1
|
Ballato M, Germanà E, Ricciardi G, Giordano WG, Tralongo P, Buccarelli M, Castellani G, Ricci-Vitiani L, D’Alessandris QG, Giuffrè G, Pizzimenti C, Fiorentino V, Zuccalà V, Ieni A, Caffo M, Fadda G, Martini M. Understanding Neovascularization in Glioblastoma: Insights from the Current Literature. Int J Mol Sci 2025; 26:2763. [PMID: 40141406 PMCID: PMC11943220 DOI: 10.3390/ijms26062763] [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: 02/06/2025] [Revised: 03/15/2025] [Accepted: 03/17/2025] [Indexed: 03/28/2025] Open
Abstract
Glioblastomas (GBMs), among the most aggressive and resilient brain tumors, characteristically exhibit high angiogenic potential, leading to the formation of a dense yet aberrant vasculature, both morphologically and functionally. With these premises, numerous expectations were initially placed on anti-angiogenic therapies, soon dashed by their limited efficacy in concretely improving patient outcomes. Neovascularization in GBM soon emerged as a complex, dynamic, and heterogeneous process, hard to manage with the classical standard of care. Growing evidence has revealed the existence of numerous non-canonical strategies of angiogenesis, variously exploited by GBM to meet its ever-increasing metabolic demand and differently involved in tumor progression, recurrence, and escape from treatments. In this review, we provide an accurate description of each neovascularization mode encountered in GBM tumors to date, highlighting the molecular players and signaling cascades primarily involved. We also detail the key architectural and functional aspects characteristic of the GBM vascular compartment because of an intricate crosstalk between the different angiogenic networks. Additionally, we explore the repertoire of emerging therapies against GBM that are currently under study, concluding with a question: faced with such a challenging scenario, could combined therapies, tailored to the patient's genetic signatures, represent an effective game changer?
Collapse
Affiliation(s)
- Mariagiovanna Ballato
- Department of Biomedical, Dental, Morphological and Functional Imaging Sciences, University of Messina, 98125 Messina, Italy; (M.B.); (E.G.); (G.R.); (W.G.G.); (P.T.)
| | - Emanuela Germanà
- Department of Biomedical, Dental, Morphological and Functional Imaging Sciences, University of Messina, 98125 Messina, Italy; (M.B.); (E.G.); (G.R.); (W.G.G.); (P.T.)
| | - Gabriele Ricciardi
- Department of Biomedical, Dental, Morphological and Functional Imaging Sciences, University of Messina, 98125 Messina, Italy; (M.B.); (E.G.); (G.R.); (W.G.G.); (P.T.)
- Istituto Clinico Polispecialistico C.O.T. Cure Ortopediche Traumatologiche s.pa., 98124 Messina, Italy
| | - Walter Giuseppe Giordano
- Department of Biomedical, Dental, Morphological and Functional Imaging Sciences, University of Messina, 98125 Messina, Italy; (M.B.); (E.G.); (G.R.); (W.G.G.); (P.T.)
| | - Pietro Tralongo
- Department of Biomedical, Dental, Morphological and Functional Imaging Sciences, University of Messina, 98125 Messina, Italy; (M.B.); (E.G.); (G.R.); (W.G.G.); (P.T.)
| | - Mariachiara Buccarelli
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, 00161 Rome, Italy; (M.B.); (G.C.); (L.R.-V.)
| | - Giorgia Castellani
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, 00161 Rome, Italy; (M.B.); (G.C.); (L.R.-V.)
| | - Lucia Ricci-Vitiani
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, 00161 Rome, Italy; (M.B.); (G.C.); (L.R.-V.)
| | | | - Giuseppe Giuffrè
- Department of Human Pathology in Adult and Developmental Age “Gaetano Barresi”, University of Messina, 98125 Messina, Italy; (G.G.); (V.F.); (V.Z.); (A.I.); (G.F.)
| | | | - Vincenzo Fiorentino
- Department of Human Pathology in Adult and Developmental Age “Gaetano Barresi”, University of Messina, 98125 Messina, Italy; (G.G.); (V.F.); (V.Z.); (A.I.); (G.F.)
| | - Valeria Zuccalà
- Department of Human Pathology in Adult and Developmental Age “Gaetano Barresi”, University of Messina, 98125 Messina, Italy; (G.G.); (V.F.); (V.Z.); (A.I.); (G.F.)
| | - Antonio Ieni
- Department of Human Pathology in Adult and Developmental Age “Gaetano Barresi”, University of Messina, 98125 Messina, Italy; (G.G.); (V.F.); (V.Z.); (A.I.); (G.F.)
| | - Maria Caffo
- Biomedical and Dental Sciences and Morphofunctional Imaging, Unit of Neurosurgery, University of Messina, 98122 Messina, Italy;
| | - Guido Fadda
- Department of Human Pathology in Adult and Developmental Age “Gaetano Barresi”, University of Messina, 98125 Messina, Italy; (G.G.); (V.F.); (V.Z.); (A.I.); (G.F.)
| | - Maurizio Martini
- Department of Human Pathology in Adult and Developmental Age “Gaetano Barresi”, University of Messina, 98125 Messina, Italy; (G.G.); (V.F.); (V.Z.); (A.I.); (G.F.)
| |
Collapse
|
2
|
Li H, Dong X, Wang L, Wen H, Qi X, Zhang K, Li Y. Genome-wide identification of Fgfr genes and function analysis of Fgfr4 in myoblasts differentiation of Lateolabrax maculatus. Gene 2024; 927:148717. [PMID: 38908457 DOI: 10.1016/j.gene.2024.148717] [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: 02/29/2024] [Revised: 05/29/2024] [Accepted: 06/19/2024] [Indexed: 06/24/2024]
Abstract
Fibroblast growth factor receptors (Fgfrs) are involved in cell proliferation, differentiation, and migration via complex signaling pathways in different tissues. Our previous studies showed that fibroblast growth factor receptor 4 (fgfr4) was detected in the most significant quantitative trait loci (QTL) for growth traits. However, studies focusing on the function of fgfr4 on the growth of bony fish are still limited. In this study, we identified seven fgfr genes in spotted sea bass (Lateolabrax maculatus) genome, namely fgfr1a, fgfr1b, fgfr2, fgfr3, fgfr4, fgfr5a, and fgfr5b. Phylogenetic analysis, syntenic analysis and gene structure analysis were conducted to further support the accuracy of our annotation and classification results. Additionally, fgfr4 showed the highest expression levels among fgfrs during the proliferation and differentiation stages of spotted sea bass myoblasts. To further study the function of fgfr4 in myogenesis, dual-fluorescence in situ hybridization (ISH) assay was conducted, and the results showed co-localization of fgfr4 with marker gene of skeletal muscle satellite cells. By treating differentiating myoblasts cultured in vitro with BLU-554, the mRNA expressions of myogenin (myog) and the numbers of myotubes formed by myoblasts increased significantly compared to negative control group. These results indicated that Fgfr4 inhibits the differentiation of myoblasts in spotted sea bass. Our findings contributed to filling a research gap on fgfr4 in bony fish myogenesis and the theoretical understanding of growth trait regulation of spotted sea bass.
Collapse
Affiliation(s)
- Hao Li
- Key Laboratory of Mariculture, Ministry of Education (KLMME), Ocean University of China, Qingdao 266003, China
| | - Ximeng Dong
- Key Laboratory of Mariculture, Ministry of Education (KLMME), Ocean University of China, Qingdao 266003, China
| | - Lingyu Wang
- Key Laboratory of Mariculture, Ministry of Education (KLMME), Ocean University of China, Qingdao 266003, China
| | - Haishen Wen
- Key Laboratory of Mariculture, Ministry of Education (KLMME), Ocean University of China, Qingdao 266003, China
| | - Xin Qi
- Key Laboratory of Mariculture, Ministry of Education (KLMME), Ocean University of China, Qingdao 266003, China
| | - Kaiqiang Zhang
- Key Laboratory of Mariculture, Ministry of Education (KLMME), Ocean University of China, Qingdao 266003, China
| | - Yun Li
- Key Laboratory of Mariculture, Ministry of Education (KLMME), Ocean University of China, Qingdao 266003, China; Sanya Oceanographic Institution, Ocean University of China, Sanya 572000, China.
| |
Collapse
|
3
|
Peng M, Deng J, Li X. Clinical advances and challenges in targeting FGF/FGFR signaling in lung cancer. Mol Cancer 2024; 23:256. [PMID: 39543657 PMCID: PMC11566285 DOI: 10.1186/s12943-024-02167-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Accepted: 10/31/2024] [Indexed: 11/17/2024] Open
Abstract
Fibroblast growth factors (FGFs) and their receptors regulate numerous cellular processes, such as metabolism and signal transduction, but can also drive tumorigenesis. Specifically, in lung cancer, the overexpression of FGFs, as well as the amplification, mutation and fusion of FGFR genes, are closely linked to the initiation, progression and resistance of the disease, suggesting that targeting FGF/FGFR is an attractive therapeutic strategy for lung cancer treatment. Nintedanib, a multitarget tyrosine kinase inhibitor (TKI) used in combination with docetaxel, has shown some success as a second-line therapy for lung cancer. However, clinical trials evaluating other FGFR inhibitors have yielded mixed results, indicating substantial complexity in targeting aberrant FGF/FGFR signaling. In this review, we describe the aberrations in FGF/FGFR signaling in lung cancer and summarize the clinical efficacy of FGFR inhibitors, such as multitarget TKIs, selective FGFR-TKIs and biological agents. We also discuss various challenges associated with FGFR targeting in lung cancer, including precision patient selection, toxicity and resistance. Finally, we provide perspectives on future directions, namely, developing novel FGFR-targeting drugs, such as FGFR degraders and more specific FGFR-TKIs, adopting combination therapy and targeting FGFs.
Collapse
Affiliation(s)
- Mei Peng
- Department of Pharmacy, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, P. R. China.
| | - Jun Deng
- Department of Pharmacy, Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University), Changsha, Hunan, 410000, P. R. China
| | - Xiangping Li
- Department of Pharmacy, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, P. R. China.
| |
Collapse
|
4
|
Giacomini A, Taranto S, Gazzaroli G, Faletti J, Capoferri D, Marcheselli R, Sciumè M, Presta M, Sacco A, Roccaro AM. The FGF/FGFR/c-Myc axis as a promising therapeutic target in multiple myeloma. J Exp Clin Cancer Res 2024; 43:294. [PMID: 39482742 PMCID: PMC11529022 DOI: 10.1186/s13046-024-03217-2] [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: 08/19/2024] [Accepted: 10/26/2024] [Indexed: 11/03/2024] Open
Abstract
Among blood cancers, multiple myeloma (MM) represents the second most common neoplasm and is characterized by the accumulation and proliferation of monoclonal plasma cells within the bone marrow. Despite the last few decades being characterized by the development of different therapeutic strategies against MM, at present such disease is still considered incurable. Although MM is highly heterogeneous in terms of genetic and molecular subtypes, about 67% of MM cases are associated with abnormal activity of the transcription factor c-Myc, which has so far revealed a protein extremely difficult to target. We have recently demonstrated that activation of fibroblast growth factor (FGF) signaling protects MM cells from oxidative stress-induced apoptosis by stabilizing the oncoprotein c-Myc. Accordingly, secretion of FGF ligands and autocrine activation of FGF receptors (FGFR) is observed in MM cells and FGFR3 genomic alterations represent some 15-20% MM cases and are associated with poor outcome. Thus, FGF/FGFR blockade may represent a promising strategy to indirectly target c-Myc in MM. On this basis, the present review aims at providing an overview of recently explored connections between the FGF/FGFR system and c-Myc oncoprotein, sustaining the therapeutic potential of targeting the FGF/FGFR/c-Myc axis in MM by using inhibitors targeting FGF ligands or FGF receptors. Importantly, the provided findings may represent the rationale for using FDA approved FGFR TK inhibitors (i.e. Pemigatinib, Futibatinib, Erdafitinib) for the treatment of MM patients presenting with an aberrant activation of this axis.
Collapse
Affiliation(s)
- Arianna Giacomini
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy.
| | - Sara Taranto
- Clinical Trial Center, Translational Research and Phase I Unit, ASST Spedali Civili Di Brescia, Brescia, Italy
| | - Giorgia Gazzaroli
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Jessica Faletti
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Davide Capoferri
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Raffaella Marcheselli
- Clinical Trial Center, Translational Research and Phase I Unit, ASST Spedali Civili Di Brescia, Brescia, Italy
| | - Margherita Sciumè
- Clinical Trial Center, Translational Research and Phase I Unit, ASST Spedali Civili Di Brescia, Brescia, Italy
| | - Marco Presta
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Antonio Sacco
- Clinical Trial Center, Translational Research and Phase I Unit, ASST Spedali Civili Di Brescia, Brescia, Italy
| | - Aldo M Roccaro
- Clinical Trial Center, Translational Research and Phase I Unit, ASST Spedali Civili Di Brescia, Brescia, Italy.
| |
Collapse
|
5
|
Jin B, Su G, Zhou X, Xu L, Wang W, Zhou T, Tan Y, Wang S, Li G. Basic Fibroblast Growth Factor Supports the Function of Limbal Niche Cells via the Wnt/β-Catenin Pathway. J Ocul Pharmacol Ther 2024; 40:571-580. [PMID: 39083404 DOI: 10.1089/jop.2024.0042] [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] [Indexed: 08/02/2024] Open
Abstract
Purpose: To test the effects and underlying mechanisms of basic fibroblast growth factor (bFGF) on the limbal niche cell (LNC) function ex vivo. Methods: By using different concentrations of bFGF (0, 4, 8, 12, and 16 ng/mL) and fibroblast growth factor receptor (FGFR) inhibitors, the effects of bFGF on LNC proliferation, expression of stem cell markers, and transcription levels of the β-catenin were investigated. Single-cell RNA sequencing (scRNA-seq) was used to analyze the action and mechanisms of FGFR subtypes and the Wnt/β-catenin pathway during LNC culture. An mature corneal epithelial cell (MCEC)/LNC three-dimensional model was constructed to verify whether bFGF activates the Wnt/β-catenin pathway in LNC by inhibiting FGFR or β-catenin targets. Results: scRNA-seq showed that FGFR1 is the main receptor in LNC, along with the molecules in the Wnt pathway, including WNT2, FZD7, LRP5, LRP6, and β-catenin. The 12 ng/mL bFGF treatment group showed higher LNC proliferation rate and transcription levels of OCT4, SOX2, NANOG, and β-catenin than any other groups (P < 0.001). In the MCEC/LNC co-culture model, MCEC/LNC treated with 12 ng/mL bFGF promoted the aggregation of the spheres than other groups, associated with increased transcription levels of P63α, WNT2, β-catenin, and a decreased transcription level of CK12 (P < 0.001). Wnt/β-catenin inhibitor LF3 treatment reversed the abovementioned effect of bFGF. Conclusions: bFGF could maintain and promote the stemness of LNC via the FGFR1/Wnt2/FZD7/LRP6 axis in a concentration-dependent manner.
Collapse
Affiliation(s)
- Bihui Jin
- Department of Ophthalmology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Guanyu Su
- Department of Ophthalmology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiao Zhou
- Department of Ophthalmology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Lingjuan Xu
- Department of Ophthalmology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Wei Wang
- Department of Ophthalmology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Tianyu Zhou
- Department of Ophthalmology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yongyao Tan
- Department of Ophthalmology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shusheng Wang
- Department of Cell and Molecular Biology & Ophthalmology, Tulane University, New Orleans, Louisiana, USA
| | - Guigang Li
- Department of Ophthalmology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
6
|
Grivas P, Garralda E, Meric-Bernstam F, Mellinghoff IK, Goyal L, Harding JJ, Dees EC, Bahleda R, Azad NS, Karippot A, Kurzrock R, Tabernero J, Kononen J, Ng MC, Mehta R, Uboha NV, Bigot F, Boni V, Bowyer SE, Breder V, Cervantes A, Chan N, Cleary JM, Dhawan M, Eefsen RL, Ewing J, Graham DM, Guren TK, Kim JW, Koynov K, Oh DY, Redman R, Yen CJ, Spetzler D, Roubaudi-Fraschini MC, Nicolas-Metral V, Ait-Sarkouh R, Zanna C, Ennaji A, Pokorska-Bocci A, Flaherty KT. Evaluating Debio 1347 in Patients with FGFR Fusion-Positive Advanced Solid Tumors from the FUZE Multicenter, Open-Label, Phase II Basket Trial. Clin Cancer Res 2024; 30:4572-4583. [PMID: 38771739 PMCID: PMC11707795 DOI: 10.1158/1078-0432.ccr-24-0012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Revised: 02/19/2024] [Accepted: 05/17/2024] [Indexed: 05/23/2024]
Abstract
PURPOSE This multicenter phase II basket trial investigated the efficacy, safety, and pharmacokinetics of Debio 1347, an investigational, oral, highly selective, ATP-competitive, small molecule inhibitor of FGFR1-3, in patients with solid tumors harboring a functional FGFR1-3 fusion. PATIENTS AND METHODS Eligible adults had a previously treated locally advanced (unresectable) or metastatic biliary tract (cohort 1), urothelial (cohort 2), or another histologic cancer type (cohort 3). Debio 1347 was administered at 80 mg once daily, continuously, in 28-day cycles. The primary endpoint was the objective response rate. Secondary endpoints included duration of response, progression-free survival, overall survival, pharmacokinetics, and incidence of adverse events. RESULTS Between March 22, 2019, and January 8, 2020, 63 patients were enrolled and treated, 30 in cohort 1, 4 in cohort 2, and 29 in cohort 3. An unplanned preliminary statistical review showed that the efficacy of Debio 1347 was lower than predicted, and the trial was terminated. In total, 3 of 58 evaluable patients had partial responses, representing an objective response rate of 5%, with a further 26 (45%) having stable disease (≥6 weeks duration). Grade ≥3 treatment-related adverse events occurred in 22 (35%) of 63 patients, with the most common being hyperphosphatemia (13%) and stomatitis (5%). Two patients (3%) discontinued treatment due to adverse events. CONCLUSIONS Debio 1347 had manageable toxicity; however, the efficacy in patients with tumors harboring FGFR fusions did not support further clinical evaluation in this setting. Our transcriptomic-based analysis characterized in detail the incidence and nature of FGFR fusions across solid tumors. See related commentary by Hage Chehade et al., p. 4549.
Collapse
MESH Headings
- Humans
- Female
- Male
- Middle Aged
- Aged
- Neoplasms/drug therapy
- Neoplasms/genetics
- Neoplasms/pathology
- Adult
- Oncogene Proteins, Fusion/genetics
- Aged, 80 and over
- Receptor, Fibroblast Growth Factor, Type 1/genetics
- Receptor, Fibroblast Growth Factor, Type 1/antagonists & inhibitors
- Receptor, Fibroblast Growth Factor, Type 3/genetics
- Receptor, Fibroblast Growth Factor, Type 3/antagonists & inhibitors
- Treatment Outcome
- Protein Kinase Inhibitors/therapeutic use
- Protein Kinase Inhibitors/adverse effects
- Protein Kinase Inhibitors/administration & dosage
Collapse
Affiliation(s)
- Petros Grivas
- Department of Medicine, Division of Hematology/Oncology, University of Washington, Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Elena Garralda
- Medical Oncology Department, Vall d’Hebron Hospital Campus and Vall d’Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - Funda Meric-Bernstam
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Ingo K. Mellinghoff
- Department of Neurology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Lipika Goyal
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA, USA
| | - James J. Harding
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Weill Cornell Medical College, New York, NY, USA
| | - E. Claire Dees
- Lineberger Comprehensive Cancer Center and Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Rastislav Bahleda
- Drug Development Department, Institut Gustave Roussy, Villejuif, France
| | - Nilo S. Azad
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | | | - Razelle Kurzrock
- Center for Personalized Cancer Therapy, University of California San Diego, Moores Cancer Center, La Jolla, CA, USA
| | - Josep Tabernero
- Medical Oncology Department, Vall d’Hebron Hospital Campus and Vall d’Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | | | - Matthew C.H. Ng
- Division of Medical Oncology, National Cancer Centre, Singapore, Singapore
| | - Rutika Mehta
- GI Medical Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL, USA
| | - Nataliya V. Uboha
- Division of Hematology and Oncology, Department of Medicine, University of Wisconsin Carbone Cancer Center, Madison, WI, USA
| | - Frédéric Bigot
- Medical Oncology Department, Institut de Cancérologie de l’Ouest, Angers, France
| | - Valentina Boni
- START Madrid, Centro Integral Oncológico Clara Campal (CIOCC), Madrid, Spain
| | | | - Valeriy Breder
- NN Blokhin National Medical Research Center of Oncology, Ministry of Health, Moscow, Russian Federation
| | - Andrés Cervantes
- Department of Medical Oncology, Hospital Clínico Universitario de Valencia, INCLIVA Biomedical Research Institute, University of Valencia, Valencia, Spain; CIBERONC, Instituto de Salud Carlos III, Madrid, Spain
| | - Nancy Chan
- Rutgers Cancer Institute of New Jersey, New Brunswick, NJ, USA
| | - James M. Cleary
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Mallika Dhawan
- Division of Hematology and Oncology, University of California, San Francisco, CA, USA
| | - Rikke L. Eefsen
- Department of Oncology, Herlev Gentofte Hospital, Herlev, Denmark
| | - James Ewing
- Ironwood Cancer and Research Centers, Scottsdale, AZ, USA
| | - Donna M. Graham
- The Christie NHS Foundation Trust, Manchester, UK; Division of Cancer Sciences, University of Manchester, Manchester, UK
| | - Tormod K. Guren
- Department of Oncology, Oslo University Hospital, Oslo, Norway
| | - Jin Won Kim
- Division of Hematology/Medical Oncology, Department, Department of Internal Medicine, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam, Republic of Korea
| | | | - Do-Youn Oh
- Seoul National University Hospital, Cancer Research Institute, Seoul National University College of Medicine, Integrated Major in Innovative Medical Science, Seoul National University Graduate School, Seoul, South Korea
| | - Rebecca Redman
- Brown Cancer Center, University of Louisville, Louisville, KY, USA
| | - Chia-Jui Yen
- Department of Oncology, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | | | | | | | | | | | | | | | - Keith T. Flaherty
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
7
|
Park JO, Feng YH, Su WC, Oh DY, Keam B, Shen L, Kim SW, Liu X, Liao H, Qing M, Zhang C, Qian J, Tang X, Li P, Triantos S, Sweiti H. Erdafitinib in Asian patients with advanced solid tumors: an open-label, single-arm, phase IIa trial. BMC Cancer 2024; 24:1006. [PMID: 39138436 PMCID: PMC11323360 DOI: 10.1186/s12885-024-12584-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Accepted: 07/01/2024] [Indexed: 08/15/2024] Open
Abstract
BACKGROUND FGFR genomic aberrations occur in approximately 5-10% of human cancers. Erdafitinib has previously demonstrated efficacy and safety in FGFR-altered advanced solid tumors, such as gliomas, thoracic, gastrointestinal, gynecological, and other rare cancers. However, its efficacy and safety in Asian patients remain largely unknown. We conducted a multicenter, open-label, single-arm phase IIa study of erdafitinib to evaluate its efficacy in Asian patients with FGFR-altered advanced cholangiocarcinoma, non-small cell lung cancer (NSCLC), and esophageal cancer. METHODS Patients with pathologically/cytologically confirmed, advanced, or refractory tumors who met molecular and study eligibility criteria received oral erdafitinib 8 mg once daily with an option for pharmacodynamically guided up-titration to 9 mg on a 28-day cycle, except for four NSCLC patients who received erdafitinib 10 mg (7 days on/7 days off) as they were recruited before the protocol amendment. The primary endpoint was investigator-assessed objective response rate per RECIST v1.1. Secondary endpoints included progression-free survival, duration of response, disease control rate, overall survival, safety, and pharmacokinetics. RESULTS Thirty-five patients (cholangiocarcinoma: 22; NSCLC: 12; esophageal cancer: 1) were enrolled. At data cutoff (November 19, 2021), the objective response rate for patients with cholangiocarcinoma was 40.9% (95% CI, 20.7-63.6); the median progression-free survival was 5.6 months (95% CI, 3.6-12.7) and median overall survival was 40.2 months (95% CI, 12.4-not estimable). No patient with RET/FGFR-altered NSCLC achieved objective response and the disease control rate was 25.0% (95% CI, 5.5-57.2%), with three patients with stable disease. The single patient with esophageal cancer achieved partial response. All patients experienced treatment-emergent adverse events, and grade ≥ 3 treatment-emergent adverse events were reported in 22 (62.9%) patients. Hyperphosphatemia was the most frequently reported treatment-emergent adverse event (all-grade, 85.7%). CONCLUSIONS Erdafitinib demonstrated efficacy in a population of Asian patients in selected advanced solid tumors, particularly in those with advanced FGFR-altered cholangiocarcinoma. Treatment was tolerable with no new safety signals. TRIAL REGISTRATION This trial is registered with ClinicalTrials.gov (NCT02699606); study registration (first posted): 04/03/2016.
Collapse
Affiliation(s)
- Joon Oh Park
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center Sungkyunkwan University School of Medicine, Seoul, Korea.
| | - Yin-Hsun Feng
- Division of Hematology and Oncology, Department of Internal Medicine, Chi-Mei Medical Center, Tainan, Taiwan
| | - Wu-Chou Su
- Department of Oncology, National Cheng Kung University Hospital, Tainan, Taiwan
| | - Do-Youn Oh
- Department of Internal Medicine, Seoul National University Hospital, Seoul, Korea
| | - Bhumsuk Keam
- Department of Internal Medicine, Cancer Research Institute, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Korea
| | - Lin Shen
- Department of GI Oncology, Peking University Cancer Hospital & Institute, Beijing, China
| | - Sang-We Kim
- Oncology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Xiufeng Liu
- Qinhuai Medical Zone, Eastern Theater General Hospital of the Chinese PLA, Nanjing, China
| | | | - Min Qing
- Janssen China R&D Center, Shanghai, China
| | | | - Jiaqi Qian
- Janssen China R&D Center, Shanghai, China
| | | | - Peng Li
- Janssen China R&D Center, Shanghai, China
| | | | | |
Collapse
|
8
|
Marin A, Morales F, Walbaum B. Fibroblast growth factor receptor signaling in estrogen receptor-positive breast cancer: mechanisms and role in endocrine resistance. Front Oncol 2024; 14:1406951. [PMID: 39040443 PMCID: PMC11260626 DOI: 10.3389/fonc.2024.1406951] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Accepted: 06/25/2024] [Indexed: 07/24/2024] Open
Abstract
Fibroblast Growth Factor Receptors (FGFRs) play a significant role in Estrogen Receptor-positive (ER+) breast cancer by contributing to tumorigenesis and endocrine resistance. This review explores the structure, signaling pathways, and implications of FGFRs, particularly FGFR1, FGFR2, FGFR3, and FGFR4, in ER+ breast cancer. FGFR1 is frequently amplified, especially in aggressive Luminal B-like tumors, and its amplification is associated with poor prognosis and treatment resistance. The co-amplification of FGFR1 with oncogenes like EIF4EBP1 and NSD3 complicates its role as a standalone oncogenic driver. FGFR2 amplification, though less common, is critical in hormone receptor regulation, driving proliferation and treatment resistance. FGFR3 and FGFR4 also contribute to endocrine resistance through various mechanisms, including the activation of alternate signaling pathways like PI3K/AKT/mTOR and RAS/RAF/MEK/ERK. Endocrine resistance remains a major clinical challenge, with around 70% of breast cancers initially hormone receptor positive. Despite the success of CDK 4/6 inhibitors in combination with endocrine therapy (ET), resistance often develops, necessitating new treatment strategies. FGFR inhibitors have shown potential in preclinical studies, but clinical trials have yielded limited success due to off-target toxicities and lack of predictive biomarkers. Current clinical trials, including those evaluating FGFR inhibitors like erdafitinib, lucitanib, and dovitinib, have demonstrated mixed outcomes, underscoring the complexity of FGFR signaling in breast cancer. The interplay between FGFR and other signaling pathways highlights the need for comprehensive molecular profiling and personalized treatment approaches. Future research should focus on identifying robust biomarkers and developing combination therapies to enhance the efficacy of FGFR-targeted treatments. In conclusion, targeting FGFR signaling in ER+ breast cancer presents both challenges and opportunities. A deeper understanding of the molecular mechanisms and resistance pathways is crucial for the successful integration of FGFR inhibitors into clinical practice, aiming to improve outcomes for patients with endocrine-resistant breast cancer.
Collapse
Affiliation(s)
- Arnaldo Marin
- Doctoral Program in Medical Sciences, Faculty of Medicine, University of Chile, Santiago, Chile
- Oncology Program, Faculty of Medicine, University of Chile, Santiago, Chile
- Department of Basic and Clinical Oncology, Faculty of Medicine, University of Chile, Santiago, Chile
| | - Fernanda Morales
- Department of Basic and Clinical Oncology, Faculty of Medicine, University of Chile, Santiago, Chile
| | - Benjamín Walbaum
- Medical Oncology Department, Hospital Clinic, Barcelona, Spain
- August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain
- Department of Hematology-Oncology, School of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| |
Collapse
|
9
|
Zhao R, Yin F, Fredimoses M, Zhao J, Fu X, Xu B, Liang M, Chen H, Liu K, Lei M, Laster KV, Li Z, Kundu JK, Dong Z, Lee MH. Targeting FGFR1 by β,β-dimethylacrylalkannin suppresses the proliferation of colorectal cancer in cellular and xenograft models. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 129:155612. [PMID: 38669968 DOI: 10.1016/j.phymed.2024.155612] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Revised: 03/05/2024] [Accepted: 04/07/2024] [Indexed: 04/28/2024]
Abstract
BACKGROUND Colorectal cancer (CRC) continues to be a major global health challenge, ranking as a top cause of cancer-related mortality. Alarmingly, the five-year survival rate for CRC patients hovers around a mere 10-30 %. The disruption of fibroblast growth factor receptor (FGFRs) signaling pathways is significantly implicated in the onset and advancement of CRC, presenting a promising target for therapeutic intervention in CRC management. Further investigation is essential to comprehensively elucidate FGFR1's function in CRC and to create potent therapies that specifically target FGFR1. PURPOSE This study aims to demonstrate the oncogenic role of FGFR1 in colorectal cancer and to explore the potential of β,β-dimethylacrylalkannin (β,β-DMAA) as a therapeutic option to inhibit FGFR1. METHODS In this research, we employed a comprehensive suite of techniques including tissue array, kinase profiling, computational docking, knockdown assay to predict and explore the inhibitor of FGFR1. Furthermore, we utilized kinase assay, pull-down, cell proliferation tests, and Patient derived xenograft (PDX) mouse models to further investigate a novel FGFR1 inhibitor and its impact on the growth of CRC. RESULTS In our research, we discovered that FGFR1 protein is markedly upregulated in colorectal cancer tissues, suggesting a significant role in regulating cellular proliferation, particularly in patients with colorectal cancer. Furthermore, we conducted a computational docking, kinase profiling analysis, simulation and identified that β,β-DMAA could directly bind with FGFR1 within ATP binding pocket domain. Cell-based assays confirmed that β,β-DMAA effectively inhibited the proliferation of colon cancer cells and also triggered cell cycle arrest, apoptosis, and altered FGFR1-mediated signaling pathways. Moreover, β,β-DMAA effectively attenuated the development of PDX tumors in mice that were FGFR1-positive, with no notable toxicity observed. In summary, our study highlights the pivotal role of FGFR1 in colorectal cancer, suggesting that inhibiting FGFR1 activity could be a promising strategy for therapeutic intervention. We present strong evidence that targeting FGFR1 with β,β-DMAA is a viable approach for the management of colorectal cancer. Given its low toxicity and high efficacy, β,β-DMAA, as an FGFR1 inhibitor, warrants further investigation in clinical settings for the treatment of FGFR1-positive tumors.
Collapse
Affiliation(s)
- Ran Zhao
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450001, China; China-US (Henan) Hormel Cancer Institute, Zhengzhou 450000, China; Tianjian Laboratory of Advanced Biomedical Sciences, Zhengzhou 450000, China
| | - Fanxiang Yin
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450001, China; China-US (Henan) Hormel Cancer Institute, Zhengzhou 450000, China; Translational Medical Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450000, China
| | | | - Jianhua Zhao
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450001, China; China-US (Henan) Hormel Cancer Institute, Zhengzhou 450000, China
| | - Xiaorong Fu
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450001, China; China-US (Henan) Hormel Cancer Institute, Zhengzhou 450000, China
| | - Beibei Xu
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450001, China; China-US (Henan) Hormel Cancer Institute, Zhengzhou 450000, China
| | - Mengrui Liang
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450001, China; China-US (Henan) Hormel Cancer Institute, Zhengzhou 450000, China
| | - Hanyong Chen
- The Hormel Institute, University of Minnesota, Austin, MN55912, USA
| | - Kangdong Liu
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450001, China; The Collaborative Innovation Center of Henan Province for Cancer Chemoprevention, Zhengzhou 450000, China
| | - Mingjuan Lei
- China-US (Henan) Hormel Cancer Institute, Zhengzhou 450000, China
| | | | - Zhi Li
- Department of General Surgery, the Affiliated Tumor Hospital of Zhengzhou University, Zhengzhou 450000, China
| | - Joydeb Kumar Kundu
- Li Ka Shing Applied Virology Institute, University of Alberta, Edmonton AB T6G 2R3, Canada
| | - Zigang Dong
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450001, China; China-US (Henan) Hormel Cancer Institute, Zhengzhou 450000, China; Tianjian Laboratory of Advanced Biomedical Sciences, Zhengzhou 450000, China.
| | - Mee-Hyun Lee
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450001, China; China-US (Henan) Hormel Cancer Institute, Zhengzhou 450000, China; The Collaborative Innovation Center of Henan Province for Cancer Chemoprevention, Zhengzhou 450000, China; College of Korean Medicine, Dongshin University, Naju 58245, Republic of Korea.
| |
Collapse
|
10
|
Liu Q, Huang J, Yan W, Liu Z, Liu S, Fang W. FGFR families: biological functions and therapeutic interventions in tumors. MedComm (Beijing) 2023; 4:e367. [PMID: 37750089 PMCID: PMC10518040 DOI: 10.1002/mco2.367] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2023] [Revised: 07/28/2023] [Accepted: 08/11/2023] [Indexed: 09/27/2023] Open
Abstract
There are five fibroblast growth factor receptors (FGFRs), namely, FGFR1-FGFR5. When FGFR binds to its ligand, namely, fibroblast growth factor (FGF), it dimerizes and autophosphorylates, thereby activating several key downstream pathways that play an important role in normal physiology, such as the Ras/Raf/mitogen-activated protein kinase kinase/extracellular signal-regulated kinase, phosphoinositide 3-kinase (PI3K)/AKT, phospholipase C gamma/diacylglycerol/protein kinase c, and signal transducer and activator of transcription pathways. Furthermore, as an oncogene, FGFR genetic alterations were found in 7.1% of tumors, and these alterations include gene amplification, gene mutations, gene fusions or rearrangements. Therefore, FGFR amplification, mutations, rearrangements, or fusions are considered as potential biomarkers of FGFR therapeutic response for tyrosine kinase inhibitors (TKIs). However, it is worth noting that with increased use, resistance to TKIs inevitably develops, such as the well-known gatekeeper mutations. Thus, overcoming the development of drug resistance becomes a serious problem. This review mainly outlines the FGFR family functions, related pathways, and therapeutic agents in tumors with the aim of obtaining better outcomes for cancer patients with FGFR changes. The information provided in this review may provide additional therapeutic ideas for tumor patients with FGFR abnormalities.
Collapse
Affiliation(s)
- Qing Liu
- Cancer CenterIntegrated Hospital of Traditional Chinese MedicineSouthern Medical UniversityGuangzhouGuangdongChina
| | - Jiyu Huang
- Cancer CenterIntegrated Hospital of Traditional Chinese MedicineSouthern Medical UniversityGuangzhouGuangdongChina
| | - Weiwei Yan
- Cancer CenterIntegrated Hospital of Traditional Chinese MedicineSouthern Medical UniversityGuangzhouGuangdongChina
| | - Zhen Liu
- Cancer CenterIntegrated Hospital of Traditional Chinese MedicineSouthern Medical UniversityGuangzhouGuangdongChina
- Key Laboratory of Protein Modification and DegradationBasic School of Guangzhou Medical UniversityGuangzhouGuangdongChina
| | - Shu Liu
- Department of Breast SurgeryThe Affiliated Hospital of Guizhou Medical UniversityGuiyangGuizhouChina
| | - Weiyi Fang
- Cancer CenterIntegrated Hospital of Traditional Chinese MedicineSouthern Medical UniversityGuangzhouGuangdongChina
| |
Collapse
|
11
|
Montero-Hidalgo AJ, Pérez-Gómez JM, Martínez-Fuentes AJ, Gómez-Gómez E, Gahete MD, Jiménez-Vacas JM, Luque RM. Alternative splicing in bladder cancer: potential strategies for cancer diagnosis, prognosis, and treatment. WILEY INTERDISCIPLINARY REVIEWS. RNA 2023; 14:e1760. [PMID: 36063028 DOI: 10.1002/wrna.1760] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Revised: 07/25/2022] [Accepted: 08/05/2022] [Indexed: 05/13/2023]
Abstract
Bladder cancer is the most common malignancy of the urinary tract worldwide. The therapeutic options to tackle this disease comprise surgery, intravesical or systemic chemotherapy, and immunotherapy. Unfortunately, a wide number of patients ultimately become resistant to these treatments and develop aggressive metastatic disease, presenting a poor prognosis. Therefore, the identification of novel therapeutic approaches to tackle this devastating pathology is urgently needed. However, a significant limitation is that the progression and drug response of bladder cancer is strongly associated with its intrinsic molecular heterogeneity. In this sense, RNA splicing is recently gaining importance as a critical hallmark of cancer since can have a significant clinical value. In fact, a profound dysregulation of the splicing process has been reported in bladder cancer, especially in the expression of certain key splicing variants and circular RNAs with a potential clinical value as diagnostic/prognostic biomarkers or therapeutic targets in this pathology. Indeed, some authors have already evidenced a profound antitumor effect by targeting some splicing factors (e.g., PTBP1), mRNA splicing variants (e.g., PKM2, HYAL4-v1), and circular RNAs (e.g., circITCH, circMYLK), which illustrates new possibilities to significantly improve the management of this pathology. This review represents the first detailed overview of the splicing process and its alterations in bladder cancer, and highlights opportunities for the development of novel diagnostic/prognostic biomarkers and their clinical potential for the treatment of this devastating cancer type. This article is categorized under: RNA Processing > Splicing Regulation/Alternative Splicing RNA in Disease and Development > RNA in Disease.
Collapse
Affiliation(s)
- Antonio J Montero-Hidalgo
- Maimonides Biomedical Research Institute of Cordoba (IMIBIC), Cordoba, 14004, Spain
- Department of Cell Biology, Physiology and Immunology, University of Cordoba, Cordoba, 14004, Spain
- Reina Sofia University Hospital (HURS), Cordoba, 14004, Spain
- CIBER Physiopathology of Obesity and Nutrition (CIBERobn), Cordoba, 14004, Spain
| | - Jesús M Pérez-Gómez
- Maimonides Biomedical Research Institute of Cordoba (IMIBIC), Cordoba, 14004, Spain
- Department of Cell Biology, Physiology and Immunology, University of Cordoba, Cordoba, 14004, Spain
- Reina Sofia University Hospital (HURS), Cordoba, 14004, Spain
- CIBER Physiopathology of Obesity and Nutrition (CIBERobn), Cordoba, 14004, Spain
| | - Antonio J Martínez-Fuentes
- Maimonides Biomedical Research Institute of Cordoba (IMIBIC), Cordoba, 14004, Spain
- Department of Cell Biology, Physiology and Immunology, University of Cordoba, Cordoba, 14004, Spain
- Reina Sofia University Hospital (HURS), Cordoba, 14004, Spain
- CIBER Physiopathology of Obesity and Nutrition (CIBERobn), Cordoba, 14004, Spain
| | - Enrique Gómez-Gómez
- Maimonides Biomedical Research Institute of Cordoba (IMIBIC), Cordoba, 14004, Spain
- Reina Sofia University Hospital (HURS), Cordoba, 14004, Spain
- Urology Service, HURS/IMIBIC, Cordoba, 14004, Spain
| | - Manuel D Gahete
- Maimonides Biomedical Research Institute of Cordoba (IMIBIC), Cordoba, 14004, Spain
- Department of Cell Biology, Physiology and Immunology, University of Cordoba, Cordoba, 14004, Spain
- Reina Sofia University Hospital (HURS), Cordoba, 14004, Spain
- CIBER Physiopathology of Obesity and Nutrition (CIBERobn), Cordoba, 14004, Spain
| | - Juan M Jiménez-Vacas
- Maimonides Biomedical Research Institute of Cordoba (IMIBIC), Cordoba, 14004, Spain
- Department of Cell Biology, Physiology and Immunology, University of Cordoba, Cordoba, 14004, Spain
- Reina Sofia University Hospital (HURS), Cordoba, 14004, Spain
- CIBER Physiopathology of Obesity and Nutrition (CIBERobn), Cordoba, 14004, Spain
| | - Raúl M Luque
- Maimonides Biomedical Research Institute of Cordoba (IMIBIC), Cordoba, 14004, Spain
- Department of Cell Biology, Physiology and Immunology, University of Cordoba, Cordoba, 14004, Spain
- Reina Sofia University Hospital (HURS), Cordoba, 14004, Spain
- CIBER Physiopathology of Obesity and Nutrition (CIBERobn), Cordoba, 14004, Spain
| |
Collapse
|
12
|
Ruan R, Li L, Li X, Huang C, Zhang Z, Zhong H, Zeng S, Shi Q, Xia Y, Zeng Q, Wen Q, Chen J, Dai X, Xiong J, Xiang X, Lei W, Deng J. Unleashing the potential of combining FGFR inhibitor and immune checkpoint blockade for FGF/FGFR signaling in tumor microenvironment. Mol Cancer 2023; 22:60. [PMID: 36966334 PMCID: PMC10039534 DOI: 10.1186/s12943-023-01761-7] [Citation(s) in RCA: 35] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Accepted: 03/14/2023] [Indexed: 03/27/2023] Open
Abstract
BACKGROUND Fibroblast growth factors (FGFs) and their receptors (FGFRs) play a crucial role in cell fate and angiogenesis, with dysregulation of the signaling axis driving tumorigenesis. Therefore, many studies have targeted FGF/FGFR signaling for cancer therapy and several FGFR inhibitors have promising results in different tumors but treatment efficiency may still be improved. The clinical use of immune checkpoint blockade (ICB) has resulted in sustained remission for patients. MAIN: Although there is limited data linking FGFR inhibitors and immunotherapy, preclinical research suggest that FGF/FGFR signaling is involved in regulating the tumor microenvironment (TME) including immune cells, vasculogenesis, and epithelial-mesenchymal transition (EMT). This raises the possibility that ICB in combination with FGFR-tyrosine kinase inhibitors (FGFR-TKIs) may be feasible for treatment option for patients with dysregulated FGF/FGFR signaling. CONCLUSION Here, we review the role of FGF/FGFR signaling in TME regulation and the potential mechanisms of FGFR-TKI in combination with ICB. In addition, we review clinical data surrounding ICB alone or in combination with FGFR-TKI for the treatment of FGFR-dysregulated tumors, highlighting that FGFR inhibitors may sensitize the response to ICB by impacting various stages of the "cancer-immune cycle".
Collapse
Affiliation(s)
- Ruiwen Ruan
- Department of Oncology, First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province, 330006, China
- Jiangxi Key Laboratory for lndividualized Cancer Therapy, 17 YongwaiStreet, Donghu District, Nanchang, Jiangxi, 330006, China
| | - Li Li
- Department of Oncology, First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province, 330006, China
- Jiangxi Key Laboratory for lndividualized Cancer Therapy, 17 YongwaiStreet, Donghu District, Nanchang, Jiangxi, 330006, China
| | - Xuan Li
- Department of Oncology, First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province, 330006, China
- Jiangxi Key Laboratory for lndividualized Cancer Therapy, 17 YongwaiStreet, Donghu District, Nanchang, Jiangxi, 330006, China
| | - Chunye Huang
- Department of Oncology, First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province, 330006, China
- Jiangxi Key Laboratory for lndividualized Cancer Therapy, 17 YongwaiStreet, Donghu District, Nanchang, Jiangxi, 330006, China
| | - Zhanmin Zhang
- Department of Oncology, First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province, 330006, China
- Jiangxi Key Laboratory for lndividualized Cancer Therapy, 17 YongwaiStreet, Donghu District, Nanchang, Jiangxi, 330006, China
| | - Hongguang Zhong
- Department of Oncology, First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province, 330006, China
- Jiangxi Key Laboratory for lndividualized Cancer Therapy, 17 YongwaiStreet, Donghu District, Nanchang, Jiangxi, 330006, China
| | - Shaocheng Zeng
- Department of Oncology, First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province, 330006, China
- Jiangxi Key Laboratory for lndividualized Cancer Therapy, 17 YongwaiStreet, Donghu District, Nanchang, Jiangxi, 330006, China
| | - Qianqian Shi
- Department of Oncology, First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province, 330006, China
- Jiangxi Key Laboratory for lndividualized Cancer Therapy, 17 YongwaiStreet, Donghu District, Nanchang, Jiangxi, 330006, China
| | - Yang Xia
- Department of Oncology, First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province, 330006, China
- Jiangxi Key Laboratory for lndividualized Cancer Therapy, 17 YongwaiStreet, Donghu District, Nanchang, Jiangxi, 330006, China
| | - Qinru Zeng
- Department of Oncology, First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province, 330006, China
- Jiangxi Key Laboratory for lndividualized Cancer Therapy, 17 YongwaiStreet, Donghu District, Nanchang, Jiangxi, 330006, China
| | - Qin Wen
- Department of Oncology, First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province, 330006, China
- Jiangxi Key Laboratory for lndividualized Cancer Therapy, 17 YongwaiStreet, Donghu District, Nanchang, Jiangxi, 330006, China
| | - Jingyi Chen
- Department of Oncology, First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province, 330006, China
- Jiangxi Key Laboratory for lndividualized Cancer Therapy, 17 YongwaiStreet, Donghu District, Nanchang, Jiangxi, 330006, China
| | - Xiaofeng Dai
- Department of Oncology, First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province, 330006, China
- Jiangxi Key Laboratory for lndividualized Cancer Therapy, 17 YongwaiStreet, Donghu District, Nanchang, Jiangxi, 330006, China
| | - Jianping Xiong
- Department of Oncology, First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province, 330006, China
- Jiangxi Key Laboratory for lndividualized Cancer Therapy, 17 YongwaiStreet, Donghu District, Nanchang, Jiangxi, 330006, China
| | - Xiaojun Xiang
- Department of Oncology, First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province, 330006, China.
- Jiangxi Key Laboratory for lndividualized Cancer Therapy, 17 YongwaiStreet, Donghu District, Nanchang, Jiangxi, 330006, China.
| | - Wan Lei
- Department of Oncology, First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province, 330006, China.
- Jiangxi Key Laboratory for lndividualized Cancer Therapy, 17 YongwaiStreet, Donghu District, Nanchang, Jiangxi, 330006, China.
| | - Jun Deng
- Department of Oncology, First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province, 330006, China.
- Jiangxi Key Laboratory for lndividualized Cancer Therapy, 17 YongwaiStreet, Donghu District, Nanchang, Jiangxi, 330006, China.
| |
Collapse
|
13
|
Joel IY, Sulaimon LA, Idris MO, Adigun TO, Adisa RA, Ademoye TA, Ogunleye MO, Olaniyi TO. Descriptor-free QSAR: effectiveness in screening for putative inhibitors of FGFR1. J Biomol Struct Dyn 2023; 41:2016-2032. [PMID: 35073829 DOI: 10.1080/07391102.2022.2026248] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
The long short-term memory (LSTM) algorithm has provided solutions to the limitations of the descriptors-utilizing QSAR models in drug design. However, the direct application of LSTM remains scarce. The effectiveness of a descriptor-free QSAR (LSTM-SM) in modeling the FGFR1 inhibitors dataset while comparing with two conventional QSAR using descriptors (126 bits Morgan fingerprint and 2 D descriptors respectively) as a baseline model was investigated in this study. The validated descriptor-free QSAR model was thereafter used to screen for active FGFR1 inhibitors in the ChemDiv database and subjected to molecular docking, induced-fit docking, QM-MM optimization, and molecular dynamics simulations to filter for compounds with high binding affinity and suggest the putative mechanism of inhibition and specificity. The LSTM-SM model performed better than conventional QSAR; having accuracy, specificity, and sensitivity of 0.92, model loss of 0.025, and AUC of 0.95. Fifteen thousand compounds were predicted as actives from the ChemDiv database and four compounds were finally selected. Of the four, two showed putatively effective binding interactions with key active site residues. Molecular dynamics simulations on these compounds in complex with the receptor further give insight into the conformational dynamics of each compound bounded to the receptor. The complexes formed are stable and exhibit a similar degree of compactness. Our findings predicted the advent of self-feature extracting machine learning algorithms of compounds, and have provided the possibility of better predictive model quality that is not necessarily limited by compound descriptors. The putative FGFR1 inhibitors, with their mechanism of inhibition and specificity, were elucidated using this approachCommunicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- I Y Joel
- University of Ilorin Molecular Diagnostic and Research Laboratory, Ilorin, Kwara State, Nigeria
| | - L A Sulaimon
- Department of Biochemistry, Faculty of Basic Medical Sciences, College of Medicine University of Lagos, Idi-araba, Lagos, Nigeria
| | - M O Idris
- School of Life Sciences, University of Science and Technology of China, Hefei, China
| | - T O Adigun
- University of Ilorin Molecular Diagnostic and Research Laboratory, Ilorin, Kwara State, Nigeria
| | - R A Adisa
- Department of Biochemistry, Faculty of Basic Medical Sciences, College of Medicine University of Lagos, Idi-araba, Lagos, Nigeria
| | - T A Ademoye
- Department of Biochemistry, Faculty of Basic Medical Sciences, College of Medicine University of Lagos, Idi-araba, Lagos, Nigeria
| | - M O Ogunleye
- Department of Biochemistry, Faculty of Basic Medical Sciences, College of Medicine University of Lagos, Idi-araba, Lagos, Nigeria
| | - T O Olaniyi
- Department of Science Laboratory Technology, Faculty of Science, Oyo State College of Agriculture and Technology, Igbo-ora, Oyo, Nigeria
| |
Collapse
|
14
|
Basu D, Pal R, Sarkar M, Barma S, Halder S, Roy H, Nandi S, Samadder A. To Investigate Growth Factor Receptor Targets and Generate Cancer Targeting Inhibitors. Curr Top Med Chem 2023; 23:2877-2972. [PMID: 38164722 DOI: 10.2174/0115680266261150231110053650] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Revised: 09/20/2023] [Accepted: 10/02/2023] [Indexed: 01/03/2024]
Abstract
Receptor tyrosine kinase (RTK) regulates multiple pathways, including Mitogenactivated protein kinases (MAPKs), PI3/AKT, JAK/STAT pathway, etc. which has a significant role in the progression and metastasis of tumor. As RTK activation regulates numerous essential bodily processes, including cell proliferation and division, RTK dysregulation has been identified in many types of cancers. Targeting RTK is a significant challenge in cancer due to the abnormal upregulation and downregulation of RTK receptors subfamily EGFR, FGFR, PDGFR, VEGFR, and HGFR in the progression of cancer, which is governed by multiple RTK receptor signalling pathways and impacts treatment response and disease progression. In this review, an extensive focus has been carried out on the normal and abnormal signalling pathways of EGFR, FGFR, PDGFR, VEGFR, and HGFR and their association with cancer initiation and progression. These are explored as potential therapeutic cancer targets and therefore, the inhibitors were evaluated alone and merged with additional therapies in clinical trials aimed at combating global cancer.
Collapse
Affiliation(s)
- Debroop Basu
- Cell and Developmental Biology Special, Department of Zoology, University of Kalyani, Kalyani, Nadia, 741235, India
| | - Riya Pal
- Cell and Developmental Biology Special, Department of Zoology, University of Kalyani, Kalyani, Nadia, 741235, IndiaIndia
| | - Maitrayee Sarkar
- Cell and Developmental Biology Special, Department of Zoology, University of Kalyani, Kalyani, Nadia, 741235, India
| | - Soubhik Barma
- Cell and Developmental Biology Special, Department of Zoology, University of Kalyani, Kalyani, Nadia, 741235, India
| | - Sumit Halder
- Cell and Developmental Biology Special, Department of Zoology, University of Kalyani, Kalyani, Nadia, 741235, India
| | - Harekrishna Roy
- Nirmala College of Pharmacy, Vijayawada, Guntur, Andhra Pradesh, India
| | - Sisir Nandi
- Global Institute of Pharmaceutical Education and Research (Affiliated to Uttarakhand Technical University), Kashipur, 244713, India
| | - Asmita Samadder
- Cell and Developmental Biology Special, Department of Zoology, University of Kalyani, Kalyani, Nadia, 741235, India
- Cytogenetics and Molecular Biology Lab., Department of Zoology, University of Kalyani, Kalyani, Nadia, 741235, India
| |
Collapse
|
15
|
FGFR Inhibitors in Cholangiocarcinoma-A Novel Yet Primary Approach: Where Do We Stand Now and Where to Head Next in Targeting This Axis? Cells 2022; 11:cells11233929. [PMID: 36497187 PMCID: PMC9737583 DOI: 10.3390/cells11233929] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Revised: 11/20/2022] [Accepted: 12/01/2022] [Indexed: 12/12/2022] Open
Abstract
Cholangiocarcinomas (CCAs) are rare but aggressive tumours with poor diagnosis and limited treatment options. Molecular targeted therapies became a promising proposal for patients after progression under first-line chemical treatment. In light of an escalating prevalence of CCA, it is crucial to fully comprehend its pathophysiology, aetiology, and possible targets in therapy. Such knowledge would play a pivotal role in searching for new therapeutic approaches concerning diseases' symptoms and their underlying causes. Growing evidence showed that fibroblast growth factor/fibroblast growth factor receptor (FGF/FGFR) pathway dysregulation is involved in a variety of processes during embryonic development and homeostasis as well as tumorigenesis. CCA is known for its close correlation with the FGF/FGFR pathway and targeting this axis has been proposed in treatment guidelines. Bearing in mind the significance of molecular targeted therapies in different neoplasms, it seems most reasonable to move towards intensive research and testing on these in the case of CCA. However, there is still a need for more data covering this topic. Although positive results of many pre-clinical and clinical studies are discussed in this review, many difficulties lie ahead. Furthermore, this review presents up-to-date literature regarding the outcomes of the latest clinical data and discussion over future directions of FGFR-directed therapies in patients with CCA.
Collapse
|
16
|
Uehara Y, Ikeda S, Kim KH, Lim HJ, Adashek JJ, Persha HE, Okamura R, Lee S, Sicklick JK, Kato S, Kurzrock R. Targeting the FGF/FGFR axis and its co-alteration allies. ESMO Open 2022; 7:100647. [PMID: 36455506 PMCID: PMC9808461 DOI: 10.1016/j.esmoop.2022.100647] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Revised: 10/09/2022] [Accepted: 10/24/2022] [Indexed: 12/03/2022] Open
Abstract
BACKGROUND We analyzed the FGF/FGFR and co-alteration cancer landscape, hypothesizing that combination therapy might be useful in the presence of co-drivers. MATERIALS AND METHODS We describe FGF/FGFR-altered pathways, prognosis, and co-alterations [cBioPortal (N = 7574)] and therapeutic outcomes [University of California San Diego Molecular Tumor Board (MTB) (N = 16)]. RESULTS Patients whose cancers harbored FGF/FGFR alterations (N = 1074) versus those without them (N = 6500) had shorter overall survival (OS) (median: 23.1 versus 26.4 months, P = 0.038) (cBioPortal). Only 6.1% (65/1074 patients) had no pathogenic co-alterations accompanying FGF/FGFR axis abnormalities. The most frequently co-altered pathways/genes involved: TP53 (70%); cell cycle (58%); PI3K (55%); and receptor tyrosine kinases and mitogen-activated protein kinase (MAPK) (65%). Harboring alterations in both FGF/FGFR and in the TP53 pathway or in the cell cycle pathway correlated with shorter OS (versus FGF/FGFR-altered without those co-altered signals) (P = 0.0001 and 0.0065). Four of 16 fibroblast growth factor receptor (FGFR) inhibitor-treated patients presented at MTB attained durable partial responses (PRs) (9, 12, 22+, and 52+ months); an additional two, stable disease (SD) of ≥6 months (13+ and 15 months) [clinical benefit rate (SD ≥ 6 months/PR) = 38%]. Importantly, six patients with cyclin pathway co-alterations received the CDK4/6 inhibitor palbociclib (75 mg p.o. 3 weeks on, 1 week off) and the multikinase FGFR inhibitor lenvatinib (10 mg p.o. daily); three (50%) achieved a PR [9 (ovarian), 12 (biliary), and 52+ months (osteosarcoma)]. Palbociclib and lenvatinib were tolerated well. CONCLUSIONS FGF/FGFR alterations portend a poor prognosis and are frequently accompanied by pathogenic co-aberrations. Malignancies harboring co-alterations that activate both cyclin and FGFR pathways can be co-targeted by CDK4/6 and FGFR inhibitors.
Collapse
Affiliation(s)
- Y Uehara
- Department of Precision Cancer Medicine, Center for Innovative Cancer Treatment, Tokyo Medical and Dental University, Tokyo; Department of Thoracic Oncology and Respiratory Medicine, Tokyo Metropolitan Cancer and Infectious Diseases Center, Komagome Hospital, Tokyo, Japan
| | - S Ikeda
- Department of Precision Cancer Medicine, Center for Innovative Cancer Treatment, Tokyo Medical and Dental University, Tokyo; Center for Personalized Cancer Therapy, Division of Hematology/Oncology, Department of Medicine, University of California San Diego, Moores Cancer Center, La Jolla, USA
| | - K H Kim
- Center for Personalized Cancer Therapy, Division of Hematology/Oncology, Department of Medicine, University of California San Diego, Moores Cancer Center, La Jolla, USA; Division of Hematology and Medical Oncology, Department of Internal Medicine, Seoul National University Boramae Medical Center, Seoul
| | - H J Lim
- Center for Personalized Cancer Therapy, Division of Hematology/Oncology, Department of Medicine, University of California San Diego, Moores Cancer Center, La Jolla, USA; Department of Internal Medicine, Veterans Health Service Medical Center, Seoul, Republic of Korea
| | - J J Adashek
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins Hospital, Baltimore
| | - H E Persha
- Purdue University College of Pharmacy, Purdue University, West Lafayette, USA
| | - R Okamura
- Center for Personalized Cancer Therapy, Division of Hematology/Oncology, Department of Medicine, University of California San Diego, Moores Cancer Center, La Jolla, USA; Department of Surgery, Kyoto University Hospital, Kyoto, Japan
| | - S Lee
- Center for Personalized Cancer Therapy, Division of Hematology/Oncology, Department of Medicine, University of California San Diego, Moores Cancer Center, La Jolla, USA
| | - J K Sicklick
- Center for Personalized Cancer Therapy and Division of Surgical Oncology, Department of Surgery, UC San Diego Moores Cancer Center, La Jolla, USA
| | - S Kato
- Center for Personalized Cancer Therapy, Division of Hematology/Oncology, Department of Medicine, University of California San Diego, Moores Cancer Center, La Jolla, USA.
| | - R Kurzrock
- WIN Consortium for Personalized Cancer Therapy, Paris, France; Medical College of Wisconsin Cancer Center and Genome Science and Precision Medicine Center, Milwaukee, USA; University of Nebraska (adjunct), Lincoln, Nebraska
| |
Collapse
|
17
|
Shreenivas AV, Kato S, Hu J, Skefos C, Sicklick J, Kurzrock R. Carcinoma of unknown primary: Molecular tumor board-based therapy. CA Cancer J Clin 2022; 72:510-523. [PMID: 36006378 PMCID: PMC10180180 DOI: 10.3322/caac.21748] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Revised: 05/20/2022] [Accepted: 06/01/2022] [Indexed: 11/18/2022] Open
Affiliation(s)
- Aditya V Shreenivas
- Division of Hematology and Medical Oncology, Medical College of Wisconsin Cancer Center, Milwaukee, Wisconsin, USA
| | - Shumei Kato
- Medical Oncology, Department of Medicine, Moores Cancer Center, University of California-San Diego, San Diego, California, USA
| | - Jingjing Hu
- Department of Pathology, Moores Cancer Center, University of California-San Diego, San Diego, California, USA
| | - Catherine Skefos
- Clinical Cancer Genetics Program, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Jason Sicklick
- Division of Surgical Oncology, Moores Cancer Center, University of California-San Diego, San Diego, California, USA
| | - Razelle Kurzrock
- Division of Hematology and Medical Oncology, Medical College of Wisconsin Cancer Center, Milwaukee, Wisconsin, USA
- Genomic Sciences and Precision Medicine Center, Medical College of Wisconsin Cancer Center, Milwaukee, Wisconsin, USA
| |
Collapse
|
18
|
Sun C, Tian X, Jia Y, Yang M, Li Y, Fernig DG. Functions of exogenous FGF signals in regulation of fibroblast to myofibroblast differentiation and extracellular matrix protein expression. Open Biol 2022; 12:210356. [PMID: 36102060 PMCID: PMC9471990 DOI: 10.1098/rsob.210356] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/05/2022] Open
Abstract
Fibroblasts are widely distributed cells found in most tissues and upon tissue injury, they are able to differentiate into myofibroblasts, which express abundant extracellular matrix (ECM) proteins. Overexpression and unordered organization of ECM proteins cause tissue fibrosis in damaged tissue. Fibroblast growth factor (FGF) family proteins are well known to promote angiogenesis and tissue repair, but their activities in fibroblast differentiation and fibrosis have not been systematically reviewed. Here we summarize the effects of FGFs in fibroblast to myofibroblast differentiation and ECM protein expression and discuss the underlying potential regulatory mechanisms, to provide a basis for the clinical application of recombinant FGF protein drugs in treatment of tissue damage.
Collapse
Affiliation(s)
- Changye Sun
- Henan Key Laboratory of Medical Tissue Regeneration, Xinxiang Medical University, Xinxiang, Henan 453003, People's Republic of China
| | - Xiangqin Tian
- Henan Key Laboratory of Medical Tissue Regeneration, Xinxiang Medical University, Xinxiang, Henan 453003, People's Republic of China
| | - Yangyang Jia
- Henan Key Laboratory of Medical Tissue Regeneration, Xinxiang Medical University, Xinxiang, Henan 453003, People's Republic of China
| | - Mingming Yang
- Department of Cardiology, Affiliated Zhongda Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu 210009, People's Republic of China
| | - Yong Li
- Department of Biochemistry, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool L69 7ZB, UK
| | - David G Fernig
- Department of Biochemistry, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool L69 7ZB, UK
| |
Collapse
|
19
|
Cho HW, Jin HS, Eom YB. FGFRL1 and FGF genes are associated with height, hypertension, and osteoporosis. PLoS One 2022; 17:e0273237. [PMID: 35980984 PMCID: PMC9387819 DOI: 10.1371/journal.pone.0273237] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Accepted: 08/04/2022] [Indexed: 11/21/2022] Open
Abstract
Hypertension and osteoporosis are two major disorders, which interact with each other. Specific genetic signals involving the fibroblast growth factor receptor-like 1 (FGFRL1) gene are related to high blood pressure and bone growth in giraffes. FGFRL1 is associated with cardiovascular system and bone formation. We performed an association study to investigate the role of FGFRL1 in hypertension, osteoporosis, and height determination in humans. In addition, we identified three kinds of phenotypes in fibroblast growth factor (FGF) genes and examined their association with the FGFRL1 gene. We identified 42 SNPs in the FGFRL1 gene associated with each trait. We then analyzed the potential functional annotation of each SNP. The FGFRL1 gene was found to be associated with height, hypertension, and osteoporosis, consistent with the results of a previous study. In addition, the FGF2, FGF4, FGF10, FGF18, and FGF22 genes were found to interact with the FGFRL1 gene. Our study suggests that both FGFRL1 and FGFRL1-related genes may determine the height and the prevalence of osteoporosis and hypertension in the Korean population.
Collapse
Affiliation(s)
- Hye-Won Cho
- Department of Medical Sciences, Graduate School, Soonchunhyang University, Asan, Chungnam, Republic of Korea
| | - Hyun-Seok Jin
- Department of Biomedical Laboratory Science, College of Life and Health Sciences, Hoseo University, Asan, Chungnam, Republic of Korea
| | - Yong-Bin Eom
- Department of Medical Sciences, Graduate School, Soonchunhyang University, Asan, Chungnam, Republic of Korea
- Department of Biomedical Laboratory Science, College of Medical Sciences, Soonchunhyang University, Asan, Chungnam, Republic of Korea
- * E-mail:
| |
Collapse
|
20
|
Laminin matrix regulates beta-cell FGFR5 expression to enhance glucose-stimulated metabolism. Sci Rep 2022; 12:6110. [PMID: 35414066 PMCID: PMC9005713 DOI: 10.1038/s41598-022-09804-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Accepted: 03/21/2022] [Indexed: 12/24/2022] Open
Abstract
We previously showed that pancreatic beta-cells plated on laminin matrix express reduced levels of FGFR1, a receptor linked to beta-cell metabolism and differentiation. Due to recent evidence that adult beta-cells also express FGFR5, a co-receptor for FGFR1, we now aim to determine the effect of laminin on FGFR5 expression and consequent effects on beta-cell metabolism. Using a genetically encoded sensor for NADPH/NADP+ redox state (Apollo-NADP+), we show overexpression of FGFR5 enhances glucose-stimulated NADPH metabolism in beta-cell lines as well as mouse and human beta-cells. This enhanced response was accompanied by increased insulin secretion as well as increased expression of transcripts for glycolytic enzymes (Glucokinase/GCK, PKM2) and the functional maturity marker Urocortin 3 (UCN3). Culturing beta-cells on laminin matrix also stimulated upregulation of endogenous FGFR5 expression, and similarly enhanced beta-cell glucose-stimulated NADPH-metabolism as well as GCK and PKM2 transcript expression. The metabolism and transcript responses triggered by laminin were disrupted by R5ΔC, a truncated receptor isoform that inhibits the FGFR5/FGFR1 signaling complex. Collectively these data reveal that beta-cells respond to laminin by increasing FGFR5 expression to enhance beta-cell glucose metabolism.
Collapse
|
21
|
Szklener K, Chmiel P, Michalski A, Mańdziuk S. New Directions and Challenges in Targeted Therapies of Advanced Bladder Cancer: The Role of FGFR Inhibitors. Cancers (Basel) 2022; 14:1416. [PMID: 35326568 PMCID: PMC8946699 DOI: 10.3390/cancers14061416] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2022] [Revised: 03/07/2022] [Accepted: 03/08/2022] [Indexed: 12/14/2022] Open
Abstract
Bladder neoplasms, including the most common urothelial carcinoma, have been an escalating problem for years, especially in highly developed countries. Recent decades have brought us a steadily growing share of this cancer in terms of both morbidity and mortality statistics. Bladder neoplasms are not only a therapeutic challenge but also an economical one due to the demanding, costly diagnostics and treatment. The treatment of urothelial cancer can be divided depending on the stage and advancement; thus, we can distinguish three main categories: non-muscle invasive bladder cancer, conventionally treated by surgical interventions; muscle invasive bladder cancer, conventionally treated with chemotherapeutics; and advanced bladder cancer with distant metastases, conventionally treated with the intensive chemotherapy in the MVAC scheme (methotrexate, vinblastine, doxorubicin, and cisplatin). Recent years have brought a breakthrough: immunotherapy and targeted therapy were discovered to be beneficial for patients disqualified from chemotherapy or patients who progressed despite treatment. This literature review summarizes the latest research into the use of targeted therapy in the treatment of advanced bladder cancer, its benefits, and its limitations.
Collapse
Affiliation(s)
- Katarzyna Szklener
- Department of Clinical Oncology and Chemotherapy, Medical University of Lublin, 8 Jaczewskiego Street, 20-090 Lublin, Poland; (P.C.); (A.M.); (S.M.)
| | | | | | | |
Collapse
|
22
|
Hindle A, Bose C, Lee J, Palade PT, Peterson CJ, Reddy PH, Awasthi S, Singh SP. Rlip Depletion Alters Oncogene Transcription at Multiple Distinct Regulatory Levels. Cancers (Basel) 2022; 14:cancers14030527. [PMID: 35158795 PMCID: PMC8833773 DOI: 10.3390/cancers14030527] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 01/07/2022] [Accepted: 01/15/2022] [Indexed: 11/16/2022] Open
Abstract
Simple Summary Rlip76 is a multifunctional membrane protein that facilitates cancer growth, and its depletion kills cancer cells. We recently found that Rlip depletion also results in broad changes to oncogene and tumor suppressor transcription. The present studies were designed to decipher the unknown downstream signaling pathways and transcriptional regulatory mechanisms driving the effect. Building on prior findings that Rlip depletion induces broad methylomic changes, we found using bioluminescence reporter assays that depletion of Rlip also exerts transcriptional control over several cancer genes through methylation-independent changes in transcription factor-mediated activation of their promoter regions and through additional as yet unidentified mechanisms. These findings have important implications for Rlip-targeted cancer therapy. Abstract Rlip76 (Rlip) is a multifunctional membrane protein that facilitates the high metabolic rates of cancer cells through the efflux of toxic metabolites and other functions. Rlip inhibition or depletion results in broad-spectrum anti-cancer effects in vitro and in vivo. Rlip depletion effectively suppresses malignancy and causes global reversion of characteristic CpG island methylomic and transcriptomic aberrations in the p53-null mouse model of spontaneous carcinogenesis through incompletely defined signaling and transcriptomic mechanisms. The methylome and transcriptome are normally regulated by the concerted actions of several mechanisms that include chromatin remodeling, promoter methylation, transcription factor interactions, and miRNAs. The present studies investigated the interaction of Rlip depletion or inhibition with the promoter methylation and transcription of selected cancer-related genes identified as being affected by Rlip depletion in our previous studies. We constructed novel promoter CpG island/luciferase reporter plasmids that respond only to CpG methylation and transcription factors. We found that Rlip depletion regulated expression by a transcription factor-based mechanism that functioned independently of promoter CpG methylation, lipid peroxidation, and p53 status.
Collapse
Affiliation(s)
- Ashly Hindle
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA; (A.H.); (C.B.); (J.L.); (C.J.P.); (P.H.R.)
| | - Chhanda Bose
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA; (A.H.); (C.B.); (J.L.); (C.J.P.); (P.H.R.)
| | - Jihyun Lee
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA; (A.H.); (C.B.); (J.L.); (C.J.P.); (P.H.R.)
- Division of Hematology & Oncology, Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
| | - Philip T. Palade
- Department of Pharmacology and Toxicology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA;
| | - Christopher J. Peterson
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA; (A.H.); (C.B.); (J.L.); (C.J.P.); (P.H.R.)
| | - P. Hemachandra Reddy
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA; (A.H.); (C.B.); (J.L.); (C.J.P.); (P.H.R.)
- Department of Pharmacology and Neuroscience, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
- Department of Public Health, Graduate School of Biomedical Sciences, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
- Department of Neurology, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
- Department of Speech, Language and Hearing Sciences, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
| | - Sanjay Awasthi
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA; (A.H.); (C.B.); (J.L.); (C.J.P.); (P.H.R.)
- Division of Hematology & Oncology, Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
- UMC Cancer Center, UMC Health System, Lubbock, TX 79415, USA
- Correspondence: (S.A.); (S.P.S.); Tel.: +1-806-743-3543 (S.A.); +1-806-743-1540 (S.P.S.)
| | - Sharda P. Singh
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA; (A.H.); (C.B.); (J.L.); (C.J.P.); (P.H.R.)
- Division of Hematology & Oncology, Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
- Correspondence: (S.A.); (S.P.S.); Tel.: +1-806-743-3543 (S.A.); +1-806-743-1540 (S.P.S.)
| |
Collapse
|
23
|
Yu L, Toriseva M, Afshan S, Cangiano M, Fey V, Erickson A, Seikkula H, Alanen K, Taimen P, Ettala O, Nurmi M, Boström PJ, Kallajoki M, Tuomela J, Mirtti T, Beumer IJ, Nees M, Härkönen P. Increased Expression and Altered Cellular Localization of Fibroblast Growth Factor Receptor-Like 1 (FGFRL1) Are Associated with Prostate Cancer Progression. Cancers (Basel) 2022; 14:cancers14020278. [PMID: 35053442 PMCID: PMC8796033 DOI: 10.3390/cancers14020278] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Revised: 12/28/2021] [Accepted: 12/29/2021] [Indexed: 02/01/2023] Open
Abstract
Simple Summary Prostate cancer (PCa) is one of the most frequently diagnosed malignancies in men. PCa is primarily regulated by androgens, but other mechanisms, such as fibroblast growth factor receptor (FGFR) signaling, are also involved. In some patients, PCa relapses after surgical removal of prostate, and androgen deprivation therapy (ADT) is used as the first-line treatment. Unfortunately, the patients often lose response to ADT and progress by other mechanisms to castration-resistant, currently non-curable PCa. In our study, we aimed to identify better diagnostic markers and therapeutic targets against PCa. We analyzed patient PCa tissue samples from radical prostatectomies and biopsies, and used physiologically relevant 3D organoids and mouse xenografts to study FGFR signaling in PCa. We found that FGFRL1, a protein belonging to the FGFR family, plays a role in PCa. Our results suggest that FGFRL1 has significant effects on PCa progression and has potential as a prognostic biomarker. Abstract Fibroblast growth factor receptors (FGFRs) 1–4 are involved in prostate cancer (PCa) regulation, but the role of FGFR-like 1 (FGFRL1) in PCa is unclear. FGFRL1 expression was studied by qRT-PCR and immunohistochemistry of patient tissue microarrays (TMAs) and correlated with clinical patient data. The effects of FGFRL1 knockdown (KD) in PC3M were studied in in vitro culture models and in mouse xenograft tumors. Our results showed that FGFRL1 was significantly upregulated in PCa. The level of membranous FGFRL1 was negatively associated with high Gleason scores (GSs) and Ki67, while increased cytoplasmic and nuclear FGFRL1 showed a positive correlation. Cox regression analysis indicated that nuclear FGFRL1 was an independent prognostic marker for biochemical recurrence after radical prostatectomy. Functional studies indicated that FGFRL1-KD in PC3M cells increases FGFR signaling, whereas FGFRL1 overexpression attenuates it, supporting decoy receptor actions of membrane-localized FGFRL1. In accordance with clinical data, FGFRL1-KD markedly suppressed PC3M xenograft growth. Transcriptomics of FGFRL1-KD cells and xenografts revealed major changes in genes regulating differentiation, ECM turnover, and tumor–stromal interactions associated with decreased growth in FGFRL1-KD xenografts. Our results suggest that FGFRL1 upregulation and altered cellular compartmentalization contribute to PCa progression. The nuclear FGFRL1 could serve as a prognostic marker for PCa patients.
Collapse
Affiliation(s)
- Lan Yu
- Institute of Biomedicine and FICAN West Cancer Centre, University of Turku and Turku University Hospital, 20520 Turku, Finland; (L.Y.); (M.T.); (S.A.); (V.F.); (P.T.); (M.N.)
| | - Mervi Toriseva
- Institute of Biomedicine and FICAN West Cancer Centre, University of Turku and Turku University Hospital, 20520 Turku, Finland; (L.Y.); (M.T.); (S.A.); (V.F.); (P.T.); (M.N.)
| | - Syeda Afshan
- Institute of Biomedicine and FICAN West Cancer Centre, University of Turku and Turku University Hospital, 20520 Turku, Finland; (L.Y.); (M.T.); (S.A.); (V.F.); (P.T.); (M.N.)
| | - Mario Cangiano
- GenomeScan, 2333 BZ Leiden, The Netherlands; (M.C.); (I.J.B.)
| | - Vidal Fey
- Institute of Biomedicine and FICAN West Cancer Centre, University of Turku and Turku University Hospital, 20520 Turku, Finland; (L.Y.); (M.T.); (S.A.); (V.F.); (P.T.); (M.N.)
| | - Andrew Erickson
- Nuffield Department of Surgical Sciences, University of Oxford, Oxford 0X3 9DU, UK;
| | - Heikki Seikkula
- Department of Urology, University of Turku and Turku University Hospital, 20520 Turku, Finland; (H.S.); (O.E.); (M.N.); (P.J.B.)
| | - Kalle Alanen
- Department of Pathology, Turku University Hospital, 20520 Turku, Finland; (K.A.); (M.K.)
| | - Pekka Taimen
- Institute of Biomedicine and FICAN West Cancer Centre, University of Turku and Turku University Hospital, 20520 Turku, Finland; (L.Y.); (M.T.); (S.A.); (V.F.); (P.T.); (M.N.)
- Department of Pathology, Turku University Hospital, 20520 Turku, Finland; (K.A.); (M.K.)
| | - Otto Ettala
- Department of Urology, University of Turku and Turku University Hospital, 20520 Turku, Finland; (H.S.); (O.E.); (M.N.); (P.J.B.)
| | - Martti Nurmi
- Department of Urology, University of Turku and Turku University Hospital, 20520 Turku, Finland; (H.S.); (O.E.); (M.N.); (P.J.B.)
| | - Peter J. Boström
- Department of Urology, University of Turku and Turku University Hospital, 20520 Turku, Finland; (H.S.); (O.E.); (M.N.); (P.J.B.)
| | - Markku Kallajoki
- Department of Pathology, Turku University Hospital, 20520 Turku, Finland; (K.A.); (M.K.)
| | - Johanna Tuomela
- Institute of Biomedicine and FICAN West Cancer Centre, University of Turku and Turku University Hospital, 20520 Turku, Finland; (L.Y.); (M.T.); (S.A.); (V.F.); (P.T.); (M.N.)
| | - Tuomas Mirtti
- HUS Diagnostic Center and Research Program in Systems Oncology (ONCOSYS), Helsinki University Hospital and University of Helsinki, 00014 Helsinki, Finland;
| | - Inès J. Beumer
- GenomeScan, 2333 BZ Leiden, The Netherlands; (M.C.); (I.J.B.)
| | - Matthias Nees
- Institute of Biomedicine and FICAN West Cancer Centre, University of Turku and Turku University Hospital, 20520 Turku, Finland; (L.Y.); (M.T.); (S.A.); (V.F.); (P.T.); (M.N.)
- Department of Biochemistry and Molecular Biology, Medical University in Lublin, 20-093 Lublin, Poland
| | - Pirkko Härkönen
- Institute of Biomedicine and FICAN West Cancer Centre, University of Turku and Turku University Hospital, 20520 Turku, Finland; (L.Y.); (M.T.); (S.A.); (V.F.); (P.T.); (M.N.)
- Correspondence: ; Tel.: +358-40-7343520
| |
Collapse
|
24
|
Yang J, Sontag D, Kung S, Minuk GY. Fibroblast Growth Factor 19 Induced Changes in Non-malignant Cholangiocytes. J Clin Transl Hepatol 2021; 9:909-916. [PMID: 34966654 PMCID: PMC8666367 DOI: 10.14218/jcth.2021.00087] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Revised: 04/21/2021] [Accepted: 05/10/2021] [Indexed: 12/04/2022] Open
Abstract
BACKGROUND AND AIMS Fibroblast growth factor (FGF)19 has been implicated in the pathogenesis of murine hepatocellular carcinoma. Whether it plays a role in the development or course of human cholangiocarcinoma remains to be determined. The aim of this study was to determine whether prolonged exposure to FGF19 results in the transformation of non-malignant human cholangiocytes into cells with malignant features. METHODS Human SV-40 transfected non-malignant H69 cholangiocytes were cultured with FGF19 (0-50 ng/mL) for 6 weeks, followed by 6 weeks with medium alone. Cell proliferation, invasion, stem cell surface markers, oncofetoprotein expression, state of differentiation, epithelial-mesenchymal transition (EMT) and interleukin (IL)-6 expression were documented at various time intervals throughout the 12-week period. RESULTS FGF19 exposure was associated with significant increases in cell proliferation, de-differentiation, EMT and IL-6 expression. However, each of these effects returned to baseline or control values during the 6-week FGF19 free follow-up period. The remaining cell properties remained unaltered. CONCLUSIONS Six weeks of FGF19 exposure did not result in the acquisition of permanent malignant features in non-malignant, human cholangiocytes.
Collapse
Affiliation(s)
- Jiaqi Yang
- Section of Hepatology, Department of Medicine, Rady College of Medicine, University of Manitoba, Winnipeg, Manitoba, Canada
| | - David Sontag
- Section of Hepatology, Department of Medicine, Rady College of Medicine, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Sam Kung
- Department of Immunology, Rady College of Medicine, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Gerald Y. Minuk
- Section of Hepatology, Department of Medicine, Rady College of Medicine, University of Manitoba, Winnipeg, Manitoba, Canada
- Department of Pharmacology and Therapeutics, Rady College of Medicine, University of Manitoba, Winnipeg, Manitoba, Canada
- Correspondence to: Gerald Y. Minuk, Morberg Family Chair in Hepatology, University of Manitoba, John Buhler Research Centre, 715 McDermot Ave., Winnipeg, MB R3E 3P4, Canada. ORCID: https://orcid.org/0000-0002-2687-940X. Tel: +1-204-789-3204, Fax: +1-204-789-3987, E-mail:
| |
Collapse
|
25
|
Mahmood HTNA, Tomas Bort E, Walker AJ, Grose RP, Chioni AM. FGF signalling facilitates cervical cancer progression. FEBS J 2021; 289:3440-3456. [PMID: 34951738 DOI: 10.1111/febs.16331] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Revised: 11/26/2021] [Accepted: 12/21/2021] [Indexed: 12/18/2022]
Abstract
Cervical cancer is one of the most frequently diagnosed cancers in women worldwide. While cervical cancer is caused by human papillomavirus (HPV), not all females infected with HPV develop the disease, suggesting that other factors might facilitate its progression. Growing evidence supports the involvement of the fibroblast growth factor receptor (FGFR) axis in several cancers, including gynecological. However, for cervical cancer, the molecular mechanisms that underpin the disease remain poorly understood, including the role of FGFR signaling. The aim of this study was to investigate FGF(R) signaling in cervical cancer through bioinformatic analysis of cell line and patient data and through detailed expression profiling, manipulation of the FGFR axis, and downstream phenotypic analysis in cell lines (HeLa, SiHa, and CaSki). Expression (protein and mRNA) analysis demonstrated that FGFR1b/c, FGFR2b/c, FGFR4, FGF2, FGF4, and FGF7 were expressed in all three lines. Interestingly, FGFR1 and 2 localized to the nucleus, supporting that nuclear FGFRs could act as transcription factors. Importantly, 2D and 3D cell cultures demonstrated that FGFR activation can facilitate cell functions correlated with invasive disease. Collectively, this study supports an association between FGFR signaling and cervical cancer progression, laying the foundations for the development of therapeutic approaches targeting FGFR in this disease.
Collapse
Affiliation(s)
| | - Elena Tomas Bort
- Centre for Tumour Biology, Barts Cancer Institute, Queen Mary University of London, UK
| | - Anthony J Walker
- School of Life Sciences Pharmacy and Chemistry, Kingston University, Kingston upon Thames, UK
| | - Richard P Grose
- Centre for Tumour Biology, Barts Cancer Institute, Queen Mary University of London, UK
| | - Athina-Myrto Chioni
- School of Life Sciences Pharmacy and Chemistry, Kingston University, Kingston upon Thames, UK
| |
Collapse
|
26
|
Chioni AM, Grose RP. Biological Significance and Targeting of the FGFR Axis in Cancer. Cancers (Basel) 2021; 13:5681. [PMID: 34830836 PMCID: PMC8616401 DOI: 10.3390/cancers13225681] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Revised: 11/08/2021] [Accepted: 11/11/2021] [Indexed: 12/15/2022] Open
Abstract
The pleiotropic effects of fibroblast growth factors (FGFs), the widespread expression of all seven signalling FGF receptors (FGFRs) throughout the body, and the dramatic phenotypes shown by many FGF/R knockout mice, highlight the diversity, complexity and functional importance of FGFR signalling. The FGF/R axis is critical during normal tissue development, homeostasis and repair. Therefore, it is not surprising that substantial evidence also pinpoints the involvement of aberrant FGFR signalling in disease, including tumourigenesis. FGFR aberrations in cancer include mutations, gene fusions, and amplifications as well as corrupted autocrine/paracrine loops. Indeed, many clinical trials on cancer are focusing on targeting the FGF/FGFR axis, using selective FGFR inhibitors, nonselective FGFR tyrosine kinase inhibitors, ligand traps, and monoclonal antibodies and some have already been approved for the treatment of cancer patients. The heterogeneous tumour microenvironment and complexity of FGFR signalling may be some of the factors responsible for the resistance or poor response to therapy with FGFR axis-directed therapeutic agents. In the present review we will focus on the structure and function of FGF(R)s, their common irregularities in cancer and the therapeutic value of targeting their function in cancer.
Collapse
Affiliation(s)
- Athina-Myrto Chioni
- School of Life Sciences Pharmacy and Chemistry, Kingston University, Penrhyn Road, Kingston upon Thames KT1 2EE, UK
| | - Richard P. Grose
- Centre for Tumour Biology, Barts Cancer Institute, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK;
| |
Collapse
|
27
|
Prete MG, Cammarota A, D’Alessio A, Zanuso V, Rimassa L. Current options and future directions of systemic therapy for advanced biliary tract cancer. EXPLORATION OF TARGETED ANTI-TUMOR THERAPY 2021; 2:416-433. [PMID: 36045701 PMCID: PMC9400707 DOI: 10.37349/etat.2021.00054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Accepted: 09/09/2021] [Indexed: 11/19/2022] Open
Abstract
Biliary tract cancers (BTCs) are aggressive tumors arising from different portions of the biliary tree and classified according to the anatomical location in intrahepatic (i) cholangiocarcinoma (CCA, iCCA), perihilar CCA (pCCA), and distal CCA (dCCA), gallbladder cancer (GBC), and ampulla of Vater cancer (AVC). Due to their silent behavior, BTCs are frequently diagnosed at advanced stages when the prognosis is poor. The available chemotherapeutic options are palliative and unfortunately, most patients will die from their disease between 6 and 18 months from diagnosis. However, over the last decade, amounting interest has been posed on the genomic landscape of BTCs and deep-sequencing studies have identified different potentially actionable driver mutations. Hence, the promising results of the early phase clinical studies with targeted agents against isocitrate dehydrogenase (IDH) 1 mutation or fibroblast growth factor (FGF) receptor (FGFR) 2 aberrations inintrahepatic tumors, and other agents against humanepidermal growth factor receptor (HER) 2 overexpression/mutations, neurotrophic tyrosine receptor kinase (NTRK) fusions or B-type Raf kinase (BRAF) mutations across different subtypes of BTCs, have paved the way for a "precision medicine" strategy for BTCs. Moreover, despite the modest results when used as monotherapy, beyond microsatellite instability-high (MSI-H) tumors, immune checkpoint inhibitors are being evaluated in combination with platinum-based chemotherapy, possibly further expanding the therapeutic landscape of advanced BTCs. This review aims to provide an overview of the approved systemic therapies, the promising results, and the ongoing studies to explore the current and future directions of advanced BTC systemic treatment.
Collapse
Affiliation(s)
- Maria Giuseppina Prete
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, 20090 Pieve Emanuele, Milan, Italy
- Medical Oncology and Hematology Unit, Humanitas Cancer Center, IRCCS Humanitas Research Hospital, Via Manzoni 56, 20089 Rozzano, Milan, Italy
| | - Antonella Cammarota
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, 20090 Pieve Emanuele, Milan, Italy
- Medical Oncology and Hematology Unit, Humanitas Cancer Center, IRCCS Humanitas Research Hospital, Via Manzoni 56, 20089 Rozzano, Milan, Italy
| | - Antonio D’Alessio
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, 20090 Pieve Emanuele, Milan, Italy
- Medical Oncology and Hematology Unit, Humanitas Cancer Center, IRCCS Humanitas Research Hospital, Via Manzoni 56, 20089 Rozzano, Milan, Italy
| | - Valentina Zanuso
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, 20090 Pieve Emanuele, Milan, Italy
- Medical Oncology and Hematology Unit, Humanitas Cancer Center, IRCCS Humanitas Research Hospital, Via Manzoni 56, 20089 Rozzano, Milan, Italy
| | - Lorenza Rimassa
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, 20090 Pieve Emanuele, Milan, Italy
- Medical Oncology and Hematology Unit, Humanitas Cancer Center, IRCCS Humanitas Research Hospital, Via Manzoni 56, 20089 Rozzano, Milan, Italy
| |
Collapse
|
28
|
Aprajita, Sharma R. Comprehending fibroblast growth factor receptor like 1: Oncogene or tumor suppressor? Cancer Treat Res Commun 2021; 29:100472. [PMID: 34689016 DOI: 10.1016/j.ctarc.2021.100472] [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: 08/07/2021] [Revised: 09/27/2021] [Accepted: 09/29/2021] [Indexed: 12/16/2022]
Abstract
Fibroblast Growth Factor Receptor Like 1 (FGFRL1) signaling has crucial role in a multitude of processes during genetic diseases, embryonic development and various types of cancer. Due to its partial structural similarity with its classical Fibroblast Growth Factor Receptor [FGFR] counterparts and lack of tyrosine kinase domain, FGFRL1 was thought to work as a decoy receptor in FGF/FGFR signaling. Later on, growing number evidences showed that expression of FGFRL1 affects major pathways like ERK1/2, Akt and others, which are dysfunctional in a wide range of human cancers. In this review, we provide an overview of the current understanding of FGFRL1 and its roles in cell differentiation, adhesion and proliferation pathways . Overexpression of FGFRL1 might lead to tumor progression and invasion. In this context, inhibitors for FGFRL1 might have therapeutic benefits in human cancer prognosis.
Collapse
Affiliation(s)
- Aprajita
- University School of Biotechnology, Guru Gobind Singh Indraprastha University, Dwarka, New Delhi, India
| | - Rinu Sharma
- University School of Biotechnology, Guru Gobind Singh Indraprastha University, Dwarka, New Delhi, India.
| |
Collapse
|
29
|
Repetto M, Crimini E, Giugliano F, Morganti S, Belli C, Curigliano G. Selective FGFR/FGF pathway inhibitors: inhibition strategies, clinical activities, resistance mutations, and future directions. Expert Rev Clin Pharmacol 2021; 14:1233-1252. [PMID: 34591728 DOI: 10.1080/17512433.2021.1947246] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Introduction: Fibroblast growth factor receptor (FGFR)/fibroblast growth factor (FGF) is a pathway characterized by recurring alterations in cancer. Its dysregulations enhance cancer cell proliferation, survival, migration and invasion, as well as angiogenesis and immune evasion.Areas covered: FGFR/FGF selective inhibitors belong to a broad class of drugs with some being approved for specific indications and others under investigation in ongoing phase I and II clinical trials. In this review, all available clinical data from trials on selective FGFR/FGF inhibitors as well as described resistance mutations and mechanisms are presented. FGFR/FGF pathway inhibitors are classified according to the mechanism they employ to dampen/suppress signaling and to the preferred FGFR binding mode when X-ray crystal structure is available.Expert opinion: Data presented suggests the general actionability of FGFR1,2,3 mutations and fusions across histologies, whereas FGFR1,2,3 amplifications alone are poor predictors of response to tyrosine kinase inhibitors. Overexpression on immunohistochemistry (IHC) of FGF19, the stimulatory ligand of FGFR4, can predict response to FGFR selective inhibitors in hepatocellular carcinoma. Whereas IHC overexpression of FGFR1,2,3 is not sufficient to predict benefit from FGFR inhibitors across solid tumors. FGFR1,2,3 mRNA overexpression can predict response even in absence of structural alteration. Data on resistance mutations suggests the need for new inhibitors to overcome gatekeeper mutations.
Collapse
Affiliation(s)
- Matteo Repetto
- Division of Early Drug Development for Innovative Therapies, European Institute of Oncology IRCCS, Milan, Italy.,Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | - Edoardo Crimini
- Division of Early Drug Development for Innovative Therapies, European Institute of Oncology IRCCS, Milan, Italy.,Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | - Federica Giugliano
- Division of Early Drug Development for Innovative Therapies, European Institute of Oncology IRCCS, Milan, Italy.,Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | - Stefania Morganti
- Division of Early Drug Development for Innovative Therapies, European Institute of Oncology IRCCS, Milan, Italy.,Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | - Carmen Belli
- Division of Early Drug Development for Innovative Therapies, European Institute of Oncology IRCCS, Milan, Italy
| | - Giuseppe Curigliano
- Division of Early Drug Development for Innovative Therapies, European Institute of Oncology IRCCS, Milan, Italy.,Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| |
Collapse
|
30
|
Surgical Result and Identification of FGFR2 Variants Using Whole-Exome Sequencing in a Chinese Family With Crouzon Syndrome. J Craniofac Surg 2021; 33:134-138. [PMID: 34538793 DOI: 10.1097/scs.0000000000008153] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
ABSTRACT Crouzon syndrome is considered as one of the most common craniosynostosis syndromes with a prevalence of 1 in 65,000 individuals, and has a close relationship with variants in fibroblast growth factor receptor 2. Here the authors described a Crouzon syndrome case, which was asked for surgery treatment for the symptom of multisuture craniosynostosis. Mild midfacial retrusion, larger head circumference, proptosis, pseudo-prognathism, and dental malposition could also be found obviously. Then fronto-orbital advancement and cranial cavity expansion were performed to the child. After whole-exome sequencing (WES) and Sanger sequencing, gene variants in the exons 2 and 3 of FGFR2 were detected. And protein tyrosine 105 replaced by cysteine in the extracellular region of FGFR2 was also detected. After operation, she presented a satisfactory anterior plagiocephaly and scaphocephaly correction, and the result was satisfied by surgeons and her parents. Variants detected using WES have further research prospect.
Collapse
|
31
|
King G, Javle M. FGFR Inhibitors: Clinical Activity and Development in the Treatment of Cholangiocarcinoma. Curr Oncol Rep 2021; 23:108. [PMID: 34269915 DOI: 10.1007/s11912-021-01100-3] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/24/2021] [Indexed: 01/06/2023]
Abstract
PURPOSE OF REVIEW Cholangiocarcinoma is an aggressive cancer with a poor prognosis and limited treatment. Gene sequencing studies have identified genetic alterations in fibroblast growth factor receptor (FGFR) in a significant proportion of cholangiocarcinoma (CCA) patients. This review will discuss the FGFR signaling pathway's role in CCA and highlight the development of therapeutic strategies targeting this pathway. RECENT FINDINGS The development of highly potent and selective FGFR inhibitors has led to the approval of pemigatinib for FGFR2 fusion or rearranged CCA. Other selective FGFR inhibitors are currently under clinical investigation and show promising activity. Despite encouraging results, the emergence of resistance is inevitable. Studies using circulating tumor DNA and on-treatment tissue biopsies have elucidated underlying mechanisms of intrinsic and acquired resistance. There is a critical need to not only develop more effective compounds, but also innovative sequencing strategies and combinations to overcome resistance to selective FGFR inhibition. Therapeutic development of precision medicine for FGFR-altered CCA is a dynamic process of involving a comprehensive understanding of tumor biology, rational clinical trial design, and therapeutic optimization. Alterations in FGFR represent a valid therapeutic target in CCA and selective FGFR inhibitors are treatment options for this patient population.
Collapse
Affiliation(s)
- Gentry King
- Division of Medical Oncology, University of Washington, Seattle, WA, USA.,Seattle Cancer Care Alliance, 825 Eastlake Avenue East, LG-465, Seattle, WA, 98109, USA.,Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Milind Javle
- Department of Gastrointestinal (GI) Medical Oncology, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Unit 0426, Houston, TX, 77030-4009, USA.
| |
Collapse
|
32
|
Azhar M, Altaf S, Uddin I, Cheng J, Wu L, Tong X, Qin W, Bao J. Towards Post-Meiotic Sperm Production: Genetic Insight into Human Infertility from Mouse Models. Int J Biol Sci 2021; 17:2487-2503. [PMID: 34326689 PMCID: PMC8315030 DOI: 10.7150/ijbs.60384] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Accepted: 05/16/2021] [Indexed: 02/06/2023] Open
Abstract
Declined quality and quantity of sperm is currently the major cause of patients suffering from infertility. Male germ cell development is spatiotemporally regulated throughout the whole developmental process. While it has been known that exogenous factors, such as environmental exposure, diet and lifestyle, et al, play causative roles in male infertility, recent progress has revealed abundant genetic mutations tightly associated with defective male germline development. In mammals, male germ cells undergo dramatic morphological change (i.e., nuclear condensation) and chromatin remodeling during post-meiotic haploid germline development, a process termed spermiogenesis; However, the molecular machinery players and functional mechanisms have yet to be identified. To date, accumulated evidence suggests that disruption in any step of haploid germline development is likely manifested as fertility issues with low sperm count, poor sperm motility, aberrant sperm morphology or combined. With the continually declined cost of next-generation sequencing and recent progress of CRISPR/Cas9 technology, growing studies have revealed a vast number of disease-causing genetic variants associated with spermiogenic defects in both mice and humans, along with mechanistic insights partially attained and validated through genetically engineered mouse models (GEMMs). In this review, we mainly summarize genes that are functional at post-meiotic stage. Identification and characterization of deleterious genetic variants should aid in our understanding of germline development, and thereby further improve the diagnosis and treatment of male infertility.
Collapse
Affiliation(s)
- Muhammad Azhar
- Division of Life Sciences and Medicine, The First Affiliated Hospital of USTC, University of Science and Technology of China, Anhui, China
| | - Saba Altaf
- Division of Life Sciences and Medicine, The First Affiliated Hospital of USTC, University of Science and Technology of China, Anhui, China
| | - Islam Uddin
- Division of Life Sciences and Medicine, The First Affiliated Hospital of USTC, University of Science and Technology of China, Anhui, China
| | - Jinbao Cheng
- The 901th hospital of Joint logistics support Force of PLA, Anhui, China
| | - Limin Wu
- Division of Life Sciences and Medicine, The First Affiliated Hospital of USTC, University of Science and Technology of China, Anhui, China
| | - Xianhong Tong
- Division of Life Sciences and Medicine, The First Affiliated Hospital of USTC, University of Science and Technology of China, Anhui, China
| | - Weibing Qin
- NHC Key Laboratory of Male Reproduction and Genetics, Family Planning Research Institute of Guangdong Province, China
| | - Jianqiang Bao
- Division of Life Sciences and Medicine, The First Affiliated Hospital of USTC, University of Science and Technology of China, Anhui, China
| |
Collapse
|
33
|
Margiotta A. All Good Things Must End: Termination of Receptor Tyrosine Kinase Signal. Int J Mol Sci 2021; 22:ijms22126342. [PMID: 34198477 PMCID: PMC8231876 DOI: 10.3390/ijms22126342] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Revised: 06/10/2021] [Accepted: 06/11/2021] [Indexed: 12/28/2022] Open
Abstract
Receptor tyrosine kinases (RTKs) are membrane receptors that regulate many fundamental cellular processes. A tight regulation of RTK signaling is fundamental for development and survival, and an altered signaling by RTKs can cause cancer. RTKs are localized at the plasma membrane (PM) and the major regulatory mechanism of signaling of RTKs is their endocytosis and degradation. In fact, RTKs at the cell surface bind ligands with their extracellular domain, become active, and are rapidly internalized where the temporal extent of signaling, attenuation, and downregulation are modulated. However, other mechanisms of signal attenuation and termination are known. Indeed, inhibition of RTKs’ activity may occur through the modulation of the phosphorylation state of RTKs and the interaction with specific proteins, whereas antagonist ligands can inhibit the biological responses mediated by the receptor. Another mechanism concerns the expression of endogenous inactive receptor variants that are deficient in RTK activity and take part to inactive heterodimers or hetero-oligomers. The downregulation of RTK signals is fundamental for several cellular functions and the homeostasis of the cell. Here, we will review the mechanisms of signal attenuation and termination of RTKs, focusing on FGFRs.
Collapse
Affiliation(s)
- Azzurra Margiotta
- Department of Biology, Faculty of Medicine, Masaryk University, 62500 Brno, Czech Republic;
- International Clinical Research Center, St. Anne’s University Hospital, 65691 Brno, Czech Republic
| |
Collapse
|
34
|
Ferguson HR, Smith MP, Francavilla C. Fibroblast Growth Factor Receptors (FGFRs) and Noncanonical Partners in Cancer Signaling. Cells 2021; 10:1201. [PMID: 34068954 PMCID: PMC8156822 DOI: 10.3390/cells10051201] [Citation(s) in RCA: 68] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Revised: 05/06/2021] [Accepted: 05/09/2021] [Indexed: 02/07/2023] Open
Abstract
Increasing evidence indicates that success of targeted therapies in the treatment of cancer is context-dependent and is influenced by a complex crosstalk between signaling pathways and between cell types in the tumor. The Fibroblast Growth Factor (FGF)/FGF receptor (FGFR) signaling axis highlights the importance of such context-dependent signaling in cancer. Aberrant FGFR signaling has been characterized in almost all cancer types, most commonly non-small cell lung cancer (NSCLC), breast cancer, glioblastoma, prostate cancer and gastrointestinal cancer. This occurs primarily through amplification and over-expression of FGFR1 and FGFR2 resulting in ligand-independent activation. Mutations and translocations of FGFR1-4 are also identified in cancer. Canonical FGF-FGFR signaling is tightly regulated by ligand-receptor combinations as well as direct interactions with the FGFR coreceptors heparan sulfate proteoglycans (HSPGs) and Klotho. Noncanonical FGFR signaling partners have been implicated in differential regulation of FGFR signaling. FGFR directly interacts with cell adhesion molecules (CAMs) and extracellular matrix (ECM) proteins, contributing to invasive and migratory properties of cancer cells, whereas interactions with other receptor tyrosine kinases (RTKs) regulate angiogenic, resistance to therapy, and metastatic potential of cancer cells. The diversity in FGFR signaling partners supports a role for FGFR signaling in cancer, independent of genetic aberration.
Collapse
Affiliation(s)
- Harriet R. Ferguson
- Division of Molecular and Cellular Function, School of Biological Science, Faculty of Biology Medicine and Health (FBMH), The University of Manchester, Manchester M13 9PT, UK;
| | - Michael P. Smith
- Division of Molecular and Cellular Function, School of Biological Science, Faculty of Biology Medicine and Health (FBMH), The University of Manchester, Manchester M13 9PT, UK;
| | - Chiara Francavilla
- Division of Molecular and Cellular Function, School of Biological Science, Faculty of Biology Medicine and Health (FBMH), The University of Manchester, Manchester M13 9PT, UK;
- Manchester Breast Centre, Manchester Cancer Research Centre, The University of Manchester, Manchester M20 4GJ, UK
| |
Collapse
|
35
|
Fonteles CSR, Finnell RH, George TM, Harshbarger RJ. Craniosynostosis: current conceptions and misconceptions. AIMS GENETICS 2021. [DOI: 10.3934/genet.2016.1.99] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
AbstractCranial bones articulate in areas called sutures that must remain patent until skull growth is complete. Craniosynostosis is the condition that results from premature closure of one or more of the cranial vault sutures, generating facial deformities and more importantly, skull growth restrictions with the ability to severely affect brain growth. Typically, craniosynostosis can be expressed as an isolated event, or as part of syndromic phenotypes. Multiple signaling mechanisms interact during developmental stages to ensure proper and timely suture fusion. Clinical outcome is often a product of craniosynostosis subtypes, number of affected sutures and timing of premature suture fusion. The present work aimed to review the different aspects involved in the establishment of craniosynostosis, providing a close view of the cellular, molecular and genetic background of these malformations.
Collapse
Affiliation(s)
- Cristiane Sá Roriz Fonteles
- Finnell Birth Defects Research Laboratory, Dell Pediatric Research Institute, The University of Texas at Austin, USA
| | - Richard H. Finnell
- Finnell Birth Defects Research Laboratory, Dell Pediatric Research Institute, The University of Texas at Austin, USA
- Department of Nutritional Sciences, Dell Pediatric Research Institute, The University of Texas at Austin, USA
| | - Timothy M. George
- Pediatric Neurosurgery, Dell Children's Medical Center, Professor, Department of Surgery, Dell Medical School, Austin, TX, USA
| | - Raymond J. Harshbarger
- Plastic Surgery, Craniofacial Team at the Dell Children's Medical Center of Central Texas, Austin, USA
| |
Collapse
|
36
|
Epstein RJ, Tian LJ, Gu YF. 2b or Not 2b: How Opposing FGF Receptor Splice Variants Are Blocking Progress in Precision Oncology. JOURNAL OF ONCOLOGY 2021; 2021:9955456. [PMID: 34007277 PMCID: PMC8110382 DOI: 10.1155/2021/9955456] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Accepted: 04/21/2021] [Indexed: 01/16/2023]
Abstract
More than ten thousand peer-reviewed studies have assessed the role of fibroblast growth factors (FGFs) and their receptors (FGFRs) in cancer, but few patients have yet benefited from drugs targeting this molecular family. Strategizing how best to use FGFR-targeted drugs is complicated by multiple variables, including RNA splicing events that alter the affinity of ligands for FGFRs and hence change the outcomes of stromal-epithelial interactions. The effects of splicing are most relevant to FGFR2; expression of the FGFR2b splice isoform can restore apoptotic sensitivity to cancer cells, whereas switching to FGFR2c may drive tumor progression by triggering epithelial-mesenchymal transition. The differentiating and regulatory actions of wild-type FGFR2b contrast with the proliferative actions of FGFR1 and FGFR3, and may be converted to mitogenicity either by splice switching or by silencing of tumor suppressor genes such as CDH1 or PTEN. Exclusive use of small-molecule pan-FGFR inhibitors may thus cause nonselective blockade of FGFR2 isoforms with opposing actions, undermining the rationale of FGFR2 drug targeting. This splice-dependent ability of FGFR2 to switch between tumor-suppressing and -driving functions highlights an unmet oncologic need for isoform-specific drug targeting, e.g., by antibody inhibition of ligand-FGFR2c binding, as well as for more nuanced molecular pathology prediction of FGFR2 actions in different stromal-tumor contexts.
Collapse
Affiliation(s)
- Richard J. Epstein
- New Hope Cancer Center, Beijing United Hospital, 9-11 Jiangtai West Rd, Chaoyang, Beijing 100015, China
- Garvan Institute of Medical Research and UNSW Clinical School, 84 Victoria St, Darlinghurst 2010 Sydney, Australia
| | - Li Jun Tian
- New Hope Cancer Center, Beijing United Hospital, 9-11 Jiangtai West Rd, Chaoyang, Beijing 100015, China
| | - Yan Fei Gu
- New Hope Cancer Center, Beijing United Hospital, 9-11 Jiangtai West Rd, Chaoyang, Beijing 100015, China
| |
Collapse
|
37
|
Santolla MF, Talia M, Maggiolini M. S100A4 Is Involved in Stimulatory Effects Elicited by the FGF2/FGFR1 Signaling Pathway in Triple-Negative Breast Cancer (TNBC) Cells. Int J Mol Sci 2021; 22:ijms22094720. [PMID: 33946884 PMCID: PMC8124532 DOI: 10.3390/ijms22094720] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 04/23/2021] [Accepted: 04/27/2021] [Indexed: 12/11/2022] Open
Abstract
Triple-negative breast cancer (TNBC) is an aggressive breast tumor subtype characterized by poor clinical outcome. In recent years, numerous advancements have been made to better understand the biological landscape of TNBC, though appropriate targets still remain to be determined. In the present study, we have determined that the expression levels of FGF2 and S100A4 are higher in TNBC with respect to non-TNBC patients when analyzing “The Invasive Breast Cancer Cohort of The Cancer Genome Atlas” (TCGA) dataset. In addition, we have found that the gene expression of FGF2 is positively correlated with S100A4 in TNBC samples. Performing quantitative PCR, Western blot, CRISPR/Cas9 genome editing, promoter studies, immunofluorescence analysis, subcellular fractionation studies, and ChIP assays, we have also demonstrated that FGF2 induces in TNBC cells the upregulation and secretion of S100A4 via FGFR1, along with the ERK1/2–AKT–c-Rel transduction signaling. Using conditioned medium from TNBC cells stimulated with FGF2, we have also ascertained that the paracrine activation of the S100A4/RAGE pathway triggers angiogenic effects in vascular endothelial cells (HUVECs) and promotes the migration of cancer-associated fibroblasts (CAFs). Collectively, our data provide novel insights into the action of the FGF2/FGFR1 axis through S100A4 toward stimulatory effects elicited in TNBC cells.
Collapse
MESH Headings
- Antigens, Neoplasm/physiology
- Cell Movement/drug effects
- Culture Media, Conditioned/pharmacology
- Female
- Fibroblast Growth Factor 2/pharmacology
- Fibroblast Growth Factor 2/physiology
- Fibroblasts/pathology
- Gene Expression Regulation, Neoplastic/physiology
- Human Umbilical Vein Endothelial Cells
- Humans
- Mitogen-Activated Protein Kinases/physiology
- Neoplasm Proteins/physiology
- Neovascularization, Pathologic/physiopathology
- Paracrine Communication
- Protein Kinase Inhibitors/pharmacology
- Proto-Oncogene Proteins c-rel/physiology
- Receptor, Fibroblast Growth Factor, Type 1/antagonists & inhibitors
- Receptor, Fibroblast Growth Factor, Type 1/genetics
- Receptor, Fibroblast Growth Factor, Type 1/physiology
- S100 Calcium-Binding Protein A4/physiology
- Signal Transduction/drug effects
- Signal Transduction/physiology
- Triple Negative Breast Neoplasms/blood supply
- Triple Negative Breast Neoplasms/physiopathology
- Tumor Cells, Cultured
Collapse
|
38
|
Sobhani N, Fassl A, Mondani G, Generali D, Otto T. Targeting Aberrant FGFR Signaling to Overcome CDK4/6 Inhibitor Resistance in Breast Cancer. Cells 2021; 10:293. [PMID: 33535617 PMCID: PMC7912842 DOI: 10.3390/cells10020293] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Revised: 01/26/2021] [Accepted: 01/29/2021] [Indexed: 01/01/2023] Open
Abstract
Breast cancer (BC) is the most common cause of cancer-related death in women worldwide. Therapies targeting molecular pathways altered in BC had significantly enhanced treatment options for BC over the last decades, which ultimately improved the lives of millions of women worldwide. Among various molecular pathways accruing substantial interest for the development of targeted therapies are cyclin-dependent kinases (CDKs)-in particular, the two closely related members CDK4 and CDK6. CDK4/6 inhibitors indirectly trigger the dephosphorylation of retinoblastoma tumor suppressor protein by blocking CDK4/6, thereby blocking the cell cycle transition from the G1 to S phase. Although the CDK4/6 inhibitors abemaciclib, palbociclib, and ribociclib gained FDA approval for the treatment of hormone receptor (HR)-positive, human epidermal growth factor receptor 2 (HER2)-negative BC as they significantly improved progression-free survival (PFS) in randomized clinical trials, regrettably, some patients showed resistance to these therapies. Though multiple molecular pathways could be mechanistically responsible for CDK4/6 inhibitor therapy resistance, one of the most predominant ones seems to be the fibroblast growth factor receptor (FGFR) pathway. FGFRs are involved in many aspects of cancer formation, such as cell proliferation, differentiation, and growth. Importantly, FGFRs are frequently mutated in BC, and their overexpression and/or hyperactivation correlates with CDK4/6 inhibitor resistance and shortened PFS in BC. Intriguingly, the inhibition of aberrant FGFR activity is capable of reversing the resistance to CDK4/6 inhibitors. This review summarizes the molecular background of FGFR signaling and discusses the role of aberrant FGFR signaling during cancer development in general and during the development of CDK4/6 inhibitor resistance in BC in particular, together with other possible mechanisms for resistance to CDK4/6 inhibitors. Subsequently, future directions on novel therapeutic strategies targeting FGFR signaling to overcome such resistance during BC treatment will be further debated.
Collapse
Affiliation(s)
- Navid Sobhani
- Department of Medicine, Section of Epidemiology and Population Sciences, Baylor College of Medicine, Houston, TX 77030, USA
| | - Anne Fassl
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02215, USA;
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Giuseppina Mondani
- Department Breast Oncoplastic Surgery Royal Cornwall Hospital, Treliske, Truro TR13LJ, UK;
| | - Daniele Generali
- Department of Medical, Surgical and Health Sciences, University of Trieste, Cattinara Hospital, 34149 Trieste, Italy;
| | - Tobias Otto
- Department of Internal Medicine III, University Hospital RWTH Aachen, Pauwelsstrasse 30, 52074 Aachen, Germany
| |
Collapse
|
39
|
Freiin von Hövel F, Kefalakes E, Grothe C. What Can We Learn from FGF-2 Isoform-Specific Mouse Mutants? Differential Insights into FGF-2 Physiology In Vivo. Int J Mol Sci 2020; 22:ijms22010390. [PMID: 33396566 PMCID: PMC7795026 DOI: 10.3390/ijms22010390] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Revised: 12/29/2020] [Accepted: 12/29/2020] [Indexed: 12/16/2022] Open
Abstract
Fibroblast growth factor 2 (FGF-2), ubiquitously expressed in humans and mice, is functionally involved in cell growth, migration and maturation in vitro and in vivo. Based on the same mRNA, an 18-kilo Dalton (kDa) FGF-2 isoform named FGF-2 low molecular weight (FGF-2LMW) isoform is translated in humans and rodents. Additionally, two larger isoforms weighing 21 and 22 kDa also exist, summarized as the FGF-2 high molecular weight (FGF-2HMW) isoform. Meanwhile, the human FGF-2HMW comprises a 22, 23, 24 and 34 kDa protein. Independent studies verified a specific intracellular localization, mode of action and tissue-specific spatiotemporal expression of the FGF-2 isoforms, increasing the complexity of their physiological and pathophysiological roles. In order to analyze their spectrum of effects, FGF-2LMW knock out (ko) and FGF-2HMWko mice have been generated, as well as mice specifically overexpressing either FGF-2LMW or FGF-2HMW. So far, the development and functionality of the cardiovascular system, bone formation and regeneration as well as their impact on the central nervous system including disease models of neurodegeneration, have been examined. This review provides a summary of the studies characterizing the in vivo effects modulated by the FGF-2 isoforms and, thus, offers a comprehensive overview of its actions in the aforementioned organ systems.
Collapse
Affiliation(s)
- Friederike Freiin von Hövel
- Institute of Neuroanatomy and Cell Biology, Hannover Medical School, Carl-Neuberg-Straße 1, D-30625 Hannover, Germany;
- Center for Systems Neuroscience (ZSN), University of Veterinary Medicine, Bünteweg 2, D-30559 Hannover, Germany;
| | - Ekaterini Kefalakes
- Center for Systems Neuroscience (ZSN), University of Veterinary Medicine, Bünteweg 2, D-30559 Hannover, Germany;
| | - Claudia Grothe
- Center for Systems Neuroscience (ZSN), University of Veterinary Medicine, Bünteweg 2, D-30559 Hannover, Germany;
- Correspondence: ; Tel.: +49-511-532-2897; Fax: +49-511-532-2880
| |
Collapse
|
40
|
Personeni N, Lleo A, Pressiani T, Colapietro F, Openshaw MR, Stavraka C, Pouptsis A, Pinato DJ, Rimassa L. Biliary Tract Cancers: Molecular Heterogeneity and New Treatment Options. Cancers (Basel) 2020; 12:3370. [PMID: 33202975 PMCID: PMC7696875 DOI: 10.3390/cancers12113370] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Revised: 11/08/2020] [Accepted: 11/11/2020] [Indexed: 12/13/2022] Open
Abstract
Most patients with biliary tract cancer (BTC) are diagnosed with advanced disease, relapse rates are high in those undergoing surgery and prognosis remains poor, while the incidence is increasing. Treatment options are limited, and chemotherapy is still the standard of care in both adjuvant and advanced disease setting. In recent years, different subtypes of BTC have been defined depending on the anatomical location and genetic and/or epigenetic aberrations. Especially for intrahepatic cholangiocarcinoma (iCCA) novel therapeutic targets have been identified, including fibroblast growth factor receptor 2 gene fusions and isocitrate dehydrogenase 1 and 2 mutations, with molecularly targeted agents having shown evidence of activity in this subgroup of patients. Additionally, other pathways are being evaluated in both iCCA and other subtypes of BTC, alongside targeting of the immune microenvironment. The growing knowledge of BTC biology and molecular heterogeneity has paved the way for the development of new therapeutic approaches that will completely change the treatment paradigm for this disease in the near future. This review provides an overview of the molecular heterogeneity of BTC and summarizes new targets and emerging therapies in development. We also discuss resistance mechanisms, open issues, and future perspectives in the management of BTC.
Collapse
Affiliation(s)
- Nicola Personeni
- Medical Oncology and Hematology Unit, Humanitas Cancer Center, Humanitas Clinical and Research Center-IRCCS, Rozzano, 20089 Milan, Italy; (N.P.); (T.P.)
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, 20090 Milan, Italy; (A.L.); (F.C.)
| | - Ana Lleo
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, 20090 Milan, Italy; (A.L.); (F.C.)
- Internal Medicine Center, Humanitas Clinical and Research Center-IRCCS, Rozzano, 20089 Milan, Italy
| | - Tiziana Pressiani
- Medical Oncology and Hematology Unit, Humanitas Cancer Center, Humanitas Clinical and Research Center-IRCCS, Rozzano, 20089 Milan, Italy; (N.P.); (T.P.)
| | - Francesca Colapietro
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, 20090 Milan, Italy; (A.L.); (F.C.)
- Internal Medicine Center, Humanitas Clinical and Research Center-IRCCS, Rozzano, 20089 Milan, Italy
| | - Mark Robert Openshaw
- Department of Surgery & Cancer, Imperial College London, Hammersmith Hospital, London W120HS, UK; (M.R.O.); (D.J.P.)
| | - Chara Stavraka
- Department of Medical Oncology, Guy’s and St Thomas’ NHS Foundation Trust, Great Maze Pond, London SE1 9RT, UK;
| | - Athanasios Pouptsis
- Department of Medical Oncology, “Euromedica” General Clinic, 54645 Thessaloniki, Greece;
| | - David James Pinato
- Department of Surgery & Cancer, Imperial College London, Hammersmith Hospital, London W120HS, UK; (M.R.O.); (D.J.P.)
| | - Lorenza Rimassa
- Medical Oncology and Hematology Unit, Humanitas Cancer Center, Humanitas Clinical and Research Center-IRCCS, Rozzano, 20089 Milan, Italy; (N.P.); (T.P.)
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, 20090 Milan, Italy; (A.L.); (F.C.)
| |
Collapse
|
41
|
Mahipal A, Tella SH, Kommalapati A, Yu J, Kim R. Prevention and treatment of FGFR inhibitor-associated toxicities. Crit Rev Oncol Hematol 2020; 155:103091. [DOI: 10.1016/j.critrevonc.2020.103091] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Revised: 07/30/2020] [Accepted: 08/17/2020] [Indexed: 12/15/2022] Open
|
42
|
Zheng C, Shi CJ, Du LJ, Jiang YH, Su JM. [Expression of fibroblast growth factor receptor like 1 protein in oral squamous cell carcinoma and its influence on tumor cell proliferation and migration]. HUA XI KOU QIANG YI XUE ZA ZHI = HUAXI KOUQIANG YIXUE ZAZHI = WEST CHINA JOURNAL OF STOMATOLOGY 2020; 38:558-565. [PMID: 33085242 DOI: 10.7518/hxkq.2020.05.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
OBJECTIVE This study aims to investigate the expression of fibroblast growth factor receptor like 1 (FGFRL1) in oral squamous cell carcinoma (OSCC) and reveals its association with tumor cell proliferation and migration. METHODS Western blot was performed to detect the expression of FGFRL1 protein in OSCC tissues, adjacent normal tissues, OSCC cell lines and normal epithelial cells. After knocking down of FGFRL1 in HN4 cells, CCK-8 and Ki67 assays were performed to detect cell proliferation, wounding healing assay and transwell were performed to detect cell-migration. Western blot was used to detect the expression of protein related to epithelial-mesenchymal transition (EMT). RESULTS The expression of FGFRL1 in OSCC tissues was higher than that in adjacent nontumor tissues, respectively (t=2.820, P=0.047 8). Moreover, the expression of FGFRL1 in OSCC cells was higher than that in HOK cells. Quantitative real-time polymerase chain reaction (qRT-PCR) showed that FGFRL1 expression of FGFRL1 RNA in HOK cells was lower than that in OSCC cells. HN4 cells transfected with FGFRL1 siRNA were included in the experimental group, whereas HN4 cells treated with NC siRNA were included in the control group. CCK-8 experiment showed no significant difference between the experimental and control groups with regard to proliferation ability at 48 h (P=0.478 1) and 72 h (P=0.334 2). Migration experiment showed that the wound healing areas in the experimental group after 12 h (P=0.022 8), 24 h (P=0.005 1), and 36 h (P=0.009 5)were smaller than that in the control group. Transwell invasion assay showed that the number of invaded cells in the experimental group after 16 h (P=0.008 7) and 24 h (P=0.008 6) were lower than that in the control group. Knocking-down FGFRL1 up-regulated the expression of E-cadherin and down-regulated the expression of N-cadherin and Vimentin in HN4 cells. CONCLUSIONS FGFRL1 expression in the OSCC tissues was significantly higher than that in the adjacent nontumor tissues. FGFRL1 expression in the OSCC cells was significantly higher than that in the HOK cells, and FGFRL1 had no effect on cell proliferation but promoted tumor cell migration and EMT.
Collapse
Affiliation(s)
- Chen Zheng
- Dept. of Stomatology, The Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou 310052, China
| | - Chao-Ji Shi
- Dept. of Oral and Maxillofacial-Head Neck Oncology, Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, National Clinical Research Center for Oral Diseases, Shanghai 200011, China
| | - Lin-Juan Du
- Dept. of Oral and Maxillofacial-Head Neck Oncology, Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, National Clinical Research Center for Oral Diseases, Shanghai 200011, China
| | - Yin-Hua Jiang
- Dept. of Stomatology, The Six Affiliated Hospital of Wenzhou Medical University, Lishui City People's Hospital, Lishui 323000, China
| | - Ji-Mei Su
- Dept. of Stomatology, The Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou 310052, China
| |
Collapse
|
43
|
Dissecting the Interaction of FGF8 with Receptor FGFRL1. Biomolecules 2020; 10:biom10101399. [PMID: 33019532 PMCID: PMC7600612 DOI: 10.3390/biom10101399] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Revised: 09/25/2020] [Accepted: 09/28/2020] [Indexed: 12/11/2022] Open
Abstract
In mammals, the novel protein fibroblast growth factor receptor-like 1 (FGFRL1) is involved in the development of metanephric kidneys. It appears that this receptor controls a crucial transition of the induced metanephric mesenchyme to epithelial renal vesicles, which further develop into functional nephrons. FGFRL1 knockout mice lack metanephric kidneys and do not express any fibroblast growth factor (FGF) 8 in the metanephric mesenchyme, suggesting that FGFRL1 and FGF8 play a decisive role during kidney formation. FGFRL1 consists of three extracellular immunoglobulin (Ig) domains (Ig1-Ig2-Ig3), a transmembrane domain and a short intracellular domain. We have prepared the extracellular domain (Ig123), the three individual Ig domains (Ig1, Ig2, Ig3) as well as all combinations containing two Ig domains (Ig12, Ig23, Ig13) in recombinant form in human cells. All polypeptides that contain the Ig2 domain (Ig123, Ig12, Ig23, Ig2) were found to interact with FGF8 with very high affinity, whereas all constructs that lack the Ig2 domain (Ig1, Ig3, Ig13) poorly interacted with FGF8 as shown by ELISA and surface plasmon resonance. It is therefore likely that FGFRL1 represents a physiological receptor for FGF8 in the kidney and that the ligand primarily binds to the Ig2 domain of the receptor. With Biacore experiments, we also measured the affinity of FGF8 for the different constructs. All constructs containing the Ig2 domain showed a rapid association and a slow dissociation phase, from which a KD of 2–3 × 10−9 M was calculated. Our data support the hypothesis that binding of FGF8 to FGFRL1 could play an important role in driving the formation of nephrons in the developing kidney.
Collapse
|
44
|
Sun Y, Li G, Zhu W, He Q, Liu Y, Chen X, Liu J, Lin J, Han-Zhang H, Yang Z, Lizaso A, Xiang J, Mao X, Liu H, Gao Y. A comprehensive pan-cancer study of fibroblast growth factor receptor aberrations in Chinese cancer patients. ANNALS OF TRANSLATIONAL MEDICINE 2020; 8:1290. [PMID: 33209870 PMCID: PMC7661893 DOI: 10.21037/atm-20-5118] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Background The prevalence and types of fibroblast growth factor receptor (FGFR) mutations vary significantly among different ethnic groups. The optimal application of FGFR inhibitors depends on these variations being comprehensively understood. However, such an analysis has yet to be conducted in Chinese patients. Methods We retrospectively screened the genomic profiling results of 10,582 Chinese cancer patients across 16 cancer types to investigate the frequency and distribution of FGFR aberrations. Results FGFR aberrations were identified in 745 patients, equating to an overall prevalence of 7.0%. A majority of the aberrations occurred on FGFR1 (56.8%), which was followed by FGFR3 (17.7%), FGFR2 (14.4%), and FGFR4 (2.8%). Further, 8.5% of patients had aberrations of more than 1 FGFR gene. The most common types of aberrations were amplification (53.7%), other mutations (38.8%), and fusions (5.6%). FGFR fusion and amplification occurred concurrently in 1.9% of the patients. FGFR aberrations were detected in 12 of the 16 cancers, with the highest prevalence belonging to colorectal cancer (CRC) (31%). Other FGFR-aberrant cancer types included stomach (16.8%), breast (14.3%), and esophageal (12.7%) cancer. Breast tumors were also more likely than other cancer types to have concurrent FGFR rearrangements and amplifications (P<0.001). In comparison with the public dataset, our cohort had a significantly higher number of FGFR aberrations in colorectal (P<0.001) and breast cancer (P=0.05). Conclusions Among the Chinese cancer patients in our study, the overall prevalence of FGFR aberrations was 7.0%. FGFR1 amplification was the most common genetic alteration in CRC, breast cancer, and lung cancer; while FGFR2 amplification was more commonly observed in gastric cancer than in other cancers in our cohort. Our study advances the understanding of the distribution of FGFR aberrations in various cancer types in the Chinese population, which will facilitate the further development of FGFR inhibitors.
Collapse
Affiliation(s)
- Yi Sun
- Department of Pathology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Gao Li
- Department of Thoracic Surgery, Hainan General Hospital, Haikou, China
| | - Wei Zhu
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, China
| | - Qiuyan He
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, China
| | - Yongchang Liu
- Department of Pathology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Xianshan Chen
- Department of Thoracic Surgery, Hainan General Hospital, Haikou, China
| | - Juan Liu
- Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital; Nanjing, China
| | - Jing Lin
- Burning Rock Biotech, Guangzhou, China
| | | | - Zheng Yang
- Department of Pathology, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | | | | | - Xinru Mao
- Burning Rock Biotech, Guangzhou, China
| | - Hao Liu
- Burning Rock Biotech, Guangzhou, China
| | - Yang Gao
- Department of Thoracic Surgery, Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
45
|
De Luca A, Esposito Abate R, Rachiglio AM, Maiello MR, Esposito C, Schettino C, Izzo F, Nasti G, Normanno N. FGFR Fusions in Cancer: From Diagnostic Approaches to Therapeutic Intervention. Int J Mol Sci 2020; 21:E6856. [PMID: 32962091 PMCID: PMC7555921 DOI: 10.3390/ijms21186856] [Citation(s) in RCA: 71] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 09/15/2020] [Accepted: 09/15/2020] [Indexed: 12/11/2022] Open
Abstract
Fibroblast growth factor receptors (FGFRs) are tyrosine kinase receptors involved in many biological processes. Deregulated FGFR signaling plays an important role in tumor development and progression in different cancer types. FGFR genomic alterations, including FGFR gene fusions that originate by chromosomal rearrangements, represent a promising therapeutic target. Next-generation-sequencing (NGS) approaches have significantly improved the discovery of FGFR gene fusions and their detection in clinical samples. A variety of FGFR inhibitors have been developed, and several studies are trying to evaluate the efficacy of these agents in molecularly selected patients carrying FGFR genomic alterations. In this review, we describe the most frequent FGFR aberrations in human cancer. We also discuss the different approaches employed for the detection of FGFR fusions and the potential role of these genomic alterations as prognostic/predictive biomarkers.
Collapse
Affiliation(s)
- Antonella De Luca
- Cell Biology and Biotherapy Unit, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131 Naples, Italy; (A.D.L.); (R.E.A.); (A.M.R.); (M.R.M.); (C.E.)
| | - Riziero Esposito Abate
- Cell Biology and Biotherapy Unit, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131 Naples, Italy; (A.D.L.); (R.E.A.); (A.M.R.); (M.R.M.); (C.E.)
| | - Anna Maria Rachiglio
- Cell Biology and Biotherapy Unit, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131 Naples, Italy; (A.D.L.); (R.E.A.); (A.M.R.); (M.R.M.); (C.E.)
| | - Monica Rosaria Maiello
- Cell Biology and Biotherapy Unit, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131 Naples, Italy; (A.D.L.); (R.E.A.); (A.M.R.); (M.R.M.); (C.E.)
| | - Claudia Esposito
- Cell Biology and Biotherapy Unit, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131 Naples, Italy; (A.D.L.); (R.E.A.); (A.M.R.); (M.R.M.); (C.E.)
| | - Clorinda Schettino
- Clinical Trials Unit, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131 Naples, Italy;
| | - Francesco Izzo
- Division of Surgical Oncology, Hepatobiliary Unit, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131 Naples, Italy;
| | - Guglielmo Nasti
- SSD Innovative Therapies for Abdominal Cancers, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131 Naples, Italy;
| | - Nicola Normanno
- Cell Biology and Biotherapy Unit, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131 Naples, Italy; (A.D.L.); (R.E.A.); (A.M.R.); (M.R.M.); (C.E.)
| |
Collapse
|
46
|
Ortmann D, Brown S, Czechanski A, Aydin S, Muraro D, Huang Y, Tomaz RA, Osnato A, Canu G, Wesley BT, Skelly DA, Stegle O, Choi T, Churchill GA, Baker CL, Rugg-Gunn PJ, Munger SC, Reinholdt LG, Vallier L. Naive Pluripotent Stem Cells Exhibit Phenotypic Variability that Is Driven by Genetic Variation. Cell Stem Cell 2020; 27:470-481.e6. [PMID: 32795399 PMCID: PMC7487768 DOI: 10.1016/j.stem.2020.07.019] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2019] [Revised: 04/10/2020] [Accepted: 07/24/2020] [Indexed: 12/11/2022]
Abstract
Variability among pluripotent stem cell (PSC) lines is a prevailing issue that hampers not only experimental reproducibility but also large-scale applications and personalized cell-based therapy. This variability could result from epigenetic and genetic factors that influence stem cell behavior. Naive culture conditions minimize epigenetic fluctuation, potentially overcoming differences in PSC line differentiation potential. Here we derived PSCs from distinct mouse strains under naive conditions and show that lines from distinct genetic backgrounds have divergent differentiation capacity, confirming a major role for genetics in PSC phenotypic variability. This is explained in part through inconsistent activity of extra-cellular signaling, including the Wnt pathway, which is modulated by specific genetic variants. Overall, this study shows that genetic background plays a dominant role in driving phenotypic variability of PSCs.
Collapse
Affiliation(s)
- Daniel Ortmann
- Wellcome Trust and MRC Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK; Department of Surgery, University of Cambridge, Cambridge, UK.
| | - Stephanie Brown
- Wellcome Trust and MRC Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK; Department of Surgery, University of Cambridge, Cambridge, UK
| | | | | | - Daniele Muraro
- Wellcome Trust and MRC Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK; Department of Surgery, University of Cambridge, Cambridge, UK; Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK
| | - Yuanhua Huang
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK; European Molecular Biology Laboratory, European Bioinformatics Institute, Wellcome Genome Campus, Hinxton, UK
| | - Rute A Tomaz
- Wellcome Trust and MRC Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK; Department of Surgery, University of Cambridge, Cambridge, UK
| | - Anna Osnato
- Wellcome Trust and MRC Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK; Department of Surgery, University of Cambridge, Cambridge, UK
| | - Giovanni Canu
- Wellcome Trust and MRC Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK; Department of Surgery, University of Cambridge, Cambridge, UK
| | - Brandon T Wesley
- Wellcome Trust and MRC Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK; Department of Surgery, University of Cambridge, Cambridge, UK
| | | | - Oliver Stegle
- European Molecular Biology Laboratory, European Bioinformatics Institute, Wellcome Genome Campus, Hinxton, UK; European Molecular Biology Laboratory, Genome Biology Unit, Heidelberg, Germany; Division of Computational Genomics and Systems Genetics, German Cancer Research, Center (DKFZ), Heidelberg, Germany
| | - Ted Choi
- Jackson Laboratory, Bar Harbor, ME, USA
| | | | | | - Peter J Rugg-Gunn
- Wellcome Trust and MRC Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK; Epigenetics Programme, Babraham Institute, Cambridge, UK
| | | | | | - Ludovic Vallier
- Wellcome Trust and MRC Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK; Department of Surgery, University of Cambridge, Cambridge, UK.
| |
Collapse
|
47
|
Huh SH, Ha L, Jang HS. Nephron Progenitor Maintenance Is Controlled through Fibroblast Growth Factors and Sprouty1 Interaction. J Am Soc Nephrol 2020; 31:2559-2572. [PMID: 32753399 DOI: 10.1681/asn.2020040401] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Accepted: 07/08/2020] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND Nephron progenitor cells (NPCs) give rise to all segments of functional nephrons and are of great interest due to their potential as a source for novel treatment strategies for kidney disease. Fibroblast growth factor (FGF) signaling plays pivotal roles in generating and maintaining NPCs during kidney development, but little is known about the molecule(s) regulating FGF signaling during nephron development. Sprouty 1 (SPRY1) is an antagonist of receptor tyrosine kinases. Although SPRY1 antagonizes Ret-GDNF signaling, which modulates renal branching, its role in NPCs is not known. METHODS Spry1, Fgf9, and Fgf20 compound mutant animals were used to evaluate kidney phenotypes in mice to understand whether SPRY1 modulates FGF signaling in NPCs and whether FGF8 functions with FGF9 and FGF20 in maintaining NPCs. RESULTS Loss of one copy of Spry1 counters effects of the loss of Fgf9 and Fgf20, rescuing bilateral renal agenesis premature NPC differentiation, NPC proliferation, and cell death defects. In the absence of SPRY1, FGF9, and FGF20, another FGF ligand, FGF8, promotes nephrogenesis. Deleting both Fgf8 and Fgf20 results in kidney agenesis, defects in NPC proliferation, and cell death. Deleting one copy of Fgf8 reversed the effect of deleting one copy of Spry1, which rescued the renal agenesis due to loss of Fgf9 and Fgf20. CONCLUSIONS SPRY1 expressed in NPCs modulates the activity of FGF signaling and regulates NPC stemness. These findings indicate the importance of the balance between positive and negative signals during NPC maintenance.
Collapse
Affiliation(s)
- Sung-Ho Huh
- Department of Neurological Sciences, University of Nebraska Medical Center, Omaha, Nebraska .,Holland Regenerative Medicine Program, University of Nebraska Medical Center, Omaha, Nebraska
| | - Ligyeom Ha
- Department of Neurological Sciences, University of Nebraska Medical Center, Omaha, Nebraska
| | - Hee-Seong Jang
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, Nebraska
| |
Collapse
|
48
|
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: 35] [Impact Index Per Article: 7.0] [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
|
49
|
Hong CS, Sun EG, Choi JN, Kim DH, Kim JH, Ryu KH, Shim HJ, Hwang JE, Bae WK, Kim HR, Kim KK, Jung C, Chung IJ, Cho SH. Fibroblast growth factor receptor 4 increases epidermal growth factor receptor (EGFR) signaling by inducing amphiregulin expression and attenuates response to EGFR inhibitors in colon cancer. Cancer Sci 2020; 111:3268-3278. [PMID: 32533590 PMCID: PMC7469799 DOI: 10.1111/cas.14526] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2020] [Revised: 06/03/2020] [Accepted: 06/06/2020] [Indexed: 02/06/2023] Open
Abstract
Fibroblast growth factor receptor 4 (FGFR4) is known to induce cancer cell proliferation, invasion, and antiapoptosis through activation of RAS/RAF/ERK and PI3K/AKT pathways, which are also known as major molecular bases of colon cancer carcinogenesis related with epidermal growth factor receptor (EGFR) signaling. However, the interaction between FGFR4 and EGFR signaling in regard to colon cancer progression is unclear. Here, we investigated a potential cross‐talk between FGFR4 and EGFR, and the effect of anti‐EGFR therapy in colon cancer treatment. To explore the biological roles of FGFR4 in cancer progression, RNA sequencing was carried out using FGFR4 transfected colon cell lines. Gene ontology data showed the upregulation of genes related to EGFR signaling, and we identified that FGFR4 overexpression secretes EGFR ligands such as amphiregulin (AREG) with consequent activation of EGFR and ErbB3. This result was also shown in in vivo study and the cooperative interaction between EGFR and FGFR4 promoted tumor growth. In addition, FGFR4 overexpression reduced cetuximab‐induced cytotoxicity and the combination of FGFR4 inhibitor (BLU9931) and cetuximab showed profound antitumor effect compared to cetuximab alone. Clinically, we found the positive correlation between FGFR4 and AREG expression in tumor tissue, but not in normal tissue, from colon cancer patients and these expressions were significantly correlated with poor overall survival in patients treated with cetuximab. Therefore, our results provide the novel mechanism of FGFR4 in connection with EGFR activation and the combination of FGFR4 inhibitor and cetuximab could be a promising therapeutic option to achieve the optimal response to anti‐EGFR therapy in colon cancer.
Collapse
Affiliation(s)
- Chang-Soo Hong
- Division of Hematology-Oncology, Department of Internal Medicine, Chonnam National University Hwasun Hospital, Chonnam National University Medical School, Hwasun, Korea
| | - Eun-Gene Sun
- Division of Hematology-Oncology, Department of Internal Medicine, Chonnam National University Hwasun Hospital, Chonnam National University Medical School, Hwasun, Korea
| | - Ji-Na Choi
- Division of Hematology-Oncology, Department of Internal Medicine, Chonnam National University Hwasun Hospital, Chonnam National University Medical School, Hwasun, Korea
| | - Dae-Hwan Kim
- Division of Hematology-Oncology, Department of Internal Medicine, Chonnam National University Hwasun Hospital, Chonnam National University Medical School, Hwasun, Korea
| | - Jo-Heon Kim
- Department of Pathology, Chonnam National University Hwasun Hospital, Hwasun, Korea
| | - Kyung-Hyun Ryu
- Department of Biological Science, Sookmyung Women's University, Seoul, Korea
| | - Hyun-Jeong Shim
- Division of Hematology-Oncology, Department of Internal Medicine, Chonnam National University Hwasun Hospital, Chonnam National University Medical School, Hwasun, Korea
| | - Jun-Eul Hwang
- Division of Hematology-Oncology, Department of Internal Medicine, Chonnam National University Hwasun Hospital, Chonnam National University Medical School, Hwasun, Korea
| | - Woo-Kyun Bae
- Division of Hematology-Oncology, Department of Internal Medicine, Chonnam National University Hwasun Hospital, Chonnam National University Medical School, Hwasun, Korea.,Combinatorial Tumor Immunotherapy MRC, Chonnam National University Medical School, Hwasun, Korea
| | - Hyeong-Rok Kim
- Department of Surgery, Chonnam National University Hwasun Hospital, Hwasun, Korea
| | - Kyung Keun Kim
- Department of Pharmacology, Chonnam National University Medical School, Hwasun, Korea
| | - Chaeyong Jung
- Department of Anatomy, Chonnam National University Medical School, Hwasun, Korea
| | - Ik-Joo Chung
- Division of Hematology-Oncology, Department of Internal Medicine, Chonnam National University Hwasun Hospital, Chonnam National University Medical School, Hwasun, Korea.,Combinatorial Tumor Immunotherapy MRC, Chonnam National University Medical School, Hwasun, Korea
| | - Sang-Hee Cho
- Division of Hematology-Oncology, Department of Internal Medicine, Chonnam National University Hwasun Hospital, Chonnam National University Medical School, Hwasun, Korea.,Combinatorial Tumor Immunotherapy MRC, Chonnam National University Medical School, Hwasun, Korea
| |
Collapse
|
50
|
Qin Q, Patel V, Galsky MD. Urothelial carcinoma: the development of FGFR inhibitors in combination with immune checkpoint inhibitors. Expert Rev Anticancer Ther 2020; 20:503-512. [PMID: 32436413 DOI: 10.1080/14737140.2020.1770600] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
INTRODUCTION The recent approval of erdafitinib and the emergence of other potent and selective fibroblast growth factor receptor (FGFR) inhibitors (FGFRi's) are shifting the treatment paradigm for patients with advanced urothelial carcinoma (UC) harboring FGFR3 alterations. Whether such therapies can, and should, be combined with immune checkpoint inhibitors (ICI's) is an area of major research interest. Areas covered: Herein, we review the FGFR signaling pathway and impact of altered FGFR signaling on UC tumorigenesis, the clinical development of FGFRi's, the rationale for FGFRi-ICI combinations, current trials, and future directions. Expert opinion: FGFR3 altered UCs are not less responsive to ICI's compared with FGFR3 wild-type (WT) tumors. However, FGFR3 altered tumors may exhibit distinct immunobiology compared with WT tumors that could potentially be exploited therapeutically. Given these considerations along with the clinical non-cross resistance of these therapeutic classes, clinical investigation of regimens combining FGFR3i and ICI is warranted.
Collapse
Affiliation(s)
- Qian Qin
- Department of Medicine, Division of Hematology and Medical Oncology, the Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai , New York, NY, USA
| | - Vaibhav Patel
- Department of Medicine, Division of Hematology and Medical Oncology, the Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai , New York, NY, USA
| | - Matthew D Galsky
- Department of Medicine, Division of Hematology and Medical Oncology, the Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai , New York, NY, USA
| |
Collapse
|