1
|
Chen X, Huang Y, Chen B, Liu H, Cai Y, Yang Y. Insight into the design of FGFR4 selective inhibitors in cancer therapy: Prospects and challenges. Eur J Med Chem 2024; 263:115947. [PMID: 37976704 DOI: 10.1016/j.ejmech.2023.115947] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Revised: 11/07/2023] [Accepted: 11/07/2023] [Indexed: 11/19/2023]
Abstract
Recently, FGFR4 has become a hot target for the treatment of cancer owing to its important role in cellular physiological processes. FGFR4 has been validated to be closely related to the occurrence of cancers, such as hepatocellular carcinoma, rhabdomyosarcoma, breast cancer and colorectal cancer. Hence, the development of FGFR4 small-molecule inhibitors is essential to further understanding the functions of FGFR4 in cancer and the treatment of FGFR4-dependent diseases. Given the particular structures of FGFR1-4, the development of FGFR4 selective inhibitors presents significant challenges. The non-conserved Cys552 in the hinge region of the FGFR4 complex becomes the key to the selectivity of FGFR4 and FGFR1/2/3 inhibitors. In this review, we systematically introduce the close relationship between FGFR4 and cancer, and conduct an in-depth analysis of the developing methodology, binding mechanism, kinase selectivity, pharmacokinetic characteristics of FGFR4 selectivity inhibitors, and their application in clinical research.
Collapse
Affiliation(s)
- Xiaolu Chen
- Department of Pharmacy, Jingzhou Hospital Affiliated to Yangtze University, Jingzhou, Hubei, 434020, China
| | - Yajiao Huang
- Department of Pharmacy, Jingzhou Hospital Affiliated to Yangtze University, Jingzhou, Hubei, 434020, China
| | - Ban Chen
- School of Biological Engineering and Food, Hubei University of Technology, Wuhan, 430068, China
| | - Huihui Liu
- Department of Pharmacy, Jingzhou Hospital Affiliated to Yangtze University, Jingzhou, Hubei, 434020, China
| | - Yuepiao Cai
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China.
| | - Yuanrong Yang
- Department of Pharmacy, Jingzhou Hospital Affiliated to Yangtze University, Jingzhou, Hubei, 434020, China.
| |
Collapse
|
2
|
Guthrie G, Vonderohe C, Burrin D. Fibroblast growth factor 15/19 expression, regulation, and function: An overview. Mol Cell Endocrinol 2022; 548:111617. [PMID: 35301051 PMCID: PMC9038700 DOI: 10.1016/j.mce.2022.111617] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Accepted: 01/18/2022] [Indexed: 12/12/2022]
Abstract
Since the discovery of fibroblast growth factor (FGF)-19 over 20 years ago, our understanding of the peptide and its role in human biology has moved forward significantly. A member of a superfamily of paracrine growth factors regulating embryonic development, FGF19 is unique in that it is a dietary-responsive endocrine hormone linked with bile acid homeostasis, glucose and lipid metabolism, energy expenditure, and protein synthesis during the fed to fasted state. FGF19 achieves this through targeting multiple tissues and signaling pathways within those tissues. The diverse functional capabilities of FGF19 is due to the unique structural characteristics of the protein and its receptor binding in various cell types. This review will cover the current literature on the protein FGF19, its target receptors, and the biological pathways they target through unique signaling cascades.
Collapse
Affiliation(s)
- Greg Guthrie
- USDA-ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, United States
| | - Caitlin Vonderohe
- USDA-ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, United States
| | - Douglas Burrin
- USDA-ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, United States.
| |
Collapse
|
3
|
He WP, Yang GP, Yang ZX, Shen HW, You ZS, Yang GF. Maelstrom promotes tumor metastasis through regulation of FGFR4 and epithelial-mesenchymal transition in epithelial ovarian cancer. J Ovarian Res 2022; 15:55. [PMID: 35513870 PMCID: PMC9074322 DOI: 10.1186/s13048-022-00992-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2022] [Accepted: 04/27/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Increasing evidence has indicated that Maelstrom (MAEL) plays an oncogenic role in various human carcinomas. However, the exact function and mechanisms by which MAEL acts in epithelial ovarian cancer (EOC) remain unclear. RESULTS This study demonstrated that MAEL was frequently overexpressed in EOC tissues and cell lines. Overexpression of MAEL was positively correlated with the histological grade of tumors, FIGO stage, and pT/pN/pM status (p < 0.05), and it also acted as an independent predictor of poor patient survival (p < 0.001). Ectopic overexpression of MAEL substantially promoted invasiveness/metastasis and induced epithelial-mesenchymal transition (EMT), whereas silencing MAEL by short hairpin RNA effectively inhibited its oncogenic function and attenuated EMT. Further study demonstrated that fibroblast growth factor receptor 4 (FGFR4) was a critical downstream target of MAEL in EOC, and the expression levels of FGFR4 were significantly associated with MAEL. (P < 0.05). CONCLUSION Our findings suggest that overexpression of MAEL plays a crucial oncogenic role in the development and progression of EOC through the upregulation of FGFR4 and subsequent induction of EMT, and also provide new insights on its potential as a therapeutic target for EOC.
Collapse
Affiliation(s)
- Wei-Peng He
- Department of Gynecology, the First Affiliated Hospital, Sun Yat-Sen University, No. 58, Zhongshan Road II, Guangzhou, 510080, China
| | - Gui-Ping Yang
- Department of Gynecology, the First Affiliated Hospital, Sun Yat-Sen University, No. 58, Zhongshan Road II, Guangzhou, 510080, China
| | - Zun-Xian Yang
- Department of Gynecology, the First Affiliated Hospital, Sun Yat-Sen University, No. 58, Zhongshan Road II, Guangzhou, 510080, China
| | - Hong-Wei Shen
- Department of Gynecology, the First Affiliated Hospital, Sun Yat-Sen University, No. 58, Zhongshan Road II, Guangzhou, 510080, China
| | - Ze-Shan You
- Department of Gynecology, the First Affiliated Hospital, Sun Yat-Sen University, No. 58, Zhongshan Road II, Guangzhou, 510080, China
| | - Guo-Fen Yang
- Department of Gynecology, the First Affiliated Hospital, Sun Yat-Sen University, No. 58, Zhongshan Road II, Guangzhou, 510080, China.
| |
Collapse
|
4
|
Kim JH, Jeong SY, Jang HJ, Park ST, Kim HS. FGFR4 Gly388Arg Polymorphism Reveals a Poor Prognosis, Especially in Asian Cancer Patients: A Meta-Analysis. Front Oncol 2021; 11:762528. [PMID: 34737965 PMCID: PMC8560792 DOI: 10.3389/fonc.2021.762528] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Accepted: 10/04/2021] [Indexed: 12/11/2022] Open
Abstract
The fibroblast growth factor-4 receptor (FGFR4) is a member of receptor tyrosine kinase. The FGFR4 Gly388Arg polymorphism in the transmembrane domain of the receptor has been shown to increase genetic susceptibility to cancers. However, its prognostic impact in cancer patients still remains controversial. Herein, we performed this meta-analysis to evaluate the clinicopathological and prognostic impacts of the FGFR4 Gly388Arg polymorphism in patients with cancer. We carried out a computerized extensive search using PubMed, Medline, and Ovid Medline databases up to July 2021. From 44 studies, 11,574 patients were included in the current meta-analysis. Regardless of the genetic models, there was no significant correlation of the FGFR4 Gly388Arg polymorphism with disease stage 3/4. In the homozygous model (Arg/Arg vs. Gly/Gly), the Arg/Arg genotype tended to show higher rate of lymph node metastasis compared with the Gly/Gly genotype (odds ratio = 1.21, 95% confidence interval (CI): 0.99-1.49, p = 0.06). Compared to patients with the Arg/Gly or Arg/Arg genotype, those with the Gly/Gly genotype had significantly better overall survival (hazard ratios (HR) = 1.19, 95% CI: 1.05-1.35, p = 0.006) and disease-free survival (HR = 1.25, 95% CI: 1.03-1.53, p = 0.02). In conclusion, this meta-analysis showed that the FGFR4 Gly388Arg polymorphism was significantly associated with worse prognosis in cancer patients. Our results suggest that this polymorphism may be a valuable genetic marker to identify patients at higher risk of recurrence or mortality.
Collapse
Affiliation(s)
- Jung Han Kim
- Division of Hemato-Oncology, Department of Internal Medicine, Kangnam Sacred-Heart Hospital, Hallym University Medical Center, College of Medicine, Hallym University, Seoul, South Korea
| | - Soo Young Jeong
- Department of Obstetrics and Gynecology, Kangnam Sacred-Heart Hospital, Hallym University Medical Center, College of Medicine, Hallym University, Seoul, South Korea
| | - Hyun Joo Jang
- Division of Gastroenterology, Department of Internal Medicine, Dongtan Sacred-Heart Hospital, Hallym University Medical Center, College of Medicine, Hallym University, Hwasung, South Korea
| | - Sung Taek Park
- Department of Obstetrics and Gynecology, Kangnam Sacred-Heart Hospital, Hallym University Medical Center, College of Medicine, Hallym University, Seoul, South Korea
| | - Hyeong Su Kim
- Division of Hemato-Oncology, Department of Internal Medicine, Kangnam Sacred-Heart Hospital, Hallym University Medical Center, College of Medicine, Hallym University, Seoul, South Korea
| |
Collapse
|
5
|
Gsur A, Baierl A, Brezina S. Colorectal Cancer Study of Austria (CORSA): A Population-Based Multicenter Study. BIOLOGY 2021; 10:722. [PMID: 34439954 PMCID: PMC8389216 DOI: 10.3390/biology10080722] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Revised: 07/26/2021] [Accepted: 07/26/2021] [Indexed: 12/14/2022]
Abstract
The Colorectal cancer Study of Austria (CORSA) is comprised more than 13,500 newly diagnosed colorectal cancer (CRC) patients, patients with high- and low-risk adenomas as well as population-based controls. The recruitment for the CORSA biobank is performed in close cooperation with the invited two-stage CRC screening project "Burgenland PREvention trial of colorectal Disease with ImmunologiCal Testing" (B-PREDICT). Annually, more than 150,000 inhabitants of the Austrian federal state Burgenland aged between 40 and 80 are invited to participate using FIT-tests as an initial screening. FIT-positive tested participants are offered a diagnostic colonoscopy and are asked to take part in CORSA, sign a written informed consent, complete questionnaires concerning dietary and lifestyle habits and provide an ethylenediaminetetraacetic acid (EDTA) blood sample as well as a stool sample. Additional CRC cases have been recruited at four hospitals in Vienna and a hospital in lower Austria. A major strength of CORSA is the population-based controls who are FIT-positive and colonoscopy-confirmed to be free of polyps and/or CRC.
Collapse
Affiliation(s)
- Andrea Gsur
- Institute of Cancer Research, Department of Medicine I, Medical University of Vienna, 1090 Vienna, Austria;
| | - Andreas Baierl
- Department of Statistics and Operations Research, University of Vienna, 1010 Vienna, Austria;
| | - Stefanie Brezina
- Institute of Cancer Research, Department of Medicine I, Medical University of Vienna, 1090 Vienna, Austria;
| |
Collapse
|
6
|
Role of FGF15 in Hepatic Surgery in the Presence of Tumorigenesis: Dr. Jekyll or Mr. Hyde? Cells 2021; 10:cells10061421. [PMID: 34200439 PMCID: PMC8228386 DOI: 10.3390/cells10061421] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Revised: 05/26/2021] [Accepted: 06/04/2021] [Indexed: 12/11/2022] Open
Abstract
The pro-tumorigenic activity of fibroblast growth factor (FGF) 19 (FGF15 in its rodent orthologue) in hepatocellular carcinoma (HCC), as well as the unsolved problem that ischemia-reperfusion (IR) injury supposes in liver surgeries, are well known. However, it has been shown that FGF15 administration protects against liver damage and regenerative failure in liver transplantation (LT) from brain-dead donors without tumor signals, providing a benefit in avoiding IR injury. The protection provided by FGF15/19 is due to its anti-apoptotic and pro-regenerative properties, which make this molecule a potentially beneficial or harmful factor, depending on the disease. In the present review, we describe the preclinical models currently available to understand the signaling pathways responsible for the apparent controversial effects of FGF15/19 in the liver (to repair a damaged liver or to promote tumorigenesis). As well, we study the potential pharmacological use that has the activation or inhibition of FGF15/19 pathways depending on the disease to be treated. We also discuss whether FGF15/19 non-pro-tumorigenic variants, which have been developed for the treatment of liver diseases, might be promising approaches in the surgery of hepatic resections and LT using healthy livers and livers from extended-criteria donors.
Collapse
|
7
|
Shiu BH, Hsieh MH, Ting WC, Chou MC, Chang LC, Huang CC, Su SC, Yang SF. Impact of FGFR4 Gene Polymorphism on the Progression of Colorectal Cancer. Diagnostics (Basel) 2021; 11:diagnostics11060978. [PMID: 34071523 PMCID: PMC8227855 DOI: 10.3390/diagnostics11060978] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 05/19/2021] [Accepted: 05/27/2021] [Indexed: 02/06/2023] Open
Abstract
Colorectal cancer (CRC) is a multifactorial malignancy, and its high incidence and mortality rate remain a global public health burden. Fibroblast growth factor receptor 4 (FGFR4) is a receptor tyrosine kinase that has been shown to play a key role in cancer development and prognosis via the activation of its downstream oncogenic signaling pathways. The present study aimed to explore the impact of FGFR4 gene polymorphisms on the risk and progression of CRC. Three FGFR4 single-nucleotide polymorphisms (SNPs), including rs1966265, rs351855, and rs7708357, were evaluated in 413 CRC cases and 413 gender- and age-matched cancer-free controls. We did not observe any significant association of three individual SNPs with the risk of CRC between the case and control group. However, while assessing the clinicopathological parameters, patients of rectal cancer possessing at least one minor allele of rs1966265 (AG and GG; AOR, 0.236; p = 0.046) or rs351855 (GA and AA; AOR, 0.191; p = 0.022) were found to develop less metastasis as compared to those who are homozygous for the major allele. Further analyses using the datasets from the Genotype-Tissue Expression (GTEx) Portal and The Cancer Genome Atlas (TCGA) revealed that rs351855 regulated FGFR4 expression in many human tissues, and increased FGFR4 levels were associated with the occurrence, advanced stage, and distal metastasis of colon adenocarcinoma. These data suggest that the amino acid change in combination with altered expression levels of FGFR4 due to genetic polymorphisms may affect CRC progression.
Collapse
Affiliation(s)
- Bei-Hao Shiu
- Institute of Medicine, Chung Shan Medical University, Taichung 402, Taiwan; (B.-H.S.); (M.-H.H.); (W.-C.T.); (M.-C.C.)
- Department of Surgery, Chung Shan Medical University Hospital, Taichung 402, Taiwan
| | - Ming-Hong Hsieh
- Institute of Medicine, Chung Shan Medical University, Taichung 402, Taiwan; (B.-H.S.); (M.-H.H.); (W.-C.T.); (M.-C.C.)
- School of Medicine, Chung Shan Medical University, Taichung 402, Taiwan
- Department of Psychiatry, Chung Shan Medical University Hospital, Taichung 402, Taiwan
| | - Wen-Chien Ting
- Institute of Medicine, Chung Shan Medical University, Taichung 402, Taiwan; (B.-H.S.); (M.-H.H.); (W.-C.T.); (M.-C.C.)
- Department of Surgery, Chung Shan Medical University Hospital, Taichung 402, Taiwan
- School of Medicine, Chung Shan Medical University, Taichung 402, Taiwan
| | - Ming-Chih Chou
- Institute of Medicine, Chung Shan Medical University, Taichung 402, Taiwan; (B.-H.S.); (M.-H.H.); (W.-C.T.); (M.-C.C.)
- Department of Surgery, Chung Shan Medical University Hospital, Taichung 402, Taiwan
- School of Medicine, Chung Shan Medical University, Taichung 402, Taiwan
| | - Lun-Ching Chang
- Department of Mathematical Sciences, Florida Atlantic University, Boca Raton, FL 33431, USA;
| | - Chi-Chou Huang
- Institute of Medicine, Chung Shan Medical University, Taichung 402, Taiwan; (B.-H.S.); (M.-H.H.); (W.-C.T.); (M.-C.C.)
- Department of Surgery, Chung Shan Medical University Hospital, Taichung 402, Taiwan
- School of Medicine, Chung Shan Medical University, Taichung 402, Taiwan
- Correspondence: (C.-C.H.); (S.-C.S.); (S.-F.Y.)
| | - Shih-Chi Su
- Whole-Genome Research Core Laboratory of Human Diseases, Chang Gung Memorial Hospital, Keelung 204, Taiwan
- Department of Dermatology, Drug Hypersensitivity Clinical and Research Center, Chang Gung Memorial Hospital, Linkou 333, Taiwan
- Correspondence: (C.-C.H.); (S.-C.S.); (S.-F.Y.)
| | - Shun-Fa Yang
- Institute of Medicine, Chung Shan Medical University, Taichung 402, Taiwan; (B.-H.S.); (M.-H.H.); (W.-C.T.); (M.-C.C.)
- Department of Medical Research, Chung Shan Medical University Hospital, Taichung 402, Taiwan
- Correspondence: (C.-C.H.); (S.-C.S.); (S.-F.Y.)
| |
Collapse
|
8
|
An updated meta-analysis of the association between fibroblast growth factor receptor 4 polymorphisms and susceptibility to cancer. Biosci Rep 2021; 40:226581. [PMID: 33017009 PMCID: PMC7584815 DOI: 10.1042/bsr20192051] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2019] [Revised: 09/30/2020] [Accepted: 09/30/2020] [Indexed: 12/24/2022] Open
Abstract
Fibroblast growth factor receptor 4 (FGFR4) is a cell surface receptor tyrosine kinases (RTKs) for FGFs. Several studies have focused on the association between FGFR4 polymorphisms and cancer development. This meta-analysis aimed to estimate the association between FGFR4 rs351855 (Gly388Arg), rs1966265 (Val10Ile), rs7708357, rs2011077, and rs376618 polymorphisms and cancer risk. Eligible studies were identified from electronic databases. All statistical analyses were achieved with the STATA 14.0 software. Pooled odds ratios (ORs) with 95% confidence intervals (CIs) were used to quantitatively estimate the association. Overall, no significant association was found among rs351855, rs2011077, and rs376618 polymorphisms with the risk of overall cancer. The rs1966265 polymorphism significantly decreased the risk of cancer in recessive (OR = 0.87, 95% CI = 0.78–0.97, P=0.009, TT vs CT+CC) genetic model. Whereas the rs7708357 polymorphism was positively associated with cancer risk in dominant (OR = 1.17, 95% CI = 1.02–1.36, P=0.028) genetic model. Stratified analysis revealed that rs351855 variant significantly increased the risk of prostate cancer in heterozygous (OR = 1.16, 95% CI = 1.02–1.32, P=0.025 AG vs GG), dominant (OR = 1.20, 95% CI = 1.06–1.35, P=0.004, AG+AA vs GG), and allele (OR = 1.22, 95% CI = 1.06–1.41, P=0.005, A vs G) genetic models. In summary, the findings of this meta-analysis indicate that rs1966265, rs7708357, and rs351855 polymorphisms are correlated to cancer development. Further well-designed studies are necessary to draw more precise conclusions.
Collapse
|
9
|
Peng T, Sun Y, Lv Z, Zhang Z, Su Q, Wu H, Zhang W, Yuan W, Zuo L, Shi L, Zhang LF, Zhou X, Mi Y. Effects of FGFR4 G388R, V10I polymorphisms on the likelihood of cancer. Sci Rep 2021; 11:1373. [PMID: 33446698 PMCID: PMC7809464 DOI: 10.1038/s41598-020-80146-y] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2020] [Accepted: 12/14/2020] [Indexed: 12/19/2022] Open
Abstract
The correlation between G388R or V10I polymorphisms of fibroblast growth factor receptor (FGFR) 4 gene and the risk of carcinoma has been investigated previously, but the results are contradictory. Odds ratios (ORs) with 95% confidence intervals (95%CIs), in silico tools, and immunohistochemical staining (IHS) were adopted to assess the association. In total, 13,793 cancer patients and 16,179 controls were evaluated in our pooled analysis. Summarization of all the studies showed that G388R polymorphism is associated with elevated susceptibility to cancer under homozygous comparison (OR = 1.21, 95%CI = 1.03–1.43, P = 0.020) and a recessive genetic model (OR = 1.21, 95%CI = 1.04–1.41, P = 0.012). In the stratification analysis by cancer type and ethnicity, similar findings were indicated for prostate cancer, breast cancer, and individuals of Asian descendant. Polyphen2 bioinformatics analysis showed that the G388R mutation is predicted to damage the protein function of FGFR4. IHS analysis indicated that FGFR4 expression is increased in advanced prostate cancer. These findings may guide personalized treatment of certain types of cancers. Up-regulation of FGFR4 may be related to a poor prognosis in prostate cancer.
Collapse
Affiliation(s)
- Tao Peng
- Department of Urology, Affiliated Hospital of Jiangnan University, Wuxi, 214000, People's Republic of China
| | - Yangyang Sun
- Department of Pathology, The Affiliated Changzhou No.2 People's Hospital of Nanjing Medical University, 29 Xinglong Road, Changzhou, 213003, People's Republic of China
| | - Zhiwei Lv
- Department of Urology, The Affiliated Changzhou No.2 People's Hospital of Nanjing Medical University, 29 Xinglong Road, Changzhou, 213003, People's Republic of China
| | - Ze Zhang
- Department of Urology, The Affiliated Changzhou No.2 People's Hospital of Nanjing Medical University, 29 Xinglong Road, Changzhou, 213003, People's Republic of China
| | - Quanxin Su
- Department of Urology, The Affiliated Changzhou No.2 People's Hospital of Nanjing Medical University, 29 Xinglong Road, Changzhou, 213003, People's Republic of China
| | - Hao Wu
- Department of Urology, The Affiliated Changzhou No.2 People's Hospital of Nanjing Medical University, 29 Xinglong Road, Changzhou, 213003, People's Republic of China
| | - Wei Zhang
- Department of Oncology, Taizhou People's Hospital, South Hailing Road 399, Taizhou, 225300, People's Republic of China
| | - Wei Yuan
- Department of Cardiology, Taizhou People's Hospital, South Hailing Road 399, Taizhou, 225300, People's Republic of China
| | - Li Zuo
- Department of Urology, The Affiliated Changzhou No.2 People's Hospital of Nanjing Medical University, 29 Xinglong Road, Changzhou, 213003, People's Republic of China
| | - Li Shi
- Department of Urology, The Affiliated Changzhou No.2 People's Hospital of Nanjing Medical University, 29 Xinglong Road, Changzhou, 213003, People's Republic of China
| | - Li-Feng Zhang
- Department of Urology, The Affiliated Changzhou No.2 People's Hospital of Nanjing Medical University, 29 Xinglong Road, Changzhou, 213003, People's Republic of China.
| | - Xiaoli Zhou
- Department of Pathology, The Affiliated Changzhou No.2 People's Hospital of Nanjing Medical University, 29 Xinglong Road, Changzhou, 213003, People's Republic of China.
| | - Yuanyuan Mi
- Department of Urology, Affiliated Hospital of Jiangnan University, Wuxi, 214000, People's Republic of China.
| |
Collapse
|
10
|
Levine KM, Ding K, Chen L, Oesterreich S. FGFR4: A promising therapeutic target for breast cancer and other solid tumors. Pharmacol Ther 2020; 214:107590. [PMID: 32492514 PMCID: PMC7494643 DOI: 10.1016/j.pharmthera.2020.107590] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Accepted: 05/26/2020] [Indexed: 02/07/2023]
Abstract
The fibroblast growth factor receptor (FGFR) signaling pathway has long been known to cancer researchers because of its role in cell survival, proliferation, migration, and angiogenesis. Dysregulation of FGFR signaling is frequently reported in cancer studies, but most of these studies focus on FGFR1-3. However, there is growing evidence implicating an important and unique role of FGFR4 in oncogenesis, tumor progression, and resistance to anti-tumor therapy in multiple types of cancer. Importantly, there are several novel FGFR4-specific inhibitors in clinical trials, making FGFR4 an attractive target for further research. In this review, we focus on assessing the role of FGFR4 in cancer, with an emphasis on breast cancer. First, the structure, physiological functions and downstream signaling pathways of FGFR4 are introduced. Next, different mechanisms reported to cause aberrant FGFR4 activation and their functions in cancer are discussed, including FGFR4 overexpression, FGF ligand overexpression, FGFR4 somatic hotspot mutations, and the FGFR4 G388R single nucleotide polymorphism. Finally, ongoing and recently completed clinical trials targeting FGFRs in cancer are reviewed, highlighting the therapeutic potential of FGFR4 inhibition for the treatment of breast cancer.
Collapse
MESH Headings
- Animals
- Female
- Humans
- Antineoplastic Agents/adverse effects
- Antineoplastic Agents/therapeutic use
- Biomarkers, Tumor/antagonists & inhibitors
- Biomarkers, Tumor/genetics
- Biomarkers, Tumor/metabolism
- Breast Neoplasms/drug therapy
- Breast Neoplasms/enzymology
- Breast Neoplasms/genetics
- Breast Neoplasms/pathology
- Gene Expression Regulation, Neoplastic
- Molecular Targeted Therapy
- Mutation
- Polymorphism, Single Nucleotide
- Protein Kinase Inhibitors/adverse effects
- Protein Kinase Inhibitors/therapeutic use
- Receptor, Fibroblast Growth Factor, Type 4/antagonists & inhibitors
- Receptor, Fibroblast Growth Factor, Type 4/genetics
- Receptor, Fibroblast Growth Factor, Type 4/metabolism
- Signal Transduction
Collapse
Affiliation(s)
- Kevin M Levine
- Women's Cancer Research Center, UPMC Hillman Cancer Center, Pittsburgh, PA, USA; Magee-Women's Research Institute, Magee-Women's Research Hospital of University of Pittsburgh Medical Center, Pittsburgh, PA, USA; Department of Pathology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Kai Ding
- Women's Cancer Research Center, UPMC Hillman Cancer Center, Pittsburgh, PA, USA; Magee-Women's Research Institute, Magee-Women's Research Hospital of University of Pittsburgh Medical Center, Pittsburgh, PA, USA; Integrative Systems Biology Program, University of Pittsburgh, Pittsburgh, PA, USA
| | - Lyuqin Chen
- Women's Cancer Research Center, UPMC Hillman Cancer Center, Pittsburgh, PA, USA; Magee-Women's Research Institute, Magee-Women's Research Hospital of University of Pittsburgh Medical Center, Pittsburgh, PA, USA; Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Steffi Oesterreich
- Women's Cancer Research Center, UPMC Hillman Cancer Center, Pittsburgh, PA, USA; Magee-Women's Research Institute, Magee-Women's Research Hospital of University of Pittsburgh Medical Center, Pittsburgh, PA, USA; Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, PA, USA.
| |
Collapse
|
11
|
Liu Y, Cao M, Cai Y, Li X, Zhao C, Cui R. Dissecting the Role of the FGF19-FGFR4 Signaling Pathway in Cancer Development and Progression. Front Cell Dev Biol 2020; 8:95. [PMID: 32154250 PMCID: PMC7044267 DOI: 10.3389/fcell.2020.00095] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Accepted: 02/04/2020] [Indexed: 12/11/2022] Open
Abstract
Fibroblast growth factor (FGF) receptor 4 (FGFR4) belongs to a family of tyrosine kinase receptor. FGFR4 is highly activated in certain types of cancer and its activation is closely associated with its specific ligand, FGF19. Indeed, FGF19-FGFR4 signaling is implicated in many cellular processes including cell proliferation, migration, metabolism, and differentiation. Since active FGF19-FGFR4 signaling acts as an oncogenic pathway in certain types of cancer, the development and therapeutic evaluation of FGFR4-specific inhibitors in cancer patients is a topic of significant interest. In this review, we aim to provide an updated overview of currently-available FGFR4 inhibitors and their ongoing clinical trials, as well as upcoming potential therapeutics. Further, we examined the possibility of enhancing the therapeutic efficiency of FGFR4 inhibitors in cancer patients. We also discussed the underlying molecular mechanisms of oncogenic activation of FGFR4 by FGF19.
Collapse
Affiliation(s)
- Yanan Liu
- Cancer and Anticancer Drug Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Meng Cao
- Cancer and Anticancer Drug Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Yuepiao Cai
- Cancer and Anticancer Drug Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Xiaokun Li
- Cancer and Anticancer Drug Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Chengguang Zhao
- Cancer and Anticancer Drug Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
- Wenzhou University-Wenzhou Medical University Collaborative Innovation Center of Biomedical, Wenzhou, China
- Institute of Life Sciences, Wenzhou University, Wenzhou, China
| | - Ri Cui
- Cancer and Anticancer Drug Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
- Wenzhou University-Wenzhou Medical University Collaborative Innovation Center of Biomedical, Wenzhou, China
- Institute of Life Sciences, Wenzhou University, Wenzhou, China
| |
Collapse
|
12
|
Expression of FGF8, FGF18, and FGFR4 in Gastroesophageal Adenocarcinomas. Cells 2019; 8:cells8091092. [PMID: 31527546 PMCID: PMC6770911 DOI: 10.3390/cells8091092] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2019] [Revised: 08/09/2019] [Accepted: 09/12/2019] [Indexed: 02/06/2023] Open
Abstract
Even though distinctive advances in the field of esophageal cancer therapy have occurred over the last few years, patients' survival rates remain poor. FGF8, FGF18, and FGFR4 have been identified as promising biomarkers in a number of cancers; however no data exist on expression of FGF8, FGF18, and FGFR4 in adenocarcinomas of the esophago-gastric junction (AEG). A preliminary analysis of the Cancer Genome Atlas (TCGA) database on FGF8, FGF18, and FGFR4 mRNA expression data of patients with AEG was performed. Furthermore, protein levels of FGF8, FGF18, and FGFR4 in diagnostic biopsies and post-operative specimens in neoadjuvantly treated and primarily resected patients using immunohistochemistry were investigated. A total of 242 patients was analyzed in this study: 87 patients were investigated in the TCGA data set analysis and 155 patients in the analysis of protein expression using immunohistochemistry. High protein levels of FGF8, FGF18, and FGFR4 were detected in 94 (60.7%), 49 (31.6%) and 84 (54.2%) patients, respectively. Multivariable Cox proportional hazard regression models revealed that high expression of FGF8 was an independent prognostic factor for diminished overall survival for all patients and for neoadjuvantly treated patients. By contrast, FGF18 overexpression was significantly associated with longer survival rates in neoadjuvantly treated patients. In addition, FGF8 protein level correlated with Mandard regression due to neoadjuvant therapy, indicating potential as a predictive marker. In summary, FGF8 and FGF18 are promising candidates for prognostic factors in adenocarcinomas of the esophago-gastric junction and new potential targets for new anti-cancer therapies.
Collapse
|
13
|
FGF8 induces therapy resistance in neoadjuvantly radiated rectal cancer. J Cancer Res Clin Oncol 2018; 145:77-86. [PMID: 30276721 PMCID: PMC6326005 DOI: 10.1007/s00432-018-2757-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2018] [Accepted: 09/19/2018] [Indexed: 01/09/2023]
Abstract
Purpose Therapy response to neoadjuvant radiochemotherapy (nRCT) of locally advanced rectal cancer varies widely so that markers predicting response are urgently needed. Fibroblast growth factor (FGF) and FGF receptor (FGFR) signaling is involved in pro-survival signaling and thereby may result in radiation resistance. Methods In a cohort of 43 rectal cancer patients, who received nRCT, we analyzed protein levels of FGF 8 and its downstream target Survivin by immunohistochemistry to assess their impact on nRCT response. In vitro resistance models were created by exposing colorectal cancer cell lines to fractionated irradiation and selecting long-term survivors. Results Our findings revealed significantly higher FGF8 and Survivin staining scores in pre-treatment biopsies as well as in surgical specimens of non-responsive compared to responsive patients. Functional studies demonstrated dose-dependent induction of FGF8 mRNA expression in mismatch-incompetent DLD1 cells already after one dose of irradiation. Surviving clones after one or two series of radiation were more resistant to an additional radiation fraction than non-irradiated controls and showed a significant increase in expression of the FGF8 receptor FGFR3 and of Survivin on both the RNA and the protein levels. Conclusion The results of this study suggest that FGF8 and Survivin contribute to radiation resistance in rectal cancer and may serve as markers to select patients who may not benefit from neoadjuvant radiotherapy. Electronic supplementary material The online version of this article (10.1007/s00432-018-2757-7) contains supplementary material, which is available to authorized users.
Collapse
|
14
|
Yamamoto M, Nomura S, Hosoi A, Nagaoka K, Iino T, Yasuda T, Saito T, Matsushita H, Uchida E, Seto Y, Goldenring JR, Kakimi K, Tatematsu M, Tsukamoto T. Established gastric cancer cell lines transplantable into C57BL/6 mice show fibroblast growth factor receptor 4 promotion of tumor growth. Cancer Sci 2018. [PMID: 29532565 PMCID: PMC5980194 DOI: 10.1111/cas.13569] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Previously no mouse gastric cancer cell lines have been available for transplantation into C57BL/6 mice. However, a gastric cancer model in immunocompetent mice would be useful for analyzing putative therapies. N-Methyl-N-nitrosourea (MNU) was given in drinking water to C57BL/6 mice and p53 heterozygous knockout mice. Only 1 tumor from a p53 knockout mouse could be cultured and the cells s.c. transplanted into a C57BL/6 mouse. We cultured this s.c. tumor, and subcloned it. mRNA expression in the most aggressive YTN16 subline was compared to the less aggressive YTN2 subline by microarray analysis, and fibroblast growth factor receptor 4 (FGFR4) in YTN16 cells was knocked out with a CRISPR/Cas9 system and inhibited by an FGFR4 selective inhibitor, BLU9931. These transplanted cell lines formed s.c. tumors in C57BL/6 mice. Four cell lines (YTN2, YTN3, YTN5, YTN16) were subcloned and established. Their in vitro growth rates were similar. However, s.c. tumor establishment rates, metastatic rates, and peritoneal dissemination rates of YTN2 and YTN3 were lower than for YTN5 and YTN16. YTN16 established 8/8 s.c. tumors, 7/8 with lung metastases, 3/8 with lymph node metastases and 5/5 with peritoneal dissemination. FGFR4 expression by YTN16 was 121-fold higher than YTN2. FGFR4-deleted YTN16 cells failed to form s.c. tumors and showed lower rates of peritoneal dissemination. BLU9931 significantly inhibited the growth of peritoneal dissemination of YTN16. These studies present the first transplantable mouse gastric cancer lines. Our results further indicate that FGFR4 is an important growth signal receptor in gastric cancer cells with high FGFR4 expression.
Collapse
Affiliation(s)
- Masami Yamamoto
- Department of Pathology, Nippon Veterinary and Life Science University, Tokyo, Japan
| | - Sachiyo Nomura
- Department of Gastrointestinal Surgery, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Akihiro Hosoi
- Department of Immunotherapeutics, The University of Tokyo Hospital, Tokyo, Japan
| | - Koji Nagaoka
- Department of Immunotherapeutics, The University of Tokyo Hospital, Tokyo, Japan
| | - Tamaki Iino
- Department of Immunotherapeutics, The University of Tokyo Hospital, Tokyo, Japan
| | - Tomohiko Yasuda
- Department of Gastrointestinal Surgery, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan.,Department of Surgery, Nippon Medical School, Tokyo, Japan
| | - Tomoko Saito
- Institute of Immunology Co., Ltd, Utsunomiya Laboratory, Genetic Modified Animal Group, Utsunomiya, Japan
| | - Hirokazu Matsushita
- Department of Immunotherapeutics, The University of Tokyo Hospital, Tokyo, Japan
| | - Eiji Uchida
- Department of Surgery, Nippon Medical School, Tokyo, Japan
| | - Yasuyuki Seto
- Department of Gastrointestinal Surgery, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - James R Goldenring
- Department of Surgery and the Epithelial Biology Center, Vanderbilt University School of Medicine, Nashville VA Medical Center, Nashville, TN, USA
| | - Kazuhiko Kakimi
- Department of Immunotherapeutics, The University of Tokyo Hospital, Tokyo, Japan
| | | | - Tetsuya Tsukamoto
- Department of Pathology, Graduate School of Medicine, Fujita Health University, Toyoake, Japan
| |
Collapse
|
15
|
Xiong SW, Ma J, Feng F, Fu W, Shu SR, Ma T, Wu C, Liu GC, Zhu J. Functional FGFR4 Gly388Arg polymorphism contributes to cancer susceptibility: Evidence from meta-analysis. Oncotarget 2018; 8:25300-25309. [PMID: 28445975 PMCID: PMC5421931 DOI: 10.18632/oncotarget.15811] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2016] [Accepted: 02/07/2017] [Indexed: 02/06/2023] Open
Abstract
Fibroblast growth factor receptor 4 (FGFR4) is a member of receptor tyrosine kinase family. A functional Gly388Arg (rs351855 G>A) polymorphism in FGFR4 gene causes a glycine-to-arginine change at codon 388 within the transmembrane domain of the receptor. Although the FGFR4 rs351855 G>A polymorphism has been implicated in cancer development, its association with cancer risk remains controversial. Here, we have systematically analyzed the association between the rs351855 G>A polymorphism and cancer risk by performing a meta-analysis of 27 studies consisting of 8,682 cases and 9,731 controls. Odds ratios (ORs) and 95% confidence intervals (CIs) were calculated to measure the strength of the association. The rs351855 G>A polymorphism was associated with an increased cancer risk under the recessive model (OR=1.19, 95% CI=1.01-1.41). Stratified analysis by cancer type indicated the rs351855 G>A polymorphism was associated with an increased risk of breast and prostate cancer, but a decreased risk of lung cancer. This meta-analysis demonstrates the FGFR rs351855 G>A polymorphism is associated with increased cancer risk and suggests it could potentially serve as a chemotherapeutic target or biomarker to screen high-risk individuals.
Collapse
Affiliation(s)
- Si-Wei Xiong
- Department of Urology, Guangzhou First People's Hospital, Guangzhou Medical University, Guangzhou 510180, Guangdong, China
| | - Jianqun Ma
- Department of Thoracic Surgery, Harbin Medical University Cancer Hospital, Harbin 150040, Heilongjiang, China
| | - Fen Feng
- Department of Gastroenterology, The First People's Hospital of Foshan (Affiliated Foshan Hospital of Sun Yat-sen University), Foshan 528000, Guangdong, China
| | - Wen Fu
- Department of Pediatric Urology, Department of Pediatric Surgery, Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou 510623, Guangdong, China
| | - Shan-Rong Shu
- Department of Gynecology and Obstetrics, The First Affiliated Hospital of Jinan University, Guangzhou 510630, Guangdong, China
| | - Tianjiao Ma
- Department of Internal Medicine, Harbin Medical University, Harbin 150081, Heilongjiang, China
| | - Caixia Wu
- Molecular Epidemiology Laboratory and Department of Laboratory Medicine, Harbin Medical University Cancer Hospital, Harbin 150040, Heilongjiang, China
| | - Guo-Chang Liu
- Department of Pediatric Urology, Department of Pediatric Surgery, Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou 510623, Guangdong, China
| | - Jinhong Zhu
- Molecular Epidemiology Laboratory and Department of Laboratory Medicine, Harbin Medical University Cancer Hospital, Harbin 150040, Heilongjiang, China
| |
Collapse
|
16
|
Fibroblast growth factor receptor 4 induced resistance to radiation therapy in colorectal cancer. Oncotarget 2018; 7:69976-69990. [PMID: 27650548 PMCID: PMC5342528 DOI: 10.18632/oncotarget.12099] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2016] [Accepted: 09/12/2016] [Indexed: 12/14/2022] Open
Abstract
In colorectal cancer (CRC), fibroblast growth factor receptor 4 (FGFR4) is upregulated and acts as an oncogene. This study investigated the impact of this receptor on the response to neoadjuvant radiotherapy by analyzing its levels in rectal tumors of patients with different responses to the therapy. Cellular mechanisms of FGFR4-induced radioresistance were analyzed by silencing or over-expressing FGFR4 in CRC cell line models. Our findings showed that the FGFR4 staining score was significantly higher in pre-treatment biopsies of non-responsive than responsive patients. Similarly, high expression of FGFR4 inhibited radiation response in cell line models. Silencing or inhibition of FGFR4 resulted in a reduction of RAD51 levels and decreased survival in radioresistant HT29 cells. Increased RAD51 expression rescued cells in the siFGFR4-group. In radiosensitive SW480 and DLD1 cells, enforced expression of FGFR4 stabilized RAD51 protein levels resulting in enhanced clearance of γ-H2AX foci and increased cell survival in the mismatch repair (MMR)-proficient SW480 cells. MMR-deficient DLD1 cells are defective in homologous recombination repair and no FGFR4-induced radioresistance was observed. Based on our results, FGFR4 may serve as a predictive marker to select CRC patients with MMR-proficient tumors who may benefit from pre-operative radiotherapy.
Collapse
|
17
|
Abstract
PURPOSE OF REVIEW Interstitial lung disease (ILD) is comprised of a heterogeneous group of disorders with highly variable natural histories and response to therapies. Pharmacogenetics focuses on the variability in drug response because of the presence of genetic factors that influence drug metabolism or disease activity. In this article, we review relevant drug-specific and disease-specific polymorphisms that may influence therapeutic response, and then highlight a recently identified drug-gene interaction in patients with idiopathic pulmonary fibrosis (IPF). RECENT FINDINGS The emergence of high-throughput genomic technology has allowed for identification of gene polymorphisms associated with susceptibility to specific disease states, including IPF and several connective tissue diseases known to cause ILD. IPF risk loci span a diverse group of genes, while most associated with connective tissue disease are critical to immune signaling. A recent pharmacogenetic analysis of patients enrolled in an IPF clinical trial identified a variant within TOLLIP to be associated with differential response to N-acetylcysteine therapy. SUMMARY Though few pharmacogenetic investigations have been conducted in patients with ILD to date, ample opportunities for pharmacogenetic exploration exist in this patient population. Such exploration will advance our understanding of specific ILDs and help usher in an era of personalized medicine.
Collapse
|
18
|
Functional FGFR4 Gly388Arg polymorphism contributes to oral squamous cell carcinoma susceptibility. Oncotarget 2017; 8:96225-96238. [PMID: 29221201 PMCID: PMC5707095 DOI: 10.18632/oncotarget.21958] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2017] [Accepted: 09/23/2017] [Indexed: 02/06/2023] Open
Abstract
Aberrations of the fibroblast growth factor receptor 4 (FGFR4) genomic region include amplification of FGFR4, activation of FGFR4 kinase domain mutations, and overexpression of FGFR4, which lead to sustained cell proliferation and contribute to tumor development. However, the association between FGFR4 single-nucleotide polymorphisms (SNPs) and risk of oral squamous cell carcinoma (OSCC) remains to be determined. We investigated the relationships between FGFR4 genetic polymorphisms, OSCC development and clinicopathological variables. We recruited a total of 955 patients with OSCC and 1191 controls. Four SNPs of FGFR4 (rs2011077, rs351855, rs7708357, and rs1966265) were examined using real-time polymerase chain reaction. We found that with the rs351855 GA genotype and a combination of the GA and AA genotypes exhibited a 1.431-fold (95% CI: 1.092-1.876) and 1.335-fold (95% CI: 1.033-1.725) higher risk of OSCC. However, patients with OSCC with a homozygous A/A genotype of FGFR4 rs351855 polymorphism had a lower risk of advanced stage OSCC (P = 0.0252). Furthermore, the patients with the FGFR4 rs351855/rs1966265 A-A haplotype had a 2.890-fold (95% confidence interval [CI]: 2.257-3.700) higher risk of OSCC than the controls. Betel quid chewers with the A-A haplotype had a considerably higher risk (95% CI: 16.159-26.937) of OSCC than did betel quid nonchewers with other haplotypes. Moreover, an additional integrated in silico analysis proposed that rs351855 G allele variant to the A allele exhibited a relatively low energy of the transmembrane region. In conclusion, our results suggest that the FGFR4 rs351855 may play a role in susceptibility for OSCC development.
Collapse
|
19
|
Englinger B, Kallus S, Senkiv J, Heilos D, Gabler L, van Schoonhoven S, Terenzi A, Moser P, Pirker C, Timelthaler G, Jäger W, Kowol CR, Heffeter P, Grusch M, Berger W. Intrinsic fluorescence of the clinically approved multikinase inhibitor nintedanib reveals lysosomal sequestration as resistance mechanism in FGFR-driven lung cancer. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2017; 36:122. [PMID: 28882160 PMCID: PMC5590147 DOI: 10.1186/s13046-017-0592-3] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/21/2017] [Accepted: 09/01/2017] [Indexed: 12/22/2022]
Abstract
Background Studying the intracellular distribution of pharmacological agents, including anticancer compounds, is of central importance in biomedical research. It constitutes a prerequisite for a better understanding of the molecular mechanisms underlying drug action and resistance development. Hyperactivated fibroblast growth factor receptors (FGFRs) constitute a promising therapy target in several types of malignancies including lung cancer. The clinically approved small-molecule FGFR inhibitor nintedanib exerts strong cytotoxicity in FGFR-driven lung cancer cells. However, subcellular pharmacokinetics of this compound and its impact on therapeutic efficacy remain obscure. Methods 3-dimensional fluorescence spectroscopy was conducted to asses cell-free nintedanib fluorescence properties. MTT assay was used to determine the impact of the lysosome-targeting agents bafilomycin A1 and chloroquine combined with nintedanib on lung cancer cell viability. Flow cytometry and live cell as well as confocal microscopy were performed to analyze uptake kinetics as well as subcellular distribution of nintedanib. Western blot was conducted to investigate protein expression. Cryosections of subcutaneous tumor allografts were generated to detect intratumoral nintedanib in mice after oral drug administration. Results Here, we report for the first time drug-intrinsic fluorescence properties of nintedanib in living and fixed cancer cells as well as in cryosections derived from allograft tumors of orally treated mice. Using this feature in conjunction with flow cytometry and confocal microscopy allowed to determine cellular drug accumulation levels, impact of the ABCB1 efflux pump and to uncover nintedanib trapping into lysosomes. Lysosomal sequestration - resulting in an organelle-specific and pH-dependent nintedanib fluorescence - was identified as an intrinsic resistance mechanism in FGFR-driven lung cancer cells. Accordingly, combination of nintedanib with agents compromising lysosomal acidification (bafilomycin A1, chloroquine) exerted distinctly synergistic growth inhibitory effects. Conclusion Our findings provide a powerful tool to dissect molecular factors impacting organismal and intracellular pharmacokinetics of nintedanib. Regarding clinical application, prevention of lysosomal trapping via lysosome-alkalization might represent a promising strategy to circumvent cancer cell-intrinsic nintedanib resistance. Electronic supplementary material The online version of this article (10.1186/s13046-017-0592-3) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Bernhard Englinger
- Institute of Cancer Research and Comprehensive Cancer Center, Department of Medicine I, Medical University of Vienna, Borschkegasse 8a, A-1090, Vienna, Austria
| | - Sebastian Kallus
- Institute of Inorganic Chemistry, University of Vienna, Waehringer Str. 42, A-1090, Vienna, Austria.,Research Cluster "Translational Cancer Therapy Research", University of Vienna, Waehringer Strasse 42, A-1090, Vienna, Austria
| | - Julia Senkiv
- Institute of Cancer Research and Comprehensive Cancer Center, Department of Medicine I, Medical University of Vienna, Borschkegasse 8a, A-1090, Vienna, Austria.,Institute of Cell Biology NAS of Ukraine, Drahomanova str 14/16, 79005, Lviv, Ukraine
| | - Daniela Heilos
- Institute of Cancer Research and Comprehensive Cancer Center, Department of Medicine I, Medical University of Vienna, Borschkegasse 8a, A-1090, Vienna, Austria.,Department of Pharmacology and Toxicology, University of Vienna, Althanstr. 14, 1090, Vienna, Austria
| | - Lisa Gabler
- Institute of Cancer Research and Comprehensive Cancer Center, Department of Medicine I, Medical University of Vienna, Borschkegasse 8a, A-1090, Vienna, Austria
| | - Sushilla van Schoonhoven
- Institute of Cancer Research and Comprehensive Cancer Center, Department of Medicine I, Medical University of Vienna, Borschkegasse 8a, A-1090, Vienna, Austria
| | - Alessio Terenzi
- Institute of Inorganic Chemistry, University of Vienna, Waehringer Str. 42, A-1090, Vienna, Austria
| | - Patrick Moser
- Institute of Cancer Research and Comprehensive Cancer Center, Department of Medicine I, Medical University of Vienna, Borschkegasse 8a, A-1090, Vienna, Austria
| | - Christine Pirker
- Institute of Cancer Research and Comprehensive Cancer Center, Department of Medicine I, Medical University of Vienna, Borschkegasse 8a, A-1090, Vienna, Austria
| | - Gerald Timelthaler
- Institute of Cancer Research and Comprehensive Cancer Center, Department of Medicine I, Medical University of Vienna, Borschkegasse 8a, A-1090, Vienna, Austria
| | - Walter Jäger
- Department of Pharmaceutical Chemistry, Division of Clinical Pharmacy and Diagnostics, University of Vienna, Althanstrasse 14, A-1090, Vienna, Austria
| | - Christian R Kowol
- Institute of Inorganic Chemistry, University of Vienna, Waehringer Str. 42, A-1090, Vienna, Austria.,Research Cluster "Translational Cancer Therapy Research", University of Vienna, Waehringer Strasse 42, A-1090, Vienna, Austria
| | - Petra Heffeter
- Institute of Cancer Research and Comprehensive Cancer Center, Department of Medicine I, Medical University of Vienna, Borschkegasse 8a, A-1090, Vienna, Austria.,Research Cluster "Translational Cancer Therapy Research", University of Vienna, Waehringer Strasse 42, A-1090, Vienna, Austria
| | - Michael Grusch
- Institute of Cancer Research and Comprehensive Cancer Center, Department of Medicine I, Medical University of Vienna, Borschkegasse 8a, A-1090, Vienna, Austria
| | - Walter Berger
- Institute of Cancer Research and Comprehensive Cancer Center, Department of Medicine I, Medical University of Vienna, Borschkegasse 8a, A-1090, Vienna, Austria. .,Research Cluster "Translational Cancer Therapy Research", University of Vienna, Waehringer Strasse 42, A-1090, Vienna, Austria.
| |
Collapse
|
20
|
Association between FGFR4 Gly388Arg polymorphism (rs351855) and cancer risk: A meta analysis including 10,584 subjects. Meta Gene 2017. [DOI: 10.1016/j.mgene.2017.04.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
|
21
|
Erdem ZN, Schwarz S, Drev D, Heinzle C, Reti A, Heffeter P, Hudec X, Holzmann K, Grasl-Kraupp B, Berger W, Grusch M, Marian B. Irinotecan Upregulates Fibroblast Growth Factor Receptor 3 Expression in Colorectal Cancer Cells, Which Mitigates Irinotecan-Induced Apoptosis. Transl Oncol 2017; 10:332-339. [PMID: 28340475 PMCID: PMC5367848 DOI: 10.1016/j.tranon.2017.02.004] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2016] [Accepted: 02/16/2017] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND: Irinotecan (IRI) is an integral part of colorectal cancer (CRC) therapy, but response rates are unsatisfactory and resistance mechanisms are still insufficiently understood. As fibroblast growth factor receptor 3 (FGFR3) mediates essential survival signals in CRC, it is a candidate gene for causing intrinsic resistance to IRI. METHODS: We have used cell line models overexpressing FGFR3 to study the receptor's impact on IRI response. For pathway blockade, a dominant-negative receptor mutant and a small molecule kinase inhibitor were employed. RESULTS: IRI exposure induced expression of FGFR3 as well as its ligands FGF8 and FGF18 both in cell cultures and in xenograft tumors. As overexpression of FGFR3 mitigated IRI-induced apoptosis in CRC cell models, this suggests that the drug itself activated a survival response. On the cellular level, the antiapoptotic protein bcl-xl was upregulated and caspase 3 activation was inhibited. Targeting FGFR3 signaling using a dominant-negative receptor mutant sensitized cells for IRI. In addition, the FGFR inhibitor PD173074 acted synergistically with the chemotherapeutic drug and significantly enhanced IRI-induced caspase 3 activity in vitro. In vivo, PD173074 strongly inhibited growth of IRI-treated tumors. CONCLUSION: Together, our results indicate that targeting FGFR3 can be a promising strategy to enhance IRI response in CRC patients.
Collapse
Affiliation(s)
- Zeynep N Erdem
- Medical University of Vienna, Department of Medicine 1, Institute of Cancer Research and Comprehensive Cancer Center Vienna, Borschkegasse 8a, 1090 Vienna, Austria
| | - Stefanie Schwarz
- Medical University of Vienna, Department of Medicine 1, Institute of Cancer Research and Comprehensive Cancer Center Vienna, Borschkegasse 8a, 1090 Vienna, Austria
| | - Daniel Drev
- Medical University of Vienna, Department of Medicine 1, Institute of Cancer Research and Comprehensive Cancer Center Vienna, Borschkegasse 8a, 1090 Vienna, Austria
| | - Christine Heinzle
- Medical University of Vienna, Department of Medicine 1, Institute of Cancer Research and Comprehensive Cancer Center Vienna, Borschkegasse 8a, 1090 Vienna, Austria
| | - Andrea Reti
- Medical University of Vienna, Department of Medicine 1, Institute of Cancer Research and Comprehensive Cancer Center Vienna, Borschkegasse 8a, 1090 Vienna, Austria
| | - Petra Heffeter
- Medical University of Vienna, Department of Medicine 1, Institute of Cancer Research and Comprehensive Cancer Center Vienna, Borschkegasse 8a, 1090 Vienna, Austria
| | - Xenia Hudec
- Medical University of Vienna, Department of Medicine 1, Institute of Cancer Research and Comprehensive Cancer Center Vienna, Borschkegasse 8a, 1090 Vienna, Austria
| | - Klaus Holzmann
- Medical University of Vienna, Department of Medicine 1, Institute of Cancer Research and Comprehensive Cancer Center Vienna, Borschkegasse 8a, 1090 Vienna, Austria
| | - Bettina Grasl-Kraupp
- Medical University of Vienna, Department of Medicine 1, Institute of Cancer Research and Comprehensive Cancer Center Vienna, Borschkegasse 8a, 1090 Vienna, Austria
| | - Walter Berger
- Medical University of Vienna, Department of Medicine 1, Institute of Cancer Research and Comprehensive Cancer Center Vienna, Borschkegasse 8a, 1090 Vienna, Austria
| | - Michael Grusch
- Medical University of Vienna, Department of Medicine 1, Institute of Cancer Research and Comprehensive Cancer Center Vienna, Borschkegasse 8a, 1090 Vienna, Austria
| | - Brigitte Marian
- Medical University of Vienna, Department of Medicine 1, Institute of Cancer Research and Comprehensive Cancer Center Vienna, Borschkegasse 8a, 1090 Vienna, Austria.
| |
Collapse
|
22
|
Xie X, Wang Z, Chen F, Yuan Y, Wang J, Liu R, Chen Q. Roles of FGFR in oral carcinogenesis. Cell Prolif 2017; 49:261-9. [PMID: 27218663 DOI: 10.1111/cpr.12260] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2016] [Accepted: 03/29/2016] [Indexed: 12/12/2022] Open
Abstract
Fibroblast growth factor receptors (FGFRs) play essential roles in organ development during the embryonic period, and regulate tissue repair in adults. Accumulating evidence suggests that alterations in FGFR signalling are involved in diverse types of cancer. In this review, we focus on aberrant regulation of FGFRs in pathogenesis of oral squamous cell carcinoma (OSCC), including altered expression and subcellular location, aberrant isoform splicing and mutations. We also provide an overview of oncogenic roles of each FGFR and its downstream signalling pathways in regulating OSCC cell proliferation and metastasis. Finally, we discuss potential application of FGFRs as anti-cancer targets in the preclinical environment and in clinical practice.
Collapse
Affiliation(s)
- Xiaoyan Xie
- State Key Laboratory of Oral Diseases, West China College of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Zhiyong Wang
- State Key Laboratory of Oral Diseases, West China College of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Fangman Chen
- State Key Laboratory of Oral Diseases, West China College of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Yao Yuan
- State Key Laboratory of Oral Diseases, West China College of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Jiayi Wang
- State Key Laboratory of Oral Diseases, West China College of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Rui Liu
- State Key Laboratory of Oral Diseases, West China College of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Qianming Chen
- State Key Laboratory of Oral Diseases, West China College of Stomatology, Sichuan University, Chengdu, 610041, China
| |
Collapse
|
23
|
Cho SH, Hong CS, Kim HN, Shin MH, Kim KR, Shim HJ, Hwang JE, Bae WK, Chung IJ. FGFR4 Arg388 Is Correlated with Poor Survival in Resected Colon Cancer Promoting Epithelial to Mesenchymal Transition. Cancer Res Treat 2016; 49:766-777. [PMID: 27857023 PMCID: PMC5512371 DOI: 10.4143/crt.2016.457] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2016] [Accepted: 10/19/2016] [Indexed: 01/01/2023] Open
Abstract
PURPOSE Fibroblast growth factor receptor 4 (FGFR4) plays an important role in cancer progression during tumor proliferation, invasion, and metastasis. This study evaluated the prognostic role of FGFR4 polymorphism in patients with resected colon cancer, including the underlying mechanism. MATERIALS AND METHODS FGFR4 polymorphism was characterized in patientswho received curative resection for stage III colon cancer. FGFR4-dependent signal pathways involving cell proliferation, invasion, and migration according to genotypes were also evaluated in transfected colon cancer cell lines. RESULTS Among a total of 273 patients, the GG of FGFR4 showed significantly better overall survival than the AG or AA, regardless of adjuvant treatment. In the group of AG or AA, combination of folinic acid, fluorouracil, and oxaliplatin (FOLFOX) resulted in better survival than fluorouracil/leucovorin or no adjuvant chemotherapy. However, in GG, there was no difference among treatment regimens. Using multivariate analyses, the Arg388 carriers, together with age, N stage, poor differentiation, absence of a lymphocyte response, and no adjuvant chemotherapy, had a significantly worse OS than patients with the Gly388 allele. In transfected colon cancer cells, overexpression of Arg388 significantly increased cell proliferation and changes in epithelial to mesenchymal transition markers compared with cells overexpressing the Gly388 allele. CONCLUSION The Arg388 allele of FGFR4 may be a biomarker and a candidate target for adjuvant treatment of patients with resected colon cancer.
Collapse
Affiliation(s)
- Sang Hee Cho
- Department of Hemato-Oncology, Chonnam National University Hwasun Hospital, Hwasun, Korea
| | - Chang Soo Hong
- Department of Hemato-Oncology, Chonnam National University Hwasun Hospital, Hwasun, Korea
| | - Hee Nam Kim
- Department of Preventive Medicine, Chonnam National Medical School, Gwangju, Korea
| | - Min Ho Shin
- Department of Preventive Medicine, Chonnam National Medical School, Gwangju, Korea
| | - Ka Rham Kim
- Department of Hemato-Oncology, Chonnam National University Hwasun Hospital, Hwasun, Korea
| | - Hyun Jeong Shim
- Department of Hemato-Oncology, Chonnam National University Hwasun Hospital, Hwasun, Korea
| | - Jun Eul Hwang
- Department of Hemato-Oncology, Chonnam National University Hwasun Hospital, Hwasun, Korea
| | - Woo Kyun Bae
- Department of Hemato-Oncology, Chonnam National University Hwasun Hospital, Hwasun, Korea
| | - Ik Joo Chung
- Department of Hemato-Oncology, Chonnam National University Hwasun Hospital, Hwasun, Korea
| |
Collapse
|
24
|
Chen L, Lei Z, Ma X, Huang Q, Zhang X, Zhang Y, Hao P, Yang M, Zhao X, Chen J, Liu G, Zheng T. Prognostic significance of fibroblast growth factor receptor 4 polymorphisms on biochemical recurrence after radical prostatectomy in a Chinese population. Sci Rep 2016; 6:33604. [PMID: 27640814 PMCID: PMC5027536 DOI: 10.1038/srep33604] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2016] [Accepted: 08/30/2016] [Indexed: 12/16/2022] Open
Abstract
Fibroblast growth factor receptor 4 (FGFR4) is a transmembrane receptor with ligand-induced tyrosine kinase activity and is involved in various biological and pathological processes. Several polymorphisms of FGFR4 are associated with the incidence and mortality of numerous cancers, including prostate cancer. In this study, we investigated whether the polymorphisms of FGFR4 influence the biochemical recurrence of prostate cancer in Chinese men after radical prostatectomy. Three common polymorphisms (rs1966265, rs2011077, and rs351855) of FGFR4 were genotyped from 346 patients with prostate cancer by using the Sequenom MassARRAY system. Kaplan–Meier curves and Cox proportional hazard models were used for survival analysis. Results showed biochemical recurrence (BCR) free survival was significantly affected by the genotypes of rs351855 but not influenced by rs1966265 and rs2011077. After adjusting for other variables in multivariable analysis, patients with rs351855 AA/AG genotypes showed significantly worse BCR-free survival than those with the GG genotype (HR = 1.873; 95% CI, 1.209–2.901; P = 0.005). Hence, FGFR4 rs351855 could be a novel independent prognostic factor of BCR after radical prostatectomy in the Chinese population. This functional polymorphism may also provide a basis for surveillance programs. Additional large-scale studies must be performed to validate the significance of this polymorphism in prostate cancer.
Collapse
Affiliation(s)
- Luyao Chen
- Department of Urology, Chinese PLA General Hospital, Beijing, China
| | - Zhengwei Lei
- Department of Urology, Chinese PLA General Hospital, Beijing, China
| | - Xin Ma
- Department of Urology, Chinese PLA General Hospital, Beijing, China
| | - Qingbo Huang
- Department of Urology, Chinese PLA General Hospital, Beijing, China
| | - Xu Zhang
- Department of Urology, Chinese PLA General Hospital, Beijing, China
| | - Yong Zhang
- Department of Urology, Puai Hospital, Wuhan, China
| | - Peng Hao
- Department of Urology, Puai Hospital, Wuhan, China
| | | | - Xuetao Zhao
- Department of Urology, Puai Hospital, Wuhan, China
| | - Jun Chen
- Department of Urology, Puai Hospital, Wuhan, China
| | - Gongxue Liu
- Department of Urology, Puai Hospital, Wuhan, China
| | - Tao Zheng
- Department of Urology, Puai Hospital, Wuhan, China
| |
Collapse
|
25
|
Telomere Transcripts Target Telomerase in Human Cancer Cells. Genes (Basel) 2016; 7:genes7080046. [PMID: 27537914 PMCID: PMC4999834 DOI: 10.3390/genes7080046] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2016] [Revised: 07/24/2016] [Accepted: 07/29/2016] [Indexed: 12/14/2022] Open
Abstract
Long non-coding transcripts from telomeres, called telomeric repeat-containing RNA (TERRA), were identified as blocking telomerase activity (TA), a telomere maintenance mechanism (TMM), in tumors. We expressed recombinant TERRA transcripts in tumor cell lines with TA and with alternative lengthening of telomeres (ALT) to study effects on TMM and cell growth. Adeno- and lentivirus constructs (AV and LV) were established for transient and stable expression of approximately 130 units of telomere hexanucleotide repeats under control of cytomegalovirus (CMV) and human RNase P RNA H1 (hH1) promoters with and without polyadenylation, respectively. Six human tumor cell lines either using telomerase or ALT were infected and analyzed for TA levels. Pre-infection cells using telomerase had 1%-3% of the TERRA expression levels of ALT cells. AV and LV expression of recombinant TERRA in telomerase positive cells showed a 1.3-2.6 fold increase in TERRA levels, and a decrease in TA of 25%-58%. Dominant-negative or small hairpin RNA (shRNA) viral expression against human telomerase reverse transcriptase (hTERT) results in senescence, not induced by TERRA expression. Population doubling time, cell viability and TL (telomere length) were not impacted by ectopic TERRA expression. Clonal growth was reduced by TERRA expression in TA but not ALT cell lines. ALT cells were not affected by treatments applied. Established cell models and tools may be used to better understand the role of TERRA in the cell, especially for targeting telomerase.
Collapse
|
26
|
Kurniawan NA, Chaudhuri PK, Lim CT. Mechanobiology of cell migration in the context of dynamic two-way cell-matrix interactions. J Biomech 2015; 49:1355-1368. [PMID: 26747513 DOI: 10.1016/j.jbiomech.2015.12.023] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2015] [Revised: 11/30/2015] [Accepted: 12/14/2015] [Indexed: 12/31/2022]
Abstract
Migration of cells is integral in various physiological processes in all facets of life. These range from embryonic development, morphogenesis, and wound healing, to disease pathology such as cancer metastasis. While cell migratory behavior has been traditionally studied using simple assays on culture dishes, in recent years it has been increasingly realized that the physical, mechanical, and chemical aspects of the matrix are key determinants of the migration mechanism. In this paper, we will describe the mechanobiological changes that accompany the dynamic cell-matrix interactions during cell migration. Furthermore, we will review what is to date known about how these changes feed back to the dynamics and biomechanical properties of the cell and the matrix. Elucidating the role of these intimate cell-matrix interactions will provide not only a better multi-scale understanding of cell motility in its physiological context, but also a more holistic perspective for designing approaches to regulate cell behavior.
Collapse
Affiliation(s)
- Nicholas A Kurniawan
- Department of Biomedical Engineering, Eindhoven University of Technology, P.O. Box 513, 5600MB Eindhoven, The Netherlands; Department of Systems Biophysics, FOM Institute AMOLF, Amsterdam, The Netherlands.
| | | | - Chwee Teck Lim
- Mechanobiology Institute, National University of Singapore, Singapore; Department of Biomedical Engineering, National University of Singapore, Singapore.
| |
Collapse
|
27
|
Ulaganathan VK, Sperl B, Rapp UR, Ullrich A. Germline variant FGFR4 p.G388R exposes a membrane-proximal STAT3 binding site. Nature 2015; 528:570-4. [PMID: 26675719 DOI: 10.1038/nature16449] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2015] [Accepted: 11/14/2015] [Indexed: 12/27/2022]
Abstract
Variant rs351855-G/A is a commonly occurring single-nucleotide polymorphism of coding regions in exon 9 of the fibroblast growth factor receptor FGFR4 (CD334) gene (c.1162G>A). It results in an amino-acid change at codon 388 from glycine to arginine (p.Gly388Arg) in the transmembrane domain of the receptor. Despite compelling genetic evidence for the association of this common variant with cancers of the bone, breast, colon, prostate, skin, lung, head and neck, as well as soft-tissue sarcomas and non-Hodgkin lymphoma, the underlying biological mechanism has remained elusive. Here we show that substitution of the conserved glycine 388 residue to a charged arginine residue alters the transmembrane spanning segment and exposes a membrane-proximal cytoplasmic signal transducer and activator of transcription 3 (STAT3) binding site Y(390)-(P)XXQ(393). We demonstrate that such membrane-proximal STAT3 binding motifs in the germline of type I membrane receptors enhance STAT3 tyrosine phosphorylation by recruiting STAT3 proteins to the inner cell membrane. Remarkably, such germline variants frequently co-localize with somatic mutations in the Catalogue of Somatic Mutations in Cancer (COSMIC) database. Using Fgfr4 single nucleotide polymorphism knock-in mice and transgenic mouse models for breast and lung cancers, we validate the enhanced STAT3 signalling induced by the FGFR4 Arg388-variant in vivo. Thus, our findings elucidate the molecular mechanism behind the genetic association of rs351855 with accelerated cancer progression and suggest that germline variants of cell-surface molecules that recruit STAT3 to the inner cell membrane are a significant risk for cancer prognosis and disease progression.
Collapse
Affiliation(s)
- Vijay K Ulaganathan
- Max Planck Institute for Biochemistry, Department of Molecular Biology, Am Klopferspitz 18, 82152, Martinsried. Germany
| | - Bianca Sperl
- Max Planck Institute for Biochemistry, Department of Molecular Biology, Am Klopferspitz 18, 82152, Martinsried. Germany
| | - Ulf R Rapp
- Max Planck Institute for Heart and Lung Research, Molecular Mechanisms of Lung Cancer, Parkstrasse 1, 61231 Bad Nauheim, Germany
| | - Axel Ullrich
- Max Planck Institute for Biochemistry, Department of Molecular Biology, Am Klopferspitz 18, 82152, Martinsried. Germany
| |
Collapse
|
28
|
Liu R, Huang S, Lei Y, Zhang T, Wang K, Liu B, Nice EC, Xiang R, Xie K, Li J, Huang C. FGF8 promotes colorectal cancer growth and metastasis by activating YAP1. Oncotarget 2015; 6:935-52. [PMID: 25473897 PMCID: PMC4359266 DOI: 10.18632/oncotarget.2822] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2014] [Accepted: 11/25/2014] [Indexed: 02/05/2023] Open
Abstract
Colorectal cancer (CRC) is a major cause of cancer-related death worldwide. The poor prognosis of CRC is mainly due to uncontrolled tumor growth and distant metastases. In this study, we found that the level of FGF8 was elevated in the great majority of CRC cases and high FGF8 expression was significantly correlated with lymph nodes metastasis and worse overall survival. Functional studies showed that FGF8 can induce a more aggressive phenotype displaying epithelial-to-mesenchymal transition (EMT) and enhanced invasion and growth in CRC cells. Consistent with this, FGF8 can also promote tumor growth and metastasis in mouse models. Bioinformatics and pathological analysis suggested that YAP1 is a potential downstream target of FGF8 in CRC cells. Molecular validation demonstrated that FGF8 fully induced nuclear localization of YAP1 and enhanced transcriptional outcomes such as the expression of CTGF and CYR61, while decreasing YAP1 expression impeded FGF-8–induced cell growth, EMT, migration and invasion, revealing that YAP1 is required for FGF8-mediated CRC growth and metastasis. Taken together, these results demonstrate that FGF8 contributes to the proliferative and metastatic capacity of CRC cells and may represent a novel candidate for intervention in tumor growth and metastasis formation.
Collapse
Affiliation(s)
- Rui Liu
- State Key Laboratory of Biotherapy/Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu, P. R. China.,State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, P. R. China
| | - Shan Huang
- State Key Laboratory of Biotherapy/Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu, P. R. China
| | - Yunlong Lei
- Department of Biochemistry and Molecular Biology, and Molecular Medicine and Cancer Research Center, Chongqing Medical University, Chongqing, P. R. China
| | - Tao Zhang
- The School of Biomedical Sciences, Chengdu Medical College, Chengdu, P. R. China
| | - Kui Wang
- State Key Laboratory of Biotherapy/Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu, P. R. China
| | - Bo Liu
- State Key Laboratory of Biotherapy/Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu, P. R. China
| | - Edouard C Nice
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, Victoria, Australia
| | - Rong Xiang
- School of Medicine, Nankai University, Tianjin, P.R. China
| | - Ke Xie
- Department of Oncology, Sichuan Provincial People's Hospital, Chengdu, P. R. China
| | - Jingyi Li
- The School of Biomedical Sciences, Chengdu Medical College, Chengdu, P. R. China
| | - Canhua Huang
- State Key Laboratory of Biotherapy/Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu, P. R. China
| |
Collapse
|
29
|
Heinzle C, Erdem Z, Paur J, Grasl-Kraupp B, Holzmann K, Grusch M, Berger W, Marian B. Is fibroblast growth factor receptor 4 a suitable target of cancer therapy? Curr Pharm Des 2015; 20:2881-98. [PMID: 23944363 DOI: 10.2174/13816128113199990594] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2013] [Accepted: 08/06/2013] [Indexed: 12/17/2022]
Abstract
Fibroblast growth factors (FGF) and their tyrosine kinase receptors (FGFR) support cell proliferation, survival and migration during embryonic development, organogenesis and tissue maintenance and their deregulation is frequently observed in cancer development and progression. Consequently, increasing efforts are focusing on the development of strategies to target FGF/FGFR signaling for cancer therapy. Among the FGFRs the family member FGFR4 is least well understood and differs from FGFRs1-3 in several aspects. Importantly, FGFR4 deletion does not lead to an embryonic lethal phenotype suggesting the possibility that its inhibition in cancer therapy might not cause grave adverse effects. In addition, the FGFR4 kinase domain differs sufficiently from those of FGFRs1-3 to permit development of highly specific inhibitors. The oncogenic impact of FGFR4, however, is not undisputed, as the FGFR4-mediated hormonal effects of several FGF ligands may also constitute a tissue-protective tumor suppressor activity especially in the liver. Therefore it is the purpose of this review to summarize all relevant aspects of FGFR4 physiology and pathophysiology and discuss the options of targeting this receptor for cancer therapy.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Brigitte Marian
- Institute of Cancer Research, Department of Medicine 1, Medical University Vienna, Borschkegasse 8a, 1090 Vienna, Austria.
| |
Collapse
|
30
|
Shim HJ, Shin MH, Kim HN, Kim JH, Hwang JE, Bae WK, Chung IJ, Cho SH. The Prognostic Significance of FGFR4 Gly388 Polymorphism in Esophageal Squamous Cell Carcinoma after Concurrent Chemoradiotherapy. Cancer Res Treat 2015; 48:71-9. [PMID: 25989802 PMCID: PMC4720088 DOI: 10.4143/crt.2015.018] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2015] [Accepted: 04/06/2015] [Indexed: 02/06/2023] Open
Abstract
Purpose The purpose of this study is to investigate the role of fibroblast growth factor receptor 4 (FGFR4) polymorphism in esophageal cancer after chemoradiotherapy (CRT). Materials and Methods Peripheral blood samples from 244 patients treated with CRT for esophageal squamous cell carcinoma were assessed for the role of FGFR4 genotype on treatment response and survival. Results A total of 94 patients were homozygous for the Gly388 allele, and 110 were heterozygous and 40 homozygous for the Arg388 allele. No significant association was found between the FGFR4 genotype and clinicopathological parameters. However, patients carrying the Gly388 allele showed a better overall response rate than Arg388 carriers (p=0.038). In addition, Gly388 allele patients at an earlier stage showed better overall survival (OS) and progression-free survival than Arg388 carriers. Among these, the Gly388 allele showed significantly improved OS compared to Arg388 carriers in the lymph node (LN) metastasis group (p=0.042) compared to the no LN metastasis group (p=0.125). However, similar survival outcomes were observed for advanced-stage disease regardless of genotype. Conclusion This result suggests that the role of FGFR4 Gly388 in treatment outcomes differs according to esophageal cancer stage. It showed a predictive role in the response of esophageal cancer patients to CRT with a better trend for OS in Gly388 than Arg388 carriers in the early stages. In particular, LN-positive early-stage patients carrying the Gly388 allele showed improved OS compared to those carrying Arg388.
Collapse
Affiliation(s)
- Hyun-Jeong Shim
- Department of Hemato-Oncology, Chonnam National University Medical School, Hwasun, Korea
| | - Min-Ho Shin
- Department of Preventive Medicine, Chonnam National University Medical School, Hwasun, Korea ; Department of Jeonnam Regional Cancer Center, Chonnam National University Medical School, Hwasun, Korea
| | - Hee-Nam Kim
- Department of Jeonnam Regional Cancer Center, Chonnam National University Medical School, Hwasun, Korea
| | - Jo-Heon Kim
- Department of Pathology, Chonnam National University Medical School, Hwasun, Korea
| | - Jun-Eul Hwang
- Department of Hemato-Oncology, Chonnam National University Medical School, Hwasun, Korea
| | - Woo-Kyun Bae
- Department of Hemato-Oncology, Chonnam National University Medical School, Hwasun, Korea
| | - Ik-Joo Chung
- Department of Hemato-Oncology, Chonnam National University Medical School, Hwasun, Korea
| | - Sang-Hee Cho
- Department of Hemato-Oncology, Chonnam National University Medical School, Hwasun, Korea
| |
Collapse
|
31
|
Ahmad A, Askari S, Befekadu R, Hahn-Strömberg V. Investigating the association between polymorphisms in connective tissue growth factor and susceptibility to colon carcinoma. Mol Med Rep 2014; 11:2493-503. [PMID: 25502877 PMCID: PMC4337474 DOI: 10.3892/mmr.2014.3083] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2014] [Accepted: 07/25/2014] [Indexed: 01/01/2023] Open
Abstract
There have been numerous studies on the gene expression of connective tissue growth factor (CTGF) in colorectal cancer, however very few have investigated polymorphisms in this gene. The present study aimed to determine whether single nucleotide polymorphisms (SNPs) in the CTGF gene are associated with a higher susceptibility to colon cancer and/or an invasive tumor growth pattern. The CTGF gene was genotyped for seven SNPs (rs6918698, rs1931002, rs9493150, rs12526196, rs12527705, rs9399005 and rs12527379) by pyrosequencing. Formalin-fixed paraffin-embedded tissue samples (n=112) from patients diagnosed with colon carcinoma, and an equal number of blood samples from healthy controls, were selected for genomic DNA extraction. The complexity index was measured using images of tumor samples (n=64) stained for cytokeratin-8. The images were analyzed and correlated with the identified CTGF SNPs and clinicopathological parameters of the patients, including age, gender, tumor penetration, lymph node metastasis, systemic metastasis, differentiation and localization of tumor. It was demonstrated that the frequency of the SNP rs6918698 GG genotype was significantly associated (P=0.05) with an increased risk of colon cancer, as compared with the GC and CC genotypes. The other six SNPs (rs1931002, rs9493150, rs12526196, rs12527705, rs9399005 and rs12527379) exhibited no significant difference in the genotype and allele frequencies between patients diagnosed with colon carcinoma and the normal healthy population. A trend was observed between genotype variation at rs6918698 and the complexity index (P=0.052). The complexity index and genotypes for any of the studied SNPs were not significantly correlated with clinical or pathological parameters of the patients. These results indicate that the rs6918698 GG genotype is associated with an increased risk of developing colon carcinoma, and genetic variations at the rs6918698 are associated with the growth pattern of the tumor. The present results may facilitate the identification of potential biomarkers of the disease in addition to drug targets.
Collapse
Affiliation(s)
- Abrar Ahmad
- Department of Clinical Medicine, Örebro University, Örebro 701 81, Sweden
| | - Shlear Askari
- Department of Clinical Medicine, Örebro University, Örebro 701 81, Sweden
| | - Rahel Befekadu
- Department of Laboratory Medicine, Section for Transfusion Medicine, Örebro University Hospital, Örebro 701 85, Sweden
| | | |
Collapse
|
32
|
Lötsch D, Steiner E, Holzmann K, Spiegl-Kreinecker S, Pirker C, Hlavaty J, Petznek H, Hegedus B, Garay T, Mohr T, Sommergruber W, Grusch M, Berger W. Major vault protein supports glioblastoma survival and migration by upregulating the EGFR/PI3K signalling axis. Oncotarget 2014; 4:1904-18. [PMID: 24243798 PMCID: PMC3875758 DOI: 10.18632/oncotarget.1264] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Despite their ubiquitous expression and high conservation during evolution, precise cellular functions of vault ribonucleoparticles, mainly built of multiple major vault protein (MVP) copies, are still enigmatic. With regard to cancer, vaults were shown to be upregulated during drug resistance development as well as malignant transformation and progression. Such in a previous study we demonstrated that human astrocytic brain tumours including glioblastoma are generally high in vault levels while MVP expression in normal brain is comparably low. However a direct contribution to the malignant phenotype in general and that of glioblastoma in particular has not been established so far. Thus we address the questions whether MVP itself has a pro-tumorigenic function in glioblastoma. Based on a large tissue collection, we re-confirm strong MVP expression in gliomas as compared to healthy brain. Further, the impact of MVP on human glioblastoma aggressiveness was analysed by using gene transfection, siRNA knock-down and dominant-negative genetic approaches. Our results demonstrate that MVP/vaults significantly support migratory and invasive competence as well as starvation resistance of glioma cells in vitro and in vivo. The enhanced aggressiveness was based on MVP-mediated stabilization of the epidermal growth factor receptor (EGFR)/phosphatidyl-inositol-3-kinase (PI3K) signalling axis. Consequently, MVP overexpression resulted in enhanced growth and brain invasion in human glioblastoma xenograft models. Our study demonstrates, for the first time, that vaults have a tumour-promoting potential by stabilizing EGFR/PI3K-mediated migration and survival pathways in human glioblastoma.
Collapse
|
33
|
Haugsten EM, Brech A, Liestøl K, Norman JC, Wesche J. Photoactivation approaches reveal a role for Rab11 in FGFR4 recycling and signalling. Traffic 2014; 15:665-83. [PMID: 24589086 DOI: 10.1111/tra.12168] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2013] [Revised: 02/25/2014] [Accepted: 03/03/2014] [Indexed: 12/24/2022]
Abstract
Fibroblast growth factor receptor 4 (FGFR4) plays important roles during development and in the adult to maintain tissue homeostasis. Moreover, overexpression of FGFR4 or activating mutations in FGFR4 has been identified as tumour-promoting events in several forms of cancer. Endocytosis is important for regulation of signalling receptors and we have previously shown that FGFR4 is mainly localized to transferrin-positive structures after ligand-induced endocytosis. Here, using a cell line with a defined pericentriolar endocytic recycling compartment, we show that FGFR4 accumulates in this compartment after endocytosis. Furthermore, using classical recycling assays and a new, photoactivatable FGFR4-PA-GFP fusion protein combined with live-cell imaging, we demonstrate that recycling of FGFR4 is dependent on Rab11. Upon Rab11b depletion, FGFR4 is trapped in the pericentriolar recycling compartment and the total levels of FGFR4 in cells are increased. Moreover, fibroblast growth factor 1 (FGF1)-induced autophosphorylation of FGFR4 as well as phosphorylation of phospholipase C (PLC)-γ is prolonged in cells depleted of Rab11. Interestingly, the activation of mitogen-activated protein kinase and AKT pathways were not prolonged but rather reduced in Rab11-depleted cells, indicating that recycling of FGFR4 is important for the nature of its signalling output. Thus, Rab11-dependent recycling of FGFR4 maintains proper levels of FGFR4 in cells and regulates FGF1-induced FGFR4 signalling.
Collapse
Affiliation(s)
- Ellen M Haugsten
- Department of Biochemistry, Institute for Cancer Research, The Norwegian Radium Hospital, Oslo University Hospital, Montebello, Oslo, 0379, Norway; Centre for Cancer Biomedicine, Faculty of Medicine, University of Oslo, Montebello, Oslo, 0379, Norway
| | | | | | | | | |
Collapse
|
34
|
Schweiger N, Hauck M, Steinhoff H, Sampl S, Reifinger M, Walter I, Kreilmeier T, Marian B, Grusch M, Berger W, Holzmann K, Kleiter M. Canine and human sarcomas exhibit predominant FGFR1 expression and impaired viability after inhibition of signaling. Mol Carcinog 2014; 54:841-52. [PMID: 24719266 DOI: 10.1002/mc.22155] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2013] [Revised: 03/18/2014] [Accepted: 03/18/2014] [Indexed: 02/03/2023]
Abstract
Fibroblast growth factor receptors (FGFRs) are important in malignant progression of several human epithelial tumors. However, little is known about FGFRs in canine or human soft tissue sarcomas. Thus, our aim was to investigate expression of FGFRs and their involvement in cell survival in sarcomas of both species. FGFR1-4 and FGFRL1 transcripts as well as IIIb/IIIc splice variants of FGFR1-3 were evaluated in 3 canine- and 6 human sarcoma cell lines and 19 spontaneous canine sarcomas by SYBRqPCR. FGFR1 protein expression was assessed by immunohistochemistry. Growth inhibitory effects of FGFR1 inhibitor PD166866 and dominant negative recombinant FGFR adenoviral expression constructs (dnFGFR) on tumor cell lines were analyzed. Profiling of multiple FGFR transcripts detected comparable co-expression in most of human and canine sarcoma cell lines and canine tumor specimens. This indicates existence of closely related regulation mechanisms for FGFR expression in sarcomas of both species. FGFR1 with splice variant IIIc was consistently expressed with highest transcript levels. In 88% of the spontaneous tumor samples a heterogeneous FGFR1 protein expression was observed. Significant growth inhibition and cell death was seen after infection with dnFGFR1 in canine and human sarcoma cells, but not with dnFGFR3 and 4. PD166866 showed selective cytotoxicity with IC50 values between 12.1 and 26.4 μM. FGFR1 inhibition blocked ligand-induced tyrosine phosphorylation of ERK1/2 mitogen-activated protein kinase isoforms. This study emphasizes the important role FGFR1, especially splice variant IIIc, likely plays in sarcomas. Inhibitory small molecules could be of potential use for targeted therapy in aggressive sarcomas of both species.
Collapse
Affiliation(s)
- Nicole Schweiger
- Department for Companion Animals and Horses, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Marlene Hauck
- Department of Medicine I, Division of Cancer Research, Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria.,Department of Clinical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, NC
| | - Heinrich Steinhoff
- Department of Medicine I, Division of Cancer Research, Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
| | - Sandra Sampl
- Department of Medicine I, Division of Cancer Research, Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
| | - Martin Reifinger
- Department of Pathobiology, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Ingrid Walter
- Vet Core Facility, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Theresa Kreilmeier
- Department for Companion Animals and Horses, University of Veterinary Medicine Vienna, Vienna, Austria.,Department of Medicine I, Division of Cancer Research, Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
| | - Brigitte Marian
- Department of Medicine I, Division of Cancer Research, Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
| | - Michael Grusch
- Department of Medicine I, Division of Cancer Research, Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
| | - Walter Berger
- Department of Medicine I, Division of Cancer Research, Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
| | - Klaus Holzmann
- Department of Medicine I, Division of Cancer Research, Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
| | - Miriam Kleiter
- Department for Companion Animals and Horses, University of Veterinary Medicine Vienna, Vienna, Austria
| |
Collapse
|
35
|
Cheng S, Al-Agha R, Araujo PB, Serri O, L. Asa S, Ezzat S. Metabolic glucose status and pituitary pathology portend therapeutic outcomes in acromegaly. PLoS One 2013; 8:e73543. [PMID: 24039977 PMCID: PMC3767813 DOI: 10.1371/journal.pone.0073543] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2013] [Accepted: 07/19/2013] [Indexed: 11/19/2022] Open
Abstract
INTRODUCTION Acromegaly is frequently associated with impaired glucose tolerance and/or diabetes. To evaluate the relationship between glucose metabolism and acromegaly disease, we evaluated 269 consecutive patients from two referral centres. METHODS Clinical presentation, pituitary tumor size and invasiveness, and pituitary pathology were captured in a dedicated database. RESULTS 131 women and 138 men with a mean age of 53.8 years were included. Of these, 201 (74.7%) presented with a macroadenoma and 18 (6.7%) with a microadenoma. Radiographic invasion was present in 91 cases (33.8%). Mean tumor diameter was 1.86 cm (0.2-4.6). Pituitary histopathologic findings revealed pure GH-producing somatotroph adenomas (SA) in 147 patients, prolactin-production by mixed lactotroph (LA) and SA or mammosomatotroph adenoma (MSA) in 46 [22.4%], acidophil stem cell adenoma in 6 [2.9%], and other diagnoses in 6 [2.9%]. Medical treatment included octreotide in 96 [36.9%] and in combination with pegvisomant or dopamine agonists in 63 [24.2%]. Nearly 80% of patients achieved IGF-1 normalization. Importantly, patients with pure somatotroph adenomas were significantly more likely to present with abnormal glucose metabolism [48.7%] than those with mixed adenomas [9.7%] [p<0.001] independent of GH/IGF-1 levels or tumor invasiveness. Abnormal glucose metabolism and pituitary pathology also remained linked following IGF-1 normalization. Moreover patients with pure SA and abnormal glucose metabolism were significantly (p<0.001) less likely to achieve disease remission despite the same therapeutic strategies. Conversely, patients with mixed adenomas were more likely (OR: 2.766 (95% CI: 1.490-5.136) to achieve disease remission. CONCLUSIONS Patients with pure somatotroph adenomas are more likely than those with mixed adenomas to exhibit abnormal glucose metabolism.
Collapse
Affiliation(s)
- Sonia Cheng
- Department of Medicine, University Health Network, Toronto, Ontario, Canada
| | - Rany Al-Agha
- Department of Medicine, University Health Network, Toronto, Ontario, Canada
| | - Paula B. Araujo
- Department of Medicine, University Health Network, Toronto, Ontario, Canada
| | - Omar Serri
- Department of Medicine, University of Montreal, Montreal, Canada
| | - Sylvia L. Asa
- Department of Pathology, University Health Network, Toronto, Ontario, Canada
| | - Shereen Ezzat
- Department of Medicine, University Health Network, Toronto, Ontario, Canada
- * E-mail:
| |
Collapse
|
36
|
Peláez-García A, Barderas R, Torres S, Hernández-Varas P, Teixidó J, Bonilla F, de Herreros AG, Casal JI. FGFR4 role in epithelial-mesenchymal transition and its therapeutic value in colorectal cancer. PLoS One 2013; 8:e63695. [PMID: 23696849 PMCID: PMC3655941 DOI: 10.1371/journal.pone.0063695] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2012] [Accepted: 04/06/2013] [Indexed: 12/16/2022] Open
Abstract
Fibroblast growth factor receptor 4 (FGFR4) is vital in early development and tissue repair. FGFR4 expression levels are very restricted in adult tissues, except in several solid tumors including colorectal cancer, which showed overexpression of FGFR4. Here, FGFR4 mutation analysis discarded the presence of activating mutations, other than Arg(388), in different colorectal cancer cell lines and tumoral samples. Stable shRNA FGFR4-silencing in SW480 and SW48 cell lines resulted in a significant decrease in cell proliferation, adhesion, cell migration and invasion. This decrease in the tumorigenic and invasive capabilities of colorectal cancer cells was accompanied by a decrease of Snail, Twist and TGFβ gene expression levels and an increase of E-cadherin, causing a reversion to a more epithelial phenotype, in three different cell lines. In addition, FGFR4-signaling activated the oncogenic SRC, ERK1/2 and AKT pathways in colon cancer cells and promoted an increase in cell survival. The relevance of FGFR4 in tumor growth was supported by two different strategies. Kinase inhibitors abrogated FGFR4-related cell growth and signaling pathways at the same extent than FGFR4-silenced cells. Specific FGFR4-targeting using antibodies provoked a similar reduction in cell growth. Moreover, FGFR4 knock-down cells displayed a reduced capacity for in vivo tumor formation and angiogenesis in nude mice. Collectively, our data support a crucial role for FGFR4 in tumorigenesis, invasion and survival in colorectal cancer. In addition, FGFR4 targeting demonstrated its applicability for colorectal cancer therapy.
Collapse
Affiliation(s)
- Alberto Peláez-García
- Department of Celular and Molecular Medicine, Centro de Investigaciones Biológicas (CIB-CSIC), Madrid, Spain
| | - Rodrigo Barderas
- Department of Celular and Molecular Medicine, Centro de Investigaciones Biológicas (CIB-CSIC), Madrid, Spain
| | - Sofía Torres
- Department of Celular and Molecular Medicine, Centro de Investigaciones Biológicas (CIB-CSIC), Madrid, Spain
| | | | - Joaquín Teixidó
- Chemokines and Cell Migration Laboratory, CIB-CSIC, Madrid, Spain
| | - Félix Bonilla
- Hospital Puerta de Hierro Majadahonda, Madrid, Spain
| | | | - J. Ignacio Casal
- Department of Celular and Molecular Medicine, Centro de Investigaciones Biológicas (CIB-CSIC), Madrid, Spain
- * E-mail:
| |
Collapse
|
37
|
Microarray analyses reveal liver metastasis-related genes in metastatic colorectal cancer cell model. J Cancer Res Clin Oncol 2013; 139:1169-78. [PMID: 23563852 DOI: 10.1007/s00432-013-1424-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2012] [Accepted: 03/22/2013] [Indexed: 02/04/2023]
Abstract
PURPOSE To study the molecular mechanisms of colorectal cancer liver metastasis. METHODS Cecal wall implantation was performed in nude mice to subclone a highly liver metastatic human colorectal cancer clone (SW1116-M) from SW1116. In vivo and in vitro assays were adopted to confirm the proliferation and metastasis potential. The human tumor metastasis PCR microarrays were used to analyze the differential gene expressions. The results were confirmed further by real-time quantitative PCR. RESULTS SW1116-M and SW1116-S5, two human colon cancer cell clones with different metastatic potential, were subcloned from SW1116. In SW1116-M, in vitro invasion, migration and in vivo metastatic potential were higher, and in vitro proliferation rate was lower than SW1116-S5. In tumor metastasis PCR microarray, 24 genes related to cell invading, adhesion, cellular growth and differentiation were found with a twofold difference between SW1116-S5 and SW1116-M. Sixteen of these, including E-cadherins, MTSS1, TRAIL and TRPM1, were up-regulated; eight genes including cathepsin L, EphB2, HGF, MET, MCAM and RORβ were down-regulated. CONCLUSIONS We have established a highly liver metastatic clone. The subsequent metastasis PCR microarray analysis identified a procedure of cellular differentiation and mesenchymal to epithelial transition (MET) in liver metastasis. The colonization to from macrometastasis is not a switch from cell cycle arrest but a result of cell differentiation and MET.
Collapse
|
38
|
The role of endosomal signaling triggered by metastatic growth factors in tumor progression. Cell Signal 2013; 25:1539-45. [PMID: 23571269 DOI: 10.1016/j.cellsig.2013.03.022] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2013] [Accepted: 03/28/2013] [Indexed: 01/12/2023]
Abstract
Within tumor microenvironment, a lot of growth factors such as hepatocyte growth factor and epidermal growth factor may induce similar signal cascade downstream of receptor tyrosine kinase (RTK) and trigger tumor metastasis synergistically. In the past decades, the intimate relationship of RTK-mediated receptor endocytosis with signal transduction was well established. In general, most RTK undergoes clathrin-dependent endocytosis and/or clathrin-independent endocytosis. The internalized receptors may sustain the signaling within early endosome, recycling to plasma membrane for subsequent ligand engagement or sorting to late endosomes/lysosome for receptor degradation. Moreover, receptor endocytosis influences signal transduction in a temporal and spatial manner for periodical and polarized cellular processes such as cell migration. The endosomal signalings triggered by various metastatic factors are quite similar in some critical points, which are essential for triggering cell migration and tumor progression. There are common regulators for receptor endocytosis including dynamin, Rab4, Rab5, Rab11 and Cbl. Moreover, many critical regulators within the RTK signal pathway such as Grb2, p38, PKC and Src were also modulators of endocytosis. In the future, these may constitute a new category of targets for prevention of tumor metastasis.
Collapse
|
39
|
Fang HM, Tian G, Zhou LJ, Zhou HY, Fang YZ. FGFR4 genetic polymorphisms determine the chemotherapy response of Chinese patients with non-small cell lung cancer. Acta Pharmacol Sin 2013; 34:549-54. [PMID: 23524567 DOI: 10.1038/aps.2012.206] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
AIM To investigate the relationship of fibroblast growth factor receptor 4 (FGFR4) gene polymorphisms with the response of Chinese patients with non-small cell lung cancer (NSCLC) to chemotherapy. METHODS A total of 629 patients with Stage III (A+B) or IV NSCLC, as well as 729 age- and gender-matched healthy controls were recruited. All the patients received platinum-based chemotherapy, and the therapeutic effects were evaluated. Three polymorphisms in the FGFR4 gene (rs351855G/A, rs145302848C/G, and rs147603016G/A) were genotyped, and the association between the 3 polymorphisms and the chemotherapy effect was analyzed using SPSS software, version 16.0. RESULTS The genotype frequencies of rs145302848C/G and rs147603016G/A were not significantly different between NSCLC patients and healthy controls on one hand, and between the responders and non-responders to the chemotherapy on the other hand. The distribution of AA genotype and A-allele of rs351855G/A was significantly lower in NSCLC patients than in healthy controls. Using patients with the GG genotype as a reference, the AA carrier had a significantly reduced risk for the development of NSCLC after normalizing to age, sex and smoking habits. In NSCLC patients, this genotype occurred more frequently in the responders to the chemotherapy than in non-responders. The chance of being a responder was significantly increased with the AA genotype as compared to G genotype. The AA genotype of rs351855G/A had a better prognosis compared with GA and GG genotype carriers: the overall survival of patients with the AA genotype of rs351855G/A was significantly longer than those with the GG+GA genotype (21.1 vs 16.5 months). CONCLUSION The rs351855G/A polymorphisms of FGFR4 gene can be used to predict the occurrence, chemotherapy response and prognosis of NSCLC.
Collapse
|