Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 8 de 8
Filtrer
Plus de filtres










Base de données
Gamme d'année
1.
J Pathol ; 2024 Jul 02.
Article de Anglais | MEDLINE | ID: mdl-38956451

RÉSUMÉ

Ovarian high-grade serous carcinoma (HGSC) originates in the fallopian tube, with secretory cells carrying a TP53 mutation, known as p53 signatures, identified as potential precursors. p53 signatures evolve into serous tubal intraepithelial carcinoma (STIC) lesions, which in turn progress into invasive HGSC, which readily spreads to the ovary and disseminates around the peritoneal cavity. We recently investigated the genomic landscape of early- and late-stage HGSC and found higher ploidy in late-stage (median 3.1) than early-stage (median 2.0) samples. Here, to explore whether the high ploidy and possible whole-genome duplication (WGD) observed in late-stage disease were determined early in the evolution of HGSC, we analysed archival formalin-fixed paraffin-embedded (FFPE) samples from five HGSC patients. p53 signatures and STIC lesions were laser-capture microdissected and sequenced using shallow whole-genome sequencing (sWGS), while invasive ovarian/fallopian tube and metastatic carcinoma samples underwent macrodissection and were profiled using both sWGS and targeted next-generation sequencing. Results showed highly similar patterns of global copy number change between STIC lesions and invasive carcinoma samples within each patient. Ploidy changes were evident in STIC lesions, but not p53 signatures, and there was a strong correlation between ploidy in STIC lesions and invasive ovarian/fallopian tube and metastatic samples in each patient. The reconstruction of sample phylogeny for each patient from relative copy number indicated that high ploidy, when present, occurred early in the evolution of HGSC, which was further validated by copy number signatures in ovarian and metastatic tumours. These findings suggest that aberrant ploidy, suggestive of WGD, arises early in HGSC and is detected in STIC lesions, implying that the trajectory of HGSC may be determined at the earliest stages of tumour development. © 2024 The Author(s). The Journal of Pathology published by John Wiley & Sons Ltd on behalf of The Pathological Society of Great Britain and Ireland.

2.
Clin Cancer Res ; 28(13): 2911-2922, 2022 07 01.
Article de Anglais | MEDLINE | ID: mdl-35398881

RÉSUMÉ

PURPOSE: Ovarian high-grade serous carcinoma (HGSC) is usually diagnosed at late stage. We investigated whether late-stage HGSC has unique genomic characteristics consistent with acquisition of evolutionary advantage compared with early-stage tumors. EXPERIMENTAL DESIGN: We performed targeted next-generation sequencing and shallow whole-genome sequencing (sWGS) on pretreatment samples from 43 patients with FIGO stage I-IIA HGSC to investigate somatic mutations and copy-number (CN) alterations (SCNA). We compared results to pretreatment samples from 52 patients with stage IIIC/IV HGSC from the BriTROC-1 study. RESULTS: Age of diagnosis did not differ between early-stage and late-stage patients (median 61.3 years vs. 62.3 years, respectively). TP53 mutations were near-universal in both cohorts (89% early-stage, 100% late-stage), and there were no significant differences in the rates of other somatic mutations, including BRCA1 and BRCA2. We also did not observe cohort-specific focal SCNA that could explain biological behavior. However, ploidy was higher in late-stage (median, 3.0) than early-stage (median, 1.9) samples. CN signature exposures were significantly different between cohorts, with greater relative signature 3 exposure in early-stage and greater signature 4 in late-stage. Unsupervised clustering based on CN signatures identified three clusters that were prognostic. CONCLUSIONS: Early-stage and late-stage HGSCs have highly similar patterns of mutation and focal SCNA. However, CN signature analysis showed that late-stage disease has distinct signature exposures consistent with whole-genome duplication. Further analyses will be required to ascertain whether these differences reflect genuine biological differences between early-stage and late-stage or simply time-related markers of evolutionary fitness. See related commentary by Yang et al., p. 2730.


Sujet(s)
Tumeurs de l'ovaire , Carcinome épithélial de l'ovaire , Femelle , Génomique , Séquençage nucléotidique à haut débit , Humains , Adulte d'âge moyen , Mutation , Tumeurs de l'ovaire/génétique , Tumeurs de l'ovaire/anatomopathologie
3.
Cancer Res ; 76(20): 6118-6129, 2016 10 15.
Article de Anglais | MEDLINE | ID: mdl-27530326

RÉSUMÉ

There is a need for transplantable murine models of ovarian high-grade serous carcinoma (HGSC) with regard to mutations in the human disease to assist investigations of the relationships between tumor genotype, chemotherapy response, and immune microenvironment. In addressing this need, we performed whole-exome sequencing of ID8, the most widely used transplantable model of ovarian cancer, covering 194,000 exomes at a mean depth of 400× with 90% exons sequenced >50×. We found no functional mutations in genes characteristic of HGSC (Trp53, Brca1, Brca2, Nf1, and Rb1), and p53 remained transcriptionally active. Homologous recombination in ID8 remained intact in functional assays. Further, we found no mutations typical of clear cell carcinoma (Arid1a, Pik3ca), low-grade serous carcinoma (Braf), endometrioid (Ctnnb1), or mucinous (Kras) carcinomas. Using CRISPR/Cas9 gene editing, we modeled HGSC by generating novel ID8 derivatives that harbored single (Trp53-/-) or double (Trp53-/-;Brca2-/-) suppressor gene deletions. In these mutants, loss of p53 alone was sufficient to increase the growth rate of orthotopic tumors with significant effects observed on the immune microenvironment. Specifically, p53 loss increased expression of the myeloid attractant CCL2 and promoted the infiltration of immunosuppressive myeloid cell populations into primary tumors and their ascites. In Trp53-/-;Brca2-/- mutant cells, we documented a relative increase in sensitivity to the PARP inhibitor rucaparib and slower orthotopic tumor growth compared with Trp53-/- cells, with an appearance of intratumoral tertiary lymphoid structures rich in CD3+ T cells. This work validates new CRISPR-generated models of HGSC to investigate its biology and promote mechanism-based therapeutics discovery. Cancer Res; 76(20); 6118-29. ©2016 AACR.


Sujet(s)
Protéine BRCA2/physiologie , Systèmes CRISPR-Cas/physiologie , Cystadénocarcinome séreux/étiologie , Modèles animaux de maladie humaine , Tumeurs de l'ovaire/étiologie , Protéine p53 suppresseur de tumeur/physiologie , Animaux , Protéine BRCA2/génétique , Lignée cellulaire tumorale , Cystadénocarcinome séreux/traitement médicamenteux , Exome , Femelle , Édition de gène , Souris , Souris de lignée C57BL , Souris knockout , Tumeurs de l'ovaire/traitement médicamenteux , Tumeurs de l'ovaire/génétique , Inhibiteurs de poly(ADP-ribose) polymérases/usage thérapeutique , Microenvironnement tumoral , Protéine p53 suppresseur de tumeur/génétique
4.
Mol Cancer Res ; 14(1): 44-55, 2016 Jan.
Article de Anglais | MEDLINE | ID: mdl-26452665

RÉSUMÉ

UNLABELLED: Homologous recombination (HR) function is critically important in high-grade serous ovarian cancer (HGSOC). HGSOC with intact HR has a worse prognosis and is less likely to respond to platinum chemotherapy and PARP inhibitors. Oncolytic adenovirus, a novel therapy for human malignancies, stimulates a potent DNA damage response that influences overall antitumor activity. Here, the importance of HR was investigated by determining the efficacy of adenovirus type 5 (Ad5) vectors in ovarian cancer. Using matched BRCA2-mutant and wild-type HGSOC cells, it was demonstrated that intact HR function promotes viral DNA replication and augments overall efficacy, without influencing viral DNA processing. These data were confirmed in a wider panel of HR competent and defective ovarian cancer lines. Mechanistically, both BRCA2 and RAD51 localize to viral replication centers within the infected cell nucleus and that RAD51 localization occurs independently of BRCA2. In addition, a direct interaction was identified between RAD51 and adenovirus E2 DNA binding protein. Finally, using functional assays of HR competence, despite inducing degradation of MRE11, Ad5 infection does not alter cellular ability to repair DNA double-strand break damage via HR. These data reveal that Ad5 redistributes critical HR components to viral replication centers and enhances cytotoxicity. IMPLICATIONS: Oncolytic adenoviral therapy may be most clinically relevant in tumors with intact HR function.


Sujet(s)
Adenoviridae/physiologie , Protéine BRCA2/métabolisme , Recombinaison homologue , Tumeurs de l'ovaire/métabolisme , Rad51 Recombinase/métabolisme , Adenoviridae/génétique , Protéines E2 d'adénovirus/métabolisme , Lignée cellulaire tumorale , Prolifération cellulaire/effets des médicaments et des substances chimiques , Cisplatine/pharmacologie , Femelle , Vecteurs génétiques/pharmacologie , Humains , Thérapie virale de cancers , Réplication virale
5.
Cancer Res ; 75(14): 2811-21, 2015 Jul 15.
Article de Anglais | MEDLINE | ID: mdl-25977332

RÉSUMÉ

Adenoviruses have been clinically tested as anticancer therapies but their utility has been severely limited by rapid, systemic cytokine release and consequent inflammatory toxicity. Here, we describe a new approach to tackling these dangerous side effects. Using human ovarian cancer cell lines as well as malignant epithelial cells harvested from the ascites of women with ovarian cancer, we show that tumor cells do not produce cytokines in the first 24 hours following in vitro infection with the oncolytic adenovirus dl922-947. In contrast, dl922-947 does induce inflammatory cytokines at early time points following intraperitoneal delivery in mice with human ovarian cancer intraperitoneal xenografts. In these animals, cytokines originate predominantly in murine tissues, especially in macrophage-rich organs such as the spleen. We use a nonreplicating adenovirus to confirm that early cytokine production is independent of adenoviral replication. Using ß3 integrin knockout mice injected intraperitoneally with dl922-947 and ß3 null murine peritoneal macrophages, we confirm a role for macrophage cell surface ß3 integrin in this dl922-947-induced inflammation. We present new evidence that co-administration of a cyclic RGD-mimetic-specific inhibitor of ß3 integrin significantly attenuates the cytokine release and inflammatory hepatic toxicity induced by dl922-947 in an intraperitoneal murine model of ovarian cancer. Importantly, we find no evidence that ß3 inhibition compromises viral infectivity and oncolysis in vitro or anticancer efficacy in vivo. By enabling safe, systemic delivery of replicating adenoviruses, this novel approach could have a major impact on the future development of these effective anticancer agents.


Sujet(s)
Adenoviridae/immunologie , Anti-inflammatoires/pharmacologie , Inflammation/prévention et contrôle , Intégrine bêta3/métabolisme , Thérapie virale de cancers/effets indésirables , Virus oncolytiques/immunologie , Tumeurs de l'ovaire/thérapie , Peptides cycliques/pharmacologie , Adenoviridae/effets des médicaments et des substances chimiques , Animaux , Anti-inflammatoires/usage thérapeutique , Antinéoplasiques/immunologie , Antinéoplasiques/usage thérapeutique , Cellules cultivées , Association thérapeutique , Femelle , Humains , Intégrine bêta3/génétique , Souris , Souris de lignée C57BL , Souris knockout , Souris nude , Virus oncolytiques/effets des médicaments et des substances chimiques , Tumeurs de l'ovaire/immunologie , Tumeurs de l'ovaire/anatomopathologie , Peptides cycliques/usage thérapeutique , Tests d'activité antitumorale sur modèle de xénogreffe
6.
Mol Oncol ; 9(4): 791-805, 2015 Apr.
Article de Anglais | MEDLINE | ID: mdl-25560085

RÉSUMÉ

Resistance to paclitaxel chemotherapy frequently develops in ovarian cancer. Oncolytic adenoviruses are a novel therapy for human malignancies that are being evaluated in early phase trials. However, there are no reliable predictive biomarkers for oncolytic adenovirus activity in ovarian cancer. We investigated the link between paclitaxel resistance and oncolytic adenovirus activity using established ovarian cancer cell line models, xenografts with de novo paclitaxel resistance and tumour samples from two separate trials. The activity of multiple Ad5 vectors, including dl922-947 (E1A CR2-deleted), dl1520 (E1B-55K deleted) and Ad5 WT, was significantly increased in paclitaxel resistant ovarian cancer in vitro and in vivo. This was associated with greater infectivity resulting from increased expression of the primary receptor for Ad5, CAR (coxsackie adenovirus receptor). This, in turn, resulted from increased CAR transcription secondary to histone modification in resistant cells. There was increased CAR expression in intraperitoneal tumours with de novo paclitaxel resistance and in tumours from patients with clinical resistance to paclitaxel. Increased CAR expression did not cause paclitaxel resistance, but did increase inflammatory cytokine expression. Finally, we identified dysregulated cell cycle control as a second mechanism of increased adenovirus efficacy in paclitaxel-resistant ovarian cancer. Ad11 and Ad35, both group B adenoviruses that utilise non-CAR receptors to infect cells, are also significantly more effective in paclitaxel-resistant ovarian cell models. Inhibition of CDK4/6 using PD-0332991 was able both to reverse paclitaxel resistance and reduce adenovirus efficacy. Thus, paclitaxel resistance increases oncolytic adenovirus efficacy via at least two separate mechanisms - if validated further, this information could have future clinical utility to aid patient selection for clinical trials.


Sujet(s)
Adenoviridae/métabolisme , Points de contrôle du cycle cellulaire/effets des médicaments et des substances chimiques , Protéine membranaire apparentée au récepteur des coxsackievirus et adénovirus/métabolisme , Résistance aux médicaments antinéoplasiques/effets des médicaments et des substances chimiques , Virus oncolytiques/métabolisme , Paclitaxel/pharmacologie , Régulation positive/effets des médicaments et des substances chimiques , Animaux , Lignée cellulaire tumorale , Cytokines/métabolisme , Femelle , Histone/métabolisme , Humains , Médiateurs de l'inflammation/métabolisme , Souris nude , Tumeurs de l'ovaire/anatomopathologie , ARN messager/génétique , ARN messager/métabolisme , Tests d'activité antitumorale sur modèle de xénogreffe
7.
Mol Ther ; 20(9): 1676-88, 2012 Sep.
Article de Anglais | MEDLINE | ID: mdl-22735379

RÉSUMÉ

Adaptive immune responses may be vital in the overall efficacy of oncolytic viruses in human malignancies. However, immune responses to oncolytic adenoviruses are poorly understood because these viruses lack activity in murine cells, which precludes evaluation in immunocompetent murine cancer models. We have evaluated human adenovirus activity in murine cells. We show that a panel of murine carcinoma cells, including CMT64, MOVCAR7, and MOSEC/ID8, can readily be infected with human adenovirus. These cells also support viral gene transcription, messenger RNA (mRNA) processing, and genome replication. However, there is a profound failure of adenovirus protein synthesis, especially late structural proteins, both in vitro and in vivo, with reduced loading of late mRNA onto ribosomes. Our data also show that in trans expression of the nonstructural late protein L4-100K increases both the amount of viral mRNA on ribosomes and the synthesis of late proteins, accompanied by reduced phosphorylation of eIF2α and improved anticancer efficacy. These results suggest that murine models that support human adenovirus replication could be generated, thus allowing evaluation of human adenoviruses in immunocompetent mice.


Sujet(s)
Adénovirus humains/génétique , Virus oncolytiques/génétique , Tumeurs de l'ovaire/thérapie , Biosynthèse des protéines , ARN viral/métabolisme , Protéines virales non structurales/génétique , Immunité acquise , Adénovirus humains/immunologie , Animaux , Lignée cellulaire tumorale , Facteur-2 d'initiation eucaryote/génétique , Facteur-2 d'initiation eucaryote/métabolisme , Femelle , Expression des gènes , Humains , Souris , Virus oncolytiques/immunologie , Tumeurs de l'ovaire/immunologie , Tumeurs de l'ovaire/anatomopathologie , Ovaire/effets des médicaments et des substances chimiques , Ovaire/immunologie , Ovaire/anatomopathologie , ARN messager/génétique , ARN messager/métabolisme , ARN viral/génétique , Ribosomes/génétique , Ribosomes/métabolisme , Spécificité d'espèce , Protéines virales non structurales/métabolisme , Réplication virale/génétique
8.
J Clin Invest ; 121(4): 1283-97, 2011 Apr.
Article de Anglais | MEDLINE | ID: mdl-21383502

RÉSUMÉ

Oncolytic adenoviruses replicate selectively within and lyse malignant cells. As such, they are being developed as anticancer therapeutics. However, the sensitivity of ovarian cancers to adenovirus cytotoxicity varies greatly, even in cells of similar infectivity. Using both the adenovirus E1A-CR2 deletion mutant dl922-947 and WT adenovirus serotype 5 in a panel of human ovarian cancer cell lines that cover a 3-log range of sensitivity, we observed profound overreplication of genomic DNA only in highly sensitive cell lines. This was associated with the presence of extensive genomic DNA damage. Inhibition of ataxia telangiectasia and Rad3-related checkpoint kinase 1 (ATR-Chk1), but not ataxia telangiectasia mutated (ATM), promoted genomic DNA damage and overreplication in resistant and partially sensitive cells. This was accompanied by increased adenovirus cytotoxicity both in vitro and in vivo in tumor-bearing mice. We also demonstrated that Cdc25A was upregulated in highly sensitive ovarian cancer cell lines after adenovirus infection and was stabilized after loss of Chk1 activity. Knockdown of Cdc25A inhibited virus-induced DNA damage in highly sensitive cells and blocked the effects of Chk1 inhibition in resistant cells. Finally, inhibition of Chk1 decreased homologous recombination repair of virus-induced genomic DNA double-strand breaks. Thus, virus-induced host cell DNA damage signaling and repair are key determinants of oncolytic adenoviral activity, and promoting unscheduled DNA synthesis and/or impeding homologous recombination repair could potentiate the effects of oncolytic adenoviruses in the treatment of ovarian cancer.


Sujet(s)
Protéines du cycle cellulaire/métabolisme , Altération de l'ADN , Thérapie virale de cancers , Virus oncolytiques/génétique , Virus oncolytiques/physiologie , Tumeurs de l'ovaire/thérapie , Protein kinases/métabolisme , Protein-Serine-Threonine Kinases/métabolisme , Animaux , Protéines mutées dans l'ataxie-télangiectasie , Lignée cellulaire tumorale , Checkpoint kinase 1 , Réparation de l'ADN , Réplication de l'ADN , Femelle , Humains , Souris , Souris nude , Mutation , Transplantation tumorale , Tumeurs de l'ovaire/génétique , Tumeurs de l'ovaire/métabolisme , Transduction du signal , Transplantation hétérologue , cdc25 Phosphatases/métabolisme
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...