1
|
Bouhniz OE, Kenani A. Potential role of genetic polymorphisms in neoadjuvant chemotherapy response in breast cancer. J Chemother 2024:1-15. [PMID: 38511398 DOI: 10.1080/1120009x.2024.2330241] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Accepted: 03/11/2024] [Indexed: 03/22/2024]
Abstract
Chemoresistance leads to treatment failure, which can arise through different mechanisms including patients' characteristics. Searching for genetic profiles as a predictor for drug response and toxicity has been extensively studied in pharmacogenomics, thus contributing to personalized medicine and providing alternative treatments. Numerous studies have demonstrated significant evidence of association between genetic polymorphisms and response to neoadjuvant chemotherapy (NAC) in breast cancer. In this review, we explored the potential impact of genetic polymorphisms in NAC primary resistance through selecting a specific clinical profile. The genetic variability within pharmacokinetics, pharmacodynamics, DNA synthesis and repair, and oncogenic signaling pathways genes could be predictive or prognostic markers for NAC resistance. The clinical implication of these results can help provide individualized treatment plans in the early stages of breast cancer treatment. Further studies are needed to determine the genetic hosts of primary chemoresistance mechanisms in order to further emphasize the implementation of genotypic approaches in personalized medicine.
Collapse
Affiliation(s)
- Om Elez Bouhniz
- Research Laboratory "Environment, Inflammation, Signaling and Pathologies" (LR18ES40), Faculty of Medicine of Monastir, University of Monastir, Monastir, Tunisia
| | - Abderraouf Kenani
- Research Laboratory "Environment, Inflammation, Signaling and Pathologies" (LR18ES40), Faculty of Medicine of Monastir, University of Monastir, Monastir, Tunisia
| |
Collapse
|
2
|
Mendivelso González DF, Sánchez Villalobos SA, Ramos AE, Montero Ovalle WJ, Serrano López ML. Single Nucleotide Polymorphisms Associated with Prostate Cancer Progression: A Systematic Review. Cancer Invest 2024; 42:75-96. [PMID: 38055319 DOI: 10.1080/07357907.2023.2291776] [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: 08/01/2023] [Accepted: 12/03/2023] [Indexed: 12/07/2023]
Abstract
BACKGROUND New biomarkers of progression in patients with prostate cancer (PCa) are needed to improve their classification and clinical management. This systematic review investigated the relationship between single nucleotide polymorphisms (SNPs) and PCa progression. METHODS A keyword search was performed in Pubmed, EMBASE, Scopus, Web of Science, and Cochrane for publications between 2007 and 2022. We included articles with adjusted and significant associations, a median follow-up greater than or equal to 24 months, patients taken to radical prostatectomy (RP) as a first therapeutic option, and results presented based on biochemical recurrence (BCR). RESULTS In the 27 articles selected, 73 SNPs were identified in 39 genes, organized in seven functional groups. Of these, 50 and 23 SNPs were significantly associated with a higher and lower risk of PCa progression, respectively. Likewise, four haplotypes were found to have a significant association with PCa progression. CONCLUSION This article highlights the importance of SNPs as potential markers of PCa progression and their possible functional relationship with some genes relevant to its development and progression. However, most variants were identified only in cohorts from two countries; no additional studies reproduce these findings.
Collapse
Affiliation(s)
| | | | | | | | - Martha Lucía Serrano López
- Cancer Biology Research Group, Instituto Nacional de Cancerología, Bogotá, Colombia
- Department of Chemistry, Universidad Nacional de Colombia, Bogotá, Colombia
| |
Collapse
|
3
|
Zhang N, Li Y. Receptor tyrosine kinases: biological functions and anticancer targeted therapy. MedComm (Beijing) 2023; 4:e446. [PMID: 38077251 PMCID: PMC10701465 DOI: 10.1002/mco2.446] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Revised: 11/23/2023] [Accepted: 11/24/2023] [Indexed: 10/16/2024] Open
Abstract
Receptor tyrosine kinases (RTKs) are a class of protein kinases that play crucial roles in various cellular processes, including cell migration, morphological differentiation, cell growth, and angiogenesis. In humans, 58 RTKs have been identified and categorized into 20 distinct families based on the composition of their extracellular regions. RTKs are primarily activated by specific ligands that bind to their extracellular region. They not only regulate tumor transformation, proliferation, metastasis, drug resistance, and angiogenesis, but also initiate and maintain the self-renewal and cloning ability of cancer stem cells. Accurate diagnosis and grading of tumors with dysregulated RTKs are essential in clinical practice. There is a growing body of evidence supporting the benefits of RTKs-targeted therapies for cancer patients, and researchers are actively exploring new targets and developing targeted agents. However, further optimization of RTK inhibitors is necessary to effectively target the diverse RTK alterations observed in human cancers. This review provides insights into the classification, structure, activation mechanisms, and expression of RTKs in tumors. It also highlights the research advances in RTKs targeted anticancer therapy and emphasizes their significance in optimizing cancer diagnosis and treatment strategies.
Collapse
Affiliation(s)
- Nan Zhang
- Chongqing University Cancer Hospital, School of MedicineChongqing UniversityChongqingChina
| | - Yongsheng Li
- Chongqing University Cancer Hospital, School of MedicineChongqing UniversityChongqingChina
- Department of Medical OncologyChongqing University Cancer HospitalChongqingChina
| |
Collapse
|
4
|
Ragothaman M, Yoo SY. Engineered Phage-Based Cancer Vaccines: Current Advances and Future Directions. Vaccines (Basel) 2023; 11:vaccines11050919. [PMID: 37243023 DOI: 10.3390/vaccines11050919] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Revised: 04/22/2023] [Accepted: 04/27/2023] [Indexed: 05/28/2023] Open
Abstract
Bacteriophages have emerged as versatile tools in the field of bioengineering, with enormous potential in tissue engineering, vaccine development, and immunotherapy. The genetic makeup of phages can be harnessed for the development of novel DNA vaccines and antigen display systems, as they can provide a highly organized and repetitive presentation of antigens to immune cells. Bacteriophages have opened new possibilities for the targeting of specific molecular determinants of cancer cells. Phages can be used as anticancer agents and carriers of imaging molecules and therapeutics. In this review, we explored the role of bacteriophages and bacteriophage engineering in targeted cancer therapy. The question of how the engineered bacteriophages can interact with the biological and immunological systems is emphasized to comprehend the underlying mechanism of phage use in cancer immunotherapy. The effectiveness of phage display technology in identifying high-affinity ligands for substrates, such as cancer cells and tumor-associated molecules, and the emerging field of phage engineering and its potential in the development of effective cancer treatments are discussed. We also highlight phage usage in clinical trials as well as the related patents. This review provides a new insight into engineered phage-based cancer vaccines.
Collapse
Affiliation(s)
- Murali Ragothaman
- BIO-IT Foundry Technology Institute, Pusan National University, Busan 46241, Republic of Korea
| | - So Young Yoo
- BIO-IT Foundry Technology Institute, Pusan National University, Busan 46241, Republic of Korea
| |
Collapse
|
5
|
Teruyama F, Kuno A, Murata Y, Nakagawa T, Shiba-Ishii A, Yuguchi S, Noguchi M. Mutational landscape of primary breast angiosarcoma with repeated resection and recurrence over a 15-year period: A case report. Pathol Int 2022; 72:457-463. [PMID: 35801418 DOI: 10.1111/pin.13257] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Revised: 06/08/2022] [Accepted: 06/12/2022] [Indexed: 01/18/2023]
Abstract
Angiosarcoma is a rare malignant tumor derived from vascular endothelial cells and has a poor prognosis. We have experienced a case of multiple breast angiosarcoma for which multiple resections had been performed during the course of its progression over a period of more than 15 years, allowing comprehensive genetic mutation analysis. Somatic mutations in several cancer-related genes were detected, but no previously reported driver gene mutations of angiosarcoma were evident. Several germline mutations associated with malignancy, such as single nucleotide polymorphisms in Fibroblast Growth Factor Receptor 4 (FGFR4) (p.Gly388Arg, rs351855), Kinase Insert Domain Receptor (KDR) (Gln472His, rs1870377) and tumor protein p53 (TP53) (p.Pro72Arg, rs1042522) were detected. Common signatures and genetic mutations were scarce in the tumor samples subjected to genetic mutational analysis. These findings suggested that this case was very probably a multiprimary angiosarcoma.
Collapse
Affiliation(s)
- Fumiya Teruyama
- Department of Pathology, Faculty of Medicine, University of Tsukuba, Ibaraki, Japan.,Graduate School of Comprehensive Human Sciences, University of Tsukuba, Ibaraki, Japan
| | - Akihiro Kuno
- Department of Anatomy and Embryology, Faculty of Medicine, University of Tsukuba, Ibaraki, Japan.,School of Integrative and Global Majors, University of Tsukuba, Ibaraki, Japan
| | - Yoshihiko Murata
- Department of Pathology, Faculty of Medicine, University of Tsukuba, Ibaraki, Japan
| | - Tomoki Nakagawa
- Department of Pathology, Faculty of Medicine, University of Tsukuba, Ibaraki, Japan
| | - Aya Shiba-Ishii
- Department of Pathology, Faculty of Medicine, University of Tsukuba, Ibaraki, Japan
| | - Shu Yuguchi
- Department of Pathology, Faculty of Medicine, University of Tsukuba, Ibaraki, Japan.,Department of Pathology, Narita Tomisato Tokushukai Hospital, Chiba, Japan
| | - Masayuki Noguchi
- Department of Pathology, Narita Tomisato Tokushukai Hospital, Chiba, Japan.,Center for Clinical and Translational Science, Shonan Kamakura General Hospital, Kanagawa, Japan
| |
Collapse
|
6
|
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
|
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
|
Aloisio A, Nisticò N, Mimmi S, Maisano D, Vecchio E, Fiume G, Iaccino E, Quinto I. Phage-Displayed Peptides for Targeting Tyrosine Kinase Membrane Receptors in Cancer Therapy. Viruses 2021; 13:649. [PMID: 33918836 PMCID: PMC8070105 DOI: 10.3390/v13040649] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Revised: 04/04/2021] [Accepted: 04/07/2021] [Indexed: 02/06/2023] Open
Abstract
Receptor tyrosine kinases (RTKs) regulate critical physiological processes, such as cell growth, survival, motility, and metabolism. Abnormal activation of RTKs and relative downstream signaling is implicated in cancer pathogenesis. Phage display allows the rapid selection of peptide ligands of membrane receptors. These peptides can target in vitro and in vivo tumor cells and represent a novel therapeutic approach for cancer therapy. Further, they are more convenient compared to antibodies, being less expensive and non-immunogenic. In this review, we describe the state-of-the-art of phage display for development of peptide ligands of tyrosine kinase membrane receptors and discuss their potential applications for tumor-targeted therapy.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Ileana Quinto
- Correspondence: (A.A.); (I.Q.): Tel.: +39-0961-3694057 (I.Q.)
| |
Collapse
|
9
|
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
|
10
|
Garcia-Recio S, Thennavan A, East MP, Parker JS, Cejalvo JM, Garay JP, Hollern DP, He X, Mott KR, Galván P, Fan C, Selitsky SR, Coffey AR, Marron D, Brasó-Maristany F, Burgués O, Albanell J, Rojo F, Lluch A, de Dueñas EM, Rosen JM, Johnson GL, Carey LA, Prat A, Perou CM. FGFR4 regulates tumor subtype differentiation in luminal breast cancer and metastatic disease. J Clin Invest 2021; 130:4871-4887. [PMID: 32573490 DOI: 10.1172/jci130323] [Citation(s) in RCA: 52] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Accepted: 06/17/2020] [Indexed: 12/14/2022] Open
Abstract
Mechanisms driving tumor progression from less aggressive subtypes to more aggressive states represent key targets for therapy. We identified a subset of luminal A primary breast tumors that give rise to HER2-enriched (HER2E) subtype metastases, but remain clinically HER2 negative (cHER2-). By testing the unique genetic and transcriptomic features of these cases, we developed the hypothesis that FGFR4 likely participates in this subtype switching. To evaluate this, we developed 2 FGFR4 genomic signatures using a patient-derived xenograft (PDX) model treated with an FGFR4 inhibitor, which inhibited PDX growth in vivo. Bulk tumor gene expression analysis and single-cell RNA sequencing demonstrated that the inhibition of FGFR4 signaling caused molecular switching. In the Molecular Taxonomy of Breast Cancer International Consortium (METABRIC) breast cancer cohort, FGFR4-induced and FGFR4-repressed signatures each predicted overall survival. Additionally, the FGFR4-induced signature was an independent prognostic factor beyond subtype and stage. Supervised analysis of 77 primary tumors with paired metastases revealed that the FGFR4-induced signature was significantly higher in luminal/ER+ tumor metastases compared with their primaries. Finally, multivariate analysis demonstrated that the FGFR4-induced signature also predicted site-specific metastasis for lung, liver, and brain, but not for bone or lymph nodes. These data identify a link between FGFR4-regulated genes and metastasis, suggesting treatment options for FGFR4-positive patients, whose high expression is not caused by mutation or amplification.
Collapse
Affiliation(s)
- Susana Garcia-Recio
- Lineberger Comprehensive Center and.,Department of Genetics, School of Medicine
| | - Aatish Thennavan
- Lineberger Comprehensive Center and.,Oral and Craniofacial Biomedicine Program, School of Dentistry, and
| | - Michael P East
- Department of Pharmacology, Department of Medicine, School of Medicine, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Joel S Parker
- Lineberger Comprehensive Center and.,Department of Genetics, School of Medicine
| | - Juan M Cejalvo
- Translational Genomics and Targeted Therapeutics in Oncology (IDIBAPS), Barcelona, Spain.,Medical Oncology Department, Hospital Clinic, Barcelona, Spain
| | - Joseph P Garay
- Lineberger Comprehensive Center and.,Department of Genetics, School of Medicine
| | - Daniel P Hollern
- Lineberger Comprehensive Center and.,Department of Genetics, School of Medicine
| | - Xiaping He
- Lineberger Comprehensive Center and.,Department of Genetics, School of Medicine
| | - Kevin R Mott
- Lineberger Comprehensive Center and.,Department of Genetics, School of Medicine
| | - Patricia Galván
- Translational Genomics and Targeted Therapeutics in Oncology (IDIBAPS), Barcelona, Spain.,Medical Oncology Department, Hospital Clinic, Barcelona, Spain
| | - Cheng Fan
- Lineberger Comprehensive Center and.,Department of Genetics, School of Medicine
| | | | | | | | - Fara Brasó-Maristany
- Translational Genomics and Targeted Therapeutics in Oncology (IDIBAPS), Barcelona, Spain.,Medical Oncology Department, Hospital Clinic, Barcelona, Spain
| | - Octavio Burgués
- GEICAM, Spanish Breast Cancer Group, Madrid, Spain.,Department of Pathology, Hospital Clínico Universitario de Valencia, Valencia, Spain
| | - Joan Albanell
- GEICAM, Spanish Breast Cancer Group, Madrid, Spain.,Centro de Investigación Biomédica en Red de Oncología (CIBERONC-ISCIII), Madrid, Spain.,IMIM Hospital del Mar Medical Research Institute, Barcelona, Spain.,Medical Oncology Department Hospital del Mar, Barcelona, Spain.,Universitat Pompeu Fabra, Barcelona, Spain
| | - Federico Rojo
- GEICAM, Spanish Breast Cancer Group, Madrid, Spain.,Centro de Investigación Biomédica en Red de Oncología (CIBERONC-ISCIII), Madrid, Spain.,Fundación Jiménez Díaz, Madrid, Spain
| | - Ana Lluch
- GEICAM, Spanish Breast Cancer Group, Madrid, Spain.,Centro de Investigación Biomédica en Red de Oncología (CIBERONC-ISCIII), Madrid, Spain.,Hospital Clínico Universitario de Valencia, Valencia, Spain.,Biomedical Research Institute INCLIVA, Universitat de València, Valencia, Spain
| | - Eduardo Martinez de Dueñas
- GEICAM, Spanish Breast Cancer Group, Madrid, Spain.,Centro de Investigación Biomédica en Red de Oncología (CIBERONC-ISCIII), Madrid, Spain.,Hospital Provincial de Castellón, Castellón, Spain
| | - Jeffery M Rosen
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas, USA
| | - Gary L Johnson
- Department of Pharmacology, Department of Medicine, School of Medicine, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Lisa A Carey
- Division of Hematology-Oncology, Department of Medicine, School of Medicine, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Aleix Prat
- Translational Genomics and Targeted Therapeutics in Oncology (IDIBAPS), Barcelona, Spain.,Medical Oncology Department, Hospital Clinic, Barcelona, Spain.,SOLTI Breast Cancer Research Group, Barcelona, Spain
| | - Charles M Perou
- Lineberger Comprehensive Center and.,Department of Genetics, School of Medicine.,Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, North Carolina, USA
| |
Collapse
|
11
|
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
|
12
|
Ardizzone A, Scuderi SA, Giuffrida D, Colarossi C, Puglisi C, Campolo M, Cuzzocrea S, Esposito E, Paterniti I. Role of Fibroblast Growth Factors Receptors (FGFRs) in Brain Tumors, Focus on Astrocytoma and Glioblastoma. Cancers (Basel) 2020; 12:E3825. [PMID: 33352931 PMCID: PMC7766440 DOI: 10.3390/cancers12123825] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Revised: 12/11/2020] [Accepted: 12/16/2020] [Indexed: 12/21/2022] Open
Abstract
Despite pharmacological treatments and surgical practice options, the mortality rate of astrocytomas and glioblastomas remains high, thus representing a medical emergency for which it is necessary to find new therapeutic strategies. Fibroblast growth factors (FGFs) act through their associated receptors (FGFRs), a family of tyrosine kinase receptors consisting of four members (FGFR1-4), regulators of tissue development and repair. In particular, FGFRs play an important role in cell proliferation, survival, and migration, as well as angiogenesis, thus their gene alteration is certainly related to the development of the most common diseases, including cancer. FGFRs are subjected to multiple somatic aberrations such as chromosomal amplification of FGFR1; mutations and multiple dysregulations of FGFR2; and mutations, translocations, and significant amplifications of FGFR3 and FGFR4 that correlate to oncogenesis process. Therefore, the in-depth study of these receptor systems could help to understand the etiology of both astrocytoma and glioblastoma so as to achieve notable advances in more effective target therapies. Furthermore, the discovery of FGFR inhibitors revealed how these biological compounds improve the neoplastic condition by demonstrating efficacy and safety. On this basis, this review focuses on the role and involvement of FGFRs in brain tumors such as astrocytoma and glioblastoma.
Collapse
Affiliation(s)
- Alessio Ardizzone
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno d’Alcontres, 98166 Messina, Italy; (A.A.); (S.A.S.); (M.C.); (S.C.); (E.E.)
| | - Sarah A. Scuderi
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno d’Alcontres, 98166 Messina, Italy; (A.A.); (S.A.S.); (M.C.); (S.C.); (E.E.)
| | - Dario Giuffrida
- Istituto Oncologico del Mediterraneo, Via Penninazzo 7, 95029 Viagrande (CT), Italy; (D.G.); (C.C.)
| | - Cristina Colarossi
- Istituto Oncologico del Mediterraneo, Via Penninazzo 7, 95029 Viagrande (CT), Italy; (D.G.); (C.C.)
| | - Caterina Puglisi
- IOM Ricerca Srl, Via Penninazzo 11, 95029 Viagrande (CT), Italy;
| | - Michela Campolo
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno d’Alcontres, 98166 Messina, Italy; (A.A.); (S.A.S.); (M.C.); (S.C.); (E.E.)
| | - Salvatore Cuzzocrea
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno d’Alcontres, 98166 Messina, Italy; (A.A.); (S.A.S.); (M.C.); (S.C.); (E.E.)
| | - Emanuela Esposito
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno d’Alcontres, 98166 Messina, Italy; (A.A.); (S.A.S.); (M.C.); (S.C.); (E.E.)
| | - Irene Paterniti
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno d’Alcontres, 98166 Messina, Italy; (A.A.); (S.A.S.); (M.C.); (S.C.); (E.E.)
| |
Collapse
|
13
|
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
|
14
|
Giacomini A, Grillo E, Rezzola S, Ribatti D, Rusnati M, Ronca R, Presta M. The FGF/FGFR system in the physiopathology of the prostate gland. Physiol Rev 2020; 101:569-610. [PMID: 32730114 DOI: 10.1152/physrev.00005.2020] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Fibroblast growth factors (FGFs) are a family of proteins possessing paracrine, autocrine, or endocrine functions in a variety of biological processes, including embryonic development, angiogenesis, tissue homeostasis, wound repair, and cancer. Canonical FGFs bind and activate tyrosine kinase FGF receptors (FGFRs), triggering intracellular signaling cascades that mediate their biological activity. Experimental evidence indicates that FGFs play a complex role in the physiopathology of the prostate gland that ranges from essential functions during embryonic development to modulation of neoplastic transformation. The use of ligand- and receptor-deleted mouse models has highlighted the requirement for FGF signaling in the normal development of the prostate gland. In adult prostate, the maintenance of a functional FGF/FGFR signaling axis is critical for organ homeostasis and function, as its disruption leads to prostate hyperplasia and may contribute to cancer progression and metastatic dissemination. Dissection of the molecular landscape modulated by the FGF family will facilitate ongoing translational efforts directed toward prostate cancer therapy.
Collapse
Affiliation(s)
- Arianna Giacomini
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy; Department of Basic Medical Sciences, Neurosciences, and Sensory Organs, University of Bari Medical School, Bari, Italy; and Italian Consortium for Biotechnology, Unit of Brescia, Brescia, Italy
| | - Elisabetta Grillo
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy; Department of Basic Medical Sciences, Neurosciences, and Sensory Organs, University of Bari Medical School, Bari, Italy; and Italian Consortium for Biotechnology, Unit of Brescia, Brescia, Italy
| | - Sara Rezzola
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy; Department of Basic Medical Sciences, Neurosciences, and Sensory Organs, University of Bari Medical School, Bari, Italy; and Italian Consortium for Biotechnology, Unit of Brescia, Brescia, Italy
| | - Domenico Ribatti
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy; Department of Basic Medical Sciences, Neurosciences, and Sensory Organs, University of Bari Medical School, Bari, Italy; and Italian Consortium for Biotechnology, Unit of Brescia, Brescia, Italy
| | - Marco Rusnati
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy; Department of Basic Medical Sciences, Neurosciences, and Sensory Organs, University of Bari Medical School, Bari, Italy; and Italian Consortium for Biotechnology, Unit of Brescia, Brescia, Italy
| | - Roberto Ronca
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy; Department of Basic Medical Sciences, Neurosciences, and Sensory Organs, University of Bari Medical School, Bari, Italy; and Italian Consortium for Biotechnology, Unit of Brescia, Brescia, Italy
| | - Marco Presta
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy; Department of Basic Medical Sciences, Neurosciences, and Sensory Organs, University of Bari Medical School, Bari, Italy; and Italian Consortium for Biotechnology, Unit of Brescia, Brescia, Italy
| |
Collapse
|
15
|
Chow WA, Yee JK, Tsark W, Wu X, Qin H, Guan M, Ross JS, Ali SM, Millis SZ. Recurrent secondary genomic alterations in desmoplastic small round cell tumors. BMC MEDICAL GENETICS 2020; 21:101. [PMID: 32393201 PMCID: PMC7216377 DOI: 10.1186/s12881-020-01034-w] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Accepted: 04/26/2020] [Indexed: 12/12/2022]
Abstract
BACKGROUND Desmoplastic small round cell tumor (DSRCT) is a rare, highly aggressive, translocation-associated soft-tissue sarcoma that primarily affects children, adolescents, and young adults, with a striking male predominance. It is characterized by t(11;22) generating a novel EWSR1-WT1 fusion gene. Secondary genomic alterations are rarely described. METHODS Tumor tissue from 83 DSRCT patients was assayed by hybrid-capture based comprehensive genomic profiling, FoundationOne® Heme next generation sequencing analysis of 406 genes and RNA sequencing of 265 genes. Tumor mutation burden was calculated from a minimum of 1.4 Mb sequenced DNA. Microsatellite instability status was determined by a novel algorithm analyzing 114 specific loci. RESULTS Comprehensive genomic profiling identified several genomically-defined DSRCT subgroups. Recurrent genomic alterations were most frequently detected in FGFR4, ARID1A, TP53, MSH3, and MLL3 genes. With the exception of FGFR4, where the genomic alterations predicted activation, most of the alterations in the remaining genes predicted gene inactivation. No DSRCT were TMB or MSI high. CONCLUSIONS In summary, recurrent secondary somatic alterations in FGFR4, ARID1A, TP53, MSH3, and MLL3 were detected in 82% of DSRCT, which is significantly greater than previously reported. These alterations may have both prognostic and therapeutic implications.
Collapse
Affiliation(s)
- Warren A Chow
- Department of Medical Oncology & Therapeutics Research, City of Hope, 1500 E. Duarte Rd, Duarte, CA, 91010, USA.
| | - Jiing-Kuan Yee
- Department of Translational Research & Cellular Therapeutics, City of Hope, Duarte, CA, USA
| | - Walter Tsark
- Center for Comparative Medicine, City of Hope, Duarte, CA, USA
| | - Xiwei Wu
- Integrative Genomics Core of Beckman Research Institute, City of Hope, Duarte, CA, USA
| | - Hanjun Qin
- Integrative Genomics Core of Beckman Research Institute, City of Hope, Duarte, CA, USA
| | - Min Guan
- Department of Medical Oncology & Therapeutics Research, City of Hope, 1500 E. Duarte Rd, Duarte, CA, 91010, USA
| | - Jeffrey S Ross
- Foundation Medicine, Inc, Cambridge, MA, USA.,Department of Pathology, Upstate Medical University, Syracuse, NY, USA
| | - Siraj M Ali
- Foundation Medicine, Inc, Cambridge, MA, USA
| | | |
Collapse
|
16
|
Ye Y, Jiang D, Li J, Han C, Wang X, Wang F, Li J. Role of fibroblast growth factor 4 in the growth and metastasis of colorectal cancer. Int J Oncol 2020; 56:1565-1573. [PMID: 32236572 DOI: 10.3892/ijo.2020.5029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2019] [Accepted: 02/20/2020] [Indexed: 11/05/2022] Open
Abstract
The role of fibroblast growth factor receptor 4 (FGFR4) in colorectal cancer (CRC) is poorly characterized. Therefore, the objective of the current study was to investigate the expression levels of FGFR4 in colorectal cancer and its prognostic value, and clarify the role of FGFR4 in the proliferation and metastasis of colorectal cancer cells. Immunohistochemistry was used to detect the association between FGFR4 expression and clinicopathological features in colorectal cancer tissues. The effect of FGFR4 silencing on tumor cell proliferation, cell cycle, apoptosis, migration and invasion was evaluated via lentiviral transfection of the colorectal cancer cell line SW620. Western blot analysis was used to detect the changes of epithelial‑mesenchymal transition (EMT) markers, following FGFR4 silencing. FGFR4 is upregulated in CRC tissues compared with normal tissues. Patients with high FGFR4 expression exhibited a lower 5‑year survival rate compared with patients with low FGFR4 expression (64 vs. 74%). FGFR4 silencing reduced proliferation, inhibited cell invasion, arrested cells in S phase and promoted apoptosis in colorectal cancer cells. FGFR4 silencing partially reversed EMT progression and FGFR4 this effect was enhanced in the presence of XAV939 (a β‑catenin inhibitor). The current data suggest that FGFR4 may be associated with prognosis in patients with colorectal cancer. In vitro functional tests revealed that FGFR4 may represent an effective therapeutic target for colorectal cancer. FGFR4 may also regulate EMT via the Wnt/β‑catenin pathway.
Collapse
Affiliation(s)
- Yanwei Ye
- Department of Gastrointestinal Surgery and Institute of Clinical Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, Henan 450003, P.R. China
| | - Dongbao Jiang
- Department of Gastrointestinal Surgery and Institute of Clinical Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, Henan 450003, P.R. China
| | - Jie Li
- Department of Gastrointestinal Surgery and Institute of Clinical Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, Henan 450003, P.R. China
| | - Chao Han
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, Henan 450003, P.R. China
| | - Xinru Wang
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, Henan 450003, P.R. China
| | - Feng Wang
- Department of Pathology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, Henan 450003, P.R. China
| | - Jingjing Li
- Department of Gastroenterology, Institute of Clinical Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, Henan 450003, P.R. China
| |
Collapse
|
17
|
Shao L, Kahraman N, Yan G, Wang J, Ozpolat B, Ittmann M. Targeting the TMPRSS2/ERG fusion mRNA using liposomal nanovectors enhances docetaxel treatment in prostate cancer. Prostate 2020; 80:65-73. [PMID: 31614005 PMCID: PMC6925833 DOI: 10.1002/pros.23918] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/15/2019] [Accepted: 10/02/2019] [Indexed: 11/07/2022]
Abstract
BACKGROUND The TMPRSS2/ERG (TE) fusion gene is present in half of the prostate cancers (PCas). The TMPRSS2 and ERG junction of the fusion messenger RNA (mRNA) constitutes a cancer-specific target. Although docetaxel-based chemotherapy is the second line of therapy following development resistance to androgen ablation therapies, it is not curative. Therefore, the development of nontoxic novel monotherapies for targeting TE mRNA in PCa patients and for increasing the clinical efficacy of docetaxel treatment are needed. METHODS We evaluated multiple approaches to enhance the delivery of TE small interfering RNA (siRNA) containing liposomes including PEGylation, topical treatment with nitroglycerin (NG) to increase permeability and retention, and three different PEG modifications: folate, RGD cyclic peptide, and a bFGF fibroblast growth factor receptor-targeting peptide. The efficacy of the optimized TE siRNA liposome in combination with docetaxel was then evaluated in vivo with or without topical NG in vivo using a VCaP xenograft model. TE fusion protein knockdown in residual tumors was assessed using Western blotting and immunohistochemistry. RESULTS In vivo therapeutic targeting of TE fusion gene by systemic delivery of RGD-peptide-coated liposomal siRNA nanovectors led to sustained target silencing, suppressed tumor growth in xenograft models and enhanced the efficacy of docetaxel chemotherapy. Simultaneous application of the vasodilator NG to the skin further increased tissue the delivery of siRNA and enhanced target knockdown. CONCLUSION TE-targeted gene silencing therapy using liposomal nanovectors is a potential therapeutic strategy as a monotherapy and to enhance the efficacy of chemotherapy in patients with advanced PCa.
Collapse
Affiliation(s)
- Longjiang Shao
- Dept. of Pathology & Immunology, Baylor College of Medicine and Michael E. DeBakey Dept. of Veterans Affairs Medical Center, Houston, Texas 77030
| | - Nermin Kahraman
- Departments of Experimental Therapeutics, The University of Texas M. D. Anderson Cancer Center, Houston,Texas 77030
| | - Ge Yan
- Dept. of Pathology & Immunology, Baylor College of Medicine and Michael E. DeBakey Dept. of Veterans Affairs Medical Center, Houston, Texas 77030
| | - Jianghua Wang
- Dept. of Pathology & Immunology, Baylor College of Medicine and Michael E. DeBakey Dept. of Veterans Affairs Medical Center, Houston, Texas 77030
| | - Bulent Ozpolat
- Departments of Experimental Therapeutics, The University of Texas M. D. Anderson Cancer Center, Houston,Texas 77030
| | - Michael Ittmann
- Dept. of Pathology & Immunology, Baylor College of Medicine and Michael E. DeBakey Dept. of Veterans Affairs Medical Center, Houston, Texas 77030
| |
Collapse
|
18
|
Akdeniz D, Tuncer SB, Kebudi R, Celik B, Kuru G, Kilic S, Sukruoglu Erdogan O, Avsar M, Buyukkapu Bay S, Tuncer S, Yazici H. Investigation of new candidate genes in retinoblastoma using the TruSight One "clinical exome" gene panel. Mol Genet Genomic Med 2019; 7:e785. [PMID: 31207142 PMCID: PMC6687622 DOI: 10.1002/mgg3.785] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2019] [Revised: 05/07/2019] [Accepted: 05/17/2019] [Indexed: 01/04/2023] Open
Abstract
BACKGROUND Retinoblastoma (Rb) is the most prevalent intraocular pediatric malignancy of the retina. Significant genetic factors are known to have a role in the development of Rb. METHODS Here, we report the mutation status of 4813 clinically significant genes in six patients with noncarrier of RB1 gene mutation and having normal RB1 promoter methylation from three families having higher risk for developing Rb in the study. RESULTS A total of 27 variants were detected in the study. Heterozygous missense variants c.1162G > A (p.Gly388Arg) in the FGFR4 gene; c.559C > T (p.Pro187Ser) in the NQO1 gene were identified. The family based evaluation of the variants showed that the variant, c.714T > G (p.Tyr238Ter), in the CLEC7A gene in first family; the variant, c.55C > T (p.Arg19Ter), in the APOC3 gene and the variant, c.1171C > T (p.Gln391Ter), in the MUTYH gene in second family; and the variant, c.211G > A (p.Gly71Arg), in the UGT1A1 gene in the third family, were found statistically significant (p < 0.05). CONCLUSION This study might be an important report on emphazing the mutational status of other genes in patients without RB1 gene mutations and having high risk for developing Rb. The study also indicates the interaction between the retinoic acid pathway and Rb oncogenesis for the first time.
Collapse
Affiliation(s)
- Demet Akdeniz
- Division of Cancer Genetics, Department of Basic Oncology, Oncology Institute, Istanbul University, Istanbul, Turkey
| | - Seref Bugra Tuncer
- Division of Cancer Genetics, Department of Basic Oncology, Oncology Institute, Istanbul University, Istanbul, Turkey
| | - Rejin Kebudi
- Division of Pediatric Hematology-Oncology, Oncology Institute, Istanbul University, Istanbul, Turkey.,Division of Pediatric Hematology-Oncology, Cerrahpasa Medical Faculty, Istanbul University-Cerrahpasa, Istanbul, Turkey
| | - Betul Celik
- Division of Cancer Genetics, Department of Basic Oncology, Oncology Institute, Istanbul University, Istanbul, Turkey
| | - Gozde Kuru
- Division of Cancer Genetics, Department of Basic Oncology, Oncology Institute, Istanbul University, Istanbul, Turkey
| | - Seda Kilic
- Division of Cancer Genetics, Department of Basic Oncology, Oncology Institute, Istanbul University, Istanbul, Turkey
| | - Ozge Sukruoglu Erdogan
- Division of Cancer Genetics, Department of Basic Oncology, Oncology Institute, Istanbul University, Istanbul, Turkey
| | - Mukaddes Avsar
- Division of Cancer Genetics, Department of Basic Oncology, Oncology Institute, Istanbul University, Istanbul, Turkey
| | - Sema Buyukkapu Bay
- Division of Pediatric Hematology-Oncology, Oncology Institute, Istanbul University, Istanbul, Turkey
| | - Samuray Tuncer
- Istanbul Medical Faculty, Department of Ophthalmology, Istanbul University, Istanbul, Turkey
| | - Hulya Yazici
- Division of Cancer Genetics, Department of Basic Oncology, Oncology Institute, Istanbul University, Istanbul, Turkey
| |
Collapse
|
19
|
Gege C, Hambruch E, Hambruch N, Kinzel O, Kremoser C. Nonsteroidal FXR Ligands: Current Status and Clinical Applications. Handb Exp Pharmacol 2019; 256:167-205. [PMID: 31197565 DOI: 10.1007/164_2019_232] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
FXR agonists have demonstrated very promising clinical results in the treatment of liver disorders such as primary biliary cirrhosis (PBC), primary sclerosing cholangitis (PSC), and nonalcoholic steatohepatitis (NASH). NASH, in particular, is one of the last uncharted white territories in the pharma landscape, and there is a huge medical need and a large potential pharmaceutical market for a NASH pharmacotherapy. Clinical efficacy superior to most other treatment options was shown by FXR agonists such as obeticholic acid (OCA) as they improved various metabolic features including liver steatosis as well as liver inflammation and fibrosis. But OCA's clinical success comes with some major liabilities such as pruritus, high-density lipoprotein cholesterol (HDLc) lowering, low-density lipoprotein cholesterol (LDLc) increase, and a potential for drug-induced liver toxicity. Some of these effects can be attributed to on-target effects exerted by FXR, but with others it is not clear whether it is FXR- or OCA-related. Therefore a quest for novel, proprietary FXR agonists is ongoing with the aim to increase FXR potency and selectivity over other proteins and to overcome at least some of the OCA-associated clinical side effects through an improved pharmacology. In this chapter we will discuss the historical and ongoing efforts in the identification and development of nonsteroidal, which largely means non-bile acid-type, FXR agonists for clinical use.
Collapse
Affiliation(s)
- Christian Gege
- Phenex Pharmaceuticals AG, Drug Discovery Research, Heidelberg, Germany
| | - Eva Hambruch
- Phenex Pharmaceuticals AG, Drug Discovery Research, Heidelberg, Germany
| | - Nina Hambruch
- Phenex Pharmaceuticals AG, Drug Discovery Research, Heidelberg, Germany
| | - Olaf Kinzel
- Phenex Pharmaceuticals AG, Drug Discovery Research, Heidelberg, Germany
| | - Claus Kremoser
- Phenex Pharmaceuticals AG, Drug Discovery Research, Heidelberg, Germany.
| |
Collapse
|
20
|
Abstract
FGF19 is a noncanonical FGF ligand that can control a broad spectrum of physiological responses, which include bile acid homeostasis, liver metabolism and glucose uptake. Many of these responses are mediated by FGF19 binding to its FGFR4/β-klotho receptor complex and controlling activation of an array of intracellular signaling events. Overactivation of the FGF19/FGFR4 axis has been implicated in tumorigenic formation, progression and metastasis, and inhibitors of this axis have recently been developed for single agent use or in combination with other anticancer drugs. Considering the critical role of this receptor complex in cancer, this review focuses on recent developments and applications of FGF19/FGFR4-targeted therapeutics.
Collapse
|
21
|
Liu J, Chen G, Liu Z, Liu S, Cai Z, You P, Ke Y, Lai L, Huang Y, Gao H, Zhao L, Pelicano H, Huang P, McKeehan WL, Wu CL, Wang C, Zhong W, Wang F. Aberrant FGFR Tyrosine Kinase Signaling Enhances the Warburg Effect by Reprogramming LDH Isoform Expression and Activity in Prostate Cancer. Cancer Res 2018; 78:4459-4470. [PMID: 29891507 DOI: 10.1158/0008-5472.can-17-3226] [Citation(s) in RCA: 70] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2017] [Revised: 03/29/2018] [Accepted: 06/04/2018] [Indexed: 12/12/2022]
Abstract
The acquisition of ectopic fibroblast growthfactor receptor 1 (FGFR1) expression is well documented in prostate cancer progression. How it contributes to prostate cancer progression is not fully understood, although it is known to confer a growth advantage and promote cell survival. Here, we report that FGFR1 tyrosine kinase reprograms the energy metabolism of prostate cancer cells by regulating the expression of lactate dehydrogenase (LDH) isozymes. FGFR1 increased LDHA stability through tyrosine phosphorylation and reduced LDHB expression by promoting its promoter methylation, thereby shifting cell metabolism from oxidative phosphorylation to aerobic glycolysis. LDHA depletion compromised, whereas LDHB depletion enhanced the tumorigenicity of prostate cancer cells. Furthermore, FGFR1 overexpression and aberrant LDH isozyme expression were associated with short overall survival and biochemical recurrence times in patients with prostate cancer. Our results indicate that ectopic FGFR1 expression reprograms the energy metabolism of prostate cancer cells, representing a hallmark change in prostate cancer progression.Significance: FGF signaling drives the Warburg effect through differential regulation of LDHA and LDHB, thereby promoting the progression of prostate cancer.Graphical Abstract: http://cancerres.aacrjournals.org/content/canres/78/16/4459/F1.large.jpg Cancer Res; 78(16); 4459-70. ©2018 AACR.
Collapse
Affiliation(s)
- Junchen Liu
- Department of Urology, Guangdong Key Laboratory of Clinical Molecular Medicine and Diagnostics, the Second Affiliated Hospital of South China University of Technology, Guangzhou, China.,Institute of Biosciences and Technology, College of Medicine, Texas A&M University, Houston, Texas
| | - Guo Chen
- Department of Urology, Guangdong Key Laboratory of Clinical Molecular Medicine and Diagnostics, the Second Affiliated Hospital of South China University of Technology, Guangzhou, China.,Institute of Biosciences and Technology, College of Medicine, Texas A&M University, Houston, Texas
| | - Zezhen Liu
- Department of Urology, Guangdong Key Laboratory of Clinical Molecular Medicine and Diagnostics, the Second Affiliated Hospital of South China University of Technology, Guangzhou, China.,Institute of Biosciences and Technology, College of Medicine, Texas A&M University, Houston, Texas
| | - Shaoyou Liu
- Department of Urology, Guangdong Key Laboratory of Clinical Molecular Medicine and Diagnostics, the Second Affiliated Hospital of South China University of Technology, Guangzhou, China.,Institute of Biosciences and Technology, College of Medicine, Texas A&M University, Houston, Texas
| | - Zhiduan Cai
- Department of Urology, Guangdong Key Laboratory of Clinical Molecular Medicine and Diagnostics, the Second Affiliated Hospital of South China University of Technology, Guangzhou, China.,Institute of Biosciences and Technology, College of Medicine, Texas A&M University, Houston, Texas
| | - Pan You
- Xianyue Hospital, Xiamen, China
| | - Yuepeng Ke
- Institute of Biosciences and Technology, College of Medicine, Texas A&M University, Houston, Texas
| | - Li Lai
- Institute of Biosciences and Technology, College of Medicine, Texas A&M University, Houston, Texas
| | - Yun Huang
- Institute of Biosciences and Technology, College of Medicine, Texas A&M University, Houston, Texas
| | | | | | - Helene Pelicano
- Departments of Translational Molecular Pathology, MD Anderson Cancer Center, Houston, Texas
| | - Peng Huang
- Departments of Translational Molecular Pathology, MD Anderson Cancer Center, Houston, Texas
| | - Wallace L McKeehan
- Institute of Biosciences and Technology, College of Medicine, Texas A&M University, Houston, Texas
| | - Chin-Lee Wu
- Departments of Pathology and Urology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Cong Wang
- Wenzhou Medical University, Wenzhou, China.
| | - Weide Zhong
- Department of Urology, Guangdong Key Laboratory of Clinical Molecular Medicine and Diagnostics, the Second Affiliated Hospital of South China University of Technology, Guangzhou, China. .,Department of Urology, Guangzhou Medical University, Guangzhou, China
| | - Fen Wang
- Institute of Biosciences and Technology, College of Medicine, Texas A&M University, Houston, Texas.
| |
Collapse
|
22
|
Pan CC, Tsuzuki T, Morii E, Fushimi H, Chen PCH, Epstein JI. Whole-exome sequencing demonstrates recurrent somatic copy number alterations and sporadic mutations in specialized stromal tumors of the prostate. Hum Pathol 2018; 76:9-16. [DOI: 10.1016/j.humpath.2017.12.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/13/2017] [Revised: 11/23/2017] [Accepted: 12/07/2017] [Indexed: 12/30/2022]
|
23
|
Wei W, You Z, Sun S, Wang Y, Zhang X, Pang D, Jiang Y. Prognostic implications of fibroblast growth factor receptor 4 polymorphisms in primary breast cancer. Mol Carcinog 2018; 57:988-996. [PMID: 29603419 DOI: 10.1002/mc.22819] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2017] [Revised: 03/20/2018] [Accepted: 03/27/2018] [Indexed: 12/14/2022]
Abstract
Fibroblast growth factor receptor 4 (FGFR4) belongs to the receptor tyrosine kinase (RTK) family, and FGFR4 polymorphisms have been implicated in both normal development and cancer, including breast cancer. In the present study, we investigated correlations between polymorphisms in FGFR4 and breast cancer prognosis. The FGFR4 SNPs rs1966265 and rs351855 were genotyped in 747 breast cancer patients using the SNaPshot method. FGFR4 protein expression was detected by immunohistochemistry in 339 samples. SNP rs351855 was correlated with FGFR4 protein expression under dominant and co-dominant models. Lymph node metastasis (LNM), ER (estrogen receptor) status, and molecular subtype were associated with high FGFR4 expression. Univariate analysis revealed rs351855 (CC/CT: P = 0.027, CC/TT: P < 0.001, CC/CT + TT: P = 0.005) to be a prognostic predictor, and multivariate analysis indicated rs351855 (CC/TT: P = 0.005) to be an independent prognostic factor. Kaplan-Meier survival analysis showed that high FGFR4 protein expression was associated with a poor prognosis. SNP rs351855 was correlated with worse outcomes, with a dose-dependent effect. The results of this study show that FGFR4 SNP rs351855 is associated with up-regulation of FGFR4 protein expression and a worse prognosis in breast cancer.
Collapse
Affiliation(s)
- Wei Wei
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, Harbin, China
| | - Zilong You
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, Harbin, China
| | - Shanshan Sun
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, Harbin, China
| | - Yuhang Wang
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, Harbin, China
| | - Xianyu Zhang
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, Harbin, China
| | - Da Pang
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, Harbin, China.,North China Translational Medicine Research and Cooperation Center (NTMRC), Harbin, China
| | - Yongdong Jiang
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, Harbin, China
| |
Collapse
|
24
|
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
|
25
|
Ezzat S, Wang R, Pintilie M, Asa SL. FGFR4 polymorphic alleles modulate mitochondrial respiration: A novel target for somatostatin analog action in pituitary tumors. Oncotarget 2018; 8:3481-3494. [PMID: 27966451 PMCID: PMC5356897 DOI: 10.18632/oncotarget.13843] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2016] [Accepted: 11/16/2016] [Indexed: 01/09/2023] Open
Abstract
We reported that a single nucleotide polymorphism (SNP) at codon 388 of the fibroblast growth factor receptor 4 (FGFR4-Gly388Arg) can result in distinct proteins that alter pituitary cell growth and function. Here, we examined the differential properties of the available therapeutic somatostatin analogs, octreotide and pasireotide, in pituitary tumor cells expressing the different FGFR4 isoforms. Consistent with their enhanced growth properties, FGFR4-R388-expressing cells show higher mitochondrial STAT3 serine phosphorylation driving basal and maximal oxygen consumption rate (OCR) than pituitary cells expressing the more common FGFR4-G388 isoform. While both somatostatin analogs reduce the OCR in FGFR4-G388 cells, pasireotide was more effective in decreasing OCR in cells expressing the variant FGFR4-R388 isoform. Down-regulation of somatostatin receptor 5 (SSTR5) abrogated the effect of pasireotide, demonstrating its involvement in mediating this action. The effects on OCR were recapitulated by introducing a constitutively active serine STAT3 but not by a tyrosine-active mutant. Moreover, pharmacologic inhibition demonstrated the role for the phosphatase PP2A in mediating the dephosphorylation of STAT3-S727 by pasireotide. Our data indicate that FGFR4 polymorphic isoforms mediate signaling that yields mitochondrial therapeutic targets of relevance to the actions of different somatostatin analogs.
Collapse
Affiliation(s)
- Shereen Ezzat
- Department of Medicine, The Endocrine Oncology Site Group, Princes Margaret Cancer Centre, and the Ontario Cancer Institute, University Health Network, Toronto, Ontario, Canada
| | - Ri Wang
- Department of Statistics, University of Waterloo, Toronto, Canada
| | - Melania Pintilie
- Department of Biostatistics, University of Toronto, Toronto, Canada
| | - Sylvia L Asa
- Department of Pathology, University Health Network, Toronto, Canada
| |
Collapse
|
26
|
Ye Y, Wang M, Li J, Shi Y, Zhang X, Zhou Y, Zhao C, Wen J. Hypoxia-Inducible Factor-1α G Polymorphism and the Risk of Cancer: A Meta-Analysis. TUMORI JOURNAL 2018. [DOI: 10.1177/1778.19290] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Affiliation(s)
- Yanwei Ye
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou
- Institute of Clinical Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou
| | - Min Wang
- Department of Function, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou
| | - Jingjing Li
- Institute of Clinical Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou
| | - Yingqiang Shi
- Department of Stomach and Soft Tissue Sarcoma Surgery, Shanghai Cancer Center, Fu Dan University, Shanghi
- Department of Oncology, Shanghai Medical College, Fu Dan University, Shanghai, China
| | - Xiefu Zhang
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou
- Institute of Clinical Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou
| | - Ye Zhou
- Department of Stomach and Soft Tissue Sarcoma Surgery, Shanghai Cancer Center, Fu Dan University, Shanghi
- Department of Oncology, Shanghai Medical College, Fu Dan University, Shanghai, China
| | - Chunlin Zhao
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou
- Institute of Clinical Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou
| | - Jianguo Wen
- Institute of Clinical Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou
| |
Collapse
|
27
|
Li Q, Zhao Q, Zhang C, Zhang P, Hu A, Zhang L, Schroder PM, Ma Y, Guo Z, Zhu X, He X. The ileal FGF15/19 to hepatic FGFR4 axis regulates liver regeneration after partial hepatectomy in mice. J Physiol Biochem 2018; 74:247-260. [PMID: 29468415 DOI: 10.1007/s13105-018-0610-8] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2017] [Accepted: 02/01/2018] [Indexed: 12/21/2022]
Abstract
Fibroblast growth factor (FGF) has been considered to modulate liver regeneration (LR) after partial hepatectomy (PH) at the tissue level. Previous studies have demonstrated that FGF15 and FGF19 induce the activation of its receptor, FGF receptor 4 (FGFR4), which can promote hepatocellular carcinoma progression and regulate liver lipid metabolism. In this study, we aimed to explore the role of the ileal FGF15/19- hepatic FGFR4 axis in the LR after PH. Male C57BL/6 mice aged 8-12 weeks were partially hepatectomized and assessed for expression of ileal FGF15/19 to hepatic FGFR4 signaling. We used recombinant human FGF19 protein and a small interfering RNA (siRNA) of FGFR4 to regulate expression of the FGF15/19-FGFR4 axis in vitro and in vivo. The proliferation and cell cycle of hepatocytes, the expression levels of FGF15/19-FGFR4 downstream molecules, liver recovery, and lipid metabolism were assessed. We found that both ileal and serum FGF15 expression were upregulated and hepatic FGFR4 was activated after PH in mice. FGF15/19 promoted cell cycle progression, enhanced proliferation, and reduced hepatic lipid accumulation of hepatocytes both in vitro and in vivo. Furthermore, the proliferative effect and lipid regulatory properties of FGF15/19 were dependent on FGFR4 in hepatocytes. In addition, ileal FGF15/19-hepatic FGFR4 transduction during hepatocyte proliferation was regulated by extracellular regulated protein kinase (ERK) 1/2. In conclusion, the ileal FGF15/19 to hepatic FGFR4 axis is activated and promotes LR after PH in mice, supporting the potential of ileal FGF15/19 to hepatic FGFR4 axis-targeted therapy to enhance LR after PH.
Collapse
Affiliation(s)
- Qiang Li
- Organ Transplant Center, The First Affiliated Hospital, Sun Yat-sen University, No. 58, Zhongshan Er Road, Guangzhou, 510080, China.,Guangdong Provincial Key Laboratory Construction Projection on Organ Donation and Transplant Immunology, Guangzhou, 510080, China
| | - Qiang Zhao
- Organ Transplant Center, The First Affiliated Hospital, Sun Yat-sen University, No. 58, Zhongshan Er Road, Guangzhou, 510080, China.,Guangdong Provincial Key Laboratory Construction Projection on Organ Donation and Transplant Immunology, Guangzhou, 510080, China
| | - Chuanzhao Zhang
- Organ Transplant Center, The First Affiliated Hospital, Sun Yat-sen University, No. 58, Zhongshan Er Road, Guangzhou, 510080, China.,Guangdong Provincial Key Laboratory Construction Projection on Organ Donation and Transplant Immunology, Guangzhou, 510080, China
| | - Peng Zhang
- Organ Transplant Center, The First Affiliated Hospital, Sun Yat-sen University, No. 58, Zhongshan Er Road, Guangzhou, 510080, China.,Guangdong Provincial Key Laboratory Construction Projection on Organ Donation and Transplant Immunology, Guangzhou, 510080, China
| | - Anbin Hu
- Organ Transplant Center, The First Affiliated Hospital, Sun Yat-sen University, No. 58, Zhongshan Er Road, Guangzhou, 510080, China.,Guangdong Provincial Key Laboratory Construction Projection on Organ Donation and Transplant Immunology, Guangzhou, 510080, China
| | - Longjuan Zhang
- Laboratory of Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China
| | - Paul M Schroder
- University of Toledo College of Medicine, Toledo, OH, 43614, USA
| | - Yi Ma
- Organ Transplant Center, The First Affiliated Hospital, Sun Yat-sen University, No. 58, Zhongshan Er Road, Guangzhou, 510080, China.,Guangdong Provincial Key Laboratory Construction Projection on Organ Donation and Transplant Immunology, Guangzhou, 510080, China
| | - Zhiyong Guo
- Organ Transplant Center, The First Affiliated Hospital, Sun Yat-sen University, No. 58, Zhongshan Er Road, Guangzhou, 510080, China. .,Guangdong Provincial Key Laboratory Construction Projection on Organ Donation and Transplant Immunology, Guangzhou, 510080, China.
| | - Xiaofeng Zhu
- Organ Transplant Center, The First Affiliated Hospital, Sun Yat-sen University, No. 58, Zhongshan Er Road, Guangzhou, 510080, China. .,Guangdong Provincial Key Laboratory Construction Projection on Organ Donation and Transplant Immunology, Guangzhou, 510080, China.
| | - Xiaoshun He
- Organ Transplant Center, The First Affiliated Hospital, Sun Yat-sen University, No. 58, Zhongshan Er Road, Guangzhou, 510080, China. .,Guangdong Provincial Key Laboratory Construction Projection on Organ Donation and Transplant Immunology, Guangzhou, 510080, China.
| |
Collapse
|
28
|
Shao L, Wang J, Karatas OF, Feng S, Zhang Y, Creighton CJ, Ittmann M. Fibroblast growth factor receptor signaling plays a key role in transformation induced by the TMPRSS2/ERG fusion gene and decreased PTEN. Oncotarget 2018; 9:14456-14471. [PMID: 29581856 PMCID: PMC5865682 DOI: 10.18632/oncotarget.24470] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2017] [Accepted: 02/03/2018] [Indexed: 12/12/2022] Open
Abstract
Prostate cancer is the most common visceral malignancy and the second leading cause of cancer deaths in US men. Correlative studies in human prostate cancers reveal a frequent association of the TMPRSS2/ERG (TE) fusion gene with loss of PTEN and studies in mouse models reveal that ERG expression and PTEN loss synergistically promote prostate cancer progression. To determine the mechanism by which ERG overexpression and PTEN loss leads to transformation, we overexpressed the TE fusion gene and knocked down PTEN in an immortalized but non-transformed prostate epithelial cell line. We show that ERG overexpression in combination with PTEN loss can transform these immortalized but non-tumorigenic cells, while either alteration alone was not sufficient to fully transform these cells. Expression microarray analysis revealed extensive changes in gene expression in cells expressing the TE fusion with loss of PTEN. Among these gene expression changes was increased expression of multiple FGF ligands and receptors. We show that activation of fibroblast growth factor receptor signaling plays a key role in transformation induced by TE fusion gene expression in association with PTEN loss. In addition, in vitro and in silico analysis reveals PTEN loss is associated with widespread increases in FGF ligands and receptors in prostate cancer. Inhibitors of FGF receptor signaling are currently entering the clinic and our results suggests that FGF receptor signaling is a therapeutic target in cancers with TE fusion gene expression and PTEN loss.
Collapse
Affiliation(s)
- Longjiang Shao
- Deptartment of Pathology & Immunology, Baylor College of Medicine, Houston, Texas 77030, USA.,Michael E. DeBakey Department of Veterans Affairs Medical Center, Houston, Texas 77030, USA
| | - Jianghua Wang
- Deptartment of Pathology & Immunology, Baylor College of Medicine, Houston, Texas 77030, USA.,Michael E. DeBakey Department of Veterans Affairs Medical Center, Houston, Texas 77030, USA
| | - Omer Faruk Karatas
- Deptartment of Pathology & Immunology, Baylor College of Medicine, Houston, Texas 77030, USA.,Michael E. DeBakey Department of Veterans Affairs Medical Center, Houston, Texas 77030, USA
| | - Shu Feng
- Deptartment of Pathology & Immunology, Baylor College of Medicine, Houston, Texas 77030, USA.,Michael E. DeBakey Department of Veterans Affairs Medical Center, Houston, Texas 77030, USA
| | - Yiqun Zhang
- Department of Medicine, Baylor College of Medicine, Houston, Texas 77030, USA.,Dan L. Duncan Cancer Center Division of Biostatistics, Baylor College of Medicine, Houston, Texas 77030, USA
| | - Chad J Creighton
- Department of Medicine, Baylor College of Medicine, Houston, Texas 77030, USA.,Dan L. Duncan Cancer Center Division of Biostatistics, Baylor College of Medicine, Houston, Texas 77030, USA
| | - Michael Ittmann
- Deptartment of Pathology & Immunology, Baylor College of Medicine, Houston, Texas 77030, USA.,Michael E. DeBakey Department of Veterans Affairs Medical Center, Houston, Texas 77030, USA
| |
Collapse
|
29
|
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
|
30
|
Bu Y, He X, Hu Q, Wang C, Xie X, Wang S. A novel cell membrane affinity sample pretreatment technique for recognition and preconcentration of active components from traditional Chinese medicine. Sci Rep 2017; 7:3569. [PMID: 28620157 PMCID: PMC5472601 DOI: 10.1038/s41598-017-03709-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2017] [Accepted: 05/03/2017] [Indexed: 12/16/2022] Open
Abstract
We describe a novel biomembrane affinity sample pretreatment technique to quickly screen and preconcentrate active components from traditional Chinese medicine (TCM), which adopts cell membrane coated silica particles (CMCSPs) as affinity ligands which benefit the biomembrane's ability to maximize simulation of drug-receptor interactions in vivo. In this study, the prepared CMCSPs formed by irreversible adsorption of fibroblast growth factor receptor 4 (FGFR4) cell membrane on the surface of silica were characterized using different spectroscopic and imaging instruments. Drug binding experiments showed the excellent adsorption rate and adsorption capacity of FGFR4/CMCSPs compared with non-coated silica particles. The FGFR4/CMCSPs were used as solid-phase extraction sorbents to pretreat the TCM Aconitum szechenyianum Gay. The resultant FGFR4/CMCSPs exhibited good performance. In addition, high selectivity and recognition ability of the FGFR4/CMCSPs were determined by selectivity experiments. Four alkaloid were screened and identified, one of these alkaloid, napellonine, showed favorable anti-tumor activity in preliminary pharmacological verification trials including cell proliferation and molecular docking assays. The proposed cell membrane affinity sample pretreatment method is a reliable, effective and time-saving method for fast screening and enriching active compounds and can be extended to pretreat other TCMs as leading compounds resources.
Collapse
Affiliation(s)
- Yusi Bu
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an, 710061, China.,Shaanxi Engineering Research Center of Cardiovascular Drugs Screening & Analysis, Xi'an, 710061, China
| | - Xiaoshuang He
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an, 710061, China.,Shaanxi Engineering Research Center of Cardiovascular Drugs Screening & Analysis, Xi'an, 710061, China
| | - Qi Hu
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an, 710061, China.,Shaanxi Engineering Research Center of Cardiovascular Drugs Screening & Analysis, Xi'an, 710061, China
| | - Cheng Wang
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an, 710061, China
| | - Xiaoyu Xie
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an, 710061, China. .,Shaanxi Engineering Research Center of Cardiovascular Drugs Screening & Analysis, Xi'an, 710061, China.
| | - Sicen Wang
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an, 710061, China. .,Shaanxi Engineering Research Center of Cardiovascular Drugs Screening & Analysis, Xi'an, 710061, China.
| |
Collapse
|
31
|
Chae YK, Ranganath K, Hammerman PS, Vaklavas C, Mohindra N, Kalyan A, Matsangou M, Costa R, Carneiro B, Villaflor VM, Cristofanilli M, Giles FJ. Inhibition of the fibroblast growth factor receptor (FGFR) pathway: the current landscape and barriers to clinical application. Oncotarget 2017; 8:16052-16074. [PMID: 28030802 PMCID: PMC5362545 DOI: 10.18632/oncotarget.14109] [Citation(s) in RCA: 231] [Impact Index Per Article: 33.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2016] [Accepted: 11/22/2016] [Indexed: 12/19/2022] Open
Abstract
The fibroblast growth factor/fibroblast growth factor receptor (FGF/FGFR) is a tyrosine kinase signaling pathway that has a fundamental role in many biologic processes including embryonic development, tissue regeneration, and angiogenesis. Increasing evidence indicates that this pathway plays a critical role in oncogenesis via gene amplification, activating mutations, or translocation in tumors of various histologies. With multiplex sequencing technology, the detection of FGFR aberrations has become more common and is tied to cancer cell proliferation, resistance to anticancer therapies, and neoangiogenesis. Inhibition of FGFR signaling appears promising in preclinical studies, suggesting a pathway of clinical interest in the development of targeted therapy. Phase I trials have demonstrated a manageable toxicity profile. Currently, there are multiple FGFR inhibitors under study with many non-selective (multi-kinase) inhibitors demonstrating limited clinical responses. As we progress from the first generation of non-selective drugs to the second generation of selective FGFR inhibitors, it is clear that FGFR aberrations do not behave uniformly across cancer types; thus, a deeper understanding of biomarker strategies is undoubtedly warranted. This review aims to consolidate data from recent clinical trials with a focus on selective FGFR inhibitors. As Phase II clinical trials emerge, concentration on patient selection as it pertains to predicting response to therapy, feasible methods for overcoming toxicity, and the likelihood of combination therapies should be utilized. We will also discuss qualities that may be desirable in future generations of FGFR inhibitors, with the hope that overcoming these current barriers will expedite the availability of this novel class of medications.
Collapse
Affiliation(s)
- Young Kwang Chae
- Developmental Therapeutics Program of the Division of Hematology Oncology, Chicago, IL, USA
- Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago, IL, USA
- Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Keerthi Ranganath
- Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | | | - Christos Vaklavas
- Division of Hematology Oncology, University of Alabama Birmingham, Birmingham, AL, USA
| | - Nisha Mohindra
- Developmental Therapeutics Program of the Division of Hematology Oncology, Chicago, IL, USA
- Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago, IL, USA
- Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Aparna Kalyan
- Developmental Therapeutics Program of the Division of Hematology Oncology, Chicago, IL, USA
- Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago, IL, USA
- Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Maria Matsangou
- Developmental Therapeutics Program of the Division of Hematology Oncology, Chicago, IL, USA
- Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago, IL, USA
- Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Ricardo Costa
- Developmental Therapeutics Program of the Division of Hematology Oncology, Chicago, IL, USA
| | - Benedito Carneiro
- Developmental Therapeutics Program of the Division of Hematology Oncology, Chicago, IL, USA
- Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago, IL, USA
- Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Victoria M. Villaflor
- Developmental Therapeutics Program of the Division of Hematology Oncology, Chicago, IL, USA
- Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago, IL, USA
- Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Massimo Cristofanilli
- Developmental Therapeutics Program of the Division of Hematology Oncology, Chicago, IL, USA
- Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago, IL, USA
- Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Francis J. Giles
- Developmental Therapeutics Program of the Division of Hematology Oncology, Chicago, IL, USA
- Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago, IL, USA
- Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| |
Collapse
|
32
|
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
|
33
|
Jiang Y, Sun S, Wei W, Ren Y, Liu J, Pang D. Association of FGFR3 and FGFR4 gene polymorphisms with breast cancer in Chinese women of Heilongjiang province. Oncotarget 2016; 6:34023-9. [PMID: 26431494 PMCID: PMC4741823 DOI: 10.18632/oncotarget.5850] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2015] [Accepted: 08/22/2015] [Indexed: 11/25/2022] Open
Abstract
BACKGROUND The fibroblast growth factor (FGF) receptor pathway is activated in many tumors. FGFR2 has been identified as a breast cancer susceptibility gene. Common variation in other FGF receptors might also affect breast cancer risk. We carried out a case-control study to investigate associations of variants in FGFR3 and FGFR4 with breast cancer in women from Heilongjiang Province. METHODS SNP rs2234909 and rs3135848 in FGFR3 and rs1966265 and rs351855 in FGFR4 were successfully genotyped in 747 breast cancer patients and 716 healthy controls using the SNaPshot method. The associations between SNPs and breast cancer were examined by logistic regression. The associations between SNPs and disease characteristics were examined by chi-square tests or one-way ANOVA as needed. RESULTS The minor alleles of rs1966265 and rs351855 in FGFR4 were strongly associated with breast cancer in the population, with odds ratios of 1.335 (95%CI = 1.154-1.545) and 1.364 (95%CI = 1.177-1.580), respectively. However, no significant associations were detected between other SNPs and breast cancer. Analyses of the disease characteristics showed that SNP rs351855 was associated with lymph-node-positive breast cancer with a dose-dependent effect of the minor allele (P = 0.008). CONCLUSIONS SNPs rs1966265 and rs351855 in FGFR4 were associated with breast cancer in a northern Chinese population.
Collapse
Affiliation(s)
- Yongdong Jiang
- Department of Breast Surgery, The Third Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Shanshan Sun
- Department of Breast Surgery, The Third Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Wei Wei
- Department of Breast Surgery, The Third Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Yanlv Ren
- Department of Breast Surgery, The Third Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Jing Liu
- Department of Anesthesiology, The Second Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Da Pang
- Department of Breast Surgery, The Third Affiliated Hospital, Harbin Medical University, Harbin, China
| |
Collapse
|
34
|
Zatelli MC, Fanciulli G, Malandrino P, Ramundo V, Faggiano A, Colao A. Predictive factors of response to mTOR inhibitors in neuroendocrine tumours. Endocr Relat Cancer 2016; 23:R173-83. [PMID: 26666705 DOI: 10.1530/erc-15-0413] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/09/2015] [Accepted: 12/14/2015] [Indexed: 12/13/2022]
Abstract
Medical treatment of neuroendocrine tumours (NETs) has drawn a lot of attention due to the recent demonstration of efficacy of several drugs on progression-free survival, including somatostatin analogs, small tyrosine kinase inhibitors and mTOR inhibitors (or rapalogs). The latter are approved as therapeutic agents in advanced pancreatic NETs and have been demonstrated to be effective in different types of NETs, with variable efficacy due to the development of resistance to treatment. Early detection of patients that may benefit from rapalogs treatment is of paramount importance in order to select the better treatment and avoid ineffective and expensive treatments. Predictive markers for therapeutic response are under intensive investigation, aiming at a tailored patient management and more appropriate resource utilization. This review summarizes the available data on the tissue, circulating and imaging markers that are potentially predictive of rapalog efficacy in NETs.
Collapse
Affiliation(s)
- Maria Chiara Zatelli
- Section of Endocrinology and Internal MedicineDepartment of Medical Sciences, University of Ferrara, Via Aldo Moro 8, 44124 Cona - Ferrara, ItalyNeuroendocrine Tumours UnitDepartment of Clinical and Experimental Medicine, University of Sassari - AOU Sassari, Sassari, ItalyEndocrinology UnitGaribaldi Nesima Medical Center, Catania, ItalyDepartment of Clinical Medicine and Surgery"Federico II" University of Naples, Naples, ItalyThyroid and Parathyroid Surgery UnitIstituto Nazionale per lo studio e la cura dei tumori "Fondazione G. Pascale" - IRCCS, Naples, Italy
| | - Giuseppe Fanciulli
- Section of Endocrinology and Internal MedicineDepartment of Medical Sciences, University of Ferrara, Via Aldo Moro 8, 44124 Cona - Ferrara, ItalyNeuroendocrine Tumours UnitDepartment of Clinical and Experimental Medicine, University of Sassari - AOU Sassari, Sassari, ItalyEndocrinology UnitGaribaldi Nesima Medical Center, Catania, ItalyDepartment of Clinical Medicine and Surgery"Federico II" University of Naples, Naples, ItalyThyroid and Parathyroid Surgery UnitIstituto Nazionale per lo studio e la cura dei tumori "Fondazione G. Pascale" - IRCCS, Naples, Italy
| | - Pasqualino Malandrino
- Section of Endocrinology and Internal MedicineDepartment of Medical Sciences, University of Ferrara, Via Aldo Moro 8, 44124 Cona - Ferrara, ItalyNeuroendocrine Tumours UnitDepartment of Clinical and Experimental Medicine, University of Sassari - AOU Sassari, Sassari, ItalyEndocrinology UnitGaribaldi Nesima Medical Center, Catania, ItalyDepartment of Clinical Medicine and Surgery"Federico II" University of Naples, Naples, ItalyThyroid and Parathyroid Surgery UnitIstituto Nazionale per lo studio e la cura dei tumori "Fondazione G. Pascale" - IRCCS, Naples, Italy
| | - Valeria Ramundo
- Section of Endocrinology and Internal MedicineDepartment of Medical Sciences, University of Ferrara, Via Aldo Moro 8, 44124 Cona - Ferrara, ItalyNeuroendocrine Tumours UnitDepartment of Clinical and Experimental Medicine, University of Sassari - AOU Sassari, Sassari, ItalyEndocrinology UnitGaribaldi Nesima Medical Center, Catania, ItalyDepartment of Clinical Medicine and Surgery"Federico II" University of Naples, Naples, ItalyThyroid and Parathyroid Surgery UnitIstituto Nazionale per lo studio e la cura dei tumori "Fondazione G. Pascale" - IRCCS, Naples, Italy
| | - Antongiulio Faggiano
- Section of Endocrinology and Internal MedicineDepartment of Medical Sciences, University of Ferrara, Via Aldo Moro 8, 44124 Cona - Ferrara, ItalyNeuroendocrine Tumours UnitDepartment of Clinical and Experimental Medicine, University of Sassari - AOU Sassari, Sassari, ItalyEndocrinology UnitGaribaldi Nesima Medical Center, Catania, ItalyDepartment of Clinical Medicine and Surgery"Federico II" University of Naples, Naples, ItalyThyroid and Parathyroid Surgery UnitIstituto Nazionale per lo studio e la cura dei tumori "Fondazione G. Pascale" - IRCCS, Naples, Italy
| | - Annamaria Colao
- Section of Endocrinology and Internal MedicineDepartment of Medical Sciences, University of Ferrara, Via Aldo Moro 8, 44124 Cona - Ferrara, ItalyNeuroendocrine Tumours UnitDepartment of Clinical and Experimental Medicine, University of Sassari - AOU Sassari, Sassari, ItalyEndocrinology UnitGaribaldi Nesima Medical Center, Catania, ItalyDepartment of Clinical Medicine and Surgery"Federico II" University of Naples, Naples, ItalyThyroid and Parathyroid Surgery UnitIstituto Nazionale per lo studio e la cura dei tumori "Fondazione G. Pascale" - IRCCS, Naples, Italy
| | | |
Collapse
|
35
|
A Polymorphism in the FGFR4 Gene Is Associated With Risk of Neuroblastoma and Altered Receptor Degradation. J Pediatr Hematol Oncol 2016; 38:131-8. [PMID: 26840079 PMCID: PMC4758892 DOI: 10.1097/mph.0000000000000506] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
BACKGROUND Outcomes for children with high-risk neuroblastoma are poor, and improved understanding of the mechanisms underlying neuroblastoma pathogenesis, recurrence, and treatment resistance will lead to improved outcomes. Aberrant growth factor receptor expression and receptor tyrosine kinase signaling are associated with the pathogenesis of many malignancies. A germline polymorphism in the FGFR4 gene is associated with increased receptor expression and activity and with decreased survival, treatment resistance, and aggressive disease for many malignancies. We therefore investigated the role of this FGFR4 polymorphism in neuroblastoma pathogenesis. MATERIALS AND METHODS Germline DNA from neuroblastoma patients and matched controls was assessed for the FGFR4 Gly/Arg388 polymorphism by RT-PCR. Allele frequencies were assessed for association with neuroblastoma patient outcomes and prognostic features. Degradation rates of the FGFR4 Arg388 and Gly388 receptors and rates of receptor internalization into the late endosomal compartment were measured. RESULTS Frequency of the FGFR4 AA genotype and the prevalence of the A allele were significantly higher in patients with neuroblastoma than in matched controls. The Arg388 receptor demonstrated slower degradation than the Gly388 receptor in neuroblastoma cells and reduced internalization into multivesicular bodies. CONCLUSIONS The FGFR4 Arg388 polymorphism is associated with an increased prevalence of neuroblastoma in children, and this association may be linked to differences in FGFR4 degradation rates. Our study provides the first evidence of a role for FGFR4 in neuroblastoma, suggesting that FGFR4 genotype and the pathways regulating FGFR4 trafficking and degradation may be relevant for neuroblastoma pathogenesis.
Collapse
|
36
|
Zhang L, Shao L, Creighton CJ, Zhang Y, Xin L, Ittmann M, Wang J. Function of phosphorylation of NF-kB p65 ser536 in prostate cancer oncogenesis. Oncotarget 2016; 6:6281-94. [PMID: 25749044 PMCID: PMC4467437 DOI: 10.18632/oncotarget.3366] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2014] [Accepted: 01/12/2015] [Indexed: 01/11/2023] Open
Abstract
Majority of prostate cancer (PCa) patients carry TMPRSS2/ERG (T/E) fusion genes and there has been tremendous interest in understanding how the T/E fusion may promote progression of PCa. We showed that T/E fusion can activate NF-kB pathway by increasing phosphorylation of NF-kB p65 Ser536 (p536), but the function of p536 has never been studied in PCa. We report here that active p536 can significantly increase cell motility and transform PNT1a cells (an immortalized normal cell line), suggesting p536 plays a critical role in promoting PCa tumorigenesis. We have discovered a set of p536 regulated genes, among which we validated the regulation of CCL2 by p536. Based on all evidence, we favor that T/E fusion, NF-kB p536 and CCL2 form a signaling chain. Finally, PNT1a cells (not tumorigenic) can form tumors in SCID mice when overexpressing of either wild type or active p65 in the presence of activated AKT, demonstrating synergistic activities of NF-kB and AKT signals in promoting PCa tumorigenesis. These findings indicate that combination therapies targeting T/E fusion, NF-kB, CCL2 and/or AKT pathways may have efficacy in T/E fusion gene expressing PCa. If successful, such targeted therapy will benefit more than half of PCa patients who carry T/E fusions.
Collapse
Affiliation(s)
- Li Zhang
- Department of Biochemistry and Molecular Biology, College of Basic Medical Sciences, Third Military Medical University, Chongqing, China.,Department of Pathology and Immunology, Baylor College of Medicine and Michael E. DeBakey Deptartment of Veterans Affairs Medical Center, Houston, Texas, USA
| | - Longjiang Shao
- Department of Pathology and Immunology, Baylor College of Medicine and Michael E. DeBakey Deptartment of Veterans Affairs Medical Center, Houston, Texas, USA
| | - Chad J Creighton
- Duncan Cancer Center Division of Biostatistics, Baylor College of Medicine, Houston, Texas, USA.,Department of Medicine, Baylor College of Medicine, Houston, Texas, USA
| | - Yiqun Zhang
- Duncan Cancer Center Division of Biostatistics, Baylor College of Medicine, Houston, Texas, USA
| | - Li Xin
- Department of Molecular & Cellular Biology, Baylor College of Medicine, Houston, Texas, USA
| | - Michael Ittmann
- Department of Pathology and Immunology, Baylor College of Medicine and Michael E. DeBakey Deptartment of Veterans Affairs Medical Center, Houston, Texas, USA
| | - Jianghua Wang
- Department of Pathology and Immunology, Baylor College of Medicine and Michael E. DeBakey Deptartment of Veterans Affairs Medical Center, Houston, Texas, USA
| |
Collapse
|
37
|
Li X, Wang C, Xiao J, McKeehan WL, Wang F. Fibroblast growth factors, old kids on the new block. Semin Cell Dev Biol 2016; 53:155-67. [PMID: 26768548 DOI: 10.1016/j.semcdb.2015.12.014] [Citation(s) in RCA: 58] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2015] [Accepted: 12/18/2015] [Indexed: 01/08/2023]
Abstract
The fibroblast growth factors (FGFs) are a family of cell intrinsic regulatory peptides that control a broad spectrum of cellular activities. The family includes canonic FGFs that elicit their activities by activating the FGF receptor (FGFR) tyrosine kinase and non-canonic members that elicit their activities intracellularly and via FGFR-independent mechanisms. The FGF signaling axis is highly complex due to the existence of multiple isoforms of both ligands and receptors, as well as cofactors that include the chemically heterogeneous heparan sulfate (HS) cofactors, and in the case of endocrine FGFs, the Klotho coreceptors. Resident FGF signaling controls embryonic development, maintains tissue homeostasis, promotes wound healing and tissue regeneration, and regulates functions of multiple organs. However, ectopic or aberrant FGF signaling is a culprit for various diseases, including congenital birth defects, metabolic disorder, and cancer. The molecular mechanisms by which the specificity of FGF signaling is achieved remain incompletely understood. Since its application as a druggable target has been gradually recognized by pharmaceutical companies and translational researchers, understanding the determinants of FGF signaling specificity has become even more important in order to get into the position to selectively suppress a particular pathway without affecting others to minimize side effects.
Collapse
Affiliation(s)
- Xiaokun Li
- College of Pharmacy, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Cong Wang
- College of Pharmacy, Wenzhou Medical University, Wenzhou, Zhejiang, China.
| | - Jian Xiao
- College of Pharmacy, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Wallace L McKeehan
- Institute of Biosciences and Technology, Texas A&M University Health Science Center, Houston, TX 77030-3303, United States
| | - Fen Wang
- Institute of Biosciences and Technology, Texas A&M University Health Science Center, Houston, TX 77030-3303, United States.
| |
Collapse
|
38
|
Li J, Ye Y, Wang M, Lu L, Han C, Zhou Y, Zhang J, Yu Z, Zhang X, Zhao C, Wen J, Kan Q. The over-expression of FGFR4 could influence the features of gastric cancer cells and inhibit the efficacy of PD173074 and 5-fluorouracil towards gastric cancer. Tumour Biol 2015; 37:6881-91. [PMID: 26662569 DOI: 10.1007/s13277-015-4411-1] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2015] [Accepted: 11/09/2015] [Indexed: 12/16/2022] Open
Abstract
The aim was to investigate the function of fibroblast growth factor receptor 4 (FGFR4) in gastric cancer (GC) and explore the treatment value of agent targeted to FGFR4. Function assays in vitro and in vivo were performed to investigate the discrepancy of biological features among the GC cells with different expression of FGFR4. GC cells were treated with the single and combination of PD173074 (PD, an inhibitor of FGFR4) and 5-fluorouracil (5-Fu). The invasion ability were stronger, and the apoptosis rates were lower in MGC803 and BGC823 cells treated with FGFR4-LV5 (over-expression of FGFR4 protein) (P < 0.05). The proliferation ability of GC cells is reduced when treated by the single and combination of 5-Fu and PD while that of the FGFR4-LV5 group was less inhibited compared with control group (P < 0.05). The apoptosis rates are remarkably increased in GC cells treated with the single and combination of 5-Fu and PD (P < 0.05). However, the apoptosis rate obviously is reduced in GC cells treated with FGFR4-LV5 compared with control group (P < 0.05). The expression of PCNA and Bcl-XL is remarkably decreased, and the expression of Caspase-3 and cleaved Caspase-3 is obviously increased in GC cells treated with the single and combination of 5-Fu and PD. The tumor volumes of nude mice in FGFR4-LV5 group were much more increased (P < 0.05). The over-expression of FGFR4 enhanced the proliferation ability of GC in vitro and in vivo. The combination of 5-Fu and PD exerted synergetic effect in weakening the proliferation ability and promoting apoptosis in GC cells, while the over-expression of FGFR4 might inhibit the efficacy of two drugs.
Collapse
Affiliation(s)
- Jingjing Li
- Department of Gastroenterology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Institute of Clinical Medicine, The First Affiliated Hospital of Zhengzhou University, 1 Jian-She Road, Zhengzhou, 450052, China
| | - Yanwei Ye
- Institute of Clinical Medicine, The First Affiliated Hospital of Zhengzhou University, 1 Jian-She Road, Zhengzhou, 450052, China.
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Zhengzhou University, 1 Jian-She Road, Zhengzhou, 450052, China.
| | - Min Wang
- Department of Function, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Lisha Lu
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Chao Han
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yubing Zhou
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Jingmin Zhang
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, China
| | - Zujiang Yu
- Department of Infectious Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Xiefu Zhang
- Institute of Clinical Medicine, The First Affiliated Hospital of Zhengzhou University, 1 Jian-She Road, Zhengzhou, 450052, China
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Zhengzhou University, 1 Jian-She Road, Zhengzhou, 450052, China
| | - Chunlin Zhao
- Institute of Clinical Medicine, The First Affiliated Hospital of Zhengzhou University, 1 Jian-She Road, Zhengzhou, 450052, China
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Zhengzhou University, 1 Jian-She Road, Zhengzhou, 450052, China
| | - Jianguo Wen
- Institute of Clinical Medicine, The First Affiliated Hospital of Zhengzhou University, 1 Jian-She Road, Zhengzhou, 450052, China
| | - Quancheng Kan
- Department of Gastroenterology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
39
|
Repana D, Ross P. Targeting FGF19/FGFR4 Pathway: A Novel Therapeutic Strategy for Hepatocellular Carcinoma. Diseases 2015; 3:294-305. [PMID: 28943626 PMCID: PMC5548263 DOI: 10.3390/diseases3040294] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2015] [Revised: 09/28/2015] [Accepted: 10/20/2015] [Indexed: 02/06/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is a lethal cancer with limited systemic therapeutic options. Liver carcinogenesis is a complex procedure and various pathways have been found to be deregulated which are potential targets for novel treatments. Aberrant signalling through FGF19 and its receptor FGFR4 seems to be the oncogenic driver for a subset of HCCs and is associated with poor prognosis. Inhibition of the pathway in preclinical models has shown antitumour activity and has triggered further evaluation of this strategy to in vivo models. This review aims to describe the role of the FGF19/FGFR4 pathway in hepatocellular carcinoma and its role as a potential predictive biomarker for novel targeted agents against FGF19/FGFR4 signalling.
Collapse
Affiliation(s)
- Dimitra Repana
- Department of Medical Oncology, Guy's and St. Thomas' NHS Foundation Trust, SE1 9RT London, UK.
| | - Paul Ross
- Department of Medical Oncology, Guy's and St. Thomas' NHS Foundation Trust, SE1 9RT London, UK.
- Department of Oncology, King's College Hospital, SE19 1RT London, UK.
| |
Collapse
|
40
|
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
|
41
|
Hyperactivated FRS2α-mediated signaling in prostate cancer cells promotes tumor angiogenesis and predicts poor clinical outcome of patients. Oncogene 2015; 35:1750-9. [PMID: 26096936 DOI: 10.1038/onc.2015.239] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2015] [Revised: 05/20/2015] [Accepted: 05/22/2015] [Indexed: 12/25/2022]
Abstract
Metastasis of tumors requires angiogenesis, which is comprised of multiple biological processes that are regulated by angiogenic factors. The fibroblast growth factor (FGF) is a potent angiogenic factor and aberrant FGF signaling is a common property of tumors. Yet, how the aberration in cancer cells contributes to angiogenesis in the tumor is not well understood. Most studies of its angiogenic signaling mechanisms have been in endothelial cells. FGF receptor substrate 2α (FRS2α) is an FGF receptor-associated protein required for activation of downstream signaling molecules that include those in the mitogen-activated protein and AKT kinase pathways. Herein, we demonstrated that overactivation and hyperactivity of FRS2α, as well as overexpression of cJUN and HIF1α, were positively correlated with vessel density and progression of human prostate cancer (PCa) toward malignancy. We also demonstrate that FGF upregulated the production of vascular endothelial growth factor A mainly by increasing expression of cJUN and HIF1α. This then promoted recruitment of endothelial cells and vessel formation for the tumor. Tumor angiogenesis in mouse PCa tissues was compromised by tissue-specific ablation of Frs2α in prostate epithelial cells. Depletion of Frs2α expression in human PCa cells and in a preclinical xenograft model, MDA PCa 118b, also significantly suppressed tumor angiogenesis accompanied with decreased tumor growth in the bone. The results underscore the angiogenic role of FRS2α-mediated signaling in tumor epithelial cells in angiogenesis. They provide a rationale for treating PCa with inhibitors of FGF signaling. They also demonstrate the potential of overexpressed FRS2α as a biomarker for PCa diagnosis, prognosis and response to therapies.
Collapse
|
42
|
Huang Y, Jin C, Hamana T, Liu J, Wang C, An L, McKeehan WL, Wang F. Overexpression of FGF9 in prostate epithelial cells augments reactive stroma formation and promotes prostate cancer progression. Int J Biol Sci 2015; 11:948-60. [PMID: 26157349 PMCID: PMC4495412 DOI: 10.7150/ijbs.12468] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2015] [Accepted: 05/15/2015] [Indexed: 12/31/2022] Open
Abstract
Bone metastasis is the major cause of morbidity and mortality of prostate cancer (PCa). Fibroblast growth factor 9 (FGF9) has been reported to promote PCa bone metastasis. However, the mechanism by which overexpression of FGF9 promotes PCa progression and metastasis is still unknown. Herein, we report that transgenic mice forced to express FGF9 in prostate epithelial cells (F9TG) developed high grade prostatic intraepithelial neoplasia (PIN) in an expression level- and time-dependent manner. Moreover, FGF9/TRAMP bigenic mice (F9TRAMP) grew advanced PCa earlier and had higher frequencies of metastasis than TRAMP littermates. We observed tumor microenvironmental changes including hypercellularity and hyperproliferation in the stromal compartment of F9TG and F9TRAMP mice. Expression of TGFβ1, a key signaling molecule overexpressed in reactive stroma, was increased in F9TG and F9TRAMP prostates. Both in vivo and in vitro data indicated that FGF9 promoted TGFβ1 expression via increasing cJun-mediated signaling. Moreover, in silico analyses showed that the expression level of FGF9 was positively associated with expression of TGFβ1 and its downstream signaling molecules in human prostate cancers. Collectively, our data demonstrated that overexpressing FGF9 in PCa cells augmented the formation of reactive stroma and promoted PCa initiation and progression.
Collapse
Affiliation(s)
- Yanqing Huang
- 1. Center for Cancer and Stem Cell Biology, Institute of Biosciences and Technology, Texas A&M Health Science Center, Houston, TX, USA
| | - Chengliu Jin
- 1. Center for Cancer and Stem Cell Biology, Institute of Biosciences and Technology, Texas A&M Health Science Center, Houston, TX, USA
| | - Tomoaki Hamana
- 1. Center for Cancer and Stem Cell Biology, Institute of Biosciences and Technology, Texas A&M Health Science Center, Houston, TX, USA
| | - Junchen Liu
- 1. Center for Cancer and Stem Cell Biology, Institute of Biosciences and Technology, Texas A&M Health Science Center, Houston, TX, USA
| | - Cong Wang
- 2. Wenzhou Medical College, Wenzhou, Zhejiang, China
| | - Lei An
- 1. Center for Cancer and Stem Cell Biology, Institute of Biosciences and Technology, Texas A&M Health Science Center, Houston, TX, USA
| | - Wallace L McKeehan
- 1. Center for Cancer and Stem Cell Biology, Institute of Biosciences and Technology, Texas A&M Health Science Center, Houston, TX, USA
| | - Fen Wang
- 1. Center for Cancer and Stem Cell Biology, Institute of Biosciences and Technology, Texas A&M Health Science Center, Houston, TX, USA ; 2. Wenzhou Medical College, Wenzhou, Zhejiang, China ; 3. Department of Molecular and Cellular Medicine, College of Medicine, Texas A&M, Health Science Center, College Station, TX, USA
| |
Collapse
|
43
|
Tiseo M, Gelsomino F, Alfieri R, Cavazzoni A, Bozzetti C, De Giorgi AM, Petronini PG, Ardizzoni A. FGFR as potential target in the treatment of squamous non small cell lung cancer. Cancer Treat Rev 2015; 41:527-39. [PMID: 25959741 DOI: 10.1016/j.ctrv.2015.04.011] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2015] [Revised: 04/12/2015] [Accepted: 04/25/2015] [Indexed: 12/31/2022]
Abstract
To date therapeutic options for squamous cell lung cancer patients remain scarce because no druggable targets have been identified so far. Aberrant signaling by FGFs (fibroblast growth factors) and FGFRs (fibroblast growth factors receptors) has been implicated in several human cancers and, particularly, in squamous non-small cell lung cancer (NSCLC). FGFR gene amplifications, somatic missense mutations, chromosomal translocations are the most frequent mechanisms able to induce aberrant activation of this pathway. Data from literature have established that the presence of an aberrant FGFR signaling has to be considered a possible negative prognostic factor but predictive of potential sensitivity to FGFR inhibitors. In the last years, clinical research efforts allowed to identify and evaluate promising FGFR inhibitors, such as monoclonal antibodies, ligand traps, non-selective or selective tyrosine kinase inhibitors. This review summarizes the current knowledge about FGFR alterations in NSCLC and the relative inhibitors in development, in particular in squamous NSCLC.
Collapse
Affiliation(s)
- Marcello Tiseo
- Division of Medical Oncology, University Hospital of Parma, Parma, Italy.
| | | | - Roberta Alfieri
- Department of Clinical and Experimental Medicine, University of Parma, Parma, Italy
| | - Andrea Cavazzoni
- Department of Clinical and Experimental Medicine, University of Parma, Parma, Italy
| | - Cecilia Bozzetti
- Division of Medical Oncology, University Hospital of Parma, Parma, Italy
| | | | | | - Andrea Ardizzoni
- Division of Medical Oncology, Sant'Orsola-Malpighi University Hospital, Bologna, Italy
| |
Collapse
|
44
|
Gallo LH, Nelson KN, Meyer AN, Donoghue DJ. Functions of Fibroblast Growth Factor Receptors in cancer defined by novel translocations and mutations. Cytokine Growth Factor Rev 2015; 26:425-49. [PMID: 26003532 DOI: 10.1016/j.cytogfr.2015.03.003] [Citation(s) in RCA: 109] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2015] [Accepted: 03/25/2015] [Indexed: 11/25/2022]
Abstract
The four receptor tyrosine kinases (RTKs) within the family of Fibroblast Growth Factor Receptors (FGFRs) are critical for normal development but also play an enormous role in oncogenesis. Mutations and/or abnormal expression often lead to constitutive dimerization and kinase activation of FGFRs, and represent the primary mechanism for aberrant signaling. Sequencing of human tumors has revealed a plethora of somatic mutations in FGFRs that are frequently identical to germline mutations in developmental syndromes, and has also identified novel FGFR fusion proteins arising from chromosomal rearrangements that contribute to malignancy. This review details approximately 200 specific point mutations in FGFRs and 40 different fusion proteins created by translocations involving FGFRs that have been identified in human cancer. This review discusses the effects of these genetic alterations on downstream signaling cascades, and the challenge of drug resistance in cancer treatment with antagonists of FGFRs.
Collapse
Affiliation(s)
- Leandro H Gallo
- Department of Chemistry and Biochemistry, Moores UCSD Cancer Center, University of California San Diego, La Jolla, CA 92093-0367, United States.
| | - Katelyn N Nelson
- Department of Chemistry and Biochemistry, Moores UCSD Cancer Center, University of California San Diego, La Jolla, CA 92093-0367, United States.
| | - April N Meyer
- Department of Chemistry and Biochemistry, Moores UCSD Cancer Center, University of California San Diego, La Jolla, CA 92093-0367, United States.
| | - Daniel J Donoghue
- Department of Chemistry and Biochemistry, Moores UCSD Cancer Center, University of California San Diego, La Jolla, CA 92093-0367, United States.
| |
Collapse
|
45
|
Fibroblast growth factor receptor 4 polymorphism is associated with liver cirrhosis in hepatocarcinoma. PLoS One 2015; 10:e0122961. [PMID: 25860955 PMCID: PMC4393280 DOI: 10.1371/journal.pone.0122961] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2014] [Accepted: 02/16/2015] [Indexed: 02/07/2023] Open
Abstract
Background Fibroblast growth factor receptor 4 (FGFR4) polymorphisms are positively correlated with tumor progression in numerous malignant tumors. However, the association between FGFR4 genetic variants and the risk of hepatocellular carcinoma (HCC) has not yet been determined. In this study, we investigated the potential associations of FGFR4 single nucleotide polymorphisms (SNPs) with HCC susceptibility and its clinicopathological characteristics. Methodology/Principal Findings Four SNPs in FGFR4 (rs1966265, rs351855, rs2011077, and rs7708357) were analyzed among 884 participants, including 595 controls and 289 patients with HCC. The samples were further analyzed to clarify the associations between these gene polymorphisms and the risk of HCC, and the impact of these SNPs on the susceptibility and clinicopathological characteristics of HCC. After adjusting for other covariants, HCC patients who carrying at least one A genotype (GA and AA) at rs351855 were observed to have a higher risk of liver cirrhosis compared with those carrying the wild-type genotype (GG) (OR: 2.113, 95% CI: 1.188–3.831). Moreover, the patients with at least one A genotype were particularly showed a high level of alpha-fetoprotein (AFP). Conclusions Our findings suggest that genetic polymorphism in FGFR4 rs351855 may be associated with the risk of HCC coupled with liver cirrhosis and may markedly increase the AFP level in Taiwanese patients with HCC. In addition, this is the first study that evaluated the risk factors associated with FGFR4 polymorphism variants in Taiwanese patients with HCC.
Collapse
|
46
|
Nakano-Tateno T, Tateno T, Hlaing MM, Zheng L, Yoshimoto K, Yamada S, Asa SL, Ezzat S. FGFR4 polymorphic variants modulate phenotypic features of Cushing disease. Mol Endocrinol 2014; 28:525-33. [PMID: 24625004 DOI: 10.1210/me.2013-1412] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Cushing disease is a potentially lethal condition resulting from hormone excess, usually due to a small pituitary tumor that fails to respond to negative feedback inhibition. A minority of patients develop larger, more aggressive tumors of the same lineage but with modest hormone excess. Here we show that a common polymorphism in the fibroblast growth factor receptor 4 (FGFR4) transmembrane domain yields receptor isoforms with distinct properties that mediate these biological differences. Forced expression of the major FGFR4-G388 variant allele supports pY-signal transducer and activator of transcription (STAT3) responses. In contrast, expression of the minor FGFR4-R388 allele enhances STAT3 serine phosphorylation, driving cellular growth. In addition, FGFR4-R388 enhances glucocorticoid receptor phosphorylation and nuclear translocation. Consistent with these findings, glucocorticoid administration resulted in enhanced hormone negative feedback in mice with knock-in of the FGFR4 variant allele. Moreover, clinical data from patients with pituitary tumors revealed that those homozygous for the R388 allele have a higher frequency of silent corticotroph macroadenomas than FGFR4-G388 carriers, who were more likely to have small but hormonally active microadenomas. These findings demonstrate that the FGFR4 transmembrane polymorphic variants can modulate cellular growth and sensitivity to glucocorticoid hormone negative feedback through distinct STAT3 modifications of relevance to the human forms of Cushing disease.
Collapse
Affiliation(s)
- Tae Nakano-Tateno
- Departments of Medicine (T.N.-T., T.T., M.M.H., L.Z., S.E.) and Laboratory Medicine and Pathobiology (T.N.-T., T.T., S.L.A.), University of Toronto, Toronto, Ontario, Canada M5S 2J7; The Endocrine Oncology Site Group (T.N.-T., T.T., S.L.A., S.E.), Princess Margaret Hospital, Toronto, Ontario, Canada M5T 2M9; Ontario Cancer Institute (T.N.-T., T.T., S.L.A., S.E.), University Health Network, Toronto, Ontario, Canada M5G-1X5; Department of Medical Pharmacology (K.Y.), Institute of Health Biosciences, The University of Tokushima Graduate School, Tokushima, 770-0855, Japan; and Hypothalamic and Pituitary Surgery (S.Y.), Toranomon Hospital, Tokyo 105-0001, Japan
| | | | | | | | | | | | | | | |
Collapse
|
47
|
Ye Y, Wang M, Hu S, Shi Y, Zhang X, Zhou Y, Zhao C, Wang G, Wen J, Zong H. Hypoxia-inducible factor-1α C1772T polymorphism and cancer risk: a meta-analysis including 18,334 subjects. Cancer Invest 2014; 32:126-35. [PMID: 24576222 DOI: 10.3109/07357907.2014.883527] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Studies on HIF 1α C1772T (P582S) polymorphism revealed a genetic susceptibility to malignant tumors, however, the results were conflicting. We conducted a meta-analysis utilizing 29 eligible case-control studies to analyze the data concerning the association between the HIF-1α C1772T polymorphism and cancer risks. There was statistical association between the HIF-1α CT/TT genotype and cancer risk (OR = 1.28, 95% CI = 1.06-1.54, P(heterogeneity) < .00001). The stability of these observations was confirmed by a one-way sensitivity analysis. Our findings suggested that CT/TT genotype was associated with increased risks of prostate cancer. Besides, the HIF-1α C1772T polymorphism most likely contributes to susceptibility to malignant tumors, especially in American population.
Collapse
Affiliation(s)
- Yanwei Ye
- 1Department of Gastrointestinal Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Fibroblast growth factor receptor 4 (FGFR4): a targetable regulator of drug resistance in colorectal cancer. Cell Death Dis 2014; 5:e1046. [PMID: 24503538 PMCID: PMC3944229 DOI: 10.1038/cddis.2014.10] [Citation(s) in RCA: 73] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2013] [Revised: 12/21/2013] [Accepted: 12/23/2013] [Indexed: 12/18/2022]
Abstract
The discovery of underlying mechanisms of drug resistance, and the development of novel agents to target these pathways, is a priority for patients with advanced colorectal cancer (CRC). We previously undertook a systems biology approach to design a functional genomic screen and identified fibroblast growth factor receptor 4 (FGFR4) as a potential mediator of drug resistance. The aim of this study was to examine the role of FGFR4 in drug resistance using RNAi and the small-molecule inhibitor BGJ398 (Novartis). We found that FGFR4 is highly expressed at the RNA and protein levels in colon cancer tumour tissue compared with normal colonic mucosa and other tumours. Silencing of FGFR4 reduced cell viability in a panel of colon cancer cell lines and increased caspase-dependent apoptosis. A synergistic interaction was also observed between FGFR4 silencing and 5-fluorouracil (5-FU) and oxaliplatin chemotherapy in colon cancer cell lines. Mechanistically, FGFR4 silencing decreased activity of the pro-survival STAT3 transcription factor and expression of the anti-apoptotic protein c-FLIP. Furthermore, silencing of STAT3 resulted in downregulation of c-FLIP protein expression, suggesting that FGFR4 may regulate c-FLIP expression via STAT3. A similar phenotype and downstream pathway changes were observed following FGFR4 silencing in cell lines resistant to 5-FU, oxaliplatin and SN38 and upon exposure of parental cells to the FGFR small-molecule inhibitor BGJ398. Our results indicate that FGFR4 is a targetable regulator of chemo-resistance in CRC, and hence inhibiting FGFR4 in combination with 5-FU and oxaliplatin is a potential therapeutic strategy for this disease.
Collapse
|
49
|
Abstract
The fibroblast growth factor receptors (FGFRs) regulate important biological processes including cell proliferation and differentiation during development and tissue repair. Over the past decades, numerous pathological conditions and developmental syndromes have emerged as a consequence of deregulation in the FGFRs signaling network. This review aims to provide an overview of FGFR family, their complex signaling pathways in tumorigenesis, and the current development and application of therapeutics targeting the FGFRs signaling for treatment of refractory human cancers.
Collapse
Affiliation(s)
- Kai Hung Tiong
- School of Postgraduate Studies and Research, International Medical University, Bukit Jalil, 57000 Kuala Lumpur, Malaysia
| | - Li Yen Mah
- School of Pharmacy, International Medical University, Bukit Jalil, 57000 Kuala Lumpur, Malaysia
- Center for Cancer and Stem Cell Research, International Medical University, 126 Jalan 19/155B, Bukit Jalil, 57000 Kuala Lumpur, Malaysia
| | - Chee-Onn Leong
- School of Pharmacy, International Medical University, Bukit Jalil, 57000 Kuala Lumpur, Malaysia
- Center for Cancer and Stem Cell Research, International Medical University, 126 Jalan 19/155B, Bukit Jalil, 57000 Kuala Lumpur, Malaysia
| |
Collapse
|
50
|
Ye YW, Hu S, Shi YQ, Zhang XF, Zhou Y, Zhao CL, Wang GJ, Wen JG, Zong H. Combination of the FGFR4 inhibitor PD173074 and 5-fluorouracil reduces proliferation and promotes apoptosis in gastric cancer. Oncol Rep 2013; 30:2777-84. [PMID: 24126887 DOI: 10.3892/or.2013.2796] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2013] [Accepted: 09/20/2013] [Indexed: 11/06/2022] Open
Abstract
Our previous findings revealed that FGFR4 may be a novel therapeutic target for gastric cancer. The aim of the present study was to explore the effects of a combination of PD173074 (PD) and 5-fluorouracil (5-Fu) on the biological behavior of gastric cancer cell lines and the relevant mechanisms involved. MKN45, a gastric cancer cell line, was treated with each single agent alone or a combination of FGF19, PD and 5-Fu. Then, a series of functional assays were performed using CCK-8 assay and flow cytometry. Western blot analysis was used to determine the expression of signaling pathway and downstream-related molecules in the MKN45 cells following the different treatments. As the concentration of PD and 5-Fu increased, the cell viability gradually decreased; the viability of the combination group was less than the viability following single administration. Western blot analysis showed that FGFR4 expression was weak in the 5-Fu-treated groups when compared with the control. PD markedly increased the apoptosis rate of MKN45 cells when compared to the control; the apoptosis rate in the cells treated with the combination of PD and 5-Fu was higher than that in the cells following single treatment. Furthermore, PD reduced the expression of p-ERK and Bcl-xl and increased caspase-3 expression. Inhibition of the activity of FGFR4 may be the main mechanisms of PD effect while 5-Fu reduced FGFR4 expression. Furthermore, the effects of the combination of 5-Fu and PD in inhibiting proliferation, increasing apoptosis and arresting cell cycle were superior to these effects following the single agent treatments, suggesting that the two drugs applied in combination may contribute to the effective treatment of gastric cancer.
Collapse
Affiliation(s)
- Yan-Wei Ye
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, P.R. China
| | | | | | | | | | | | | | | | | |
Collapse
|