Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Dis Model Mech ; 8(4): 393-401, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25713296

RESUMEN

Treatment of non-small cell lung cancer (NSCLC) is based on histological analysis and molecular profiling of targetable driver oncogenes. Therapeutic responses are further defined by the landscape of passenger mutations, or loss of tumor suppressor genes. We report here a thorough study to address the physiological role of the putative lung cancer tumor suppressor EPH receptor A3 (EPHA3), a gene that is frequently mutated in human lung adenocarcinomas. Our data shows that homozygous or heterozygous loss of EphA3 does not alter the progression of murine adenocarcinomas that result from Kras mutation or loss of Trp53, and we detected negligible postnatal expression of EphA3 in adult wild-type lungs. Yet, EphA3 was expressed in the distal mesenchyme of developing mouse lungs, neighboring the epithelial expression of its Efna1 ligand; this is consistent with the known roles of EPH receptors in embryonic development. However, the partial loss of EphA3 leads only to subtle changes in epithelial Nkx2-1, endothelial Cd31 and mesenchymal Fgf10 RNA expression levels, and no macroscopic phenotypic effects on lung epithelial branching, mesenchymal cell proliferation, or abundance and localization of CD31-positive endothelia. The lack of a discernible lung phenotype in EphA3-null mice might indicate lack of an overt role for EPHA3 in the murine lung, or imply functional redundancy between EPHA receptors. Our study shows how biological complexity can challenge in vivo functional validation of mutations identified in sequencing efforts, and provides an incentive for the design of knock-in or conditional models to assign the role of EPHA3 mutation during lung tumorigenesis.


Asunto(s)
Carcinogénesis/genética , Genes Supresores de Tumor , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patología , Morfogénesis/genética , Receptor EphA3/genética , Adenocarcinoma/genética , Adenocarcinoma/patología , Adenocarcinoma del Pulmón , Animales , Carcinogénesis/patología , Regulación del Desarrollo de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Humanos , Pulmón/embriología , Pulmón/patología , Mesodermo/metabolismo , Ratones , Mutación , Proteínas Proto-Oncogénicas p21(ras)/metabolismo , Receptor EphA3/metabolismo , Proteína p53 Supresora de Tumor/metabolismo
2.
J Gene Med ; 13(5): 253-61, 2011 May.
Artículo en Inglés | MEDLINE | ID: mdl-21520358

RESUMEN

BACKGROUND: Oncolytic adenoviruses are an attractive strategy for treating cancers resistant to conventional treatments. However, their systemic utility could be limited as a result of the high prevalence of pre-existing immunity towards the vector. Furthermore, neutralizing antibodies (NAbs) may prevent successful intravenous readministration of the same agent. Previous preclinical reports indicate that the NAb response can be partially overcome by modifying the adenoviral fiber knob. However, to date, this strategy has not been evaluated in human patients. METHODS: Twenty-four human patients with advanced cancer were treated with two cycles of oncolytic adenoviruses, featuring three capsid variants: unmodified adenovirus serotype 5 (Ad5), serotype 5 with RGD motif in the HI-loop of the fiber knob (Ad5-RGD) and serotype 5 carrying fiber knob from serotype 3 (Ad5/3). A virus with different fiber structure was used for the second round of treatment and patient serum was analyzed for a neutralizing effect. RESULTS: All patients developed NAbs against the virus that they were treated with. However, the magnitude and velocity of the response varied considerably. When measured just before the second treatment cycle, a differential in serum NAb titer against the first versus second virus was seen in 83% of cases, suggesting that even minor changes in the fiber knob can circumvent neutralization in cancer patients. No correlation between NAb titers and outcome variables was observed. CONCLUSIONS: The results obtained in the present study extend previous preclinical reports into human cancer patients and suggest that modification of the fiber knob is a feasible strategy for circumventing the NAb response in patients receiving multiple rounds of oncolytic adenoviruses.


Asunto(s)
Adenoviridae/inmunología , Anticuerpos Neutralizantes/inmunología , Anticuerpos Antivirales/inmunología , Proteínas de la Cápside/inmunología , Vectores Genéticos/inmunología , Neoplasias , Viroterapia Oncolítica , Adenoviridae/genética , Adolescente , Adulto , Anciano , Terapia Genética , Humanos , Persona de Mediana Edad , Neoplasias/inmunología , Neoplasias/terapia , Resultado del Tratamiento , Adulto Joven
3.
Cancer Res ; 70(11): 4297-309, 2010 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-20484030

RESUMEN

Granulocyte macrophage colony-stimulating factor (GMCSF) can mediate antitumor effects by recruiting natural killer cells and by induction of tumor-specific cytotoxic T-cells through antigen-presenting cells. Oncolytic tumor cell-killing can produce a potent costimulatory danger signal and release of tumor epitopes for antigen-presenting cell sampling. Therefore, an oncolytic adenovirus coding for GMCSF was engineered and shown to induce tumor-specific immunity in an immunocompetent syngeneic hamster model. Subsequently, 20 patients with advanced solid tumors refractory to standard therapies were treated with Ad5-D24-GMCSF. Of the 16 radiologically evaluable patients, 2 had complete responses, 1 had a minor response, and 5 had disease stabilization. Responses were frequently seen in injected and noninjected tumors. Treatment was well tolerated and resulted in the induction of both tumor-specific and virus-specific immunity as measured by ELISPOT and pentamer analysis. This is the first time that oncolytic virus-mediated antitumor immunity has been shown in humans. Ad5-D24-GMCSF is promising for further clinical testing.


Asunto(s)
Adenoviridae/genética , Factor Estimulante de Colonias de Granulocitos/genética , Inmunoterapia/métodos , Neoplasias/terapia , Viroterapia Oncolítica/métodos , Adenoviridae/inmunología , Adenoviridae/metabolismo , Animales , Cricetinae , Epítopos de Linfocito T/inmunología , Factor Estimulante de Colonias de Granulocitos/biosíntesis , Factor Estimulante de Colonias de Granulocitos/inmunología , Humanos , Proteínas Inhibidoras de la Apoptosis , Proteínas Asociadas a Microtúbulos/inmunología , Neoplasias/genética , Neoplasias/inmunología , Neoplasias/virología , Survivin , Linfocitos T/inmunología , Transfección
4.
Clin Cancer Res ; 16(11): 3035-43, 2010 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-20501623

RESUMEN

PURPOSE: Twenty-one patients with cancer were treated with a single round of oncolytic adenovirus ICOVIR-7. EXPERIMENTAL DESIGN: ICOVIR-7 features an RGD-4C modification of the fiber HI-loop of serotype 5 adenovirus for enhanced entry into tumor cells. Tumor selectivity is mediated by an insulator, a modified E2F promoter, and a Rb-binding site deletion of E1A, whereas replication is optimized with E2F binding hairpins and a Kozak sequence. ICOVIR-7 doses ranged from 2 x 10(10) to 1 x 10(12) viral particles. All patients had advanced and metastatic solid tumors refractory to standard therapies. RESULTS: ICOVIR-7 treatment was well tolerated with mild to moderate fever, fatigue, elevated liver transaminases, chills, and hyponatremia. One patient had grade 3 anemia but no other serious side effects were seen. At baseline, 9 of 21 of patients had neutralizing antibody titers against the ICOVIR-7 capsid. Treatment resulted in neutralizing antibody titer induction within 4 weeks in 16 of 18 patients. No elevations of serum proinflammatory cytokine levels were detected. Viral genomes were detected in the circulation in 18 of 21 of patients after injection and 7 of 15 of the samples were positive 2 to 4 weeks later suggesting viral replication. CONCLUSIONS: Overall, objective evidence of antitumor activity was seen in 9 of 17 evaluable patients. In radiological analyses, 5 of 12 evaluable patients had stabilization or reduction in tumor size. These consisted of one partial response, two minor responses and two cases of stable disease, all occurring in patients who had progressive disease before treatment. In summary, ICOVIR-7 treatment is apparently safe, resulting in anticancer activity, and is therefore promising for further clinical testing.


Asunto(s)
Adenoviridae , Neoplasias/terapia , Viroterapia Oncolítica/métodos , Adolescente , Adulto , Anciano , Anticuerpos Antivirales/análisis , Niño , Femenino , Humanos , Interleucinas/sangre , Masculino , Persona de Mediana Edad , Viroterapia Oncolítica/efectos adversos , Retratamiento , Resultado del Tratamiento , Replicación Viral
6.
J Gene Med ; 10(7): 744-53, 2008 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-18338835

RESUMEN

BACKGROUND: Cyclo-oxygenase 2 (Cox-2) is expressed in many types of tumors, but typically undetectable in normal tissues. However, Cox-2 is known to be induced following infection by many microbial agents, which might threaten the tumor selectivity of the Cox-2 promoter in the context of virotherapy or viral gene delivery. Cox-2 expression is regulated in part post-transcriptionally by stimulation or inhibition of mRNA degradation by 3'-untranslated region (3'-UTR) AU-rich elements. In the present study, we investigated the induction of the Cox-2 promoter both in normal and tumor cells after adenovirus infection and explored the utility of AU-rich elements for regaining promoter selectivity. METHODS: Nontumor and tumor cells were transfected in vitro and in vivo with plasmids containing the Cox-2 or cytomegalovirus immediate early promoter driving luciferase (with or without 3'-UTR elements) followed by adenoviral infection. Selectivity and activity of the promoters and 3'-UTR elements were analysed by luciferase assay and in-vivo imaging. RESULTS: The Cox-2 promoter was induced in both normal and tumor cells following infection with E1 containing replicative adenoviruses but not in the absence of E1. Utilization of AU-rich elements counteracted promoter induction in vitro and in vivo in nonmalignant cells but not in cancer cells, thus increasing the selectivity of the approach ten-fold without loss of potency. CONCLUSIONS: Adenoviral infection induces the Cox-2 promoter in normal and tumor cells, which might compromise specificity of the promoter. Utilization of AU-rich destabilization elements can rescue the tumor selectivity of the promoter.


Asunto(s)
Infecciones por Adenovirus Humanos/metabolismo , Ciclooxigenasa 2/genética , Regulación de la Expresión Génica/genética , Regiones Promotoras Genéticas/genética , Regiones no Traducidas 3'/metabolismo , Infecciones por Adenovirus Humanos/genética , Línea Celular Tumoral , Cartilla de ADN/genética , Humanos , Luciferasas , Plásmidos/genética , Transfección
7.
Gynecol Oncol ; 108(1): 166-72, 2008 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-17950450

RESUMEN

OBJECTIVE: Oncolytic adenoviruses capable of replication selectively in tumor cells are an appealing approach for the treatment of neoplastic diseases refractory to conventional therapies. The aim of this study was to evaluate the effect of dose and scheduling of a tropism-modified, adenovirus serotype 3 receptor-targeted, Rb/p16 pathway-selective replication-competent adenovirus, Ad5/3-delta24, against human ovarian adenocarcinoma. As oncolytic viruses and chemotherapy can have synergistic interactions, the antitumor efficacy of Ad5/3-delta24 was also studied in combination with epirubicin and gemcitabine, common second-line treatment options for platinum-resistant ovarian cancer. METHODS: Orthotopic murine models of peritoneally disseminated ovarian cancer were utilized to compare survival of mice treated with either a single viral dose or weekly delivery. The lowest effective dose of intraperitoneal Ad5/3-delta24 was determined. Combinations of Ad5/3-delta24 and gemcitabine or epirubicin were studied in vitro as well as in vivo. RESULTS: Treatment outcome after administration of a single dose of Ad5/3-delta24 was as effective as delivery of several weekly doses. Our results also demonstrate that a single intraperitoneal injection of 100 viral particles significantly increased the survival of mice compared to untreated animals. Further, combining Ad5/3-delta24 with either gemcitabine or epirubicin resulted in greater therapeutic benefit than either agent alone. CONCLUSION: These preclinical data suggest that Ad5/3-delta24 represents a promising treatment strategy for advanced ovarian cancer as a single agent or in combination with chemotherapy.


Asunto(s)
Adenocarcinoma/terapia , Adenoviridae/fisiología , Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Desoxicitidina/análogos & derivados , Epirrubicina/farmacología , Viroterapia Oncolítica/métodos , Neoplasias Ováricas/terapia , Adenocarcinoma/tratamiento farmacológico , Adenocarcinoma/virología , Adenoviridae/genética , Animales , Antibióticos Antineoplásicos/administración & dosificación , Antibióticos Antineoplásicos/farmacología , Antimetabolitos Antineoplásicos/administración & dosificación , Antimetabolitos Antineoplásicos/farmacología , Línea Celular Tumoral , Terapia Combinada , Desoxicitidina/administración & dosificación , Desoxicitidina/farmacología , Esquema de Medicación , Epirrubicina/administración & dosificación , Femenino , Humanos , Ratones , Ratones SCID , Neoplasias Ováricas/tratamiento farmacológico , Neoplasias Ováricas/virología , Replicación Viral , Ensayos Antitumor por Modelo de Xenoinjerto , Gemcitabina
8.
Mol Cancer Ther ; 6(10): 2728-36, 2007 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-17938266

RESUMEN

Renal cancer is a common and deadly disease that lacks curative treatments when metastatic. Here, we have used oncolytic adenoviruses, a promising developmental approach whose safety has recently been validated in clinical trials. Although preliminary clinical efficacy data exist for selected tumor types, potency has generally been less than impressive. One important reason may be that expression of the primary receptor, coxsackie-adenovirus receptor, is often low on many or most advanced tumors, although not evaluated in detail with renal cancer. Here, we tested if fluorescence-assisted cell sorting could be used to predict efficacy of a panel of infectivity-enhanced capsid-modified marker gene expressing adenoviruses in renal cancer cell lines, clinical specimens, and subcutaneous and orthotopic murine models of peritoneally metastatic renal cell cancer. The respective selectively oncolytic adenoviruses were tested for killing of tumor cells in these models, and biodistribution after locoregional delivery was evaluated. In vivo replication was analyzed with noninvasive imaging. Ad5/3-Delta24, Ad5-Delta24RGD, and Ad5.pK7-Delta24 significantly increased survival of mice compared with mock or wild-type virus and 50% of Ad5/3-Delta24 treated mice were alive at 320 days. Because renal tumors are often highly vascularized, we investigated if results could be further improved by adding bevacizumab, a humanized antivascular endothelial growth factor antibody. The combination was well tolerated but did not improve survival, suggesting that the agents may be best used in sequence instead of together. These results set the stage for clinical testing of oncolytic adenoviruses for treatment of metastatic renal cancer currently lacking other treatment options.


Asunto(s)
Adenoviridae/genética , Cápside/metabolismo , Carcinoma de Células Renales/terapia , Neoplasias Renales/terapia , Viroterapia Oncolítica , Neoplasias Peritoneales/terapia , Transgenes/fisiología , Inhibidores de la Angiogénesis/uso terapéutico , Animales , Anticuerpos Monoclonales/uso terapéutico , Anticuerpos Monoclonales Humanizados , Apoptosis/efectos de los fármacos , Bevacizumab , Cápside/química , Carcinoma de Células Renales/secundario , Terapia Combinada , Proteína de la Membrana Similar al Receptor de Coxsackie y Adenovirus , Modelos Animales de Enfermedad , Femenino , Citometría de Flujo , Técnicas de Transferencia de Gen , Proteoglicanos de Heparán Sulfato/metabolismo , Humanos , Neoplasias Renales/patología , Luciferasas/metabolismo , Ratones , Ratones Desnudos , Ratones SCID , Neoplasias Peritoneales/secundario , Receptores Virales/metabolismo , Células Tumorales Cultivadas , Factor A de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Factor A de Crecimiento Endotelial Vascular/inmunología , Replicación Viral , Ensayos Antitumor por Modelo de Xenoinjerto
9.
Hum Gene Ther ; 18(7): 627-41, 2007 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-17604566

RESUMEN

Systemic adenoviral delivery into tumors is inefficient because of liver sequestration of intravenously administered virus. One potential solution for improving bioavailability is the use of carrier cells such as human mesenchymal stem cells (MSCs), which have been suggested to have inherent tumor tropism. Here we investigated the capacity of capsid-modified adenoviruses to infect and replicate in MSCs. Further, biodistribution and tumor-killing efficacy of MSCs loaded with oncolytic adenoviruses were evaluated in orthotopic murine models of lung and breast cancer. In vitro, heparan sulfate proteoglycan- and alpha(v)beta integrin-targeted viruses enhanced gene delivery to bone marrow- and adipose tissue-derived MSCs up to 11,000-fold over adenovirus serotype 5 (Ad5). Infectivity-enhanced oncolytic adenoviruses showed notably higher rates of cytolysis of in vitro-passaged MSCs in comparison with wild-type virus. In vivo, intravenously injected MSCs homed primarily to the lungs, and virus was released into advanced orthotopic breast and lung tumors for therapeutic efficacy and increased survival. When the same dose of virus was injected intravenously without MSCs, only transduction of the liver was seen. These results suggest that MSCs loaded with oncolytic adenoviruses might be a useful approach for improving the bioavailability of systemically administered oncolytic adenoviruses.


Asunto(s)
Adenoviridae/genética , Neoplasias de la Mama/terapia , Terapia Genética/métodos , Neoplasias Pulmonares/terapia , Células Madre Mesenquimatosas/fisiología , Viroterapia Oncolítica , Tropismo , Animales , Supervivencia Celular , Células Cultivadas , Femenino , Humanos , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas/citología , Ratones , Ratones Desnudos , Transducción Genética , Replicación Viral
10.
Mol Cancer Ther ; 6(2): 742-51, 2007 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-17308070

RESUMEN

Hormone refractory metastatic prostate cancer is a deadly disease that currently lacks curative treatments. Conditionally replicating adenoviruses (CRAds) are promising new agents against cancer due to their innate capability to cause oncolysis of tumor cells. Their antitumor effect is determined in part by their capacity for infecting cancer cells. However, the respective primary receptor, the coxsackie-adenovirus receptor (CAR), is variably expressed in many cancer types. We created Ad5/3Delta24hCG, a novel CRAd retargeted to the adenovirus serotype 3 receptor, which has been reported to be highly expressed in tumors. Furthermore, we added a transgene for the beta-chain of human chorionic gonadotropin (hCGbeta), whose expression was tightly coupled to virus replication. Ad5/3Delta24hCG was found effective in killing prostate cancer cells, and oncolysis was seen in concordance with hCGbeta production. In a s.c. in vivo model of hormone refractory prostate cancer, Ad5/3Delta24hCG treatment resulted in statistically significant tumor growth inhibition. Moreover, i.v. injection of Ad5/3Delta24hCG prolonged the survival of mice with hormone refractory prostate cancer metastatic to the lung. Detection of hCGbeta in serum samples confirmed viral replication in vivo. Infection of human clinical samples of cancerous and normal prostatic tissue resulted in effective hCGbeta production in cancer tissue, whereas it remained low in nonmalignant tissue, suggesting cancer-specific replication. These results suggest that Ad5/3Delta24hCG is a potent virus for the treatment of hormone refractory prostate cancer in vitro and in vivo. These preclinical data set the stage for translation into clinical studies.


Asunto(s)
Adenoviridae/fisiología , Gonadotropina Coriónica/farmacología , Neoplasias Pulmonares/terapia , Viroterapia Oncolítica , Neoplasias de la Próstata/terapia , Replicación Viral , Animales , Supervivencia Celular , Células Cultivadas , Gonadotropina Coriónica/genética , Proteína de la Membrana Similar al Receptor de Coxsackie y Adenovirus , ADN Viral/genética , Terapia Genética , Vectores Genéticos , Humanos , Riñón/metabolismo , Riñón/virología , Neoplasias Pulmonares/secundario , Neoplasias Pulmonares/virología , Masculino , Ratones , Ratones Desnudos , Oligopéptidos/metabolismo , Neoplasias de la Próstata/metabolismo , Neoplasias de la Próstata/virología , Receptores Virales/metabolismo , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
11.
Int J Cancer ; 121(1): 165-74, 2007 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-17315187

RESUMEN

Conditionally replicating oncolytic adenoviruses represent a promising developmental strategy for the treatment of cancer refractory to current treatments, such as hormone refractory metastatic breast cancer. In clinical cancer trials, adenoviral agents have been well tolerated, but gene transfer has been insufficient for clinical benefit. One of the main reasons may be the deficiency of the primary adenovirus receptor, and therefore viral capsid modifications have been employed. Another obstacle to systemic delivery is rapid clearance of virus by hepatic Kupffer cells and subsequent inadequate bioavailability. In this study, we compared several capsid-modified oncolytic adenoviruses for the treatment of breast cancer with and without Kupffer cell inactivation. Replication deficient capsid-modified viruses were analyzed for their gene transfer efficacy in vitro in breast cancer cell lines and clinical samples and in vivo in orthotopic models of breast cancer. The effect of Kupffer cell depleting agents on gene transfer efficacy in vivo was evaluated. An aggressive lung metastatic model was developed to study the effect of capsid-modified oncolytic adenoviruses on survival. Capsid-modified viruses displayed increased gene transfer and cancer cell killing in vitro and resulted in increased survival in an orthotopic model of lung metastatic breast cancer in mice. Biodistribution of viruses was favorable, tumor burden and treatment response could be monitored repeatedly. Kuppfer cell inactivation led to enhanced systemic gene delivery, but did not increase the survival of mice. These results facilitate clinical translation of oncolytic adenoviruses for the treatment of hormone refractory metastatic breast cancer.


Asunto(s)
Adenoviridae/patogenicidad , Neoplasias de la Mama/patología , Neoplasias de la Mama/terapia , Viroterapia Oncolítica , Animales , Neoplasias de la Mama/virología , Proteínas de la Cápside/genética , Proteínas de la Cápside/metabolismo , Línea Celular , Modelos Animales de Enfermedad , Gadolinio , Técnicas de Transferencia de Gen , Hormonas/metabolismo , Humanos , Liposomas , Ratones , Metástasis de la Neoplasia/patología , Poli I , Tasa de Supervivencia , Replicación Viral , Ensayos Antitumor por Modelo de Xenoinjerto
12.
J Gene Med ; 9(1): 3-9, 2007 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-17149790

RESUMEN

BACKGROUND: Adenoviruses can cause severe toxicity in immunocompromised individuals. Although clinical trials have confirmed the potency and safety of selectively oncolytic adenoviruses for treatment of advanced cancers, increasingly effective agents could result in more toxicity and therefore it would be useful if replication could be abrogated if necessary. METHODS: We analyzed the effect of chlorpromazine, an inhibitor of clathrin-dependent endocytosis and apigenin, a cell cycle regulator, on adenovirus replication and toxicity. First, we evaluated the in vitro replication of a tumor targeted Rb-p16 pathway selective oncolytic adenovirus (Ad5/3-Delta24) and a wild-type adenovirus in normal cells, fresh liver samples and in ovarian cancer cell lines. Further, we analyzed the in vitro cell killing efficacy of adenoviruses in the presence and absence of the substances. Moreover, the effect on in vivo efficacy, replication and liver toxicity of the adenoviruses was evaluated. RESULTS: We demonstrate in vitro and in vivo reduction of adenovirus replication and associated toxicity with chlorpromazine and apigenin. Effective doses were well within what would be predicted safe in humans. CONCLUSIONS: Chlorpromazine and apigenin might reduce the replication of adenovirus, which could provide a safety switch in case replication-associated side effects are encountered in patients. In addition, these substances could be useful for the treatment of systemic adenoviral infections in immunosuppressed patients.


Asunto(s)
Adenoviridae/efectos de los fármacos , Apigenina/farmacología , Clorpromazina/farmacología , Virus Oncolíticos , Replicación Viral/efectos de los fármacos , Adenocarcinoma/tratamiento farmacológico , Adenocarcinoma/virología , Adenoviridae/genética , Animales , Femenino , Humanos , Hígado/efectos de los fármacos , Hígado/metabolismo , Hígado/virología , Ratones , Ratones Desnudos , Viroterapia Oncolítica/métodos , Neoplasias Ováricas/tratamiento farmacológico , Neoplasias Ováricas/virología , Pruebas de Toxicidad , Células Tumorales Cultivadas
13.
Cancer ; 107(7): 1578-88, 2006 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-16948124

RESUMEN

BACKGROUND: Variable expression of the coxsackie and adenovirus receptor (CAR) has limited gene transfer efficacy to many types of tumors. Consequently, tropism-modified adenoviruses have been developed for enhanced infectivity. To the authors' knowledge, targeting approaches for nonsmall cell lung cancer (NSCLC) have not been comprehensively evaluated. The current hypothesis was that modified adenoviruses could be used for increasing gene transfer to and killing of NSCLC cells in vitro and in vivo. METHODS: Ten NSCLC cell lines were analyzed to represent the different NSCLC histologies. Because clinical tumors may differ from established cell lines, 6 clinical specimens fresh from patients were analyzed. For in vivo studies, a novel orthotopic murine model of advanced lung cancer was developed. Because tumor response is difficult to quantitate in orthotopic models, noninvasive imaging of green fluorescent protein (GFP) was utilized as a surrogate for tumor size measurements. RESULTS: Adenoviruses whose capsids were modified with RGD-4C, the serotype 3 knob, or polylysine displayed increased gene transfer to NSCLC cell lines and clinical samples in comparison to serotype 5 viruses. Conditionally replicating oncolytic adenoviruses (CRAds) with the same modifications showed enhanced therapeutic efficiency in vitro and in vivo. The median survival of mice treated with Ad5.pK7-Delta24 or Ad5-Delta24RGD increased 37% (P<.01). GFP imaging allowed noninvasive individualized detection of response and recurrence. CONCLUSIONS: Targeting of adenovirus to heterologous receptors can improve killing of NSCLC cells. Utilization of clinical samples and an orthotopic model of advanced lung cancer may provide clinically relevant translational data.


Asunto(s)
Adenoviridae/genética , Cápside/metabolismo , Carcinoma de Pulmón de Células no Pequeñas/terapia , Neoplasias Pulmonares/terapia , Viroterapia Oncolítica , Animales , Cápside/química , Femenino , Fluorometría , Técnicas de Transferencia de Gen , Proteínas Fluorescentes Verdes/análisis , Proteínas Fluorescentes Verdes/genética , Humanos , Ratones , Ratones Endogámicos , Oligopéptidos/química , Receptores Virales/metabolismo , Replicación Viral
14.
Clin Cancer Res ; 12(10): 3137-44, 2006 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-16707613

RESUMEN

PURPOSE: Metastatic gastric cancer remains a common and devastating disease without curative treatment. Recent proof-of-concept clinical trials have validated gene therapy with adenoviruses as an effective and safe modality for the treatment of cancer. However, expression of the primary coxsackie-adenovirus receptor is variable in advanced cancers, and therefore, the use of heterologous receptors could be advantageous. EXPERIMENTAL DESIGN: Here, we used capsid-modified adenoviruses for increasing the transduction and subsequent antitumor efficacy. 5/3 chimeric viruses have a serotype 3 knob which allows binding to a receptor distinct from coxsackie-adenovirus receptor. The fiber of Ad5lucRGD is modified with an integrin-targeted motif. Polylysine motifs, pK7 and pK21, bind to heparan sulfates. Oncolytic adenoviruses replicate in and kill tumor cells selectively. Gastric cancer cell lines and fresh clinical samples from patients were infected with transductionally targeted viruses. Capsid-modified oncolytic adenoviruses were used in cell killing experiments. To test viral transduction and therapeutic efficacy in vivo, we developed orthotopic mouse models featuring i.p. disseminated human gastric cancer, which allowed the evaluation of biodistribution and antitumor efficacy in a system similar to humans. RESULTS: Capsid modifications benefited gene transfer efficiency and cell killing in gastric cancer cell lines and clinical samples in vitro and in vivo. Modified oncolytic adenoviruses significantly increased the survival of mice with orthotopic gastric cancer. CONCLUSIONS: These preclinical data set the stage for the clinical evaluation of safety and efficacy in patients with disease refractory to current modalities.


Asunto(s)
Adenoviridae/patogenicidad , Cápside/metabolismo , Técnicas de Transferencia de Gen , Terapia Genética/métodos , Viroterapia Oncolítica , Neoplasias Gástricas/terapia , Animales , Muerte Celular , Supervivencia Celular , Proteína de la Membrana Similar al Receptor de Coxsackie y Adenovirus , Femenino , Ratones , Neoplasias Experimentales , Receptores Virales , Neoplasias Gástricas/patología , Transducción Genética , Células Tumorales Cultivadas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...