Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
1.
Gynecol Oncol ; 139(1): 118-26, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26232337

ABSTRACT

OBJECTIVE: Endometrial carcinoma (EC) is the sixth most common cancer in women and therapies are limited for advanced and recurrent disease. Patient-derived tumor xenograft (PDTX) models are becoming popular tools in translational research because of their histological and genetic similarity to the original tumors and the ability to predict therapeutic response to treatments. Here, we established and characterized a panel of 24 EC PDTX models which includes the major histological and genetic subtypes observed in patients. METHODS: Fresh tumor tissues collected from primary, metastatic and recurrent type I and type II EC patients were engrafted in immunocompromised mice. Histology, vimentin, and cytokeratin expression were evaluated, together with Microsatellite instability (MSI), mutation profiling by Whole Exome Sequencing and copy number profiling by Whole Genome Low Coverage Sequencing. The efficacy of both PI3K and MEK inhibitors was evaluated in a model of endometrioid carcinoma harboring PTEN, PIK3CA and KRAS mutations. RESULTS: We observed good similarity between primary tumors and the corresponding xenografts, at histological and genetic level. Among the engrafted endometrioid models, we found a significant enrichment of MSI and POLE mutated tumors, compared to non-engrafted samples. Combination treatment with NVP-BEZ235 and AZD6244 showed the possibility to stabilize the tumor growth in one model originated from a patient who already received several lines of chemotherapy. CONCLUSION: The established EC PDTX models, resembling the original human tumors, promise to be useful for preclinical evaluation of novel combination and targeted therapies in specific EC subgroups.


Subject(s)
Endometrial Neoplasms/drug therapy , Endometrial Neoplasms/pathology , Xenograft Model Antitumor Assays/methods , Animals , Endometrial Neoplasms/enzymology , Endometrial Neoplasms/genetics , Female , Humans , Mice , Molecular Targeted Therapy , Neoplasm Transplantation , Protein Kinase Inhibitors/pharmacology
2.
Gynecol Oncol ; 138(2): 378-82, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26050920

ABSTRACT

OBJECTIVES: New treatment options for advanced and recurrent endometrial carcinoma (EC) are necessary. Epidemiological studies showed that diabetic patients using metformin have reduced risks of endometrial cancer (EC) incidence. Moreover, pre- and clinical studies demonstrated an antitumor effect by metformin, with and without additional treatments, for different solid malignancies. However, cancer cell-autonomous effects of metformin on EC have not been fully characterized yet. The aim of this study was to investigate the effect of metformin, with or without carboplatin, on patient-derived primary endometrioid EC cells xenografted in nude mice, to assess its ability to reduce or impair growth in already established tumors. METHODS: Two xenograft models were established by subcutaneous inoculation of primary endometrioid EC cell suspensions. Tumors were allowed to grow and then mice were treated with metformin (250 mg/kg, daily, p.o.), carboplatin (50 mg/kg, 1×/week, i.p.), or the combination of both compounds at the same concentration as single treatment, for three weeks. Effects of metformin treatment on the tumor mass were determined by tumor growth follow-up. Metformin influences on AMPK/mTOR cell signaling were evaluated by investigating AKT, AMPK and S6 phosphorylation levels. RESULTS: In vivo, metformin did not affect the growth of EC tumors established from patient-derived primary cultures and the phosphorylation of AKT, AMPK and S6. In addition, no enhanced antitumor effect was determined by combining metformin and carboplatin treatments. CONCLUSIONS: Metformin, at clinically relevant concentrations, did not show effects on the growth of already established tumors. Adding metformin to carboplatin did not have synergistic effects.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Carcinoma, Endometrioid/drug therapy , Endometrial Neoplasms/drug therapy , Metformin/pharmacology , Animals , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Carboplatin/administration & dosage , Carcinoma, Endometrioid/pathology , Endometrial Neoplasms/pathology , Female , Humans , Metformin/administration & dosage , Mice , Mice, Nude , Xenograft Model Antitumor Assays
3.
Gynecol Oncol ; 138(1): 165-73, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25933683

ABSTRACT

OBJECTIVES: Endometrial carcinoma (EC) is the most common gynecological cancer in the Western World. Treatment options are limited for advanced and recurrent disease. Therefore, new treatment options are necessary. Inhibition of the PI3K/AKT/mTOR and/or the Ras/Raf/MEK pathways is suggested to be clinically relevant. However, the knowledge about the effect of combination targeted therapy in EC is limited. The aim of this study was to investigate the effect of these therapies on primary endometrioid EC cell cultures in vitro and in vivo. METHODS: Primary endometrioid EC cell cultures were incubated with Temsirolimus (mTORC1 inhibitor), NVP-BKM120 (pan-PI3K inhibitor), NVP-BEZ235 (pan-PI3K/mTOR inhibitor), or AZD6244 (MEK1/2 inhibitor) as single treatment. In vitro, the effect of NVP-BEZ235 with or without AZD6244 was determined for cell viability, cell cycle arrest, apoptosis induction, and cell signaling. In vivo, the effect of NVP-BEZ35 was investigated for 2 subcutaneous xenograft models of the corresponding primary cultures. RESULTS: NVP-BEZ235 was the most potent PI3K/AKT/mTOR pathway inhibitor. NVP-BEZ235 and AZD6244 reduced cell viability and induced cell cycle arrest and apoptosis, by reduction of p-AKT, p-S6, and p-ERK levels. Combination treatment showed a synergistic effect. In vivo, NVP-BEZ235 reduced tumor growth and inhibited p-S6 expression. The effects of the compounds were independent of the mutation profile of the cell cultures used. CONCLUSIONS: A synergistic antitumor effect was shown for NVP-BEZ235 and AZD6244 in primary endometrioid EC cells in vitro. In addition, NVP-BEZ235 induced reduction of tumor growth in vivo. Therefore, targeted therapies seem an interesting strategy to further evaluate in clinical trials.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Benzimidazoles/pharmacology , Carcinoma, Endometrioid/drug therapy , Endometrial Neoplasms/drug therapy , Imidazoles/pharmacology , Quinolines/pharmacology , Animals , Benzimidazoles/administration & dosage , Carcinoma, Endometrioid/enzymology , Drug Synergism , Endometrial Neoplasms/enzymology , Female , Humans , Imidazoles/administration & dosage , MAP Kinase Kinase Kinases/antagonists & inhibitors , MAP Kinase Signaling System/drug effects , Mice , Phosphatidylinositol 3-Kinases/metabolism , Phosphoinositide-3 Kinase Inhibitors , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Proto-Oncogene Proteins c-akt/metabolism , Quinolines/administration & dosage , TOR Serine-Threonine Kinases/antagonists & inhibitors , TOR Serine-Threonine Kinases/metabolism , Tumor Cells, Cultured , Xenograft Model Antitumor Assays , raf Kinases/antagonists & inhibitors , raf Kinases/metabolism , ras Proteins/antagonists & inhibitors , ras Proteins/metabolism
4.
Int J Gynecol Cancer ; 25(3): 363-71, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25695543

ABSTRACT

OBJECTIVE: Well-characterized, low-passage, primary cell cultures established directly from patient tumors are an important tool for drug screening because these cultures faithfully recapitulate the genomic features of primary tumors. Here, we aimed to establish these cell cultures from primary endometrial carcinomas (ECs) and to develop subcutaneous and orthotopic xenograft models as a model to validate promising treatment options for EC in the in vivo setting. METHODS: Primary cell cultures of EC tumors were established and validated by analysing histologic and genetic characteristics, telomerase activity, and in vitro and in vivo growth characteristics. Using these primary cell cultures, subcutaneous and orthotopic mouse models were subsequently established. RESULTS: We established and characterized 7 primary EC cell cultures and corresponding xenograft models of different types of endometrioid tumors. Interestingly, we observed that the chance to successfully establish a primary cell culture seems higher for microsatellite instable than microsatellite stable tumors. For the first time, we also established an orthotopic murine model for EC derived from a primary cell culture. In contrast to EC cell lines, grafted tumor cultures preserved the original tumor structure and mimicked all histologic features. They also established abdominal and distant metastases, reflecting the tumorigenic behavior in the clinical setting. Remarkably, the established cell cultures and xenograft tumors also preserved the genetic characteristics of the primary tumor. CONCLUSIONS: The established EC cultures reflect the epithelial genetic characteristics of the primary tumor. Therefore, they provide an appropriate model to investigate EC biology and apply high-throughput drug screening experiments. In addition, the established murine xenograft models, in particular the orthotopic model, will be useful to validate promising therapeutic strategies in vivo, as the grafted tumors closely resemble the primary tumors from which they were derived. Microsatellite instable status seems to determine the success rate of establishing primary cell cultures.


Subject(s)
Carcinoma/genetics , Disease Models, Animal , Endometrial Neoplasms/genetics , Endometrial Neoplasms/pathology , Microsatellite Instability , Adult , Aged , Animals , Carcinoma/chemistry , Carcinoma/secondary , Cell Proliferation , Endometrial Neoplasms/chemistry , Female , Humans , Keratins/analysis , Mice , Mice, Nude , Microsatellite Repeats , Middle Aged , Neoplasm Transplantation/methods , Primary Cell Culture , Receptors, Estrogen/analysis , Receptors, Progesterone/analysis , Tumor Cells, Cultured , Vimentin/analysis
5.
Hum Genet ; 134(2): 231-45, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25487306

ABSTRACT

Several studies have reported associations between multiple cancer types and single-nucleotide polymorphisms (SNPs) on chromosome 5p15, which harbours TERT and CLPTM1L, but no such association has been reported with endometrial cancer. To evaluate the role of genetic variants at the TERT-CLPTM1L region in endometrial cancer risk, we carried out comprehensive fine-mapping analyses of genotyped and imputed SNPs using a custom Illumina iSelect array which includes dense SNP coverage of this region. We examined 396 SNPs (113 genotyped, 283 imputed) in 4,401 endometrial cancer cases and 28,758 controls. Single-SNP and forward/backward logistic regression models suggested evidence for three variants independently associated with endometrial cancer risk (P = 4.9 × 10(-6) to P = 7.7 × 10(-5)). Only one falls into a haplotype previously associated with other cancer types (rs7705526, in TERT intron 1), and this SNP has been shown to alter TERT promoter activity. One of the novel associations (rs13174814) maps to a second region in the TERT promoter and the other (rs62329728) is in the promoter region of CLPTM1L; neither are correlated with previously reported cancer-associated SNPs. Using TCGA RNASeq data, we found significantly increased expression of both TERT and CLPTM1L in endometrial cancer tissue compared with normal tissue (TERT P = 1.5 × 10(-18), CLPTM1L P = 1.5 × 10(-19)). Our study thus reports a novel endometrial cancer risk locus and expands the spectrum of cancer types associated with genetic variation at 5p15, further highlighting the importance of this region for cancer susceptibility.


Subject(s)
Chromosomes, Human, Pair 5/genetics , Genetic Loci , Membrane Proteins/genetics , Models, Genetic , Neoplasm Proteins/genetics , Polymorphism, Single Nucleotide , Telomerase/genetics , Chromosomes, Human, Pair 5/metabolism , Databases, Nucleic Acid , Female , Gene Expression Regulation, Neoplastic/genetics , Haplotypes , Humans , Membrane Proteins/biosynthesis , Neoplasm Proteins/biosynthesis , Promoter Regions, Genetic , Risk Factors , Telomerase/biosynthesis
6.
Histopathology ; 65(3): 371-88, 2014 Sep.
Article in English | MEDLINE | ID: mdl-25353038

ABSTRACT

AIMS: To check the usefulness of a standardized protocol of PTEN FISH in 31 endometrial carcinomas (ECs) in comparison with SNP array (SNPA), multiplex ligation-dependent probe amplification (MLPA), and immunohistochemistry. METHODS AND RESULTS: Fluorescence in-situ hybridization analysis showed two PTEN copies in 17 cases, three copies in nine cases, hemizygous deletion in two cases, and diverse cell populations with different PTEN copy number in three cases. A good correlation was seen between FISH and SNPA, particularly in cases with three copies. FISH identified two cases with entire deletion of chromosome 10, but did not identify a focal deletion of PTEN. Five cases with PTEN deletion and duplication of the second allele by SNPA were interpreted as normal by FISH. Concordance between FISH and MLPA was seen in 15 cases with two copies, and in two cases with PTEN deletion. Six cases were interpreted as amplified by MLPA, but showed polyploidy by FISH. FISH was superior to SNPA and MLPA in assessing the tumours with diverse cell populations with different PTEN copies. CONCLUSIONS: The results show good concordance between FISH, SNPA and MLPA. SNPA was superior in tumours with deletion of one copy and duplication of the second allele. FISH was superior in assessing tumour heterogeneity.


Subject(s)
Endometrial Neoplasms/genetics , In Situ Hybridization, Fluorescence , PTEN Phosphohydrolase/genetics , Carcinoma, Endometrioid/genetics , Carcinoma, Endometrioid/metabolism , Carcinoma, Endometrioid/pathology , Endometrial Neoplasms/metabolism , Endometrial Neoplasms/pathology , Female , Gene Dosage , Genes, Tumor Suppressor , Humans , Immunohistochemistry , Multiplex Polymerase Chain Reaction , PTEN Phosphohydrolase/metabolism , Polymorphism, Single Nucleotide , Reproducibility of Results
7.
Mol Cancer ; 13: 223, 2014 Sep 27.
Article in English | MEDLINE | ID: mdl-25261936

ABSTRACT

BACKGROUND: About 20% of patients diagnosed with endometrial cancer (EC) are considered high-risk with unfavorable prognosis. In the framework of the European Network for Individualized Treatment in EC (ENITEC), we investigated the presence and phenotypic features of Circulating Tumor Cells (CTC) in high-risk EC patients. METHODS: CTC isolation was carried out in peripheral blood samples from 34 patients, ranging from Grade 3 Stage IB to Stage IV carcinomas and recurrences, and 27 healthy controls using two methodologies. Samples were subjected to EpCAM-based immunoisolation using the CELLection™ Epithelial Enrich kit (Invitrogen, Dynal) followed by RTqPCR analysis. The phenotypic determinants of endometrial CTC in terms of pathogenesis, hormone receptor pathways, stem cell markers and epithelial to mesenchymal transition (EMT) drivers were asked. Kruskal-Wallis analysis followed by Dunn's post-test was used for comparisons between groups. Statistical significance was set at p < 0.05. RESULTS: EpCAM-based immunoisolation positively detected CTC in high-risk endometrial cancer patients. CTC characterization indicated a remarkable plasticity phenotype defined by the expression of the EMT markers ETV5, NOTCH1, SNAI1, TGFB1, ZEB1 and ZEB2. In addition, the expression of ALDH and CD44 pointed to an association with stemness, while the expression of CTNNB1, STS, GDF15, RELA, RUNX1, BRAF and PIK3CA suggested potential therapeutic targets. We further recapitulated the EMT phenotype found in endometrial CTC through the up-regulation of ETV5 in an EC cell line, and validated in an animal model of systemic dissemination the propensity of these CTC in the accomplishment of metastasis. CONCLUSIONS: Our results associate the presence of CTC with high-risk EC. Gene-expression profiling characterized a CTC-plasticity phenotype with stemness and EMT features. We finally recapitulated this CTC-phenotype by over-expressing ETV5 in the EC cell line Hec1A and demonstrated an advantage in the promotion of metastasis in an in vivo mouse model of CTC dissemination and homing.


Subject(s)
Endometrial Neoplasms/genetics , Endometrial Neoplasms/pathology , Gene Expression Profiling , Neoplastic Cells, Circulating/metabolism , Neoplastic Cells, Circulating/pathology , Aged , Animals , Cell Separation , DNA-Binding Proteins/metabolism , Disease Models, Animal , Epithelial-Mesenchymal Transition/genetics , Female , Gene Expression Regulation, Neoplastic , Humans , Mice, Nude , Neoplasm Metastasis , Phenotype , Risk Factors , Transcription Factors/metabolism
8.
Elife ; 3: e02725, 2014 Aug 01.
Article in English | MEDLINE | ID: mdl-25085081

ABSTRACT

DNA replication errors that persist as mismatch mutations make up the molecular fingerprint of mismatch repair (MMR)-deficient tumors and convey them with resistance to standard therapy. Using whole-genome and whole-exome sequencing, we here confirm an MMR-deficient mutation signature that is distinct from other tumor genomes, but surprisingly similar to germ-line DNA, indicating that a substantial fraction of human genetic variation arises through mutations escaping MMR. Moreover, we identify a large set of recurrent indels that may serve to detect microsatellite instability (MSI). Indeed, using endometrial tumors with immunohistochemically proven MMR deficiency, we optimize a novel marker set capable of detecting MSI and show it to have greater specificity and selectivity than standard MSI tests. Additionally, we show that recurrent indels are enriched for the 'DNA double-strand break repair by homologous recombination' pathway. Consequently, DSB repair is reduced in MMR-deficient tumors, triggering a dose-dependent sensitivity of MMR-deficient tumor cultures to DSB inducers.


Subject(s)
Biomarkers, Tumor/genetics , DNA Breaks, Double-Stranded , Endometrial Neoplasms/genetics , INDEL Mutation , Microsatellite Repeats , Ovarian Neoplasms/genetics , Base Pair Mismatch , DNA Fingerprinting , DNA Mismatch Repair , Endometrial Neoplasms/diagnosis , Endometrial Neoplasms/pathology , Female , Homologous Recombination , Humans , Microsatellite Instability , Neoplasm Staging , Ovarian Neoplasms/diagnosis , Ovarian Neoplasms/pathology , Sensitivity and Specificity
9.
Oncol Rep ; 29(2): 413-8, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23232836

ABSTRACT

Placental growth factor (PlGF), a homolog of vascular endothelial growth factor (VEGF), exerts pleiotropic functions in cancer by affecting tumor cells as well as endothelial and inflammatory cells. Moreover, PlGF expression correlates with tumor stage, recurrence, metastasis and patient outcome in different types of cancer. Recently, administration of anti-PlGF therapy reduced tumor growth and metastasis in preclinical tumor models. In the present study, we evaluated the diagnostic and prognostic value of systemic and local expression of PlGF in primary endometrial carcinomas. PlGF levels in tumor lysates (n=128) and serum (n=88) of patients with primary endometrial cancer were determined using ELISA. PlGF mRNA expression in endometrial carcinoma tissues was quantified by quantitative qRT-PCR. Results were compared to endometrial cancer stage and grade. Systemic PlGF levels were not altered in patients with endometrial cancer (FIGO stage I-II-III) as compared to healthy controls. Only in FIGO stage IV patients, serum PlGF levels were slightly increased. Local PlGF mRNA and protein expression in endometrial tumors progressively increased with tumor grade. In endometrioid carcinomas, PlGF mRNA expression was significantly increased in endometrioid grade 3 tumors as compared to normal endometrial tissue. PlGF protein expression was significantly increased in endometrioid grade 2 and 3 carcinomas and in serous carcinomas as compared to normal endometrial tissue. Our study showed that systemic/serum PlGF levels cannot be used as a diagnostic or prognostic marker in endometrial cancer. However, the increased local expression of PlGF, primarily in high-grade carcinomas, underscores the possibility for preclinical assessment of anti-PlGF therapy in endometrial cancer.


Subject(s)
Biomarkers, Tumor/metabolism , Carcinoma/metabolism , Carcinoma/pathology , Endometrial Neoplasms/metabolism , Endometrial Neoplasms/pathology , Pregnancy Proteins/metabolism , Adult , Aged , Aged, 80 and over , Analysis of Variance , Biomarkers, Tumor/blood , Carcinoma/blood , Case-Control Studies , Endometrial Neoplasms/blood , Female , Humans , Middle Aged , Multivariate Analysis , Neoplasm Grading , Neoplasm Staging , Placenta Growth Factor , Pregnancy Proteins/blood , RNA, Messenger/metabolism , Statistics, Nonparametric
10.
Cancer Epidemiol Biomarkers Prev ; 22(2): 216-23, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23221126

ABSTRACT

BACKGROUND: Experimental and epidemiologic evidence have suggested that chronic inflammation may play a critical role in endometrial carcinogenesis. METHODS: To investigate this hypothesis, a two-stage study was carried out to evaluate single-nucleotide polymorphisms (SNP) in inflammatory pathway genes in association with endometrial cancer risk. In stage I, 64 candidate pathway genes were identified and 4,542 directly genotyped or imputed SNPs were analyzed among 832 endometrial cancer cases and 2,049 controls, using data from the Shanghai Endometrial Cancer Genetics Study. Linkage disequilibrium of stage I SNPs significantly associated with endometrial cancer (P < 0.05) indicated that the majority of associations could be linked to one of 24 distinct loci. One SNP from each of the 24 loci was then selected for follow-up genotyping. Of these, 21 SNPs were successfully designed and genotyped in stage II, which consisted of 10 additional studies including 6,604 endometrial cancer cases and 8,511 controls. RESULTS: Five of the 21 SNPs had significant allelic odds ratios (ORs) and 95% confidence intervals (CI) as follows: FABP1, 0.92 (0.85-0.99); CXCL3, 1.16 (1.05-1.29); IL6, 1.08 (1.00-1.17); MSR1, 0.90 (0.82-0.98); and MMP9, 0.91 (0.87-0.97). Two of these polymorphisms were independently significant in the replication sample (rs352038 in CXCL3 and rs3918249 in MMP9). The association for the MMP9 polymorphism remained significant after Bonferroni correction and showed a significant association with endometrial cancer in both Asian- and European-ancestry samples. CONCLUSIONS: These findings lend support to the hypothesis that genetic polymorphisms in genes involved in the inflammatory pathway may contribute to genetic susceptibility to endometrial cancer. Impact statement: This study adds to the growing evidence that inflammation plays an important role in endometrial carcinogenesis.


Subject(s)
Asian People/genetics , Biomarkers, Tumor/genetics , Endometrial Neoplasms/etiology , Genetic Predisposition to Disease , Inflammation/complications , Polymorphism, Single Nucleotide/genetics , Case-Control Studies , China/epidemiology , Endometrial Neoplasms/epidemiology , Female , Follow-Up Studies , Gene Expression Profiling , Humans , Inflammation/genetics , Linkage Disequilibrium , Middle Aged , Neoplasm Staging , Oligonucleotide Array Sequence Analysis , Prognosis , Risk Factors
11.
Gynecol Oncol ; 128(2): 327-34, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23219661

ABSTRACT

OBJECTIVE: Endometrial cancer patients may benefit from systemic adjuvant chemotherapy, alone or in combination with targeted therapies. Prognostic and predictive markers are needed, however, to identify patients amenable for these therapies. METHODS: Primary endometrial tumors were genotyped for >100 hot spot mutations in genes potentially acting as prognostic or predictive markers. Mutations were correlated with tumor characteristics in a discovery cohort, replicated in independent cohorts and finally, confirmed in the overall population (n=1063). RESULTS: PIK3CA, PTEN and KRAS mutations were most frequently detected, respectively in 172 (16.2%), 164 (15.4%) and 161 (15.1%) tumors. Binary logistic regression revealed that PIK3CA mutations were more common in high-grade tumors (OR=2.03; P=0.001 for grade 2 and OR=1.89; P=0.012 for grade 3 compared to grade 1), whereas a positive TP53 status correlated with type II tumors (OR=11.92; P<0.001) and PTEN mutations with type I tumors (OR=19.58; P=0.003). Conversely, FBXW7 mutations correlated with positive lymph nodes (OR=3.38; P=0.045). When assessing the effects of individual hot spot mutations, the H1047R mutation in PIK3CA correlated with high tumor grade and reduced relapse-free survival (HR=2.18; P=0.028). CONCLUSIONS: Mutations in PIK3CA, TP53, PTEN and FBXW7 correlate with high tumor grade, endometrial cancer type and lymph node status, whereas PIK3CA H1047R mutations serve as prognostic markers for relapse-free survival in endometrial cancer patients.


Subject(s)
Biomarkers, Tumor/genetics , Endometrial Neoplasms/genetics , Mutation , Aged , Carcinoma, Endometrioid/genetics , Carcinoma, Endometrioid/pathology , Cell Cycle Proteins/genetics , Class I Phosphatidylinositol 3-Kinases , DNA, Neoplasm/chemistry , DNA, Neoplasm/genetics , Endometrial Neoplasms/pathology , F-Box Proteins/genetics , F-Box-WD Repeat-Containing Protein 7 , Female , High-Throughput Nucleotide Sequencing , Humans , Middle Aged , Neoplasm Grading , Neoplasm Staging , PTEN Phosphohydrolase/genetics , Phosphatidylinositol 3-Kinases/genetics , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins p21(ras) , Tumor Suppressor Protein p53/genetics , Ubiquitin-Protein Ligases/genetics , ras Proteins/genetics
12.
Cancer Epidemiol Biomarkers Prev ; 21(6): 980-7, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22426144

ABSTRACT

BACKGROUND: Genome-wide association studies (GWAS) have identified more than 100 genetic loci for various cancers. However, only one is for endometrial cancer. METHODS: We conducted a three-stage GWAS including 8,492 endometrial cancer cases and 16,596 controls. After analyzing 585,963 single-nucleotide polymorphisms (SNP) in 832 cases and 2,682 controls (stage I) from the Shanghai Endometrial Cancer Genetics Study, we selected the top 106 SNPs for in silico replication among 1,265 cases and 5,190 controls from the Australian/British Endometrial Cancer GWAS (stage II). Nine SNPs showed results consistent in direction with stage I with P < 0.1. These nine SNPs were investigated among 459 cases and 558 controls (stage IIIa) and six SNPs showed a direction of association consistent with stages I and II. These six SNPs, plus two additional SNPs selected on the basis of linkage disequilibrium and P values in stage II, were investigated among 5,936 cases and 8,166 controls from an additional 11 studies (stage IIIb). RESULTS: SNP rs1202524, near the CAPN9 gene on chromosome 1q42.2, showed a consistent association with endometrial cancer risk across all three stages, with ORs of 1.09 [95% confidence interval (CI), 1.03-1.16] for the A/G genotype and 1.17 (95% CI, 1.05-1.30) for the G/G genotype (P = 1.6 × 10(-4) in combined analyses of all samples). The association was stronger when limited to the endometrioid subtype, with ORs (95% CI) of 1.11 (1.04-1.18) and 1.21 (1.08-1.35), respectively (P = 2.4 × 10(-5)). CONCLUSIONS: Chromosome 1q42.2 may host an endometrial cancer susceptibility locus. IMPACT: This study identified a potential genetic locus for endometrial cancer risk.


Subject(s)
Endometrial Neoplasms/genetics , Genome-Wide Association Study/methods , Case-Control Studies , Chromosomes, Human, Pair 14 , Endometrial Neoplasms/pathology , Female , Genetic Loci , Genetic Predisposition to Disease , Humans , Neoplasm Staging , Polymorphism, Single Nucleotide , Risk Factors
13.
Anticancer Res ; 31(6): 2367-71, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21737666

ABSTRACT

Endometrial stromal sarcoma (ESS) with primitive neuroectodermal differentiation is a very uncommon entity. Such a case presenting as stage IIIc (International Federation of Gynaecology and Obstetrics (FIGO) 2010) disease in a 51-year-old female is described. Microscopy suggested a small blue round cell tumour. Cytogenetic and multicolour fluorescent in situ hybridisation (M-FISH) analysis revealed a complex karyotype with the presence of unbalanced t(10;17)(q22;p13) translocation, indicating ESS. Peripheral Ewing´s sarcoma was excluded based on FISH and RT-PCR fusion transcripts analysis. After surgical staging, the patient received bleomycin-etoposide-cisplatin combination chemotherapy. A detailed analysis of the histopathology and genetic findings forms the basis of this report.


Subject(s)
Chromosomes, Human, Pair 10 , Chromosomes, Human, Pair 17 , Neuroectodermal Tumors/pathology , Sarcoma, Endometrial Stromal/genetics , Translocation, Genetic , Uterine Neoplasms/genetics , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Bleomycin/administration & dosage , Cell Differentiation/physiology , Cisplatin/administration & dosage , Etoposide/administration & dosage , Female , Humans , Neuroectodermal Tumors/drug therapy , Neuroectodermal Tumors/genetics , Phenotype , Sarcoma, Endometrial Stromal/drug therapy , Sarcoma, Endometrial Stromal/pathology , Uterine Neoplasms/drug therapy , Uterine Neoplasms/pathology
14.
Int J Gynecol Cancer ; 21(6): 1071-7, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21792013

ABSTRACT

BACKGROUND: In non-small cell lung cancer, expression of excision repair cross-complementation group 1 (ERCC1) and p53 correlates with platinum resistance and class III ß-tubulin with resistance to taxanes. The potential to personalize treatment in endometrial cancer remains uninvestigated. METHODS: Patients received platinum-based chemotherapy, with or without paclitaxel. Patients were divided into 2 groups: group A (n = 33) consisted of patients with early-stage endometrial cancer treated with adjuvant chemotherapy. Group B (n = 116) included cases with primary advanced or recurrent disease. Immunohistochemistry was performed to analyze the expression of ERCC1 and p53, for all cases, and class III ß-tubulin for cases treated with paclitaxel. The findings were correlated with response according to Response Criteria in Solid Tumors; recurrence-free, disease-specific survival; and established prognostic markers. RESULTS: The mean age of 149 patients was 64 years (range, 31-84 years). Distribution of histopathologic subtypes was as follows: 44 endometrioid (30%), 92 serous/clear cell (62%), and 13 carcinosarcomas (8%).In group A, 11 (33%) and 19 patients (58%) showed expression for ERCC1 and p53, respectively. Seven (78%) of nine patients receiving paclitaxel were positive for class III ß-tubulin. There was no correlation between expression of ERCC1, p53, or class III ß-tubulin and recurrence or survival. In group B, 25 (22%) and 61 patients (64%) were positive for ERCC1 and p53, respectively. Fifty-two (74%) of seventy patients receiving paclitaxel were positive for class III ß-tubulin. Only p53 expression correlated with survival (P = 0.01). CONCLUSIONS: In contrast to theoretical assumptions, the current study did not reveal evidence that the expression of ERCC1 and class III ß-tubulin predicts response to cytotoxic treatment and patient outcome in endometrial cancer.


Subject(s)
Endometrial Neoplasms/drug therapy , Neoplasm Recurrence, Local/drug therapy , Adult , Aged , Aged, 80 and over , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , DNA Repair , DNA-Binding Proteins/metabolism , Disease-Free Survival , Drug Resistance, Neoplasm , Endometrial Neoplasms/blood , Endometrial Neoplasms/metabolism , Endometrial Neoplasms/pathology , Endonucleases/metabolism , Female , Humans , Immunohistochemistry , Middle Aged , Neoplasm Recurrence, Local/blood , Neoplasm Recurrence, Local/metabolism , Neoplasm Recurrence, Local/pathology , Neoplasm Staging , Paclitaxel/administration & dosage , Retrospective Studies , Tubulin/metabolism , Tumor Suppressor Protein p53/metabolism
15.
Nat Genet ; 43(5): 451-4, 2011 May.
Article in English | MEDLINE | ID: mdl-21499250

ABSTRACT

Endometrial cancer is the most common malignancy of the female genital tract in developed countries. To identify genetic variants associated with endometrial cancer risk, we performed a genome-wide association study involving 1,265 individuals with endometrial cancer (cases) from Australia and the UK and 5,190 controls from the Wellcome Trust Case Control Consortium. We compared genotype frequencies in cases and controls for 519,655 SNPs. Forty seven SNPs that showed evidence of association with endometrial cancer in stage 1 were genotyped in 3,957 additional cases and 6,886 controls. We identified an endometrial cancer susceptibility locus close to HNF1B at 17q12 (rs4430796, P = 7.1 × 10(-10)) that is also associated with risk of prostate cancer and is inversely associated with risk of type 2 diabetes.


Subject(s)
Endometrial Neoplasms/genetics , Hepatocyte Nuclear Factor 1-beta/genetics , Adolescent , Adult , Aged , Aged, 80 and over , Case-Control Studies , Diabetes Mellitus, Type 2/genetics , Female , Genetic Predisposition to Disease , Genetic Variation , Genome-Wide Association Study , Humans , Male , Middle Aged , Odds Ratio , Polymorphism, Single Nucleotide , Prostatic Neoplasms/genetics , Risk Factors , Young Adult
16.
Cancer Res ; 70(16): 6537-47, 2010 Aug 15.
Article in English | MEDLINE | ID: mdl-20682798

ABSTRACT

Treatment of bone metastases is largely symptomatic and is still an unmet medical need. Current therapies mainly target the late phase of tumor-induced osteoclast activation and hereby inhibit further metastatic growth. This treatment method is, however, less effective in preventing initial tumor engraftment, a process that is supposed to depend on the bone microenvironment. We explored whether bone-derived placental growth factor (PlGF), a homologue of vascular endothelial growth factor-A, regulates osteolytic metastasis. Osteogenic cells secrete PlGF, the expression of which is enhanced by bone-metastasizing breast tumor cells. Selective neutralization of host-derived PlGF by anti-mouse PlGF (alphaPlGF) reduced the incidence, number, and size of bone metastases, and preserved bone mass. alphaPlGF did not affect metastatic tumor angiogenesis but inhibited osteoclast formation by preventing the upregulation of the osteoclastogenic cytokine receptor activator of NF-kappaB ligand in osteogenic cells, as well as by blocking the autocrine osteoclastogenic activity of PlGF. alphaPlGF also reduced the engraftment of tumor cells in the bone and inhibited their interaction with matrix components in the metastatic niche. alphaPlGF therefore inhibits not only the progression of metastasis but also the settlement of tumor in the bone. These findings identify novel properties of PlGF and suggest that alphaPlGF might offer opportunities for adjuvant therapy of bone metastasis.


Subject(s)
Bone Neoplasms/secondary , Breast Neoplasms/pathology , Osteoclasts/pathology , Pregnancy Proteins/antagonists & inhibitors , Pregnancy Proteins/metabolism , Animals , Antibodies, Monoclonal/pharmacology , Bone Neoplasms/metabolism , Breast Neoplasms/metabolism , Cell Differentiation/physiology , Cell Line, Tumor , Female , Humans , Lung Neoplasms/secondary , Male , Mice , Mice, Inbred C57BL , Mice, Nude , Osteoclasts/metabolism , Placenta Growth Factor , Transplantation, Heterologous
17.
Cell ; 141(1): 178-90, 2010 Apr 02.
Article in English | MEDLINE | ID: mdl-20371353

ABSTRACT

Our findings that PlGF is a cancer target and anti-PlGF is useful for anticancer treatment have been challenged by Bais et al. Here we take advantage of carcinogen-induced and transgenic tumor models as well as ocular neovascularization to report further evidence in support of our original findings of PlGF as a promising target for anticancer therapies. We present evidence for the efficacy of additional anti-PlGF antibodies and their ability to phenocopy genetic deficiency or silencing of PlGF in cancer and ocular disease but also show that not all anti-PlGF antibodies are effective. We also provide additional evidence for the specificity of our anti-PlGF antibody and experiments to suggest that anti-PlGF treatment will not be effective for all tumors and why. Further, we show that PlGF blockage inhibits vessel abnormalization rather than density in certain tumors while enhancing VEGF-targeted inhibition in ocular disease. Our findings warrant further testing of anti-PlGF therapies.


Subject(s)
Neovascularization, Physiologic/drug effects , Pregnancy Proteins/antagonists & inhibitors , Pregnancy Proteins/metabolism , Angiogenesis Inhibitors/therapeutic use , Animals , Antibodies, Monoclonal/therapeutic use , Carcinoma, Hepatocellular/blood supply , Carcinoma, Hepatocellular/prevention & control , Choroid/blood supply , Disease Models, Animal , Eye Diseases/pathology , Humans , Liver Neoplasms, Experimental/blood supply , Liver Neoplasms, Experimental/prevention & control , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Transgenic , Papilloma/blood supply , Papilloma/chemically induced , Papilloma/prevention & control , Placenta Growth Factor , Skin Neoplasms/blood supply , Skin Neoplasms/chemically induced , Skin Neoplasms/prevention & control
18.
EMBO J ; 29(2): 424-41, 2010 Jan 20.
Article in English | MEDLINE | ID: mdl-20010698

ABSTRACT

Vascular endothelial growth factor (VEGF) and beta-catenin both act broadly in embryogenesis and adulthood, including in the skeletal and vascular systems. Increased or deregulated activity of these molecules has been linked to cancer and bone-related pathologies. By using novel mouse models to locally increase VEGF levels in the skeleton, we found that embryonic VEGF over-expression in osteo-chondroprogenitors and their progeny largely pheno-copied constitutive beta-catenin activation. Adult induction of VEGF in these cell populations dramatically increased bone mass, associated with aberrant vascularization, bone marrow fibrosis and haematological anomalies. Genetic and pharmacological interventions showed that VEGF increased bone mass through a VEGF receptor 2- and phosphatidyl inositol 3-kinase-mediated pathway inducing beta-catenin transcriptional activity in endothelial and osteoblastic cells, likely through modulation of glycogen synthase kinase 3-beta phosphorylation. These insights into the actions of VEGF in the bone and marrow environment underscore its power as pleiotropic bone anabolic agent but also warn for caution in its therapeutic use. Moreover, the finding that VEGF can modulate beta-catenin activity may have widespread physiological and clinical ramifications.


Subject(s)
Bone and Bones/metabolism , Bone and Bones/pathology , Gene Expression Regulation, Developmental , Vascular Endothelial Growth Factor A/metabolism , beta Catenin/metabolism , Animals , Bone and Bones/embryology , Cell Differentiation , Cell Proliferation , Cells, Cultured , Endothelial Cells/cytology , Humans , Mesoderm/cytology , Mice , Mice, Transgenic , Morphogenesis , Osteoblasts/cytology , Phosphatidylinositol 3-Kinases/metabolism , Stem Cells/cytology , Stromal Cells/cytology , Vascular Endothelial Growth Factor A/genetics , beta Catenin/genetics
19.
J Clin Invest ; 116(5): 1230-42, 2006 May.
Article in English | MEDLINE | ID: mdl-16614757

ABSTRACT

Current therapies for delayed- or nonunion bone fractures are still largely ineffective. Previous studies indicated that the VEGF homolog placental growth factor (PlGF) has a more significant role in disease than in health. Therefore we investigated the role of PlGF in a model of semi-stabilized bone fracture healing. Fracture repair in mice lacking PlGF was impaired and characterized by a massive accumulation of cartilage in the callus, reminiscent of delayed- or nonunion fractures. PlGF was required for the early recruitment of inflammatory cells and the vascularization of the fracture wound. Interestingly, however, PlGF also played a role in the subsequent stages of the repair process. Indeed in vivo and in vitro findings indicated that PlGF induced the proliferation and osteogenic differentiation of mesenchymal progenitors and stimulated cartilage turnover by particular MMPs. Later in the process, PlGF was required for the remodeling of the newly formed bone by stimulating osteoclast differentiation. As PlGF expression was increased throughout the process of bone repair and all the important cell types involved expressed its receptor VEGFR-1, the present data suggest that PlGF is required for mediating and coordinating the key aspects of fracture repair. Therefore PlGF may potentially offer therapeutic advantages for fracture repair.


Subject(s)
Bone Remodeling , Cartilage/cytology , Fracture Healing , Mesoderm/cytology , Pregnancy Proteins/physiology , Animals , Cartilage/metabolism , Cell Differentiation , Cell Proliferation , Inflammation , Mice , Mice, Transgenic , Models, Biological , Osteoclasts/cytology , Placenta Growth Factor , Vascular Endothelial Growth Factor Receptor-1/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...