Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Biotechnol Bioeng ; 118(9): 3522-3532, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-33818758

RESUMO

Virus-based biologicals are one of the most promising biopharmaceuticals of the 21st century medicine and play a significant role in the development of innovative therapeutic, prophylactic, and clinical applications. Oncolytic virus manufacturing scale can range from 5 L in research and development up to 50 L for clinical studies and reach hundreds of liters for commercial scale. The inherent productivity and high integration potential of periodic counter-current chromatography (PCC) offer a transversal solution to decrease equipment footprint and the reduction of several non-value-added unit operations. We report on the design of an efficient PCC process applied to the intermediate purification of oncolytic adenovirus. The developed ion-exchange chromatographic purification method was carried out using a four-column setup for three different scenarios: (i) variation in the feedstock, (ii) potential use of a post-load washing step to improve virus recovery, and (iii) stability during extended operation. Obtained virus recoveries (57%-86%) and impurity reductions (>80% DNA, and >70% total protein) match or overcome batch purification. Regarding process stability and automation, our results show that not only the dynamic control strategy used is able to suppress perturbations in the sample inlet but also allows for unattended operation in the case of ion exchange capture.


Assuntos
Produtos Biológicos/isolamento & purificação , Vírus Oncolíticos/isolamento & purificação , Células A549 , Distribuição Contracorrente , Humanos
2.
BMC Vet Res ; 16(1): 307, 2020 Aug 25.
Artigo em Inglês | MEDLINE | ID: mdl-32843040

RESUMO

BACKGROUND: Cancer is a leading cause of mortality for both humans and dogs. As spontaneous canine cancers appear to be relevant models of human cancers, developing new therapeutic approaches could benefit both species. Oncolytic virotherapy is a promising therapeutic approach in cancer treatment. TG6002 is a recombinant oncolytic vaccinia virus deleted in the thymidine kinase and ribonucleotide reductase genes and armed with the suicide gene FCU1 that encodes a protein which catalyses the conversion of the non-toxic 5-fluorocytosine into the toxic metabolite 5-fluorouracil. Previous studies have shown the ability of TG6002 to infect and replicate in canine tumor cell lines, and demonstrated its oncolytic potency in cell lines, xenograft models and canine mammary adenocarcinoma explants. Moreover, 5-fluorouracil synthesis has been confirmed in fresh canine mammary adenocarcinoma explants infected with TG6002 with 5-fluorocytosine. This study aims at assessing the safety profile and viral shedding after unique or repeated intramuscular injections of TG6002 in seven healthy Beagle dogs. RESULTS: Repeated intramuscular administrations of TG6002 at the dose of 5 × 107 PFU/kg resulted in no clinical or biological adverse effects. Residual TG6002 in blood, saliva, urine and feces of treated dogs was not detected by infectious titer assay nor by qPCR, ensuring the safety of the virus in the dogs and their environment. CONCLUSIONS: These results establish the good tolerability of TG6002 in healthy dogs with undetectable viral shedding after multiple injections. This study supports the initiation of further studies in canine cancer patients to evaluate the oncolytic potential of TG6002 and provides critical data for clinical development of TG6002 as a human cancer therapy.


Assuntos
Produtos Biológicos/administração & dosagem , Vírus Oncolíticos/isolamento & purificação , Vaccinia virus/isolamento & purificação , Eliminação de Partículas Virais , Animais , Produtos Biológicos/efeitos adversos , Cães , Injeções Intramusculares/veterinária , Masculino , Terapia Viral Oncolítica
3.
Curr Gene Ther ; 18(6): 366-374, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30411681

RESUMO

INTRODUCTION: Oncolytic virus therapy is currently considered as a promising therapeutic approach for cancer treatment. Adenovirus is well-known and extensively characterized as an oncolytic agent. The increasing number of clinical trials using this virus generates the demand for the development of a well-established purification approach. Triton X-100 is commonly used in cell lysis buffer preparations. The addition of this surfactant in the list of substances with the very high concern of the Registration, Evaluation, Authorization and Restriction of Chemicals (REACH) regulation promoted the research for effective alternatives. METHODS: In this work, a purification strategy for oncolytic adenovirus compatible with phase I clinical trials, using an approved surfactant - Polysorbate 20 was developed. The proposed downstream train, composed by clarification, concentration using tangential flow filtration, intermediate purification with anion exchange chromatography, followed by a second concentration and a final polishing step was evaluated for both Triton X-100 and Polysorbate 20 processes. The impact of cell lysis with Polysorbate20 and Triton X-100 for each downstream step was evaluated in terms of product recovery and impurities removal. Overall, 61 ± 4% of infectious viral particles were recovered. Depletion of host cell proteins and ds-DNA was 99.9% and 97.1%, respectively. RESULTS & CONCLUSION: The results indicated that Polysorbate 20 can be used as a replacement for Triton X-100 during cell lysis with no impact on product recovery, potency, and purity. Moreover, the developed process is scalable and able to provide a highly purified product to be used in phase I and II clinical trials.


Assuntos
Adenoviridae/isolamento & purificação , Vírus Oncolíticos/isolamento & purificação , Polissorbatos , Células A549 , Adenoviridae/patogenicidade , Filtração/métodos , Humanos , Octoxinol , Vírus Oncolíticos/patogenicidade
4.
Virol J ; 15(1): 65, 2018 04 10.
Artigo em Inglês | MEDLINE | ID: mdl-29631630

RESUMO

BACKGROUND: Endometrial cancer (EC) is one of the most common gynecological malignancies globally. Although progress has been made in surgical and other adjuvant therapies, there is still a great need to develop new approaches to further reduce the incidence and mortality of EC. Oncolytic virotherapy offers a novel promising option of cancer treatment and has demonstrated good efficacy in preclinical models and clinical trials. However, only few oncolytic viruses have been tested for EC treatment. In this study, the potential of an oncolytic coxsackievirus B3 (CV-B3) strain 2035A (CV-B3/2035A) was investigated as a novel biotherapeutic agent against EC. METHODS: Human EC cell lines (Ishikawa, HEC-1-A and HEC-1-B) were infected with CV-B3/2035A, and viral replication and cytotoxic effects were evaluated in vitro. CV-B3/2035A-induced oncolysis was also investigated in nude mice bearing EC xenografts in vivo and in patient-derived EC samples ex vivo. RESULTS: Human EC cell lines expressing different levels of CAR and DAF were all susceptible to infection by CV-B3/2035A and supported efficient viral replication in vitro. In the EC xenograft/nude mouse model, both intratumoral and intravenous administrations of CV-B3-2035A exerted significant therapeutic effects against pre-established EC tumors without causing significant treatment-related toxicity and mortality in nude mice. Moreover, CV-B3/2035A treatment resulted in decreased viability of patient-derived EC samples ex vivo. CONCLUSIONS: CV-B3/2035A showed oncolytic activity in human EC cell lines both in vitro and in vivo as well as in patient-derived EC samples ex vivo and thus could be used as an alternative virotherapy agent for the treatment of EC.


Assuntos
Neoplasias do Endométrio/terapia , Neoplasias do Endométrio/virologia , Enterovirus/fisiologia , Terapia Viral Oncolítica , Vírus Oncolíticos/fisiologia , Animais , Linhagem Celular Tumoral , Neoplasias do Endométrio/patologia , Enterovirus/isolamento & purificação , Feminino , Humanos , Camundongos Endogâmicos BALB C , Camundongos Nus , Transplante de Neoplasias , Vírus Oncolíticos/isolamento & purificação , Receptores Virais/metabolismo , Replicação Viral , Ensaios Antitumorais Modelo de Xenoenxerto
6.
Vaccine ; 34(42): 5082-5089, 2016 09 30.
Artigo em Inglês | MEDLINE | ID: mdl-27614781

RESUMO

Vaccinia virus (VV) is an oncolytic virus that is currently being evaluated as a promising cancer vaccine in several phase I, II and III clinical trials. Although several quality control tests are performed on each new batch of virus, these do not routinely include a systematic characterization of virus particle homogeneity, or relate the infectious titer to the total number of submicron sized particles (SSPs) present in the sample. SSPs are comprised of infectious virus and non-infectious viral particles, but also cell contaminants derived from the virus isolation procedures, such as cellular vesicles and debris. Here we have employed flow virometry (FV) analysis and sorting to isolate and characterize distinct SSP populations in therapeutic oncolytic VV preparations. We show that VV preparations contain SSPs heterogeneous in size and include large numbers of non-infectious VV particles. Furthermore, we used FV to illustrate how VV has a propensity to aggregate over time and under various handling and storage procedures. Accordingly, we find that together the infectious titer, the total number of SSPs, the number of viral genomes and the level of particle aggregation in a sample constitute useful parameters that greatly facilitate inter-sample assessment of physical quality, and also provides a means to monitor sample deterioration over time. Additionally, we have successfully employed FV sorting to further isolate virus from other particles by identifying a lipophilic dye that preferentially stains VV over other SSPs in the sample. Overall, we demonstrate that FV is a fast and effective tool that can be used to perform quality, and consistency control assessments of oncolytic VV vaccine preparations.


Assuntos
Citometria de Fluxo/métodos , Vírus Oncolíticos , Vaccinia virus , Vírion/isolamento & purificação , Vesículas Extracelulares , Humanos , Terapia Viral Oncolítica , Vírus Oncolíticos/isolamento & purificação , Vaccinia virus/isolamento & purificação , Replicação Viral
7.
J Gen Virol ; 97(9): 2166-2179, 2016 09.
Artigo em Inglês | MEDLINE | ID: mdl-27417501

RESUMO

Pancreatic ductal adenocarcinoma (PDA) is one of the leading causes of cancer-related deaths worldwide and the development of new treatment strategies for PDA patients is of crucial importance. Virotherapy uses natural or engineered oncolytic viruses (OVs) to selectively kill tumour cells. Due to their genetic heterogeneity, PDA cells are highly variable in their permissiveness to various OVs. The avian influenza A virus (IAV) H7N3 A/turkey/Italy/2962/03 is a potent inducer of apoptosis in PDA cells previously shown to be resistant to other OVs (Kasloff et al., 2014), suggesting that it might be effective against specific subclasses of pancreatic cancer. To improve the selectivity of the avian influenza isolate for PDA cells, here confirmed deficient for IFN response, we engineered a truncation in the NS1 gene that is the major virus-encoded IFN antagonist. The recombinant virus (NS1-77) replicated efficiently in PDA cells, but was attenuated in non-malignant pancreatic ductal cells, in which it induced a potent IFN response that acted upon bystander uninfected cancer cells, triggering their death. The engineered virus displayed an enhanced ability to debulk a PDA-derived tumour in xenograft mouse model. Our results highlight the possibility of selecting an IAV strain from the diverse natural avian reservoir on the basis of its inherent oncolytic potency in specific PDA subclasses and, through engineering, improve its safety, selectivity and debulking activity for cancer treatment.


Assuntos
Adenocarcinoma/terapia , Carcinoma Ductal Pancreático/terapia , Vírus da Influenza A Subtipo H7N3/isolamento & purificação , Terapia Viral Oncolítica/métodos , Vírus Oncolíticos/isolamento & purificação , Animais , Linhagem Celular Tumoral , Modelos Animais de Doenças , Xenoenxertos , Humanos , Fatores Imunológicos/genética , Fatores Imunológicos/metabolismo , Vírus da Influenza A Subtipo H7N3/genética , Vírus da Influenza A Subtipo H7N3/fisiologia , Camundongos , Camundongos SCID , Vírus Oncolíticos/genética , Deleção de Sequência , Resultado do Tratamento , Proteínas não Estruturais Virais/genética , Proteínas não Estruturais Virais/metabolismo , Replicação Viral
8.
Viruses ; 8(6)2016 06 14.
Artigo em Inglês | MEDLINE | ID: mdl-27314377

RESUMO

Oncolytic adenoviruses (Ads) have been shown to be safe and have great potential for the treatment of solid tumors. However, the therapeutic efficacy of Ads is antagonized by limited spread within solid tumors. To develop Ads with enhanced spread, viral particles of an E1-wildtype Ad5 dl309 was repeatedly treated with UV type C irradiation and selected for the efficient replication and release from cancer cells. After 72 cycles of treatment and cancer selection, AdUV was isolated. This vector has displayed many favorable characteristics for oncolytic therapy. AdUV was shown to lyse cancer cells more effectively than both E1-deleted and E1-wildtype Ads. This enhanced cancer cell lysis appeared to be related to increased AdUV replication in and release from infected cancer cells. AdUV-treated A549 cells displayed greater expression of the autophagy marker LC3-II during oncolysis and formed larger viral plaques upon cancer cell monolayers, indicating increased virus spread among cancer cells. This study indicates the potential of this approach of irradiation of entire viral particles for the development of oncolytic viruses with designated therapeutic properties.


Assuntos
Adenoviridae/crescimento & desenvolvimento , Adenoviridae/isolamento & purificação , Vírus Oncolíticos/crescimento & desenvolvimento , Vírus Oncolíticos/isolamento & purificação , Inoculações Seriadas , Raios Ultravioleta , Adenoviridae/genética , Adenoviridae/efeitos da radiação , Linhagem Celular Tumoral , Sobrevivência Celular , Humanos , Vírus Oncolíticos/genética , Vírus Oncolíticos/efeitos da radiação , Ensaio de Placa Viral
9.
Viruses ; 8(3): 72, 2016 Mar 07.
Artigo em Inglês | MEDLINE | ID: mdl-26959050

RESUMO

The development of oncolytic viruses has led to an emerging new class of cancer therapeutics. Although the safety profile has been encouraging, the transition of oncolytic viruses to the clinical setting has been a slow process due to modifications. Therefore, a new generation of more potent oncolytic viruses needs to be exploited, following our better understanding of the complex interactions between the tumor, its microenvironment, the virus, and the host immune response. The conventional method for creation of tumor-targeted oncolytic viruses is based on homologous recombination. However, the creation of new mutant oncolytic viruses with large genomes remains a challenge due to the multi-step process and low efficiency of homologous recombination. The CRISPR-associated endonuclease Cas9 has hugely advanced the potential to edit the genomes of various organisms due to the ability of Cas9 to target a specific genomic site by a single guide RNA. In this review, we discuss the CRISPR-Cas9 system as an efficient viral editing method for the creation of new oncolytic viruses, as well as its potential future applications in the development of oncolytic viruses. Further, this review discusses the potential of off-target effects as well as CRISPR-Cas9 as a tool for basic research into viral biology.


Assuntos
Sistemas CRISPR-Cas , Biologia Molecular/métodos , Vírus Oncolíticos/genética , Vírus Oncolíticos/isolamento & purificação , Recombinação Genética , Virologia/métodos
10.
Viruses ; 7(12): 6251-78, 2015 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-26633466

RESUMO

Viruses that specifically replicate in tumor over normal cells offer promising cancer therapies. Oncolytic viruses (OV) not only kill the tumor cells directly; they also promote anti-tumor immunotherapeutic responses. Other major advantages of OVs are that they dose-escalate in tumors and can be genetically engineered to enhance potency and specificity. Unmodified wild type reovirus is a propitious OV currently in phase I-III clinical trials. This review summarizes modifications to reovirus that may improve potency and/or specificity during oncolysis. Classical genetics approaches have revealed reovirus variants with improved adaptation towards tumors or with enhanced ability to establish specific steps of virus replication and cell killing among transformed cells. The recent emergence of a reverse genetics system for reovirus has provided novel strategies to fine-tune reovirus proteins or introduce exogenous genes that could promote oncolytic activity. Over the next decade, these findings are likely to generate better-optimized second-generation reovirus vectors and improve the efficacy of oncolytic reotherapy.


Assuntos
Descoberta de Drogas/métodos , Terapia Viral Oncolítica/métodos , Vírus Oncolíticos/isolamento & purificação , Vírus Oncolíticos/fisiologia , Reoviridae/isolamento & purificação , Reoviridae/fisiologia , Ensaios Clínicos como Assunto
11.
Biotechnol Prog ; 31(6): 1600-12, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26289142

RESUMO

Anchorage-dependent cell cultures are used for the production of viruses, viral vectors, and vaccines, as well as for various cell therapies and tissue engineering applications. Most of these applications currently rely on planar technologies for the generation of biological products. However, as new cell therapy product candidates move from clinical trials towards potential commercialization, planar platforms have proven to be inadequate to meet large-scale manufacturing demand. Therefore, a new scalable platform for culturing anchorage-dependent cells at high cell volumetric concentrations is urgently needed. One promising solution is to grow cells on microcarriers suspended in single-use bioreactors. Toward this goal, a novel bioreactor system utilizing an innovative Vertical-Wheel™ technology was evaluated for its potential to support scalable cell culture process development. Two anchorage-dependent human cell types were used: human lung carcinoma cells (A549 cell line) and human bone marrow-derived mesenchymal stem cells (hMSC). Key hydrodynamic parameters such as power input, mixing time, Kolmogorov length scale, and shear stress were estimated. The performance of Vertical-Wheel bioreactors (PBS-VW) was then evaluated for A549 cell growth and oncolytic adenovirus type 5 production as well as for hMSC expansion. Regarding the first cell model, higher cell growth and number of infectious viruses per cell were achieved when compared with stirred tank (ST) bioreactors. For the hMSC model, although higher percentages of proliferative cells could be reached in the PBS-VW compared with ST bioreactors, no significant differences in the cell volumetric concentration and expansion factor were observed. Noteworthy, the hMSC population generated in the PBS-VW showed a significantly lower percentage of apoptotic cells as well as reduced levels of HLA-DR positive cells. Overall, these results showed that process transfer from ST bioreactor to PBS-VW, and scale-up was successfully carried out for two different microcarrier-based cell cultures. Ultimately, the data herein generated demonstrate the potential of Vertical-Wheel bioreactors as a new scalable biomanufacturing platform for microcarrier-based cell cultures of complex biopharmaceuticals.


Assuntos
Adenoviridae/fisiologia , Reatores Biológicos , Técnicas de Cultura de Células/instrumentação , Células-Tronco Mesenquimais/citologia , Vírus Oncolíticos/fisiologia , Adenoviridae/isolamento & purificação , Técnicas de Cultura de Células/métodos , Linhagem Celular Tumoral , Desenho de Equipamento , Humanos , Hidrodinâmica , Vírus Oncolíticos/isolamento & purificação
12.
BMB Rep ; 48(8): 454-60, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-26058397

RESUMO

Naturally occurring reoviruses are live replication-proficient viruses that specifically infect human cancer cells while sparing their normal counterpart. Since the discovery of reoviruses in 1950s, they have shown various degrees of safety and efficacy in pre-clinical or clinical applications for human anti-cancer therapeutics. I have recently discovered that cellular tumor suppressor genes are also important in determining reoviral tropism. Carcinogenesis is a multi-step process involving the accumulation of both oncogene and tumor suppressor gene abnormalities. Reoviruses can exploit abnormal cellular tumor suppressor signaling for their oncolytic specificity and efficacy. Many tumor suppressor genes such as p53, ataxia telangiectasia mutated (ATM), and retinoblastoma associated (RB) are known to play important roles in genomic fidelity/maintenance. Thus, a tumor suppressor gene abnormality could affect host genomic integrity and likely disrupt intact antiviral networks due to the accumulation of genetic defects which in turn could result in oncolytic reovirus susceptibility. This review outlines the discovery of oncolytic reovirus strains, recent progresses in elucidating the molecular connection between oncogene/tumor suppressor gene abnormalities and reoviral oncotropism, and their clinical implications. Future directions in the utility of reovirus virotherapy is also proposed in this review.


Assuntos
Neoplasias/terapia , Neoplasias/virologia , Terapia Viral Oncolítica/métodos , Vírus Oncolíticos/fisiologia , Reoviridae/fisiologia , Humanos , Terapia Viral Oncolítica/tendências , Vírus Oncolíticos/isolamento & purificação
13.
Virol J ; 11: 91, 2014 May 16.
Artigo em Inglês | MEDLINE | ID: mdl-24886301

RESUMO

BACKGROUND: Newcastle disease virus (NDV), a single-stranded RNA virus of the family Paramyxoviridae, is a candidate virotherapy agent in cancer treatment. Promising responses were observed in clinical studies. Despite its high potential, the possibility of the virus to develop a persistent form of infection in cancer cells has not been investigated. Occurrence of persistent infection by NDV in cancer cells may cause the cells to be less susceptible to the virus killing. This would give rise to a population of cancer cells that remains viable and resistant to treatment. RESULTS: During infection experiment in a series of colorectal cancer cell lines, we adventitiously observed a development of persistent infection by NDV in SW480 cells, but not in other cell lines tested. This cell population, designated as SW480P, showed resistancy towards NDV killing in a re-infection experiment. The SW480P cells retained NDV genome and produced virus progeny with reduced plaque forming ability. CONCLUSION: These observations showed that NDV could develop persistent infection in cancer cells and this factor needs to be taken into consideration when using NDV in clinical settings.


Assuntos
Vírus da Doença de Newcastle/crescimento & desenvolvimento , Vírus Oncolíticos/crescimento & desenvolvimento , Linhagem Celular Tumoral , Neoplasias Colorretais , Humanos , Vírus da Doença de Newcastle/isolamento & purificação , Vírus Oncolíticos/isolamento & purificação
14.
Hum Gene Ther ; 24(12): 1029-41, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24099555

RESUMO

Computed tomography (CT) is the most commonly used radiological response evaluation method in contemporary oncology. However, it may not be optimally suitable for assessment of oncolytic virus treatments because of paradoxical inflammatory tumor swellings, which result from virus treatments, particularly when viruses are armed with immunostimulatory molecules. Here we investigated the prognostic utility of CT and [(18)F]-fluorodeoxyglucose (FDG) positron emission tomography (PET) in oncolytic virus treatments. We also investigated possible appearance of false-positive FDG signals in FDG-PET imaging of humans and hamsters treated with oncolytic adenoviruses. First, immunocompetent Syrian hamsters were treated with intratumoral adenovirus injections, tumor growth was followed up, and [(18)F]-FDG-uptake was quantitated with small animal PET/CT. Second, we describe a retrospective patient series, essentially 17 individual case reports, of advanced cancer patients treated with oncolytic adenoviruses in the context of an Advanced Therapy Access Program (ATAP) who underwent radiological response evaluation with both contrast-enhanced CT and FDG-PET. Third, we collected a retrospective case series of radiological response and survival data of 182 patients treated with oncolytic adenoviruses in ATAP to evaluate the prognostic reliability of CT and FDG-PET. Overall, responses in CT and FDG-PET correlated well with each other and were equally reliable as prognostic markers for long survival after oncolytic adenovirus treatment. Interestingly, we observed that new FDG-avid lymph nodes appearing in FDG-PET after virus treatments may represent inflammatory responses and therefore should not be interpreted as treatment failure in the absence of other signs or verification of disease progression. We also observed indications that FDG-PET might be more sensitive in detection of responses than tumor size.


Assuntos
Adenoviridae/isolamento & purificação , Terapia Genética , Neoplasias/terapia , Vírus Oncolíticos/isolamento & purificação , Adenoviridae/química , Animais , Cricetinae , Fluordesoxiglucose F18/química , Humanos , Neoplasias/genética , Vírus Oncolíticos/química , Vírus Oncolíticos/genética , Tomografia por Emissão de Pósitrons , Compostos Radiofarmacêuticos/química , Tomografia Computadorizada de Emissão
15.
Curr Protoc Hum Genet ; Chapter 12: Unit 12.14, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23853080

RESUMO

Oncolytic adenoviruses are designed to take advantage of the virus' native ability to replicate in cancer cells to induce oncolysis. Subsequently, the released viral progeny spread and kill the neighboring cancer cells. These characteristics, together with the ability of adenovirus to infect a broad spectrum of cells, its well understood replication machinery, and relative ease of manufacture have led to the intensive use of adenovirus as an anticancer agent. This unit describes cloning strategies, procedures to turn the intended design into virus, and quality analyses of resultant adenoviral vectors. Most of these procedures were optimized especially for oncolytic adenoviral vectors.


Assuntos
Adenovírus Humanos/genética , Genoma Viral , Terapia Viral Oncolítica/métodos , Vírus Oncolíticos/genética , Adenovírus Humanos/isolamento & purificação , Linhagem Celular Transformada , Linhagem Celular Tumoral , Vetores Genéticos/genética , Células HEK293 , Humanos , Mutação , Neoplasias/patologia , Neoplasias/terapia , Neoplasias/virologia , Vírus Oncolíticos/isolamento & purificação , Regiões Promotoras Genéticas/genética , Transfecção , Vírion/genética , Replicação Viral
16.
Gene Ther ; 20(1): 102-11, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-22357510

RESUMO

Newcastle disease virus (NDV) is an oncolytic paramyxovirus with a nonsegmented single-stranded RNA genome. In this report, a recombinant oncolytic NDV was passaged in human tumor xenografts and reisolated and characterized after two rounds of bioselection. Several isolates could be recovered that differed from the parental virus with respect to virus spread in tumor cells and the ability to form syncytia in human tumor cells. Three isolates were identified that demonstrated superior oncolytic potency compared with the parental virus as measured by increased oncolytic potency in confluent tumor cell monolayers, in tumor cell spheroids and in a mouse xenograft tumor model. The surface proteins F and HN were sequence analyzed and characterized for fusogenicity. The present study demonstrates that in vivo NDV bioselection can enable the isolation of novel, oncolytic NDV and thus represents a powerful methodology for the development of highly potent oncolytic viruses.


Assuntos
Vírus da Doença de Newcastle/genética , Vírus Oncolíticos/genética , Seleção Genética , Animais , Linhagem Celular Tumoral , Células Gigantes , Hemaglutininas/genética , Humanos , Camundongos , Camundongos Nus , Transplante de Neoplasias , Vírus da Doença de Newcastle/isolamento & purificação , Vírus Oncolíticos/isolamento & purificação , Transplante Heterólogo , Proteínas Virais de Fusão/genética
17.
J Gen Virol ; 94(Pt 4): 726-737, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23239570

RESUMO

RH2 is a novel oncolytic herpes simplex virus type 1 (HSV-1) produced by simultaneous infection with neurovirulent γ134.5 gene-deficient HSV-1 R849 derived from strain F and the spontaneously occurring, fusogenic HSV-1 HF in cell culture. The genome of RH2 was studied using Genome Sequencer FLX. RH2 comprised 149 64 bp and it was shown that the lacZ gene was inserted into the γ134.5 gene of R849. Comparison of ORFs revealed that RH2 had 100 % identity with strain F in 21/58 unique long (UL) genes (36.2%) and 1/13 unique short (US) genes (7.7%). RH2 had 100% amino acid identity with HF10 in 24/58 UL genes (41.4%) and 9/13 US genes (69.2%). Twelve genes, including UL27 (gB), US4 (gG) and UL6 (gD), had amino acid changes unique to RH2. Amino acid changes in gB occurred at positions 459 (T→A) and 817 (L→P). Other unique features were the amino acids missing in UL36 (VP1/2) and UL46 (VP11/12). Thus, RH2 is an HF10-based vector preserving the fusogenic amino acid changes of gB but lacking the γ134.5 gene. RH2 is expected to be a version of HF10 useful for the treatment of brain tumours as well as oral squamous cell carcinoma. Spontaneously occurring HSV-1 mutants may also be useful clinically, as their genome sequences can easily be determined by this genome sequencing system.


Assuntos
DNA Viral/química , DNA Viral/genética , Genoma Viral , Herpesvirus Humano 1/genética , Vírus Oncolíticos/genética , Substituição de Aminoácidos , Genes Virais , Herpesvirus Humano 1/isolamento & purificação , Humanos , Dados de Sequência Molecular , Vírus Oncolíticos/isolamento & purificação , Análise de Sequência de DNA , Homologia de Sequência de Aminoácidos
18.
Anal Chem ; 84(5): 2548-56, 2012 Mar 06.
Artigo em Inglês | MEDLINE | ID: mdl-22324738

RESUMO

DNA aptamers are promising immunoshielding agents that could protect oncolytic viruses (OVs) from neutralizing antibodies (nAbs) and increase the efficiency of cancer treatment. In the present Article, we introduce a novel technology for electrochemical differentiation of epitope-specific aptamers (eDEA) without selecting aptamers against individual antigenic determinants. For this purpose, we selected DNA aptamers that can bind noncovalently to an intact oncolytic virus, vaccinia virus (VACV), which can selectively replicate in and kill only tumor cells. The aptamers were integrated as a recognition element into a multifunctional electrochemical aptasensor. The developed aptasensor was used for the linear quantification of the virus in the range of 500-3000 virus particles with a detection limit of 330 virions. Also, the aptasensor was employed to compare the binding affinities of aptamers to VACV and to estimate the degree of protection of VACV using the anti-L1R neutralizing antibody in a displacement assay fashion. Three anti-VACV aptamer clones, vac2, vac4, and vac6, showed the best immunoprotection results and can be applied for enhanced delivery of VACV. Another two sequences, vac5 and vac46, exhibited high affinities to VACV without shielding it from nAb and can be further utilized in sandwich bioassays.


Assuntos
Aptâmeros de Nucleotídeos/metabolismo , Técnicas Eletroquímicas , Epitopos/metabolismo , Anticorpos Neutralizantes/imunologia , Aptâmeros de Nucleotídeos/imunologia , Epitopos/imunologia , Corantes Fluorescentes/química , Humanos , Vírus Oncolíticos/imunologia , Vírus Oncolíticos/isolamento & purificação , Vaccinia virus/imunologia , Vaccinia virus/isolamento & purificação
19.
Methods Mol Biol ; 797: 1-19, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-21948465

RESUMO

Herpes simplex virus-1 (HSV-1) is an enveloped, double-stranded DNA virus that has been used with modification as an oncolytic virus against a number of tumor types. Modifications that make HSV-1 replication--conditional, i.e., selectively divide in replicating cells make it fulfill a prerequisite criteria for oncolytic viruses. Other appealing features of HSV-1 as an oncolytic virus include its large, modifiable genome; its sensitivity to antiviral agents, such as ganciclovir; and its lack of host cell integration. Here, we review the methods of HSV-1 engineering, through traditional recombination techniques as well as through bacterial artificial chromosome (BAC) technology. We then describe protocols for titering, amplification, and purification of engineered HSV-1-derived oncolytic viruses.


Assuntos
Engenharia Genética/métodos , Herpesvirus Humano 1/genética , Terapia Viral Oncolítica , Vírus Oncolíticos/genética , Replicação Viral/genética , Antivirais/farmacologia , Cromossomos Artificiais Bacterianos/genética , Ganciclovir/farmacologia , Vetores Genéticos , Herpesvirus Humano 1/efeitos dos fármacos , Herpesvirus Humano 1/isolamento & purificação , Humanos , Vírus Oncolíticos/efeitos dos fármacos , Vírus Oncolíticos/isolamento & purificação , Recombinação Genética/genética
20.
Methods Mol Biol ; 797: 35-52, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-21948467

RESUMO

Oncolytic (replication-competent) adenoviruses (Ads) represent the most advanced platform for cancer gene therapy. These viral vectors ablate tumors by killing tumor cells in the process of virus replication. As progeny virions are released, they infect remaining cancer cells, generating a bystander effect. Ads engineered for increased cancer specificity produce less damage to normal tissues. First-generation oncolytic Ads have demonstrated acceptable levels of safety while the efficacy was observed only in combination with chemotherapy and/or radiation. Second-generation oncolytic Ads are armed with therapeutic transgenes to increase release, spread, and bystander effect for enhancing the efficacy. Third-generation oncolytic Ads are armed vectors with capsid modifications for transductional detargeting from normal tissues and targeting to cancer cells. Chemical modification of the capsid additionally improves therapeutic window. Here, we describe methods for generation and characterization of advanced-generation oncolytic Ads.


Assuntos
Adenoviridae/química , Adenoviridae/genética , Terapia Genética , Neoplasias/terapia , Terapia Viral Oncolítica , Vírus Oncolíticos/química , Vírus Oncolíticos/genética , Adenoviridae/isolamento & purificação , Animais , Capsídeo/química , Vetores Genéticos , Humanos , Camundongos , Camundongos Nus , Vírus Oncolíticos/isolamento & purificação
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...