Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 88
Filtrer
1.
Cancers (Basel) ; 13(23)2021 Nov 30.
Article de Anglais | MEDLINE | ID: mdl-34885153

RÉSUMÉ

The poly(ADP-ribose) binding protein CHFR regulates cellular responses to mitotic stress. The deubiquitinase UBC13, which regulates CHFR levels, has been associated with better overall survival in paclitaxel-treated ovarian cancer. Despite the extensive use of taxanes in the treatment of ovarian cancer, little is known about expression of CHFR itself in this disease. In the present study, tissue microarrays containing ovarian carcinoma samples from 417 women who underwent initial surgical debulking were stained with anti-CHFR antibody and scored in a blinded fashion. CHFR levels, expressed as a modified H-score, were examined for association with histology, grade, time to progression (TTP) and overall survival (OS). In addition, patient-derived xenografts from 69 ovarian carcinoma patients were examined for CHFR expression and sensitivity to paclitaxel monotherapy. In clinical ovarian cancer specimens, CHFR expression was positively associated with serous histology (p = 0.0048), higher grade (p = 0.000014) and higher stage (p = 0.016). After correction for stage and debulking, there was no significant association between CHFR staining and overall survival (p = 0.62) or time to progression (p = 0.91) in patients with high grade serous cancers treated with platinum/taxane chemotherapy (N = 249). Likewise, no association between CHFR expression and paclitaxel sensitivity was observed in ovarian cancer PDXs treated with paclitaxel monotherapy. Accordingly, differences in CHFR expression are unlikely to play a major role in paclitaxel sensitivity of high grade serous ovarian cancer.

2.
Nat Commun ; 11(1): 5173, 2020 10 14.
Article de Anglais | MEDLINE | ID: mdl-33057068

RÉSUMÉ

In ovarian cancer (OC), IL-17-producing T cells (Th17s) predict improved survival, whereas regulatory T cells predict poorer survival. We previously developed a vaccine whereby patient-derived dendritic cells (DCs) are programmed to induce Th17 responses to the OC antigen folate receptor alpha (FRα). Here we report the results of a single-arm open-label phase I clinical trial designed to determine vaccine safety and tolerability (primary outcomes) and recurrence-free survival (secondary outcome). Immunogenicity is also evaluated. Recruitment is complete with a total of 19 Stage IIIC-IV OC patients in first remission after conventional therapy. DCs are generated using our Th17-inducing protocol and are pulsed with HLA class II epitopes from FRα. Mature antigen-loaded DCs are injected intradermally. All patients have completed study-related interventions. No grade 3 or higher adverse events are seen. Vaccination results in the development of Th1, Th17, and antibody responses to FRα in the majority of patients. Th1 and antibody responses are associated with prolonged recurrence-free survival. Antibody-dependent cell-mediated cytotoxic activity against FRα is also associated with prolonged RFS. Of 18 patients evaluable for efficacy, 39% (7/18) remain recurrence-free at the time of data censoring, with a median follow-up of 49.2 months. Thus, vaccination with Th17-inducing FRα-loaded DCs is safe, induces antigen-specific immunity, and is associated with prolonged remission.


Sujet(s)
Vaccins anticancéreux/administration et posologie , Cellules dendritiques/transplantation , Récidive tumorale locale/épidémiologie , Tumeurs de l'ovaire/thérapie , Cellules Th17/immunologie , Sujet âgé , Vaccins anticancéreux/effets indésirables , Vaccins anticancéreux/immunologie , Survie sans rechute , Femelle , Récepteur-1 des folates/immunologie , Humains , Immunité humorale , Injections intradermiques , Interféron gamma/métabolisme , Interleukine-17/métabolisme , Adulte d'âge moyen , Récidive tumorale locale/immunologie , Récidive tumorale locale/prévention et contrôle , Tumeurs de l'ovaire/immunologie , Tumeurs de l'ovaire/mortalité , Cellules Th17/métabolisme , Transplantation autologue/effets indésirables , Transplantation autologue/méthodes
3.
Nat Commun ; 10(1): 3935, 2019 09 02.
Article de Anglais | MEDLINE | ID: mdl-31477716

RÉSUMÉ

Mucinous ovarian carcinoma (MOC) is a unique subtype of ovarian cancer with an uncertain etiology, including whether it genuinely arises at the ovary or is metastatic disease from other organs. In addition, the molecular drivers of invasive progression, high-grade and metastatic disease are poorly defined. We perform genetic analysis of MOC across all histological grades, including benign and borderline mucinous ovarian tumors, and compare these to tumors from other potential extra-ovarian sites of origin. Here we show that MOC is distinct from tumors from other sites and supports a progressive model of evolution from borderline precursors to high-grade invasive MOC. Key drivers of progression identified are TP53 mutation and copy number aberrations, including a notable amplicon on 9p13. High copy number aberration burden is associated with worse prognosis in MOC. Our data conclusively demonstrate that MOC arise from benign and borderline precursors at the ovary and are not extra-ovarian metastases.


Sujet(s)
Adénocarcinome mucineux/génétique , Carcinome épithélial de l'ovaire/génétique , Analyse de profil d'expression de gènes/méthodes , Tumeurs de l'ovaire/génétique , Adénocarcinome mucineux/classification , Adénocarcinome mucineux/métabolisme , Carcinome épithélial de l'ovaire/classification , Carcinome épithélial de l'ovaire/métabolisme , Études de cohortes , Femelle , Régulation de l'expression des gènes tumoraux , Humains , Mutation , Tumeurs de l'ovaire/classification , Tumeurs de l'ovaire/métabolisme , Analyse de séquence d'ADN/méthodes , Analyse de survie
4.
Gynecol Oncol ; 154(3): 495-504, 2019 09.
Article de Anglais | MEDLINE | ID: mdl-31204077

RÉSUMÉ

OBJECTIVE: This study is designed to identify genes and pathways that could promote metastasis to the bowel in high-grade serous ovarian cancer (OC) and evaluate their associations with clinical outcomes. METHODS: We performed RNA sequencing of OC primary tumors (PTs) and their corresponding bowel metastases (n = 21 discovery set; n = 18 replication set). Differentially expressed genes (DEGs) were those expressed at least 2-fold higher in bowel metastases (BMets) than PTs in at least 30% of patients (P < .05) with no increased expression in paired benign bowel tissue and were validated with quantitative reverse transcription PCR. Using an independent OC cohort (n = 333), associations between DEGs in PTs and surgical and clinical outcomes were performed. Immunohistochemistry and mouse xenograft studies were performed to confirm the role of LRRC15 in promoting metastasis. RESULTS: Among 27 DEGs in the discovery set, 21 were confirmed in the replication set: SFRP2, Col11A1, LRRC15, ADAM12, ADAMTS12, MFAP5, LUM, PLPP4, FAP, POSTN, GRP, MMP11, MMP13, C1QTNF3, EPYC, DIO2, KCNA1, NETO1, NTM, MYH13, and PVALB. Higher expression of more than half of the genes in the PT was associated with an increased requirement for bowel resection at primary surgery and an inability to achieve complete cytoreduction. Increased expression of LRRC15 in BMets was confirmed by immunohistochemistry and knockdown of LRRC15 significantly inhibited tumor progression in mice. CONCLUSIONS: We identified 21 genes that are overexpressed in bowel metastases among patients with OC. Our findings will help select potential molecular targets for the prevention and treatment of malignant bowel obstruction in OC.


Sujet(s)
Carcinome épithélial de l'ovaire/génétique , Carcinome épithélial de l'ovaire/anatomopathologie , Tumeurs de l'intestin/génétique , Tumeurs de l'intestin/secondaire , Tumeurs de l'ovaire/génétique , Tumeurs de l'ovaire/anatomopathologie , Animaux , Lignée cellulaire tumorale , Études de cohortes , Femelle , Techniques de knock-down de gènes , Hétérogreffes , Séquençage nucléotidique à haut débit , Humains , Protéines membranaires/génétique , Souris , Souris nude , ARN tumoral/génétique , Transcriptome , Régulation positive
5.
Cancer Epidemiol Biomarkers Prev ; 27(9): 1101-1109, 2018 09.
Article de Anglais | MEDLINE | ID: mdl-29967001

RÉSUMÉ

Background: Endometrioid carcinoma (EC) and clear cell carcinoma (CC) histotypes of epithelial ovarian cancer are understudied compared with the more common high-grade serous carcinomas (HGSC). We therefore sought to characterize EC and CC transcriptomes in relation to HGSC.Methods: Following bioinformatics processing and gene abundance normalization, differential expression analysis of RNA sequence data collected on fresh-frozen tumors was completed with nonparametric statistical analysis methods (55 ECs, 19 CCs, 112 HGSCs). Association of gene expression with progression-free survival (PFS) was completed with Cox proportional hazards models. Eight additional multi-histotype expression array datasets (N = 852 patients) were used for replication.Results: In the discovery set, tumors generally clustered together by histotype. Thirty-two protein-coding genes were differentially expressed across histotype (P < 1 × 10-10) and showed similar associations in replication datasets, including MAP2K6, KIAA1324, CDH1, ENTPD5, LAMB1, and DRAM1 Nine genes associated with PFS (P < 0.0001) showed similar associations in replication datasets. In particular, we observed shorter PFS time for CC and EC patients with high gene expression for CCNB2, CORO2A, CSNK1G1, FRMD8, LIN54, LINC00664, PDK1, and PEX6, whereas, the converse was observed for HGSC patients.Conclusions: The results suggest important histotype differences that may aid in the development of treatment options, particularly those for patients with EC or CC.Impact: We present replicated findings on transcriptomic differences and how they relate to clinical outcome for two of the rarer ovarian cancer histotypes of EC and CC, along with comparison with the common histotype of HGSC. Cancer Epidemiol Biomarkers Prev; 27(9); 1101-9. ©2018 AACR.


Sujet(s)
Adénocarcinome à cellules claires/génétique , Marqueurs biologiques tumoraux/génétique , Carcinome endométrioïde/génétique , Cystadénocarcinome séreux/génétique , Tumeurs de l'ovaire/génétique , Transcriptome , Adénocarcinome à cellules claires/anatomopathologie , Adulte , Sujet âgé , Sujet âgé de 80 ans ou plus , Carcinome endométrioïde/anatomopathologie , Cystadénocarcinome séreux/anatomopathologie , Femelle , Études de suivi , Humains , Adulte d'âge moyen , Tumeurs de l'ovaire/anatomopathologie , Pronostic , Taux de survie , Jeune adulte
6.
Clin Cancer Res ; 24(13): 3014-3025, 2018 07 01.
Article de Anglais | MEDLINE | ID: mdl-29545464

RÉSUMÉ

Purpose: Folate receptor alpha (FR) is overexpressed in several cancers. Endogenous immunity to the FR has been demonstrated in patients and suggests the feasibility of targeting FR with vaccine or other immune therapies. CD4 helper T cells are central to the development of coordinated immunity, and prior work shows their importance in protecting against relapse. Our previous identification of degenerate HLA-class II epitopes from human FR led to the development of a broad coverage epitope pool potentially useful in augmenting antigen-specific immune responses in most patients.Patients and Methods: We conducted a phase I clinical trial testing safety and immunogenicity of this vaccine, enrolling patients with ovarian cancer or breast cancer who completed conventional treatment and who showed no evidence of disease. Patients were initially treated with low-dose cyclophosphamide and then vaccinated 6 times, monthly. Immunity and safety were examined during the vaccine period and up to 1 year later.Results: Vaccination was well tolerated in all patients. Vaccine elicited or augmented immunity in more than 90% of patients examined. Unlike recall immunity to tetanus toxoid (TT), FR T-cell responses developed slowly over the course of vaccination with a median time to maximal immunity in 5 months. Despite slow development of immunity, responsiveness appeared to persist for at least 12 months.Conclusions: The results demonstrate that it is safe to augment immunity to the FR tumor antigen, and the developed vaccine is testable for therapeutic activity in most patients whose tumors express FR, regardless of HLA genotype. Clin Cancer Res; 24(13); 3014-25. ©2018 AACR.


Sujet(s)
Tumeurs du sein/immunologie , Tumeurs du sein/thérapie , Vaccins anticancéreux/immunologie , Récepteur-1 des folates/immunologie , Tumeurs de l'ovaire/immunologie , Tumeurs de l'ovaire/thérapie , Vaccins sous-unitaires/immunologie , Adulte , Sujet âgé , Séquence d'acides aminés , Tumeurs du sein/diagnostic , Vaccins anticancéreux/administration et posologie , Association thérapeutique , Cytokines/métabolisme , Épitopes/composition chimique , Épitopes/immunologie , Femelle , Antigènes d'histocompatibilité de classe II/immunologie , Humains , Immunité , Immunogénicité des vaccins , Numération des lymphocytes , Adulte d'âge moyen , Métastase tumorale , Stadification tumorale , Tumeurs de l'ovaire/diagnostic , Peptides/composition chimique , Peptides/immunologie , Sous-populations de lymphocytes T/immunologie , Sous-populations de lymphocytes T/métabolisme , Résultat thérapeutique , Vaccination , Vaccins sous-unitaires/administration et posologie
7.
Mayo Clin Proc ; 93(3): 307-320, 2018 03.
Article de Anglais | MEDLINE | ID: mdl-29502561

RÉSUMÉ

OBJECTIVE: To evaluate myeloid differentiation primary response gene 88 (MyD88) and Toll-like receptor 4 (TLR4) expression in relation to clinical features of epithelial ovarian cancer, histologic subtypes, and overall survival. PATIENTS AND METHODS: We conducted centralized immunohistochemical staining, semi-quantitative scoring, and survival analysis in 5263 patients participating in the Ovarian Tumor Tissue Analysis consortium. Patients were diagnosed between January 1, 1978, and December 31, 2014, including 2865 high-grade serous ovarian carcinomas (HGSOCs), with more than 12,000 person-years of follow-up time. Tissue microarrays were stained for MyD88 and TLR4, and staining intensity was classified using a 2-tiered system for each marker (weak vs strong). RESULTS: Expression of MyD88 and TLR4 was similar in all histotypes except clear cell ovarian cancer, which showed reduced expression compared with other histotypes (P<.001 for both). In HGSOC, strong MyD88 expression was modestly associated with shortened overall survival (hazard ratio [HR], 1.13; 95% CI, 1.01-1.26; P=.04) but was also associated with advanced stage (P<.001). The expression of TLR4 was not associated with survival. In low-grade serous ovarian cancer (LGSOC), strong expression of both MyD88 and TLR4 was associated with favorable survival (HR [95% CI], 0.49 [0.29-0.84] and 0.44 [0.21-0.89], respectively; P=.009 and P=.02, respectively). CONCLUSION: Results are consistent with an association between strong MyD88 staining and advanced stage and poorer survival in HGSOC and demonstrate correlation between strong MyD88 and TLR4 staining and improved survival in LGSOC, highlighting the biological differences between the 2 serous histotypes.


Sujet(s)
Carcinome épithélial de l'ovaire/métabolisme , Facteur de différenciation myéloïde-88/métabolisme , Tumeurs de l'ovaire/métabolisme , Récepteur de type Toll-4/métabolisme , Adulte , Sujet âgé , Marqueurs biologiques tumoraux/métabolisme , Carcinome épithélial de l'ovaire/mortalité , Femelle , Humains , Immunohistochimie/méthodes , Adulte d'âge moyen , Tumeurs de l'ovaire/mortalité , Analyse de survie , Analyse sur puce à tissus/méthodes
8.
Front Oncol ; 8: 58, 2018.
Article de Anglais | MEDLINE | ID: mdl-29594039

RÉSUMÉ

BACKGROUND: Most pelvic high-grade serous (HGS) carcinomas have been proposed to arise from tubal primaries that progress rapidly to advanced disease. However, the temporal sequence of ovarian and peritoneal metastases is not well characterized. METHODS: To establish the sequence of metastases, phylogenetic relationships among the ovarian and peritoneal carcinomas were determined from single-nucleotide variations (SNVs) in nine tumor regions from each patient with pelvic HGS carcinomas. Somatic SNVs from each tumor sample were used to reconstruct phylogenies of samples from each patient. Variant allele frequencies were used to reconstruct subclone phylogenies in each tumor sample. RESULTS: We show that pelvic HGS carcinomas are highly heterogeneous, only sharing less than 4% of somatic SNVs among all nine carcinoma implants in one patient. TP53 mutations are found in all nine carcinoma implants in each patient. The phylogenetic analyses reveal that peritoneal metastases arose from early branching events that preceded branching events for ovarian carcinomas in some patients. Finally, subclone phylogenies indicate the presence of multiple subclones at each tumor implant and early tumor clones in peritoneal implants. CONCLUSION: The genetic evidence that peritoneal implants arose before or concurrently with ovarian implants is consistent with the emerging concept of the extra-ovarian origin of pelvic HGS cancer. Our results challenge the concept of stepwise spatial progression from the fallopian primary to ovarian carcinomas to peritoneal dissemination and suggest an alternative progression model where peritoneal spreading of early clones occurs before or in parallel with ovarian metastases.

9.
Cancer Inform ; 17: 1176935118755341, 2018.
Article de Anglais | MEDLINE | ID: mdl-29434467

RÉSUMÉ

High-grade serous ovarian cancer (HGSOC) is a complex disease in which initiation and progression have been associated with copy number alterations, epigenetic processes, and, to a lesser extent, germline variation. We hypothesized that, when summarized at the gene level, tumor methylation and germline genetic variation, alone or in combination, influence tumor gene expression in HGSOC. We used Elastic Net (ENET) penalized regression method to evaluate these associations and adjust for somatic copy number in 3 independent data sets comprising tumors from more than 470 patients. Penalized regression models of germline variation, with or without methylation, did not reveal a role in HGSOC gene expression. However, we observed significant association between regional methylation and expression of 5 genes (WDPCP, KRT6C, BRCA2, EFCAB13, and ZNF283). CpGs retained in ENET model for BRCA2 and ZNF283 appeared enriched in several regulatory elements, suggesting that regularized regression may provide a novel utility for integrative genomic analysis.

10.
Genet Epidemiol ; 41(8): 898-914, 2017 12.
Article de Anglais | MEDLINE | ID: mdl-29119601

RÉSUMÉ

X-chromosome inactivation (XCI) epigenetically silences transcription of an X chromosome in females; patterns of XCI are thought to be aberrant in women's cancers, but are understudied due to statistical challenges. We develop a two-stage statistical framework to assess skewed XCI and evaluate gene-level patterns of XCI for an individual sample by integration of RNA sequence, copy number alteration, and genotype data. Our method relies on allele-specific expression (ASE) to directly measure XCI and does not rely on male samples or paired normal tissue for comparison. We model ASE using a two-component mixture of beta distributions, allowing estimation for a given sample of the degree of skewness (based on a composite likelihood ratio test) and the posterior probability that a given gene escapes XCI (using a Bayesian beta-binomial mixture model). To illustrate the utility of our approach, we applied these methods to data from tumors of ovarian cancer patients. Among 99 patients, 45 tumors were informative for analysis and showed evidence of XCI skewed toward a particular parental chromosome. For 397 X-linked genes, we observed tumor XCI patterns largely consistent with previously identified consensus states based on multiple normal tissue types. However, 37 genes differed in XCI state between ovarian tumors and the consensus state; 17 genes aberrantly escaped XCI in ovarian tumors (including many oncogenes), whereas 20 genes were unexpectedly inactivated in ovarian tumors (including many tumor suppressor genes). These results provide evidence of the importance of XCI in ovarian cancer and demonstrate the utility of our two-stage analysis.


Sujet(s)
Tumeurs épithéliales épidermoïdes et glandulaires/génétique , Tumeurs de l'ovaire/génétique , Adulte , Allèles , Théorème de Bayes , Carcinome épithélial de l'ovaire , Chromosomes X humains , Femelle , Gènes liés au chromosome X , Génotype , Humains , Modèles génétiques , Tumeurs épithéliales épidermoïdes et glandulaires/anatomopathologie , Tumeurs de l'ovaire/anatomopathologie , Polymorphisme de nucléotide simple , ARN tumoral/composition chimique , ARN tumoral/isolement et purification , ARN tumoral/métabolisme , Analyse de séquence d'ARN , Inactivation du chromosome X
11.
JAMA Oncol ; 3(12): e173290, 2017 12 01.
Article de Anglais | MEDLINE | ID: mdl-29049607

RÉSUMÉ

Importance: Cytotoxic CD8+ tumor-infiltrating lymphocytes (TILs) participate in immune control of epithelial ovarian cancer; however, little is known about prognostic patterns of CD8+ TILs by histotype and in relation to other clinical factors. Objective: To define the prognostic role of CD8+ TILs in epithelial ovarian cancer. Design, Setting, and Participants: This was a multicenter observational, prospective survival cohort study of the Ovarian Tumor Tissue Analysis Consortium. More than 5500 patients, including 3196 with high-grade serous ovarian carcinomas (HGSOCs), were followed prospectively for over 24 650 person-years. Exposures: Following immunohistochemical analysis, CD8+ TILs were identified within the epithelial components of tumor islets. Patients were grouped based on the estimated number of CD8+ TILs per high-powered field: negative (none), low (1-2), moderate (3-19), and high (≥20). CD8+ TILs in a subset of patients were also assessed in a quantitative, uncategorized manner, and the functional form of associations with survival was assessed using penalized B-splines. Main Outcomes and Measures: Overall survival time. Results: The final sample included 5577 women; mean age at diagnosis was 58.4 years (median, 58.2 years). Among the 5 major invasive histotypes, HGSOCs showed the most infiltration. CD8+ TILs in HGSOCs were significantly associated with longer overall survival; median survival was 2.8 years for patients with no CD8+ TILs and 3.0 years, 3.8 years, and 5.1 years for patients with low, moderate, or high levels of CD8+ TILs, respectively (P value for trend = 4.2 × 10−16). A survival benefit was also observed among women with endometrioid and mucinous carcinomas, but not for those with the other histotypes. Among HGSOCs, CD8+ TILs were favorable regardless of extent of residual disease following cytoreduction, known standard treatment, and germline BRCA1 pathogenic mutation, but were not prognostic for BRCA2 mutation carriers. Evaluation of uncategorized CD8+ TIL counts showed a near-log-linear functional form. Conclusions and Relevance: This study demonstrates the histotype-specific nature of immune infiltration and provides definitive evidence for a dose-response relationship between CD8+ TILs and HGSOC survival. That the extent of infiltration is prognostic, not merely its presence or absence, suggests that understanding factors that drive infiltration will be the key to unraveling outcome heterogeneity in this cancer.


Sujet(s)
Antigènes CD8/métabolisme , Carcinome épithélial de l'ovaire/traitement médicamenteux , Cystadénocarcinome séreux/immunologie , Lymphocytes TIL/immunologie , Tumeurs de l'ovaire/immunologie , Protéine BRCA2/génétique , Carcinome épithélial de l'ovaire/immunologie , Carcinome épithélial de l'ovaire/anatomopathologie , Études de cohortes , Cystadénocarcinome séreux/génétique , Cystadénocarcinome séreux/anatomopathologie , Femelle , Humains , Adulte d'âge moyen , Mutation , Grading des tumeurs , Tumeurs de l'ovaire/génétique , Tumeurs de l'ovaire/anatomopathologie , Études prospectives , Analyse de survie , Résultat thérapeutique
12.
Cancer Res ; 77(23): 6667-6678, 2017 12 01.
Article de Anglais | MEDLINE | ID: mdl-28993412

RÉSUMÉ

Ligation of programmed cell death-1 (PD-1) in the tumor microenvironment is known to inhibit effective adaptive antitumor immunity. Blockade of PD-1 in humans has resulted in impressive, durable regression responses in select tumor types. However, durable responses have been elusive in ovarian cancer patients. PD-1 was recently shown to be expressed on and thereby impair the functions of tumor-infiltrating murine and human myeloid dendritic cells (TIDC) in ovarian cancer. In the present work, we characterize the regulation of PD-1 expression and the effects of PD-1 blockade on TIDC. Treatment of TIDC and bone marrow-derived dendritic cells (DC) with IL10 led to increased PD-1 expression. Both groups of DCs also responded to PD-1 blockade by increasing production of IL10. Similarly, treatment of ovarian tumor-bearing mice with PD-1 blocking antibody resulted in an increase in IL10 levels in both serum and ascites. While PD-1 blockade or IL10 neutralization as monotherapies were inefficient, combination of these two led to improved survival and delayed tumor growth; this was accompanied by augmented antitumor T- and B-cell responses and decreased infiltration of immunosuppressive MDSC. Taken together, our findings implicate compensatory release of IL10 as one of the adaptive resistance mechanisms that undermine the efficacy of anti-PD-1 (or anti-PD-L1) monotherapies and prompt further studies aimed at identifying such resistance mechanisms. Cancer Res; 77(23); 6667-78. ©2017 AACR.


Sujet(s)
Interleukine-10/métabolisme , Interleukine-10/pharmacologie , Tumeurs de l'ovaire/immunologie , Récepteur-1 de mort cellulaire programmée/antagonistes et inhibiteurs , Récepteur-1 de mort cellulaire programmée/biosynthèse , Animaux , Lymphocytes B/immunologie , Lignée cellulaire tumorale , Cellules dendritiques/immunologie , Résistance aux médicaments antinéoplasiques , Femelle , Humains , Souris , Souris de lignée C57BL , Tumeurs de l'ovaire/traitement médicamenteux , Interférence par ARN , Petit ARN interférent/génétique , Facteur de transcription STAT-3/antagonistes et inhibiteurs , Facteur de transcription STAT-3/génétique , Lymphocytes T/immunologie , Microenvironnement tumoral/immunologie
13.
Genes Cancer ; 8(5-6): 589-599, 2017 May.
Article de Anglais | MEDLINE | ID: mdl-28740577

RÉSUMÉ

BACKGROUND: Limited effectiveness of therapeutic agents targeting epidermal growth factor receptor (EGFR) in clinical trials using unselected ovarian cancer patients has prompted efforts to more effectively stratify patients who might best benefit from these therapies. A series of studies that have evaluated immunohistochemical (IHC) staining of EGFR in ovarian cancer biopsies has produced unclear results as to the utility of this measure as a prognostic biomarker. Here, we used one of the largest, single institution cohorts to date to determine possible associations of EGFR expression with patient outcome. METHODS: We performed IHC staining of EGFR in tissue microarrays including nearly 500 patient tumor samples. Staining was classified by subcellular localization (membranous, cytoplasmic) or by automated image analysis algorithms. We also performed a literature review to place these results in the context of previous studies. RESULTS: No significant associations were found between EGFR subcellular localization or expression and histology, stage, grade, or outcome. These results were broadly consistent with the consensus of the reviewed literature. CONCLUSIONS: These results suggest that IHC staining for EGFR may not be a useful prognostic biomarker for ovarian cancer patients. Future studies should pursue other staining methods or analysis in combination with other pathway mediators.

14.
Anticancer Res ; 37(7): 3673-3677, 2017 07.
Article de Anglais | MEDLINE | ID: mdl-28668859

RÉSUMÉ

AIM: To identify preoperative nutritional and inflammatory markers that predict perioperative outcomes in patients with ovarian cancer (OC). PATIENTS AND METHODS: Fifty patients who underwent primary debulking for advanced (stage III/IV) OC were selected from a cohort of patients who underwent surgery between 2002 and 2009. We analyzed C-reactive protein (CRP), interleukin-6 (IL6) and albumin and their impact on mortality and surgical outcomes. RESULTS: Two patients were excluded since they did not have adequate measurements of CRP and IL6. Among the remaining patients, 25 (52%) were ≥70 years old. Nine (19%), 12 (25%) and 12 (25%) patients had low serum albumin (<3.0 g/dl), elevated CRP (≥70 mg/l) and elevated IL6 (≥24 pg/ml), respectively. Age was a significant predictor of non-home discharge (p=0.01). Low serum albumin (<3.0 g/dl) was a predictor of death within 6 month (p=0.03). Elevated CRP (≥70 mg/l) was a predictor of non-home discharge (p=0.02), death within 6 months (p=0.02), death within 12 months (p=0.04), and longer hospital stay (p=0.01). Elevated IL6 (≥24 pg/ml) was a predictor of non-home discharge (p=0.002) and surgical complications (p=0.02), and also associated with longer hospital stay (p=0.03). CONCLUSION: Poor nutrition and high inflammatory status negatively influence surgical and oncological outcomes of patients with OC. These preoperative markers can be used for selection of patients for neoadjuvant chemotherapy at high risk of short survival, non-home discharge and long hospital stay.


Sujet(s)
Marqueurs biologiques/sang , Inflammation/sang , État nutritionnel/physiologie , Tumeurs de l'ovaire/sang , Tumeurs de l'ovaire/chirurgie , Sujet âgé , Marqueurs biologiques/métabolisme , Protéine C-réactive/métabolisme , Femelle , Humains , Inflammation/métabolisme , Interleukine-6/sang , Interleukine-6/métabolisme , Tumeurs de l'ovaire/métabolisme , Période périopératoire , Projets pilotes , Sérumalbumine/métabolisme
15.
Neoplasia ; 19(8): 628-636, 2017 Aug.
Article de Anglais | MEDLINE | ID: mdl-28658648

RÉSUMÉ

Interest in preclinical drug development for ovarian cancer has stimulated development of patient-derived xenograft (PDX) or tumorgraft models. However, the unintended formation of human lymphoma in severe combined immunodeficiency (SCID) mice from Epstein-Barr virus (EBV)-infected human lymphocytes can be problematic. In this study, we have characterized ovarian cancer PDXs which developed human lymphomas and explore methods to suppress lymphoproliferative growth. Fresh human ovarian tumors from 568 patients were transplanted intraperitoneally in SCID mice. A subset of PDX models demonstrated atypical patterns of dissemination with mediastinal masses, hepatosplenomegaly, and CD45-positive lymphoblastic atypia without ovarian tumor engraftment. Expression of human CD20 but not CD3 supported a B-cell lineage, and EBV genomes were detected in all lymphoproliferative tumors. Immunophenotyping confirmed monoclonal gene rearrangements consistent with B-cell lymphoma, and global gene expression patterns correlated well with other human lymphomas. The ability of rituximab, an anti-CD20 antibody, to suppress human lymphoproliferation from a patient's ovarian tumor in SCID mice and prevent growth of an established lymphoma led to a practice change with a goal to reduce the incidence of lymphomas. A single dose of rituximab during the primary tumor heterotransplantation process reduced the incidence of CD45-positive cells in subsequent PDX lines from 86.3% (n = 117 without rituximab) to 5.6% (n = 160 with rituximab), and the lymphoma rate declined from 11.1% to 1.88%. Taken together, investigators utilizing PDX models for research should routinely monitor for lymphoproliferative tumors and consider implementing methods to suppress their growth.


Sujet(s)
Syndromes lymphoprolifératifs/étiologie , Syndromes lymphoprolifératifs/prévention et contrôle , Seconde tumeur primitive/étiologie , Seconde tumeur primitive/prévention et contrôle , Tumeurs de l'ovaire/anatomopathologie , Animaux , Antinéoplasiques immunologiques/pharmacologie , Biopsie , Évolution clonale , Modèles animaux de maladie humaine , Infections à virus Epstein-Barr/complications , Infections à virus Epstein-Barr/virologie , Femelle , Analyse de profil d'expression de gènes , Hétérogreffes , Humains , Immunohistochimie , Lymphocytes/immunologie , Lymphocytes/métabolisme , Lymphocytes/anatomopathologie , Syndromes lymphoprolifératifs/traitement médicamenteux , Souris , Seconde tumeur primitive/traitement médicamenteux , Rituximab/pharmacologie
16.
Oncotarget ; 8(29): 46891-46899, 2017 Jul 18.
Article de Anglais | MEDLINE | ID: mdl-28423358

RÉSUMÉ

Gene fusions play a critical role in some cancers and can serve as important clinical targets. In epithelial ovarian cancer (EOC), the contribution of fusions, especially by histological type, is unclear. We therefore screened for recurrent fusions in a histologically diverse panel of 220 EOCs using RNA sequencing. The Pipeline for RNA-Sequencing Data Analysis (PRADA) was used to identify fusions and allow for comparison with The Cancer Genome Atlas (TCGA) tumors. Associations between fusions and clinical prognosis were evaluated using Cox proportional hazards regression models. Nine recurrent fusions, defined as occurring in two or more tumors, were observed. CRHR1-KANSL1 was the most frequently identified fusion, identified in 6 tumors (2.7% of all tumors). This fusion was not associated with survival; other recurrent fusions were too rare to warrant survival analyses. One recurrent in-frame fusion, UBAP1-TGM7, was unique to clear cell (CC) EOC tumors (in 10%, or 2 of 20 CC tumors). We found some evidence that CC tumors harbor more fusions on average than any other EOC histological type, including high-grade serous (HGS) tumors. CC tumors harbored a mean of 7.4 fusions (standard deviation [sd] = 7.4, N = 20), compared to HGS EOC tumors mean of 2.0 fusions (sd = 3.3, N = 141). Few fusion genes were detected in endometrioid tumors (mean = 0.24, sd = 0.74, N = 55) or mucinous tumors (mean = 0.25, sd = 0.5, N = 4) tumors. To conclude, we identify one fusion at 10% frequency in the CC EOC subtype, but find little evidence for common (> 5% frequency) recurrent fusion genes in EOC overall, or in HGS subtype-specific EOC tumors.


Sujet(s)
Réarrangement des gènes , Études d'associations génétiques , Tumeurs épithéliales épidermoïdes et glandulaires/génétique , Tumeurs épithéliales épidermoïdes et glandulaires/anatomopathologie , Protéines de fusion oncogènes/génétique , Tumeurs de l'ovaire/génétique , Tumeurs de l'ovaire/anatomopathologie , Phénotype , Marqueurs biologiques tumoraux , Carcinome épithélial de l'ovaire , Femelle , Régulation de l'expression des gènes tumoraux , Humains , Grading des tumeurs , Pronostic , Translocation génétique
17.
Clin Cancer Res ; 23(15): 4077-4085, 2017 Aug 01.
Article de Anglais | MEDLINE | ID: mdl-28280090

RÉSUMÉ

Purpose: Here we assess whether molecular subtyping identifies biological features of tumors that correlate with survival and surgical outcomes of high-grade serous ovarian cancer (HGSOC).Experimental Design: Consensus clustering of pooled mRNA expression data from over 2,000 HGSOC cases was used to define molecular subtypes of HGSOCs. This de novo classification scheme was then applied to 381 Mayo Clinic HGSOC patients with detailed survival and surgical outcome information.Results: Five molecular subtypes of HGSOC were identified. In the pooled dataset, three subtypes were largely concordant with prior studies describing proliferative, mesenchymal, and immunoreactive tumors (concordance > 70%), and the group of tumors previously described as differentiated type was segregated into two new types, one of which (anti-mesenchymal) had downregulation of genes that were typically upregulated in the mesenchymal subtype. Molecular subtypes were significantly associated with overall survival (P < 0.001) and with rate of optimal surgical debulking (≤1 cm, P = 1.9E-4) in the pooled dataset. Among stage III-C or IV Mayo Clinic patients, molecular subtypes were also significantly associated with overall survival (P = 0.001), as well as rate of complete surgical debulking (no residual disease; 16% in mesenchymal tumors compared with >28% in other subtypes; P = 0.02).Conclusions: HGSOC tumors may be categorized into five molecular subtypes that associate with overall survival and the extent of residual disease following debulking surgery. Because mesenchymal tumors may have features that were associated with less favorable surgical outcome, molecular subtyping may have future utility in guiding neoadjuvant treatment decisions for women with HGSOC. Clin Cancer Res; 23(15); 4077-85. ©2017 AACR.


Sujet(s)
Cystadénocarcinome séreux/anatomopathologie , Protéines tumorales/génétique , Maladie résiduelle/anatomopathologie , Tumeurs de l'ovaire/anatomopathologie , Adulte , Sujet âgé , Sujet âgé de 80 ans ou plus , Cystadénocarcinome séreux/traitement médicamenteux , Cystadénocarcinome séreux/génétique , Cystadénocarcinome séreux/chirurgie , Survie sans rechute , Femelle , Régulation de l'expression des gènes tumoraux , Humains , Adulte d'âge moyen , Traitement néoadjuvant , Grading des tumeurs , Maladie résiduelle/traitement médicamenteux , Maladie résiduelle/génétique , Maladie résiduelle/chirurgie , Tumeurs de l'ovaire/traitement médicamenteux , Tumeurs de l'ovaire/génétique , Tumeurs de l'ovaire/chirurgie , Pronostic , Résultat thérapeutique
18.
Gynecol Oncol ; 143(2): 379-388, 2016 Nov.
Article de Anglais | MEDLINE | ID: mdl-27614696

RÉSUMÉ

OBJECTIVE: Poly(ADP-ribose) polymerase (PARP) inhibitors have yielded encouraging responses in high-grade serous ovarian carcinomas (HGSOCs), but the optimal treatment setting remains unknown. We assessed the effect of niraparib on HGSOC patient-derived xenograft (PDX) models as well as the relationship between certain markers of homologous recombination (HR) status, including BRCA1/2 mutations and formation of RAD51 foci after DNA damage, and response of these PDXs to niraparib in vivo. METHODS: Massively parallel sequencing was performed on HGSOCs to identify mutations contributing to HR deficiency. HR pathway integrity was assessed using fluorescence microscopy-based RAD51 focus formation assays. Effects of niraparib (MK-4827) on treatment-naïve PDX tumor growth as monotherapy, in combination with carboplatin/paclitaxel, and as maintenance therapy were assessed by transabdominal ultrasound. Niraparib responses were correlated with changes in levels of poly(ADP-ribose), PARP1, and repair proteins by western blotting. RESULTS: Five PDX models were evaluated in vivo. Tumor regressions were induced by single-agent niraparib in one of two PDX models with deleterious BRCA2 mutations and in a PDX with RAD51C promoter methylation. Diminished formation of RAD51 foci failed to predict response, but Artemis loss was associated with resistance. Niraparib generally failed to enhance responses to carboplatin/paclitaxel chemotherapy, but maintenance niraparib therapy delayed progression in a BRCA2-deficient PDX. CONCLUSIONS: Mutations in HR genes are neither necessary nor sufficient to predict response to niraparib. Assessment of repair status through multiple complementary assays is needed to guide PARP inhibitor therapy, design future clinical trials and identify ovarian cancer patients most likely to benefit from PARP inhibition.


Sujet(s)
Recombinaison homologue , Indazoles/usage thérapeutique , Tumeurs de l'ovaire/traitement médicamenteux , Pipéridines/usage thérapeutique , Inhibiteurs de poly(ADP-ribose) polymérases/usage thérapeutique , Protéines de liaison à l'ADN/analyse , Protéines de liaison à l'ADN/génétique , Femelle , Gène BRCA2 , Humains , Tumeurs de l'ovaire/génétique , Régions promotrices (génétique)
19.
Tumour Biol ; 37(10): 13279-13286, 2016 Oct.
Article de Anglais | MEDLINE | ID: mdl-27460076

RÉSUMÉ

The tumor-associated inflammatory microenvironment may play a pivotal role in epithelial ovarian cancer (EOC) carcinogenesis and outcomes, but a detailed profile in patient-derived tumors is needed. Here, we investigated the expression of TLR4- and MyD88-associated markers in tumors from over 500 EOC patients using immunohistochemical staining. We demonstrate that high expression of TLR4 and MyD88 predicts poorer overall survival in patients with EOC; most likely, this is due to their association with serous histology and features of high tumor burden and aggressiveness, including stage, grade, and ascites at surgery. Combined TLR4 and MyD88 expression appears to serve as an independent risk factor for shortened survival time, even after covariate adjustment (both moderate HR 1.1 [95 % CI 0.7-1.8], both strong HR 2.1 [95 % CI 1.1-3.8], both weak as referent; p = 0.027). We reveal that in EOC tissues with elevated expression of both TLR4 and MyD88 and activated NF-κB signaling pathway, expression of hsp60, hsp70, beta 2 defensin, and HMGB1 are also enriched. In total, these results suggest that activation of TLR4/MyD88/NF-κB signaling by endogenous ligands may contribute to an inflammatory microenvironment that drives a more aggressive phenotype with poorer clinical outcome in EOC patients.


Sujet(s)
Inflammation/complications , Inflammation/métabolisme , Facteur de différenciation myéloïde-88/métabolisme , Tumeurs épithéliales épidermoïdes et glandulaires/étiologie , Tumeurs épithéliales épidermoïdes et glandulaires/métabolisme , Tumeurs de l'ovaire/étiologie , Tumeurs de l'ovaire/métabolisme , Récepteur de type Toll-4/métabolisme , Microenvironnement tumoral , Adulte , Sujet âgé , Sujet âgé de 80 ans ou plus , Marqueurs biologiques , Carcinome épithélial de l'ovaire , Transformation cellulaire néoplasique , Femelle , Expression des gènes , Humains , Immunohistochimie , Inflammation/anatomopathologie , Matrix metalloproteinase 9/métabolisme , Adulte d'âge moyen , Facteur de différenciation myéloïde-88/génétique , Facteur de transcription NF-kappa B/métabolisme , Grading des tumeurs , Stadification tumorale , Tumeurs épithéliales épidermoïdes et glandulaires/mortalité , Tumeurs épithéliales épidermoïdes et glandulaires/anatomopathologie , Tumeurs de l'ovaire/mortalité , Tumeurs de l'ovaire/anatomopathologie , Pronostic , Transduction du signal , Récepteur de type Toll-4/génétique , Microenvironnement tumoral/génétique , Jeune adulte
20.
Sci Rep ; 6: 29831, 2016 07 20.
Article de Anglais | MEDLINE | ID: mdl-27436510

RÉSUMÉ

Recently, the use of a liquid biopsy has shown promise in monitoring tumor burden. While point mutations have been extensively studied, chromosomal rearrangements have demonstrated greater tumor specificity. Such rearrangements can be identified in the tumor and subsequently detected in the plasma of patients using quantitative PCR (qPCR). In this study we used a whole-genome mate-pair protocol to characterize a landscape of genomic rearrangements in the primary tumors of ten ovarian cancer patients. Individualized tumor-specific primer panels of aberrant chromosomal junctions were identified for each case and detected by qPCR within the cell-free DNA. Selected chromosomal junctions were detected in pre-surgically drawn blood in eight of the ten patients. Of these eight, three demonstrated the continued presence of circulating tumor DNA (ctDNA) post-surgery, consistent with their documented presence of disease, and in five ctDNA was undetectable in the post-surgical blood collection, consistent with their lack of detectable disease. The ctDNA fraction was calculated using a novel algorithm designed for the unique challenges of quantifying ctDNA using qPCR to allow observations of real-time tumor dynamics. In summary, a panel of individualized junctions derived from tumor DNA could be an effective way to monitor cancer patients for relapse and therapeutic efficacy using cfDNA.


Sujet(s)
Acides nucléiques acellulaires/analyse , Aberrations des chromosomes , ADN tumoral/analyse , Tumeurs de l'ovaire/génétique , Acides nucléiques acellulaires/sang , Acides nucléiques acellulaires/génétique , ADN tumoral/sang , ADN tumoral/génétique , Femelle , Humains , Récidive tumorale locale/diagnostic , Récidive tumorale locale/génétique , /méthodes , Tumeurs de l'ovaire/sang , Tumeurs de l'ovaire/chirurgie , Réaction de polymérisation en chaine en temps réel/méthodes , Reproductibilité des résultats , Sensibilité et spécificité , Charge tumorale/génétique
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...