101
|
FitzGerald LM, Karlins E, Karyadi DM, Kwon EM, Koopmeiners JS, Stanford JL, Ostrander EA. Association of FGFR4 genetic polymorphisms with prostate cancer risk and prognosis. Prostate Cancer Prostatic Dis 2008; 12:192-7. [PMID: 18762813 DOI: 10.1038/pcan.2008.46] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
The fibroblast growth factor receptor 4 (FGFR4) is thought to be involved in many critical cellular processes and has been associated with prostate cancer risk. Four single nucleotide polymorphisms (SNPs) within or near FGFR4 were analyzed in a population-based study of 1458 prostate cancer patients and 1352 age-matched controls. We found no evidence to suggest that any of the FGFR4 SNP genotypes were associated with prostate cancer risk or with disease aggressiveness, Gleason score or stage. A weak association was seen between rs351855 and prostate cancer-specific mortality. Subset analysis of cases that had undergone radical prostatectomy revealed an association between rs351855 and prostate cancer risk. Although our results confirm an association between FGFR4 and prostate cancer risk in radical prostatectomy cases, they suggest that the role of FGFR4 in disease risk and outcomes at a population-based level appears to be minor.
Collapse
Affiliation(s)
- L M FitzGerald
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | | | | | | | | | | | | |
Collapse
|
102
|
Abstract
Tumour expression of the urokinase plasminogen activator correlates with invasive capacity. Consequently, inhibition of this serine protease by physiological inhibitors should decrease invasion and metastasis. However, of the two main urokinase inhibitors, high tumour levels of the type 1 inhibitor actually promote tumour progression, whereas high levels of the type 2 inhibitor decrease tumour growth and metastasis. We propose that the basis of this apparently paradoxical action of two similar serine protease inhibitors lies in key structural differences controlling interactions with components of the extracellular matrix and endocytosis-signalling co-receptors.
Collapse
Affiliation(s)
- David R Croucher
- Cancer Research Program, Garvan Institute of Medical Research, 384 Victoria Street, Darlinghurst, New South Wales, Australia 2010
| | | | | | | |
Collapse
|
103
|
Ansell A, Farnebo L, Grénman R, Roberg K, Thunell LK. Polymorphism of FGFR4 in cancer development and sensitivity to cisplatin and radiation in head and neck cancer. Oral Oncol 2008; 45:23-9. [PMID: 18487077 DOI: 10.1016/j.oraloncology.2008.03.007] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2008] [Revised: 03/12/2008] [Accepted: 03/13/2008] [Indexed: 02/07/2023]
Abstract
The aim of this study was to investigate the predisposition of the FGFR4 Gly/Arg polymorphism for development of head and neck squamous cell carcinoma (HNSCC) and, furthermore, to examine if the FGFR4 Arg(388) allele can be associated with resistance to chemo- and radiotherapy. When analysing 110 tumour biopsies a significant 1.7-fold increased risk to develop HNSCC in individuals carrying the Gly(388) allele (p=0.026) was found. Moreover a 2-fold increased risk for males harbouring the Gly(388) allele (p=0.031) to develop HNSCC was detected. In 39 HNSCC cell lines the role of the Arg(388) allele for radiation and cisplatin sensitivity was investigated. Our results show no role of the Arg(388) allele for the radiosensitivity (p=0.996) but indicate a tendency to increased cisplatin sensitivity (p=0.141). When screening the transmembrane and kinase domains in the FGFR4 gene a novel mutation, probably generating a truncated protein lacking exons 14-18, was found in six of eight selected cell lines. Taken together, we have here identified a marker that predicts the risk to develop HNSCC and possibly the sensitivity to cisplatin as well as a novel mutation in the FGFR4 gene.
Collapse
Affiliation(s)
- Anna Ansell
- Division of Otorhinolaryngology, University Hospital, SE-58185 Linköping, Sweden
| | | | | | | | | |
Collapse
|
104
|
Cotton LM, O'Bryan MK, Hinton BT. Cellular signaling by fibroblast growth factors (FGFs) and their receptors (FGFRs) in male reproduction. Endocr Rev 2008; 29:193-216. [PMID: 18216218 PMCID: PMC2528845 DOI: 10.1210/er.2007-0028] [Citation(s) in RCA: 84] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2007] [Accepted: 11/29/2007] [Indexed: 12/25/2022]
Abstract
The major function of the reproductive system is to ensure the survival of the species by passing on hereditary traits from one generation to the next. This is accomplished through the production of gametes and the generation of hormones that function in the maturation and regulation of the reproductive system. It is well established that normal development and function of the male reproductive system is mediated by endocrine and paracrine signaling pathways. Fibroblast growth factors (FGFs), their receptors (FGFRs), and signaling cascades have been implicated in a diverse range of cellular processes including: proliferation, apoptosis, cell survival, chemotaxis, cell adhesion, motility, and differentiation. The maintenance and regulation of correct FGF signaling is evident from human and mouse genetic studies which demonstrate that mutations leading to disruption of FGF signaling cause a variety of developmental disorders including dominant skeletal diseases, infertility, and cancer. Over the course of this review, we will provide evidence for differential expression of FGFs/FGFRs in the testis, male germ cells, the epididymis, the seminal vesicle, and the prostate. We will show that this signaling cascade has an important role in sperm development and maturation. Furthermore, we will demonstrate that FGF/FGFR signaling is essential for normal epididymal function and prostate development. To this end, we will provide evidence for the involvement of the FGF signaling system in the regulation and maintenance of the male reproductive system.
Collapse
Affiliation(s)
- Leanne M Cotton
- Department of Cell Biology, University of Virginia Health Sciences Center, Charlottesville, VA 22908, USA
| | | | | |
Collapse
|
105
|
Knowles MA. Novel therapeutic targets in bladder cancer: mutation and expression of FGF receptors. Future Oncol 2008; 4:71-83. [PMID: 18241002 DOI: 10.2217/14796694.4.1.71] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Bladder cancer presents several challenges in clinical management. For the large group of noninvasive tumors, key problems are the development of multiple recurrences that require long-term surveillance and a lack of effective therapies to prevent recurrence. For the smaller group of poor prognosis patients with invasive disease, novel therapies are urgently needed. The identification of mutations of FGF receptor 3 (FGFR3) in most noninvasive bladder tumors and the recent finding of overexpression of this receptor not only in superficial tumors but also in many invasive bladder cancers has generated optimism that therapies targeting this receptor tyrosine kinase may have major application in the treatment of urothelial cancers. There is little information on the other members of this receptor family apart from FGFR2, which is implicated as a tumor suppressor. Recent preclinical evaluations of FGFR3 as a therapeutic target have provided a strong impetus for the development of targeted agents for clinical use.
Collapse
Affiliation(s)
- Margaret A Knowles
- Cancer Research UK Clinical Centre, Section of Oncology, Leeds Institute of Molecular Medicine, St James's University Hospital, Beckett Street, Leeds, LS9 7TF, UK.
| |
Collapse
|
106
|
Sahadevan K, Darby S, Leung HY, Mathers ME, Robson CN, Gnanapragasam VJ. Selective over-expression of fibroblast growth factor receptors 1 and 4 in clinical prostate cancer. J Pathol 2007; 213:82-90. [PMID: 17607666 DOI: 10.1002/path.2205] [Citation(s) in RCA: 89] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Fibroblast growth factor receptors (FGFRs) mediate the tumourigenic effects of FGFs in prostate cancer. These receptors are therefore potential therapeutic targets in the development of inhibitors to this pathway. To identify the most relevant targets, we simultaneously investigated FGFR1-4 expression using a prostate cancer tissue microarray (TMA) and in laser capture microdissected (LCM) prostate epithelial cells. In malignant prostates (n = 138) we observed significant FGFR1 and FGFR4 protein over-expression in comparison with benign prostates (n = 58; p < 0.0001). FGFR1 was expressed at high levels in the majority of tumours (69% of grade 3 or less, 74% of grade 4 and 70% of grade 5), while FGFR4 was strongly expressed in 83% of grade 5 cancers but in only 25% of grade 1-3 cancers (p < 0.0001). At the transcript level we observed a similar pattern, with FGFR1 and FGFR4 mRNA over-expressed in malignant epithelial cells compared to benign cells (p < 0.0005 and p < 0.05, respectively). While total FGFR2 was increased in some cancers, there was no association between expression and tumour grade or stage. Transcript analysis, however, revealed a switch in the predominant isoform expressed from FGFR2IIIb to FGFR2IIIc among malignant epithelial cells. In contrast, protein and transcript expression of FGFR3 was very similar between benign and cancer biopsies. The functional effect of targeting FGFR4 in prostate cancer cells has not previously been investigated. In in vitro experiments, suppression of FGFR4 by RNA interference effectively blocked prostate cancer cell proliferation (p < 0.0001) and invasion (p < 0.001) in response to exogenous stimulation. This effect was evident regardless of whether the cells expressed the FGFR4 Arg388 or Gly388 allele. In parallel experiments, FGFR3 suppression had no discernible effect on cancer cell behaviour. These results suggest evidence of selective over-expression of FGFR1 and FGFR4 in clinical prostate cancer and support the notion of targeted inhibition of these receptors to disrupt FGF signalling.
Collapse
MESH Headings
- Case-Control Studies
- Cell Line, Tumor
- Cell Proliferation
- Gene Expression Profiling
- Gene Expression Regulation, Neoplastic
- Humans
- Immunohistochemistry
- Male
- Microdissection
- Microscopy, Confocal
- Oligonucleotide Array Sequence Analysis
- Polymorphism, Single Nucleotide
- Prostatic Neoplasms/genetics
- Prostatic Neoplasms/metabolism
- Protein Isoforms/genetics
- RNA Interference
- RNA, Small Interfering/pharmacology
- Receptor, Fibroblast Growth Factor, Type 1/analysis
- Receptor, Fibroblast Growth Factor, Type 1/genetics
- Receptor, Fibroblast Growth Factor, Type 1/metabolism
- Receptor, Fibroblast Growth Factor, Type 2/analysis
- Receptor, Fibroblast Growth Factor, Type 2/genetics
- Receptor, Fibroblast Growth Factor, Type 2/metabolism
- Receptor, Fibroblast Growth Factor, Type 3/analysis
- Receptor, Fibroblast Growth Factor, Type 3/genetics
- Receptor, Fibroblast Growth Factor, Type 3/metabolism
- Receptor, Fibroblast Growth Factor, Type 4/analysis
- Receptor, Fibroblast Growth Factor, Type 4/genetics
- Receptor, Fibroblast Growth Factor, Type 4/metabolism
- Receptors, Fibroblast Growth Factor/analysis
- Receptors, Fibroblast Growth Factor/genetics
- Receptors, Fibroblast Growth Factor/metabolism
- Transcription, Genetic
Collapse
Affiliation(s)
- K Sahadevan
- Urology Research Group, Northern Institute for Cancer Research, Medical School, Newcastle University, Framlington Place, Newcastle upon Tyne NE2 4HH, UK
| | | | | | | | | | | |
Collapse
|
107
|
Entz-Werle N, Lavaux T, Metzger N, Stoetzel C, Lasthaus C, Marec P, Kalifa C, Brugieres L, Pacquement H, Schmitt C, Tabone MD, Gentet JC, Lutz P, Babin A, Oudet P, Gaub MP, Perrin-Schmitt F. Involvement of MET/TWIST/APC combination or the potential role of ossification factors in pediatric high-grade osteosarcoma oncogenesis. Neoplasia 2007; 9:678-88. [PMID: 17786187 PMCID: PMC1950438 DOI: 10.1593/neo.07367] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2007] [Revised: 06/29/2007] [Accepted: 07/03/2007] [Indexed: 11/18/2022] Open
Abstract
Dysregulated cell growth or differentiation due to misexpression of developmental critical factors seems to be a decisive event in oncogenesis. As osteosarcomas are histologically defined by malignant osteoblasts producing an osteoid component, we prospected in pediatric osteosarcomas treated with OS94 protocol the genomic status of several genes implied in ossification processes. In 91 osteosarcoma cases, we focused on the analysis of the fibroblast growth factor receptors (FGFRs) TWIST, APC, and MET by allelotyping, real-time quantitative polymerase chain reaction, gene sequencing, and protein polymorphism study. Our study supports the frequent role of TWIST, APC, and MET as osteosarcoma markers (50%, 62%, and 50%, respectively). TWIST and MET were mainly found to be deleted, and no additional APC mutation was identified. Surprisingly, FGFRs are abnormal in only < 30%. Most of these factors and their abnormalities seem to be linked more or less to one clinical subgroup, but the most significant correlation is the link of MET, TWIST, and APC abnormalities to a worse outcome and their combination within abnormal tumors. A wider cohort is mandatory to define more robust molecular conclusions, but these results are to be considered as the beginning of a more accurate basis for diagnosis, in search of targeted therapies, and to further characterize prognostic markers.
Collapse
Affiliation(s)
- Natacha Entz-Werle
- Institut National de la Santé et de la Recherche Médicale U682, Strasbourg, France.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
108
|
Desnoyers LR, Pai R, Ferrando RE, Hötzel K, Le T, Ross J, Carano R, D'Souza A, Qing J, Mohtashemi I, Ashkenazi A, French DM. Targeting FGF19 inhibits tumor growth in colon cancer xenograft and FGF19 transgenic hepatocellular carcinoma models. Oncogene 2007; 27:85-97. [PMID: 17599042 DOI: 10.1038/sj.onc.1210623] [Citation(s) in RCA: 205] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Although fibroblast growth factor 19 (FGF19) can promote liver carcinogenesis in mice its involvement in human cancer is not well characterized. Here we report that FGF19 and its cognate receptor FGF receptor 4 (FGFR4) are coexpressed in primary human liver, lung and colon tumors and in a subset of human colon cancer cell lines. To test the importance of FGF19 for tumor growth, we developed an anti-FGF19 monoclonal antibody that selectively blocks the interaction of FGF19 with FGFR4. This antibody abolished FGF19-mediated activity in vitro and inhibited growth of colon tumor xenografts in vivo and effectively prevented hepatocellular carcinomas in FGF19 transgenic mice. The efficacy of the antibody in these models was linked to inhibition of FGF19-dependent activation of FGFR4, FRS2, ERK and beta-catenin. These findings suggest that the inactivation of FGF19 could be beneficial for the treatment of colon cancer, liver cancer and other malignancies involving interaction of FGF19 and FGFR4.
Collapse
MESH Headings
- Animals
- Antibodies, Blocking/therapeutic use
- Antibodies, Monoclonal/therapeutic use
- Antineoplastic Agents/pharmacology
- Carcinoma, Hepatocellular/drug therapy
- Carcinoma, Hepatocellular/genetics
- Carcinoma, Hepatocellular/immunology
- Carcinoma, Squamous Cell/drug therapy
- Carcinoma, Squamous Cell/immunology
- Carcinoma, Squamous Cell/metabolism
- Colonic Neoplasms/drug therapy
- Colonic Neoplasms/genetics
- Colonic Neoplasms/immunology
- Fibroblast Growth Factors/antagonists & inhibitors
- Fibroblast Growth Factors/biosynthesis
- Fibroblast Growth Factors/genetics
- Fibroblast Growth Factors/immunology
- Gene Targeting/methods
- HCT116 Cells
- HT29 Cells
- Humans
- Liver Neoplasms, Experimental/drug therapy
- Liver Neoplasms, Experimental/immunology
- Lung Neoplasms/drug therapy
- Lung Neoplasms/genetics
- Lung Neoplasms/immunology
- Mice
- Mice, Inbred BALB C
- Mice, Nude
- Mice, Transgenic
- Neoplasm Transplantation
- Receptor, Fibroblast Growth Factor, Type 4/biosynthesis
- Receptor, Fibroblast Growth Factor, Type 4/genetics
- Receptor, Fibroblast Growth Factor, Type 4/metabolism
- Transplantation, Heterologous
- Xenograft Model Antitumor Assays/methods
Collapse
Affiliation(s)
- L R Desnoyers
- 1Department of Molecular Oncology, Genentech Inc., South San Francisco, CA, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
109
|
Matakidou A, el Galta R, Rudd MF, Webb EL, Bridle H, Eisen T, Houlston RS. Further observations on the relationship between the FGFR4 Gly388Arg polymorphism and lung cancer prognosis. Br J Cancer 2007; 96:1904-7. [PMID: 17519899 PMCID: PMC2359960 DOI: 10.1038/sj.bjc.6603816] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
The Gly388Arg polymorphism in the fibroblast growth factor receptor 4 (FGFR4) gene has been reported to influence prognosis in a wide variety of cancer types. To determine whether Gly388Arg is a marker for lung cancer prognosis, we genotyped 619 lung cancer patients with incident disease and examined the relationship between genotype and overall survival. While we employed a comprehensive set of statistical tests, including those sensitive to the detection of differences in early survival, our data provide little evidence to support the tenet that the FGFR4 Gly388Arg polymorphism is a clinically useful marker for lung cancer prognosis.
Collapse
MESH Headings
- Amino Acid Substitution
- Arginine
- Carcinoma, Non-Small-Cell Lung/genetics
- Carcinoma, Non-Small-Cell Lung/mortality
- Carcinoma, Non-Small-Cell Lung/pathology
- Carcinoma, Non-Small-Cell Lung/therapy
- Carcinoma, Small Cell/genetics
- Carcinoma, Small Cell/pathology
- Carcinoma, Small Cell/therapy
- Combined Modality Therapy
- Glycine
- Humans
- Lung Neoplasms/genetics
- Lung Neoplasms/mortality
- Lung Neoplasms/pathology
- Lung Neoplasms/therapy
- Middle Aged
- Neoplasm Staging/mortality
- Polymorphism, Single Nucleotide
- Prognosis
- Receptor, Fibroblast Growth Factor, Type 4/genetics
- Survival Analysis
Collapse
Affiliation(s)
- A Matakidou
- Section of Cancer Genetics, Institute of Cancer Research, Surrey, UK
| | - R el Galta
- Section of Cancer Genetics, Institute of Cancer Research, Surrey, UK
| | - M F Rudd
- Section of Cancer Genetics, Institute of Cancer Research, Surrey, UK
| | - E L Webb
- Section of Cancer Genetics, Institute of Cancer Research, Surrey, UK
| | - H Bridle
- Department of Oncology, University of Cambridge, Cambridge, UK
| | - T Eisen
- Department of Oncology, University of Cambridge, Cambridge, UK
| | - R S Houlston
- Section of Cancer Genetics, Institute of Cancer Research, Surrey, UK
- Section of Cancer Genetics, Brookes Lawley Building, Institute of Cancer Research, Surrey SM2 5NG, UK. E-mail:
| | | |
Collapse
|
110
|
Pal NR, Aguan K, Sharma A, Amari SI. Discovering biomarkers from gene expression data for predicting cancer subgroups using neural networks and relational fuzzy clustering. BMC Bioinformatics 2007; 8:5. [PMID: 17207284 PMCID: PMC1770936 DOI: 10.1186/1471-2105-8-5] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2006] [Accepted: 01/06/2007] [Indexed: 11/10/2022] Open
Abstract
Background The four heterogeneous childhood cancers, neuroblastoma, non-Hodgkin lymphoma, rhabdomyosarcoma, and Ewing sarcoma present a similar histology of small round blue cell tumor (SRBCT) and thus often leads to misdiagnosis. Identification of biomarkers for distinguishing these cancers is a well studied problem. Existing methods typically evaluate each gene separately and do not take into account the nonlinear interaction between genes and the tools that are used to design the diagnostic prediction system. Consequently, more genes are usually identified as necessary for prediction. We propose a general scheme for finding a small set of biomarkers to design a diagnostic system for accurate classification of the cancer subgroups. We use multilayer networks with online gene selection ability and relational fuzzy clustering to identify a small set of biomarkers for accurate classification of the training and blind test cases of a well studied data set. Results Our method discerned just seven biomarkers that precisely categorized the four subgroups of cancer both in training and blind samples. For the same problem, others suggested 19–94 genes. These seven biomarkers include three novel genes (NAB2, LSP1 and EHD1 – not identified by others) with distinct class-specific signatures and important role in cancer biology, including cellular proliferation, transendothelial migration and trafficking of MHC class antigens. Interestingly, NAB2 is downregulated in other tumors including Non-Hodgkin lymphoma and Neuroblastoma but we observed moderate to high upregulation in a few cases of Ewing sarcoma and Rabhdomyosarcoma, suggesting that NAB2 might be mutated in these tumors. These genes can discover the subgroups correctly with unsupervised learning, can differentiate non-SRBCT samples and they perform equally well with other machine learning tools including support vector machines. These biomarkers lead to four simple human interpretable rules for the diagnostic task. Conclusion Although the proposed method is tested on a SRBCT data set, it is quite general and can be applied to other cancer data sets. Our scheme takes into account the interaction between genes as well as that between genes and the tool and thus is able find a very small set and can discover novel genes. Our findings suggest the possibility of developing specialized microarray chips or use of real-time qPCR assays or antibody based methods such as ELISA and western blot analysis for an easy and low cost diagnosis of the subgroups.
Collapse
Affiliation(s)
- Nikhil R Pal
- Electronics and Communication Sciences Unit, Indian Statistical Institute, 203, B. T. Road, Calcutta – 700108, India
| | - Kripamoy Aguan
- Neurogenetics Laboratory, RIKEN Brain Science Institute, Saitama, Japan
| | - Animesh Sharma
- Department of Computer Science and Electrical Engineering, University of Missouri, Kansas city, USA
| | - Shun-ichi Amari
- Lab for Mathematical Neuroscience, RIKEN Brain Science Institute, Saitama, Japan
| |
Collapse
|
111
|
Yang YC, Lu ML, Rao JY, Wallerand H, Cai L, Cao W, Pantuck A, Dalbagni G, Reuter V, Figlin RA, Belldegrun A, Cordon-Cardo C, Zhang ZF. Joint association of polymorphism of the FGFR4 gene and mutation TP53 gene with bladder cancer prognosis. Br J Cancer 2006; 95:1455-8. [PMID: 17088904 PMCID: PMC2360734 DOI: 10.1038/sj.bjc.6603456] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The impact of the fibroblast growth factor receptor 4 (FGFR4) Gly388Arg polymorphism on bladder cancer is unknown. We found no clear correlations between the FGFR4 genotype and risk of bladder cancer or pathological parameters. Neither the polymorphism nor TP53 mutation status was an independent predictor of prognosis, but they might act jointly on the disease-specific survival of patients.
Collapse
Affiliation(s)
- Y C Yang
- Department of Epidemiology, UCLA School of Public Health and Jonsson Comprehensive Cancer Center, Los Angeles, CA 90095, USA
| | - M L Lu
- Department of Pathology, Memorial Sloan-Kettering Cancer Center, New York, NY 10021, USA
| | - J Y Rao
- Department of Epidemiology, UCLA School of Public Health and Jonsson Comprehensive Cancer Center, Los Angeles, CA 90095, USA
- Department of Pathology and Laboratory Medicine, UCLA School of Medicine, Los Angeles, CA 90095, USA
| | - H Wallerand
- EMI INSERM 03-37 and Service d'Urologie, Université Paris XII, AP-HP, Hôpital Henri Mondor, 94000 Créteil, France
| | - L Cai
- Department of Epidemiology, Fujian Medical University, Fuzhou, Fujian, PR China
| | - W Cao
- Department of Epidemiology, UCLA School of Public Health and Jonsson Comprehensive Cancer Center, Los Angeles, CA 90095, USA
| | - A Pantuck
- Department of Urology, UCLA School of Medicine, Los Angeles, CA 90095, USA
| | - G Dalbagni
- Department of Surgery, Memorial Sloan-Kettering Cancer Center, New York, NY 10021, USA
| | - V Reuter
- Department of Pathology, Memorial Sloan-Kettering Cancer Center, New York, NY 10021, USA
| | - R A Figlin
- Department of Medicine, Hematology-Oncology, UCLA School of Medicine, Los Angeles, CA 90095, USA
| | - A Belldegrun
- Department of Urology, UCLA School of Medicine, Los Angeles, CA 90095, USA
| | - C Cordon-Cardo
- Department of Pathology, Memorial Sloan-Kettering Cancer Center, New York, NY 10021, USA
| | - Z F Zhang
- Department of Epidemiology, UCLA School of Public Health and Jonsson Comprehensive Cancer Center, Los Angeles, CA 90095, USA
- E-mail:
| |
Collapse
|
112
|
Wang J, Cai Y, Penland R, Chauhan S, Miesfeld RL, Ittmann M. Increased expression of the metastasis-associated gene Ehm2 in prostate cancer. Prostate 2006; 66:1641-52. [PMID: 16927306 DOI: 10.1002/pros.20474] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
BACKGROUND Alterations of fibroblast growth factors and their receptors contribute to prostate cancer progression by enhancing cell survival, motility, and proliferation. The expression of the FGFR-4 Arg(388) variant is correlated with the occurrence of pelvic lymph node metastasis and biochemical (PSA) recurrence in men undergoing radical prostatectomy. Ehm2 is an androgen-regulated gene that has been associated with metastasis in other systems, so we sought to determine if it is expressed in prostate cancer and if the FGFR-4 Arg(388) variant can increase its expression. METHODS Expression of Ehm2 was examined by quantitative RT-PCR and Western blotting in prostate cell lines and by quantitative RT-PCR, in situ hybridization, and immunohistochemistry in prostate tissues. The effect of Ehm2 expression on collagen IV adhesion was tested by transient overexpression and RNA interference. RESULTS Ehm2 expression is upregulated in prostate cancer cell lines and prostate cancer tissues. Expression of the FGFR-4 Arg(388) variant results in increased expression of Ehm2. Increased expression of Ehm2 leads to decreased adhesion to collagen IV, which has been associated with metastasis in cancers. Analysis of tissue microarrays revealed that increased Ehm2 expression is associated with biochemical recurrence after radical prostatectomy, which is indicative of more aggressive disease. CONCLUSIONS Ehm2 is overexpressed in prostate cancer and may enhance disease progression and metastasis.
Collapse
Affiliation(s)
- Jianghua Wang
- Department of Pathology of Baylor College of Medicine and Michael E. DeBakey Department of Veterans Affairs Medical Center, Houston, Texas 77030, USA
| | | | | | | | | | | |
Collapse
|
113
|
Jung V, Wullich B, Kamradt J, Stöckle M, Unteregger G. An improved in vitro model to characterize invasive growing cancer cells simultaneously by function and genetic aberrations. Toxicol In Vitro 2006; 21:183-90. [PMID: 17126525 DOI: 10.1016/j.tiv.2006.09.021] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2006] [Revised: 09/12/2006] [Accepted: 09/14/2006] [Indexed: 11/17/2022]
Abstract
Invasion into the surrounding tissue and bone metastasis is a common feature of advanced prostate cancer. Chromosomal and other genetic or epigenetic abnormalities were aligned to this behaviour mostly by using permanent cell lines, paraffin embedded tissue or primary tumour samples. Both attempts fail to reflect either the original situation or functional information in the patient's tissue. Thus, we developed an improved in vitro assay to follow invasion of prostate cancer cells derived from fresh samples of radical prostatectomy specimens. Fresh tumour samples were applied onto Matrigeltrade mark-coated invasion chambers using a cocultivation model. Invasive growing cells were harvested from the bottom of the membrane or from the underlying gel and further characterized using comparative genomic hybridization. Prostate cancer cells have the capability to invasively grow through the barrier of a Matrigeltrade mark and could easily be sampled in a pad of Matrigeltrade mark. Comparative genomic hybridization revealed characteristic chromosomal aberrations of the invasive growing cells. Noteworthy is their ability to spheroid formation, which allows for further cell propagation by standard cell culture methods. Thus, our improved invasion model is a tool for the sampling of invasive growing cancer cells from fresh human tumour material allowing for functional as well as genetic studies.
Collapse
Affiliation(s)
- V Jung
- Department of Urology and Pediatric Urology, University Clinic of the Saarland, Homburg/Saar, Germany
| | | | | | | | | |
Collapse
|
114
|
Thussbas C, Nahrig J, Streit S, Bange J, Kriner M, Kates R, Ulm K, Kiechle M, Hoefler H, Ullrich A, Harbeck N. FGFR4 Arg388 allele is associated with resistance to adjuvant therapy in primary breast cancer. J Clin Oncol 2006; 24:3747-55. [PMID: 16822847 DOI: 10.1200/jco.2005.04.8587] [Citation(s) in RCA: 105] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
PURPOSE A recent study presented first evidence that a single nucleotide polymorphism (SNP) at codon 388 of fibroblast growth factor receptor 4 (FGFR4) gene, causing a transmembrane domain missense mutation (Gly388Arg), is associated with disease outcome in node-positive breast cancer. This article addresses the clinical relevance of this SNP, FGFR4 genotype, phenotype, and HER2 regarding patient outcome and influence of adjuvant systemic therapy in a substantial primary breast cancer collective (n = 372; median follow-up, 94.5 months). METHODS Polymerase chain reaction restriction fragment length polymorphism analysis of germ-line polymorphism was performed in uninvolved lymph nodes; FGFR4 and HER2 expression were assessed immunohistochemically in tissue microarrays. RESULTS In 51% of patients, homo- or heterozygous Arg388 allele was present. No correlation existed between FGFR4 genotype and expression or HER2 status. In node-negative patients, FGFR4 genotype was not correlated with disease outcome. In node-positive patients, however, FGFR4 Arg388 was significantly associated with poor disease-free survival (DFS; P = .02) and overall survival (OS; P = .04). Notably, this association seems to be attributable to relatively poor therapy response in Arg388 carriers, reflected in their significantly shorter DFS (P = .02) and OS (P = .045) among patients receiving adjuvant systemic therapy. It is also seen as a significant interaction term in a multivariate proportional hazards model with Arg388 carriers having only about half as much benefit from adjuvant systemic therapy as wild-type carriers. CONCLUSION According to this study, FGFR4 Arg388 genotype is a marker for breast cancer progression in patients with adjuvant systemic therapy, particularly chemotherapy, and thus may indicate therapy resistance.
Collapse
Affiliation(s)
- Christoph Thussbas
- Department of Obstetrics and Gynecology, Institute of Pathology, and Institute for Medical Statistics and Epidemiology, Technical University of Munich, Munich, Germany
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
115
|
Streit S, Mestel DS, Schmidt M, Ullrich A, Berking C. FGFR4 Arg388 allele correlates with tumour thickness and FGFR4 protein expression with survival of melanoma patients. Br J Cancer 2006; 94:1879-86. [PMID: 16721364 PMCID: PMC2361343 DOI: 10.1038/sj.bjc.6603181] [Citation(s) in RCA: 60] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
A single nucleotide polymorphism in the gene for FGFR4 (−Arg388) has been associated with progression in various types of human cancer. Although fibroblast growth factors (FGFs) belong to the most important growth factors in melanoma, expression of FGF receptor subtype 4 has not been investigated yet. In this study, the protein expression of this receptor was analysed in 137 melanoma tissues of different progression stages by immunohistochemistry. FGFR4 protein was expressed in 45% of the specimens and correlated with pTNM tumour stages (UICC, P=0.023 and AJCC, P=0.046), presence of microulceration (P=0.009), tumour vascularity (P=0.001), metastases (P=0.025), number of primary tumours (P=0.022), overall survival (P=0.047) and disease-free survival (P=0.024). Furthermore, FGFR4 Arg388 polymorphism was analysed in 185 melanoma patients by polymerase chain reaction-restriction fragment length polymorphism (PCR-RFLP). The Arg388 allele was detected in 45% of the melanoma patients and was significantly associated with tumour thickness (by Clark's level of invasion (P=0.004) and by Breslow in mm (P=0.02)) and the tumour subtype nodular melanoma (P=0.002). However, there was no correlation of the FGFR4 Arg388 allele with overall and disease-free survival. In conclusion, the Arg388 genotype and the protein expression of FGFR4 may be potential markers for progression of melanoma.
Collapse
Affiliation(s)
- S Streit
- Department of Molecular Biology, Max-Planck-Institute of Biochemistry, Am Klopferspitz 18a, Martinsried D-82152, Germany
| | - D S Mestel
- Department of Dermatology, Ludwig-Maximilian University of Munich, Frauenlobstr. 9-11, Munich D-80337, Germany
| | - M Schmidt
- Munich Cancer Registry/IBE, Ludwig-Maximilian University of Munich, Marchioninistr. 15, Munich D-81377, Germany
| | - A Ullrich
- Department of Molecular Biology, Max-Planck-Institute of Biochemistry, Am Klopferspitz 18a, Martinsried D-82152, Germany
| | - C Berking
- Department of Dermatology, Ludwig-Maximilian University of Munich, Frauenlobstr. 9-11, Munich D-80337, Germany
- E-mail:
| |
Collapse
|
116
|
Culig Z. Endocrine regulation of prostate cancer growth. Expert Rev Endocrinol Metab 2006; 1:379-389. [PMID: 30764076 DOI: 10.1586/17446651.1.3.379] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Prostate cancer is one of the most frequently diagnosed malignant neoplasms in the Western world. The focus of this article is on regulation of proliferation, apoptosis, angiogenesis and differentiation by steroid and peptide hormones. Steroid hormones, in particular androgens and estrogens, exert their effects through respective receptors. The androgen receptor is functional in all stages of the disease and it can be activated in a ligand-dependent and -independent manner. In prostate cancer, expression of some androgen receptor coactivators increases during tumor progression. Interleukins and transforming growth factor-β are multifunctional regulators of prostate growth and proliferation. Signal transduction cascades of fibroblast growth factors and insulin-like growth factors are responsible for increased survival and angiogenesis in prostate cancer. In clinical specimens obtained from prostate cancer, there is an increased phosphorylation of mitogen-activated protein kinase and Akt kinase, whose action in the regulation of cell survival is redundant.
Collapse
Affiliation(s)
- Zoran Culig
- a Associate Professor, Innsbruck Medical University, Department of Urology, Anichstrasse 35, A-6020 Innsbruck, Austria.
| |
Collapse
|
117
|
Wang J, Thompson B, Ren C, Ittmann M, Kwabi-Addo B. Sprouty4, a suppressor of tumor cell motility, is down regulated by DNA methylation in human prostate cancer. Prostate 2006; 66:613-24. [PMID: 16388505 DOI: 10.1002/pros.20353] [Citation(s) in RCA: 68] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
PURPOSE Alterations of fibroblast growth factors (FGFs) and their receptors contribute to prostate cancer progression by enhancing cellular proliferation, survival, and motility. The Sprouty gene family negatively regulates FGF signaling and may limit the ability of FGFs to enhance tumor progression. Sprouty1 is down regulated in human prostate cancers and Sprouty1 expression can markedly inhibit prostate cancer proliferation in vitro. Sprouty4 has been shown to negatively regulate both proliferation and cell migration in other systems. We therefore examined whether Sprouty4 expression was altered in prostate cancer. EXPERIMENTAL DESIGN Expression of Sprouty4 was examined by in situ hybridization and quantitative RT-PCR. Methylation of the Sprouty4 gene promoter was assessed using bisulfite modification and sequencing. The effect of Sprouty4 expression on cell migration was determined using an in vitro wounding assay. RESULTS By in situ hybridization Sprouty4 is expressed in normal prostatic epithelial cells and is decreased in a subset of prostate cancers. Quantitative RT-PCR confirms that Sprouty4 expression is decreased in approximately one half of prostate cancers. Analysis of the 5'-regulatory region revealed a CpG island approximately 1 kb upstream of the transcription initiation site, the proximal portion of which was preferentially methylated in prostate cancer tissues. More than one half of all prostate cancer DNAs were methylated in this region and methylation was significantly correlated with decreased Sprouty4 expression as determined by quantitative RT-PCR. When overexpressed in prostate cancer cell lines, Sprouty4 did not inhibit cell proliferation but did inhibit cell migration. CONCLUSIONS Sprouty4 expression is down regulated in human prostate cancer by DNA methylation and this decreased expression may contribute to increased cell migration.
Collapse
Affiliation(s)
- Jianghua Wang
- Department of Pathology, Baylor College of Medicine and Michael E. DeBakey, Department of Veterans Affairs Medical Center, Houston, Texas 77030, USA
| | | | | | | | | |
Collapse
|
118
|
Kanda S, Miyata Y, Kanetake H. Current status and perspective of antiangiogenic therapy for cancer: urinary cancer. Int J Clin Oncol 2006; 11:90-107. [PMID: 16622744 DOI: 10.1007/s10147-006-0565-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2006] [Indexed: 12/27/2022]
Abstract
Angiogenesis is considered a prerequisite for solid tumor growth. Antiangiogenic therapy reduces tumor size and extends host survival in a number of preclinical animal models. However, in humans antiangiogenic therapy is a poor promoter of tumor regression and has shown minimal effect on patient survival. In urinary cancers, such as renal cell cancer, prostate cancer, and bladder cancer, advanced refractory disease is a good candidate for antiangiogenic therapy because of its resistance to ordinary chemotherapy, radiotherapy, and hormonal therapy. Unique characteristics of molecular mechanisms underlie the induction of angiogenesis in urinary cancers. In this review, we summarize these unique mechanisms and review the results of clinical trials of antiangiogenic therapy for these cancers, discussing prospects and problems relating to antiangiogenic therapy.
Collapse
Affiliation(s)
- Shigeru Kanda
- Department of Molecular Microbiology and Immunology, Division of Endothelial Cell Biology, Nagasaki University Graduate School of Biomedical Science, 1-7-1 Sakamoto, Nagasaki 852-8501, Japan.
| | | | | |
Collapse
|
119
|
Spinola M, Leoni V, Pignatiello C, Conti B, Ravagnani F, Pastorino U, Dragani TA. Functional FGFR4 Gly388Arg polymorphism predicts prognosis in lung adenocarcinoma patients. J Clin Oncol 2005; 23:7307-11. [PMID: 16061909 DOI: 10.1200/jco.2005.17.350] [Citation(s) in RCA: 84] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
PURPOSE Fibroblast growth factor receptor 4 (FGFR4) is a member of a family of transmembrane receptors with ligand-induced tyrosine kinase activity. The Gly388Arg polymorphism in the FGFR4 gene was reported to modulate cancer cell migration in vitro and to be associated with breast, colon, and prostate cancer prognostic parameters. The purpose of this study was to investigate the involvement of the FGFR4 polymorphism in lung tumorigenesis. PATIENTS AND METHODS A case-control study was performed including 274 patients with histologically confirmed lung adenocarcinoma and 401 healthy control subjects from general population. mRNA expression analysis was carried out in healthy lung of cancer patients. RESULTS Patients with the Arg/Arg or Gly/Arg genotype compared to those with a Gly/Gly genotype had an earlier age at cancer onset (median age, 60.2 v 63.4 years), higher proportion of poor clinical stage disease (hazard ratio [HR], 2.3; 95% CI, 1.4 to 3.9; P = .002), of nodal involvement (HR, 1.9; 95% CI, 1.1 to 3.2; P = .027), or of short-term survivors (HR, 1.6; 95% CI, 1.1 to 2.3; P = .008). In healthy lungs, FGFR4 did not show allele-specific expression and mRNA levels were not associated with genotype. CONCLUSION This study suggests that FGFR4 Gly388Arg polymorphism may predict prognosis in lung adenocarcinoma.
Collapse
Affiliation(s)
- Monica Spinola
- Department of Experimental Oncology, Istituto Nazionale Tumori, Via G. Venezian 1, 20133 Milan, Italy
| | | | | | | | | | | | | |
Collapse
|