Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 3 de 3
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Nat Commun ; 15(1): 493, 2024 Jan 12.
Artigo em Inglês | MEDLINE | ID: mdl-38216554

RESUMO

Measles virus (MV) vaccine strains have shown significant preclinical antitumor activity against glioblastoma (GBM), the most lethal glioma histology. In this first in human trial (NCT00390299), a carcinoembryonic antigen-expressing oncolytic measles virus derivative (MV-CEA), was administered in recurrent GBM patients either at the resection cavity (Group A), or, intratumorally on day 1, followed by a second dose administered in the resection cavity after tumor resection on day 5 (Group B). A total of 22 patients received study treatment, 9 in Group A and 13 in Group B. Primary endpoint was safety and toxicity: treatment was well tolerated with no dose-limiting toxicity being observed up to the maximum feasible dose (2×107 TCID50). Median OS, a secondary endpoint, was 11.6 mo and one year survival was 45.5% comparing favorably with contemporary controls. Other secondary endpoints included assessment of viremia, MV replication and shedding, humoral and cellular immune response to the injected virus. A 22 interferon stimulated gene (ISG) diagonal linear discriminate analysis (DLDA) classification algorithm in a post-hoc analysis was found to be inversely (R = -0.6, p = 0.04) correlated with viral replication and tumor microenvironment remodeling including proinflammatory changes and CD8 + T cell infiltration in post treatment samples. This data supports that oncolytic MV derivatives warrant further clinical investigation and that an ISG-based DLDA algorithm can provide the basis for treatment personalization.


Assuntos
Glioblastoma , Terapia Viral Oncolítica , Vírus Oncolíticos , Humanos , Vírus do Sarampo/genética , Antígeno Carcinoembrionário/genética , Recidiva Local de Neoplasia/terapia , Vacina contra Sarampo , Microambiente Tumoral
2.
PLoS Genet ; 14(1): e1007179, 2018 01.
Artigo em Inglês | MEDLINE | ID: mdl-29364907

RESUMO

Integration of human papillomavirus (HPV) genomes into cellular chromatin is common in HPV-associated cancers. Integration is random, and each site is unique depending on how and where the virus integrates. We recently showed that tandemly integrated HPV16 could result in the formation of a super-enhancer-like element that drives transcription of the viral oncogenes. Here, we characterize the chromatin landscape and genomic architecture of this integration locus to elucidate the mechanisms that promoted de novo super-enhancer formation. Using next-generation sequencing and molecular combing/fiber-FISH, we show that ~26 copies of HPV16 are integrated into an intergenic region of chromosome 2p23.2, interspersed with 25 kb of amplified, flanking cellular DNA. This interspersed, co-amplified viral-host pattern is frequent in HPV-associated cancers and here we designate it as Type III integration. An abundant viral-cellular fusion transcript encoding the viral E6/E7 oncogenes is expressed from the integration locus and the chromatin encompassing both the viral enhancer and a region in the adjacent amplified cellular sequences is strongly enriched in the super-enhancer markers H3K27ac and Brd4. Notably, the peak in the amplified cellular sequence corresponds to an epithelial-cell-type specific enhancer. Thus, HPV16 integration generated a super-enhancer-like element composed of tandem interspersed copies of the viral upstream regulatory region and a cellular enhancer, to drive high levels of oncogene expression.


Assuntos
Regulação Viral da Expressão Gênica , Genes Virais , Papillomavirus Humano 16/genética , Papillomavirus Humano 16/patogenicidade , Fatores de Transcrição/metabolismo , Integração Viral/fisiologia , Células Cultivadas , Elementos Facilitadores Genéticos , Células HCT116 , Células HeLa , Células Hep G2 , Interações Hospedeiro-Patógeno/genética , Células Endoteliais da Veia Umbilical Humana , Papillomavirus Humano 16/metabolismo , Humanos , Células K562 , Vírus Oncogênicos/genética , Vírus Oncogênicos/patogenicidade , Papillomaviridae/genética , Papillomaviridae/metabolismo , Papillomaviridae/patogenicidade , Ligação Proteica , Multimerização Proteica , Regulação para Cima/genética
3.
mBio ; 7(5)2016 09 13.
Artigo em Inglês | MEDLINE | ID: mdl-27624132

RESUMO

UNLABELLED: In cancer cells associated with human papillomavirus (HPV) infections, the viral genome is very often found integrated into the cellular genome. The viral oncogenes E6 and E7 are transcribed from the viral promoter, and integration events that alter transcriptional regulation of this promoter contribute to carcinogenic progression. In this study, we detected highly enriched binding of the super-enhancer markers Brd4, MED1, and H3K27ac, visible as a prominent nuclear focus by immunofluorescence, at the tandemly integrated copies of HPV16 in cells of the cervical neoplasia cell line W12 subclone 20861. Tumor cells are often addicted to super-enhancer-driven oncogenes and are particularly sensitive to disruption of transcription factor binding to the enhancers. Treatment of 20861 cells with bromodomain inhibitors displaced Brd4 from the HPV integration site, greatly decreased E6/E7 transcription, and inhibited cellular proliferation. Thus, Brd4 activates viral transcription at this integration site, and strong selection for E6/E7 expression can drive the formation of a super-enhancer-like element to promote oncogenesis. IMPORTANCE: Oncogenic human papillomaviruses play an essential role in the development of cervical cancer, and growth of these cancer cells requires continued expression of the viral E6 and E7 oncogenes. Integration of the virus into the host genome often results in deregulation of E6 and E7 expression, which provides a selective growth advantage and increases genetic instability of infected cells. We show here that tandemly integrated copies of the viral genome can form a super-enhancer-like element that drives E6/E7 transcription. Targeted disruption of factors binding to this element decreases viral transcription and causes cell death. Thus, cancer cells that harbor integrated HPV could be targeted by therapeutics that disrupt super-enhancer function.


Assuntos
Interações Hospedeiro-Patógeno , Papillomavirus Humano 16/fisiologia , Proteínas Nucleares/metabolismo , Proteínas Oncogênicas Virais/biossíntese , Proteínas E7 de Papillomavirus/biossíntese , Proteínas Repressoras/biossíntese , Fatores de Transcrição/metabolismo , Transcrição Gênica , Integração Viral , Proteínas de Ciclo Celular , Linhagem Celular Tumoral , Expressão Gênica , Histonas/metabolismo , Humanos , Subunidade 1 do Complexo Mediador/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...