Next Article in Journal
Screening Services in a Community Pharmacy in Poznan (Poland) to Increase Early Detection of Hypertension
Previous Article in Journal
The Impact of Spinopelvic Mobility on Arthroplasty: Implications for Hip and Spine Surgeons
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Editorial

Promises of Fibroblast Growth Factor Receptor–Directed Therapy in Tailored Cancer Treatment

by
Sharmila Fagoonee
1,* and
Rinaldo Pellicano
2
1
Institute of Biostructure and Bioimaging (CNR), Molecular Biotechnology Center, 10126 Turin, Italy
2
Unit of Gastroenterology, Molinette Hospital, 10123 Turin, Italy
*
Author to whom correspondence should be addressed.
J. Clin. Med. 2020, 9(8), 2570; https://doi.org/10.3390/jcm9082570
Submission received: 28 July 2020 / Accepted: 6 August 2020 / Published: 8 August 2020
(This article belongs to the Section Clinical Laboratory Medicine)
Cancer, one of the deadliest and undefeatable diseases, involves the deregulated growth of cells with the conferment of a high potential to metastasize. It is a multifaceted disease with around 18 million cases diagnosed annually in the world [1]. In recent years, the discovery of oncogenic mechanisms regarding molecular drivers of cancer progression and response to treatment has led to the development of novel target therapies. In this regard, receptor tyrosine kinases (RTKs) have become an attractive therapeutic target. RTKs play a pivotal role in essential processes such as cell survival, migration, proliferation, and differentiation. Due to their importance in cellular homeostasis, the expression and activity of RTKs is finely tuned, and any aberrancy in their signalling leads to disease development. Chromosomal rearrangements, gene amplifications, gain of function, alternative splicing, and autocrine activation are among the most frequent causes of oncogenic RTK signalling [2,3]. Among the RTKs, the fibroblast growth factor–fibroblast growth factor receptor (FGF–FGFR) axis has attracted considerable attention as a druggable target, and several therapeutic strategies have been developed for its selective targeting in cancer [4].
FGFR was first recognized as an important entity in embryogenesis, and abnormalities affecting components of its pathway were associated with developmental defects and congenital malformations [5]. Since then, numerous studies have shown the fundamental role of the FGF–FGFR signalling pathway in various crucial biological processes including tissue regeneration and angiogenesis. Moreover, deregulation of FGFR signalling, through the above-cited oncogenic mechanisms, has been detected in cancer. For instance, aberrant FGF signalling, via upregulation of FGFR 1, 2, and 4 expression, has been implicated in cholangiocarcinoma (CCA) progression [6]. Several studies have also identified different FGFR2 gene fusions in CCA cases, including FGFR–BICCI, FGFR2–AHCYL1, FGFR2–TACC3, and FGFR2–KIAA 1598 [7]. Importantly, in intrahepatic CCA, FGFR2 fusions have been observed in 10% to 16% of patients by DNA sequencing of tumour samples [8]. These aberrations contribute to cancer cell proliferation, resistance to anticancer therapies, and neoangiogenesis; for example, the activation of downstream oncogenes (such as, enhanced FRS2 and STAT3 signalling) or the loss of microRNA regulation (such as loss of miR-99a, which modulates FGFR3 expression) following FGFR3–TCCC3 fusion are among the observed causes of such havoc [9].
Due to the high redundancy and pleiotropic effects of the FGF–FGFR signalling pathway, it has been challenging to design drugs targeting this axis. Therapeutic approaches to selectively block the FGF–FGFR system in human cancer rely on novel strategies and drugs, which are currently being employed in preclinical studies and clinical trials [10]. Tyrosine kinase inhibitors (TKIs), such as small molecules, antagonistic antibodies, or peptide inhibitors or FGF ligand traps, with high antitumour activity and specific toxicity profiles have been developed. Cytotoxic drugs can also be efficiently delivered into cancer cells through FGFRs. For instance, antibody–drug or ligand–drug conjugates or anti-FGFR aptamers can cause rapid downregulation of surface FGFRs and lead to cellular-trafficking-dependant lysosomal receptor degradation. Interestingly, in some cancers such as biliary tract cancers, FGFR genetic aberrations appear to be the most important predictor of sensitivity to small-molecule TKIs, such as NVP-BGJ398, a pan-FGFR inhibitor [11].
The development of resistance against applied therapeutics is a serious unresolved problem in cancer treatment. Tumour cells may evolve ways to bypass the FGFR–FGF signalling by overexpressing other RTK members (bypass signalling) or by inducing mutations in the inhibitor-binding pocket in the FGFR (gatekeeper mutations) [12]. In these cases, the use of combination therapies and novel FGFR therapies may help in overcoming cancer cell resistance. Moreover, FGFRs, which are not exposed on the cell membrane, may act independently of the tyrosine kinase activity. In fact, FGFRs have been described in several subcellular organelles, such as the endosome, nucleus, and mitochondria. Nuclear and mitochondrial FGFRs are capable of promoting tumourigenesis by modulating gene expression or cellular metabolism. Porębska et al. concisely reviewed how the trafficking of FGFRs can be targeted for selective cancer treatment [2]. As tyrosine kinase activity is not involved, the intracellular FGFRs cannot be targeted using classical anticancer drugs. New strategies aimed at inhibiting the cellular translocation of FGFRs are required. One strategy may involve the selective targeting of the processing proteases present at the plasma membrane [2].
Despite the effort in developing FGFR-pathway-targeting drugs, there are still barriers to their clinical application [13]. Epithelial-to-mesenchymal transition may emerge as a consequence of chronic exposure to FGFR inhibitors, hence resulting in resistance [14]. Additional layers of complication arise from alternative splicing of FGFRs by RNA-binding proteins causing alterations in ligand recognition. For instance, epithelial splicing regulatory factor-1 (ESRP1) was shown to promote the expression of the epithelial isoform of FGFR2, FGFR-2-IIIb (which binds specific ligands of the paracrine FGF family such as FGF-7), at the expense of the mesenchymal isoform FGFR-2-IIIc [3,15]. Both FGFR2 and ESRP1 are frequently amplified and demethylated in gastric cancer, hence leading to increased expression of ESRP1 and the FGFR isoform IIIb [16]. A class switch from FGFR-2-IIIb to FGFR-2-IIIc has been observed during the progression of prostate and bladder cancer, with FGFR-2-IIIc being associated with a more malignant phenotype [17]. Interestingly, restoration of FGFR-2-IIIb in prostate cancer cells enhanced their sensitivity to radiation and chemotherapy, indicating that modulating alternative splicing may be a possible alternative to selectively target the FGFR–FGF pathway [16].
It is becoming increasingly clear that the consequence of FGFR aberrations is not uniform across cancer types. Molecular testing such as next-generation sequencing is employed to analyse tumours to detect actionable FGF/FGFR alterations, but biopsy location and tumour heterogeneity can limit this approach. There is, thus, an urgent need to develop biomarkers to choose the most appropriate treatment strategies and to frequently and non-invasively monitor response to targeted therapies [18]. Analysis of serum or urine circulating tumour DNA, faecal microRNAs, or extracellular vesicles released from tumour cells may overcome these limitations [19,20,21]. Endocrine FGFs may also be applied as serum biomarkers of cancer as recently reviewed in prostate cancer [15]. These data are encouraging and warrant further studies regarding the optimal timing and conditions for measuring these biomolecules.
As phase II clinical trials emerge, patient selection as well as methods for predicting response to therapy and for overcoming toxicity become essential. Future generations of FGFR inhibitors will hopefully overcome current barriers and expedite the availability of these new medications for blocking the progression of cancers that rely on the FGFR–FGF axis [13]. Understanding the FGFR status of tumours, as well as their natural history, molecular mechanisms, prognostic impact, and response to FGFR-targeting is crucial to offer precision-medicine-based therapy for their effective management. Thus, FGFR studies are an excellent example of how in-depth exploration at a molecular level could render the design of individualized therapy plans feasible [22].

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Bray, F.; Ferlay, J.; Soerjomataram, I.; Siegel, R.L.; Torre, L.A.; Jemal, A. Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J. Clin. 2018, 68, 394–424. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  2. Porębska, N.; Latko, M.; Kucińska, M.; Zakrzewska, M.; Otlewski, J.; Opaliński, Ł. Targeting Cellular Trafficking of Fibroblast Growth Factor Receptors as a Strategy for Selective Cancer Treatment. J. Clin. Med. 2018, 8, 7. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Fagoonee, S.; Picco, G.; Orso, F.; Arrigoni, A.; Longo, D.L.; Forni, M.; Scarfò, I.; Cassenti, A.; Piva, R.; Cassoni, P.; et al. The RNA-binding protein ESRP1 promotes human colorectal cancer progression. Oncotarget 2017, 8, 10007–10024. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Porta, R.; Borea, R.; Coelho, A.; Khan, S.; Araújo, A.; Reclusa, P.; Franchina, T.; Van Der Steen, N.; Van Dam, P.; Ferri, J.; et al. FGFR a promising druggable target in cancer: Molecular biology and new drugs. Crit. Rev. Oncol. Hematol. 2017, 113, 256–267. [Google Scholar] [CrossRef] [Green Version]
  5. Hajihosseini, M.K.; Wilson, S.; De Moerlooze, L.; Dickson, C. A splicing switch and gain-of-function mutation in FgfR2-IIIc hemizygotes causes Apert/Pfeiffer-syndrome-like phenotypes. Proc. Natl. Acad. Sci. USA 2001, 98, 3855–3860. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  6. Rizvi, S.; Yamada, D.; Hirsova, P.; Bronk, S.F.; Werneburg, N.W.; Krishnan, A.; Salim, W.; Zhang, L.; Trushina, E.; Truty, M.J.; et al. A Hippo and Fibroblast Growth Factor Receptor Autocrine Pathway in Cholangiocarcinoma. J. Biol. Chem. 2016, 291, 8031–8047. [Google Scholar] [CrossRef] [Green Version]
  7. Rizvi, S.; Borad, M.J. The rise of the FGFR inhibitor in advanced biliary cancer: The next cover of time magazine? J. Gastrointest. Oncol. 2016, 7, 789–796. [Google Scholar] [CrossRef] [Green Version]
  8. Jain, A.; Borad, M.J.; Kelley, R.K.; Wang, Y.; Abdelwahab, R.; Mericbernstam, F.; Baggerly, K.A.; Kaseb, A.; Alshamsi, H.O.; Ahn, D.H.; et al. Cholangiocarcinoma With FGFR Genetic Aberrations: A Unique Clinical Phenotype. JCO Precis. Oncol. 2018, 1–12. [Google Scholar] [CrossRef]
  9. Parker, B.C.; Engels, M.; Annala, M.; Zhang, W. Emergence of FGFR family gene fusions as therapeutic targets in a wide spectrum of solid tumours. J. Pathol. 2014, 232, 4–15. [Google Scholar] [CrossRef]
  10. Giacomini, A.; Chiodelli, P.; Matarazzo, S.; Rusnati, M.; Presta, M.; Ronca, R. Blocking the FGF/FGFR system as a “two-compartment” antiangiogenic/antitumor approach in cancer therapy. Pharmacol. Res. 2016, 107, 172–185. [Google Scholar] [CrossRef]
  11. Katoh, M. Fibroblast growth factor receptors as treatment targets in clinical oncology. Nat. Rev. Clin. Oncol. 2019, 16, 105–122. [Google Scholar] [CrossRef] [PubMed]
  12. Dai, S.; Zhou, Z.; Chen, Z.; Xu, G.; Chen, Y. Fibroblast Growth Factor Receptors (FGFRs): Structures and Small Molecule Inhibitors. Cells 2019, 8, 614. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. Chae, Y.K.; Ranganath, K.; Hammerman, P.S.; Vaklavas, C.; Mohindra, N.; Kalyan, A.; Matsangou, M.; Costa, R.; Carneiro, B.; Villaflor, V.M.; et al. Inhibition of the fibroblast growth factor receptor (FGFR) pathway: The current landscape and barriers to clinical application. Oncotarget 2017, 8, 16052–16074. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Grygielewicz, P.; Dymek, B.; Bujak, A.; Gunerka, P.; Stanczak, A.; Lamparska-Przybysz, M.; Wieczorek, M.; Dzwonek, K.; Zdzalik, D. Epithelial-mesenchymal transition confers resistance to selective FGFR inhibitors in SNU-16 gastric cancer cells. Gastric Cancer 2016, 19, 53–62. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  15. Teishima, J.; Hayashi, T.; Nagamatsu, H.; Shoji, K.; Shikuma, H.; Yamanaka, R.; Sekino, Y.; Goto, K.; Inoue, S.; Matsubara, A. Fibroblast Growth Factor Family in the Progression of Prostate Cancer. J. Clin. Med. 2019, 8. [Google Scholar] [CrossRef] [Green Version]
  16. Teles, S.P.; Oliveira, P.; Ferreira, M.; Carvalho, J.; Ferreira, P.; Oliveira, C. Integrated Analysis of Structural Variation and RNA Expression of FGFR2 and Its Splicing Modulator ESRP1 Highlight the ESRP1amp-FGFR2norm-FGFR2-IIIchigh Axis in Diffuse Gastric Cancer. Cancers 2019, 12, 70. [Google Scholar] [CrossRef] [Green Version]
  17. Tiong, K.H.; Mah, L.Y.; Leong, C.O. Functional roles of fibroblast growth factor receptors (FGFRs) signaling in human cancers. Apoptosis 2013, 18, 1447–1468. [Google Scholar] [CrossRef] [Green Version]
  18. Caviglia, G. Detection of gastrointestinal cancer: The role of cancer biomarkers. Minerva Biotecnol. 2019, 31, 1–2. [Google Scholar] [CrossRef]
  19. Han, X.; Wang, J.; Sun, Y. Circulating Tumor DNA as Biomarkers for Cancer Detection. Genom. Proteom. Bioinform. 2017, 15, 59–72. [Google Scholar] [CrossRef]
  20. Xiao, Y.; Zhong, J.; Zhong, B.; Huang, J.; Jiang, L.; Jiang, Y.; Yuan, J.; Sun, J.; Dai, L.; Yang, C.; et al. Exosomes as potential sources of biomarkers in colorectal cancer. Cancer Lett. 2020, 476, 13–22. [Google Scholar] [CrossRef]
  21. Francavilla, A.; Tarallo, S.; Pardini, B.; Naccarati, A. Fecal microRNAs as non-invasive biomarkers for the detection of colorectal cancer: A systematic review. Minerva Biotecnol. 2019, 31, 30–42. [Google Scholar] [CrossRef]
  22. Actis, G.C.; Pellicano, R. Chronic (inflammatory) diseases: Precision medicine versus comprehensive understanding? Minerva Med. 2018, 109, 150–151. [Google Scholar] [PubMed]

Share and Cite

MDPI and ACS Style

Fagoonee, S.; Pellicano, R. Promises of Fibroblast Growth Factor Receptor–Directed Therapy in Tailored Cancer Treatment. J. Clin. Med. 2020, 9, 2570. https://doi.org/10.3390/jcm9082570

AMA Style

Fagoonee S, Pellicano R. Promises of Fibroblast Growth Factor Receptor–Directed Therapy in Tailored Cancer Treatment. Journal of Clinical Medicine. 2020; 9(8):2570. https://doi.org/10.3390/jcm9082570

Chicago/Turabian Style

Fagoonee, Sharmila, and Rinaldo Pellicano. 2020. "Promises of Fibroblast Growth Factor Receptor–Directed Therapy in Tailored Cancer Treatment" Journal of Clinical Medicine 9, no. 8: 2570. https://doi.org/10.3390/jcm9082570

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop