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.
Mol Ther Oncolytics ; 21: 340-355, 2021 Jun 25.
Artigo em Inglês | MEDLINE | ID: mdl-34141871

RESUMO

Advanced pancreatic cancer is characterized by few treatment options and poor outcomes. Oncolytic virotherapy and chemotherapy involve complementary pharmacodynamics and could synergize to improve therapeutic efficacy. Likewise, multimodality treatment may cause additional toxicity, and new agents have to be safe. Balancing both aims, we generated an oncolytic measles virus for 5-fluorouracil-based chemovirotherapy of pancreatic cancer with enhanced tumor specificity through microRNA-regulated vector tropism. The resulting vector encodes a bacterial prodrug convertase, cytosine deaminase-uracil phosphoribosyl transferase, and carries synthetic miR-148a target sites in the viral F gene. Combination of the armed and targeted virus with 5-fluorocytosine, a prodrug of 5-fluorouracil, resulted in cytotoxicity toward both infected and bystander pancreatic cancer cells. In pancreatic cancer xenografts, a single intratumoral injection of the virus induced robust in vivo expression of prodrug convertase. Based on intratumoral transgene expression kinetics, we devised a chemovirotherapy regimen to assess treatment efficacy. Concerted multimodality treatment with intratumoral virus and systemic prodrug administration delayed tumor growth and prolonged survival of xenograft-bearing mice. Our results demonstrate that 5-fluorouracil-based chemovirotherapy with microRNA-sensitive measles virus is an effective strategy against pancreatic cancer at a favorable therapeutic index that warrants future clinical translation.

2.
J Gen Virol ; 101(4): 399-409, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-32053093

RESUMO

Oncolytic virotherapy is an emerging treatment option for numerous cancers, with several virus families currently being evaluated in clinical trials. More specifically, vaccine-strain measles virus has arisen as a promising candidate for the treatment of different tumour types in several early clinical trials. Replicating viruses, and especially RNA viruses without proofreading polymerases, can rapidly adapt to varying environments by selecting quasispecies with advantageous genetic mutations. Subsequently, these genetic alterations could potentially weaken the safety profile of virotherapy. In this study, we demonstrate that, following an extended period of virus replication in producer or cancer cell lines, the quasispecies consensus sequence of vaccine strain-derived measles virus accrues a remarkably small number of mutations throughout the nonsegmented negative-stranded RNA genome. Interestingly, we detected a nonrandom distribution of genetic alterations within the genome, with an overall decreasing frequency of mutations from the 3' genome start to its 5' end. Comparing the serially passaged viruses to the parental virus on producer cells, we found that the acquired consensus mutations did not drastically change viral replication kinetics or cytolytic potency. Collectively, our data corroborate the genomic stability and excellent safety profile of oncolytic measles virus, thus supporting its continued development and clinical translation as a promising viro-immunotherapeutic.


Assuntos
Instabilidade Genômica , Vírus do Sarampo/genética , Quase-Espécies/genética , Animais , Linhagem Celular Tumoral , Sobrevivência Celular , Chlorocebus aethiops , Humanos , Vírus do Sarampo/crescimento & desenvolvimento , Mutação , Terapia Viral Oncolítica , Inoculações Seriadas , Células Vero , Virulência/genética
3.
Mol Ther Oncolytics ; 9: 30-40, 2018 Jun 29.
Artigo em Inglês | MEDLINE | ID: mdl-29988512

RESUMO

Measles viruses derived from the live-attenuated Edmonton-B vaccine lineage are currently investigated as novel anti-cancer therapeutics. In this context, tumor specificity and oncolytic potency are key determinants of the therapeutic index. Here, we describe a systematic and comprehensive analysis of a recently developed post-entry targeting strategy based on the incorporation of microRNA target sites (miRTS) into the measles virus genome. We have established viruses with target sites for different microRNA species in the 3' untranslated regions of either the N, F, H, or L genes and generated viruses harboring microRNA target sites in multiple genes. We report critical importance of target-site positioning with proximal genomic positions effecting maximum vector control. No relevant additional effect of six versus three miRTS copies for the same microRNA species in terms of regulatory efficiency was observed. Moreover, we demonstrate that, depending on the microRNA species, viral mRNAs containing microRNA target sites are directly cleaved and/or translationally repressed in presence of cognate microRNAs. In conclusion, we report highly efficient control of measles virus replication with various miRTS positions for development of safe and efficient cancer virotherapy and provide insights into the mechanisms underlying microRNA-mediated vector control.

4.
J Gen Virol ; 98(9): 2248-2257, 2017 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-28809150

RESUMO

Viruses from the diverse family of Paramyxoviridae include important pathogens and are applied in gene therapy and for cancer treatment. The Tupaia paramyxovirus (TPMV), isolated from the kidney of a tree shrew, does not infect human cells and neutralizing antibodies against other Paramyxoviridae do not cross-react with TPMV. Here, we present a vector system for de novo generation of infectious TPMV that allows for insertion of additional genes as well as targeting using antibody single-chain variable fragments. We show that the recombinant TPMV specifically infect cells expressing the targeted receptor and replicate in human cells. This vector system provides a valuable tool for both basic research and therapeutic applications.


Assuntos
Técnicas de Transferência de Genes , Vetores Genéticos/genética , Paramyxoviridae/genética , Animais , Linhagem Celular , Vetores Genéticos/fisiologia , Humanos , Paramyxoviridae/fisiologia , Transgenes , Tupaia/virologia
5.
Mol Ther Methods Clin Dev ; 3: 16018, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27088104

RESUMO

Oncolytic viruses (OVs) are unique anticancer agents based on their pleotropic modes of action, which include, besides viral tumor cell lysis, activation of antitumor immunity. A panel of diverse viruses, often genetically engineered, has advanced to clinical investigation, including phase 3 studies. This diversity of virotherapeutics not only offers interesting opportunities for the implementation of different therapeutic regimens but also poses challenges for clinical translation. Thus, manufacturing processes and regulatory approval paths need to be established for each OV individually. This review provides an overview of clinical-grade manufacturing procedures for OVs using six virus families as examples, and key challenges are discussed individually. For example, different virus features with respect to particle size, presence/absence of an envelope, and host species imply specific requirements for measures to ensure sterility, for handling, and for determination of appropriate animal models for toxicity testing, respectively. On the other hand, optimization of serum-free culture conditions, increasing virus yields, development of scalable purification strategies, and formulations guaranteeing long-term stability are challenges common to several if not all OVs. In light of the recent marketing approval of the first OV in the Western world, strategies for further upscaling OV manufacturing and optimizing product characterization will receive increasing attention.

6.
Mol Ther ; 22(11): 1949-59, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25156126

RESUMO

We hypothesized that the combination of oncolytic virotherapy with immune checkpoint modulators would reduce tumor burden by direct cell lysis and stimulate antitumor immunity. In this study, we have generated attenuated Measles virus (MV) vectors encoding antibodies against CTLA-4 and PD-L1 (MV-aCTLA-4 and MV-aPD-L1). We characterized the vectors in terms of growth kinetics, antibody expression, and cytotoxicity in vitro. Immunotherapeutic effects were assessed in a newly established, fully immunocompetent murine model of malignant melanoma, B16-CD20. Analyses of tumor-infiltrating lymphocytes and restimulation experiments indicated a favorable immune profile after MV-mediated checkpoint modulation. Therapeutic benefits in terms of delayed tumor progression and prolonged median overall survival were observed for animals treated with vectors encoding anti-CTLA-4 and anti-PD-L1, respectively. Combining systemic administration of antibodies with MV treatment also improved therapeutic outcome. In vivo oncolytic efficacy against human tumors was studied in melanoma xenografts. MV-aCTLA-4 and MV-aPD-L1 were equally efficient as parental MV in this model, with high rates of complete tumor remission (> 80%). Furthermore, we could demonstrate lysis of tumor cells and transgene expression in primary tissue from melanoma patients. The current results suggest rapid translation of combining immune checkpoint modulation with oncolytic viruses into clinical application.


Assuntos
Antígeno B7-H1/metabolismo , Antígeno CTLA-4/metabolismo , Linfócitos do Interstício Tumoral/metabolismo , Melanoma Experimental/terapia , Vírus Oncolíticos/imunologia , Animais , Vetores Genéticos/administração & dosagem , Vírus do Sarampo/genética , Vírus do Sarampo/imunologia , Vírus do Sarampo/metabolismo , Melanoma Experimental/imunologia , Camundongos , Terapia Viral Oncolítica , Vírus Oncolíticos/genética , Vírus Oncolíticos/metabolismo , Resultado do Tratamento
7.
PLoS One ; 9(3): e90508, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24598703

RESUMO

Multiple types of oncolytic viruses are currently under investigation in clinical trials. To optimize therapeutic outcomes it is believed that the plethora of different tumor types will require a diversity of different virus types. Sendai virus (SeV), a murine parainfluenza virus, displays a broad host range, enters cells within minutes and already has been applied safely as a gene transfer vector in gene therapy patients. However, SeV spreading naturally is abrogated in human cells due to a lack of virus activating proteases. To enable oncolytic applications of SeV we here engineered a set of novel recombinant vectors by a two-step approach: (i) introduction of an ubiquitously recognized cleavage-motive into SeV fusion protein now enabling continuous spreading in human tissues, and (ii) profound attenuation of these rSeV by the knockout of viral immune modulating accessory proteins. When employing human hepatoma cell lines, newly generated SeV variants now reached high titers and induced a profound tumor cell lysis. In contrast, virus release from untransformed human fibroblasts or primary human hepatocytes was found to be reduced by about three log steps in a time course experiment which enables the cumulation of kinetic differences of the distinct phases of viral replication such as primary target cell infection, target cell replication, and progeny virus particle release. In a hepatoma xenograft animal model we found a tumor-specific spreading of our novel recombinant SeV vectors without evidence of biodistribution into non-malignant tissues. In conclusion, we successfully developed novel tumor-selective oncolytic rSeV vectors, constituting a new tool for virotherapy of solid tumors being ready for further preclinical and clinical development to address distinct tumor types.


Assuntos
Neoplasias/terapia , Terapia Viral Oncolítica , Vírus Sendai/genética , Sequência de Aminoácidos , Animais , Sequência de Bases , Linhagem Celular Tumoral , Sobrevivência Celular , Chlorocebus aethiops , Vetores Genéticos , Humanos , Camundongos Endogâmicos BALB C , Camundongos Nus , Proteínas de Neoplasias/metabolismo , Transplante de Neoplasias , Neoplasias/enzimologia , Vírus Oncolíticos/genética , Vírus Oncolíticos/fisiologia , Peptídeo Hidrolases/metabolismo , Engenharia de Proteínas , Proteólise , Vírus Sendai/fisiologia , Células Vero , Carga Viral , Proteínas Virais/metabolismo , Replicação Viral
8.
Proc Natl Acad Sci U S A ; 111(5): E554-62, 2014 Feb 04.
Artigo em Inglês | MEDLINE | ID: mdl-24449891

RESUMO

Aptazymes are small, ligand-dependent self-cleaving ribozymes that function independently of transcription factors and can be customized for induction by various small molecules. Here, we introduce these artificial riboswitches for regulation of DNA and RNA viruses. We hypothesize that they represent universally applicable tools for studying viral gene functions and for applications as a safety switch for oncolytic and live vaccine viruses. Our study shows that the insertion of artificial aptazymes into the adenoviral immediate early gene E1A enables small-molecule-triggered, dose-dependent inhibition of gene expression. Aptazyme-mediated shutdown of E1A expression translates into inhibition of adenoviral genome replication, infectious particle production, and cytotoxicity/oncolysis. These results provide proof of concept for the aptazyme approach for effective control of biological outcomes in eukaryotic systems, specifically in virus infections. Importantly, we also demonstrate aptazyme-dependent regulation of measles virus fusion protein expression, translating into potent reduction of progeny infectivity and virus spread. This not only establishes functionality of aptazymes in fully cytoplasmic genetic systems, but also implicates general feasibility of this strategy for application in viruses with either DNA or RNA genomes. Our study implies that gene regulation by artificial riboswitches may be an appealing alternative to Tet- and other protein-dependent gene regulation systems, based on their small size, RNA-intrinsic mode of action, and flexibility of the inducing molecule. Future applications range from gene analysis in basic research to medicine, for example as a safety switch for new generations of efficiency-enhanced oncolytic viruses.


Assuntos
Vírus de DNA/genética , Vírus de DNA/fisiologia , Regulação Viral da Expressão Gênica , Vírus de RNA/genética , Vírus de RNA/fisiologia , Riboswitch/genética , Replicação Viral/genética , Adenoviridae/genética , Adenoviridae/patogenicidade , Adenoviridae/fisiologia , Proteínas E1A de Adenovirus/genética , Proteínas E1A de Adenovirus/metabolismo , Linhagem Celular , Vírus de DNA/patogenicidade , Genes Virais/genética , Ligantes , Vírus do Sarampo/genética , Vírus do Sarampo/patogenicidade , Vírus do Sarampo/fisiologia , Vírus Oncolíticos/genética , Vírus Oncolíticos/fisiologia , Vírus de RNA/patogenicidade , RNA Catalítico/metabolismo , Vírion/fisiologia , Internalização do Vírus
9.
Int J Cancer ; 134(1): 235-43, 2014 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-23797800

RESUMO

Therapy-induced senescence (TIS) as a permanent growth arrest can be induced by various stimuli, including anticancer compounds. TIS emerged as a promising strategy to overcome resistance phenomena. However, senescent cancer cells might regain proliferation activity in vivo or even secrete tumor-promoting cytokines. Therefore, successful exploitation of TIS in cancer treatment simultaneously requires the development of effective strategies to eliminate senescent cancer cells. Virotherapy aims to selectively hit tumor cells, thus a combination with senescence-inducing drugs was explored. As a model, we chose measles vaccine virus (MeV), which does not interfere with cellular senescence by itself. In different tumor cell types, such as hepatoma, pancreatic and mammary gland carcinoma, we demonstrate efficient viral replication and lysis after TIS by gemcitabine, doxorubicin or taxol. Applying real time imaging, we even found an accelerated lysis of senescent cancer cells, supporting an enhanced viral replication with an increase in cell-associated and released infectious MeV particles. In summary, we show as a proof-of-concept that senescent tumor cells can be efficiently exploited as virus host cells by oncolytic MeV. These observations open up a new field for preclinical and clinical research to further investigate TIS and oncolytic viruses as an attractive combinatorial future treatment approach.


Assuntos
Senescência Celular/fisiologia , Resistencia a Medicamentos Antineoplásicos/fisiologia , Vírus do Sarampo/fisiologia , Terapia Viral Oncolítica/métodos , Vírus Oncolíticos/fisiologia , Antineoplásicos/farmacologia , Linhagem Celular Tumoral , Senescência Celular/efeitos dos fármacos , Citometria de Fluxo , Humanos , Vacina contra Sarampo/uso terapêutico , Neoplasias/terapia , Neoplasias/virologia
10.
Vaccine ; 31(37): 3888-93, 2013 Aug 20.
Artigo em Inglês | MEDLINE | ID: mdl-23831325

RESUMO

We developed a novel vaccine platform based on a paramyxoviral, genome replication-deficient Sendai virus vector that can express heterologous genes inserted into the genome. To validate the novel approach in vivo, we generated a combined vaccine candidate against human respiratory syncytial virus (RSV) and human parainfluenza virus type 3 (PIV3). The present study compares two different methods of displaying heterologous antigens: (i) the RSV fusion (F) protein, encoded as a secretable version in an additional transcription unit, serves as an antigen only after being expressed in infected cells; (ii) PIV3 fusion (F) and hemagglutinin-neuraminidase (HN) genes, replacing Sendai counterparts in the vector genome, are also expressed as structural components on the surface of vaccine particles. The efficacy of this prototype vaccine was assessed in a mouse model after mucosal administration. The vaccine candidate was able to elicit specific mucosal, humoral and T cell-mediated immune responses against RSV and PIV3. However, PIV3 antigen display on the vaccine particles' surface induced higher antibody titers than the RSV antigen, being expressed only after cell infection. Consequently, this construct induced an adequate neutralizing antibody response only to PIV3. Finally, replicating virus particles were not detected in the lungs of immunized mice, confirming the genome stability and replication deficiency of this vaccine vector in vivo. Both factors can contribute substantially to the safety profile of vaccine candidates. In conclusion, this replication-deficient Sendai vector represents an efficient platform that can be used for vaccine developments against various viral pathogens.


Assuntos
Vírus Sendai/genética , Vacinas Sintéticas/genética , Vacinas Sintéticas/imunologia , Animais , Anticorpos Neutralizantes/sangue , Feminino , Vetores Genéticos , Genoma Viral , Proteína HN/genética , Imunidade Humoral , Camundongos , Camundongos Endogâmicos BALB C , Vírus da Parainfluenza 3 Humana/genética , Vírus da Parainfluenza 3 Humana/imunologia , Vírus da Parainfluenza 3 Humana/patogenicidade , Vacinas contra Vírus Sincicial Respiratório/genética , Vacinas contra Vírus Sincicial Respiratório/imunologia , Vírus Sincicial Respiratório Humano/genética , Vírus Sincicial Respiratório Humano/patogenicidade , Linfócitos T/imunologia , Vacinas Sintéticas/farmacologia , Proteínas Virais de Fusão/genética , Replicação Viral
11.
Hum Gene Ther ; 24(7): 644-54, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23642239

RESUMO

Oncolytic measles viruses (MV) derived from the live attenuated vaccine strain have been engineered for increased antitumor activity, and are currently under investigation in clinical phase 1 trials. Approaches with other viral vectors have shown that insertion of immunomodulatory transgenes enhances the therapeutic potency. In this study, we engineered MV for expression of the cytokine granulocyte-macrophage colony-stimulating factor (GM-CSF). For the first time, therapeutic efficacy and adaptive immune response in the context of MV oncolysis could be evaluated in the previously established immunocompetent murine colon adenocarcinoma model MC38cea. MC38cea cells express the human carcinoembryonic antigen (CEA), allowing for infection with retargeted MV. Intratumoral application of MV-GMCSF significantly delayed tumor progression and prolonged median overall survival compared with control virus-treated mice. Importantly, more than one-third of mice treated with MV-GMCSF showed complete tumor remission and rejected successive tumor reengraftment, demonstrating robust long-term protection. An enhanced cell-mediated tumor-specific immune response could be detected by lactate dehydrogenase assay and interferon-γ enzyme-linked immunospot assay. Furthermore, MV-GMCSF treatment correlated with increased abundance of tumor-infiltrating CD3(+) lymphocytes analyzed by quantitative microscopy of tumor sections. These findings underline the potential of oncolytic, GM-CSF-expressing MV as an effective therapeutic cancer vaccine actively recruiting adaptive immune responses for enhanced therapeutic impact and tumor elimination. Thus, the treatment benefit of this combined immunovirotherapy approach has direct implications for future clinical trials.


Assuntos
Adenocarcinoma/prevenção & controle , Neoplasias do Colo/prevenção & controle , Fator Estimulador de Colônias de Granulócitos e Macrófagos/imunologia , Vírus do Sarampo/imunologia , Terapia Viral Oncolítica/métodos , Vírus Oncolíticos/imunologia , Imunidade Adaptativa/imunologia , Adenocarcinoma/genética , Adenocarcinoma/imunologia , Análise de Variância , Animais , Chlorocebus aethiops , Neoplasias do Colo/genética , Neoplasias do Colo/imunologia , ELISPOT , Engenharia Genética/métodos , Fator Estimulador de Colônias de Granulócitos e Macrófagos/genética , Fator Estimulador de Colônias de Granulócitos e Macrófagos/metabolismo , Imuno-Histoquímica , L-Lactato Desidrogenase/metabolismo , Vírus do Sarampo/genética , Camundongos , Vírus Oncolíticos/genética , Células Vero
12.
Hum Gene Ther ; 24(5): 554-64, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-23550539

RESUMO

Cholangiocarcinoma (CC) is curable only in early stages by complete surgical resection. Thus, in advanced disease stages in which a complete removal of the tumor mass is no longer possible and palliative chemotherapy achieves only modest success, therapeutics employing new methods of action are desperately needed. Oncolytic viruses employed in clinical studies have been shown to spread preferentially in cancer cells. Beyond that, virotherapeutic cell killing can be enhanced by virus-based expression of suicide genes. We engineered a measles vaccine virus (MeV) vector expressing super cytosine deaminase (SCD), a fusion protein of yeast cytosine deaminase and uracil phosphoribosyltransferase, which converts the prodrug 5-fluorocytosine (5-FC) to 5-fluorouracil (5-FU) and subsequently to 5-fluorouridine-monophosphate. This novel vector was evaluated using three different human-derived CC cell lines. In vitro, all CC cell lines were found to be permissive to MeV infection. Partial blocking of MeV-mediated oncolysis could be overcome by employment of the SCD transgene together with administration of 5-FC. In vivo, intratumoral application of SCD-armed MeV together with a systemic 5-FC treatment showed a significant reduction in tumor size in a TFK-1 xenograft mouse model when compared with virus-only treatment. In a second animal experiment employing a HuCCT1 xenograft tumor model, an enhanced SCD-armed MeV vector, in which the SCD transgene was expressed from a different genomic position, led not only to reduced tumor volumes, but also to a significant survival benefit. On the basis of these encouraging preclinical data on employment of SCD-armed MeV for the virotherapeutic treatment of chemotherapy-resistant CC, a clinical virotherapy trial is set up currently.


Assuntos
Neoplasias dos Ductos Biliares/terapia , Colangiocarcinoma/genética , Colangiocarcinoma/terapia , Terapia Viral Oncolítica , Vírus Oncolíticos/genética , Animais , Neoplasias dos Ductos Biliares/genética , Neoplasias dos Ductos Biliares/virologia , Linhagem Celular Tumoral , Colangiocarcinoma/patologia , Fluoruracila/administração & dosagem , Terapia Genética , Humanos , Vacina contra Sarampo/administração & dosagem , Vírus do Sarampo/genética , Camundongos
13.
J Invest Dermatol ; 133(4): 1034-42, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23223133

RESUMO

Effective treatment modalities for advanced melanoma are desperately needed. An innovative approach is virotherapy, in which viruses are engineered to infect cancer cells, resulting in tumor cell lysis and an amplification effect by viral replication and spread. Ideally, tumor selectivity of these oncolytic viruses is already determined during viral cell binding and entry, which has not been reported for melanoma. We engineered an oncolytic measles virus entering melanoma cells through the high molecular weight melanoma-associated antigen (HMWMAA) and proved highly specific infection and spread in melanoma cells. We further enhanced this oncolytic virus by inserting the FCU1 gene encoding the yeast-derived prodrug convertases cytosine deaminase and uracil phosphoribosyltransferase. Combination treatment with armed and retargeted MV-FCU1-αHMWMAA and the prodrug 5-fluorocytosine (5-FC) led to effective prodrug conversion to 5-fluorouracil, extensive cytotoxicity to melanoma cells, and excessive bystander killing of noninfected cells. Importantly, HMWMAA-retargeted MV showed antitumor activity in a human xenograft mouse model, which was further increased by the FCU1/5-FC prodrug activation system. Finally, we demonstrated susceptibility of melanoma skin metastasis biopsies to HMWMAA-retargeted MV. The highly selective, entry-targeted and armed oncolytic virus MV-FCU1-αHMWMAA may become a potent building block of future melanoma therapies.


Assuntos
Vírus do Sarampo/genética , Melanoma/tratamento farmacológico , Terapia Viral Oncolítica/métodos , Pró-Fármacos/farmacocinética , Neoplasias Cutâneas/tratamento farmacológico , Animais , Antígenos de Neoplasias/metabolismo , Antimetabólitos/metabolismo , Biópsia , Linhagem Celular Tumoral , Chlorocebus aethiops , Terapia Combinada , Feminino , Flucitosina/metabolismo , Engenharia Genética/métodos , Genoma Viral/genética , Humanos , Melanoma/secundário , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Proteínas Recombinantes/genética , Neoplasias Cutâneas/patologia , Células Vero , Ensaios Antitumorais Modelo de Xenoenxerto
14.
Open Virol J ; 6: 73-81, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22787520

RESUMO

In the beginning of a paramyxovirus infection after cell entry viral survival depends on efficient primary (1°) transcription and on the stability of only one input nucleocapsid. Here we examined the influence of the viral polymerase co-factor phosphoprotein P on the very early phase of an infection, i.e. before progeny nucleocapsids are synthesized. We used a novel set-up with Sendai virus (SeV) mutants incapable of genome replication: SeV-ΔP with the entire P ORF deleted, SeV-PΔ2-77 with the deletion of aa 2-77. These mutants allow maintaining the state of the very beginning of an infection when statistically one viral genome is present in the cell. This single genome serves as template for transcription. During SeV-ΔP infections only early 1° transcription takes place at low levels. However, when the truncated P protein is expressed in SeV-PΔ2-77 infections, 1° transcription levels rise significantly up to an 8-fold increased amount of viral mRNA. This shows that the P protein is able to support transcription and thereby mediates the transition from early to late 1° transcription. Importantly, nucleocapsids of both mutants could be shown to remain stable and functional for at least 5 days - even without de novo P protein synthesis. These results describe a novel function of the P protein: enhancing viral gene expression even before genome replication has started. Thus, the since long postulated supportive function of the P protein is not related to stabilization of the nucleocapsid but rather enhances the processivity of the viral polymerase during late 1° and secondary (2°) transcription and genome replication.

15.
Mol Ther ; 19(6): 1097-106, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21468006

RESUMO

Oncolytic measles viruses (MV) derived from the live attenuated vaccine strain have been engineered for increased tumor-cell specificity, and are currently under investigation in clinical trials including a phase I study for glioblastoma multiforme (GBM). Recent preclinical studies have shown that the cellular tropism of several viruses can be controlled by inserting microRNA-target sequences into their genomes, thereby inhibiting spread in tissues expressing cognate microRNAs. Since neuron-specific microRNA-7 is downregulated in gliomas but highly expressed in normal brain tissue, we engineered a microRNA-sensitive virus containing target sites for microRNA-7 in the 3'-untranslated region of the viral fusion gene. In presence of microRNA-7 this modification inhibits translation of envelope proteins, restricts viral spread, and progeny production. Even though highly attenuated in presence of microRNA-7, this virus retained full efficacy against glioblastoma xenografts. Furthermore, microRNA-mediated inhibition protected genetically modified mice susceptible to MV infection from a potentially lethal intracerebral challenge. Importantly, endogenous microRNA-7 expression in primary human brain resections tightly restricted replication and spread of microRNA-sensitive virus. This is proof-of-concept that tropism restriction by tissue-specific microRNAs can be adapted to oncolytic MV to regulate viral replication and gene expression to maximize tumor specificity without compromising oncolytic efficacy.


Assuntos
Sobrevivência Celular/fisiologia , Vírus do Sarampo/genética , MicroRNAs/genética , Vírus Oncolíticos/fisiologia , Animais , Neoplasias Encefálicas/terapia , Linhagem Celular , Linhagem Celular Tumoral , Sobrevivência Celular/genética , Chlorocebus aethiops , Feminino , Vetores Genéticos/genética , Glioblastoma/terapia , Glioma/terapia , Humanos , Immunoblotting , Técnicas In Vitro , Vírus do Sarampo/fisiologia , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Terapia Viral Oncolítica , Vírus Oncolíticos/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Células Vero , Ensaios Antitumorais Modelo de Xenoenxerto
16.
Cell Immunol ; 247(2): 85-94, 2007 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-17904538

RESUMO

Respiratory syncytial virus (RSV) causes severe respiratory disease in infants and a vaccine is highly desirable. The fusion (F) protein of RSV is an important vaccine target, but the contribution of F-specific T cells to successful vaccination remains unclear. We studied the immune response to vaccination of mice with a recombinant Sendai virus expressing RSV F (rSeV F). rSeV F induced protective neutralizing antibody and RSV F-specific CTL responses. T cell immunity was stronger than that induced by recombinant vaccinia virus (rVV F), a well characterized reference vector. Vaccination of antibody-deficient mice showed that vaccine-induced RSV F-specific T cells were sufficient for protective immunity. rSeV F induced T cell immunity in the presence of neutralizing antibodies, which did not impair the vaccine response. Although the F protein only contains a subdominant CTL epitope, vaccination with rSeV F is sufficient to induce protective T cell immunity against RSV in mice.


Assuntos
Engenharia Genética , Infecções por Vírus Respiratório Sincicial/imunologia , Infecções por Vírus Respiratório Sincicial/prevenção & controle , Vírus Sinciciais Respiratórios/imunologia , Vírus Sendai/genética , Vírus Sendai/imunologia , Linfócitos T/imunologia , Animais , Anticorpos/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Infecções por Vírus Respiratório Sincicial/patologia , Solubilidade , Linfócitos T/efeitos dos fármacos , Proteínas Virais de Fusão/genética , Proteínas Virais de Fusão/imunologia
17.
J Virol ; 81(24): 13835-44, 2007 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-17855540

RESUMO

Among the members of the paramyxovirus family, the transcription process and the components involved have been studied under in vitro conditions thus far. Here, we reexamined the function of the viral RNA-dependent RNA polymerase through infection studies with Sendai virus (SeV) N and P deletion (Delta) mutants. To elucidate solely transcription-specific processes, all virus mutants also were rendered deficient in genome replication. Using mutant SeV DeltaP, the earlier suspected supplemental role of P protein was clearly demonstrated to be essential during viral gene expression. Moreover, when SeV DeltaN or DeltaN PDelta2-77 (with the 5' end of the P gene deleted) mutant was used for infections, a completely unexpected new and essential role for N protein was discovered for viral gene expression. In the early phases of an infection and in the absence of de novo viral protein synthesis, primary transcription occurs at hardly detectable levels. In contrast, if newly synthesized N protein is present, primary viral transcription reaches normal levels. From our data, we conclude that de novo synthesis of SeV N and P proteins is a key step for viral gene expression that facilitates the transition from preliminary to normal primary transcriptional activity.


Assuntos
Regulação Viral da Expressão Gênica , Proteínas do Nucleocapsídeo/biossíntese , Fosfoproteínas/biossíntese , RNA Viral/metabolismo , Vírus Sendai/metabolismo , Proteínas Virais/metabolismo , Animais , Linhagem Celular , Chlorocebus aethiops , Deleção de Genes , Proteínas do Nucleocapsídeo/genética , Fosfoproteínas/genética , RNA Viral/genética , Recombinação Genética , Vírus Sendai/genética , Vírus Sendai/patogenicidade , Transcrição Gênica , Células Vero , Proteínas Virais/biossíntese , Proteínas Virais/genética
18.
J Immunol ; 175(8): 4971-80, 2005 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-16210599

RESUMO

Delivery of Ags to dendritic cells (DCs) plays a pivotal role in the induction of efficient immune responses ranging from immunity to tolerance. The observation that certain viral pathogens are able to infect DCs has led to a concept in which applications of recombinant viruses are used for Ag delivery with the potential benefit of inducing potent Ag-specific T cell responses directed against multiple epitopes. As a prerequisite for such an application, the infection of DCs by recombinant viruses should not interfere with their stimulatory capacity. In this context, we could show that an emerging negative-strand RNA viral vector system based on the Sendai virus (SeV) is able to efficiently infect monocyte-derived human DCs (moDCs). However, after infection with SeV wild type, both the response of DCs to bacterial LPS as a powerful mediator of DC maturation and the allostimulatory activity were severely impaired. Interestingly, using various recombinant SeV vectors that were devoid of single viral genes, we were able to identify the SeV matrix (M) protein as a key component in moDC functional impairment after viral infection. Consequently, use of M-deficient SeV vectors preserved the allostimulatory activity in infected moDCs despite an efficient expression of all other virally encoded genes, thereby identifying M-deficient vectors as a highly potent tool for the genetic manipulation of DCs.


Assuntos
Células Dendríticas/imunologia , Células Dendríticas/virologia , Deleção de Genes , Vetores Genéticos/imunologia , Vírus Sendai/imunologia , Proteínas da Matriz Viral/deficiência , Proteínas da Matriz Viral/genética , Apoptose/imunologia , Diferenciação Celular/imunologia , Células Cultivadas , Citocinas/metabolismo , Citocinas/fisiologia , Epitopos de Linfócito T/imunologia , Epitopos de Linfócito T/metabolismo , Humanos , Imunidade Inata , Mediadores da Inflamação/metabolismo , Mediadores da Inflamação/fisiologia , Lipopolissacarídeos/imunologia , Vírus Sendai/genética , Proteínas da Matriz Viral/fisiologia , Vírion/genética , Vírion/imunologia
19.
J Virol ; 79(16): 10467-77, 2005 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-16051839

RESUMO

Entry of most paramyxoviruses is accomplished by separate attachment and fusion proteins that function in a cooperative manner. Because of this close interdependence, it was not possible with most paramyxoviruses to replace either of the two protagonists by envelope glycoproteins from related paramyxoviruses. By using reverse genetics of Sendai virus (SeV), we demonstrate that chimeric respiratory syncytial virus (RSV) fusion proteins containing either the cytoplasmic domain of the SeV fusion protein or in addition the transmembrane domain were efficiently incorporated into SeV particles provided the homotypic SeV-F was deleted. In the presence of SeV-F, the chimeric glycoproteins were incorporated with significantly lower efficiency, indicating that determinants in the SeV-F ectodomain exist that contribute to glycoprotein uptake. Recombinant SeV in which the homotypic fusion protein was replaced with chimeric RSV fusion protein replicated in a trypsin-independent manner and was neutralized by antibodies directed to RSV-F. However, replication of this virus also relied on the hemagglutinin-neuraminidase (HN) as pretreatment of cells with neuraminidase significantly reduced the infection rate. Finally, recombinant SeV was generated with chimeric RSV-F as the only envelope glycoprotein. This virus was not neutralized by antibodies to SeV and did not use sialic acids for attachment. It replicated more slowly than hybrid virus containing HN and produced lower virus titers. Thus, on the one hand RSV-F can mediate infection in an autonomous way while on the other hand it accepts support by a heterologous attachment protein.


Assuntos
Proteína HN/fisiologia , Proteínas Recombinantes de Fusão/fisiologia , Vírus Sinciciais Respiratórios/fisiologia , Vírus Sendai/genética , Proteínas Virais de Fusão/fisiologia , Animais , Anticorpos Antivirais/imunologia , Chlorocebus aethiops , Células HeLa , Humanos , Recombinação Genética , Vírus Sendai/fisiologia , Tripsina/farmacologia , Células Vero , Replicação Viral
20.
J Gen Virol ; 86(Pt 8): 2305-2314, 2005 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-16033978

RESUMO

Induction of apoptosis during Sendai virus (SeV) infection has previously been documented to be triggered by initiator caspases (for strain F) or by a contribution of the cellular protein TIAR (T-cell-activated intracellular antigen-related) (for strain Z). Here, evidence was provided that both TIAR and caspases are simultaneously involved in apoptosis induction as a result of infection with SeV strain F. SeV F infection induced death in all tested cell lines, which could only be partially prevented through the pan-caspase inhibitor z-VAD-fmk. However, infection of seven different cell lines with the SeV mutant Fctr48z overexpressing a TIAR-sequestering RNA from the modified leader resulted in a cell type-dependent reduced cytopathic effect (CPE); in an earlier study a similar mutant derived from SeV Z was shown to prevent the induction of any CPE. Finally, blocking of caspases through z-VAD-fmk combined with Fctr48z infection led to complete abrogation of CPE, clearly demonstrating the existence of two separate mechanisms inducing cell death during SeV F infections. Interestingly, a cell type-specific interference between these two mechanisms could be detected during infection with the mutant virus Fctr48z: RNA transcribed from the mutated leader was able to trans-dominantly inhibit caspase-mediated apoptosis. Thus, virus-expressed factors enabling a well-balanced ratio of suppression and triggering of apoptosis seem to be essential for optimal virus replication.


Assuntos
Apoptose/fisiologia , RNA Viral , Vírus Sendai/fisiologia , Clorometilcetonas de Aminoácidos/farmacologia , Animais , Caspases/genética , Caspases/fisiologia , Linhagem Celular , Efeito Citopatogênico Viral , Dano ao DNA/genética , Dano ao DNA/fisiologia , Humanos , Mutação , Especificidade de Órgãos , Proteínas de Ligação a RNA/genética , Proteínas de Ligação a RNA/fisiologia , Infecções por Respirovirus/virologia , Vírus Sendai/genética , Vírus Sendai/patogenicidade
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...