Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
Mais filtros

Bases de dados
Tipo de documento
País de afiliação
Intervalo de ano de publicação
1.
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
2.
Int J Cancer ; 136(9): 2228-40, 2015 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-25303768

RESUMO

Antibody therapy of solid cancers is well established, but suffers from unsatisfactory tumor penetration of large immunoglobulins or from low serum retention of antibody fragments. Oncolytic viruses are in advanced clinical development showing excellent safety, but suboptimal potency due to limited virus spread within tumors. Here, by developing an immunoRNase-encoding oncolytic adenovirus, we combine viral oncolysis with intratumoral genetic delivery of a small antibody-fusion protein for targeted bystander killing of tumor cells (viro-antibody therapy). Specifically, we explore genetic delivery of a small immunoRNase consisting of an EGFR-binding scFv antibody fragment fused to the RNase Onconase (ONC(EGFR)) that induces tumor cell death by RNA degradation after cellular internalization. Onconase is a frog RNase that combines lack of immunogenicity and excellent safety in patients with high tumor killing potency due to its resistance to the human cytosolic RNase inhibitor. We show that ONC(EGFR) expression by oncolytic adenoviruses is feasible with an optimized, replication-dependent gene expression strategy. Virus-encoded ONC(EGFR) induces potent and EGFR-dependent bystander killing of tumor cells. Importantly, the ONC(EGFR)-encoding oncolytic adenovirus showed dramatically increased cytotoxicity specifically to EGFR-positive tumor cells in vitro and significantly enhanced therapeutic activity in a mouse xenograft tumor model. The latter demonstrates that ONC(EGFR) is expressed at levels sufficient to trigger tumor cell killing in vivo. The established ONC(EGFR)-encoding oncolytic adenovirus represents a novel agent for treatment of EGFR-positive tumors. This viro-antibody therapy platform can be further developed for targeted/personalized cancer therapy by exploiting antibody diversity to target further established or emerging tumor markers or combinations thereof.


Assuntos
Adenoviridae/genética , Antineoplásicos/administração & dosagem , Antineoplásicos/metabolismo , Vetores Genéticos/genética , Vírus Oncolíticos/genética , Ribonucleases/administração & dosagem , Ribonucleases/metabolismo , Animais , Anticorpos Antivirais , Linhagem Celular Tumoral , Receptores ErbB/metabolismo , Feminino , Terapia Genética/métodos , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Terapia Viral Oncolítica/métodos , RNA/metabolismo , Replicação Viral/efeitos dos fármacos , Ensaios Antitumorais Modelo de Xenoenxerto
3.
Nucleic Acids Res ; 40(21): e167, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-22885302

RESUMO

Therapeutic gene transfer by replication-defective viral vectors or, for cancer treatment, by replication-competent oncolytic viruses shows high promise for treatment of major diseases. To ensure safety, timing or dosing in patients, external control of therapeutic gene expression is desirable or even required. In this study, we explored the potential of artificial aptazymes, ligand-dependent self-cleaving ribozymes, as an innovative tool for regulation of therapeutic gene expression. Importantly, aptazymes act on RNA intrinsically, independent of regulatory protein-nucleic acid interactions and stoichiometry, are non-immunogenic and of small size. These are key advantages compared with the widely used inducible promoters, which were also reported to lose regulation at high copy numbers, e.g. after replication of oncolytic viruses. We characterized aptazymes in therapeutic gene transfer utilizing adenovectors (AdVs), adeno-associated vectors (AAVs) and oncolytic adenoviruses (OAds), which are all in advanced clinical testing. Our results show similar aptazyme-mediated regulation of gene expression by plasmids, AdVs, AAVs and OAds. Insertion into the 5'-, 3'- or both untranslated regions of several transgenes resulted in ligand-responsive gene expression. Notably, aptazyme regulation was retained during OAd replication and spread. In conclusion, our study demonstrates the fidelity of aptazymes in viral vectors and oncolytic viruses and highlights the potency of riboswitches for medical applications.


Assuntos
Adenoviridae/genética , Regulação da Expressão Gênica , Vírus Oncolíticos/genética , RNA Catalítico/genética , Riboswitch , Adenoviridae/fisiologia , Linhagem Celular Tumoral , Vírus Defeituosos/genética , Dependovirus/genética , Vetores Genéticos , Genoma Viral , Humanos , RNA Catalítico/metabolismo , Transdução Genética , Transgenes , Regiões não Traduzidas , Replicação Viral
4.
PLoS One ; 9(4): e95723, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24760010

RESUMO

Adenoviral gene therapy and oncolysis would critically benefit from targeted cell entry by genetically modified capsids. This requires both the ablation of native adenovirus tropism and the identification of ligands that remain functional in virus context. Here, we establish cell type-specific entry of HAdV-5-based vectors by genetic ligand insertion into a chimeric fiber with shaft and knob domains of the short HAdV-41 fiber (Ad5T/41sSK). This fiber format was reported to ablate transduction in vitro and biodistribution to the liver in vivo. We show that the YSA peptide, binding to the pan-cancer marker EphA2, can be inserted into three positions of the chimeric fiber, resulting in strong transduction of EphA2-positive but not EphA2-negative cells of human melanoma biopsies and of tumor xenografts after intratumoral injection. Transduction was blocked by soluble YSA peptide and restored for EphA2-negative cells after recombinant EphA2 expression. The YSA peptide could also be inserted into three positions of a CAR binding-ablated HAdV-5 fiber enabling specific transduction; however, the Ad5T/41sSK format was superior in vivo. In conclusion, we establish an adenovirus capsid facilitating functional insertion of targeting peptides and a novel adenovirus using the tumor marker EphA2 as receptor with high potential for cancer gene therapy and viral oncolysis.


Assuntos
Adenoviridae/metabolismo , Receptor EphA2/metabolismo , Animais , Linhagem Celular , Feminino , Humanos , Melanoma/metabolismo , Melanoma/terapia , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Ensaios Antitumorais Modelo de Xenoenxerto
5.
Hum Gene Ther ; 22(4): 389-404, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-20939692

RESUMO

Key challenges facing cancer therapy are the development of tumor-specific drugs and potent multimodal regimens. Oncolytic adenoviruses possess the potential to realize both aims by restricting virus replication to tumors and inserting therapeutic genes into the virus genome, respectively. A major effort in this regard is to express transgenes in a tumor-specific manner without affecting virus replication. Using both luciferase as a sensitive reporter and genetic prodrug activation, we show that promoter control of E1A facilitates highly selective expression of transgenes inserted into the late transcription unit. This, however, required multistep optimization of late transgene expression. Transgene insertion via internal ribosome entry site (IRES), splice acceptor (SA), or viral 2A sequences resulted in replication-dependent expression. Unexpectedly, analyses in appropriate substrates and with matching control viruses revealed that IRES and SA, but not 2A, facilitated indirect transgene targeting via tyrosinase promoter control of E1A. Transgene expression via SA was more selective (up to 1,500-fold) but less effective than via IRES. Notably, we also revealed transgene-dependent interference with splicing. Hence, the prodrug convertase FCU1 (a cytosine deaminase-uracil phosphoribosyltransferase fusion protein) was expressed only after optimizing the sequence surrounding the SA site and mutating a cryptic splice site within the transgene. The resulting tyrosinase promoter-regulated and FCU1-encoding adenovirus combined effective oncolysis with targeted prodrug activation therapy of melanoma. Thus, prodrug activation showed potent bystander killing and increased cytotoxicity of the virus up to 10-fold. We conclude that armed oncolytic viruses can be improved substantially by comparing and optimizing strategies for targeted transgene expression, thereby implementing selective and multimodal cancer therapies.


Assuntos
Adenoviridae , Genes Virais , Vetores Genéticos , Mutagênese Insercional/genética , Vírus Oncolíticos , Transcrição Gênica , Transgenes/genética , Adenoviridae/genética , Adenoviridae/metabolismo , Processamento Alternativo/genética , Antineoplásicos/farmacologia , Efeito Espectador/efeitos dos fármacos , Efeito Espectador/genética , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/genética , Regulação da Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica/genética , Ordem dos Genes , Células HEK293 , Humanos , Melanoma/terapia , Monofenol Mono-Oxigenase/genética , Vírus Oncolíticos/genética , Vírus Oncolíticos/metabolismo , Pró-Fármacos/metabolismo , Regiões Promotoras Genéticas/genética , Sequências Reguladoras de Ácido Nucleico
6.
PLoS One ; 6(11): e27934, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-22140489

RESUMO

Adenoviruses (Ads), especially HAdV-5, have been genetically equipped with tumor-restricted replication potential to enable applications in oncolytic cancer therapy. Such oncolytic adenoviruses have been well tolerated in cancer patients, but their anti-tumor efficacy needs to be enhanced. In this regard, it should be considered that cancer cells, dependent on their tissue of origin, can differ substantially from the normal host cells to which Ads are adapted by complex virus-host interactions. Consequently, viral replication efficiency, a key determinant of oncolytic activity, might be suboptimal in cancer cells. Therefore, we have analyzed both the replication kinetics of HAdV-5 and the virus-induced transcriptome in human bronchial epithelial cells (HBEC) in comparison to cancer cells. This is the first report on genome-wide expression profiling of Ads in their native host cells. We found that E1A expression and onset of viral genome replication are most rapid in HBEC and considerably delayed in melanoma cells. In squamous cell lung carcinoma cells, we observed intermediate HAdV-5 replication kinetics. Infectious particle production, viral spread and lytic activity of HAdV-5 were attenuated in melanoma cells versus HBEC. Expression profiling at the onset of viral genome replication revealed that HAdV-5 induced the strongest changes in the cellular transcriptome in HBEC, followed by lung cancer and melanoma cells. We identified prominent regulation of genes involved in cell cycle and DNA metabolism, replication and packaging in HBEC, which is in accord with the necessity to induce S phase for viral replication. Strikingly, in melanoma cells HAdV-5 triggered opposing regulation of said genes and, in contrast to lung cancer cells, no weak S phase induction was detected when using the E2F promoter as reporter. Our results provide a rationale for improving oncolytic adenoviruses either by adaptation of viral infection to target tumor cells or by modulating tumor cell functions to better support viral replication.


Assuntos
Adenovírus Humanos/genética , Adenovírus Humanos/fisiologia , Neoplasias/virologia , Vírus Oncolíticos/genética , Vírus Oncolíticos/fisiologia , Transcriptoma/genética , Replicação Viral/genética , Infecções por Adenovirus Humanos/virologia , Brônquios/patologia , Linhagem Celular Tumoral , Análise por Conglomerados , Efeito Citopatogênico Viral , Regulação para Baixo/genética , Fator de Transcrição E2F1/genética , Células Epiteliais/metabolismo , Células Epiteliais/virologia , Regulação Viral da Expressão Gênica , Genes Reporter/genética , Genoma Viral/genética , Humanos , Anotação de Sequência Molecular , Regiões Promotoras Genéticas/genética , Fase S , Regulação para Cima/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA