RESUMO
italic>Salmonella has emerged as a promising tumor-targeting strategy in recent years due to its good tumor targeting ability and certain safety. In order to further optimize its therapeutic effect, scientists have tried to modify Salmonella, including its attenuation and drug loading. This paper summarizes the mechanism and research progress of Salmonella-mediated targeted tumor therapy, and introduces the strategies and related progress of its modification and optimization. At the same time, the advantages, current challenges and future development directions of Salmonella-mediated tumor therapy are summarized.
RESUMO
Liver cancer is one of the most common cancers,and its common surgical treatment methods include tran-scatheter arterial chemoembolization,radiofrequency ablation,and liver transplantation surgery.However,the treatment effect of these surgeries on patients with mid-to late-stage liver cancer is not ideal.In recent years,with the continuous development of tumor gene therapy and tumor immunology,tumor treatment methods have transitioned from traditional models to targeted onco-lytic virus therapy.With the advantages of fast replication,the oncolytic virus can kill tumor cells without damaging other normal cells and realize the targeted treatment of liver cancer through mechanisms such as activating the immune system and improving the tumor microenvironment.In addition,immunotherapy can reduce tumor recurrence and metastasis,thereby exerting therapeutic effects on liver cancer.This article reviews the research progress of oncolytic virus and immunotherapy for liver cancer,aiming to provide a reference for the clinical treatment of liver cancer.
RESUMO
Objective To explore the significance of the combined strategy of oncolytic virus infection, immune effector cell supplement, and immune checkpoint blocking in the treatment of gastric cancer. Methods A tumor-bearing nude mouse model of gastric cancer was treated with recombinant oncolytic vaccinia virus carrying the CXCL9 gene of T lymphocyte chemokine and IL-7gene (VV-IL7-CXCL9) obtained in previous experiments. We established a triple-intervention group of recombinant oncolytic vaccinia virus intratumoral injection+cytotoxic T lymphocyte (CTL)+immune checkpoint blocker (PD-1 monoclonal antibody), a single-intervention group of three measures, a dual-intervention group, and a blank control group. After the intervention, tumor-growth curve method, tumor-inhibition rate method, and bioluminescence method were used to detect the tumor treatment effect. ELISA was used to detect CXCL9 and IL-7 molecule concentration in the serum and tissue homogenate of animals in each group. Results The therapeutic results of animal models showed that the tumor growth in the triple-intervention group of recombinant oncolytic poxvirus+CTL+immune checkpoint blocker (PD-1 monoclonal antibody) was significantly slower than those in the other intervention and the blank control group. Conclusion The combination of recombinant oncolytic poxvirus intratumoral injection+CTL+immune checkpoint blocker (PD-1 monoclonal antibody) exert a strong antitumor effect in intervention experiments on gastric-cancer animal models.
RESUMO
Objective:To investigate whether IL-7-secreting oncolytic herpes simplex virus(HSV)could activate CD8+T cells and inhibit the growth of hepatocellular carcinoma.Methods:The expression of IL-7 was detected by Western blot.The in vitro cleavage of tumor cells by tumor oncolytic virus HSV and HSV-IL-7 were detected by crystal violet staining.The tumor inhibition ability of HSV-IL-7 and HSV were detected in subcutaneous transplanted tumor model.Levels of IL-7,IFN-γ and TNF-α in serum and tumor tissues were determined by ELISA.The infiltration of CD8+T cells in tumor tissues was detected by immunohistochemistry.Flow cytometry was used to detect Granzyme B secretion in CD8+T cells infiltrated by tumor.Results:Tumor cells infected with HSV-IL-7 expressed high level of IL-7.Both HSV and HSV-IL-7 can effectively lyse B16-F10,CT-26 and H22 tumor cell lines in a dose-dependent manner in vitro.HSV-IL-7 could significantly inhibit the growth of H22 hepatoma cells in vivo(P<0.01)and prolong the survival time of tumor-bearing mice(P<0.001).HSV-IL-7 could significantly increase the IL-7 content in tumor sites(P<0.000 1),and effectively increase the number of tumor infiltrating CD8+T cells(P<0.001).HSV-IL-7 significantly enhanced Granzyme B secretion of tumor-infiltrating CD8+T cells and IFN-γ and TNF-α in tumor tissues(P<0.000 1).Conclusion:HSV-IL-7 has well tumor inhibition activity in vivo and in vitro.It also can activate the anti-tumor activity of CD8+T cells in vivo by secreting IL-7,inhibit tumor growth and prolong the survival time of tumor-bearing mice.
RESUMO
Viruses are closely related to the occurrence of human diseases, and the infection of some viruses will lead to the occurrence and development of tumors, such as EB virus, hepatitis B virus, HPV virus, etc. On the contrary, some viruses can also be used for the treatment of tumors, such as oncolytic viruses. Lung cancer is a malignant tumor with the highest incidence rate and mortality rate in the world. In recent years, the relationship between virus and lung cancer has attracted more and more attention. Therefore, this article will review the mechanism and therapeutic impact of viruses on lung cancer.
RESUMO
Targeting multiple immune mechanisms may overcome therapy resistance and further improve cancer immunotherapy for humans. Here, we describe the application of virus-like vesicles (VLV) for delivery of three immunomodulators alone and in combination, as a promising approach for cancer immunotherapy. VLV vectors were designed to deliver single chain interleukin (IL)-12, short-hairpin RNA (shRNA) targeting programmed death ligand 1 (PD-L1), and a dominant-negative form of IL-17 receptor A (dn-IL17RA) as a single payload or as a combination payload. Intralesional delivery of the VLV vector expressing IL-12 alone, as well as the trivalent vector (designated CARG-2020) eradicated large established tumors. However, only CARG-2020 prevented tumor recurrence and provided long-term survival benefit to the tumor-bearing mice, indicating a benefit of the combined immunomodulation. The abscopal effects of CARG-2020 on the non-injected contralateral tumors, as well as protection from the tumor cell re-challenge, suggest immune-mediated mechanism of protection and establishment of immunological memory. Mechanistically, CARG-2020 potently activates Th1 immune mechanisms and inhibits expression of genes related to T cell exhaustion and cancer-promoting inflammation. The ability of CARG-2020 to prevent tumor recurrence and to provide survival benefit makes it a promising candidate for its development for human cancer immunotherapy.
RESUMO
Abstract Introduction: Gastric cancer is the fifth most frequently diagnosed type of cancer and the fourth leading cause of cancer mortality worldwide. Oncolytic viruses are a potential agent for cancer treatment. Objective: To evaluate the penetration capacity, selectivity, and oncolytic efficiency of rotavirus Wt1-5 using an ex vivo infection model in tumor samples obtained from patients diagnosed with gastric adenocarcinoma. Materials and methods: Experimental laboratory study performed on explants of diffuse and intestinal-subtype gastric adenocarcinomas collected at the Hospital Universitario de La Samaritana (Bogotá D.C., Colombia). These explants were infected with rotavirus Wt1-5, and immunocytochemistry tests were used to assess its penetration and diffusion capacity through the tumor microenvironment, as well as its potential as an oncolytic virus. Data are described using means and standard deviations. In addition, a bivariate analysis was performed using the Mann-Whitney U test to determine differences between the data from the evaluated assays and the control used in each assay. A statistical significance level of p<0.05 was considered. Results: At 12 hours post infection (h.p.i), rotavirus Wt1-5 had disseminated in all tumor layers, promoting the infection of tumor cells and resulting in necrosis of tumor tissue after 48 h.p.i. On the other hand, adjacent non-tumor tissues showed no evidence of infection with this rotavirus or tissue lysis (p<0.05). Conclusions: Explant culture is a useful model for studying and predicting ex vivo infectious behavior. Rotavirus Wt1-5 selectively and efficiently infects tumor cells in gastric adenocarcinoma explants, both diffuse and intestinal.
Resumen Introducción. A nivel mundial, el cáncer gástrico es el quinto cáncer más comúnmente diagnosticado y la cuarta causa de mortalidad por cáncer. Los virus oncolíticos son un agente terapéutico potencial para el cáncer. Objetivo. Evaluar la capacidad de penetración, la selectividad y la eficiencia oncolítica del rotavirus Wt1-5 mediante un modelo de infección ex vivo en muestras tumorales obtenidas de pacientes con diagnóstico de adenocarcinoma gástrico. Materiales y métodos. Estudio experimental de laboratorio realizado en explantes de adenocarcinoma gástricos de subtipo difuso e intestinal recolectados en el Hospital Universitario de La Samaritana (Bogotá D.C., Colombia). Estos explantes se infectaron con el rotavirus Wt1-5 y, mediante pruebas inmunohistoquímicas, se evaluó su capacidad de penetración y difusión a través del microambiente tumoral, así como su potencial como virus oncolítico. Los datos se describen usando medias y desviaciones estándar. Además, se realizó un análisis bivariado mediante la prueba de U de Mann-Whitney para determinar las diferencias entre los datos de los ensayos evaluados y el control empleado en cada uno. Se consideró un nivel de significancia estadística de p<0.05. Resultados. A las 12 horas post infección (h.p.i) se observó que el rotavirus Wt1-5 se había diseminado en todas las capas del tumor, lo cual favoreció la infección de las células tumorales y generó necrosis del tejido tumoral a partir de las 48 h.p.i. Por otro lado, los tejidos no tumorales adyacentes no mostraron evidencia de infección con este rotavirus, ni lisis tisular (p<0.05). Conclusiones. El cultivo de explantes es un modelo útil para estudiar y predecir el comportamiento infeccioso ex vivo. El rotavirus Wt1-5 infecta de manera selectiva y eficiente las células tumorales en explantes de adeno-carcinoma gástrico, tanto del subtipo difuso como del subtipo intestinal.
RESUMO
@#In recent years, the chimeric antigen receptor T-cell (CAR-T) therapy has achieved breakthrough progress in the treatment of hematologic malignancies. However, when it comes to solid tumors, numerous challenges persist.These include limited CAR-T cell infiltration, susceptibility to T cell exhaustion, off-target effects, and more.Thus, novel therapeutic strategies are imperative to enhance the efficacy of CAR-T therapy for solid tumors. In comparison to standalone CAR-T approaches, the combination of CAR-T with other tumor treatment modalities has demonstrated remarkable effectiveness in both preclinical and clinical research.This review article summarizes the advancements in combining CAR-T with various solid tumor treatments: antibody drugs, oncolytic viruses, tumor vaccines, and nanomedicines.The objective is to furnish a theoretical foundation and novel perspectives for the development of innovative CAR-T combination strategies tailored for solid tumor therapy.
RESUMO
Aims@#Hypoxia is believed to be one of the key components contributing to the clinical resistance of cancer therapies. Alternative strategies are under investigation to overcome this resistance and the oncolytic virus stands amongst the others. Newcastle disease virus (NDV) has been demonstrated to possess oncolytic activity against cancer cells. The present study investigated the effects of oncolytic NDV strain AF2240 and V4-UPM on osteosarcoma cells (Saos-2) under normoxic and hypoxic conditions. @*Methodology and results@#Results showed that the NDV strain AF2240 and V4-UPM could infect and kill normoxic and hypoxic Saos-2 cells equally well by inducing hypoxia-independent apoptosis, and S-phase cell cycle arrest under the microscopy examination, cell viability assay, Annexin V apoptosis assay and cell cycle analysis experiments. However, the Velogenic NDV strain AF2240 excelled over the lentogenic NDV V4-UPM with increased oncolytic effects in Saos-2 cells.@*Conclusion, significance and impact of study@#In a nutshell, normoxia or hypoxia microenvironment has little effect on NDV-induced oncolysis of Saos-2 cancer cells which poses as a potential agent for the treatment of resistant cancer.
RESUMO
Glioma is a highly aggressive malignant tumor of the central nervous system that necessitates active treatment through surgery,radiotherapy,and chemotherapy.However,the prognosis of high-grade gliomas[World Health Organization(WHO)classification of central nervous system tumorsgrade Ⅲ-Ⅳ]remains poor,thus new treatment strategies are urgently needed.Oncolytic virus(OV)therapy is a kind of immunotherapy that can specifically infect and effectively kill tumor cells while activating anti-tumor immunity.The oncolytic herpes simplex virus(oHSV)is expected to emerge as a new adjuvant treatment for glioma due to its unique advantages.This article reviews the current understanding of oHSV,the anti-tumor mechanism of OV,the current clinical research status of oHSV targeted therapy for glioma,the research progress of oHSV collaborative anti-tumor strategy,and the existing problems in oHSV anti-glioma research,aiming to provide valuable insights for the treatment of glioma.
RESUMO
Since the oncolytic herpes simplex virus T-VEC was approved in the United States for the treatment of malignant melanoma in 2015, there has been increasing interests in the oncolytic virus therapy. The oncolytic virus therapy also occupies a certain position in the treatment research process of non-small cell lung cancer(NSCLC). Based on the rapid development of genetic engineering and protein engineering, researchers have designed many recombinant oncolytic viruses targeting various specific sites to further improve their targeting and oncolytic effect in order to alleviate symptoms and even cure NSCLC patients. This review introduces the two major classifications of oncolytic viruses, wild type and gene-edited, and how they achieve tumor lysis by specifically targeting and killing tumor cells. We focus on the research progress of oncolytic virus applied alone to treat NSCLC, or combined with chemotherapy, immunotherapies such as chimeric antigen receptor (CAR)-T cell therapy, immune checkpoint inhibitors and other current hot research to treat NSCLC. At the same time, we summarize and discuss the issue of targeted transport, which is of high concern in the academic field of oncolytic virus therapy, and point out that the use of extracellular vesicles as drug carriers has a good potential for development. Finally, we analyze the existing problems and future application prospects in the context of existing basic and clinical studies, to expend new approaches for the treatment of NSCLC, so that it is no longer limited to traditional therapies.
RESUMO
This study constructed a nano-drug delivery system, A3@GMH, by co-delivering the stapled anoplin peptide(Ano-3, A3) with the light-harvesting material graphene oxide(GO), and evaluated its oncolytic immunotherapy effect on triple-negative breast cancer(TNBC). A3@GMH was prepared using an emulsion template method and its physicochemical properties were characterized. The in vivo and in vitro photothermal conversion abilities of A3@GMH were investigated using an infrared thermal imager. The oncoly-tic activity of A3@GMH against TNBC 4T1 cells was evaluated through cell counting kit-8(CCK-8), lactate dehydrogenase(LDH) release, live/dead cell staining, and super-resolution microscopy. The targeting properties of A3@GMH on 4T1 cells were assessed using a high-content imaging system and flow cytometry. In vitro and in vivo studies were conducted to investigate the antitumor mechanism of A3@GMH in combination with photothermal therapy(PTT) through inducing immunogenic cell death(ICD) in 4T1 cells. The results showed that the prepared A3@GMH exhibited distinct mesoporous and coated structures with an average particle size of(308.9±7.5) nm and a surface potential of(-6.79±0.58) mV. The encapsulation efficiency and drug loading of A3 were 23.9%±0.6% and 20.5%±0.5%, respectively. A3@GMH demonstrated excellent photothermal conversion ability and biological safety. A3@GMH actively mediated oncolytic features such as 4T1 cell lysis and LDH release, as well as ICD effects, and showed enhanced in vitro antitumor activity when combined with PTT. In vivo, A3@GMH efficiently induced ICD effects with two rounds of PTT, activated the host's antitumor immune response, and effectively suppressed tumor growth in 4T1 tumor-bearing mice, achieving an 88.9% tumor inhibition rate with no apparent toxic side effects. This study suggests that the combination of stapled anoplin peptide and PTT significantly enhances the oncolytic immunotherapy for TNBC and provides a basis for the innovative application of anti-tumor peptides derived from TCM in TNBC treatment.
Assuntos
Humanos , Animais , Camundongos , Terapia Fototérmica , Neoplasias de Mama Triplo Negativas/patologia , Peptídeos Catiônicos Antimicrobianos , Imunoterapia/métodos , Linhagem Celular Tumoral , Fototerapia/métodos , Nanopartículas/químicaRESUMO
Pancreatic cancer is one of the most common tumors in digestive system, which is characterized by insidious clinical symptoms, strong invasion, easy metastasis and high mortality. In recent years, immunotherapy is a new direction to the treatment of solid tumors, but its applica-tion in pancreatic cancer is limited by tumor microenvironment of pancreatic cancer. The authors systematically analyze the tumor microenvironment of pancreatic cancer, summarize the clinical researches related to pancreatic cancer immunotherapy, and discuss the prospect of pancreatic cancer immunotherapy.
RESUMO
Objective:To study the effect of splenic lymphocytes isolated from mouse models of colorectal carcinoma with liver metastases induced by oncolytic herpes simplex virus type Ⅱ(oHSV2) on the growth of pulmonary metastases of colorectal carcinoma.Methods:A total of 18 6-week-old BALB/c female mice were selected, colorectal carcinoma cell line CT26 of mice in logarithmic phase was inoculated at the right back (2×10 5 per mouse) and spleen (1×10 5 per mouse) of mice, and tumor cells had hematogenous metastasis to liver through splenic vein. CT26 colorectal carcinoma with liver metastases model was constructed. All mice were respectively divided into oHSV2 group and phosphatic buffered saline (PBS) group, 9 mice in each group according to the random number table method. Mice in oHSV2 group were treated with subcutaneous intratumoral multi-point injection of 100 μl oHSV2 (the multiplicity of infection was 1) for 6 cycles, while mice in PBS group were treated with subcutaneous intratumoral multi-point injection of 100 μl PBS for 6 cycles in total, once injection every other day; the survival of mice was analyzed by using Kaplan-Meier method and tumor growth was observed. The mice of both groups in mouse models of colorectal carcinoma with liver metastases were killed on day 20 and their splenic lymphocytes were isolated. After investigation of the most suitable inoculation number and the optimal observation time of colorectal carcinoma with pulmonary metastases CT26 cell lines, 9 6-week-old BALB/c female mice were divided into the experimental group, the negative control group and the blank control group according to the random number table method, with 3 mice in each group. Mice in the experimental group were injected with splenic lymphocytes (4×10 7 per mouse) and CT26 cells (2×10 5 per mouse) isolated from mouse models induced by oHSV2 via the tail vein, mice in the negative control group were injected with splenic lymphocytes (4×10 7 per mouse) and CT26 cells (2×10 5 per mouse) isolated from normal mice with same weeks old via the tail vein, and mice in the blank control group were injected with only CT26 cells (2×10 5 per mouse) via the tail vein. The above 3 groups were executed on day 10 after inoculation, and tumor growth, histopathological changes of mice were also observed; the survival of mice was analyzed by using Kaplan-Meier method. Results:In models of colorectal carcinoma with liver metastases, liver metastases lesions were not detected in 7 mice and 2 mice had 1-2 liver metastases lesions with long diameter less than 2 mm of oHSV2 group; in PBS group, 9 mice all had multiple liver metastases lesions with tumor long diameter ranging from 1 to 10 mm. And mice in oHSV2 group survived much longer than that of mice in PBS group ( P < 0.001). In models of pulmonary metastases, the optimal number of CT26 cells in mouse tail vein was 2×10 5 per mouse; the best observation time point was day 10 after tail vein injection. On day 24 after inoculation, all mice in the negative control group and the blank control group died, while mice in the experimental group all survived on day 60, and the difference of the overall survival in the above 3 groups was statistically significant ( P = 0.007). HE staining results showed that the lung tissues of the experimental group did not show clear tumor cells, whereas the lung tissues of the negative control group and the blank control group showed extensive diffuse tumor cells. Conclusions:Splenic lymphocytes produced by oHSV2 induction in mouse models of colorectal carcinoma with liver metastases can effectively inhibit the development of pulmonary metastases in colorectal carcinoma CT26 cell of mice.
RESUMO
Gene editing technology CRISPR/Cas9 and its derivative editing technologies including base editor and prime editor can precisely edit the target genome sequences, having been widely used in tumor therapy and achieved remarkable clinical results in tumor immunotherapy, human papilloma virus infection treatment and oncolytic virotherapy, providing a new means for tumor therapy.
RESUMO
Objective To explore whether PI3K inhibitor combined with oncolytic virus can play an effective oncolytic effect on osteosarcoma. Methods The cytotoxicity to tumor cells was detected by MTT method, and the mechanism of enhancing the anti-tumor activity was explored by observation of the swelling of endoplasmic reticulum using electron microscope and the expression of apoptosis-related proteins using Western blotting. The tumor clearance ability of the combination of the PI3k inhibitor ZSTK474 and vesicular stomatitis virus A51 (VSVA51) was verified by anti-tumor experiment in vivo. The apoptosis of tumor cells was verified by immunohistochemistry. Results PI3K inhibitor could be used as sensitizers of oncolytic VSVA51, and confirmed that the)' promoted the strong apoptosis of osteosarcoma cells by aggravating the stress of endoplasmic reticulum in tumor cells (P < 0 . 01). In vivo experiments also showed that PI3K inhibitors combined with VSVA51 could significantly promote the oncolytic effect of osteosarcoma (P<0.001), and this combination therapy enhanced the infiltration of immune cells in the tumor (P<0.001). Conclusion PI3K inhibitors combined with oncolytic virus is a potential therapy for osteosarcoma.
RESUMO
Abstract Background: Viruses are being used as alternative and complementary tools for treating cancers. Oncolytic viruses exhibit tumor tropism, ability to enhance anti-tumor immunity and ability to be used in combination with conventional chemotherapy and radiotherapy. We have recently selected some rotavirus isolates which are adapted to efficiently infect and kill tumor cell lines. Aim: We tested five tumor cell-adapted rotavirus isolates for their ability to infect the human adenocarcinoma cell line MCF-7. Methods: Cell surface membrane-associated proteins mediating virus particle attachment were characterized using ELISA, immunoprecipitation, FACS analysis, and antibody blocking. Results: It was found that heat shock proteins (HSPs) such as Hsp90, Hsp70, Hsp60, and Hsp40 are expressed on the cell surface forming complexes with protein disulfide isomerase (PDI), integrin β3, and heat shock cognate protein 70 (Hsc70) in lipid raft microdomains. Interaction of rotavirus isolates with these cellular proteins was further confirmed by a competition assay and an inhibition assay involving the HSPs tested. Conclusion: Our findings suggest that the tumor cell-adapted rotavirus isolates studied here offer a promising tool for killing tumor cells, thus encouraging further research into this topic, including animal models.
Resumen Antecedentes: Los virus se utilizan como herramientas alternativas y complementarias para el tratamiento del cáncer. Los virus oncolíticos exhiben tropismo por tumores, capacidad para intensificar la inmunidad antitumoral y la capacidad para utilizarse en combinación con quimioterapia y radioterapia convencionales. Recientemente, hemos seleccionado algunos aislamientos de rotavirus que están adaptados para infectar y eliminar de manera eficiente líneas de células tumorales. Objetivo: Se ensayaron cinco aislamientos de rotavirus adaptados a células tumorales para determinar su capacidad para infectar la línea celular de adenocarcinoma humano MCF-7. Métodos: Las proteínas asociadas a la membrana de la superficie celular que median la unión de partículas de virus se caracterizaron mediante ELISA, inmunoprecipitación, análisis FACS y bloqueo de anticuerpos. Resultados: Se encontró que las proteínas de choque térmico (HSPs) como Hsp90, Hsp70, Hsp60 y Hsp40 se expresan en la superficie celular formando complejos con la proteína disulfuro isomerasa (PDI), la integrina β3 y la proteína análoga de choque térmico 70 (Hsc70) en microdominios lipídicos (rafts). La interacción de los aislamientos de rotavirus con estas proteínas celulares se confirmó adicionalmente mediante un ensayo de competición y un ensayo de inhibición que incluía las HSP ensayadas. Conclusión: Nuestros hallazgos sugieren que los aislamientos de rotavirus adaptados a las células tumorales estudiados aquí ofrecen una herramienta prometedora para eliminar las células tumorales, lo que estimula más investigaciones sobre este tema, incluidos los modelos animales.
Assuntos
Humanos , Adenocarcinoma , Rotavirus , Vírus Oncolíticos , Proteínas de Choque Térmico , Adenocarcinoma/terapia , Proteínas de Choque Térmico HSC70 , Células MCF-7RESUMO
OBJECTIVE:To study the effects and mechanism of oncolytic virus M 1(called M 1 virus for short )inducing the apoptosis of cervical cancer C-33A cells. METHODS :MTT assay was used to detect survival rate of C- 33A cells that were treated with different titers (0,0.001,0.01,0.1,1,10 PFU/cell)of M 1 virus. C- 33A cells were divided into control group (0 PFU/cell), low-dose,medium-dose and high-dose groups of M 1 virus(0.001,0.01,0.1 PFU/cell). After treated with corresponding titers of M1 virus for 48 h,flow cytometry was used to detect the apoptotic rate and infection rate of cells;Western blot was performed to detect the protein expression of C/EBP homologous proteins (CHOP),caspase-12,caspase-3 and cleaved-caspase- 3. RESULTS : After treated with different titers of M 1 virus,the survival rate of C- 33A cells decreased significantly (P<0.01),and showed a dose-dependent tr end. Compared with control group ,the apoptotic rate and infection rate of cells in M 1 virus groups as well as the protein expression of CHOP ,caspase-12 and cleaved-caspase- 3(except for medium-dose group )in M 1 virus medium-dose and high-dose groups were increased significantly (P<0.01). CONCLUSIONS :M1 virus can induce the apoptosis of cervical cancer C-33A cells ,and its mechanism may be related to the activation of endoplasmic reticulum stress pathway.
RESUMO
@#[Abstract] Objective: To explore the anti-tumor activity of oncolytic adenovirus co-expressing lymphocyte activation gene 3 (LAG-3) antibody (aLAG) against glioblastoma. Methods: aLAG sequence was inserted into the skeleton of oncolytic adenovirus Ad3 to obtain recombinant oncolytic adenovirus (Ad3-aLAG). The expression of aLAG in infected gliblastoma GL261 cells was detected by WB. The cytotoxicity of recombinant oncolytic adenovirus against glioblastoma was detected by MTT method. The tumor inhibitory activity of recombinant oncolytic adenovirus against glioblastoma in vivo was evaluated with mice subcutaneous xenograft model. Tumor infiltrating T cells were detected by immunohistochemical staining, and the levels of cytokines TNF-α and IFN-γ secreted by tumor infiltrating T cells were detected by Flow cytometry. Results: The recombinant oncolytic adenovirus was successfully constructed, which could effectively express aLAG and kill GL261 cells in vitro (all P<0.01). Experimental results of mice subcutaneous xenograft model showed that the tumor inhibition ability of recombinant oncolytic adenovirus Ad3-aLAG was stronger than that of Ad3 oncolytic adenovirus (P<0.01), and Ad3-aLAG could effectively enhance the infiltration of CD3+ T cells in tumor tissue (P<0.01) and enhance the IFN-γ secretion ability of infiltrating T cells (P<0.01). Conclusion: Ad3-aLAG recombinant oncolytic adenovirus can significantly inhibit the growth of glioblastoma cells in vivo and in vitro, and enhance the anti-tumor immune response in vivo, which is promising to provide a new scheme for the treatment of glioblastoma.
RESUMO
BACKGROUND@#Lung cancer is one of the leading causes of cancer-related morbidity and mortality. Oncolytic virotherapy is an emerging therapeutic modality that utilizes replication-competent viruses to destroy cancers. As a powerful tool to kill tumor cells with excellent safety profile, attenuated measles virus of the Edmonston strain (MV-Edm) has been widely applied in the development of tumor therapy and preclinical trials. The aim of this study was to investigate the synergistic effect of nuclear factor kappa B (NF-κB) signaling pathway inhibitor and oncolytic measles virus vaccine against lung cancer and the involved mechanisms.@*METHODS@#Using Western blot to detect MV-Edm infection of A549 and H1299 were infected by MV-Edm alone or used the NF-κB pathway inhibitor PS1145/cell autophagy related siRNA, expression level of p-IκBα, IκBα, PARP and BAX were determined by western blot. Using flow cytometry to analysis the rate of apoptosis, and using MTT [3-(4,5)-dimethylthiahiazo(-z-y1)-3,5-di-phenytetrazoliumromide] method to detect the cell survival rate.@*RESULTS@#Inhibition of cell autophagy could obviously inhibit the MV-Edm infection induced the NF-κB pathway activation in A549 and H1299. In MV-Edm infected A549 and H1299, p-IκBα level increased and IκBα level decreased over infection time, compared with control group. Inhibition of the NF-κB pathway by PS1145 could promote the apoptosis of MV-Edm infected A549 and H1299 and amplify the tumor killing effect.@*CONCLUSIONS@#The combination of NF-κB signaling pathway inhibitor pS1145 and oncolytic measles virus vaccine strains can promote the apoptosis of human lung cancer cells A549 and H1299 and enhance their oncolytic effect.