Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 63
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Viruses ; 15(7)2023 06 29.
Artículo en Inglés | MEDLINE | ID: mdl-37515160

RESUMEN

Patients with stage IV gastric cancer suffer from dismal outcomes, a challenge especially in many Asian populations and for which new therapeutic options are needed. To explore this issue, we used oncolytic reovirus in combination with currently used chemotherapeutic drugs (irinotecan, paclitaxel, and docetaxel) for the treatment of gastric and other gastrointestinal cancer cells in vitro and in a mouse model. Cell viability in vitro was quantified by WST-1 assays in human cancer cell lines treated with reovirus and/or chemotherapeutic agents. The expression of reovirus protein and caspase activity was determined by flow cytometry. For in vivo studies, athymic mice received intratumoral injections of reovirus in combination with irinotecan or paclitaxel, after which tumor size was monitored. In contrast to expectations, we found that reoviral oncolysis was only poorly correlated with Ras pathway activation. Even so, the combination of reovirus with chemotherapeutic agents showed synergistic cytopathic effects in vitro, plus enhanced reovirus replication and apoptosis. In vivo experiments showed that reovirus alone can reduce tumor size and that the combination of reovirus with chemotherapeutic agents enhances this effect. Thus, we find that oncolytic reovirus therapy is effective against gastric cancer. Moreover, the combination of reovirus and chemotherapeutic agents synergistically enhanced cytotoxicity in human gastric cancer cell lines in vitro and in vivo. Our data support the use of reovirus in combination with chemotherapy in further clinical trials, and highlight the need for better biomarkers for reoviral oncolytic responsiveness.


Asunto(s)
Viroterapia Oncolítica , Virus Oncolíticos , Orthoreovirus , Reoviridae , Neoplasias Gástricas , Ratones , Animales , Humanos , Irinotecán , Neoplasias Gástricas/terapia , Línea Celular Tumoral , Reoviridae/fisiología , Paclitaxel
2.
J Virol ; 97(5): e0030923, 2023 05 31.
Artículo en Inglés | MEDLINE | ID: mdl-37070982

RESUMEN

Coxsackievirus A21 (CVA21) is a naturally occurring RNA virus that, in preclinical studies and clinical trials, has demonstrated promising potential in treating a range of malignancies. Other oncolytic viruses, such as adenovirus, vesicular stomatitis virus, herpesvirus, and vaccinia virus, all can be engineered to carry one or more transgenes for various purposes, including immune modulation, virus attenuation, and induction of apoptosis of tumor cells. However, it remained unknown whether CVA21 can express therapeutic or immunomodulatory payloads due to its small size and high mutation rate. Using reverse genetics techniques, we demonstrated that a transgene encoding a truncated green fluorescent protein (GFP) of up to 141 amino acids (aa) can be successfully carried in the 5' end of the coding region. Furthermore, a chimeric virus carrying an eel fluorescent protein, UnaG (139 aa), was also made and shown to be stable, and it maintained efficient tumor cell-killing activity. Similar to other oncolytic viruses, the likelihood of delivering CVA21 by the intravenous route is low due to issues like blood absorption, neutralizing antibodies, and liver clearance. To address this problem, we designed the CVA21 cDNA under the control of a weak RNA polymerase II promoter, and subsequently, a stable cell pool in 293T cells was made by integrating the resulting CVA21 cDNA into the cell genome. We showed that the cells are viable and able to persistently generate rCVA21 de novo. The carrier cell approach described here may pave the way to designing new cell therapy strategies by arming with oncolytic viruses. IMPORTANCE As a naturally occurring virus, coxsackievirus A21 is a promising oncolytic virotherapy modality. In this study, we first used reverse genetics to determine whether A21 can stably carry transgenes and found that it could express up to 141 amino acids of foreign GFP. The chimeric virus carrying another fluorescent eel protein UnaG (139 amino acids) gene also appeared to be stable over at least 7 passages. Our results provided guidance on how to select and engineer therapeutic payloads for future A21 anticancer research. Second, the challenges of delivering oncolytic viruses by the intravenous route hamper the broader use of oncolytic viruses in the clinic. Here, we used A21 to show that cells could be engineered to stably carry and persistently release the virus by harboring the viral cDNA in the genome. The approach we presented here may pave a new way for oncolytic virus administration using cells as carriers.


Asunto(s)
Enterovirus Humano A , Virus Oncolíticos , Aminoácidos/genética , Línea Celular Tumoral , ADN Complementario , Enterovirus Humano A/genética , Viroterapia Oncolítica/métodos , Virus Oncolíticos/genética , Transgenes
3.
Mol Ther ; 30(2): 644-661, 2022 02 02.
Artículo en Inglés | MEDLINE | ID: mdl-34547462

RESUMEN

Preclinical and clinical studies have validated the antitumor effects of several oncolytic viruses (OVs). However, the efficacy of OVs is limited when they are administered as monotherapies. Combination therapy is a promising direction for oncolytic virotherapy in the future. A high dose of vitamin C (VitC) exerts anticancer effects by triggering the accretion of substantial amounts of reactive oxygen species (ROS). OVs can induce immunogenic tumor cell death and elicit an antitumor immune response. ROS play an important role in immunogenic cell death (ICD). This study aimed to explore whether high-dose VitC in combination with oncolytic adenoviruses (oAds) exhibited a synergistic antitumor effect. High-dose VitC synergized with oAds against tumor by enhancing immunogenic tumor cell death. Combination therapy with high-dose VitC and oAds significantly increased the number of T cells in the tumor microenvironment (TME) and promoted the activation of T cells. Furthermore, the antitumor effect of the combination therapy was CD8+ T cell dependent. In addition, combination therapy with high-dose VitC and oAds reprogramed the immunosuppressive TME. Our study provides a new strategy for combination therapy of OVs.


Asunto(s)
Neoplasias , Viroterapia Oncolítica , Virus Oncolíticos , Adenoviridae/genética , Humanos , Muerte Celular Inmunogénica , Neoplasias/terapia , Virus Oncolíticos/fisiología , Microambiente Tumoral
4.
Viruses ; 13(6)2021 06 05.
Artículo en Inglés | MEDLINE | ID: mdl-34198859

RESUMEN

Oncolytic viruses have emerged as a promising strategy for cancer therapy due to their dual ability to selectively infect and lyse tumor cells and to induce systemic anti-tumor immunity. Among various candidate viruses, coxsackievirus group B (CVBs) have attracted increasing attention in recent years. CVBs are a group of small, non-enveloped, single-stranded, positive-sense RNA viruses, belonging to species human Enterovirus B in the genus Enterovirus of the family Picornaviridae. Preclinical studies have demonstrated potent anti-tumor activities for CVBs, particularly type 3, against multiple cancer types, including lung, breast, and colorectal cancer. Various approaches have been proposed or applied to enhance the safety and specificity of CVBs towards tumor cells and to further increase their anti-tumor efficacy. This review summarizes current knowledge and strategies for developing CVBs as oncolytic viruses for cancer virotherapy. The challenges arising from these studies and future prospects are also discussed in this review.


Asunto(s)
Enterovirus Humano B/genética , Ingeniería Genética , Vectores Genéticos/genética , Virus Oncolíticos/genética , Animales , Ensayos Clínicos como Asunto , Evaluación Preclínica de Medicamentos , Enterovirus Humano B/fisiología , Ingeniería Genética/métodos , Terapia Genética/efectos adversos , Terapia Genética/métodos , Humanos , Neoplasias/terapia , Viroterapia Oncolítica/efectos adversos , Viroterapia Oncolítica/métodos , Resultado del Tratamiento , Replicación Viral
5.
Viruses ; 13(6)2021 06 11.
Artículo en Inglés | MEDLINE | ID: mdl-34208264

RESUMEN

Oncolytic virotherapy (OV) is an emerging class of immunotherapeutic drugs. Their mechanism of action is two-fold: direct cell lysis and unmasking of the cancer through immunogenic cell death, which allows the immune system to recognize and eradicate tumours. Breast cancer is the most common cancer in women and is challenging to treat with immunotherapy modalities because it is classically an immunogenically "cold" tumour type. This provides an attractive niche for OV, given viruses have been shown to turn "cold" tumours "hot," thereby opening a plethora of treatment opportunities. There has been a number of pre-clinical attempts to explore the use of OV in breast cancer; however, these have not led to any meaningful clinical trials. This review considers both the potential and the barriers to OV in breast cancer, namely, the limitations of monotherapy and the scope for combination therapy, improving viral delivery and challenges specific to the breast cancer population (e.g., tumour subtype, menopausal status, age).


Asunto(s)
Neoplasias de la Mama/terapia , Terapia Genética , Viroterapia Oncolítica , Animales , Protocolos de Quimioterapia Combinada Antineoplásica/efectos adversos , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Neoplasias de la Mama/etiología , Estudios Clínicos como Asunto , Terapia Combinada , Evaluación Preclínica de Medicamentos , Femenino , Terapia Genética/efectos adversos , Terapia Genética/métodos , Vectores Genéticos/administración & dosificación , Vectores Genéticos/genética , Humanos , Viroterapia Oncolítica/efectos adversos , Viroterapia Oncolítica/métodos , Virus Oncolíticos/genética , Resultado del Tratamiento
6.
Viruses ; 13(4)2021 04 10.
Artículo en Inglés | MEDLINE | ID: mdl-33920168

RESUMEN

Oncolytic reovirus preferentially targets and kills cancer cells via the process of oncolysis, and additionally drives clinically favorable antitumor T cell responses that form protective immunological memory against cancer relapse. This two-prong attack by reovirus on cancers constitutes the foundation of its use as an anticancer oncolytic agent. Unfortunately, the efficacy of these reovirus-driven antitumor effects is influenced by the highly suppressive tumor microenvironment (TME). In particular, the myeloid cell populations (e.g., myeloid-derived suppressive cells and tumor-associated macrophages) of highly immunosuppressive capacities within the TME not only affect oncolysis but also actively impair the functioning of reovirus-driven antitumor T cell immunity. Thus, myeloid cells within the TME play a critical role during the virotherapy, which, if properly understood, can identify novel therapeutic combination strategies potentiating the therapeutic efficacy of reovirus-based cancer therapy.


Asunto(s)
Células Mieloides/fisiología , Neoplasias/terapia , Viroterapia Oncolítica , Virus Oncolíticos/patogenicidad , Reoviridae/patogenicidad , Línea Celular Tumoral , Ensayos Clínicos como Asunto , Humanos , Neoplasias/inmunología , Orthoreovirus/fisiología , Microambiente Tumoral/inmunología
7.
Cancer Treat Res Commun ; 27: 100323, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33530025

RESUMEN

Human telomerase reverse transcriptase (hTERT) is an enzyme that is critically involved in elongating and maintaining telomeres length to control cell life span and replicative potential. Telomerase activity is continuously expressed in human germ-line cells and most cancer cells, whereas it is suppressed in most somatic cells. In normal cells, by reducing telomerase activity and progressively shortening the telomeres, the cells progress to the senescence or apoptosis process. However, in cancer cells, telomere lengths remain constant due to telomerase's reactivation, and cells continue to proliferate and inhibit apoptosis, and ultimately lead to cancer development and human death due to metastasis. Studies demonstrated that several DNA and RNA oncoviruses could interact with telomerase by integrating their genome sequence within the host cell telomeres specifically. Through the activation of the hTERT promoter and lengthening the telomere, these cells contributes to cancer development. Since oncoviruses can activate telomerase and increase hTERT expression, there are several therapeutic strategies based on targeting the telomerase of cancer cells like telomerase-targeted peptide vaccines, hTERT-targeting dendritic cells (DCs), hTERT-targeting gene therapy, and hTERT-targeting CRISPR/Cas9 system that can overcome tumor-mediated toleration mechanisms and specifically apoptosis in cancer cells. This study reviews available data on the molecular structure of telomerase and the role of oncoviruses and telomerase interaction in cancer development and telomerase-dependent therapeutic approaches to conquest the cancer cells.


Asunto(s)
Neoplasias/genética , Proteínas Oncogénicas Virales/metabolismo , Retroviridae/patogenicidad , Telomerasa/metabolismo , Animales , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Apoptosis/genética , Senescencia Celular/genética , Modelos Animales de Enfermedad , Terapia Genética/métodos , Interacciones Microbiota-Huesped/genética , Humanos , Ratones , Neoplasias/terapia , Neoplasias/virología , Proteínas Oncogénicas Virales/genética , Viroterapia Oncolítica/métodos , Virus Oncolíticos/genética , Virus Oncolíticos/inmunología , Regiones Promotoras Genéticas , Retroviridae/genética , Telomerasa/antagonistas & inhibidores , Telómero/metabolismo , Homeostasis del Telómero
8.
Int J Mol Sci ; 21(20)2020 Oct 09.
Artículo en Inglés | MEDLINE | ID: mdl-33050329

RESUMEN

Malignant brain tumors remain incurable diseases. Although much effort has been devoted to improving patient outcome, multiple factors such as the high tumor heterogeneity, the strong tumor-induced immunosuppressive microenvironment, and the low mutational burden make the treatment of these tumors especially challenging. Thus, novel therapeutic strategies are urgent. Oncolytic viruses (OVs) are biotherapeutics that have been selected or engineered to infect and selectively kill cancer cells. Increasingly, preclinical and clinical studies demonstrate the ability of OVs to recruit T cells and induce durable immune responses against both virus and tumor, transforming a "cold" tumor microenvironment into a "hot" environment. Besides promising clinical results as a monotherapy, OVs can be powerfully combined with other cancer therapies, helping to overcome critical barriers through the creation of synergistic effects in the fight against brain cancer. Although many questions remain to be answered to fully exploit the therapeutic potential of OVs, oncolytic virotherapy will clearly be part of future treatments for patients with malignant brain tumors.


Asunto(s)
Neoplasias Encefálicas/genética , Neoplasias Encefálicas/terapia , Terapia Genética , Vectores Genéticos/genética , Viroterapia Oncolítica , Virus Oncolíticos/genética , Animales , Biomarcadores de Tumor , Ensayos Clínicos como Asunto , Terapia Combinada , Evaluación Preclínica de Medicamentos , Técnicas de Transferencia de Gen , Terapia Genética/métodos , Vectores Genéticos/administración & dosificación , Humanos , Viroterapia Oncolítica/métodos , Transducción Genética , Resultado del Tratamiento
9.
Appl Microbiol Biotechnol ; 104(19): 8231-8242, 2020 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-32816087

RESUMEN

Oncolytic virus (OV) immunotherapy is characterized by viruses which specifically target cancer cells and cause their cytolysis. They provide a unique and promising new tool for the eradication of cancer as they interact with and affect the tumor microenvironment (TME), vasculature, and immune system. Advancements of genetic engineering have allowed for these viruses to be armed in such a way to have enhanced targeting, strong immunomodulation properties, and an ability to modify the TME. However, there are still major limitations in their use, mostly due to difficulties in delivering the viral particles to the tumors and in ensuring that the immunomodulatory properties are able to stimulate the host immune response to mount a complete response. Using novel delivery systems and using OVs as a complementary therapy in a combinatorial treatment have shown some significant successes. In this review, we discuss the major issues and difficulties in using OVs as anti-tumor agents and some of the strategies put in place so far to overcome these limitations. KEY POINTS: • Oncolytic viruses (OVs) infect cancer cells and cause their cytolysis. • The major limitations in using OVs as anti-tumor therapy were discussed. • The potential strategies to overcome these limitations were summarized.


Asunto(s)
Neoplasias , Viroterapia Oncolítica , Virus Oncolíticos , Humanos , Inmunomodulación , Inmunoterapia , Neoplasias/terapia , Virus Oncolíticos/genética , Microambiente Tumoral
10.
Biochim Biophys Acta Rev Cancer ; 1874(1): 188385, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32554098

RESUMEN

As a promising area of tumor treatment, immunotherapies, such as immune checkpoint inhibitors, have been applied to various types of cancer. However, many patients do not respond to such therapies. Increasing application of tumor ablation therapy, a minimally invasive treatment, has been observed in the clinic. Although it can boost the anti-tumor immune response of patients in many ways, ablation alone is not sufficient to remove the tumor completely or stop tumor recurrence in the long term. Currently, there is emerging research focusing on ablation in combination with immunotherapy, aiming to confirm the therapeutic value of this treatment for cancer patients. Hence, in this article, we review the classification, guideline recommendations, and immunomodulatory effects of ablation therapy, as well as the pre-clinical and clinical research of this combination therapy.


Asunto(s)
Técnicas de Ablación/métodos , Antineoplásicos Inmunológicos/uso terapéutico , Hipertermia Inducida/métodos , Neoplasias/terapia , Viroterapia Oncolítica/métodos , Técnicas de Ablación/normas , Animales , Antineoplásicos Inmunológicos/farmacología , Antígeno B7-H1/antagonistas & inhibidores , Antígeno B7-H1/inmunología , Antígeno CTLA-4/antagonistas & inhibidores , Antígeno CTLA-4/inmunología , Ensayos Clínicos como Asunto , Terapia Combinada/métodos , Terapia Combinada/normas , Modelos Animales de Enfermedad , Humanos , Hipertermia Inducida/normas , Neoplasias/inmunología , Viroterapia Oncolítica/normas , Virus Oncolíticos/inmunología , Guías de Práctica Clínica como Asunto , Receptor de Muerte Celular Programada 1/antagonistas & inhibidores , Receptor de Muerte Celular Programada 1/inmunología , Resultado del Tratamiento
11.
J Immunol Res ; 2020: 8459496, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32411806

RESUMEN

NK cells are lymphocytes with antitumor properties and can directly lyse tumor cells in a non-MHC-restricted manner. However, the tumor microenvironment affects the immune function of NK cells, which leads to immune evasion. This may be related to the pathogenesis of some diseases. Therefore, great efforts have been made to improve the immunotherapy effect of natural killer cells. NK cells from different sources can meet different clinical needs, in order to minimize the inhibition of NK cells and maximize the response potential of NK cells, for example, modification of NK cells can increase the number of NK cells in tumor target area, change the direction of NK cells, and improve their targeting ability to malignant cells. Checkpoint blocking is also a promising strategy for NK cells to kill tumor cells. Combination therapy is another strategy for improving antitumor ability, especially in combination with oncolytic viruses and nanomaterials. In this paper, the mechanisms affecting the activity of NK cells were reviewed, and the therapeutic potential of different basic NK cell strategies in tumor therapy was focused on. The main strategies for improving the immune function of NK cells were described, and some new strategies were proposed.


Asunto(s)
Inmunoterapia/métodos , Células Asesinas Naturales/trasplante , Neoplasias/terapia , Animales , Antígenos de Neoplasias/inmunología , Antineoplásicos Inmunológicos/administración & dosificación , Terapia Combinada/métodos , Modelos Animales de Enfermedad , Sistemas de Liberación de Medicamentos/métodos , Humanos , Inhibidores de Puntos de Control Inmunológico/administración & dosificación , Evasión Inmune/efectos de los fármacos , Memoria Inmunológica , Células Asesinas Naturales/efectos de los fármacos , Células Asesinas Naturales/inmunología , Células Asesinas Naturales/efectos de la radiación , Magnetoterapia , Ratones , Nanomedicina/métodos , Nanopartículas/administración & dosificación , Neoplasias/inmunología , Virus Oncolíticos/inmunología , Receptores Quiméricos de Antígenos/inmunología , Microambiente Tumoral/efectos de los fármacos , Microambiente Tumoral/inmunología , Microambiente Tumoral/efectos de la radiación
12.
Biochem Pharmacol ; 177: 113986, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32330494

RESUMEN

Virotherpay is emerging as a promising strategy against cancer, and three oncolytic viruses (OVs) have gained approval in different countries for the treatment of several cancer types. Beyond the capability to selectively infect, replicate and lyse cancer cells, OVs act through a multitude of events, including modification of the tumour micro/macro-environment as well as a complex modulation of the anti-tumour immune response by activation of danger signals and immunogenic cell death pathways. Most OVs show limited effects, depending on the viral platform and the interactions with the host. OVs used as monotherapy only in a minority of patients elicited a full response. Better outcomes were obtained using OVs in combination with other treatments, such as immune therapy or chemotherapy, suggesting that the full potential of OVs can be unleashed in combination with other treatment modalities. Here, we report the main described combination of OVs with conventional chemotherapeutic agents: platinum salts, mitotic inhibitors, anthracyclines and other antibiotics, anti-metabolites, alkylating agents and topoisomerase inhibitors. Additionally, our work provides an overview of OV combination with targeted therapies: histone deacetylase inhibitors, kinase inhibitors, monoclonal antibodies, inhibitors of DNA repair, inhibitors of the proteasome complex and statins that demonstrated enhanced OV anti-neoplastic activity. Although further studies are required to assess the best combinations to translate the results in the clinic, it is clear that combined therapies, acting with complementary mechanisms of action might be useful to target cancer lesions resistant to currently available treatments.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Terapia Combinada/métodos , Inmunoterapia/métodos , Neoplasias/terapia , Viroterapia Oncolítica/métodos , Virus Oncolíticos/genética , Alquilantes/uso terapéutico , Antibióticos Antineoplásicos/uso terapéutico , Antimetabolitos Antineoplásicos/uso terapéutico , Antimitóticos/uso terapéutico , Inhibidores de Histona Desacetilasas/uso terapéutico , Humanos , Neoplasias/genética , Neoplasias/inmunología , Neoplasias/patología , Virus Oncolíticos/inmunología , Compuestos de Platino/uso terapéutico , Inhibidores de Proteínas Quinasas/uso terapéutico , Inhibidores de Topoisomerasa/uso terapéutico , Microambiente Tumoral/efectos de los fármacos , Microambiente Tumoral/genética , Microambiente Tumoral/inmunología
13.
Mol Biol Rep ; 47(3): 1691-1702, 2020 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-31970625

RESUMEN

Phytotherapy has been used to treat a different type of diseases including cancer for a long time, and it was a source for different active anti-tumor agents. Oncolytic Newcastle disease virus (AMHA1) are very promising anti-tumor therapy. Nevertheless, NDV-based monotherapeutics have not been very useful to some resistant tumors. Thus, the efficiency of oncolytic NDV must enhance by combining NDV with other novel therapies. The current study aimed to determine the possibility of improving the oncolytic effect induced by NDV through Rheum ribes rhizomes extract administration in vitro and in vivo. Methods, the in vitro study include exposure of the crude extract of Rheum ribes alone or NDV alone or combination of both agents for 72 h. The cancer cells tested were murine mammary adenocarcinoma AMN3, Human Rhabdomyosarcoma RD, and Human Glioblastoma AMGM5, and using rat embryo fibroblast REF as normal control cells. MTT cell viability assay was used and analyzed for possible synergism using the Chou-Talalay analysis method. In vivo experiment included study the combination and the monotherapeutic modalities in the transplanted murine mammary adenocarcinoma AM3 line and tumor sections analyzed by histopathology. Results, Combination therapy of NDV-R. ribes showed enhanced oncolytic activity on cancer cells. With no cytotoxicity on normal cells. In vivo study showed that monotherapeutic modalities had lower growth inhibitory effect on transplanted tumors in mice in compare to combination therapy. Histopathological examination revealed the broader area of necrosis in tumors treated by combination therapy. In conclusion, the novel combination recommended for clinical application for cancer therapy.


Asunto(s)
Adenocarcinoma/terapia , Neoplasias Mamarias Experimentales/terapia , Viroterapia Oncolítica/métodos , Extractos Vegetales/farmacología , Rheum/química , Rizoma/química , Adenocarcinoma/patología , Animales , Antineoplásicos/farmacología , Línea Celular , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Terapia Combinada/métodos , Femenino , Humanos , Neoplasias Mamarias Experimentales/patología , Ratones , Virus de la Enfermedad de Newcastle/fisiología , Virus Oncolíticos/fisiología , Ratas , Resultado del Tratamiento
14.
Methods Mol Biol ; 2058: 285-293, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-31486046

RESUMEN

Oncolytic viral immunotherapy based on the MG1 Maraba platform has undergone extensive preclinical evaluation, resulting in the advancement of two programs into clinical trials. MG1 Maraba encoding tumor antigens (tumor associated antigens or viral antigens) are used to boost antitumor immunity, while MG1 Maraba infects tumors, causes oncolysis and transforms the tumor microenvironment. An overview of MG1 Maraba clinical development is outlined here, along with general considerations relating to the design of clinical trials for complex biologic products such as oncolytic viral immunotherapies. These include choice of patient population, optimized treatment regimen, and endpoints which provide early signals of activity and inform the late-stage development path of these agents with novel mechanisms of action.


Asunto(s)
Vectores Genéticos/genética , Virus Oncolíticos/genética , Rhabdoviridae/genética , Investigación Biomédica Traslacional , Animales , Ensayos Clínicos como Asunto , Evaluación Preclínica de Medicamentos , Ingeniería Genética , Terapia Genética/métodos , Humanos , Inmunoterapia/métodos , Neoplasias/genética , Neoplasias/inmunología , Neoplasias/terapia , Viroterapia Oncolítica/métodos , Virus Oncolíticos/inmunología , Proyectos de Investigación , Rhabdoviridae/inmunología
15.
Oncol Rep ; 41(3): 1509-1520, 2019 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-30569160

RESUMEN

Oncolytic viruses are genetically engineered viruses designed for the treatment of solid tumors, and are often coupled with the antitumor immunity of the host. The challenge of using oncolytic herpes simplex virus (oHSV) as an efficacious oncolytic agent is the potential host tissue damage caused by the production of a range of cytokines following intratumoral oHSV injection. An HSV­suppressor of cytokine signaling 4 (SOCS4) recombinant virus was created to investigate whether it inhibits cytokine storm. Recombinant HSV­SOCS4 and HSV­1(F) were used to infect mice, and levels of several representative cytokines, including monocyte chemoattractant protein­1, interleukin (IL)­1ß, tumor necrosis factor­α, IL­6 and interferon γ, in serum and bronchoalveolar lavage fluid (BALF) of infected mice were determined, and immune cells in BALF and spleen were enumerated. Lung damage, virus titers in the lung, body weight and survival rates of infected mice were also determined and compared between the two groups. The cytokine concentration of HSV­SOCS4­infected mice was significantly decreased compared with that of HSV­1(F)­infected mice in BALF and serum, and a smaller number of cluster of differentiation (CD)11b+ cells of BALF, and CD8+CD62L+ T cells and CD4+CD62L+ T cells of the spleen were also identified in HSV­SOCS4­infected mice. HSV­SOCS4­infected mice exhibited slight lung damage, a decrease in body weight loss and a 100% survival rate. The results of the present study indicated that SOCS4 protein may be a useful regulator to inhibit cytokine overproduction, and that HSV­SOCS4 may provide a possible solution to control cytokine storm and its consequences following induction by oncolytic virus treatment.


Asunto(s)
Citocinas/inmunología , Vectores Genéticos/inmunología , Viroterapia Oncolítica/efectos adversos , Virus Oncolíticos/inmunología , Proteínas Supresoras de la Señalización de Citocinas/inmunología , Animales , Antineoplásicos Inmunológicos/efectos adversos , Antineoplásicos Inmunológicos/inmunología , Productos Biológicos/efectos adversos , Productos Biológicos/inmunología , Líquido del Lavado Bronquioalveolar/citología , Líquido del Lavado Bronquioalveolar/inmunología , Chlorocebus aethiops , Citocinas/metabolismo , Evaluación Preclínica de Medicamentos , Femenino , Vectores Genéticos/genética , Herpesvirus Humano 1/inmunología , Pulmón/citología , Pulmón/inmunología , Ratones , Ratones Endogámicos BALB C , Modelos Animales , Neoplasias/tratamiento farmacológico , Viroterapia Oncolítica/métodos , Virus Oncolíticos/genética , Proteínas Recombinantes/genética , Proteínas Recombinantes/inmunología , Proteínas Supresoras de la Señalización de Citocinas/genética , Linfocitos T/inmunología , Células Vero
16.
Nat Commun ; 9(1): 4342, 2018 10 18.
Artículo en Inglés | MEDLINE | ID: mdl-30337542

RESUMEN

Oncolytic virotherapy is a promising therapeutic strategy that uses replication-competent viruses to selectively destroy malignancies. However, the therapeutic effect of certain oncolytic viruses (OVs) varies among cancer patients. Thus, it is necessary to overcome resistance to OVs through rationally designed combination strategies. Here, through an anticancer drug screening, we show that DNA-dependent protein kinase (DNA-PK) inhibition sensitizes cancer cells to OV M1 and improves therapeutic effects in refractory cancer models in vivo and in patient tumour samples. Infection of M1 virus triggers the transcription of interferons (IFNs) and the activation of the antiviral response, which can be abolished by pretreatment of DNA-PK inhibitor (DNA-PKI), resulting in selectively enhanced replication of OV M1 within malignancies. Furthermore, DNA-PK inhibition promotes the DNA damage response induced by M1 virus, leading to increased tumour cell apoptosis. Together, our study identifies the combination of DNA-PKI and OV M1 as a potential treatment for cancers.


Asunto(s)
Antivirales/farmacología , Daño del ADN , Proteína Quinasa Activada por ADN/antagonistas & inhibidores , Virus Oncolíticos/fisiología , Animales , Antineoplásicos/farmacología , Línea Celular Tumoral , Terapia Combinada , Proteína Quinasa Activada por ADN/metabolismo , Evaluación Preclínica de Medicamentos , Estrés del Retículo Endoplásmico/efectos de los fármacos , Humanos , Ratones Endogámicos BALB C , Ratones Desnudos , Modelos Biológicos , Viroterapia Oncolítica , Inhibidores de Proteínas Quinasas/farmacología , Ratas
17.
Mol Pharm ; 15(11): 4777-4800, 2018 11 05.
Artículo en Inglés | MEDLINE | ID: mdl-30226786

RESUMEN

Toll-like Receptor 4 (TLR4) agonists have had a long journey in the field of cancer immunotherapy. Nevertheless, despite the remarkable number of the TLR4 ligands that have gone through various preclinical and clinical stages, only two (Bacillus Calmette-Guérin (BCG) and monophosphoryl lipid A (MPLA)) have hitherto obtained the FDA approval for clinical application in cancer treatment. This paper provides a comprehensive review of the TLR4 agonists' journey as cancer active immunotherapeutics. Following a brief discussion of the rationale behind the use of TLR ligands in cancer immunotherapy, we will initially focus on the forerunner of the TLR4 agonists, bacterial lipopolysaccharide (LPS). Within this context, the potentials and shortcomings of immunotherapy with this agent will be addressed, the strategies that have been devised to enhance the associated therapeutic outcome will be discussed, and the consequent achievements and shortcomings will be summarized. Subsequently, further and perhaps less well-known, molecular, bacterial, and viral TLR4 agonists with potential for cancer immunotherapy will be introduced, and if present, the outcome of the preclinical and clinical investigations of these agents will be reviewed. Finally, a look will be cast upon the promising souvenirs of the relatively new arena of nanotechnology, where TLR4 activating nanoparticulate systems will be proposed as potential candidates for the future development of this field.


Asunto(s)
Inmunoterapia/métodos , Neoplasias/tratamiento farmacológico , Neoplasias/terapia , Receptor Toll-Like 4/agonistas , Adyuvantes Inmunológicos/uso terapéutico , Animales , Vacuna BCG/uso terapéutico , Ensayos Clínicos como Asunto , Evaluación Preclínica de Medicamentos , Humanos , Inmunidad Innata , Inmunoterapia/efectos adversos , Ligandos , Lípido A/análogos & derivados , Lípido A/uso terapéutico , Nanopartículas/uso terapéutico , Neoplasias/inmunología , Virus Oncolíticos/inmunología , Virus Sincitiales Respiratorios/inmunología , Receptor Toll-Like 4/inmunología , Resultado del Tratamiento
18.
Am J Clin Dermatol ; 19(5): 657-670, 2018 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-29961183

RESUMEN

The field of tumor immunology has faced many complex challenges over the last century, but the approval of immune checkpoint inhibitors (anti-cytotoxic T-lymphocyte-associated protein 4 [CTLA4] and anti-programmed cell death-1 [PD-1]/PD-ligand 1 [PD-L1]) and talimogene laherparepvec (T-VEC) for the treatment of metastatic melanoma have awakened a new wave of interest in cancer immunotherapy. Additionally, combinations of vaccines and oncolytic viral therapies with immune checkpoint inhibitors and other systemic agents seem to be promising synergistic strategies to further boost the immune response against cancer. These combinations are undergoing clinical investigation, and if successful, will hopefully soon become available to patients. Here, we review key basic concepts of tumor-induced immune suppression in malignant melanoma, the historical perspective around vaccine development in melanoma, and advances in oncolytic viral therapies. We also discuss the emerging role for combination approaches with different immunomodulatory agents as well as new developments in personalized immunization approaches.


Asunto(s)
Antineoplásicos Inmunológicos/uso terapéutico , Vacunas contra el Cáncer/uso terapéutico , Inmunoterapia/métodos , Melanoma/terapia , Neoplasias Cutáneas/terapia , Animales , Antineoplásicos Inmunológicos/farmacología , Antígeno B7-H1/antagonistas & inhibidores , Antígeno B7-H1/inmunología , Antígeno CTLA-4/antagonistas & inhibidores , Antígeno CTLA-4/inmunología , Vacunas contra el Cáncer/inmunología , Ensayos Clínicos Fase II como Asunto , Ensayos Clínicos Fase III como Asunto , Terapia Combinada/métodos , Modelos Animales de Enfermedad , Evaluación Preclínica de Medicamentos , Humanos , Melanoma/genética , Melanoma/inmunología , Viroterapia Oncolítica/métodos , Virus Oncolíticos/inmunología , Medicina de Precisión/métodos , Neoplasias Cutáneas/genética , Neoplasias Cutáneas/inmunología , Resultado del Tratamiento
19.
Immunotherapy ; 10(11): 971-986, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-29900755

RESUMEN

The standard of care for early hepatobiliary cancers (HBC) includes surgical resection. Liver transplantations or locoregional therapies are beneficial in early hepatocellular carcinoma (HCC) under certain circumstances. Systemic treatments have some benefit in advanced HBC, though long-term prognosis remains poor. We evaluated the role of oncolytic viruses in the treatment of HBCs through a systematic literature review. The recombinant vaccinia virus JX-594 improved median survival in patients with local/metastatic HCC more strongly at high dose than at low dose (14.1 vs 6.7 months; p = 0.08) in a Phase II study. A Phase III study with JX-594 and sorafenib in advanced HCC is ongoing. No survival benefit in HCC was seen with two other recombinant adenoviruses (Ad-TK and DL1520). Several preclinical trials using oncolytic viruses in HBC showed promising results, warranting clinical studies.


Asunto(s)
Carcinoma Hepatocelular/terapia , Neoplasias Hepáticas/terapia , Viroterapia Oncolítica , Virus Oncolíticos/fisiología , Sorafenib/uso terapéutico , Virus Vaccinia/genética , Animales , Ensayos Clínicos como Asunto , Evaluación Preclínica de Medicamentos , Eliminación Hepatobiliar , Humanos , Resultado del Tratamiento
20.
Front Immunol ; 9: 866, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29755464

RESUMEN

Oncolytic viral therapy is a new promising strategy against cancer. Oncolytic viruses (OVs) can replicate in cancer cells but not in normal cells, leading to lysis of the tumor mass. Beside this primary effect, OVs can also stimulate the immune system. Tumors are an immuno-suppressive environment in which the immune system is silenced in order to avoid the immune response against cancer cells. The delivery of OVs into the tumor wakes up the immune system so that it can facilitate a strong and durable response against the tumor itself. Both innate and adaptive immune responses contribute to this process, producing an immune response against tumor antigens and facilitating immunological memory. However, viruses are recognized by the immune system as pathogens and the consequent anti-viral response could represent a big hurdle for OVs. Finding a balance between anti-tumor and anti-viral immunity is, under this new light, a priority for researchers. In this review, we provide an overview of the various ways in which different components of the immune system can be allied with OVs. We have analyzed the different immune responses in order to highlight the new and promising perspectives leading to increased anti-tumor response and decreased immune reaction to the OVs.


Asunto(s)
Interacciones Microbiota-Huesped/inmunología , Neoplasias/terapia , Viroterapia Oncolítica/métodos , Virus Oncolíticos/inmunología , Inmunidad Adaptativa , Antígenos de Neoplasias/inmunología , Humanos , Inmunidad Innata , Neoplasias/inmunología , Viroterapia Oncolítica/efectos adversos , Resultado del Tratamiento
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA