Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
1.
Mol Oncol ; 15(1): 91-103, 2021 01.
Article in English | MEDLINE | ID: mdl-33016563

ABSTRACT

In the era of personalized medicine, where transition from organ-based to individualized genetic diagnosis takes place, the tailoring of treatment in cancer becomes increasingly important. This is particularly true for high-grade, advanced FIGO stage serous adenocarcinomas of the ovary (OC), fallopian tube (TC), and peritoneum (PC), which are currently all treated identically. We analyzed three independent patient cohorts using histopathologically classified diagnosis and various molecular approaches (transcriptomics, immunohistochemistry, next-generation sequencing, fluorescent and chromogenic in situ hybridization). Using multivariate Cox regression model, we found that PC is more aggressive compared with advanced-stage OC independent of residual disease as shown by an earlier relapse-free survival in two large cohorts (HR: 2.63, CI: 1.59-4.37, P < 0.001, and HR: 1.66, CI: 1.04-2.63, P < 0.033). In line with these findings, transcriptomic data revealed differentially expressed gene signatures identifying PC as high stromal response tumors. The third independent cohort (n = 4054) showed a distinction between these cancer types for markers suggested to be predictive for chemotherapy drug response. Our findings add additional evidence that ovarian and peritoneal cancers are epidemiologically and molecularly distinct diseases. Moreover, our data also suggest consideration of the tumor-sampling site for future diagnosis and treatment decisions.


Subject(s)
Ovarian Neoplasms/pathology , Peritoneal Neoplasms/pathology , Age Factors , Aged , Cohort Studies , Female , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Humans , Immunity/genetics , Middle Aged , Molecular Targeted Therapy , Neoplasm Grading , Neoplasm Invasiveness , Neoplasm Recurrence, Local/pathology , Neoplasm Staging , Ovarian Neoplasms/genetics , Ovarian Neoplasms/immunology , Peritoneal Neoplasms/genetics , Peritoneal Neoplasms/immunology , Stromal Cells/pathology , Treatment Outcome
3.
Swiss Med Wkly ; 148: w14647, 2018 Jul 16.
Article in English | MEDLINE | ID: mdl-30141520

ABSTRACT

OBJECTIVE: The methods used to diagnose and classify ovarian cancer have changed over the past decade. We used hospital registry data to assess the incidence, treatment durations and hospital costs of ovarian cancer in Switzerland. METHODS: We carried out a retrospective analysis of a hospital registry covering all inpatient care episodes in Switzerland between 1998 and 2012. Ovarian cancer incidence was assessed by identifying patients with a first ovarian cancer diagnosis as the main reason for hospital stay after an event-free period. We assessed the duration and cost of ovarian cancer treatment sequences as well as the evolution of hospital patient volume over time. RESULTS: The average age-adjusted incidence rate was 14.6 per 100,000 women per year between 2004 and 2012. This rate is substantially higher (+35.5%) than the corresponding rate published by the National Institute for Cancer Epidemiology and Registration (NICER). Hospital patient volume was low in most cases, with more than 40% of patients treated in hospitals with fewer than 20 cases per year. However, the share of patients treated in hospitals with more than 30 cases per year has increased substantially since 2009. CONCLUSIONS: We found a substantial difference between the ovarian cancer incidence estimate based on hospital registry data and the corresponding estimate by NICER. The reasons for this substantial difference should be carefully explored. A case-wise comparison could determine whether the difference is due to over- or under-reporting in one of the two registries. The low ovarian cancer patient volume in many hospitals is in conflict with the numbers required for certified specialised cancer centres. The recent increase in patient volume in specialised cancer centres, however, might reflect a growing understanding of the needs and requirements of comprehensive cancer care.


Subject(s)
Carcinoma, Ovarian Epithelial/epidemiology , Carcinoma, Ovarian Epithelial/surgery , Length of Stay/statistics & numerical data , Ovarian Neoplasms/epidemiology , Ovarian Neoplasms/surgery , Registries , Carcinoma, Ovarian Epithelial/therapy , Chemotherapy, Adjuvant , Female , Hospitals/statistics & numerical data , Humans , Incidence , Length of Stay/economics , Middle Aged , Ovarian Neoplasms/therapy , Retrospective Studies , Switzerland/epidemiology
4.
Oncoimmunology ; 5(2): e1062969, 2016 Feb.
Article in English | MEDLINE | ID: mdl-27057429

ABSTRACT

T-cell bispecific antibodies (TCBs) are a novel therapeutic tool designed to selectively recruit T-cells to tumor cells and simultaneously activate them. However, it is currently unknown whether the dysfunctional state of T-cells, embedded into the tumor microenvironment, imprints on the therapeutic activity of TCBs. We performed a comprehensive analysis of activation and effector functions of tumor-infiltrating T-cells (TILs) in different tumor types, upon stimulation by a TCB targeting folate receptor 1 and CD3 (FolR1-TCB). We observed a considerable heterogeneity in T-cell activation, cytokine production and tumor cell killing upon exposure to FolR1-TCB among different FolR1-expressing tumors. Of note, tumors presenting with a high frequency of PD-1hi TILs displayed significantly impaired tumor cell killing and T-cell function. Further characterization of additional T-cell inhibitory receptors revealed that PD-1hi TILs defined a T-cell subset with particularly high levels of multiple inhibitory receptors compared with PD-1int and PD-1neg T-cells. PD-1 blockade could restore cytokine secretion but not cytotoxicity of TILs in a subset of patients with scarce PD-1hi expressing cells; in contrast, patients with abundance of PD-1hi expressing T-cells did not benefit from PD-1 blockade. Our data highlight that FolR1-TCB is a promising novel immunotherapeutic treatment option which is capable of activating intratumoral T-cells in different carcinomas. However, its therapeutic efficacy may be substantially hampered by a pre-existing dysfunctional state of T-cells, reflected by abundance of intratumoral PD-1hi T-cells. These findings present a rationale for combinatorial approaches of TCBs with other therapeutic strategies targeting T-cell dysfunction.

5.
BMC Mol Biol ; 15: 24, 2014 Oct 07.
Article in English | MEDLINE | ID: mdl-25294702

ABSTRACT

BACKGROUND: The GBGT1 gene encodes the globoside alpha-1,3-N-acetylgalactosaminyltransferase 1. This enzyme catalyzes the last step in the multi-step biosynthesis of the Forssman (Fs) antigen, a pentaglycosyl ceramide of the globo series glycosphingolipids. While differential GBGT1 mRNA expression has been observed in a variety of human tissues being highest in placenta and ovary, the expression of GBGT1 and the genes encoding the glycosyltransferases and glycosidases involved in the biosynthesis of Fs as well as the possible involvement of DNA methylation in transcriptional regulation of GBGT1 expression have not yet been investigated. RESULTS: RT-qPCR profiling showed high GBGT1 expression in normal ovary surface epithelial (HOSE) cell lines and low GBGT1 expression in all (e.g. A2780, SKOV3) except one (OVCAR3) investigated ovarian cancer cell lines, a finding that was confirmed by Western blot analysis. Hierarchical cluster analysis showed that GBGT1 was even the most variably expressed gene of Fs biosynthesis-relevant glycogenes and among the investigated cell lines, whereas NAGA which encodes the alpha-N-acetylgalactosaminidase hydrolyzing Fs was not differentially expressed. Bisulfite- and COBRA-analysis of the CpG island methylation status in the GBGT1 promoter region demonstrated high or intermediate levels of GBGT1 DNA methylation in all ovarian cancer cell lines (except for OVCAR3) but marginal levels of DNA methylation in the two HOSE cell lines. The extent of DNA methylation inversely correlated with GBGT1 mRNA and protein expression. Bioinformatic analysis of GBGT1 in The Cancer Genome Atlas ovarian cancer dataset demonstrated that this inverse correlation was also found in primary ovarian cancer tissue samples confirming our cell line-based findings. Restoration of GBGT1 mRNA and protein expression in low GBGT1-expressing A2780 cells was achieved by 5-aza-2'-deoxycytidine treatment and these treated cells exhibited increased helix pomatia agglutinin-staining, reflecting the elevated presence of Fs disaccharide on these cells. CONCLUSIONS: GBGT1 expression is epigenetically silenced through promoter hypermethylation in ovarian cancer. Our findings not only suggest an involvement of DNA methylation in the synthesis of Fs antigen but may also explain earlier studies showing differential GBGT1 expression in various human tissue samples and disease stages.


Subject(s)
DNA Methylation , Gene Expression Regulation, Neoplastic , N-Acetylgalactosaminyltransferases/genetics , Ovarian Neoplasms/genetics , Azacitidine/analogs & derivatives , Azacitidine/pharmacology , Cell Line, Tumor , Decitabine , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Ovary/metabolism , Promoter Regions, Genetic
6.
Cancer Immunol Immunother ; 63(9): 925-38, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24906866

ABSTRACT

In addition to direct tumor cell cytotoxicity, chemotherapy can mediate tumor reduction through immune modulation of the tumor microenvironment to promote anti-tumor immunity. Mature dendritic cells (DCs) play key roles in priming robust immune responses in tumor-bearing hosts. Here, we screened a panel of 21 anticancer agents with defined molecular targets for their ability to induce direct maturation of DCs. We identified ansamitocin P3, a microtubule-depolymerizing agent, as a potent inducer of phenotypic and functional maturation of DCs. Exposure of both murine spleen-derived and human monocyte-derived DCs to ansamitocin P3 triggered up-regulation of maturation markers and production of pro-inflammatory cytokines, resulting in an enhanced T cell stimulatory capacity. Local administration of ansamitocin P3 induced maturation of skin Langerhans cells in vivo and promoted antigen uptake and extensive homing of tumor-resident DCs to tumor-draining lymph nodes. When used as an adjuvant in a specific vaccination approach, ansamitocin P3 dramatically increased activation of antigen-specific T cells. Finally, we demonstrate that ansamitocin P3, due to its immunomodulatory properties, acts in synergy with antibody-mediated blockade of the T cell inhibitory receptors PD-1 and CTLA-4. The combination treatment was most effective and induced durable growth inhibition of established tumors. Mechanistically, we observed a reduced regulatory T cell frequency and improved T cell effector function at the tumor site. Taken together, our study unravels an immune-based anti-tumor mechanism exploited by microtubule-depolymerizing agents, including ansamitocin P3, and paves the way for future clinical trials combining this class of agents with immunotherapy.


Subject(s)
Dendritic Cells/drug effects , Maytansine/analogs & derivatives , Neoplasms, Experimental/drug therapy , Neoplasms, Experimental/immunology , Tubulin Modulators/pharmacology , Animals , B7-2 Antigen/biosynthesis , B7-2 Antigen/immunology , CD11 Antigens/immunology , CTLA-4 Antigen/antagonists & inhibitors , CTLA-4 Antigen/immunology , Cell Line, Tumor , Dendritic Cells/immunology , Humans , Interferon-gamma/immunology , Lymph Nodes/drug effects , Lymph Nodes/immunology , Lymphocyte Activation/drug effects , Maytansine/pharmacology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microtubules/drug effects , Microtubules/metabolism , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Programmed Cell Death 1 Receptor/immunology , T-Lymphocytes/drug effects , T-Lymphocytes/immunology
7.
J Ovarian Res ; 7: 60, 2014.
Article in English | MEDLINE | ID: mdl-24936210

ABSTRACT

Ovarian cancer is the fifth most common cause of cancer death in women and the leading cause of death from gynaecological malignancies. Of the 75% women diagnosed with locally advanced or disseminated disease, only 30% will survive five years following treatment. This poor prognosis is due to the following reasons: limited understanding of the tumor origin, unclear initiating events and early developmental stages of ovarian cancer, lack of reliable ovarian cancer-specific biomarkers, and drug resistance in advanced cases. In the past, in vitro studies using cell line models have been an invaluable tool for basic, discovery-driven cancer research. However, numerous issues including misidentification and cross-contamination of cell lines have hindered research efforts. In this study we examined all ovarian cancer cell lines available from cell banks. Hereby, we identified inconsistencies in the reporting, difficulties in the identification of cell origin or clinical data of the donor patients, restricted ethnic and histological type representation, and a lack of tubal and peritoneal cancer cell lines. We recommend that all cell lines should be distributed via official cell banks only with strict guidelines regarding the minimal available information required to improve the quality of ovarian cancer research in future.


Subject(s)
Ovarian Neoplasms/pathology , Animals , Cell Line, Tumor , Drug Screening Assays, Antitumor , Female , Humans , Ovarian Neoplasms/genetics , Reproducibility of Results
8.
Cancer Immunol Res ; 2(8): 741-55, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24916470

ABSTRACT

Antibody-drug conjugates (ADC) are emerging as powerful treatment strategies with outstanding target-specificity and high therapeutic activity in patients with cancer. Brentuximab vedotin represents a first-in-class ADC directed against CD30(+) malignancies. We hypothesized that its sustained clinical responses could be related to the stimulation of an anticancer immune response. In this study, we demonstrate that the dolastatin family of microtubule inhibitors, from which the cytotoxic component of brentuximab vedotin is derived, comprises potent inducers of phenotypic and functional dendritic cell (DC) maturation. In addition to the direct cytotoxic effect on tumor cells, dolastatins efficiently promoted antigen uptake and migration of tumor-resident DCs to the tumor-draining lymph nodes. Exposure of murine and human DCs to dolastatins significantly increased their capacity to prime T cells. Underlining the requirement of an intact host immune system for the full therapeutic benefit of dolastatins, the antitumor effect was far less pronounced in immunocompromised mice. We observed substantial therapeutic synergies when combining dolastatins with tumor antigen-specific vaccination or blockade of the PD-1-PD-L1 and CTLA-4 coinhibitory pathways. Ultimately, treatment with ADCs using dolastatins induces DC homing and activates cellular antitumor immune responses in patients. Our data reveal a novel mechanism of action for dolastatins and provide a strong rationale for clinical treatment regimens combining dolastatin-based therapies, such as brentuximab vedotin, with immune-based therapies.


Subject(s)
Dendritic Cells/immunology , Depsipeptides/pharmacology , Neoplasms/immunology , Tubulin Modulators/pharmacology , Animals , Antibodies/therapeutic use , Antigens/immunology , Brentuximab Vedotin , CTLA-4 Antigen/antagonists & inhibitors , Cancer Vaccines/therapeutic use , Cell Line , Cells, Cultured , Cytokines/immunology , Dendritic Cells/cytology , Dendritic Cells/drug effects , Depsipeptides/therapeutic use , Humans , Immunoconjugates/therapeutic use , Mice, Inbred C57BL , Mice, Transgenic , Neoplasms/pathology , Neoplasms/therapy , Ovalbumin/immunology , Programmed Cell Death 1 Receptor/antagonists & inhibitors , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , Tubulin Modulators/therapeutic use , Tumor Burden/drug effects
9.
Curr Probl Dermatol ; 45: 216-24, 2014.
Article in English | MEDLINE | ID: mdl-24643189

ABSTRACT

Human papillomavirus (HPV) infections affect women in every age group and in various benign, premalignant as well as malignant gynecological conditions. As a benign condition, condylomata acuminata of the whole female genital tract can be observed, transmitted by low risk HPV types 6 and 11, whilst dysplastic changes of the vulva appear as vulvar intraepithelial neoplasia, of the vagina as vaginal intraepithelial neoplasia and of the cervix as cervical intraepithelial neoplasia, and are caused by high risk HPV types most notably 16 and 18. These dysplastic changes give rise to precursor lesions of vulvar and cervical cancer, both driven via immune regression and potentially hormonal changes by promoting the malignant transformation profile of HPV subtypes. Attributes which can support this process are smoking, immunodeficiency, vitamin deficiency, stress, vaginal infections and hormonal influence. The causal relationship between persistent infection with high-risk HPV genotypes and vulvar and cervical cancer has been clearly demonstrated and is stronger than the relationship observed between smoking and lung cancer. New global cancer prevention can be envisaged by implementing vaccination against HPV in young women, with 2 vaccines currently approved by the Food and Drug Administration: the quadrivalent Gardasil (HPV-6, -11, -16, -18) and the bivalent Cervarix (HPV-16, -18).


Subject(s)
Carcinoma/therapy , Genital Neoplasms, Female/therapy , Papillomavirus Infections/therapy , Adult , Aged , Aged, 80 and over , Antiviral Agents/therapeutic use , Brachytherapy/methods , Carcinoma/prevention & control , Carcinoma/virology , Combined Modality Therapy/methods , Female , Genital Neoplasms, Female/prevention & control , Genital Neoplasms, Female/virology , Humans , Middle Aged , Papillomavirus Infections/complications , Papillomavirus Infections/prevention & control , Papillomavirus Vaccines/therapeutic use , Young Adult
10.
Int J Mol Med ; 33(4): 784-94, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24535703

ABSTRACT

Vulvar melanoma is the second most common vulvar cancer. Patients with vulvar melanoma usually present with the disease at a late stage and have a poor prognosis. The prognostic predictors reported in the literature are not unequivocal and the role of lichen sclerosus and c-KIT mutations in the aetiology of vulvar melanoma is unclear. Breslow staging currently seems to be the most adequate predictor of prognosis. We thus performed a clinicopathological and literature review to identify suitable predictors of prognosis and survival and investigated the expression of c-KIT (by immunohistochemistry) in patients with vulvar melanoma (n=33) from the Gynaecological Cancer Centres of the Royal Hospital for Women (Sydney, Australia) and John Hunter Hospital (Newcastle, Australia). Our series of 33 patients fitted the expected clinical profile of older women: delayed presentation, high stage, limited response to treatment and poor prognosis. We identified 3 patients (9.1%) with lichen sclerosus associated with melanoma in situ, although no lichen sclerosus was found in the areas of invasive melanoma. No patient had vulvar nevi. We identified a) Breslow's depth, b) an absence of any of the pathological risk factors, such as satellitosis, in-transit metastasis, lymphovascular space invasion (LVSI) and dermal mitosis, c) removal of inguino-femoral lymph nodes, d) lateral margin of >1 cm, and e) c-KIT expression as valuable prognostic predictors for disease-free survival. We conclude that c-KIT expression is, apart from Breslow's depth, another valuable predictor of prognosis and survival. Lichen sclerosus may be associated with vulvar melanoma.


Subject(s)
Melanoma/metabolism , Melanoma/pathology , Proto-Oncogene Proteins c-kit/metabolism , Vulvar Neoplasms/metabolism , Vulvar Neoplasms/pathology , Adult , Aged , Aged, 80 and over , Biomarkers, Tumor/metabolism , Cell Proliferation , Cohort Studies , Female , Humans , Immunohistochemistry , Kaplan-Meier Estimate , Melanocytes/metabolism , Melanocytes/pathology , Melanoma/immunology , Middle Aged , Molecular Targeted Therapy , Multivariate Analysis , Prognosis , Risk Factors , Treatment Outcome , Vulvar Neoplasms/immunology
11.
PLoS One ; 8(1): e54362, 2013.
Article in English | MEDLINE | ID: mdl-23326605

ABSTRACT

Aberrant Wnt signalling is implicated in numerous human cancers, and understanding the effects of modulation of pathway members may lead to the development of novel therapeutics. Expression of secreted frizzled related protein 4 (SFRP4), an extracellular modulator of the Wnt signalling pathway, is progressively lost in more aggressive ovarian cancer phenotypes. Here we show that recombinant SFRP4 (rSFRP4) treatment of a serous ovarian cancer cell line results in inhibition of ß-catenin dependent Wnt signalling as measured by TOP/FOP Wnt reporter assay and decreased transcription of Wnt target genes, Axin2, CyclinD1 and Myc. In addition, rSFRP4 treatment significantly increased the ability of ovarian cancer cells to adhere to collagen and fibronectin, and decreased their ability to migrate across an inflicted wound. We conclude that these changes in cell behaviour may be mediated via mesenchymal to epithelial transition (MET), as rSFRP4 treatment also resulted in increased expression of the epithelial marker E-cadherin, and reduced expression of Vimentin and Twist. Combined, these results indicate that modulation of a single upstream gatekeeper of Wnt signalling can have effects on downstream Wnt signalling and ovarian cancer cell behaviour, as mediated through epithelial to mesenchymal plasticity (EMP). This raises the possibility that SFRP4 may be used both diagnostically and therapeutically in epithelial ovarian cancer.


Subject(s)
Neoplasms, Glandular and Epithelial/genetics , Ovarian Neoplasms/genetics , Proto-Oncogene Proteins , Recombinant Proteins , Wnt Signaling Pathway/genetics , Carcinoma, Ovarian Epithelial , Cell Line, Tumor , Cell Movement/genetics , Epithelial-Mesenchymal Transition , Female , Genetic Vectors , Humans , Molecular Targeted Therapy , Neoplasms, Glandular and Epithelial/pathology , Ovarian Neoplasms/pathology , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Transfection , beta Catenin/genetics , beta Catenin/metabolism
12.
PLoS One ; 7(2): e31885, 2012.
Article in English | MEDLINE | ID: mdl-22363760

ABSTRACT

BACKGROUND: Activation of the Wnt signaling pathway is implicated in aberrant cellular proliferation in various cancers. In 40% of endometrioid ovarian cancers, constitutive activation of the pathway is due to oncogenic mutations in ß-catenin or other inactivating mutations in key negative regulators. Secreted frizzled-related protein 4 (SFRP4) has been proposed to have inhibitory activity through binding and sequestering Wnt ligands. METHODOLOGY/PRINCIPAL FINDINGS: We performed RT-qPCR and Western-blotting in primary cultures and ovarian cell lines for SFRP4 and its key downstream regulators activated ß-catenin, ß-catenin and GSK3ß. SFRP4 was then examined by immunohistochemistry in a cohort of 721 patients and due to its proposed secretory function, in plasma, presenting the first ELISA for SFRP4. SFRP4 was most highly expressed in tubal epithelium and decreased with malignant transformation, both on RNA and on protein level, where it was even more profound in the membrane fraction (p<0.0001). SFRP4 was expressed on the protein level in all histotypes of ovarian cancer but was decreased from borderline tumors to cancers and with loss of cellular differentiation. Loss of membrane expression was an independent predictor of poor survival in ovarian cancer patients (p = 0.02 unadjusted; p = 0.089 adjusted), which increased the risk of a patient to die from this disease by the factor 1.8. CONCLUSIONS/SIGNIFICANCE: Our results support a role for SFRP4 as a tumor suppressor gene in ovarian cancers via inhibition of the Wnt signaling pathway. This has not only predictive implications but could also facilitate a therapeutic role using epigenetic targets.


Subject(s)
Ovarian Neoplasms/metabolism , Ovarian Neoplasms/pathology , Proto-Oncogene Proteins/metabolism , Ascites/metabolism , Ascites/pathology , Cell Line, Tumor , Cell Membrane/metabolism , Cell Transformation, Neoplastic , Cohort Studies , Female , Gene Expression Regulation, Neoplastic , Humans , Immunohistochemistry , Neoplasm Invasiveness , Ovarian Neoplasms/blood , Ovarian Neoplasms/genetics , Phenotype , Prognosis , Proto-Oncogene Proteins/blood , Proto-Oncogene Proteins/genetics , Survival Analysis , Treatment Outcome
13.
J Clin Pathol ; 60(6): 622-6, 2007 Jun.
Article in English | MEDLINE | ID: mdl-16822880

ABSTRACT

BACKGROUND: Currently, no specific immunohistochemical markers are available to differentiate primary mucinous epithelial ovarian cancer (MOC) from adenocarcinomas originating at other sites that have metastasised to the ovary, which may have an impact on patient management and prognosis. AIM: To investigate the expression of two intestinal markers, galectin 4 and meprin alpha, in mucinous carcinomas of the ovary and gastrointestinal tract. METHODS: Using immunohistochemical analysis, the expression of galectin 4 and meprin alpha was investigated in 10 MOCs and in 38 mucinous adenocarcinomas of colon, pancreas, stomach and appendix, the most common sites of origin of ovarian metastases. RESULTS: Total cytoplasmic galectin 4 expression was relatively consistent between the different carcinomas. Membranous meprin alpha expression was significantly lower in MOCs compared with gastrointestinal carcinomas. Moreover, meprin alpha expression showed greater discrimination between the ovarian and gastrointestinal carcinomas than the cytokeratins CK7 and CK20, the current standard immunohistochemical markers used to determine the tissue origin of mucinous carcinomas involving the ovaries. CONCLUSIONS: Meprin alpha is a useful additional marker in differentiating primary from secondary mucinous adenocarcinomas of the ovary.


Subject(s)
Adenocarcinoma, Mucinous/diagnosis , Biomarkers, Tumor/metabolism , Gastrointestinal Neoplasms/pathology , Metalloendopeptidases/metabolism , Ovarian Neoplasms/diagnosis , Adenocarcinoma/diagnosis , Adenocarcinoma/metabolism , Adenocarcinoma/secondary , Adenocarcinoma, Mucinous/metabolism , Diagnosis, Differential , Female , Galectin 4/metabolism , Humans , Immunoenzyme Techniques , Keratin-20/metabolism , Keratin-7/metabolism , Neoplasm Proteins/metabolism , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/secondary
14.
Clin Cancer Res ; 10(13): 4427-36, 2004 Jul 01.
Article in English | MEDLINE | ID: mdl-15240533

ABSTRACT

PURPOSE: A better understanding of the molecular pathways underlying the development of epithelial ovarian cancer (EOC) is critical to identify ovarian tumor markers for use in diagnostic or therapeutic applications. The aims of this study were to integrate the results from 14 transcript profiling studies of EOC to identify novel biomarkers and to examine their expression in early and late stages of the disease. EXPERIMENTAL DESIGN: A database incorporating genes identified as being highly up-regulated in each study was constructed. Candidate tumor markers were selected from genes that overlapped between studies and by evidence of surface membrane or secreted expression. The expression patterns of three integral membrane proteins, discoidin domain receptor 1 (DDR1), claudin 3 (CLDN3), and epithelial cell adhesion molecule, all of which are involved in cell adhesion, were evaluated in a cohort of 158 primary EOC using immunohistochemistry. RESULTS: We confirmed that these genes are highly overexpressed in all histological subtypes of EOC compared with normal ovarian surface epithelium, identifying DDR1 and CLDN3 as new biomarkers of EOC. Furthermore, we determined that these genes are also expressed in ovarian epithelial inclusion cysts, a site of metaplastic changes within the normal ovary, in borderline tumors and in low-grade and stage cancer. A trend toward an association between low CLDN3 expression and poor patient outcome was also observed. CONCLUSIONS: These results suggest that up-regulation of DDR1, CLDN3, and epithelial cell adhesion molecule are early events in the development of EOC and have potential application in the early detection of disease.


Subject(s)
Antigens, Neoplasm/biosynthesis , Cell Adhesion Molecules/biosynthesis , Membrane Proteins/biosynthesis , Ovarian Neoplasms/metabolism , Ovary/metabolism , Receptor Protein-Tyrosine Kinases/biosynthesis , Biomarkers, Tumor , Cell Adhesion , Cell Line, Tumor , Cell Membrane/metabolism , Claudin-3 , Cohort Studies , DNA, Complementary/metabolism , Databases as Topic , Discoidin Domain Receptor 1 , Disease Progression , Disease-Free Survival , Epithelial Cell Adhesion Molecule , Epithelium/metabolism , Female , Genetic Markers , Humans , Immunohistochemistry , Oligonucleotide Array Sequence Analysis , RNA, Messenger/metabolism , Time Factors , Treatment Outcome , Up-Regulation
SELECTION OF CITATIONS
SEARCH DETAIL
...