Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 87
Filtrar
Mais filtros

Medicinas Complementares
Tipo de documento
Intervalo de ano de publicação
1.
Viruses ; 15(7)2023 06 29.
Artigo em Inglês | MEDLINE | ID: mdl-37515160

RESUMO

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.


Assuntos
Terapia Viral Oncolítica , Vírus Oncolíticos , Orthoreovirus , Reoviridae , Neoplasias Gástricas , Camundongos , Animais , Humanos , Irinotecano , Neoplasias Gástricas/terapia , Linhagem Celular Tumoral , Reoviridae/fisiologia , Paclitaxel
2.
J Virol ; 97(5): e0030923, 2023 05 31.
Artigo em Inglês | MEDLINE | ID: mdl-37070982

RESUMO

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.


Assuntos
Enterovirus Humano A , Vírus Oncolíticos , Aminoácidos/genética , Linhagem Celular Tumoral , DNA Complementar , Enterovirus Humano A/genética , Terapia Viral Oncolítica/métodos , Vírus Oncolíticos/genética , Transgenes
3.
Cardiovasc Intervent Radiol ; 45(12): 1812-1821, 2022 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-35902397

RESUMO

PURPOSE: To investigate the effect of radiofrequency hyperthermia (RFH)-enhanced oncolytic immuno-virotherapy on in vitro pancreatic adenocarcinoma cell line and in vivo rat pancreatic cancer model. MATERIALS AND METHODS: Rat pancreatic adenocarcinoma cell line and 24 Lewis rats with orthotopic pancreatic adenocarcinomas underwent treatment with either (1) oncolytic virotherapy (talimogene laherparepvec [T-VEC]) plus RFH at 42 °C for 30 min; (2) oncolytic virotherapy-only; (3) RFH-only; or (4) saline (control). MTS assays and flow cytometry were used to analyze tumor cell viability and apoptosis levels 24 h after treatment. In the in vivo studies, bioluminescence optical/x-ray imaging and ultrasound imaging was used to assess tumor viability and size 7 and 14 days after treatment. Histopathologic analysis was performed after hematoxylin and eosin staining, TUNEL, Ki-67, and immunohistochemical staining with CD8 and ANK61. RESULTS: Combination therapy (T-VEC + RFH) induced decreased cell viability and increased cell apoptosis compared to T-VEC alone, RFH alone, or control. Optical/x-ray imaging and ultrasound imaging demonstrated decreased tumor bioluminescent signal and tumor volume relative to baseline after combination therapy compared to T-VEC alone, RFH alone, or control. Histopathology demonstrated decreased tumor volume and cell proliferation, increased CD8+ T cell and NK cell infiltration in tumors treated with the combination therapy compared to other three groups. CONCLUSION: RFH enhances locally delivered oncolytic immuno-virotherapy for pancreatic adenocarcinoma, with decreased cell viability and increased apoptosis observed after combination therapy in vitro, and decreased cell viability and tumor volume and increased immune cell infiltrate observed after combination therapy in vivo.


Assuntos
Adenocarcinoma , Hipertermia Induzida , Melanoma , Terapia Viral Oncolítica , Neoplasias Pancreáticas , Ratos , Animais , Terapia Viral Oncolítica/métodos , Adenocarcinoma/diagnóstico por imagem , Adenocarcinoma/terapia , Neoplasias Pancreáticas/diagnóstico por imagem , Neoplasias Pancreáticas/terapia , Melanoma/patologia , Hipertermia Induzida/métodos , Ratos Endogâmicos Lew , Neoplasias Pancreáticas
4.
Mol Ther ; 30(2): 644-661, 2022 02 02.
Artigo em Inglês | MEDLINE | ID: mdl-34547462

RESUMO

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.


Assuntos
Neoplasias , Terapia Viral Oncolítica , Vírus Oncolíticos , Adenoviridae/genética , Humanos , Morte Celular Imunogênica , Neoplasias/terapia , Vírus Oncolíticos/fisiologia , Microambiente Tumoral
5.
Viruses ; 13(6)2021 06 11.
Artigo em Inglês | MEDLINE | ID: mdl-34208264

RESUMO

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).


Assuntos
Neoplasias da Mama/terapia , Terapia Genética , Terapia Viral Oncolítica , Animais , Protocolos de Quimioterapia Combinada Antineoplásica/efeitos adversos , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Neoplasias da Mama/etiologia , Estudos Clínicos como Assunto , Terapia Combinada , Avaliação Pré-Clínica de Medicamentos , Feminino , Terapia Genética/efeitos adversos , Terapia Genética/métodos , Vetores Genéticos/administração & dosagem , Vetores Genéticos/genética , Humanos , Terapia Viral Oncolítica/efeitos adversos , Terapia Viral Oncolítica/métodos , Vírus Oncolíticos/genética , Resultado do Tratamento
6.
Viruses ; 13(6)2021 06 05.
Artigo em Inglês | MEDLINE | ID: mdl-34198859

RESUMO

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.


Assuntos
Enterovirus Humano B/genética , Engenharia Genética , Vetores Genéticos/genética , Vírus Oncolíticos/genética , Animais , Ensaios Clínicos como Assunto , Avaliação Pré-Clínica de Medicamentos , Enterovirus Humano B/fisiologia , Engenharia Genética/métodos , Terapia Genética/efeitos adversos , Terapia Genética/métodos , Humanos , Neoplasias/terapia , Terapia Viral Oncolítica/efeitos adversos , Terapia Viral Oncolítica/métodos , Resultado do Tratamento , Replicação Viral
7.
Phytomedicine ; 89: 153611, 2021 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-34144429

RESUMO

BACKGROUND: Although the breast cancer mortality has slowed down from 2008 to 2017, breast cancer incidence rate continues to rise and thus, new and/or improved treatments are highly needed. Among them, oncolytic virotherapy which has the ability of facilitating the antitumor adaptive immunity, appears as a promising anticancer therapy. Oncolytic measles virus (MV) is particularly suitable for targeting breast cancer due to the upregulation of MV's receptor nectin-4. Nonetheless, with limited clinical success currently, ways of boosting MV-induced breast cancer oncolysis are therefore necessary. Oncolytic virotherapy alone and combined with chemotherapeutic drugs are two strategic areas with intensive development for the search of anticancer drugs. Considering that baicalein (BAI) and cinnamaldehyde (CIN) have demonstrated antitumor properties against multiple cancers including breast cancer, they could be good partners for MV-based oncolytic virotherapy. PURPOSE: To assess the in vitro effect of BAI and CIN with MV and assess their combination effects. METHODS: We examined the combinatorial cytotoxic effect of oncolytic MV and BAI or CIN on MCF-7 breast cancer cells. Potential anti-MV activities of the phytochemicals were first investigated in vitro to determine the optimal combination model. Synergism of MV and BAI or CIN was then evaluated in vitro by calculating the combination indices. Finally, cell cycle analysis and apoptosis assays were performed to confirm the mechanism of synergism. RESULTS: Overall, the viral sensitization combination modality using oncolytic MV to first infect MCF-7 breast cancer cells followed by drug treatment with BAI or CIN was found to produce significantly enhanced tumor killing. Further mechanistic studies showed that the combinations 'MV-BAI' and 'MV-CIN' display synergistic anti-breast cancer effect, mediated by elevated apoptosis. CONCLUSION: We demonstrated, for the first time, effective combination of oncolytic MV with BAI or CIN that could be further explored and potentially developed into novel therapeutic strategies targeting nectin-4-marked breast cancer cells.


Assuntos
Acroleína/análogos & derivados , Neoplasias da Mama , Flavanonas/farmacologia , Terapia Viral Oncolítica , Acroleína/farmacologia , Neoplasias da Mama/terapia , Moléculas de Adesão Celular , Linhagem Celular Tumoral , Feminino , Humanos , Vírus do Sarampo , Ensaios Antitumorais Modelo de Xenoenxerto
8.
Clin Adv Hematol Oncol ; 19(4): 246-260, 2021 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-33989273

RESUMO

In the vast majority of cases, cutaneous melanoma presents as localized disease and is treated with wide excision and sentinel lymph node biopsy, with shared decision making regarding completion lymph node dissection and adjuvant systemic therapy. The treatment of recurrent and in-transit disease is more complex, with further options for regional and systemic therapies and multiple variables to be factored into decisions. Rates of overall and complete response to regional therapies can be quite high in carefully chosen patients, which limits the need for systemic therapies and their inherent side effects. Ongoing trials aim to assess the efficacy of combination regional and systemic therapies and assist in deciding among these options. This review discusses the treatment of primary melanoma and regional nodal disease and offers an in-depth discussion of options for the treatment of recurrent melanoma and in-transit melanoma.


Assuntos
Melanoma/terapia , Recidiva Local de Neoplasia/terapia , Animais , Corantes/uso terapêutico , Gerenciamento Clínico , Fator Estimulador de Colônias de Granulócitos e Macrófagos/uso terapêutico , Humanos , Interleucina-2/uso terapêutico , Melanoma/diagnóstico , Recidiva Local de Neoplasia/diagnóstico , Estadiamento de Neoplasias , Terapia Viral Oncolítica , Rosa Bengala/uso terapêutico , Biópsia de Linfonodo Sentinela
9.
Viruses ; 13(4)2021 04 10.
Artigo em Inglês | MEDLINE | ID: mdl-33920168

RESUMO

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.


Assuntos
Células Mieloides/fisiologia , Neoplasias/terapia , Terapia Viral Oncolítica , Vírus Oncolíticos/patogenicidade , Reoviridae/patogenicidade , Linhagem Celular Tumoral , Ensaios Clínicos como Assunto , Humanos , Neoplasias/imunologia , Orthoreovirus/fisiologia , Microambiente Tumoral/imunologia
10.
Mol Med Rep ; 23(5)2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-33760188

RESUMO

Programmed cell death protein­1 (PD­1)/programmed death protein ligand­1 (PD­L1) inhibitors for treatment of a various types of cancers have revolutionized cancer immunotherapy. However, PD­1/PD­L1 inhibitors are associated with a low response rate and are only effective on a small number of patients with cancer. Development of an anti­PD­1/PD­L1 sensitizer for improving response rate and effectiveness of immunotherapy is a challenge. The present study reviews the synergistic effects of PD­1/PD­L1 inhibitor with oncolytic virus, tumor vaccine, molecular targeted drugs, immunotherapy, chemotherapy, radiotherapy, intestinal flora and traditional Chinese medicine, to provide information for development of effective combination therapies.


Assuntos
Antígeno B7-H1/genética , Inibidores de Checkpoint Imunológico/uso terapêutico , Neoplasias/terapia , Receptor de Morte Celular Programada 1/genética , Antígeno B7-H1/antagonistas & inibidores , Antígeno B7-H1/imunologia , Vacinas Anticâncer/imunologia , Vacinas Anticâncer/uso terapêutico , Humanos , Inibidores de Checkpoint Imunológico/imunologia , Imunoterapia/tendências , Neoplasias/genética , Neoplasias/imunologia , Terapia Viral Oncolítica/tendências , Receptor de Morte Celular Programada 1/antagonistas & inibidores , Receptor de Morte Celular Programada 1/imunologia
11.
Cancer Treat Res Commun ; 27: 100323, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33530025

RESUMO

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.


Assuntos
Neoplasias/genética , Proteínas Oncogênicas Virais/metabolismo , Retroviridae/patogenicidade , Telomerase/metabolismo , Animais , Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico , Apoptose/genética , Senescência Celular/genética , Modelos Animais de Doenças , Terapia Genética/métodos , Interações entre Hospedeiro e Microrganismos/genética , Humanos , Camundongos , Neoplasias/terapia , Neoplasias/virologia , Proteínas Oncogênicas Virais/genética , Terapia Viral Oncolítica/métodos , Vírus Oncolíticos/genética , Vírus Oncolíticos/imunologia , Regiões Promotoras Genéticas , Retroviridae/genética , Telomerase/antagonistas & inibidores , Telômero/metabolismo , Homeostase do Telômero
12.
Curr Pharm Biotechnol ; 22(9): 1141-1148, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33069195

RESUMO

onventional cancer therapeutic approaches broadly include chemotherapy, radiation therapy and surgery. These established approaches have evolved over several decades of clinical experience. For a complex disease like cancer, satisfactory treatment remains an enigma for the simple fact that the causal factors for cancer are extremely diverse. In order to overcome existing therapeutic limitations, consistent scientific endeavors have evolved several potential therapeutic approaches, majority of which focuses essentially on targeted drug delivery, minimal concomitant ramification, and selective high cytotoxicity. The current review focuses on highlighting some of these potential alternatives that are currently in various stages of in vitro, in vivo, and clinical trials. These include physical, chemical and biological entities that are avidly being explored for therapeutic alternatives. Some of these entities include suicide gene, micro RNA, modulatory peptides, ultrasonic waves, free radicals, nanoparticles, phytochemicals, and gene knockout, and stem cells. Each of these techniques may be exploited exclusively and in combination with conventional therapeutic approaches thereby enhancing the therapeutic efficacy of the treatment. The review intends to briefly discuss the mechanism of action, pros, and cons of potential alternatives to conventional therapeutic approaches.


Assuntos
Neoplasias/terapia , Sistemas de Liberação de Medicamentos , Genes Transgênicos Suicidas/genética , Humanos , MicroRNAs/uso terapêutico , Nanomedicina , Nanopartículas/química , Neoplasias/tratamento farmacológico , Terapia Viral Oncolítica , Fármacos Fotossensibilizantes/uso terapêutico
13.
Int J Mol Sci ; 21(20)2020 Oct 09.
Artigo em Inglês | MEDLINE | ID: mdl-33050329

RESUMO

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.


Assuntos
Neoplasias Encefálicas/genética , Neoplasias Encefálicas/terapia , Terapia Genética , Vetores Genéticos/genética , Terapia Viral Oncolítica , Vírus Oncolíticos/genética , Animais , Biomarcadores Tumorais , Ensaios Clínicos como Assunto , Terapia Combinada , Avaliação Pré-Clínica de Medicamentos , Técnicas de Transferência de Genes , Terapia Genética/métodos , Vetores Genéticos/administração & dosagem , Humanos , Terapia Viral Oncolítica/métodos , Transdução Genética , Resultado do Tratamento
14.
Appl Microbiol Biotechnol ; 104(19): 8231-8242, 2020 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-32816087

RESUMO

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.


Assuntos
Neoplasias , Terapia Viral Oncolítica , Vírus Oncolíticos , Humanos , Imunomodulação , Imunoterapia , Neoplasias/terapia , Vírus Oncolíticos/genética , Microambiente Tumoral
15.
Biochim Biophys Acta Rev Cancer ; 1874(1): 188385, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32554098

RESUMO

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.


Assuntos
Técnicas de Ablação/métodos , Antineoplásicos Imunológicos/uso terapêutico , Hipertermia Induzida/métodos , Neoplasias/terapia , Terapia Viral Oncolítica/métodos , Técnicas de Ablação/normas , Animais , Antineoplásicos Imunológicos/farmacologia , Antígeno B7-H1/antagonistas & inibidores , Antígeno B7-H1/imunologia , Antígeno CTLA-4/antagonistas & inibidores , Antígeno CTLA-4/imunologia , Ensaios Clínicos como Assunto , Terapia Combinada/métodos , Terapia Combinada/normas , Modelos Animais de Doenças , Humanos , Hipertermia Induzida/normas , Neoplasias/imunologia , Terapia Viral Oncolítica/normas , Vírus Oncolíticos/imunologia , Guias de Prática Clínica como Assunto , Receptor de Morte Celular Programada 1/antagonistas & inibidores , Receptor de Morte Celular Programada 1/imunologia , Resultado do Tratamento
16.
Biochem Pharmacol ; 177: 113986, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-32330494

RESUMO

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.


Assuntos
Anticorpos Monoclonais/uso terapêutico , Terapia Combinada/métodos , Imunoterapia/métodos , Neoplasias/terapia , Terapia Viral Oncolítica/métodos , Vírus Oncolíticos/genética , Alquilantes/uso terapêutico , Antibióticos Antineoplásicos/uso terapêutico , Antimetabólitos Antineoplásicos/uso terapêutico , Antimitóticos/uso terapêutico , Inibidores de Histona Desacetilases/uso terapêutico , Humanos , Neoplasias/genética , Neoplasias/imunologia , Neoplasias/patologia , Vírus Oncolíticos/imunologia , Compostos de Platina/uso terapêutico , Inibidores de Proteínas Quinases/uso terapêutico , Inibidores da Topoisomerase/uso terapêutico , Microambiente Tumoral/efeitos dos fármacos , Microambiente Tumoral/genética , Microambiente Tumoral/imunologia
17.
Expert Opin Biol Ther ; 20(6): 635-652, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-32067509

RESUMO

Introduction: Immune checkpoint inhibitors (ICI) have dramatically improved the outcome for cancer patients across multiple tumor types. However the response rates to ICI monotherapy remain relatively low, in part due to some tumors cultivating an inherently 'cold' immune microenvironment. Oncolytic viruses (OV) have the capability to promote a 'hotter' immune microenvironment which can improve the efficacy of ICI.Areas covered: In this article we conducted a literature search through Pubmed/Medline to identify relevant articles in both the pre-clinical and clinical settings for combining OVs with ICIs and discuss the impact of this approach on treatment as well as changes within the tumor microenvironment. We also explore the future directions of this novel combination strategy.Expert opinion: The imminent results of the Phase 3 study combining pembrolizumab with or without T-Vec injection are eagerly awaited. OV/ICI combinations remain one of the most promising avenues to explore in the success of cancer immunotherapy.


Assuntos
Inibidores de Checkpoint Imunológico/uso terapêutico , Neoplasias/terapia , Terapia Viral Oncolítica/métodos , Adenoviridae/fisiologia , Anticorpos Monoclonais Humanizados/uso terapêutico , Terapia Combinada , Enterovirus/fisiologia , Humanos , Neoplasias/tratamento farmacológico , Neoplasias/patologia , Orthoreovirus/fisiologia , Vaccinia virus/fisiologia
18.
Mol Biol Rep ; 47(3): 1691-1702, 2020 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-31970625

RESUMO

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.


Assuntos
Adenocarcinoma/terapia , Neoplasias Mamárias Experimentais/terapia , Terapia Viral Oncolítica/métodos , Extratos Vegetais/farmacologia , Rheum/química , Rizoma/química , Adenocarcinoma/patologia , Animais , Antineoplásicos/farmacologia , Linhagem Celular , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Terapia Combinada/métodos , Feminino , Humanos , Neoplasias Mamárias Experimentais/patologia , Camundongos , Vírus da Doença de Newcastle/fisiologia , Vírus Oncolíticos/fisiologia , Ratos , Resultado do Tratamento
19.
Methods Mol Biol ; 2058: 285-293, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-31486046

RESUMO

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.


Assuntos
Vetores Genéticos/genética , Vírus Oncolíticos/genética , Rhabdoviridae/genética , Pesquisa Translacional Biomédica , Animais , Ensaios Clínicos como Assunto , Avaliação Pré-Clínica de Medicamentos , Engenharia Genética , Terapia Genética/métodos , Humanos , Imunoterapia/métodos , Neoplasias/genética , Neoplasias/imunologia , Neoplasias/terapia , Terapia Viral Oncolítica/métodos , Vírus Oncolíticos/imunologia , Projetos de Pesquisa , Rhabdoviridae/imunologia
20.
Oncol Rep ; 41(3): 1509-1520, 2019 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-30569160

RESUMO

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.


Assuntos
Citocinas/imunologia , Vetores Genéticos/imunologia , Terapia Viral Oncolítica/efeitos adversos , Vírus Oncolíticos/imunologia , Proteínas Supressoras da Sinalização de Citocina/imunologia , Animais , Antineoplásicos Imunológicos/efeitos adversos , Antineoplásicos Imunológicos/imunologia , Produtos Biológicos/efeitos adversos , Produtos Biológicos/imunologia , Líquido da Lavagem Broncoalveolar/citologia , Líquido da Lavagem Broncoalveolar/imunologia , Chlorocebus aethiops , Citocinas/metabolismo , Avaliação Pré-Clínica de Medicamentos , Feminino , Vetores Genéticos/genética , Herpesvirus Humano 1/imunologia , Pulmão/citologia , Pulmão/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Modelos Animais , Neoplasias/tratamento farmacológico , Terapia Viral Oncolítica/métodos , Vírus Oncolíticos/genética , Proteínas Recombinantes/genética , Proteínas Recombinantes/imunologia , Proteínas Supressoras da Sinalização de Citocina/genética , Linfócitos T/imunologia , Células Vero
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA