Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
1.
J Cancer Res Clin Oncol ; 139(2): 201-9, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23015072

ABSTRACT

PURPOSE: Targeted therapies using the anti-EGFR antibodies panitumumab (Pmab) or cetuximab (Cmab) are currently restricted to patients with metastatic colorectal adenocarcinoma whose tumours do not show a mutation in KRAS. However, recent retrospective studies indicated that patients with tumours mutated in codon 13 of KRAS may benefit from treatment with Cmab in contrast to patients with tumours mutated in KRAS codon 12. METHODS: To study the functional impact of the subtype of KRAS mutations on the efficiency of EGFR-targeted therapies, we correlated the KRAS mutation status of 15 colorectal carcinoma cell lines with the in vitro sensitivity of these cells to Cmab/Pmab. Mutations in the potential predictive biomarkers BRAF and PIK3CA as well as protein expression of EGFR and PTEN were also determined. RESULTS: Four out of seven KRAS-mutated cell lines were characterised by the p.G13D mutation. Treatment of these cells using Cmab/Pmab induced a significant growth inhibition in contrast to cell lines showing a KRAS mutation at codon 12 or 61. Out of the eight KRAS wild-type cell lines, five were insensitive to Cmab/Pmab. These cell lines were characterised either by BRAF mutation or by absence of EGFR or PTEN protein expression. CONCLUSIONS: Since KRAS p.G13D-mutated tumour cells may respond to EGFR-targeted therapy, we suggest including subtype analysis of KRAS mutations in prospective clinical trials. In KRAS wild-type tumour cells, BRAF mutations and loss of EGFR or PTEN expression may lead to resistance to EGFR-targeted therapy and should be considered as additional negative predictive biomarkers.


Subject(s)
Antibodies, Monoclonal/pharmacology , Antineoplastic Agents/pharmacology , Colorectal Neoplasms/genetics , Drug Resistance, Neoplasm/genetics , Mutation , Proto-Oncogene Proteins/genetics , ras Proteins/genetics , Antibodies, Monoclonal, Humanized , Cell Line, Tumor , Cell Proliferation/drug effects , Cetuximab , Class I Phosphatidylinositol 3-Kinases , Colorectal Neoplasms/metabolism , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/genetics , ErbB Receptors/metabolism , Gene Expression Regulation, Neoplastic , Humans , Panitumumab , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins B-raf/genetics , Proto-Oncogene Proteins B-raf/metabolism , Proto-Oncogene Proteins p21(ras) , ras Proteins/metabolism
2.
Clin Sarcoma Res ; 2(1): 24, 2012 Dec 18.
Article in English | MEDLINE | ID: mdl-23249693

ABSTRACT

BACKGROUND: Chemokine receptor CXCR4, together with its ligand CXCL12, plays critical roles in cancer progression, including growth, metastasis and angiogenesis. Ewing sarcoma is a sarcoma with poor prognosis despite current therapies, particularly for patients with advanced-stage disease. Lungs and bone (marrow), organs of predilection for (primary/metastatic) Ewing sarcoma, represent predominant CXCL12 sources. METHODS: To gain insight into the role of the CXCR4-CXCL12 axis in Ewing sarcoma, CXCR4, CXCL12 and hypoxia-inducible factor-1α protein expression was studied in therapy-naïve and metastatic tumors by immunohistochemistry. CXCR4 function was assessed in vitro, by flow cytometry and proliferation/ cell viability assays, in the presence of recombinant CXCL12 and/or CXCR4-antagonist AMD3100 or under hypoxic conditions. RESULTS: Whereas CXCR4 was predominantly expressed by tumor cells, CXCL12 was observed in both tumor and stromal areas. Survival analysis revealed an (expression level-dependent) negative impact of CXCR4 expression (p < 0.04). A role for the CXCR4-CXCL12 axis in Ewing sarcoma growth was suggested by our observations that i) CXCR4 expression correlated positively with tumor volume at diagnosis (p = 0.013), ii) CXCL12 was present within the microenvironment of virtually all cases, iii) CXCL12 induced proliferation of CXCR4-positive Ewing sarcoma cell lines, which could be abrogated by AMD3100. CXCR4 expression was not correlated with occurrence of metastatic disease. Also, therapy-naïve tumors demonstrated higher CXCR4 expression as compared to metastases (p = 0.027). Evaluation of in vivo hypoxia-inducible factor-1α expression and culture of cells under hypoxic conditions revealed no role for hypoxia in CXCR4 expression. CONCLUSIONS: Together, our results imply a crucial role for the CXCR4-CXCL12 axis in auto- and/or paracrine growth stimulation. Integration of CXCR4-targeting strategies into first- and/or second-line treatment regimens may represent a promising treatment option for Ewing sarcoma.

3.
J Pathol ; 226(5): 796-805, 2012 Apr.
Article in English | MEDLINE | ID: mdl-21960059

ABSTRACT

Identification of factors to detect chemotherapy-resistant tumours at diagnosis is a first priority for risk-adapted therapy in the oncology of children and young adults, where more individualized, effective, and less toxic treatments are highly desirable. In this study, we analysed the miRNAs discriminating Ewing's sarcoma (EWS) patients with different clinical outcomes in order to identify new indicators of prognosis. miRNA expression was investigated in 49 primary EWSs by using the Agilent human miRNA microarray v.2 and/or qRT-PCR. Statistical power of the samples studied for miRNA expression was verified, indicating adequate sample size. Microarray analysis defined a signature of five miRNAs (miR-34a, miR-23a, miR-92a, miR-490-3p, and miR-130b) as an independent predictor of risk for disease progression and survival. Validation analysis in the extended sample set indicated that both miR-34a and miR-490-3p achieved sufficient statistical power to predict prognosis. Results were particularly robust for miR-34a, which appeared associated with either event-free or overall survival and emerged as a significant predictor also after multivariate analysis. Patients with the highest expression of miR-34a did not experience adverse events in 5 years; in contrast, patients with the lowest expression recurred within 2 years. High expression of miR34a can be detected also in paraffin-embedded tissues by in situ hybridization, thus contributing to an easy routine evaluation of this miRNA. Functional analysis of miR-34a in EWS cell lines indicated that when miR-34a expression was enforced, cells were less proliferative, less malignant, and sensitized to doxorubicin and vincristine. Expression of miR-34a could be increased in p53wt cells by treatment with nutlin-3a. Accordingly, nutlin-3a synergizes with doxorubicin. Overall, our data indicate that miR-34a expression is a strong predictor of outcome in EWS. Restoration of miR-34a activity may be useful to decrease malignancy and increase tumour sensitivity to current drugs, so sparing excessive long-term toxicity to EWS patients.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Biomarkers, Tumor/metabolism , Bone Neoplasms/drug therapy , Bone Neoplasms/metabolism , MicroRNAs/metabolism , Sarcoma, Ewing/drug therapy , Sarcoma, Ewing/metabolism , Bone Neoplasms/genetics , Bone Neoplasms/mortality , Bone Neoplasms/pathology , Cell Line, Tumor , Disease-Free Survival , Dose-Response Relationship, Drug , Doxorubicin/pharmacology , Drug Resistance, Neoplasm/genetics , Drug Synergism , Humans , Imidazoles/pharmacology , In Situ Hybridization , Inhibitory Concentration 50 , Kaplan-Meier Estimate , Multivariate Analysis , Oligonucleotide Array Sequence Analysis , Paraffin Embedding , Piperazines/pharmacology , Proportional Hazards Models , Real-Time Polymerase Chain Reaction , Reproducibility of Results , Retrospective Studies , Risk Assessment , Risk Factors , Sarcoma, Ewing/genetics , Sarcoma, Ewing/mortality , Sarcoma, Ewing/pathology , Time Factors , Transfection , Treatment Outcome , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Vincristine/pharmacology
4.
Lab Invest ; 91(8): 1195-205, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21519327

ABSTRACT

Cancer cell lines represent in vitro models for studying malignancies, general cell biology, drug discovery and more. Whether they can be considered as exact representative models of the parental tumors remains uncertain given the acquisition of additional ex vivo changes of the cells and the lack of tissue architecture and stroma. Previously, within the EuroBoNeT consortium, we characterized a collection of bone sarcoma cell lines on genomic and proteomic level. Here, we address the phenotypical and functional characterization of the unique set of osteosarcoma cell lines (n=19) in vitro and in vivo. For functional analysis of differentiation capacity, cells were stimulated towards osteoblasts, adipocytes and chondrocytes. Furthermore, all cell lines were injected subcutaneously and intramuscularly into nude mice to assay their in vivo tumor formation capacity as well as for phenotypical analysis of the tumors. All formed tumors were further characterized histologically and immunohistochemically. Out of 19 cell lines, 17 (89%) showed adipogenic differentiation, 13/19 (68%) could differentiate towards osteoblasts and in 6/19 (32%) cell lines chondrogenic differentiation was evident. About half of the cell lines (8/19, 42%) produced tumors in vivo after subcutaneous and intramuscular injections. Several cell lines showed invasion into adjacent tissues and one tumor developed several lung metastases. The use of cell lines, especially in cancer research, is of paramount importance. Here, we identify comprehensively characterized osteosarcoma cell lines, which robustly represent clinical osteosarcoma providing researchers useful in vitro and in vivo models to study the genetics and functional characteristics of this highly malignant neoplasm.


Subject(s)
Bone Neoplasms/metabolism , Osteosarcoma/metabolism , Sarcoma, Experimental/metabolism , Animals , Bone Neoplasms/pathology , Cell Differentiation , Cell Line, Tumor , Gene Expression Profiling , Humans , Immunohistochemistry , Mice , Mice, Nude , Neoplasm Metastasis , Neoplasm Transplantation , Osteosarcoma/pathology , Sarcoma, Experimental/pathology
5.
Pediatr Blood Cancer ; 57(3): 406-14, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21254373

ABSTRACT

BACKGROUND: Alveolar Rhabdomyosarcomas (RMA) are characterized by chromosomal translocations, fusing the PAX3 or PAX7 gene with FKHR in about 85%. Previous studies have suggested that the fusion type is associated with prognosis. In order to investigate the predictive value of the PAX-FKHR fusion status on disease outcome of patients with RMA treated in the CWS trials we performed a retrospective analysis. PROCEDURE: Between 1986 and 2004, out of 446 patients with RMA treated in four consecutive CWS trials, tumor samples from 126 patients were available for RT-PCR analysis. Survival depending on fusion status in context with known clinical risk-factors was analyzed. RESULTS: Out of 126 samples, 121 had adequate quality for PAX-FKHR fusion status analysis. PAX-FKHR fusions were detected in 101 samples: 60% PAX3-FKHR and 24% PAX7-FKHR fusions, 17% were fusion-negative. There was no significant difference in survival between patients with PAX3-FKHR versus PAX7-FKHR positive tumors. The fusion transcript negative cohort showed a more favorable outcome than the fusion transcript positive cohort among patients with metastatic disease. From the established clinical risk-factors none was associated with a significantly higher risk of failure or death in a multivariate analysis. CONCLUSIONS: PAX-FKHR fusion type was not a significant predictor for survival in our analysis. More extensive molecular analyses are needed to identify features with prognostic relevance and useful therapeutic impact.


Subject(s)
Oncogene Proteins, Fusion/analysis , Predictive Value of Tests , Rhabdomyosarcoma, Alveolar/diagnosis , Adolescent , Child , Child, Preschool , Clinical Trials as Topic , Female , Humans , Male , Prognosis , Retrospective Studies , Rhabdomyosarcoma, Alveolar/genetics , Rhabdomyosarcoma, Alveolar/pathology , Young Adult
6.
BMC Cancer ; 10: 372, 2010 Jul 16.
Article in English | MEDLINE | ID: mdl-20637078

ABSTRACT

BACKGROUND: Hypoxia regulates gene expression via the transcription factor HIF (Hypoxia-Inducible Factor). Little is known regarding HIF expression and function in primary bone sarcomas. We describe HIF expression and phenotypic effects of hypoxia, hypoglycaemia and HIF in Ewing's sarcoma and osteosarcoma. METHODS: HIF-1alpha and HIF-2alpha immunohistochemistry was performed on a Ewing's tumour tissue array. Ewing's sarcoma and osteosarcoma cell lines were assessed for HIF pathway induction by Western blot, luciferase assay and ELISA. Effects of hypoxia, hypoglycaemia and isoform-specific HIF siRNA were assessed on proliferation, apoptosis and migration. RESULTS: 17/56 Ewing's tumours were HIF-1alpha-positive, 15 HIF-2alpha-positive and 10 positive for HIF-1alpha and HIF-2alpha. Expression of HIF-1alpha and cleaved caspase 3 localised to necrotic areas. Hypoxia induced HIF-1alpha and HIF-2alpha in Ewing's and osteosarcoma cell lines while hypoglycaemia specifically induced HIF-2alpha in Ewing's. Downstream transcription was HIF-1alpha-dependent in Ewing's sarcoma, but regulated by both isoforms in osteosarcoma. In both cell types hypoglycaemia reduced cellular proliferation by >or= 45%, hypoxia increased apoptosis and HIF siRNA modulated hypoxic proliferation and migration. CONCLUSIONS: Co-localisation of HIF-1alpha and necrosis in Ewing's sarcoma suggests a role for hypoxia and/or hypoglycaemia in in vivo induction of HIF. In vitro data implicates hypoxia as the primary HIF stimulus in both Ewing's and osteosarcoma, driving effects on proliferation and apoptosis. These results provide a foundation from which to advance understanding of HIF function in the pathobiology of primary bone sarcomas.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Bone Neoplasms/pathology , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Hypoxia/pathology , Osteosarcoma/pathology , Sarcoma, Ewing/pathology , Apoptosis , Basic Helix-Loop-Helix Transcription Factors/antagonists & inhibitors , Basic Helix-Loop-Helix Transcription Factors/genetics , Blotting, Western , Bone Neoplasms/genetics , Bone Neoplasms/metabolism , Cell Adhesion , Cell Line, Tumor , Cell Movement , Cell Proliferation , Enzyme-Linked Immunosorbent Assay , Humans , Hypoxia/genetics , Hypoxia/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/antagonists & inhibitors , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Immunoenzyme Techniques , Osteosarcoma/genetics , Osteosarcoma/metabolism , Phenotype , RNA, Messenger/genetics , RNA, Small Interfering/pharmacology , Reverse Transcriptase Polymerase Chain Reaction , Sarcoma, Ewing/genetics , Sarcoma, Ewing/metabolism
7.
J Clin Oncol ; 28(12): 1982-8, 2010 Apr 20.
Article in English | MEDLINE | ID: mdl-20308673

ABSTRACT

PURPOSE EWS-ETS fusion genes are the driving force in Ewing's sarcoma pathogenesis. Because of the variable breakpoint locations in the involved genes, there is heterogeneity in fusion RNA and protein architecture. Since previous retrospective studies suggested prognostic differences among patients expressing different EWS-FLI1 fusion types, the impact of fusion RNA architecture on disease progression and relapse was studied prospectively within the Euro-E.W.I.N.G. 99 clinical trial. PATIENTS AND METHODS Among 1,957 patients who registered before January 1, 2007, 703 primary tumors were accessible for the molecular biology study. Fusion type was assessed by polymerase chain reaction on frozen (n = 578) or paraffin-embedded materials (n = 125). The primary end point was the time to disease progression or relapse. Results After exclusion of noninformative patients, 565 patients were entered into the prognostic factor analysis comparing type 1 (n = 296), type 2 (n = 133), nontype 1/nontype 2 EWS-FLI1 (n = 91) and EWS-ERG fusions (n = 45). Median follow-up time was 4.5 years. The distribution of sex, age, tumor volume, tumor site, disease extension, or histologic response did not differ between the four fusion type groups. We did not observe any significant prognostic value of the fusion type on the risk of progression or relapse. The only slight difference was that the risk of progression or relapse associated with nontype 1/nontype 2 EWS-FLI1 fusions was 1.38 (95% CI, 0.96 to 2.0) times higher than risk associated with other fusion types, but it was not significant (P = .10). CONCLUSION In contrast to retrospective studies, the prospective evaluation did not confirm a prognostic benefit for type 1 EWS-FLI1 fusions.


Subject(s)
Bone Neoplasms/genetics , Gene Expression Regulation, Neoplastic , Oncogene Proteins, Fusion/genetics , Proto-Oncogene Protein c-fli-1/genetics , Sarcoma, Ewing/genetics , Transcription Factors/genetics , Adult , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Biopsy , Bone Neoplasms/mortality , Bone Neoplasms/pathology , Bone Neoplasms/therapy , Chi-Square Distribution , Disease Progression , Europe , Female , Genetic Predisposition to Disease , Humans , Kaplan-Meier Estimate , Male , Neoplasm Recurrence, Local , Paraffin Embedding , Phenotype , Proportional Hazards Models , Prospective Studies , RNA-Binding Protein EWS , Radiotherapy, Adjuvant , Reverse Transcriptase Polymerase Chain Reaction , Risk Assessment , Risk Factors , Sarcoma, Ewing/mortality , Sarcoma, Ewing/secondary , Sarcoma, Ewing/therapy , Stem Cell Transplantation , Time Factors , Treatment Outcome , Young Adult
8.
Genes Chromosomes Cancer ; 49(1): 40-51, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19787792

ABSTRACT

Usage of cancer cell lines has repeatedly generated conflicting results provoked by differences among subclones or contamination with mycoplasm or other immortal mammalian cells. To overcome these limitations, we decided within the EuroBoNeT consortium to characterize a common set of cell lines including osteosarcomas (OS), Ewing sarcomas (ES), and chondrosarcomas (CS). DNA fingerprinting was used to guarantee the identity of all of the cell lines and to distinguish subclones of osteosarcoma cell line HOS. Screening for homozygous loss of 38 tumor suppressor genes by MLPA revealed deletion of CDKN2A as the most common event (15/36), strictly associated with absence of the CDKN2A (p16) protein. Ten cell lines showed missense mutations of the TP53 gene while another set of nine cell lines showed mutations resulting in truncation of the TP53 protein. Cells harboring missense mutations expressed high levels of nuclear TP53, while cell lines with nonsense mutations showed weak/absent staining for TP53. TP53(wt) cell lines usually expressed the protein in 2-10% of the cells. However, seven TP53(wt) osteosarcomas were negative for both mRNA and protein expression. Our analyses shed light on the correlation between immunohistochemical and genetic data for CDKN2A and TP53, and confirm the importance of these signaling pathways. The characterization of a substantial number of cell lines represents an important step to supply research groups with proven models for further advanced studies on tumor biology and may help to make results from different laboratories more comparable.


Subject(s)
Biomedical Research , Bone Neoplasms/pathology , Cell Line, Tumor , Animals , Cooperative Behavior , Cyclin-Dependent Kinase Inhibitor p16 , Europe , Humans , Tumor Suppressor Protein p53
9.
Genes Chromosomes Cancer ; 48(8): 679-93, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19441093

ABSTRACT

Osteosarcomas are the most common primary malignant tumor of bone, and almost all conventional osteosarcomas are high-grade tumors with complex karyotypes. We have examined DNA copy number changes in 36 osteosarcoma tumors and 20 cell lines using microarray-based comparative genomic hybridization. The most frequent minimal recurrent regions of gain identified in the tumor samples were in 1q21.2-q21.3 (78% of the samples), 1q21.3-q22 (78%), and 8q22.1 (72%). Minimal recurrent regions in 10q22.1-q22.2 (81%), 6q16.1 (67%), 13q14.2 (67%), and 13q21.1 (67%) were most frequently lost. A small region in 3q13.31 (2.1 Mb) containing the gene limbic system-associated membrane protein (LSAMP) was frequently deleted (56%). LSAMP has previously been reported to be a candidate tumor suppressor gene in other cancer types. The deletion was validated using fluorescence in situ hybridization, and the expression level and promoter methylation status of LSAMP were investigated using quantitative real-time reverse transcription PCR and methylation-specific PCR, respectively. LSAMP showed low expression compared to two normal bone samples in 6/15 tumors and 5/9 cell lines with deletion of 3q13.31, and also in 5/14 tumors and 3/11 cell lines with normal copy number or gain. Partial or full methylation of the investigated CpG island was identified in 3/30 tumors and 7/20 cell lines. Statistical analyses revealed that loss of 11p15.4-p15.3 and low expression of LSAMP (both P = 0.011) were significantly associated with poor survival. Our results show that LSAMP is a novel candidate tumor suppressor gene in osteosarcomas.


Subject(s)
Bone Neoplasms/genetics , Cell Adhesion Molecules, Neuronal/genetics , Comparative Genomic Hybridization/methods , Genes, Tumor Suppressor , Oligonucleotide Array Sequence Analysis/methods , Osteosarcoma/genetics , Adolescent , Adult , Aged , Bone Neoplasms/metabolism , Cell Adhesion Molecules, Neuronal/metabolism , Cell Line, Tumor , Child , Chromosome Aberrations , Cluster Analysis , CpG Islands/genetics , DNA Methylation , Data Interpretation, Statistical , Female , GPI-Linked Proteins , Gene Dosage , Gene Expression Profiling , Humans , In Situ Hybridization, Fluorescence , Kaplan-Meier Estimate , Male , Middle Aged , Osteosarcoma/metabolism , Reproducibility of Results , Reverse Transcriptase Polymerase Chain Reaction
10.
J Pathol ; 218(2): 222-31, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19274709

ABSTRACT

Ewing sarcoma (EWS) is a tumour most commonly arising in bone, although on occasion in soft tissue, with a poor prognosis in patients with refractory or relapsed disease, despite multimodal therapy. Immunotherapeutic strategies based on tumour-reactive T and/or natural killer cells may improve the treatment of advanced-stage EWS. Since cellular immune recognition critically depends on human leukocyte antigen (HLA) expression, knowledge about HLA expression in EWS is crucial in the design of cellular immunotherapeutic strategies. Constitutive and IFNgamma-induced HLA class I expression was analysed in EWS cell lines (n = 6) by flow cytometry, using antibodies against both monomorphic and allele-specific antigens. Expression of antigen processing pathway components and beta-2 microglobulin (beta2m) was assessed by western blot. Expression of class II transactivator (CIITA), and its contribution to HLA class II expression, was evaluated by qRT-PCR, transduction assays, and flow cytometry. beta2m/HLA class I and class II expression was validated in EWS tumours (n = 67) by immunofluorescence. Complete or partial absence of HLA class I expression was observed in 79% of EWS tumours. Lung metastases consistently lacked HLA class I and sequential tumours demonstrated a tendency towards decreased expression upon disease progression. Together with absent or low constitutive expression levels of specific HLA class I loci and alleles, and differential induction of identical alleles by IFNgamma in different cell lines, these results may reflect the existence of an immune escape mechanism. Inducible expression of TAP-1/-2, tapasin, LMP-2/-7, and the beta2m/HLA class I complex by IFNgamma suggests that regulatory mechanisms are mainly responsible for heterogeneity in constitutive class I expression. EWSs lack IFNgamma-inducible HLA class II, due to lack of functional CIITA. The majority of EWS tumours, particularly if advanced-stage, exhibit complete or partial absence of both classes of HLA. This knowledge will be instrumental in the design of cellular immunotherapeutic strategies for advanced-stage EWS.


Subject(s)
Bone Neoplasms/immunology , Histocompatibility Antigens Class I/analysis , Sarcoma, Ewing/immunology , Adolescent , Antigen Presentation , Biomarkers/analysis , Blotting, Western , Bone Neoplasms/mortality , Bone Neoplasms/secondary , Cell Line, Tumor , Flow Cytometry , HLA-DR Antigens/genetics , Histocompatibility Antigens Class II/genetics , Humans , Immunization , Immunohistochemistry , Interferon-gamma/pharmacology , Kaplan-Meier Estimate , Nuclear Proteins/genetics , Sarcoma, Ewing/mortality , Sarcoma, Ewing/secondary , Statistics, Nonparametric , Trans-Activators/genetics , Tumor Cells, Cultured , Tumor Escape , beta 2-Microglobulin/analysis
11.
Hum Pathol ; 40(6): 893-7, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19269015

ABSTRACT

Peripheral primitive neuroectodermal tumor/Ewing's tumors are rare bone and soft tissue malignancies with a highly aggressive clinical course and early metastases occurring at multiple peripheral sites. Here, we present for the first time a case of a 46-year-old man with a primary peripheral primitive neuroectodermal tumor/Ewing's tumor of the testis. The diagnosis of peripheral primitive neuroectodermal tumor/Ewing's tumor was established by histology, immunohistochemistry, and molecular pathology. The tumor revealed a rapid progress in 2 months' time. Therefore, the patient was included in the EURO-E.W.I.N.G.99 study and was placed on chemotherapy. However, the tumor progressed during ongoing therapy, and the patient died in March 2008. In conclusion, though being reported here for the first time, peripheral primitive neuroectodermal tumor/Ewing's tumors should be considered in the differential diagnosis of blue round cell tumors of the testis. A rapid and correct diagnosis of this entity is crucial for fast and accurate therapy, which is stressed by the fatal case presented here.


Subject(s)
Neuroectodermal Tumors, Primitive, Peripheral/pathology , Sarcoma, Ewing/pathology , Testicular Neoplasms/pathology , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Diagnosis, Differential , Fatal Outcome , Humans , Male , Middle Aged , Neuroectodermal Tumors, Primitive, Peripheral/diagnosis , Neuroectodermal Tumors, Primitive, Peripheral/drug therapy , Sarcoma, Ewing/diagnosis , Sarcoma, Ewing/drug therapy , Testicular Neoplasms/diagnosis , Testicular Neoplasms/drug therapy
12.
Cancer Res ; 68(17): 7100-9, 2008 Sep 01.
Article in English | MEDLINE | ID: mdl-18757425

ABSTRACT

Although p53 is the most frequently mutated gene in cancer, half of human tumors retain wild-type p53, whereby it is unknown whether normal p53 function is compromised by other cancer-associated alterations. One example is Ewing's sarcoma family tumors (ESFT), where 90% express wild-type p53. ESFT are characterized by EWS-FLI1 oncogene fusions. Studying 6 ESFT cell lines, silencing of EWS-FLI1 in a wild-type p53 context resulted in increased p53 and p21(WAF1/CIP1) levels, causing cell cycle arrest. Using a candidate gene approach, HEY1 was linked to p53 induction. HEY1 was rarely expressed in 59 primary tumors, but consistently induced upon EWS-FLI1 knockdown in ESFT cell lines. The NOTCH signaling pathway targets HEY1, and we show NOTCH2 and NOTCH3 to be expressed in ESFT primary tumors and cell lines. Upon EWS-FLI1 silencing, NOTCH3 processing accompanied by nuclear translocation of the activated intracellular domain was observed in all but one p53-mutant cell line. In cell lines with the highest HEY1 induction, NOTCH3 activation was the consequence of JAG1 transcriptional induction. JAG1 modulation by specific siRNA, NOTCH-processing inhibition by either GSI or ectopic NUMB1, and siRNA-mediated HEY1 knockdown all inhibited p53 and p21(WAF1/CIP1) induction. Conversely, forced expression of JAG1, activated NOTCH3, or HEY1 induced p53 and p21(WAF1/CIP1). These results indicate that suppression of EWS-FLI1 reactivates NOTCH signaling in ESFT cells, resulting in p53-dependent cell cycle arrest. Our data link EWS-FLI1 to the NOTCH and p53 pathways and provide a plausible basis both for NOTCH tumor suppressor effects and oncogenesis of cancers that retain wild-type p53.


Subject(s)
Genes, p53 , Oncogene Proteins, Fusion/physiology , Receptors, Notch/physiology , Sarcoma, Ewing/metabolism , Transcription Factors/physiology , Base Sequence , Basic Helix-Loop-Helix Transcription Factors/genetics , Blotting, Western , Cell Cycle Proteins/genetics , Cell Line, Tumor , Cyclin-Dependent Kinase Inhibitor p21/genetics , DNA Primers , Fluorescent Antibody Technique , Gene Silencing , Humans , Oligonucleotide Array Sequence Analysis , Oncogene Proteins, Fusion/genetics , Proto-Oncogene Protein c-fli-1 , RNA-Binding Protein EWS , Reverse Transcriptase Polymerase Chain Reaction , Transcription Factors/genetics
13.
Eur J Cancer ; 44(5): 699-709, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18294840

ABSTRACT

In Ewing's sarcoma family of tumours (ESFT), the clinically most adverse prognostic parameters are the presence of tumour metastasis at time of diagnosis and poor response to neoadjuvant chemotherapy. To identify genes differentially regulated between metastatic and localised tumours, we analysed 27 ESFT specimens using Affymetrix microarrays. Functional annotation of differentially regulated genes revealed 29 over-represented pathways including PDGF, TP53, NOTCH, and WNT1-signalling. Regression of primary tumours (n=20) induced by polychemotherapy was found to be correlated with the expression of genes involved in angiogenesis, apoptosis, ubiquitin proteasome pathway, and PI3 kinase and p53 pathways. These findings could be confirmed by in vitro cytotoxicity assays. A set of 46 marker genes correctly classifies these 20 tumours as responding versus non-responding. We conclude that expression signatures of initial tumour biopsies can help to identify ESFT patients at high risk to develop tumour metastasis or to suffer from a therapy refractory cancer.


Subject(s)
Bone Neoplasms/genetics , Gene Expression Profiling/methods , Microarray Analysis/methods , Sarcoma, Ewing/genetics , Adolescent , Adult , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Bone Neoplasms/drug therapy , Cell Communication/genetics , Cell Line, Tumor , Child , Child, Preschool , Drug Resistance, Neoplasm/genetics , Female , Genes, p53 , Humans , Male , Mutation, Missense/genetics , Sarcoma, Ewing/drug therapy , Sarcoma, Ewing/secondary
14.
Cancer Biol Ther ; 6(11): 1700-3, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17986871

ABSTRACT

Renal cell carcinoma (RCC) occurring in renal allografts after cadaveric kidney transplantation has rarely been observed. RCC accounts for 2.3% of all malignancies in the general population, but up to 4.8% of malignancies in renal transplant recipients. Most have been reported in the patient's own diseased kidneys, whereas RCC in the renal allograft occur in only 10%. Here, we describe an organ-preserving surgical technique of a malignant renal tumor in a kidney allograft using a harmonic scalpel (Ultracision) for tumor enucleation. Furthermore we demonstrate by DNA microsatellite analysis the tumor's genetic origin as donor related. Collectively, we suggest that patients with a well defined low grade RCC in the kidney allograft and altogether low malignancy and good allograft function should only undergo an organ-preserving procedure and short-term postoperative screening.


Subject(s)
Carcinoma, Renal Cell/surgery , Kidney Neoplasms/surgery , Kidney Transplantation , Nephrons/surgery , Carcinoma, Renal Cell/genetics , Carcinoma, Renal Cell/pathology , Female , Humans , Kidney Neoplasms/genetics , Kidney Neoplasms/pathology , Magnetic Resonance Imaging , Microsatellite Repeats , Middle Aged , Transplantation, Homologous
15.
Clin Cancer Res ; 13(2 Pt 1): 488-97, 2007 Jan 15.
Article in English | MEDLINE | ID: mdl-17255270

ABSTRACT

PURPOSE: To characterize the prognostic and predictive impact of protein expression profiles in high-risk breast cancer patients who had previously been shown to benefit from high-dose chemotherapy (HDCT) in comparison to dose-dense chemotherapy (DDCT). EXPERIMENTAL DESIGN: The expression of 34 protein markers was evaluated using tissue microarrays containing paraffin-embedded breast cancer samples from 236 patients who were randomized to the West German Study Group AM01 trial. RESULTS: (a) 24 protein markers of the initial panel of 34 markers were sufficient to identify five profile clusters (subtypes) by K-means clustering: luminal-A (27%), luminal-B (12%), HER-2 (21%), basal-like (13%) cluster, and a so-called "multiple marker negative" (MMN) cluster (27%) characterized by the absence of specifying markers. (b) After DDCT, HER-2 and basal-like groups had significantly worse event-free survival [EFS; hazard ratio (HR), 3.6 [95% confidence interval (95% CI), 1.65-8.18; P = 0.001] and HR, 3.7 (95% CI, 1.68-8.48; P < 0.0001), respectively] when compared with both luminal groups. (c) After HDCT, the HR was 1.5 (95% CI, 0.76-3.05) for EFS in the HER-2 subgroup and 1.1 (95% CI, 0.37-3.32) in the basal-like subgroup, which indicates a better outcome for patients in the HER-2 and basal-like subgroups who received HDCT. The MMN cluster showed a trend to a better EFS after HDCT compared with DDCT. CONCLUSIONS: Protein expression profiling in high-risk breast cancers identified five subtypes, which differed with respect to survival and response to chemotherapy: In contrast to luminal-A and luminal-B subtypes, HER-2 and basal-like subgroups had a significant predictive benefit, and the MMN cluster had a trend to a predictive benefit, both from HDCT when compared with DDCT.


Subject(s)
Gene Expression Profiling/methods , Gene Expression Regulation, Neoplastic , Adult , Aged , Antineoplastic Agents/pharmacology , Biomarkers, Tumor , Cluster Analysis , Disease-Free Survival , Female , Germany , Humans , Middle Aged , Oligonucleotide Array Sequence Analysis , Prognosis , Risk
16.
Oncol Rep ; 17(2): 399-407, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17203180

ABSTRACT

Despite tremendous effort and progress in the diagnostics of pancreatic cancer with respect to imaging techniques and molecular genetics, only very few patients can be cured by surgery leading to a 5-year survival rate of only 3%. Especially the lack of chemotherapeutical options in this entity requires a better understanding of the molecular mechanisms leading to pancreatic carcinoma growth and progression in order to develop novel treatment regimens. To identify signaling pathways that are critical for this tumor entity, we compared six well-established pancreatic cancer cell lines (Capan-1, Capan-2, HUP-T3, HUP-T4, KCL-MOH, PaTu-8903) with colon cancer cell lines and tumor cell lines of non-epithelial origin by expression profiling. For this purpose we employed Human Genome Focus Arrays representing about 8500 well annotated human genes. We identified 353 genes with significantly high expression in the group of pancreatic carcinomas. Based on Gene Ontology annotations these genes are especially involved in Rho protein signal transduction, proteasome activator activity, cell motility, apoptotic program, and cell-cell adhesion processes indicating these pathways to be interesting candidates for the design of targeted therapies. Most pancreatic carcinomas are characterized by mutations in the TP53 and the KRAS genes and the absence of microsatellite instability, which could also be confirmed for our panel of pancreatic carcinoma cell lines. Looking for individual differences within this group that may be responsible for more or less aggressive behavior, we identified genomic amplifications at the 8q22.1 and the 8q24.22 loci to be associated with enhanced gene transcription. Because we have previously shown that gains of genomic material from the long arm of chromosome 8 have an adverse effect on the outcome of pancreatic carcinoma patients, we conclude that functional analysis of amplified genes at 8q22 and/or 8q24 may lead to an improved understanding of pancreatic carcinoma progression.


Subject(s)
Chromosomes, Human, Pair 8 , Gene Expression Regulation, Neoplastic , Genome, Human , Mutation , Pancreatic Neoplasms/metabolism , Apoptosis , Cell Line, Tumor , Chromosome Mapping , Disease Progression , Female , Genes, p53 , Genes, ras , Humans , Microsatellite Repeats , Oligonucleotide Array Sequence Analysis , Transcription, Genetic
17.
Neoplasia ; 8(7): 613-22, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16867224

ABSTRACT

Clear cell sarcoma of soft tissue (CCSST) represents a highly malignant tumor of the musculoskeletal system that is characterized by the chromosomal translocation t(12;22)(q13;q12) of the Ewing sarcoma gene (EWSR1) and activating transcription factor 1 (ATF1). In a former microarray expression study, we identified ERBB3, a member of the epidermal growth factor receptor (EGFR) family, as a promising new diagnostic marker in the differential diagnosis of CCSST. Here we show that, besides ErbB3, all CCSST cell lines (n = 8) also express the ErbB2 receptor or the ErbB4 receptor, representing an adequate coreceptor of ErbB3. The phosphorylation status of ErbB3 revealed these receptor pairs to be either constitutively activated in CCSST cells with high neuregulin-1 (NRG1) expression (n = 4) or activatable by exogenic NRG1 in cells showing low amounts of NRG1 mRNA (n = 4). Exogenous NRG1 stimulated the growth of a subset of CCSST cells but did not affect the kinetics of another subset. This difference was not strictly dependent on endogenous NRG1 expression; however, the growth-inhibiting effect of the pan-ErbB tyrosine kinase inhibitor CI-1033 or PD158780 clearly correlated with NRG1 expression indicating an autocrine growth stimulation loop which may constitute an interesting target of new therapeutic strategies in this tumor entity.


Subject(s)
Gene Expression Regulation, Neoplastic , Neuregulin-1/metabolism , Receptor, ErbB-3/metabolism , Sarcoma, Clear Cell/metabolism , Signal Transduction , Soft Tissue Neoplasms/metabolism , Alleles , Cell Line, Tumor , Humans , Kinetics , Nucleic Acid Hybridization , Oligonucleotide Array Sequence Analysis , Oligonucleotides/chemistry , Phosphorylation , Pyrimidines/pharmacology
18.
Diagn Mol Pathol ; 15(2): 83-9, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16778588

ABSTRACT

The t(11;22) translocation is a diagnostic hallmark of various small round-cell tumors. This study correlates the performance of fluorescence in situ hybridization (FISH) and reverse transcription polymerase chain reaction (RT-PCR) in the detection of this translocation analyzing paraffin-embedded tissue specimens. As negative control samples, 10 cases of normal colon mucosa and 10 cases of colon carcinoma tissue were analyzed by FISH to determine a valid cutoff value for the diagnosis of a t(11;22) translocation. The mean number of false-positive nuclei differed significantly between disomic and polysomic control group cases (P=0.002). Therefore, the cutoff value was determined considering the pitfall polysomy. The analysis group consisted of 20 cases from the University of Düsseldorf and 10 cases from the University of Bonn. These cases were analyzed using PCR (Düsseldorf) and FISH (Bonn) using a single-blinded approach. Twenty-two cases (73.3%) were concordant in both methods. Five cases (16.7%) were discrepant, showing a positive result in FISH whereas PCR was negative. Three cases (10.0%) were analyzed by FISH, and PCR failed for nonoptimized tissue preparation. In conclusion, the detection of t(11;22) translocation is critically dependent on a thoroughly defined cutoff value for FISH and on appropriate tissue preparation for both methods. We recommend FISH as a sensitive screening tool in the detection of t(11;22) followed by subsequent PCR amplification of the specific chimeric transcript.


Subject(s)
Chromosomes, Human, Pair 11/genetics , Chromosomes, Human, Pair 22/genetics , In Situ Hybridization, Fluorescence/methods , Reverse Transcriptase Polymerase Chain Reaction/methods , Translocation, Genetic , Adult , Aged , Base Sequence , Carcinoma, Small Cell/diagnosis , Carcinoma, Small Cell/genetics , Case-Control Studies , Colonic Neoplasms/diagnosis , Colonic Neoplasms/genetics , DNA Primers/genetics , Humans , In Situ Hybridization, Fluorescence/statistics & numerical data , Predictive Value of Tests , Reverse Transcriptase Polymerase Chain Reaction/statistics & numerical data , Sarcoma, Small Cell/diagnosis , Sarcoma, Small Cell/genetics , Single-Blind Method
19.
Hum Pathol ; 37(2): 205-11, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16426921

ABSTRACT

The proto-oncogene c-KIT (CD117) is highly expressed in normal breast epithelium and is decreased in invasive breast cancer. In this study, we analyzed the protein expression and the mutational status of c-KIT in ductal carcinoma in situ (DCIS) of the breast and correlated these findings with nuclear grade, architectural pattern, and expression of HER-2, estrogen receptor (ER)-alpha, and progesterone receptor (PR). C-KIT, HER-2, ER, and PR expression were analyzed immunohistochemically in 106 cases of paraffin-embedded DCIS (85 pure DCIS and 21 DCIS with concurrent carcinoma). Direct sequencing of exons 9 and 11 of the c-KIT gene was performed to analyze the hot spot mutational regions in representative cases. C-KIT expression was found in 55 (52.8%) of all DCIS, correlating with high nuclear grade (P < .0001), comedonecrosis (P < .0001), and solid growth pattern (P = .001). Furthermore, c-KIT expression was strongly associated with HER-2 positivity (P < .0001) and was significantly lower in ER- or PR-positive cases (P = .001 and P = .006, respectively). C-KIT expression alone or co-expression with HER-2 in pure DCIS did not differ significantly from DCIS with invasive component (P = .09). Mutational analysis in 6 c-KIT-positive DCIS revealed no activating mutations in exons 9 or 11. Our findings suggest that the expression of c-KIT protein might define a subset of poorly differentiated, HER-2-positive DCIS with decreased expression of steroid hormone receptors, comedonecrosis, and a solid growth pattern. The implications of c-KIT and HER-2 co-expression for breast carcinogenesis should be further evaluated.


Subject(s)
Breast Neoplasms/metabolism , Carcinoma, Intraductal, Noninfiltrating/metabolism , Proto-Oncogene Proteins c-kit/biosynthesis , Receptor, ErbB-2/biosynthesis , Adolescent , Adult , Aged , Aged, 80 and over , Carcinoma, Intraductal, Noninfiltrating/classification , Female , Humans , In Situ Hybridization, Fluorescence , Middle Aged , Neoplasm Invasiveness , Proto-Oncogene Mas , Receptors, Estrogen/analysis , Receptors, Progesterone/analysis
20.
Oncol Rep ; 14(3): 751-8, 2005 Sep.
Article in English | MEDLINE | ID: mdl-16077987

ABSTRACT

Since most tumours escape replicative senescence by re-activation of the enzyme telomerase, telomerase is a promising target in the treatment of cancer and a promising marker for diagnosis and therapeutic response. We evaluated the effects of doxorubicin, one of the most active drugs in the treatment of Ewing's sarcoma, on telomerase in the human Ewing's sarcoma cell line STA-ET-1 in vitro and in STA-ET-1 xenografts in vivo. Telomerase activity (TA) was examined by TRAP-assay and real-time PCR. Real-time PCR was also used to quantify the mRNA expression of the catalytic subunit of telomerase (hTERT). In vitro growth inhibition was determined by the MTT-assay. Tumour xenografts were analyzed for tumour volume, apoptosis, necrosis, and proliferation. Doxorubicin concentrations that inhibited in vitro growth of STA-ET-1 by 50% compared to untreated controls ranged between 0.14 microM after 24 h and 0.01 microM after 72 h. Compared to untreated controls doxorubicin reduced TA in STA-ET-1 at toxic concentrations, but increased TA at non-toxic concentrations. In comparison with untreated xenografts, TA was reduced to 65% and hTERT expression dropped to 25% within 72 h in xenografts treated with 17.5 mg/kg doxorubicin i.p.; both recovered to initial values after 264 h. The rate of proliferating cells dropped to 70% within 96 h and increased thereafter. The highest rates of necrosis and apoptosis were seen after 96 h. hTERT expression co-varied significantly with proliferation but not with TA, apoptosis, and necrosis. No correlation was observed between TA, proliferation, apoptosis and necrosis. The results suggest doxorubicin induces down-regulation of hTERT gene expression that at least in part modulates TA in these tumours.


Subject(s)
Antibiotics, Antineoplastic/pharmacology , Doxorubicin/pharmacology , Sarcoma, Ewing/drug therapy , Telomerase/genetics , Animals , Antibiotics, Antineoplastic/therapeutic use , Apoptosis/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Dose-Response Relationship, Drug , Doxorubicin/therapeutic use , Gene Expression Regulation, Enzymologic/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Humans , Mice , Mice, Nude , RNA, Messenger/genetics , RNA, Messenger/metabolism , Sarcoma, Ewing/enzymology , Sarcoma, Ewing/pathology , Telomerase/metabolism , Time Factors , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL
...