Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
1.
Colomb. med ; 52(1): e2024196, Jan.-Mar. 2021. tab, graf
Article in English | LILACS | ID: biblio-1249638

ABSTRACT

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.


Subject(s)
Humans , Adenocarcinoma , Rotavirus , Oncolytic Viruses , Heat-Shock Proteins , Adenocarcinoma/therapy , HSC70 Heat-Shock Proteins , MCF-7 Cells
2.
Biomédica (Bogotá) ; 40(2): 362-381, abr.-jun. 2020. graf
Article in English | LILACS | ID: biblio-1124231

ABSTRACT

Introduction: Cancer is the second leading cause of death in the United States, surpassed only by cardiovascular disease. However, cancer has now overtaken cardiovascular disease as the main cause of death in 12 countries in Western Europe. The burden of cancer is posing a major challenge to health care systems worldwide and demanding improvements in methods for cancer prevention, diagnosis, and treatment. Alternative and complementary strategies for orthodox surgery, radiotherapy, and chemotherapy need to be developed. Objective: To determine the oncolytic potential of tumor cell-adapted rotavirus in terms of their ability to infect and lysate murine myeloma Sp2/0-Ag14 cells. Materials and methods: We inoculated rotaviruses Wt1-5, WWM, TRUYO, ECwt-O, and WTEW in Sp2/0-Ag14 cells and we examined their infectious effects by immunocytochemistry, immunofluorescence, flow cytometry, and DNA fragmentation assays. Results: Rotavirus infection involved the participation of some heat shock proteins, of protein disulfide isomerase (PDI), and integrin ß3. We detected the accumulation of viral antigens within the virus-inoculated cells and in the culture medium in all the rotavirus isolates examined. The rotavirus-induced cell death mechanism in Sp2/0-Ag14 cells involved changes in cell membrane permeability, chromatin condensation, and DNA fragmentation, which were compatible with cytotoxicity and apoptosis. Conclusions: The ability of the rotavirus isolates Wt1-5, WWM, TRUYO, ECwt-O, and WTEW to infect and cause cell death of Sp2/0-Ag14 cells through mechanisms that are compatible with virus-induced apoptosis makes them potential candidates as oncolytic agents.


Introducción. El cáncer es la segunda causa de muerte en los Estados Unidos, solamente superado por la enfermedad cardiovascular. Sin embargo, el cáncer aventaja a la enfermedad cardiovascular como primera causa de muerte en doce países de Europa occidental. Se requieren mejores métodos de prevención, diagnóstico y tratamiento para afrontar el gran desafío que el cáncer representa mundialmente para los sistemas de salud, y se necesita desarrollar estrategias alternativas y complementarias a la cirugía, la radioterapia y la quimioterapia convencionales. Objetivo. Evaluar el potencial oncolítico de rotavirus adaptados a células tumorales por su capacidad para infectar y lisar células Sp2/0-Ag14 de mieloma de ratón. Materiales y métodos. Los aislamientos de rotavirus Wt1-5, WWM, TRUYO, ECwt-O y WTEW se inocularon en células Sp2/0-Ag14 y se examinaron sus efectos infecciosos mediante inmunocitoquímica, inmunofluorescencia, citometría de flujo y ensayos de fragmentación del ADN. Resultados. La infección con los rotavirus Wt1-5, WWM, TRUYO, ECwt-O y WTEW implicó la participación de algunas proteínas de choque térmico, la proteína disulfuro isomerasa y la integrina ß3. La acumulación de antígenos virales intracelulares y extracelulares se detectó en todos los virus utilizados. Los mecanismos de muerte inducidos por los rotavirus en células Sp2/0-Ag14 indujeron cambios en la permeabilidad de la membrana celular, la condensación de cromatina y la fragmentación de ADN, los cuales fueron compatibles con citotoxicidad y apoptosis. Conclusiones. La capacidad de los rotavirus estudiados para infectar y causar la muerte de células Sp2/0-Ag14 mediante mecanismos compatibles con la apoptosis inducida viralmente los convierte en candidatos potenciales para ser utilizados como agentes oncolíticos.


Subject(s)
Oncolytic Viruses , Neoplasms/therapy , Rotavirus Infections
3.
Chinese Journal of Biotechnology ; (12): 1269-1276, 2020.
Article in Chinese | WPRIM | ID: wpr-826850

ABSTRACT

Human adenoviruses are widespread causative agent that induces respiratory diseases, epidemic keratoconjunctivitis and other related diseases. Adenoviruses are commonly used in experimental and clinical areas. It is one of the most commonly used virus vectors in gene therapy, and it has attracted a lot of attention and has a high research potential in tumor gene therapy and virus oncolytic. Here, we summarize the biological characteristics, epidemiology and current application of adenovirus, in order to provide reference for engineering application of adenovirus.


Subject(s)
Humans , Adenovirus Infections, Human , Epidemiology , Virology , Adenoviruses, Human , Genetics , Genetic Engineering , Methods , Genetic Vectors , Oncolytic Virotherapy , Oncolytic Viruses , Genetics , Virus Replication
4.
Journal of Liver Cancer ; : 1-11, 2019.
Article in English | WPRIM | ID: wpr-765710

ABSTRACT

Systemic target therapeutic drugs, such as sorafenib, lenvatinib, or regorafenib are the only drugs that are known to be effective against advanced hepatocellular carcinoma (HCC). However, these agents show a limited efficacy in killing residual tumors. Immunotherapy is an alternative approach to this treatment and has been used to successfully treat different cancers, including HCC. HCC is an inflammation-induced cancer and represents a very interesting target for immunotherapeutics. Immunotherapies aim to reverse the immune tolerance and suppression found in tumor microenvironments and include approaches, such as adoptive cell therapy, immune checkpoint inhibition, and cancer vaccination. Adoptive cell therapy uses autologous natural killer or cytokine-induced killer cells by cultivating them ex vivo and subsequently reinfusing them into the patient. Immune checkpoint inhibitors reactivate tumor-specific T cells by suppressing checkpoint-mediated inhibitory signaling. Cancer vaccination induces a tumor-specific immune response by activating effector T lymphocytes. A wide range of potential immunotherapy-related adverse events occur; therefore, a multidisciplinary collaborative management is required across the clinical spectrum. This review summarizes the current status of immunotherapy for HCC and provides a perspective on its future applications.


Subject(s)
Humans , Carcinoma, Hepatocellular , Cell- and Tissue-Based Therapy , Cytokine-Induced Killer Cells , Homicide , Immune Tolerance , Immunotherapy , Neoplasm, Residual , Oncolytic Viruses , T-Lymphocytes , Tumor Microenvironment , Vaccination
5.
Osong Public Health and Research Perspectives ; (6): 246-252, 2019.
Article in English | WPRIM | ID: wpr-760706

ABSTRACT

OBJECTIVES: Vesicular stomatitis virus (VSV) is under development as an oncolytic virus due to its preferential replication in cancer cells and oncolytic activity, however the viral components responsible have not yet been determined. In this study the effects of VSV wild-type (wt) and M51R-mutant matrix proteins (M51R-mMP) on apoptosis, pyroptosis, necroptosis, and autophagy pathways, in an esophagus cancer cell line (KYSE-30) were investigated. METHODS: The KYSE-30 cells were transfected with pcDNA3.1 plasmids encoding wt or M51R-mMP, and apoptosis, pyroptosis, necroptosis, and autophagy were evaluated 48 and 72 hours after transfection. RESULTS: KYSE-30 cells transfected with VSV wt and M51R-mMPs significantly reduced cell viability to < 50% at 72 hours post-transfection. M51R-MP significantly increased the concentration of caspase-8 and caspase-9 at 48 and 72 hours post-transfection, respectively ( p < 0.05). In contrast, no significant changes were detected following transfection with the VSV wt plasmid. Moreover, VSV wt and M51R-mMP transfected cells did not change the expression of caspase-3. VSV wt and M51R-mMPs did not mMP change caspase-1 expression (a marker of pyroptosis) at 48 and 72 hours post-transfection. However, M51R-mMP and VSV wt transfected cells significantly increased RIP-1 (a marker of necroptosis) expression at 72 hours post-infection ( p < 0.05). Beclin-1, a biomarker of autophagy, was also induced by transfection with VSV wt or M51R-mMPs at 48 hours post-transfection. CONCLUSION: The results in this study indicated that VSV exerts oncolytic activity in KYSE-30 tumor cells through different cell death pathways, suggesting that M51R-mMP may potentially be used to enhance oncolysis.


Subject(s)
Apoptosis , Autophagy , Carcinoma, Squamous Cell , Caspase 3 , Caspase 8 , Caspase 9 , Cell Death , Cell Line , Cell Survival , Epithelial Cells , Esophageal Neoplasms , Oncolytic Viruses , Plasmids , Pyroptosis , Transfection , Vesicular Stomatitis , Viral Structures
6.
IBJ-Iranian Biomedical Journal. 2017; 21 (4): 206-217
in English | IMEMR | ID: emr-189230

ABSTRACT

Background: Oncolytic herpes simplex virus [oHSV]-based vectors lacking gamma 34.5 gene, are considered as ideal templates to construct efficient vectors for [targeted] cancer gene therapy. Herein, we reported the construction of three single/dually-flourescence labeled and gamma 34.5-deleted, recombinant HSV-1 vectors for rapid generation and easy selection/isolation of different HSV-Based vectors


Methods: Generation of recombinant viruses was performed with conventional homologous recombination methods using green fluorescent protein [GFP] and BleCherry harboring shuttle vectors. Viruses were isolated by direct fluorescence observation and standard plaque purifying methods and confirmed by PCR and sequencing and flow cytometry. XTT and plaque assay titration were performed on Vero, U87MG, and T98 GBM cell lines


Results: We generated three recombinant viruses, HSV-GFP, HSV-GR [Green-Red], and HSV-Red. The HSV-GFP showed two log higher titer [1010 PFU] than wild type [108 PFU]. In contrast, HSV-GR and HSV-Red showed one log lower titer [107 PFU] than parental HSV. Cytotoxicity analysis showed that HSV-GR and HSV-Red can lyse target tumor cells at multiplicity of infection of 10 and 1 [P<0.001]. Moreover, HSV-GFP showed higher infection potency [98%] in comparison with HSV-GR [82%]


Conclusion: Our oHSVs provide a simple and an efficient platform for construction and rapid isolation of 2[nd] and 3[rd] generation oHSVs by replacing the inserted dyes with transgenes and also for rapid identification via fluorescence activated cell sorting. These vectors can also be used for tracing the efficacy of therapeutic agents on target cells, imaging of neural or tumoral cells in vitro/in vivo and as oncolytic agents in cancer therapy


Subject(s)
Oncolytic Viruses , Homologous Recombination , Flow Cytometry , Microscopy, Fluorescence
7.
Journal of Taibah University Medical Sciences. 2016; 11 (3): 187-193
in English | IMEMR | ID: emr-180217

ABSTRACT

Oncolytic viruses [OVs] are an emerging class of novel anti-cancer therapeutic agents that selectively infect and destroy cancerous tissues without damaging normal cells. With the recent US Food and Drug Administration [FDA] approval of Herpes Virus [T-VEC] for the treatment of advanced melanoma, oncolytic virotherapy has gained more attention for further development as a novel form of immunotherapy. A viable approach to maximize the efficacy of OVs involves arming them with immuneenhancing cytokines that are capable of boosting the host's immune response to effectively attack tumour cells. Interleukin-12 [IL-12] is a powerful cytokine with potent antitumour activities that activates both innate and adaptive anti-tumour responses. Several studies have demonstrated that IL-12-expressing OVs improve the therapeutic index in pre-clinical tumour models by activating and recruiting dendritic cells [DCs], cytotoxic natural killer [NK] cells and cytotoxic T cells, which subsequently improve tumour clearance. In this review, the immunological mechanisms of IL-12eexpressing viruses are discussed


Subject(s)
Oncolytic Viruses , Antineoplastic Agents , Immunotherapy , Interleukin-12
8.
Journal of Bacteriology and Virology ; : 126-131, 2015.
Article in English | WPRIM | ID: wpr-194339

ABSTRACT

Naturally occurring reoviruses are live replication-proficient viruses specifically infecting human cancer cell while sparing normal counterpart. Since the discovery of reoviruses in 1950s, reoviruses have shown various degrees of safety and efficacy in pre-clinical or clinical application for human anti-cancer therapeutics. I have recently shown that cellular tumor suppressor genes, such as p53, ATM (Ataxia telangiectasia mutated), and RB (Retinoblastoma associated), are important in determining reoviral oncotropism. Thus, it is interesting to examine whether the aberrancy of c-Myc expression, whose normal function also plays an important role in the maintenance of genomic integrity, could affect reoviral oncolytic tropism. Hs68 cells are non-tumorigenic normal cells and resistant to reoviral cytopathic effects. Importantly, I found that c-Myc overexpression in human HS68 cells effectively induced reovirus cytophatic effects compared to mock expressed cells as shown by the typical reoviral cytophathology and an increased level of caspase-3 activity. Taken together, overexpression of c-Myc could play an important role in determining reoviral oncolytic tropism.


Subject(s)
Humans , Caspase 3 , Genes, Tumor Suppressor , Oncogenes , Oncolytic Viruses , Telangiectasis , Tropism
9.
Iranian Journal of Cancer Prevention. 2013; 6 (2): 101-107
in English | IMEMR | ID: emr-127021

ABSTRACT

New cancer therapies with novel mechanisms and functions are needed to treat patients with different cancers. Virotherapy is a good scenario for such treatment. The advantages of virotherapy include the potential lack of cross resistance with standard therapies and the ability to cause tumor destruction by numerous mechanisms. Oncolytic virus not only possesses unique mechanisms of action that are distinct from other treatment modalities, its self-perpetuating nature provides an ideal platform for therapeutic transgenic insertion. In this review article, a variety of oncolytic viruses in cancer gene therapy will be described


Subject(s)
Neoplasms , Oncolytic Viruses , Genetic Therapy
10.
Article in English | IMSEAR | ID: sea-144784

ABSTRACT

Cancer is one of the major causes of death worldwide. In spite of achieving significant successes in medical sciences in the past few decades, the number of deaths due to cancer remains unchecked. The conventional chemotherapy and radiotherapy have limited therapeutic index and a plethora of treatment related side effects. This situation has provided an impetus for search of novel therapeutic strategies that can selectively destroy the tumour cells, leaving the normal cells unharmed. Viral oncotherapy is such a promising treatment modality that offers unique opportunity for tumour targeting. Numerous viruses with inherent anti-cancer activity have been identified and are in different phases of clinical trials. In the era of modern biotechnology and with better understanding of cancer biology and virology, it has become feasible to engineer the oncolytic viruses (OVs) to increase their tumour selectivity and enhance their oncolytic activity. In this review, the mechanisms by which oncolytic viruses kill the tumour cells have been discussed as also the development made in virotherapy for cancer treatment with emphasis on their tumour specific targeting.


Subject(s)
Apoptosis , Humans , Neoplasms/drug therapy , Neoplasms/radiotherapy , Neoplastic Stem Cells , Oncolytic Viruses/pathogenicity , Oncolytic Viruses/metabolism , Oncolytic Virotherapy/methods
11.
Chinese Journal of Oncology ; (12): 89-95, 2012.
Article in Chinese | WPRIM | ID: wpr-335337

ABSTRACT

<p><b>OBJECTIVE</b>The aim of this study was to construct a new oncolytic virus oHSV2hGM-CSF and evaluate its oncolytic activity in vitro and in vivo in parallel with oHSV1hGM-CSF.</p><p><b>METHODS</b>oHSV2hGM-CSF was a replication-competent, attenuated HSV2 based on the HG52 virus (an HSV2 strain). It was engineered to be specific for cancer by deletion of the viral genes ICP34.5 and ICP47 and insertion of the gene encoding hGM-CSF. To measure the in vitro killing effect of the virus, 15 human tumor cell lines (HeLa, Eca-109, PG, HepG2, SK/FU, CNE-2Z, PC-3, SK-OV3, A-549, 786-0, MCF-7, Hep-2, HT-29, SK-Mel-28, U87-MG) and mouse melanoma (B16R) cell line were seeded into 24-well plates and infected with viruses at MOI = 1 (multiplicity of infection, MOI), or left uninfected. The cells were harvested 24 and 48 hours post infection, and observed under the microscope. For animal studies, the oncolytic viruses were administered intratumorally (at 3-day interval) at a dose of 2.3 x 10(6) PFU (plaque forming unit, PFU) for three times when the tumor volume reached 7-8 mm3. The tumor volume was measured at 3-day intervals and animal survival was recorded.</p><p><b>RESULTS</b>Both oHSV2hCM-CSFand oHSV1hGM-CSF induced widespread cytopathic effects at 24 h after infection. OHSV2hGM-CSF, by contrast, produced more plaques with a syncytial phenotype than oHSV1hGM-CSF. In the in vitro killing experiments for the cell lines HeLa, HepG2, SK-Mel-28, B16R and U87-MG, oHSV2hGM-CSF eradicated significantly more cells than oHSV1hGM-CSF under the same conditions. For the mouse experiments, it was observed that oHSV2hGM-CSF significantly inhibited the tumor growth. At 15 days after B16R tumor cells inoculation, the tumor volumes of the PBS, oHSV1hGCM-CSF and oHSV2hGM-CSF groups were (374.7 +/- 128.24) mm3, (128.23 +/- 45.32) mm3 (P < 0.05, vs. PBS group) or (10.06 +/- 5.1) mm3 (P < 0.01, vs. PBS group), respectively (mean +/- error). The long term therapeutic effect of oHSV2hGM-CSF on the B16R animal model was evaluated by recording animal survival over 110 days after tumor cells inoculation whereas all the mice in the PBS group died by day 22 (P < 0.01). The anti-tumor mechanism of the newly constructed oHSV2hGM-CSF against B16R cell tumor appeared to include the directly oncolytic activity and the induction of anti-tumor immunity to some degree.</p><p><b>CONCLUSION</b>The findings of our study demonstrate that the newly constructed oHSV2hGM-CSF has potent anti-tumor activity in vitro to many tumor cell lines and in vive to the transplanted B16R tumor models.</p>


Subject(s)
Animals , Female , Humans , Mice , Cell Line, Tumor , Gene Deletion , Genetic Engineering , Granulocyte-Macrophage Colony-Stimulating Factor , Genetics , Herpesvirus 2, Human , Genetics , Allergy and Immunology , Immediate-Early Proteins , Genetics , Metabolism , Melanoma, Experimental , Pathology , Therapeutics , Virology , Mice, Inbred C57BL , Oncolytic Virotherapy , Methods , Oncolytic Viruses , Genetics , Physiology , Random Allocation , Tumor Burden , Viral Proteins , Genetics , Metabolism , Xenograft Model Antitumor Assays
12.
Chinese Journal of Otorhinolaryngology Head and Neck Surgery ; (12): 672-676, 2012.
Article in Chinese | WPRIM | ID: wpr-262512

ABSTRACT

<p><b>OBJECTIVE</b>To evaluate the antitumor efficacy of Ad-TD-RFP for human nasopharyngeal carcinoma cells (C666-1) in vitro and in vivo.</p><p><b>METHODS</b>The oncolytic effects of Ad-TD-RFP and control virus dl11520 on C666-1 cells were determined by cytotoxicity assay (MTS assay). Viral replication of Ad-TD-RFP and dl11520 was detected at different time points (24 h, 48 h, 72 h and 96 h) by tissue culture infective dose (TCID(50)) in C666-1 cells implanted subcutaneously into the flank in each of BALB/c nude mice. The xenografts were injected intratumorally with Ad-TD-RFP or dl1520 to investigate their effects on tumor growth.</p><p><b>RESULTS</b>The concentration for 50% of maximal effect (EC(50)) values of Ad-TD-RFP and dl1520 were (107.6 ± 3.2) pt/cell and (174.1 ± 4.0) pt/cell, respectively (t = 22.6, P < 0.001). The Ad-TD-RFP replication was 3-14 folds more than dl1520 replication at four time points (24 h, 48 h, 72 h and 96 h) in C666-1 cells (t values were 33.6, 23.4, 20.8 and 17.3, respectively, P < 0.001). The average tumor volumes of PBS group, dl1520 group and Ad-TD-RFP group were (1765.5 ± 713.9) mm(3), (1036.9 ± 623.8) mm(3), and (420.8 ± 238.7) mm(3), respectively (F = 12.0, P < 0.05) on day 67 after treatment.</p><p><b>CONCLUSIONS</b>The antitumour efficacy of the novel oncolytic adenovirus Ad-TD-RFP for human nasopharyngeal carcinoma C666-1 cells is superior to that of dl1520 in vitro and in vivo. The outcome of this study provides an experimental basis for the treatment of human nasopharyngeal carcinoma by viral gene therapy.</p>


Subject(s)
Animals , Female , Humans , Mice , Adenoviridae , Classification , Genetics , Carcinoma , Cell Line, Tumor , Genetic Vectors , Mice, Inbred BALB C , Mice, Nude , Nasopharyngeal Neoplasms , Therapeutics , Oncolytic Virotherapy , Oncolytic Viruses , Genetics , Xenograft Model Antitumor Assays
13.
Chinese Journal of Cancer ; (12): 831-841, 2011.
Article in English | WPRIM | ID: wpr-294455

ABSTRACT

Oncolytic herpes simplex virus (HSV) can replicate in and kill cancer cells without harming normal tissue. G47delta is a third-generation HSV vector. In this study, the therapeutic effects of G47delta on human nasopharyngeal carcinoma (NPC) were determined in vitro and in vivo. The human NPC cell lines CNE-2 and SUNE-1, primary normal nasopharyngeal epithelial cells (NPECs), and immortalized nasopharyngeal cells NP-69 and NPEC2/Bmi1 were infected with G47delta at different multiplicities of infection (MOIs). The survival of infected cells was observed daily. Two subcutaneous models of NPC were established with CNE-2 and SUNE-1 in Balb/c nude mice. G47delta or virus buffer as control was injected into the subcutaneous tumors. Tumor size was measured twice a week, and animals were euthanized when the diameter of their tumors exceeded 18 mm or when the animals appeared moribund. For the NPC cell lines CNE-2 and SUNE-1, more than 85% and 95% of cells were killed on day 5 after G47delta infection at MOI = 0.01 and MOI = 0.1, respectively. Similar results were observed for an immortalized cell line NPEC2/Bmi-1. A moderate effect of G47delta was also found on another immortalized cell line NP-69, of which only 27.7% and 75.9% of cells were killed at MOI = 0.01 and MOI = 0.1, respectively. On the contrary, there was almost no effect observed on NPECs. The in vivo experiments showed that tumors in mice in the G47delta-treated group regressed completely, and the mice exhibited much longer survival time than those in the control groups. Our results suggest that the potential therapeutic effects of G47delta would be applicable for treatment of NPC patients in the future.


Subject(s)
Animals , Female , Humans , Mice , Apoptosis , Carcinoma , Cell Line, Tumor , Mice, Inbred BALB C , Mice, Nude , Nasopharyngeal Neoplasms , Pathology , Therapeutics , Virology , Oncolytic Virotherapy , Methods , Oncolytic Viruses , Physiology , Simplexvirus , Physiology , Xenograft Model Antitumor Assays
14.
Chinese Journal of Oncology ; (12): 822-826, 2011.
Article in Chinese | WPRIM | ID: wpr-320129

ABSTRACT

<p><b>OBJECTIVE</b>To study the effect of CEA gene regulation on the anti-tumor activity of oncolytic adenovirus H101 to esophageal carcinoma, and to explore the intrinsic factors influencing H101 sensitivity.</p><p><b>METHODS</b>Stable human esophageal cancer cell line EC9706 cells with lower (EC9706-SCEA) and higher CEA expression (EC9706-CEA) were chosen, thawed and cultured, and then to analyse the influence of CEA expressed at different levels on cell growth. The cytotoxic effect of H101 was assayed by in vitro and nude mouse in vivo.</p><p><b>RESULTS</b>The cell growth experiment showed that the population doubling time of EC9706-SCEA, EC9706-CEA and EC9706 cells were (30.9 ± 2.0) h, (31.1 ± 2.5) h and (29.1 ± 2.6) h, respectively, showing no significant difference among them (P > 0.05). The cytotoxic activity of H101 was higher on EC9706-SCEA than on other four groups, when MOI was ≥ 0.01 PFU (P < 0.05). The mouse experiment showed that H101 inhibited the growth of transplanted tumors in all experimental groups. Its effect on CEA-silenced tumors (inhibition rate was 61.5% to 74.5%) was significantly higher than that on CEA-overexpression tumors (32.3% to 38.5%) and control EC9706 transplanted tumors (35.5% to 44.8%). There was a significant difference between them (P < 0.05).</p><p><b>CONCLUSIONS</b>The results in vitro and in vivo experiments show that H101 can enhance the cytotoxic effect on EC9706 cells with lower CEA expression. To silence the expression of CEA may provide a novel strategy for target gene therapy of esophageal carcinoma.</p>


Subject(s)
Animals , Female , Humans , Mice , Adenoviridae , Physiology , Carcinoembryonic Antigen , Genetics , Metabolism , Cell Line, Tumor , Cell Proliferation , Esophageal Neoplasms , Metabolism , Pathology , Therapeutics , Gene Silencing , Mice, Inbred BALB C , Mice, Nude , Neoplasm Transplantation , Oncolytic Virotherapy , Oncolytic Viruses , Physiology , RNA, Messenger , Metabolism , RNA, Small Interfering , Genetics , Tumor Burden
15.
Acta Pharmaceutica Sinica ; (12): 1476-1482, 2011.
Article in Chinese | WPRIM | ID: wpr-323098

ABSTRACT

To establish a detection method of oncolytic adenovirus/p53 and standard of quality control, human telomerase reverse transcriptase (hTERT) promoter, CMV fusion promoter containing hypoxia reaction element (HRE) and p53 gene were identified by vector DNA restriction enzyme digestion and PCR analysis. The result conformed that all modified regions were in consistent with theoretical ones. Particle number was 2.0 x 10(11) mL(-1) determined by UV (A260). Infectious titer was 5.0 x 10(10) IU mL(-1) analyzed by TCID50. In vitro p53 gene expression in human lung cancer cell H1299 was determined by ELISA, and A450 ratio of nucleoprotein in virus infection group to control group was 5.2. Antitumor potency was evaluated by cytotoxicity assay using human lung cancer cell A549, and the MOI(IC50) of this gene therapy preparation was 1.0. The tumor cells targeted replication ability of recombinant virus was determined by TCID50 titer ratio of filial generation virus between human lung cancer cell A549 and human diploid epidermal fibrolast BJ cells after infected by virus with same MOI. TCID50 titer ratio of tumor cell infection group to normal cell infection control group was 398. The IE-HPLC purity of virus was 99.5%. There was less than 1 copy of wild type adenovirus within 1 x 10(7) VP recombinant virus. Other quality control items were complied with corresponding requirements in the guidance for human somatic cell therapy and gene therapy and Chinese pharmacopeia volume III. The detection method of oncolytic adenovirus/p53 was successfully established for quality control standard. The study also provided reference for quality control of other oncolytic viral vector products.


Subject(s)
Humans , Adenoviridae , Genetics , Metabolism , Physiology , Cell Line, Tumor , Gene Expression Regulation, Neoplastic , Genes, p53 , Genetic Therapy , Genetic Vectors , Neoplasms , Metabolism , Pathology , Virology , Oncolytic Viruses , Genetics , Metabolism , Physiology , Quality Control , Recombinant Fusion Proteins , Genetics , Metabolism , Transfection , Virus Replication
16.
Journal of Zhejiang University. Medical sciences ; (6): 226-230, 2010.
Article in Chinese | WPRIM | ID: wpr-259212

ABSTRACT

<p><b>OBJECTIVE</b>To investigate the inhibitive effects of chimeric oncolytic adenovirus SG235 on leukemia cells in vitro.</p><p><b>METHODS</b>The ability of SG235 to infect leukemia cells and the expression of CD46 on blasts from the patient with leukemia were detected by flow cytometry (FACS). The cytotoxicity of the virus was evaluated by MTT assay. Apoptosis induced by SG235 was detected with Annexin-V/PI staining and TUNEL assay followed by FACS analysis.</p><p><b>RESULT</b>The majority of leukemia cells from the patient with acute leukemia was CD46-positive. GFP-positive cells were 45.1%, 35.7%, 54.2%, 37.0%, 30.1%, %67.1, 17.2% and 33.1% in Mutz-1, Kasumi-1, K562, HL60, Molt- 4, RPMI8226, L428, and Jurkat cell lines treated with SG235-EGFP vector at MOI (multiplicity of infection) of 50 for 48 h.SG235 treatment resulted in marked growth inhibition and apoptosis of Kassumi-1 cells, and also significantly inhibited expression of p-Akt.</p><p><b>CONCLUSION</b>The chimeric oncolytic adenovirus SG235 can infect leukemia cell effectively and results in the growth inhibition and apoptosis of Kasumi-1 cells in vitro.</p>


Subject(s)
Humans , Adenoviridae , Genetics , Apoptosis , Cell Line, Tumor , Cell Proliferation , Genetic Vectors , Leukemia , Genetics , Metabolism , Pathology , Membrane Cofactor Protein , Metabolism , Oncolytic Viruses , Transfection
17.
Chinese Journal of Biotechnology ; (12): 1031-1036, 2010.
Article in Chinese | WPRIM | ID: wpr-292175

ABSTRACT

Naturally existing Newcastle disease virus (NDV) can specifically execute oncolytic ability in clinical studies. Reports from clinical trials using NDV as oncolytic agents showed promise and warrant results in cancer therapy. In recent years, reverse genetics technology has been used widely in the studies of NDV virology. More recently, the technology was applied to optimize the oncolytic efficacy of NDV, for instance, modification of the F gene, and expression of GM-CSF, IFN-gamma, IL-2 or TNF-alpha. NDV is widely investigated in cancer therapy and will definitely offer a prosperous future for clinical cancer therapeutics. We reviewed the developments of cancer therapy by recombinant NDV using reverse genetics technology, as well as our own experience in this domain.


Subject(s)
Animals , Humans , Neoplasms , Pathology , Therapeutics , Newcastle disease virus , Genetics , Physiology , Oncolytic Virotherapy , Methods , Oncolytic Viruses , Genetics , Physiology , Recombination, Genetic
18.
Journal of Biomedical Engineering ; (6): 389-394, 2010.
Article in Chinese | WPRIM | ID: wpr-341612

ABSTRACT

In order to evaluate the biological activity in vitro and the antitumor effects of 131I-conditionally replicating oncolytic adenovirus KH901 on HepG2 human hepatoma xenografts, the leves of GM-CSF expression were determined by ELISA method. A panel of tumor and normal cells was infected with recombinant adenovirus KH901 at MOI of 10 PPC. The medium was harvested to determine the bioactivity of GM-CSF after 24 hours. Nude mice bearing HepG2 human hepatoma xenografts were given 131-KH901. Antitumor effects were assessed using endpoints of tumor growth delay. The data showed that after 24 hours 131-KH901 replicated hugely in tumor cells and produced significant amount of GM-CSF 183.27 +/- 6.90 pg/ml, while producing very small amount of GM-CSF 20.44 +/- 0.77 pg/ml in normal cells. In the treatment of tumor, 131I-KH901 showed higher restraint rate (71.3%) compared to 131I (22.7%) or KH901 (52.7%). Therefore, 131-KH901 can inhibit the growth of human hepatoma cell in nude mice and it may be a potential drug for treating liver cancer.


Subject(s)
Animals , Female , Humans , Male , Mice , Adenoviridae , Genetics , Metabolism , Granulocyte-Macrophage Colony-Stimulating Factor , Genetics , Metabolism , Hep G2 Cells , Iodine Radioisotopes , Liver Neoplasms, Experimental , Diagnostic Imaging , Pathology , Virology , Mice, Nude , Oncolytic Virotherapy , Oncolytic Viruses , Genetics , Metabolism , Radionuclide Imaging
19.
National Journal of Andrology ; (12): 679-683, 2010.
Article in Chinese | WPRIM | ID: wpr-295019

ABSTRACT

<p><b>OBJECTIVE</b>To construct an oncolytic adenovirus with the DD3 promoter regulation, expressing small hairpin RNA and targeting the SATB1 gene (SATBI-shRNA), and to evaluate its potential for inhibiting the growth of human prostatic carcinoma cells (LNCaP) in vitro.</p><p><b>METHODS</b>SATB1-shRNA expression cassettes containing the H1 promoter were produced by PCR from pSilencer3. 1-SATB1 and inserted into the pZD55 vector, and the recombinant plasmid pZD55-SATB1-shRNA was constructed, pZD55SATB1-shRNA and pZXC2-DD3-E1A were digested with EcoRV and Xba I , and the obtained expression cassettes linked each other to construct the recombinant plasmid pDD3-ZD55-SATB1, which was cotransfected with the pBHGE3 packaging plasmids mixture into 293 cells by Effectence. The recombined adenoviruses DD3-ZD55-SATB1 were identified by PCR. Viruses were propagated on HEK293 cells and purified by standard techniques, and the functional PFU titers determined by plaque assay on 293 cells. The antitumor potential of DD3-ZD55-SATB1 to LNCaP was evaluated by the crystal violet dye method. The expression level of the E1A gene was detected by Western blot, and that of the SATB1 gene in the adenovirus-infected LNCaP cells by both Western blot and RT-PCR.</p><p><b>RESULTS</b>An oncolytic adenovirus expressing SATB1-shRNA with the DD3 promoter regulation, DD3-ZD55-SATB1, was constructed successfully, whose functional PFU titer was 1.2 x 10(10) PFU/ml. DD3-ZD55-SATB1 showed an obvious cytopathic effect and a selective expression of E1A in the adenovirus-infected LNCaP cells; it exhibited a high LNCaP-targetability and specific SATB1-silencing effect.</p><p><b>CONCLUSION</b>The successful construction of the oncolytic adenovirus DD3-ZD55-SATB1 offers a new tool for researches on the gene therapy for human prostate cancer.</p>


Subject(s)
Humans , Male , Adenoviridae , Genetics , Carcinoma , Therapeutics , Cell Line, Tumor , Genetic Vectors , Matrix Attachment Region Binding Proteins , Genetics , Oncolytic Virotherapy , Methods , Oncolytic Viruses , Genetics , Promoter Regions, Genetic , Prostatic Neoplasms , Therapeutics , RNA Interference , RNA, Small Interfering , Genetics
20.
Article in English | IMSEAR | ID: sea-135930

ABSTRACT

Cancer is a major cause of deaths in humans. Though there has been significant progress in cancer therapy, the limited efficacy and toxicities of current chemo- and radiotherapies have provided an impetus for the search of new therapeutics. A therapeutic approach, which uses viruses for the treatment of cancer termed, oncolytic virotherapy has recently emerged. Newcastle disease virus (NDV) is one such virus with an inherent oncolytic property. NDV causes a highly infectious disease in poultry worldwide. In humans it is reported to have oncolytic and immuno-stimulatory effects. It specifically replicates in tumour cells while sparing normal cells and cause oncolysis. For many years different strains of the NDV have been investigated for treatment of various human cancers. Recent advances in reverse genetics provided investigators the tools to produce recombinant NDV with improved oncolytic property.


Subject(s)
Animals , Apoptosis , Humans , Neoplasms/pathology , Neoplasms/therapy , Newcastle disease virus/genetics , Newcastle disease virus/physiology , Oncolytic Virotherapy/methods , Oncolytic Viruses/genetics , Oncolytic Viruses/physiology
SELECTION OF CITATIONS
SEARCH DETAIL