Fluorescentes Verdes , Humanos , Proteínas Luminiscentes/genética , Ratones , Ratones Desnudos , Modelos Animales , Mucina-1/análisis , Fragmentos de Péptidos/análisis , Regiones Promotoras Genéticas , Distribución Aleatoria , Trasplante Heterólogo , Células Tumorales Cultivadas , Factor de Necrosis Tumoral alfa/biosíntesis , Replicación Viral
17.
Clin Microbiol Infect ; 8(9): 551-63, 2002 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-12427216

RESUMEN

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.


Asunto(s)
Terapia Genética/métodos , Vectores Genéticos , Neoplasias/terapia , Simplexvirus/genética , Antineoplásicos/uso terapéutico , Ensayos Clínicos Fase I como Asunto , Terapia Combinada , Virus Defectuosos/genética , Técnicas de Transferencia de Gen , Humanos , Neoplasias/tratamiento farmacológico , Neoplasias/radioterapia , Simplexvirus/enzimología , Transcripción Genética , Replicación Viral
18.
Gene Ther ; 9(1): 75-80, 2002 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-11850725

RESUMEN

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.


Asunto(s)
Terapia Genética/métodos , Vacunas contra el Virus del Herpes Simple/administración & dosificación , Herpesvirus Humano 1 , Herpesvirus Humano 2 , Neoplasias Hepáticas Experimentales/terapia , Animales , Terapia Combinada , Humanos , Neoplasias Hepáticas Experimentales/radioterapia , Ratones , Ratones Desnudos , Trasplante de Neoplasias , Células Tumorales Cultivadas , Replicación Viral/efectos de la radiación
20.
Prostate Cancer Prostatic Dis ; 5(4): 296-303, 2002.
Artículo en Inglés | MEDLINE | ID: mdl-12627215

RESUMEN

The limiting factor for radiation (RT) dose-escalation is normal tissue toxicity. In dose-escalation studies, it is important to determine the factors associated with toxicity and the length of follow-up period after which a particular RT dose is considered safe. We analyzed 449 prostate cancer patients treated with RT at our institution and followed for a median of 27 months. Genitourinary (GU) and gastrointerological (GI) complications were graded and analyzed using three different statistical models. Univariate and multivariate analyses were conducted for factors associated with toxicity. There was no RTOG grade 4 or 5 toxicity. Only 23 patients (5%) experienced grade 3 toxicity. After treatment, there was an initial rapid decline in the risk of toxicity following treatment, followed by an increase or stabilization of the toxicity with time of follow-up. The breakpoints between the two periods were 2 y (any toxicity) and 1 y (high toxicity) for GU and 9 months (any toxicity, high toxicity) for GI. Age, dose, fraction size, duration of treatment and hospital of treatment emerge as important factors in the probability of developing toxicity. Our study shows that delivering conventional doses using conformal techniques is associated with minimal high-grade toxicity. However, even within a narrow dose range and fraction size used, differences do emerge which should lead one to be cautious in extending the results of dose escalation study to the community practice without a sufficient follow-up.


Asunto(s)
Adenocarcinoma/radioterapia , Enfermedades Gastrointestinales/etiología , Enfermedades Urogenitales Masculinas/etiología , Neoplasias de la Próstata/radioterapia , Traumatismos por Radiación/etiología , Radioterapia Conformacional/efectos adversos , Población Negra , Chicago/epidemiología , Fraccionamiento de la Dosis de Radiación , Estudios de Seguimiento , Enfermedades Gastrointestinales/epidemiología , Humanos , Masculino , Enfermedades Urogenitales Masculinas/epidemiología , Traumatismos por Radiación/epidemiología , Dosificación Radioterapéutica , Índice de Severidad de la Enfermedad , Factores Socioeconómicos , Población Blanca
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...