Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 57
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Int J Mol Sci ; 20(3)2019 Jan 31.
Artículo en Inglés | MEDLINE | ID: mdl-30709038

RESUMEN

Oncolytic adenoviruses can trigger lysis of tumor cells, induce an antitumor immune response, bypass classical chemotherapeutic resistance strategies of tumors, and provide opportunities for combination strategies. A major challenge is the development of scalable production methods for viral seed stocks and sufficient quantities of clinical grade viruses. Because of promising clinical signals in a compassionate use program (Advanced Therapy Access Program) which supported further development, we chose the oncolytic adenovirus ONCOS-401 as a testbed for a new approach to scale up. We found that the best viral production conditions in both T-175 flasks and HYPERFlasks included A549 cells grown to 220,000 cells/cm² (80% confluency), with ONCOS-401 infection at 30 multiplicity of infection (MOI), and an incubation period of 66 h. The Lysis A harvesting method with benzonase provided the highest viral yield from both T-175 and HYPERFlasks (10,887 ± 100 and 14,559 ± 802 infectious viral particles/cell, respectively). T-175 flasks and HYPERFlasks produced up to 2.1 × 108 ± 0.2 and 1.75 × 108 ± 0.08 infectious particles of ONCOS-401 per cm² of surface area, respectively. Our findings suggest a suitable stepwise process that can be applied to optimizing the initial production of other oncolytic viruses.


Asunto(s)
Adenoviridae/crecimiento & desarrollo , Virus Oncolíticos/crecimiento & desarrollo , Cultivo de Virus/instrumentación , Células A549 , Animales , Técnicas de Cultivo Celular por Lotes/instrumentación , Humanos , Carga Viral , Cultivo de Virus/métodos , Replicación Viral
2.
J Med Virol ; 90(10): 1669-1673, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-29797583

RESUMEN

Oncolytic adenoviral immunotherapy activates the innate immune system with subsequent induction of adaptive tumor-specific immune responses to fight cancer. Hence, oncolytic viruses do not only eradicate cancer cells by direct lysis, but also generate antitumor immune response, allowing for long-lasting cancer control and tumor reduction. Their therapeutic effect can be further enhanced by arming the oncolytic adenovirus with costimulatory transgenes and/or coadministration with other antitumor therapies. ONCOS-102 has already been found to be well tolerated and efficacious against some types of treatment-refractory tumors, including mesothelin-positive ovarian cancer (NCT01598129). It induced local and systemic CD8+ T-cell immunity and upregulated programmed death ligand 1. These results strongly advocate the use of ONCOS-102 in combination with other therapeutic strategies in advanced and refractory tumors, especially those expressing the mesothelin antigen. The in vivo work presented herein describes the ability of the oncolytic adenovirus ONCOS-102 to induce mesothelin-specific T-cells after the administration of the virus in bagg albino (BALB/c) mice with mesothelin-positive tumors. We also demonstrate the effectiveness of the interferon-γ the enzyme-linked immunospot (ELISPOT) assay to detect the induction of T-cells recognizing mesothelin, hexon, and E1A antigens in ONCOS-102-treated mesothelioma-bearing BALB/c mice. Thus, the ELISPOT assay could be useful to monitor the progress of therapy with ONCOS-102.


Asunto(s)
Adenoviridae/crecimiento & desarrollo , Anticuerpos Antineoplásicos/sangre , Mesotelioma/terapia , Viroterapia Oncolítica , Virus Oncolíticos/crecimiento & desarrollo , Animales , Modelos Animales de Enfermedad , Ensayo de Immunospot Ligado a Enzimas , Humanos , Mesotelina , Ratones Endogámicos BALB C , Linfocitos T/inmunología , Resultado del Tratamiento
3.
Int J Cancer ; 139(8): 1883-93, 2016 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-27287512

RESUMEN

Malignant mesothelioma (MM) is a rare cancer type caused mainly by asbestos exposure. The median overall survival time of a mesothelioma cancer patient is less than 1-year from diagnosis. Currently there are no curative treatment modalities for malignant mesothelioma, however treatments such as surgery, chemotherapy and radiotherapy can help to improve patient prognosis and increase life expectancy. Pemetrexed-Cisplatin is the only standard of care (SoC) chemotherapy for malignant mesothelioma, but the median PFS/OS (progression-free survival/overall survival) from the initiation of treatment is only up to 12 months. Therefore, new treatment strategies against malignant mesothelioma are in high demand. ONCOS-102 is a dual targeting, chimeric oncolytic adenovirus, coding for human GM-CSF. The safety and immune activating properties of ONCOS-102 have already been assessed in phase 1 study (NCT01598129). In this preclinical study, we evaluated the antineoplastic activity of combination treatment with SoC chemotherapy (Pemetrexed, Cisplatin, Carboplatin) and ONCOS-102 in xenograft BALB/c model of human malignant mesothelioma. We demonstrated that ONCOS-102 is able to induce immunogenic cell death of human mesothelioma cell lines in vitro and showed anti-tumor activity in the treatment of refractory H226 malignant pleural mesothelioma (MPM) xenograft model. While chemotherapy alone showed no anti-tumor activity in the mesothelioma mouse model, ONCOS-102 was able to slow down tumor growth. Interestingly, a synergistic anti-tumor effect was seen when ONCOS-102 was combined with chemotherapy regimens. These findings give a rationale for the clinical testing of ONCOS-102 in combination with first-line chemotherapy in patients suffering from malignant mesothelioma.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Neoplasias Pulmonares/terapia , Mesotelioma/terapia , Viroterapia Oncolítica/métodos , Adenoviridae/genética , Adenoviridae/inmunología , Adenoviridae/fisiología , Animales , Carboplatino/administración & dosificación , Línea Celular Tumoral , Terapia Combinada , Factor Estimulante de Colonias de Granulocitos y Macrófagos/administración & dosificación , Factor Estimulante de Colonias de Granulocitos y Macrófagos/genética , Factor Estimulante de Colonias de Granulocitos y Macrófagos/inmunología , Humanos , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/inmunología , Neoplasias Pulmonares/virología , Mesotelioma/tratamiento farmacológico , Mesotelioma/inmunología , Mesotelioma/virología , Mesotelioma Maligno , Ratones , Ratones Endogámicos BALB C , Pemetrexed/administración & dosificación , Replicación Viral , Ensayos Antitumor por Modelo de Xenoinjerto
4.
Int J Cancer ; 137(7): 1775-83, 2015 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-25821063

RESUMEN

Metastatic melanoma is refractory to irradiation and chemotherapy, but amenable to immunological approaches such as immune-checkpoint-inhibiting antibodies or adoptive cell therapies. Oncolytic virus replication is an immunogenic phenomenon, and viruses can be armed with immunostimulatory molecules. Therefore, oncolytic immuno-virotherapy of malignant melanoma is an appealing approach, which was recently validated by a positive phase 3 trial. We investigated the potency of oncolytic adenovirus Ad5/3-D24-GMCSF on a panel of melanoma cell lines and animal models, and summarized the melanoma-specific human data from the Advanced Therapy Access Program (ATAP). The virus effectively eradicated human melanoma cells in vitro and subcutaneous SK-MEL-28 melanoma xenografts in nude mice when combined with low-dose cyclophosphamide. Furthermore, virally-expressed granulocyte-macrophage colony-stimulating factor (GM-CSF) stimulated the differentiation of human monocytes into macrophages. In contrast to human cells, RPMI 1846 hamster melanoma cells exhibited no response to oncolytic viruses and the chimeric 5/3 fiber failed to increase the efficacy of transduction, suggesting limited utility of the hamster model in the context of viruses with this capsid. In ATAP, treatments appeared safe and well-tolerated. Four out of nine melanoma patients treated were evaluable for possible therapy benefit with modified RECIST criteria: one patient had minor response, two had stable disease, and one had progressive disease. Two patients were alive at 559 and 2,149 days after treatment. Ad5/3-D24-GMCSF showed promising efficacy in preclinical studies and possible antitumor activity in melanoma patients refractory to other forms of therapy. This data supports continuing the clinical development of oncolytic adenoviruses for treatment of malignant melanoma.


Asunto(s)
Factor Estimulante de Colonias de Granulocitos y Macrófagos/genética , Melanoma/terapia , Viroterapia Oncolítica/métodos , Adenoviridae/genética , Animales , Diferenciación Celular/fisiología , Línea Celular Tumoral , Cricetinae , Ciclofosfamida/farmacología , Femenino , Factor Estimulante de Colonias de Granulocitos y Macrófagos/biosíntesis , Humanos , Macrófagos/patología , Macrófagos/virología , Melanoma/tratamiento farmacológico , Melanoma/genética , Melanoma/virología , Ratones , Ratones Desnudos , Monocitos/patología , Monocitos/virología , Distribución Aleatoria , Ensayos Antitumor por Modelo de Xenoinjerto
5.
Int J Cancer ; 135(3): 720-30, 2014 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-24374597

RESUMEN

Sarcomas are a relatively rare cancer, but often incurable at the late metastatic stage. Oncolytic immunotherapy has gained attention over the past years, and a wide range of oncolytic viruses have been delivered via intratumoral injection with positive safety and promising efficacy data. Here, we report preclinical and clinical results from treatment of sarcoma with oncolytic adenovirus Ad5/3-D24-GMCSF (CGTG-102). Ad5/3-D24-GMCSF is a serotype chimeric oncolytic adenovirus coding for human granulocyte-macrophage colony-stimulating factor (GM-CSF). The efficacy of Ad5/3-D24-GMCSF was evaluated on a panel of soft-tissue sarcoma (STS) cell lines and in two animal models. Sarcoma specific human data were also collected from the Advanced Therapy Access Program (ATAP), in preparation for further clinical development. Efficacy was seen in both in vitro and in vivo STS models. Fifteen patients with treatment-refractory STS (13/15) or primary bone sarcoma (2/15) were treated in ATAP, and treatments appeared safe and well-tolerated. A total of 12 radiological RECIST response evaluations were performed, and two cases of minor response, six cases of stable disease and four cases of progressive disease were detected in patients progressing prior to virus treatment. Overall, the median survival time post treatment was 170 days. One patient is still alive at 1,459 days post virus treatment. In summary, Ad5/3-D24-GMCSF appears promising for the treatment of advanced STS; a clinical trial for treatment of refractory injectable solid tumors including STS is ongoing.


Asunto(s)
Adenoviridae/genética , Terapia Genética , Factor Estimulante de Colonias de Granulocitos y Macrófagos/genética , Viroterapia Oncolítica , Sarcoma/terapia , Animales , Femenino , Factor Estimulante de Colonias de Granulocitos y Macrófagos/metabolismo , Humanos , Inyecciones Intralesiones , Mesocricetus , Ratones , Ratones Desnudos , Pronóstico , Sarcoma/sangre , Sarcoma/mortalidad , Tasa de Supervivencia , Células Tumorales Cultivadas , Replicación Viral , Ensayos Antitumor por Modelo de Xenoinjerto
6.
Mol Ther ; 21(6): 1212-23, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23546299

RESUMEN

Oncolytic adenoviruses and certain chemotherapeutics can induce autophagy and immunogenic cancer cell death. We hypothesized that the combination of oncolytic adenovirus with low-dose temozolomide (TMZ) is safe, effective, and capable of inducing antitumor immune responses. Metronomic low-dose cyclophosphamide (CP) was added to selectively reduce regulatory T-cells. Preclinically, combination therapy inhibited tumor growth, increased autophagy, and triggered immunogenic cell death as indicated by elevated calreticulin, adenosine triphosphate (ATP) release, and nuclear protein high-mobility group box-1 (HMGB1) secretion. A total of 41 combination treatments given to 17 chemotherapy-refractory cancer patients were well tolerated. We observed anti- and proinflammatory cytokine release, evidence of virus replication, and induction of neutralizing antibodies. Tumor cells showed increased autophagy post-treatment. Release of HMGB1 into serum--a possible indicator of immune response--increased in 60% of treatments, and seemed to correlate with tumor-specific T-cell responses, observed in 10/15 cases overall (P = 0.0833). Evidence of antitumor efficacy was seen in 67% of evaluable treatments with a trend for increased survival over matched controls treated with virus only. In summary, the combination of oncolytic adenovirus with low-dose TMZ and metronomic CP increased tumor cell autophagy, elicited antitumor immune responses, and showed promising safety and efficacy.


Asunto(s)
Adenoviridae/genética , Antineoplásicos/farmacología , Autofagia/efectos de los fármacos , Dacarbazina/análogos & derivados , Neoplasias/terapia , Viroterapia Oncolítica/métodos , Adenosina Trifosfato/metabolismo , Adenoviridae/fisiología , Adolescente , Adulto , Anciano , Animales , Anticuerpos Neutralizantes/sangre , Calreticulina/metabolismo , Muerte Celular/efectos de los fármacos , Línea Celular Tumoral , Niño , Terapia Combinada/métodos , Ciclofosfamida/farmacología , Citocinas/sangre , ADN Viral/sangre , Dacarbazina/farmacología , Relación Dosis-Respuesta a Droga , Femenino , Proteína HMGB1/sangre , Proteína HMGB1/metabolismo , Humanos , Inmunohistoquímica , Masculino , Ratones , Ratones Desnudos , Microscopía Electrónica , Persona de Mediana Edad , Virus Oncolíticos/genética , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/metabolismo , Temozolomida , Replicación Viral , Ensayos Antitumor por Modelo de Xenoinjerto , Adulto Joven
7.
J Immunother Cancer ; 12(3)2024 Mar 08.
Artículo en Inglés | MEDLINE | ID: mdl-38458776

RESUMEN

BACKGROUND: Cancer immunotherapy relies on using the immune system to recognize and eradicate cancer cells. Adaptive immunity, which consists of mainly antigen-specific cytotoxic T cells, plays a pivotal role in controlling cancer progression. However, innate immunity is a necessary component of the cancer immune response to support an immunomodulatory state, enabling T-cell immunosurveillance. METHODS: Here, we elucidated and exploited innate immune cells to sustain the generation of antigen-specific T cells on the use of our cancer vaccine platform. We explored a previously developed oncolytic adenovirus (AdCab) encoding for a PD-L1 (Programmed-Death Ligand 1) checkpoint inhibitor, which consists of a PD-1 (Programmed Cell Death Protein 1) ectodomain fused to an IgG/A cross-hybrid Fc. We coated AdCab with major histocompatibility complex (MHC-I)-restricted tumor peptides, generating a vaccine platform (named PeptiCab); the latter takes advantage of viral immunogenicity, peptide cancer specificity to prime T-cell responses, and antibody-mediated effector functions. RESULTS: As proof of concept, PeptiCab was used in murine models of melanoma and colon cancer, resulting in tumor growth control and generation of systemic T-cell-mediated antitumor responses. In specific, PeptiCab was able to generate antitumor T effector memory cells able to secrete various inflammatory cytokines. Moreover, PeptiCab was able to polarize neutrophils to attain an antigen-presenting phenotype by upregulating MHC-II, CD80 and CD86 resulting in an enhanced T-cell expansion. CONCLUSION: Our data suggest that exploiting innate immunity activates T-cell antitumor responses, enhancing the efficiency of a vaccine platform based on oncolytic adenovirus coated with MHC-I-restricted tumor peptides.


Asunto(s)
Neoplasias , Receptores de IgG , Humanos , Animales , Ratones , Inmunidad Adaptativa , Linfocitos T Citotóxicos , Citocinas/metabolismo , Neoplasias/terapia , Neoplasias/patología
8.
Int J Biol Macromol ; 262(Pt 1): 129926, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38331062

RESUMEN

The emergence of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) posed a threat to public health and the global economy, necessitating the development of various vaccination strategies. Mutations in the SPIKE protein gene, a crucial component of mRNA and adenovirus-based vaccines, raised concerns about vaccine efficacy, prompting the need for rapid vaccine updates. To address this, we leveraged PeptiCRAd, an oncolytic vaccine based on tumor antigen decorated oncolytic adenoviruses, creating a vaccine platform called PeptiVAX. First, we identified multiple CD8 T-cell epitopes from highly conserved regions across coronaviruses, expanding the range of T-cell responses to non-SPIKE proteins. We designed short segments containing the predicted epitopes presented by common HLA-Is in the global population. Testing the immunogenicity, we characterized T-cell responses to candidate peptides in peripheral blood mononuclear cells (PBMCs) from pre-pandemic healthy donors and ICU patients. As a proof of concept in mice, we selected a peptide with epitopes predicted to bind to murine MHC-I haplotypes. Our technology successfully elicited peptide-specific T-cell responses, unaffected by the use of unarmed adenoviral vectors or adeno-based vaccines encoding SPIKE. In conclusion, PeptiVAX represents a fast and adaptable SARS-CoV-2 vaccine delivery system that broadens T-cell responses beyond the SPIKE protein, offering potential benefits for vaccine effectiveness.


Asunto(s)
COVID-19 , Vacunas Virales , Humanos , Ratones , Animales , Vacunas contra la COVID-19 , COVID-19/prevención & control , Glicoproteína de la Espiga del Coronavirus/genética , Leucocitos Mononucleares , SARS-CoV-2 , Péptidos/química , Epítopos de Linfocito T
9.
Mol Ther ; 20(1): 221-9, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22044933

RESUMEN

Calcium channel blockers including verapamil have been proposed to enhance release and antitumor efficacy of oncolytic adenoviruses in preclinical studies but this has not been studied in humans before. Here, we studied if verapamil leads to increased replication of oncolytic adenovirus in cancer patients, as measured by release of virions from tumor cells into the systemic circulation. The study was conducted as a matched case-control study of advanced cancer patients treated with oncolytic adenoviruses with or without verapamil. We observed that verapamil increased mean virus titers present in blood after treatment (P < 0.05). The frequency or severity of adverse events was not increased, nor were cytokine responses or neutralizing antibody levels different between groups. Signs of possible treatment-related clinical benefits were observed in both groups, but there was no significant difference in responses or survival. Thus, our data suggests that the combination of verapamil with oncolytic adenoviruses is safe and well tolerated. Moreover, verapamil treatment seems to result in higher virus titers in blood, indicating enhanced overall replication in tumors. A randomized trial is needed to confirm these findings and to study if enhanced replication results in benefits to patients.


Asunto(s)
Adenoviridae/genética , Bloqueadores de los Canales de Calcio/uso terapéutico , Vectores Genéticos , Neoplasias/terapia , Virus Oncolíticos/genética , Verapamilo/uso terapéutico , Adenoviridae/inmunología , Adolescente , Adulto , Anciano , Anticuerpos Neutralizantes/inmunología , Anticuerpos Antivirales/inmunología , Bloqueadores de los Canales de Calcio/efectos adversos , Terapia Combinada , ADN Viral/sangre , Sinergismo Farmacológico , Femenino , Terapia Genética , Vectores Genéticos/administración & dosificación , Vectores Genéticos/efectos adversos , Humanos , Mediadores de Inflamación/sangre , Masculino , Persona de Mediana Edad , Estadificación de Neoplasias , Neoplasias/mortalidad , Neoplasias/patología , Viroterapia Oncolítica/efectos adversos , Virus Oncolíticos/inmunología , Análisis de Supervivencia , Resultado del Tratamiento , Verapamilo/efectos adversos , Carga Viral/efectos de los fármacos , Adulto Joven
10.
Mol Ther ; 20(9): 1821-30, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22871667

RESUMEN

Twenty-five patients with chemotherapy refractory cancer were treated with a fully serotype 3-based oncolytic adenovirus Ad3-hTERT-E1A. In mice, Ad3 induced higher amounts of cytokines but less liver damage than Ad5 or Ad5/3. In humans, the only grade 3 adverse reactions were self-limiting cytopenias and generally the safety profile resembled Ad5-based oncolytic viruses. Patients that had been previously treated with Ad5 viruses presented longer lasting lymphocytopenia but no median increase in Ad3-specific T-cells in blood, suggesting immunological activity against antigens other than Ad3 hexon. Frequent alterations in antitumor T-cells in blood were seen regardless of previous virus exposure. Neutralizing antibodies against Ad3 increased in all patients, whereas Ad5 neutralizing antibodies remained stable. Treatment with Ad3-hTERT-E1A resulted in re-emergence of Ad5 viruses from previous treatments into blood and vice versa. Signs of possible efficacy were seen in 11/15 (73%) patients evaluable for tumor markers, four of which were treated only intravenously. Particularly promising results were seen in breast cancer patients and especially those receiving concomitant trastuzumab. Taken together, Ad3-hTERT-E1A seems safe for further clinical testing or development of armed versions. It offers an immunologically attractive alternative, with possible pharmacodynamic differences and a different receptor compared to Ad5.


Asunto(s)
Adenoviridae/inmunología , Terapia Genética , Neoplasias/terapia , Viroterapia Oncolítica , Virus Oncolíticos/inmunología , Adenoviridae/genética , Adulto , Anciano , Animales , Anticuerpos Monoclonales Humanizados/administración & dosificación , Anticuerpos Monoclonales Humanizados/uso terapéutico , Anticuerpos Antivirales/biosíntesis , Anticuerpos Antivirales/inmunología , Antineoplásicos/administración & dosificación , Antineoplásicos/uso terapéutico , Biomarcadores de Tumor/genética , Biomarcadores de Tumor/inmunología , Neoplasias de la Mama/genética , Neoplasias de la Mama/inmunología , Neoplasias de la Mama/mortalidad , Neoplasias de la Mama/terapia , Resistencia a Antineoplásicos , Femenino , Genes Virales , Vectores Genéticos , Humanos , Masculino , Ratones , Persona de Mediana Edad , Neoplasias/genética , Neoplasias/inmunología , Neoplasias/mortalidad , Virus Oncolíticos/genética , Tasa de Supervivencia , Linfocitos T/inmunología , Trastuzumab , Replicación Viral
11.
Int J Cancer ; 131(1): 253-63, 2012 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-21834073

RESUMEN

Conventional cancer treatments often have little impact on the course of advanced pancreatic cancer. Although cancer gene therapy with adenoviruses is a promising developmental approach, the primary receptor is poorly expressed in pancreatic cancers which might compromise efficacy and thus targeting to other receptors could be beneficial. Extended stealth delivery, combination with standard chemotherapy or circumvention of host antiadenoviral immune response might improve efficacy further. In this work, capsid-modified adenoviruses were studied for transduction of cell lines and clinical normal and tumor tissue samples. The respective oncolytic viruses were tested for oncolytic activity in vitro and in vivo. Survival was studied in a peritoneally disseminated pancreas cancer model, with or without concurrent gemcitabine while silica implants were utilized for extended intraperitoneal virus delivery. Immunocompetent mice and Syrian hamsters were used to study the effect of silica mediated delivery on antiviral immune responses and subsequent in vivo gene delivery. Capsid modifications selectively enhanced gene transfer to malignant pancreatic cancer cell lines and clinical samples. The respective oncolytic viruses resulted in increased cell killing in vitro, which translated into a survival benefit in mice. Early proinfammatory cytokine responses and formation of antiviral neutralizing antibodies was partially avoided with silica implants. The implant also shielded the virus from pre-existing neutralizing antibodies, while increasing the pancreas/liver gene delivery ratio six-fold. In conclusion, capsid modified adenoviruses would be useful for testing in pancreatic cancer trials. Silica implants might increase the safety and efficacy of the approach.


Asunto(s)
Adenovirus Humanos , Desoxicitidina/análogos & derivados , Viroterapia Oncolítica/métodos , Virus Oncolíticos , Neoplasias Pancreáticas/terapia , Adenovirus Humanos/genética , Adenovirus Humanos/inmunología , Animales , Anticuerpos Neutralizantes , Cápside/química , Proteínas de la Cápside/genética , Proteínas de la Cápside/metabolismo , Línea Celular Tumoral , Terapia Combinada , Cricetinae , Desoxicitidina/farmacología , Desoxicitidina/uso terapéutico , Femenino , Humanos , Ratones , Virus Oncolíticos/genética , Virus Oncolíticos/inmunología , Neoplasias Pancreáticas/tratamiento farmacológico , Neoplasias Pancreáticas/inmunología , Gel de Sílice , Transfección , Gemcitabina
12.
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
13.
Mol Ther ; 19(10): 1858-66, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21792178

RESUMEN

Oncolytic adenoviruses are an emerging experimental approach for treatment of tumors refractory to available modalities. Although preclinical results have been promising, and clinical safety has been excellent, it is also apparent that tumors can become virus resistant. The resistance mechanisms acquired by advanced tumors against conventional therapies are increasingly well understood, which has allowed development of countermeasures. To study this in the context of oncolytic adenovirus, we developed two in vivo models of acquired resistance, where initially sensitive tumors eventually gain resistance and relapse. These models were used to investigate the phenomenon on RNA and protein levels using two types of analysis of microarray data, quantitative reverse transcriptase-polymerase chain reaction and immunohistochemistry. Interferon (IFN) signaling pathways were found upregulated and Myxovirus resistance protein A (MxA) expression was identified as a marker correlating with resistance, while transplantation experiments suggested a role for tumor stroma in maintaining resistance. Furthermore, pathway analysis suggested potential therapeutic targets in oncolytic adenovirus-resistant cells. Improved understanding of the antiviral phenotype causing tumor recurrence is of key importance in order to improve treatment of advanced tumors with oncolytic adenoviruses. Given the similarities between mechanisms of action, this finding might be relevant for other oncolytic viruses as well.


Asunto(s)
Adenoviridae/fisiología , Interferones/biosíntesis , Viroterapia Oncolítica , Animales , Secuencia de Bases , Línea Celular Tumoral , Cartilla de ADN , Femenino , Humanos , Inmunohistoquímica , Ratones , Ratones SCID , Análisis de Secuencia por Matrices de Oligonucleótidos , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Trasplante Heterólogo
14.
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
15.
Front Immunol ; 13: 826164, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35493448

RESUMEN

Oncolytic Viruses (OVs) work through two main mechanisms of action: the direct lysis of the virus-infected cancer cells and the release of tumor antigens as a result of the viral burst. In this sc.enario, the OVs act as in situ cancer vaccines, since the immunogenicity of the virus is combined with tumor antigens, that direct the specificity of the anti-tumor adaptive immune response. However, this mechanism in some cases fails in eliciting a strong specific T cell response. One way to overcome this problem and enhance the priming efficiency is the production of genetically modified oncolytic viruses encoding one or more tumor antigens. To avoid the long and expensive process related to the engineering of the OVs, we have exploited an approach based on coating OVs (adenovirus and vaccinia virus) with tumor antigens. In this work, oncolytic viruses encoding tumor antigens and tumor antigen decorated adenoviral platform (PeptiCRAd) have been used as cancer vaccines and evaluated both for their prophylactic and therapeutic efficacy. We have first tested the oncolytic vaccines by exploiting the OVA model, moving then to TRP2, a more clinically relevant tumor antigen. Finally, both approaches have been investigated in tumor neo-antigens settings. Interestingly, both genetically modified oncolytic adenovirus and PeptiCRAd elicited T cells-specific anti-tumor responses. However, in vitro cross-representation experiments, showed an advantage of PeptiCRAd as regards the fast presentation of the model epitope SIINFEKL from OVA in an immunogenic rather than tolerogenic fashion. Here two approaches used as cancer oncolytic vaccines have been explored and characterized for their efficacy. Although the generation of specific anti-tumor T cells was elicited in both approaches, PeptiCRAd retains the advantage of being rapidly adaptable by coating the adenovirus with a different set of tumor antigens, which is crucial in personalized cancer vaccines clinical setting.


Asunto(s)
Vacunas contra el Cáncer , Neoplasias , Viroterapia Oncolítica , Virus Oncolíticos , Adenoviridae , Antígenos de Neoplasias , Humanos , Virus Oncolíticos/genética , Péptidos , Medicina de Precisión , Vacunas de Subunidad
16.
Elife ; 112022 03 22.
Artículo en Inglés | MEDLINE | ID: mdl-35314027

RESUMEN

Besides the isolation and identification of major histocompatibility complex I-restricted peptides from the surface of cancer cells, one of the challenges is eliciting an effective antitumor CD8+ T-cell-mediated response as part of therapeutic cancer vaccine. Therefore, the establishment of a solid pipeline for the downstream selection of clinically relevant peptides and the subsequent creation of therapeutic cancer vaccines are of utmost importance. Indeed, the use of peptides for eliciting specific antitumor adaptive immunity is hindered by two main limitations: the efficient selection of the most optimal candidate peptides and the use of a highly immunogenic platform to combine with the peptides to induce effective tumor-specific adaptive immune responses. Here, we describe for the first time a streamlined pipeline for the generation of personalized cancer vaccines starting from the isolation and selection of the most immunogenic peptide candidates expressed on the tumor cells and ending in the generation of efficient therapeutic oncolytic cancer vaccines. This immunopeptidomics-based pipeline was carefully validated in a murine colon tumor model CT26. Specifically, we used state-of-the-art immunoprecipitation and mass spectrometric methodologies to isolate >8000 peptide targets from the CT26 tumor cell line. The selection of the target candidates was then based on two separate approaches: RNAseq analysis and HEX software. The latter is a tool previously developed by Jacopo, 2020, able to identify tumor antigens similar to pathogen antigens in order to exploit molecular mimicry and tumor pathogen cross-reactive T cells in cancer vaccine development. The generated list of candidates (26 in total) was further tested in a functional characterization assay using interferon-γ enzyme-linked immunospot (ELISpot), reducing the number of candidates to six. These peptides were then tested in our previously described oncolytic cancer vaccine platform PeptiCRAd, a vaccine platform that combines an immunogenic oncolytic adenovirus (OAd) coated with tumor antigen peptides. In our work, PeptiCRAd was successfully used for the treatment of mice bearing CT26, controlling the primary malignant lesion and most importantly a secondary, nontreated, cancer lesion. These results confirmed the feasibility of applying the described pipeline for the selection of peptide candidates and generation of therapeutic oncolytic cancer vaccine, filling a gap in the field of cancer immunotherapy, and paving the way to translate our pipeline into human therapeutic approach.


Asunto(s)
Vacunas contra el Cáncer , Neoplasias , Adenoviridae , Animales , Antígenos de Neoplasias , Linfocitos T CD8-positivos , Vacunas contra el Cáncer/uso terapéutico , Línea Celular Tumoral , Inmunoterapia/métodos , Ratones , Neoplasias/tratamiento farmacológico , Péptidos
17.
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
18.
J Virol ; 84(2): 856-66, 2010 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19906926

RESUMEN

Oncolytic vaccinia viruses have shown compelling results in preclinical cancer models and promising preliminary safety and antitumor activity in early clinical trials. However, to facilitate systemic application it would be useful to improve tumor targeting and antitumor efficacy further. Here we report the generation of vvdd-VEGFR-1-Ig, a targeted and armed oncolytic vaccinia virus. Tumor targeting was achieved by deletion of genes for thymidine kinase and vaccinia virus growth factor, which are necessary for replication in normal but not in cancer cells. Given the high vascularization typical of kidney cancers, we armed the virus with the soluble vascular endothelial growth factor (VEGF) receptor 1 protein for an antiangiogenic effect. Systemic application of high doses of vvdd-VEGFR-1-Ig resulted in cytokine induction in an immunocompromised mouse model. Upon histopathological analysis, splenic extramedullary hematopoiesis was seen in all virus-injected mice and was more pronounced in the vvdd-VEGFR-1-Ig group. Analysis of the innate immune response after intravenous virus injection revealed high transient and dose-dependent cytokine elevations. When medium and low doses were used for intratumoral or intravenous injection, vvdd-VEGFR-1-Ig exhibited a stronger antitumor effect than the unarmed control. Furthermore, expression of VEGFR-1-Ig was confirmed, and a concurrent antiangiogenic effect was seen. In an immunocompetent model, systemic vvdd-VEGFR-1-Ig exhibited superior antitumor efficacy compared to the unarmed control virus. In conclusion, the targeted and armed vvdd-VEGFR-1-Ig has promising anticancer activity in renal cell cancer models. Extramedullary hematopoiesis may be a sensitive indicator of vaccinia virus effects in mice.


Asunto(s)
Inhibidores de la Angiogénesis , Neoplasias Renales/terapia , Viroterapia Oncolítica/métodos , Virus Oncolíticos , Virus Vaccinia , Receptor 1 de Factores de Crecimiento Endotelial Vascular , Inhibidores de la Angiogénesis/administración & dosificación , Inhibidores de la Angiogénesis/genética , Inhibidores de la Angiogénesis/metabolismo , Animales , Línea Celular Tumoral , Modelos Animales de Enfermedad , Humanos , Inmunocompetencia , Huésped Inmunocomprometido , Riñón/citología , Riñón/virología , Neoplasias Renales/inmunología , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Virus Oncolíticos/genética , Virus Oncolíticos/metabolismo , Timidina Quinasa/genética , Timidina Quinasa/metabolismo , Resultado del Tratamiento , Virus Vaccinia/genética , Virus Vaccinia/metabolismo , Receptor 1 de Factores de Crecimiento Endotelial Vascular/administración & dosificación , Receptor 1 de Factores de Crecimiento Endotelial Vascular/genética , Receptor 1 de Factores de Crecimiento Endotelial Vascular/metabolismo
19.
Mol Pharm ; 8(1): 12-28, 2011 Feb 07.
Artículo en Inglés | MEDLINE | ID: mdl-21126047

RESUMEN

Although results of cancer treatment have improved steadily, metastatic solid tumors can be cured only rarely and therefore new modalities are needed. Tumors often become apoptosis-resistant and capable of excluding drugs during therapy. Similar mechanisms of resistance apply to many treatment regimens, and cross-resistance between different chemotherapeutics often limits the treatment options. Therefore, loss of efficacy may occur simultaneously for different chemotherapeutics. One experimental strategy with an increasing amount of clinical evidence is oncolytic viruses, which replicate preferentially in tumor cells by taking advantage of cancer-specific cellular changes. Adenoviruses are the most widely clinically used oncolytic agents. Replication of oncolytic virus per se kills tumor cells, but oncolysis can also activate the immune system, which may play a role in tumor control. Viruses can be modified in a variety of ways to improve their selectivity and efficacy. The adenovirus genome can be easily engineered to incorporate different tumor targeting mechanisms and therapeutic transgenes for improved antitumor properties. Here we review the available preclinical and clinical data on use of oncolytic adenoviruses in humans.


Asunto(s)
Adenoviridae/fisiología , Neoplasias/terapia , Virus Oncolíticos/fisiología , Adenoviridae/genética , Terapia Genética , Humanos , Viroterapia Oncolítica , Virus Oncolíticos/genética
20.
Mol Pharm ; 8(1): 93-103, 2011 Feb 07.
Artículo en Inglés | MEDLINE | ID: mdl-20964369

RESUMEN

Oncolytic adenoviruses are an emerging treatment option for advanced and refractory cancer. Such patients are often treated with corticosteroids to ameliorate tumor associated symptoms. Thus, it is important to evaluate whether safety is affected by immunosuppression possibly induced by corticosteroids. Concurrent low-dose cyclophosphamide, appealing for its immunomodulatory effects, could also impact safety. In a retrospective case-control study, we evaluated the effect of systemic corticosteroid use in cancer patients receiving oncolytic virotherapy. Four treatment groups were identified: (1) oncolytic adenovirus with oral glucocorticoids, (2) virus alone, (3) virus with glucocorticoids and cyclophosphamide and (4) virus with cyclophosphamide. Adverse events, neutralizing antibody titers, viral DNA in circulation and tumor responses were evaluated. The most common adverse effects were grade 1-2 fatigue, nausea, fever and abdominal pain. Common asymptomatic findings included self-limiting grade 1-3 hyponatremia and aspartate aminotransferase increase. Safety was good and no significant differences were observed between the groups. All patients had an increase in neutralizing antibody titers post-treatment, and no trends for differences between groups were observed. There were fewer post-treatment virus genomes circulating in patients receiving glucocorticoids when compared to their control groups. Overall, glucocorticoid use in cancer patients receiving oncolytic adenovirus, with or without low-dose cyclophosphamide, seems safe.


Asunto(s)
Adenoviridae/genética , Glucocorticoides/uso terapéutico , Neoplasias/tratamiento farmacológico , Neoplasias/terapia , Viroterapia Oncolítica/métodos , Adulto , Anciano , Femenino , Humanos , Masculino , Persona de Mediana Edad , Viroterapia Oncolítica/efectos adversos , Estudios Retrospectivos , Resultado del Tratamiento
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA