Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
1.
PLoS One ; 12(8): e0182715, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28796812

RESUMEN

The purpose of this work was to carry out preclinical toxicity and bio-distribution studies required for regulatory approval of a clinical trial application for Phase I clinical studies of ONCOS-102 (Ad5/3-D24-GM-CSF) for therapy of advanced cancers (NCT01598129). The study design, route of administration and dosage differs from the clinical protocol and in more detail, investigate bio-distribution and toxicological profile of ONCOS-102 treatment in animal model. The study was carried out in 300 hamsters divided into nine test groups-three bio-distribution groups and six groups for analysis of toxicity. Hamsters received ONCOS-102 by intracardial, intraperitoneal or subcutaneous injections. Additionally, one group was administered twice a week with intraperitoneal injections of Cyclophosphamide. The control animals were administered with NaCl solution without ONCOS-102 in the same volume and the same way. No adverse effects of repeated administration of ONCOS-102 including body weight, food consumption, hematology and clinical chemistry parameters, histopathology and bio-accumulation were observed in the course of 6-month administration and following 3- month recovery period. All obtained findings indicate the treatment clinically safe.


Asunto(s)
Adenoviridae/genética , Factor Estimulante de Colonias de Granulocitos y Macrófagos/genética , Neoplasias/terapia , Viroterapia Oncolítica , Virus Oncolíticos/genética , Animales , Cricetinae , Femenino , Masculino , Mesocricetus , Terapia Molecular Dirigida , Distribución Tisular
2.
Mol Ther ; 23(5): 964-973, 2015 05.
Artículo en Inglés | MEDLINE | ID: mdl-25655312

RESUMEN

The quality of the antitumor immune response is decisive when developing new immunotherapies for cancer. Oncolytic adenoviruses cause a potent immunogenic stimulus and arming them with costimulatory molecules reshapes the immune response further. We evaluated peripheral blood T-cell subsets of 50 patients with refractory solid tumors undergoing treatment with oncolytic adenovirus. These data were compared to changes in antiviral and antitumor T cells, treatment efficacy, overall survival, and T-cell subsets in pre- and post-treatment tumor biopsies. Treatment caused a significant (P < 0.0001) shift in T-cell subsets in blood, characterized by a proportional increase of CD8(+) cells, and decrease of CD4(+) cells. Concomitant treatment with cyclophosphamide and temozolomide resulted in less CD4(+) decrease (P = 0.041) than cyclophosphamide only. Interestingly, we saw a correlation between T-cell changes in peripheral blood and the tumor site. This correlation was positive for CD8(+) and inverse for CD4(+) cells. These findings give insight to the interconnections between peripheral blood and tumor-infiltrating lymphocyte (TIL) populations regarding oncolytic virotherapy. In particular, our data suggest that induction of T-cell response is not sufficient for clinical response in the context of immunosuppressive tumors, and that peripheral blood T cells have a complicated and potentially misleading relationship with TILs.


Asunto(s)
Adenoviridae , Terapia Genética , Neoplasias/inmunología , Neoplasias/terapia , Viroterapia Oncolítica , Virus Oncolíticos , Subgrupos de Linfocitos T/inmunología , Adenoviridae/genética , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/metabolismo , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , Niño , Femenino , Humanos , Recuento de Linfocitos , Linfocitos Infiltrantes de Tumor/inmunología , Linfocitos Infiltrantes de Tumor/metabolismo , Masculino , Persona de Mediana Edad , Neoplasias/diagnóstico , Neoplasias/genética , Virus Oncolíticos/genética , Subgrupos de Linfocitos T/metabolismo , Transducción Genética , Transgenes , Adulto Joven
3.
Cancer Res ; 72(7): 1621-31, 2012 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-22323527

RESUMEN

The immunosuppressive environment of advanced tumors is a primary obstacle to the efficacy of immunostimulatory and vaccine approaches. Here, we report an approach to arm an oncolytic virus with CD40 ligand (CD40L) to stimulate beneficial immunologic responses in patients. A double-targeted chimeric adenovirus controlled by the hTERT promoter and expressing CD40L (CGTG-401) was constructed and nine patients with progressing advanced solid tumors refractory to standard therapies were treated intratumorally. No serious adverse events resulting in patient hospitalization occurred. Moderate or no increases in neutralizing antibodies were seen, suggesting effective Th1 immunologic effects. An assessment of the blood levels of virus indicated 17.5% of the samples (n = 40) were positive at a low level early after treatment, but not thereafter. In contrast, high levels of virus, CD40L, and RANTES were documented locally at the tumor. Peripheral blood mononuclear cells were analyzed by IFN-γ ELISPOT analysis and induction of both survivin-specific and adenovirus-specific T cells was seen. Antitumor T-cell responses were even more pronounced when assessed by intracellular cytokine staining after stimulation with tumor type-specific peptide pools. Of the evaluable patients, 83% displayed disease control at 3 months and in both cases in which treatment was continued the effect was sustained for at least 8 months. Injected and noninjected lesions responded identically. Together, these findings support further clinical evaluation of CGTG-401.


Asunto(s)
Adenoviridae/genética , Ligando de CD40/genética , Inmunoterapia , Neoplasias/terapia , Viroterapia Oncolítica , Adenoviridae/inmunología , Adulto , Anticuerpos Antivirales/análisis , Ligando de CD40/análisis , Quimiocina CCL5/análisis , Femenino , Humanos , Interferón gamma/biosíntesis , Masculino , Persona de Mediana Edad , Imagen Multimodal , Neoplasias/inmunología , Viroterapia Oncolítica/efectos adversos , Tomografía de Emisión de Positrones , Telomerasa/genética , Células TH1/inmunología , Tomografía Computarizada por Rayos X , Virión/aislamiento & purificación
4.
Int J Cancer ; 130(8): 1937-47, 2012 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-21630267

RESUMEN

The safety of oncolytic viruses for treatment of cancer has been shown in clinical trials while antitumor efficacy has often remained modest. As expression of the coxsackie-adenovirus receptor may be variable in advanced tumors, we developed Ad5-D24-RGD, a p16/Rb pathway selective oncolytic adenovirus featuring RGD-4C modification of the fiber. This allows viral entry through alpha-v-beta integrins frequently highly expressed in advanced tumors. Advanced tumors are often immunosuppressive which results in lack of tumor eradication despite abnormal epitopes being present. Granulocyte-macrophage colony stimulating factor (GMCSF) is a potent activator of immune system with established antitumor properties. To stimulate antitumor immunity and break tumor associated immunotolerance, we constructed Ad5-RGD-D24-GMCSF, featuring GMCSF controlled by the adenoviral E3 promoter. Preliminary safety of Ad5-D24-RGD and Ad5-RGD-D24-GMCSF for treatment of human cancer was established. Treatments with Ad5-D24-RGD (N = 9) and Ad5-RGD-D24-GMCSF (N = 7) were well tolerated. Typical side effects were grade 1-2 fatigue, fever and injection site pain. 77% (10/13) of evaluable patients showed virus in circulation for at least 2 weeks. In 3 out of 6 evaluable patients, disease previously progressing stabilized after a single treatment with Ad5-RGD-D24-GMCSF. In addition, 2/3 patients had stabilization or reduction in tumor marker levels. All patients treated with Ad5-D24-RGD showed disease progression in radiological analysis, although 3/6 had temporary reduction or stabilization of marker levels. Induction of tumor and adenovirus specific immunity was demonstrated with ELISPOT in Ad5-RGD-D24-GMCSF treated patients. RGD modified oncolytic adenoviruses with or without GMCSF seem safe for further clinical development.


Asunto(s)
Factor Estimulante de Colonias de Granulocitos y Macrófagos/metabolismo , Neoplasias/terapia , Oligopéptidos/metabolismo , Viroterapia Oncolítica/métodos , Adenoviridae/genética , Adulto , Anciano , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Línea Celular Tumoral , ADN Viral/genética , Resistencia a Antineoplásicos , Fatiga/etiología , Femenino , Fiebre/etiología , Vectores Genéticos/administración & dosificación , Vectores Genéticos/genética , Factor Estimulante de Colonias de Granulocitos y Macrófagos/genética , Humanos , Integrinas/metabolismo , Masculino , Persona de Mediana Edad , Neoplasias/metabolismo , Neoplasias/virología , Oligopéptidos/genética , Viroterapia Oncolítica/efectos adversos , Virus Oncolíticos/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Resultado del Tratamiento , Carga Viral , Replicación Viral/genética
5.
Mol Ther ; 19(9): 1737-46, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21673660

RESUMEN

Patients with advanced solid tumors refractory to and progressing after conventional therapies were treated with three different regimens of low-dose cyclophosphamide (CP) in combination with oncolytic adenovirus. CP was given with oral metronomic dosing (50 mg/day, N = 21), intravenously (single 1,000 mg dose, N = 7) or both (N = 7). Virus was injected intratumorally. Controls (N = 8) received virus without CP. Treatments were well tolerated and safe regardless of schedule. Antibody formation and virus replication were not affected by CP. Metronomic CP (oral and oral + intravenous schedules) decreased regulatory T cells (T(regs)) without compromising induction of antitumor or antiviral T-cell responses. Oncolytic adenovirus given together with metronomic CP increased cytotoxic T cells and induced Th1 type immunity on a systemic level in most patients. All CP regimens resulted in higher rates of disease control than virus only (all P < 0.0001) and the best progression-free (PFS) and overall survival (OS) was seen in the oral + intravenous group. One year PFS and OS were 53 and 42% (P = 0.0016 and P < 0.02 versus virus only), respectively, both which are unusually high for chemotherapy refractory patients. We conclude that low-dose CP results in immunological effects appealing for oncolytic virotherapy. While these first-in-human data suggest good safety, intriguing efficacy and extended survival, the results should be confirmed in a randomized trial.


Asunto(s)
Antineoplásicos/administración & dosificación , Ciclofosfamida/administración & dosificación , Neoplasias/tratamiento farmacológico , Viroterapia Oncolítica/métodos , Linfocitos T Reguladores/inmunología , Adenoviridae/genética , Adolescente , Adulto , Anciano , Animales , Antineoplásicos/inmunología , Antineoplásicos/uso terapéutico , Línea Celular Tumoral , Niño , Terapia Combinada , Cricetinae , Ciclofosfamida/inmunología , Ciclofosfamida/uso terapéutico , Relación Dosis-Respuesta a Droga , Esquema de Medicación , Femenino , Vectores Genéticos , Humanos , Masculino , Mesocricetus , Persona de Mediana Edad , Neoplasias/inmunología , Resultado del Tratamiento , Adulto Joven
6.
Ann Med ; 43(2): 151-63, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21261555

RESUMEN

BACKGROUND: Successful tumor targeting of systemically administered oncolytic adenoviruses may be hindered by interactions with blood components. MATERIALS AND METHODS: Blood distribution of oncolytic adenoviruses featuring type 5 adenovirus fiber, 5/3 capsid chimerism, or RGD-4C in the fiber knob was investigated in vitro and in patients with refractory solid tumors. RESULTS: Virus titers and prevalence in serum of patients increased over the first post-treatment week, suggesting replication. Detection of low virus loads was more sensitive in blood clots than in serum, although viral levels > 500 viral particles/mL did not differ significantly between both sample types. While adenovirus bound to erythrocytes, platelets, granulocytes, and peripheral blood mononuclear cells in vitro, the virus was mainly detectable in erythrocytes and granulocytes in cancer patients. Taken together with a temporary post-treatment decrease in thrombocyte counts, platelet activation by adenovirus and subsequent clearance seem likely to occur in humans. Fiber modifications had limited observed effect on virus distribution in blood cell compartments. Neutrophils, monocytes and cytotoxic T lymphocytes were the major leukocyte subpopulations interacting with adenoviruses. CONCLUSION: Serum and blood clots are relevant to estimate oncolytic adenovirus replication. Insight into viral interactions with blood cells may contribute to the development of new strategies for tumor delivery.


Asunto(s)
Adenoviridae/metabolismo , Neoplasias/terapia , Viroterapia Oncolítica/métodos , Coagulación Sanguínea , Plaquetas/metabolismo , Eritrocitos/metabolismo , Granulocitos/metabolismo , Humanos , Monocitos/metabolismo , Neoplasias/patología , Neutrófilos/metabolismo , Virus Oncolíticos/metabolismo , Linfocitos T Citotóxicos/metabolismo , Replicación Viral
7.
Mol Ther ; 18(10): 1874-84, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20664527

RESUMEN

Augmenting antitumor immunity is a promising way to enhance the potency of oncolytic adenoviral therapy. Granulocyte-macrophage colony-stimulating factor (GMCSF) can mediate antitumor effects by recruiting natural killer cells and by induction of tumor-specific CD8(+) cytotoxic T-lymphocytes. Serotype 5 adenoviruses (Ad5) are commonly used in cancer gene therapy. However, expression of the coxsackie-adenovirus receptor is variable in many advanced tumors and preclinical data have demonstrated an advantage for replacing the Ad5 knob with the Ad3 knob. Here, a 5/3 capsid chimeric and p16-Rb pathway selective oncolytic adenovirus coding for GMCSF was engineered and tested preclinically. A total of 21 patients with advanced solid tumors refractory to standard therapies were then treated intratumorally and intravenously with Ad5/3-D24-GMCSF, which was combined with low-dose metronomic cyclophosphamide to reduce regulatory T cells. No severe adverse events occurred. Analysis of pretreatment samples of malignant pleural effusion and ascites confirmed the efficacy of Ad5/3-D24-GMCSF in transduction and cell killing. Evidence of biological activity of the virus was seen in 13/21 patients and 8/12 showed objective clinical benefit as evaluated by radiology with Response Evaluation Criteria In Solid Tumors (RECIST) criteria. Antiadenoviral and antitumoral immune responses were elicited after treatment. Thus, Ad5/3-D24-GMCSF seems safe in treating cancer patients and promising signs of efficacy were seen.


Asunto(s)
Adenoviridae/genética , Terapia Genética/métodos , Factor Estimulante de Colonias de Granulocitos y Macrófagos/metabolismo , Neoplasias/terapia , Viroterapia Oncolítica/métodos , Adolescente , Adulto , Anciano , Animales , Línea Celular , Línea Celular Tumoral , Cricetinae , Ciclofosfamida/uso terapéutico , Femenino , Factor Estimulante de Colonias de Granulocitos y Macrófagos/genética , Humanos , Inmunosupresores/uso terapéutico , Masculino , Mesocricetus , Persona de Mediana Edad , Neoplasias/tratamiento farmacológico , Ensayos Antitumor por Modelo de Xenoinjerto , Adulto Joven
8.
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
9.
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
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...