Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Mol Ther ; 22(5): 986-98, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24448161

RESUMEN

Human and canine cancer share similarities such as genetic and molecular aspects, biological complexity, tumor epidemiology, and targeted therapeutic treatment. Lack of good animal models for human adenovirotherapy has spurred the use of canine adenovirus 2-based oncolytic viruses. We have constructed a canine oncolytic virus that mimics the characteristics of our previously published human adenovirus ICOVIR17: expression of E1a controlled by E2F sites, deletion of the pRb-binding site of E1a, insertion of an RGD integrin-binding motif at the fiber Knob, and expression of hyaluronidase under the major late promoter/IIIa protein splicing acceptor control. Preclinical studies showed selectivity, increased cytotoxicity, and strong hyaluronidase activity. Intratumoral treatment of canine osteosarcoma and melanoma xenografts in mice resulted in inhibition of tumor growth and prolonged survival. Moreover, we treated six dogs with different tumor types, including one adenoma, two osteosarcomas, one mastocitoma, one fibrosarcoma, and one neuroendocrine hepatic carcinoma. No virus-associated adverse effects were observed, but toxicity associated to tumor lysis, including disseminated intravascular coagulation and systemic failure, was found in one case. Two partial responses and two stable diseases warrant additional clinical testing.


Asunto(s)
Melanoma/terapia , Viroterapia Oncolítica , Virus Oncolíticos/genética , Osteosarcoma/terapia , Adenovirus Caninos/genética , Animales , Perros , Humanos , Hialuronoglucosaminidasa/genética , Hialuronoglucosaminidasa/uso terapéutico , Melanoma/genética , Melanoma/veterinaria , Ratones , Osteosarcoma/genética , Osteosarcoma/veterinaria , Ensayos Antitumor por Modelo de Xenoinjerto
2.
Mol Ther ; 20(1): 54-62, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-21863000

RESUMEN

Tumor-associated stromal cells constitute a major hurdle in the antitumor efficacy with oncolytic adenoviruses. To overcome this biological barrier, an in vitro bioselection of a mutagenized AdwtRGD stock in human cancer-associated fibroblasts (CAFs) was performed. Several rounds of harvest at early cytopathic effect (CPE) followed by plaque isolation led us to identify one mutant with large plaque phenotype, enhanced release in CAFs and enhanced cytotoxicity in CAF and several tumor cell lines. Whole genome sequencing and functional mapping identified the truncation of the last 17 amino acids in C-terminal end of the i-leader protein as the mutation responsible for this phenotype. Similar mutations have been previously isolated in two independent bioselection processes in tumor cell lines. Importantly, our results establish the enhanced antitumor activity in vivo of the i-leader C-terminal truncated mutants, especially in a desmotic fibroblast-embedded lung carcinoma model in mice. These results indicate that the i-leader truncation represents a promising trait to improve virotherapy with oncolytic adenoviruses.


Asunto(s)
Adenoviridae/genética , Fibroblastos/metabolismo , Virus Oncolíticos/genética , Células del Estroma/metabolismo , Proteínas E3 de Adenovirus/genética , Animales , Apoptosis/genética , Efecto Citopatogénico Viral/genética , Modelos Animales de Enfermedad , Fibroblastos/patología , Humanos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/mortalidad , Neoplasias Pulmonares/terapia , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Ratones Desnudos , Mutación , Viroterapia Oncolítica , Selección Genética , Células del Estroma/patología , Microambiente Tumoral , Replicación Viral/genética , Ensayos Antitumor por Modelo de Xenoinjerto
3.
J Immunother Cancer ; 10(3)2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-35338084

RESUMEN

BACKGROUND: VCN-01 is an oncolytic adenovirus (Ad5 based) designed to replicate in cancer cells with dysfunctional RB1 pathway, express hyaluronidase to enhance virus intratumoral spread and facilitate chemotherapy and immune cells extravasation into the tumor. This phase I clinical trial was aimed to find the maximum tolerated dose/recommended phase II dose (RP2D) and dose-limiting toxicity (DLT) of the intravenous delivery of the replication-competent VCN-01 adenovirus in patients with advanced cancer. METHODS: Part I: patients with advanced refractory solid tumors received one single dose of VCN-01. Parts II and III: patients with pancreatic adenocarcinoma received VCN-01 (only in cycle 1) and nab-paclitaxel plus gemcitabine (VCN-concurrent on day 1 in Part II, and 7 days before chemotherapy in Part III). Patients were required to have anti-Ad5 neutralizing antibody (NAbs) titers lower than 1/350 dilution. Pharmacokinetic and pharmacodynamic analyses were performed. RESULTS: 26% of the patients initially screened were excluded based on high NAbs levels. Sixteen and 12 patients were enrolled in Part I and II, respectively: RP2D were 1×1013 viral particles (vp)/patient (Part I), and 3.3×1012 vp/patient (Part II). Fourteen patients were included in Part III: there were no DLTs and the RP2D was 1×1013 vp/patient. Observed DLTs were grade 4 aspartate aminotransferase increase in one patient (Part I, 1×1013 vp), grade 4 febrile neutropenia in one patient and grade 5 thrombocytopenia plus enterocolitis in another patient (Part II, 1×1013 vp). In patients with pancreatic adenocarcinoma overall response rate were 50% (Part II) and 50% (Part III). VCN-01 viral genomes were detected in tumor tissue in five out of six biopsies (day 8). A second viral plasmatic peak and increased hyaluronidase serum levels suggested replication after intravenous injection in all patients. Increased levels of immune biomarkers (interferon-γ, soluble lymphocyte activation gene-3, interleukin (IL)-6, IL-10) were found after VCN-01 administration. CONCLUSIONS: Treatment with VCN-01 is feasible and has an acceptable safety. Encouraging biological and clinical activity was observed when administered in combination with nab-paclitaxel plus gemcitabine to patients with pancreatic adenocarcinoma. TRIAL REGISTRATION NUMBER: NCT02045602.


Asunto(s)
Adenocarcinoma , Neoplasias Pancreáticas , Adenocarcinoma/patología , Adenoviridae/genética , Albúminas , Protocolos de Quimioterapia Combinada Antineoplásica/efectos adversos , Desoxicitidina/análogos & derivados , Humanos , Hialuronoglucosaminidasa/uso terapéutico , Paclitaxel , Neoplasias Pancreáticas/tratamiento farmacológico , Gemcitabina , Neoplasias Pancreáticas
4.
Mol Ther ; 18(7): 1275-83, 2010 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-20442708

RESUMEN

Successful virotherapy requires efficient virus spread within tumors. We tested whether the expression of hyaluronidase, an enzyme which dissociates the extracellular matrix (ECM), could enhance the intratumoral distribution of an oncolytic adenovirus and improve its therapeutic activity. As a proof of concept, we demonstrated that intratumoral coadministration of hyaluronidase in mice-bearing tumor xenografts improves the antitumor activity of an oncolytic adenovirus. Next, we constructed a replication-competent adenovirus expressing a soluble form of the human sperm hyaluronidase (PH20) under the control of the major late promoter (MLP) (AdwtRGD-PH20). Intratumoral treatment of human melanoma xenografts with AdwtRGD-PH20 resulted in degradation of hyaluronan (HA), enhanced viral distribution, and induced tumor regression in all treated tumors. Finally, the PH20 cDNA was inserted in an oncolytic adenovirus that selectively kills pRb pathway-defective tumor cells. The antitumoral activity of the novel oncolytic adenovirus expressing PH20 (ICOVIR17) was compared to that of the parental virus ICOVIR15. ICOVIR17 showed more antitumor efficacy following intratumoral and systemic administration in mice with prestablished tumors, along with an improved spread of the virus within the tumor. Importantly, a single intravenous dose of ICOVIR17 induced tumor regression in 60% of treated tumors. These results indicate that ICOVIR17 is a promising candidate for clinical testing.


Asunto(s)
Adenoviridae/fisiología , Regulación Enzimológica de la Expresión Génica , Hialuronoglucosaminidasa/metabolismo , Adenoviridae/genética , Animales , Bovinos , Línea Celular , Línea Celular Tumoral , Cricetinae , Femenino , Humanos , Hialuronoglucosaminidasa/genética , Inmunohistoquímica , Masculino , Melanoma/terapia , Mesocricetus , Ratones , Ratones Endogámicos BALB C , Viroterapia Oncolítica , Ensayos Antitumor por Modelo de Xenoinjerto
5.
Mol Ther ; 18(5): 903-11, 2010 May.
Artículo en Inglés | MEDLINE | ID: mdl-20179683

RESUMEN

The therapeutic potential of oncolytic adenoviruses is limited by the rate of adenovirus release. Based on the observation that several viruses induce cell death and progeny release by disrupting intracellular calcium homeostasis, we hypothesized that the alteration in intracellular calcium concentration induced by verapamil could improve the rate of virus release and spread, eventually enhancing the antitumoral activity of oncolytic adenoviruses. Our results indicate that verapamil substantially enhanced the release of adenovirus from a variety of cell types resulting in an improved cell-to-cell spread and cytotoxicity. Furthermore, the combination of the systemic administration of an oncolytic adenovirus (ICOVIR-5) with verapamil in vivo greatly improved its antitumoral activity in two different tumor xenograft models without affecting the selectivity of this virus. Overall, our findings indicate that verapamil provides a new, safe, and versatile way to improve the antitumoral potency of oncolytic adenoviruses in the clinical setting.


Asunto(s)
Adenoviridae/fisiología , Viroterapia Oncolítica/métodos , Virus Oncolíticos/fisiología , Verapamilo/uso terapéutico , Adenoviridae/efectos de los fármacos , Adenoviridae/genética , Animales , Western Blotting , Línea Celular , Línea Celular Tumoral , Vectores Genéticos , Humanos , Masculino , Ratones , Ratones Endogámicos BALB C , Neoplasias/terapia , Virus Oncolíticos/efectos de los fármacos , Virus Oncolíticos/genética , Liberación del Virus/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto
6.
Mol Ther ; 18(11): 1960-71, 2010 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-20808288

RESUMEN

Oncolytic adenoviruses are promising anticancer agents due to their ability to self-amplify at the tumor mass. However, tumor stroma imposes barriers difficult to overcome by these agents. Transgene expression is a valuable strategy to counteract these limitations and to enhance antitumor activity. For this purpose, the genetic backbone in which the transgene is inserted should be optimized to render transgene expression compatible with the adenovirus replication cycle and to keep genome size within the encapsidation size limit. In order to design a potent and selective oncolytic adenovirus that keeps intact all the viral functions with minimal increase in genome size, we inserted palindromic E2F-binding sites into the endogenous E1A promoter. The insertion of these sites controlling E1A-Δ24 results in a low systemic toxicity profile in mice. Importantly, the E2F-binding sites also increased the cytotoxicity and the systemic antitumor activity relative to wild-type adenovirus in all cancer models tested. The low toxicity and the increased potency results in improved antitumor efficacy after systemic injection and increased survival of mice carrying tumors. Furthermore, the constrained genome size of this backbone allows an efficient and potent expression of transgenes, indicating that this virus holds promise for overcoming the limitations of oncolytic adenoviral therapy.


Asunto(s)
Adenoviridae/genética , Proteínas E1A de Adenovirus/genética , Terapia Genética , Neoplasias/terapia , Virus Oncolíticos/fisiología , Regiones Promotoras Genéticas/genética , Proteína de Retinoblastoma/fisiología , Proteínas E1A de Adenovirus/metabolismo , Animales , Western Blotting , Efecto Citopatogénico Viral , Factor de Transcripción E2F1/metabolismo , Vectores Genéticos , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Neoplasias/genética , Neoplasias/virología , ARN Mensajero/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal , Transgenes/fisiología , Células Tumorales Cultivadas , Replicación Viral , Ensayos Antitumor por Modelo de Xenoinjerto
7.
J Control Release ; 332: 517-528, 2021 04 10.
Artículo en Inglés | MEDLINE | ID: mdl-33675877

RESUMEN

Tumor targeting and intratumoral virus spreading are key features for successful oncolytic virotherapy. VCN-11 is a novel oncolytic adenovirus, genetically modified to express hyaluronidase (PH20) and display an albumin-binding domain (ABD) on the hexon. ABD allows the virus to self-coat with albumin when entering the bloodstream and evade neutralizing antibodies (NAbs). Here, we validate VCN-11 mechanism of action and characterize its toxicity. VCN-11 replication, hyaluronidase activity and binding to human albumin to evade NAbs was evaluated. Toxicity and efficacy of VCN-11 were assessed in mice and hamsters. Tumor targeting, and antitumor activity was analyzed in the presence of NAbs in several tumor models. VCN-11 induced 450 times more cytotoxicity in tumor cells than in normal cells. VCN-11 hyaluronidase production was confirmed by measuring PH20 activity in vitro and in virus-infected tumor areas in vivo. VCN-11 evaded NAbs from different sources and tumor targeting was demonstrated in the presence of high levels of NAbs in vivo, whereas the control virus without ABD was neutralized. VCN-11 showed a low toxicity profile in athymic nude mice and Syrian hamsters, allowing treatments with high doses and fractionated administrations without major toxicities (up to 1.2x1011vp/mouse and 7.5x1011vp/hamster). Fractionated intravenous administrations improved circulation kinetics and tumor targeting. VCN-11 antitumor efficacy was demonstrated in the presence of NAbs against Ad5 and itself. Oncolytic adenovirus VCN-11 disrupts tumor matrix and displays antitumor effects even in the presence of NAbs. These features make VCN-11 a safe promising candidate to test re-administration in clinical trials.


Asunto(s)
Viroterapia Oncolítica , Virus Oncolíticos , Adenoviridae , Animales , Anticuerpos Neutralizantes , Línea Celular Tumoral , Cricetinae , Hialuronoglucosaminidasa , Ratones , Ratones Desnudos , Virus Oncolíticos/genética , Replicación Viral , Ensayos Antitumor por Modelo de Xenoinjerto
8.
J Immunother Cancer ; 9(11)2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-35149591

RESUMEN

BACKGROUND: Pancreatic ductal adenocarcinoma (PDAC) is characterized by dense desmoplastic stroma that limits the delivery of anticancer agents. VCN-01 is an oncolytic adenovirus designed to replicate in cancer cells with a dysfunctional RB1 pathway and express hyaluronidase. Here, we evaluated the mechanism of action of VCN-01 in preclinical models and in patients with pancreatic cancer. METHODS: VCN-01 replication and antitumor efficacy were evaluated alone and in combination with standard chemotherapy in immunodeficient and immunocompetent preclinical models using intravenous or intratumoral administration. Hyaluronidase activity was evaluated by histochemical staining and by measuring drug delivery into tumors. In a proof-of-concept clinical trial, VCN-01 was administered intratumorally to patients with PDAC at doses up to 1×1011 viral particles in combination with chemotherapy. Hyaluronidase expression was measured in serum by an ELISA and its activity within tumors by endoscopic ultrasound elastography. RESULTS: VCN-01 replicated in PDAC models and exerted antitumor effects which were improved when combined with chemotherapy. Hyaluronidase expression by VCN-01 degraded tumor stroma and facilitated delivery of a variety of therapeutic agents such as chemotherapy and therapeutic antibodies. Clinically, treatment was generally well-tolerated and resulted in disease stabilization of injected lesions. VCN-01 was detected in blood as secondary peaks and in post-treatment tumor biopsies, indicating virus replication. Patients had increasing levels of hyaluronidase in sera over time and decreased tumor stiffness, suggesting stromal disruption. CONCLUSIONS: VCN-01 is an oncolytic adenovirus with direct antitumor effects and stromal disruption capabilities, representing a new therapeutic agent for cancers with dense stroma. TRIAL REGISTRATION NUMBER: EudraCT number: 2012-005556-42 and NCT02045589.


Asunto(s)
Adenoviridae/genética , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Carcinoma Ductal Pancreático/terapia , Viroterapia Oncolítica/métodos , Neoplasias Pancreáticas/terapia , Células del Estroma/efectos de los fármacos , Albúminas/administración & dosificación , Animales , Carcinoma Ductal Pancreático/genética , Carcinoma Ductal Pancreático/patología , Terapia Combinada , Desoxicitidina/administración & dosificación , Desoxicitidina/análogos & derivados , Femenino , Humanos , Masculino , Mesocricetus , Ratones , Ratones Endogámicos C57BL , Paclitaxel/administración & dosificación , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/patología , Pronóstico , Gemcitabina
9.
Cancer Res ; 67(17): 8255-63, 2007 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-17804740

RESUMEN

During 2007, approximately 200,000 people in the United States will be diagnosed with brain tumors. Gliomas account for 77% of primary malignant brain tumors, and the prognosis has hardly changed in the past 20 years, with only 30% of patients with malignant glioma surviving 5 years after diagnosis. Oncolytic adenoviruses are promising therapies for the treatment of gliomas. Here, report the antiglioma activity of the tumor-selective ICOVIR-5 adenovirus, which encompasses an early 1A adenoviral (E1A) deletion in the retinoblastoma (Rb) protein-binding region, substitution of the E1A promoter for E2F-responsive elements, and an RGD-4C peptide motif inserted into the adenoviral fiber to enhance adenoviral tropism. Mechanistic studies showed a dramatic addiction of ICOVIR-5 to the E2F1 oncogene in vitro and in vivo. This addiction was mediated by the occupancy of the ectopic adenoviral E2F1-responsive elements by the endogenous E2F1 protein resulting in high level of E1A expression in cancer cells and potent antiglioma effect. Importantly, we showed for the first time the ability of oncolytic adenoviruses to enhance E2F transcriptional activity in vivo, and we provided direct evidence of the interaction of the E2F1 protein with native and ectopic adenovirus promoters. Restoration of Rb function led to the association of Rb/E2F1 repressor complexes with ICOVIR-5 ectopic E2F1 promoter and subsequent down-modulation of E1A, dramatically impairing adenoviral replication. In xenografted mice, intratumoral injection of ICOVIR-5 resulted in a significant improvement of the median survival (P < 0.0001), and furthermore, led to 37% of long-term survivors free of disease. The antitumor activity of ICOVIR-5 suggests that it has the potential to be an effective agent in the treatment of gliomas.


Asunto(s)
Adenoviridae , Proteínas E1A de Adenovirus/genética , Factor de Transcripción E2F1/metabolismo , Glioma/terapia , Viroterapia Oncolítica/métodos , Adenoviridae/genética , Adenoviridae/fisiología , Animales , Regulación Viral de la Expresión Génica , Glioma/patología , Glioma/virología , Humanos , Ratones , Ratones Desnudos , Virus Oncolíticos/genética , Virus Oncolíticos/fisiología , Proteína de Retinoblastoma/genética , Proteína de Retinoblastoma/metabolismo , Resultado del Tratamiento , Células Tumorales Cultivadas , Replicación Viral , Ensayos Antitumor por Modelo de Xenoinjerto
10.
Sci Rep ; 9(1): 14368, 2019 10 07.
Artículo en Inglés | MEDLINE | ID: mdl-31591461

RESUMEN

Last advances in the treatment of pediatric tumors has led to an increase of survival rates of children affected by primitive neuroectodermal tumors, however, still a significant amount of the patients do not overcome the disease. In addition, the survivors might suffer from severe side effects caused by the current standard treatments. Oncolytic virotherapy has emerged in the last years as a promising alternative for the treatment of solid tumors. In this work, we study the anti-tumor effect mediated by the oncolytic adenovirus VCN-01 in CNS-PNET models. VCN-01 is able to infect and replicate in PNET cell cultures, leading to a cytotoxicity and immunogenic cell death. In vivo, VCN-01 increased significantly the median survival of mice and led to long-term survivors in two orthotopic models of PNETs. In summary, these results underscore the therapeutic effect of VCN-01 for rare pediatric cancers such as PNETs, and warrants further exploration on the use of this virus to treat them.


Asunto(s)
Infecciones por Adenoviridae/genética , Adenoviridae/genética , Tumores Neuroectodérmicos Primitivos/genética , Virus Oncolíticos/genética , Infecciones por Adenoviridae/terapia , Infecciones por Adenoviridae/virología , Animales , Línea Celular Tumoral , Niño , Preescolar , Modelos Animales de Enfermedad , Humanos , Estimación de Kaplan-Meier , Ratones , Tumores Neuroectodérmicos Primitivos/terapia , Tumores Neuroectodérmicos Primitivos/virología , Viroterapia Oncolítica , Pediatría , Tasa de Supervivencia , Ensayos Antitumor por Modelo de Xenoinjerto
11.
Hum Gene Ther ; 30(3): 352-364, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30234393

RESUMEN

Oncolytic viruses represent a unique type of agents that combine self-amplification, lytic, and immunostimulatory properties against tumors. A local and locoregional clinical benefit has been demonstrated upon intratumoral injections of an oncolytic herpes virus in melanoma patients, leading to its approval in the United States and Europe for patients without visceral disease (up to stage IVM1a). However, in order to debulk and change the local immunosuppressive environment of tumors that cannot be injected directly, oncolyitc viruses need to be administered systemically. Among different viruses, adenovirus has been extensively used in clinical trials but with few evidences of activity upon systemic administration. Preclinical efficacy of a single intravenous administration of our oncolytic adenovirus ICOVIR5, an adenovirus type 5 responsive to the retinoblastoma pathway commonly deregulated in tumors, led us to use this virus in a dose-escalation phase 1 trial in metastatic melanoma patients. The results in 12 patients treated with a single infusion of a dose up to 1 × 1013 viral particles show that ICOVIR5 can reach melanoma metastases upon a single intravenous administration but fails to induce tumor regressions. These results support the systemic administration of armed oncolytic viruses to treat disseminated cancer.


Asunto(s)
Adenoviridae/genética , Terapia Genética , Vectores Genéticos/genética , Melanoma/genética , Melanoma/terapia , Viroterapia Oncolítica , Virus Oncolíticos/genética , Administración Intravenosa , Animales , Biopsia , Citocinas/metabolismo , Orden Génico , Terapia Genética/efectos adversos , Terapia Genética/métodos , Vectores Genéticos/administración & dosificación , Humanos , Melanoma/diagnóstico , Ratones , Viroterapia Oncolítica/efectos adversos , Viroterapia Oncolítica/métodos , Distribución Tisular , Resultado del Tratamiento , Replicación Viral
12.
Sci Transl Med ; 11(476)2019 01 23.
Artículo en Inglés | MEDLINE | ID: mdl-30674657

RESUMEN

Retinoblastoma is a pediatric solid tumor of the retina activated upon homozygous inactivation of the tumor suppressor RB1 VCN-01 is an oncolytic adenovirus designed to replicate selectively in tumor cells with high abundance of free E2F-1, a consequence of a dysfunctional RB1 pathway. Thus, we reasoned that VCN-01 could provide targeted therapeutic activity against even chemoresistant retinoblastoma. In vitro, VCN-01 effectively killed patient-derived retinoblastoma models. In mice, intravitreous administration of VCN-01 in retinoblastoma xenografts induced tumor necrosis, improved ocular survival compared with standard-of-care chemotherapy, and prevented micrometastatic dissemination into the brain. In juvenile immunocompetent rabbits, VCN-01 did not replicate in retinas, induced minor local side effects, and only leaked slightly and for a short time into the blood. Initial phase 1 data in patients showed the feasibility of the administration of intravitreous VCN-01 and resulted in antitumor activity in retinoblastoma vitreous seeds and evidence of viral replication markers in tumor cells. The treatment caused local vitreous inflammation but no systemic complications. Thus, oncolytic adenoviruses targeting RB1 might provide a tumor-selective and chemotherapy-independent treatment option for retinoblastoma.


Asunto(s)
Adenoviridae/fisiología , Terapia Molecular Dirigida , Virus Oncolíticos/fisiología , Proteína de Retinoblastoma/metabolismo , Retinoblastoma/metabolismo , Transducción de Señal , Animales , Línea Celular Tumoral , Citotoxicidad Inmunológica , Humanos , Ratones , Metástasis de la Neoplasia , Conejos , Retinoblastoma/inmunología , Retinoblastoma/patología , Análisis de Supervivencia , Distribución Tisular , Investigación Biomédica Traslacional , Resultado del Tratamiento , Replicación Viral , Ensayos Antitumor por Modelo de Xenoinjerto
13.
Hum Gene Ther ; 17(9): 929-40, 2006 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-16972761

RESUMEN

Deletion of viral functions that can be complemented by the specific phenotype of tumor cells is a common strategy to design oncolytic viruses. For example, enhanced mRNA cytoplasmic export in tumor cells phenocopies the adenovirus E1B-55K function and renders mutants of this protein tumor selective. Also, an activated RB pathway complements specific E1A functions that can be deleted to produce oncolytic viruses. In this paper we demonstrate that an adenoviral mutant deleted in virus-associated I (VAI) and VAII RNAs (Ad-VAdel) has oncotropism characterized by 100-fold replication deficiency compared with wild-type adenovirus in normal cells and an unaffected ability to replicate and kill different types of tumor cells. This mutant also displays active antitumoral activity in vivo. In contrast, this oncotropism is less evident in a mutant expressing an inactive form of VAI (Adsub719) because VAII RNA expression is upregulated. The mRNA translation promoted by VA RNA genes can be phenocopied in tumor cells with the activation of signal transduction pathways, such as the Ras pathway.


Asunto(s)
Adenoviridae/genética , Genes Virales , Neoplasias/patología , Adenoviridae/fisiología , Secuencia de Bases , Línea Celular , Cartilla de ADN , Humanos , Mutación , Neoplasias/virología , Replicación Viral
14.
Cancer Res ; 63(17): 5544-50, 2003 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-14500393

RESUMEN

Adenovirus synthesize proteins that interact with oncogene and tumor suppressor gene products to set the cell for virus replication. Mutant viruses defective in these functions replicate selectively in cancer cells and represent new tools to treat cancer. We report a selectivity strategy based on deletions of adenovirus Virus-Associated (VA) RNAs. In normal cells, these RNAs are necessary for virus replication because they inactivate the RNA-dependent protein kinase protein kinase R, a kinase that otherwise would block protein translation in response to infection. However, downstream effectors of Ras can also inactivate protein kinase R, and therefore, the need for VA RNA genes should be bypassed in cells with an active Ras pathway. We demonstrate here that a VAI RNA mutant presents a Ras-dependent replication and can be used for oncolytic virotherapy of pancreatic tumors.


Asunto(s)
Adenovirus Humanos/fisiología , ARN Viral/genética , Proteínas ras/fisiología , Adenovirus Humanos/genética , Efecto Citopatogénico Viral/genética , Activación Enzimática , Humanos , Neoplasias Pancreáticas/terapia , Neoplasias Pancreáticas/virología , Células Tumorales Cultivadas , Replicación Viral/genética , eIF-2 Quinasa/metabolismo
16.
PLoS One ; 11(1): e0147211, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26808201

RESUMEN

Despite the recent advances in the development of antitumor therapies, the prognosis for patients with malignant gliomas remains dismal. Therapy with tumor-selective viruses is emerging as a treatment option for this devastating disease. In this study we characterize the anti-glioma effect of VCN-01, an improved hyaluronidase-armed pRB-pathway-selective oncolytic adenovirus that has proven safe and effective in the treatment of several solid tumors. VCN-01 displayed a significant cytotoxic effect on glioma cells in vitro. In vivo, in two different orthotopic glioma models, a single intra-tumoral administration of VCN-01 increased overall survival significantly and led to long-term survivors free of disease.


Asunto(s)
Adenovirus Humanos/fisiología , Neoplasias Encefálicas/terapia , Glioma/terapia , Viroterapia Oncolítica , Virus Oncolíticos/fisiología , Proteínas E1A de Adenovirus/genética , Adenovirus Humanos/genética , Animales , Neoplasias Encefálicas/patología , Línea Celular Tumoral , Efecto Citopatogénico Viral , Factor de Transcripción E2F1/genética , Genes Sintéticos , Genes Virales , Glioma/patología , Humanos , Hialuronoglucosaminidasa/genética , Inyecciones Intralesiones , Ratones , Células Madre Neoplásicas , Oligopéptidos/genética , Virus Oncolíticos/genética , Organismos Modificados Genéticamente , Replicación Viral , Ensayos Antitumor por Modelo de Xenoinjerto
17.
Clin Cancer Res ; 22(9): 2217-25, 2016 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-26603261

RESUMEN

PURPOSE: Osteosarcoma is the most common malignant bone tumor in children and adolescents. Despite aggressive chemotherapy, more than 30% of patients do not respond and develop bone or lung metastasis. Oncolytic adenoviruses engineered to specifically destroy cancer cells are a feasible option for osteosarcoma treatment. VCN-01 is a replication-competent adenovirus specifically engineered to replicate in tumors with a defective RB pathway, presents an enhanced infectivity through a modified fiber and an improved distribution through the expression of a soluble hyaluronidase. The aim of this study is to elucidate whether the use of VCN-01 would be an effective therapeutic strategy for pediatric osteosarcoma. EXPERIMENTAL DESIGN: We used osteosarcoma cell lines established from patients with metastatic disease (531MII, 678R, 588M, and 595M) and a commercial cell line (143B). MTT assays were carried out to evaluate the cytotoxicity of VCN-01. Hexon assays were used to evaluate the replication of the virus. Western blot analysis was performed to assess the expression levels of viral proteins and autophagic markers. The antitumor effect of VCN-01 was evaluated in orthotopic and metastatic osteosarcoma murine animal models. RESULTS: This study found that VCN-01, a new generation genetically modified oncolytic adenovirus, administered locally or systemically, had a potent antisarcoma effect in vitro and in vivo in mouse models of intratibial and lung metastatic osteosarcoma. Moreover, VCN-01 administration showed a safe toxicity profile. CONCLUSIONS: These results uncover VCN-01 as a promising strategy for osteosarcoma, setting the bases to propel a phase I/II trial for kids with this disease. Clin Cancer Res; 22(9); 2217-25. ©2015 AACR.


Asunto(s)
Adenoviridae/genética , Neoplasias Óseas/virología , Virus Oncolíticos/genética , Osteosarcoma/virología , Animales , Neoplasias Óseas/patología , Línea Celular , Línea Celular Tumoral , Femenino , Terapia Genética/métodos , Células HEK293 , Humanos , Neoplasias Pulmonares/patología , Neoplasias Pulmonares/virología , Ratones , Ratones Desnudos , Viroterapia Oncolítica/métodos , Pediatría , Replicación Viral/genética , Ensayos Antitumor por Modelo de Xenoinjerto/métodos
18.
Clin Cancer Res ; 21(6): 1406-18, 2015 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-25391696

RESUMEN

PURPOSE: Tumor targeting upon intravenous administration and subsequent intratumoral virus dissemination are key features to improve oncolytic adenovirus therapy. VCN-01 is a novel oncolytic adenovirus that combines selective replication conditional to pRB pathway deregulation, replacement of the heparan sulfate glycosaminoglycan putative-binding site KKTK of the fiber shaft with an integrin-binding motif RGDK for tumor targeting, and expression of hyaluronidase to degrade the extracellular matrix. In this study, we evaluate the safety and efficacy profile of this novel oncolytic adenovirus. EXPERIMENTAL DESIGN: VCN-01 replication and potency were assessed in a panel of tumor cell lines. VCN-01 tumor-selective replication was evaluated in human fibroblasts and pancreatic islets. Preclinical toxicity, biodistribution, and efficacy studies were conducted in mice and Syrian hamsters. RESULTS: Toxicity and biodistribution preclinical studies support the selectivity and safety of VCN-01. Antitumor activity after intravenous or intratumoral administration of the virus was observed in all tumor models tested, including melanoma and pancreatic adenocarcinoma, both in immunodeficient mice and immunocompetent hamsters. CONCLUSIONS: Oncolytic adenovirus VCN-01 characterized by the expression of hyaluronidase and the RGD shaft retargeting ligand shows an efficacy-toxicity prolife in mice and hamsters by intravenous and intratumoral administration that warrants clinical testing.


Asunto(s)
Adenocarcinoma/terapia , Melanoma/terapia , Viroterapia Oncolítica/métodos , Neoplasias Pancreáticas/terapia , Adenoviridae/genética , Animales , Sitios de Unión/genética , Proteínas Portadoras/genética , Línea Celular , Línea Celular Tumoral , Cricetinae , Matriz Extracelular/metabolismo , Femenino , Células HEK293 , Proteoglicanos de Heparán Sulfato/genética , Proteoglicanos de Heparán Sulfato/metabolismo , Humanos , Hialuronoglucosaminidasa/biosíntesis , Hialuronoglucosaminidasa/genética , Islotes Pancreáticos/virología , Masculino , Mesocricetus , Ratones , Ratones Endogámicos BALB C , Viroterapia Oncolítica/efectos adversos , Virus Oncolíticos/genética , Unión Proteica , Ensayos Antitumor por Modelo de Xenoinjerto
19.
Methods Mol Biol ; 246: 121-38, 2004.
Artículo en Inglés | MEDLINE | ID: mdl-14970588

RESUMEN

Cell transduction in vitro is only the first step toward proving that a genetherapy vector can be useful to treat tumors. However, tumor targeting in vivo is now the milestone for gene therapy to succeed against disseminated cancer. Therefore, most valuable information is obtained from studies of vector biodistribution. Owing to the hepatotropism of adenoviral vectors, a particularly important parameter is the tumor/liver ratio. This ratio can be given at the level of gene expression if the amount of transgene expression is measured. To optimize the targeting, however, the levels of viral particles that reach the tumor compared to other organs must be studied. Most of this chapter deals with methods to quantify the virus fate in tumor-bearing animals. We present a radioactive labeling method that can be used to study biodistribution. After a small section dealing with tumor models, we describe methods to quantify different parameters related to adenovirus-mediated tumor targeting.


Asunto(s)
Adenoviridae/genética , Técnicas de Transferencia de Gen , Vectores Genéticos , Neoplasias/genética , Animales , Secuencia de Bases , Separación Celular , Técnicas de Cultivo , Cartilla de ADN , Citometría de Flujo , Inmunohistoquímica , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos
20.
Hum Gene Ther Methods ; 24(6): 372-80, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24020980

RESUMEN

The contamination of adenovirus (Ad) stocks with adeno-associated viruses (AAV) is usually unnoticed, and it has been associated with lower Ad yields upon large-scale production. During Ad propagation, AAV contamination needs to be detected routinely by polymerase chain reaction without symptomatic suspicion. In this study, we describe that the coinfection of either Ad wild type 5 or oncolytic Ad with AAV results in a large-plaque phenotype associated with an accelerated release of Ad from coinfected cells. This accelerated release was accompanied with the expected decrease in Ad yields in two out of three cell lines tested. Despite this lower Ad yield, coinfection with AAV accelerated cell death and enhanced the cytotoxicity mediated by Ad propagation. Intratumoral coinjection of Ad and AAV in two xenograft tumor models improved antitumor activity and mouse survival. Therefore, we conclude that accidental or intentional AAV coinfection has important implications for Ad-mediated virotherapy.


Asunto(s)
Dependovirus/genética , Viroterapia Oncolítica/métodos , Virus Oncolíticos/genética , Transfección/métodos , Animales , Línea Celular Tumoral , Contaminación de ADN , Femenino , Células HEK293 , Humanos , Ratones , Ratones Endogámicos BALB C , Neoplasias Experimentales/terapia , Transfección/normas , Ensayos Antitumor por Modelo de Xenoinjerto
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA