Hostname: page-component-8448b6f56d-cfpbc Total loading time: 0 Render date: 2024-04-23T09:27:53.683Z Has data issue: false hasContentIssue false

FGFR3 targeting strategies for achondroplasia

Published online by Cambridge University Press:  23 April 2012

Melanie B. Laederich
Affiliation:
Shriners Hospital for Children, Research Center, Portland, OR, USA Departments of Cell & Developmental Biology and Molecular & Medical Genetics, Oregon Health & Science University, Portland, OR, USA
William A. Horton*
Affiliation:
Shriners Hospital for Children, Research Center, Portland, OR, USA Departments of Cell & Developmental Biology and Molecular & Medical Genetics, Oregon Health & Science University, Portland, OR, USA
*
*Corresponding author: William A. Horton, Research Center, Shriners Hospital for Children, 3101 SW Sam Jackson Park Road, Portland, OR 97239, USA. E-mail: wah@shcc.org

Abstract

Mutations that exaggerate signalling of the receptor tyrosine kinase fibroblast growth factor receptor 3 (FGFR3) give rise to achondroplasia, the most common form of dwarfism in humans. Here we review the clinical features, genetic aspects and molecular pathogenesis of achondroplasia and examine several therapeutic strategies designed to target the mutant receptor or its signalling pathways, including the use of kinase inhibitors, blocking antibodies, physiologic antagonists, RNAi and chaperone inhibitors. We conclude by discussing the challenges of treating growth plate disorders in children.

Type
Review Article
Copyright
Copyright © Cambridge University Press 2012

Access options

Get access to the full version of this content by using one of the access options below. (Log in options will check for institutional or personal access. Content may require purchase if you do not have access.)

References

References

1Horton, W.A., Hall, J.G. and Hecht, J.T. (2007) Achondroplasia. Lancet 370, 162-172CrossRefGoogle ScholarPubMed
2Kozma, C. (2006) Dwarfs in ancient Egypt. American Journal of Medical Genetics A 140, 303-311CrossRefGoogle ScholarPubMed
3Rimoin, D.L. (1975) The chondrodystrophies. Advances in Human Genetics 5, 1-118Google ScholarPubMed
4Shiang, R. et al. (1994) Mutations in the transmembrane domain of FGFR3 cause the most common genetic form of dwarfism, achondroplasia. Cell 78, 335-342CrossRefGoogle ScholarPubMed
5Rousseau, F. et al. (1994) Mutations in the gene encoding fibroblast growth factor receptor-3 in achondroplasia. Nature 371, 252-254CrossRefGoogle ScholarPubMed
6Bellus, G.A. et al. (1995) Achondroplasia is defined by recurrent G380R mutations of FGFR3. American Journal of Human Genetics 56, 368-373Google ScholarPubMed
7Wynn, J. et al. (2007) Mortality in achondroplasia study: a 42-year follow-up. American Journal of Medical Genetics Part A 143A, 2502-2511CrossRefGoogle ScholarPubMed
8Velinov, M. et al. (1994) The gene for achondroplasia maps to the telomeric region of chromosome 4p. Nature Genetics 6, 314-317CrossRefGoogle Scholar
9Bellus, G.A. et al. (1995) A recurrent mutation in the tyrosine kinase domain of fibroblast growth factor receptor 3 causes hypochondroplasia. Nature Genetics 10, 357-359CrossRefGoogle ScholarPubMed
10Tavormina, P.L. et al. (1995) Thanatophoric dysplasia (types I and II) caused by distinct mutations in fibroblast growth factor receptor 3. Nature Genetics 9, 321-328Google Scholar
11Vajo, Z., Francomano, C.A. and Wilkin, D.J. (2000) The molecular and genetic basis of fibroblast growth factor receptor 3 disorders: the achondroplasia family of skeletal dysplasias, Muenke craniosynostosis, and Crouzon syndrome with acanthosis nigricans. Endocrine Reviews 21, 23-39Google ScholarPubMed
12Dodurga, Y. et al. (2011) Incidence of fibroblast growth factor receptor 3 gene (FGFR3) A248C, S249C, G372C, and T375C mutations in bladder cancer. Genetics and Molecular Research: GMR 10, 86-95Google Scholar
13Sonvilla, G. et al. (2010) Fibroblast growth factor receptor 3-IIIc mediates colorectal cancer growth and migration. British Journal of Cancer 102, 1145-1156CrossRefGoogle ScholarPubMed
14Chesi, M. et al. (1997) Frequent translocation t(4;14)(p16.3;q32.3) in multiple myeloma is associated with increased expression and activating mutations of fibroblast growth factor receptor 3. Nature Genetics 16, 260-264Google Scholar
15Goriely, A. et al. (2005) Gain-of-function amino acid substitutions drive positive selection of FGFR2 mutations in human spermatogonia. Proceedings of the National Academy Science of the United States of America 102, 6051-6056Google Scholar
16Wilkie, A.O. (2005) Bad bones, absent smell, selfish testes: the pleiotropic consequences of human FGF receptor mutations. Cytokine and Growth Factor Reviews 16, 187-203CrossRefGoogle ScholarPubMed
17Eswarakumar, V.P., Lax, I. and Schlessinger, J. (2005) Cellular signaling by fibroblast growth factor receptors. Cytokine and Growth Factor Reviews 16, 139-149CrossRefGoogle ScholarPubMed
18Ornitz, D.M. (2005) FGF signaling in the developing endochondral skeleton. Cytokine and Growth Factor Reviews 16, 205-213CrossRefGoogle ScholarPubMed
19Chen, H. et al. (2007) A molecular brake in the kinase hinge region regulates the activity of receptor tyrosine kinases. Molecular Cell 27, 717-730CrossRefGoogle ScholarPubMed
20Colvin, J.S. et al. (1996) Skeletal overgrowth and deafness in mice lacking fibroblast growth factor receptor 3. Nature Genetics 12, 390-397CrossRefGoogle ScholarPubMed
21Deng, C. et al. (1996) Fibroblast growth factor receptor 3 is a negative regulator of bone growth. Cell 84, 911-921Google Scholar
22Naski, M.C. et al. (1998) Repression of hedgehog signaling and BMP4 expression in growth plate cartilage by fibroblast growth factor receptor 3. Development 125, 4977-4988CrossRefGoogle ScholarPubMed
23Garofalo, S. et al. (1999) Skeletal dysplasia and defective chondrocyte differentiation by targeted overexpression of fibroblast growth factor 9 in transgenic mice. Journal of Bone and Mineral Research: The Official Journal of the American Society for Bone and Mineral Research 14, 1909-1915Google Scholar
24Itoh, N. and Ornitz, D.M. (2008) Functional evolutionary history of the mouse Fgf gene family. Developmental Dynamics: An Official Publication of the American Association of Anatomists 237, 18-27CrossRefGoogle ScholarPubMed
25Ornitz, D.M. et al. (1996) Receptor specificity of the fibroblast growth factor family. Journal of Biological Chemistry 271, 15292-15297Google Scholar
26Liu, Z. et al. (2002) Coordination of chondrogenesis and osteogenesis by fibroblast growth factor 18. Genes and Development 16, 859-869CrossRefGoogle ScholarPubMed
27Ohbayashi, N. et al. (2002) FGF18 is required for normal cell proliferation and differentiation during osteogenesis and chondrogenesis. Genes and Development 16, 870-879CrossRefGoogle ScholarPubMed
28Liu, Z. et al. (2007) FGF18 is required for early chondrocyte proliferation, hypertrophy and vascular invasion of the growth plate. Developmental Biology 302, 80-91Google Scholar
29Krejci, P. et al. (2007) Fibroblast growth factors 1, 2, 17, and 19 are the predominant FGF ligands expressed in human fetal growth plate cartilage. Pediatric Research 61, 267-272CrossRefGoogle Scholar
30Chuang, C.Y. et al. (2010) Heparan sulfate-dependent signaling of fibroblast growth factor 18 by chondrocyte-derived perlecan. Biochemistry 49, 5524-5532CrossRefGoogle ScholarPubMed
31Plotnikov, A.N. et al. (1999) Structural basis for FGF receptor dimerization and activation. Cell 98, 641-650Google Scholar
32Hart, K.C., Robertson, S.C. and Donoghue, D.J. (2001) Identification of tyrosine residues in constitutively activated fibroblast growth factor receptor 3 involved in mitogenesis, Stat activation, and phosphatidylinositol 3-kinase activation. Molecular Biology of the Cell 12, 931-942Google Scholar
33Mohammadi, M., Olsen, S.K. and Ibrahimi, O.A. (2005) Structural basis for fibroblast growth factor receptor activation. Cytokine and Growth Factor Reviews 16, 107-137CrossRefGoogle ScholarPubMed
34Horton, W.A. and Degnin, C.R. (2009) FGFs in endochondral skeletal development. Trends in Endocrinology and Metabolism 20, 341-348Google Scholar
35Bae, J.H. and Schlessinger, J. (2010) Asymmetric tyrosine kinase arrangements in activation or autophosphorylation of receptor tyrosine kinases. Molecules and Cells 29, 443-448Google Scholar
36Su, W.C. et al. (1997) Activation of Stat1 by mutant fibroblast growth-factor receptor in thanatophoric dysplasia type II dwarfism. Nature 386, 288-292Google Scholar
37Legeai-Mallet, L. et al. (1998) Fibroblast growth factor receptor 3 mutations promote apoptosis but do not alter chondrocyte proliferation in thanatophoric dysplasia. Journal of Biological Chemistry 273, 13007-13014Google Scholar
38Sahni, M. et al. (1999) FGF signaling inhibits chondrocyte proliferation and regulates bone development through the STAT-1 pathway. Genes and Development 13, 1361-1366Google Scholar
39Hart, K.C. et al. (2000) Transformation and Stat activation by derivatives of FGFR1, FGFR3, and FGFR4. Oncogene 19, 3309-3320Google Scholar
40Choi, D.Y. et al. (2001) Fibroblast growth factor receptor 3 induces gene expression primarily through Ras-independent signal transduction pathways. Journal of Biological Chemistry 276, 5116-5122Google Scholar
41Murakami, S. et al. (2004) Constitutive activation of MEK1 in chondrocytes causes Stat1-independent achondroplasia-like dwarfism and rescues the Fgfr3-deficient mouse phenotype. Genes and Development 18, 290-305Google Scholar
42Zhang, R. et al. (2006) Constitutive activation of MKK6 in chondrocytes of transgenic mice inhibits proliferation and delays endochondral bone formation. Proceedings of the National Academy Science of the United States of America 103, 365-370Google Scholar
43Minina, E. et al. (2002) Interaction of FGF, Ihh/Pthlh, and BMP signaling integrates chondrocyte proliferation and hypertrophic differentiation. Developmental Cell 3, 439-449Google Scholar
44Dailey, L. et al. (2003) A network of transcriptional and signaling events is activated by FGF to induce chondrocyte growth arrest and differentiation. Journal of Cell Biology 161, 1053-1066Google Scholar
45Schibler, L. et al. (2009) New insight on FGFR3-related chondrodysplasias molecular physiopathology revealed by human chondrocyte gene expression profiling. PLoS ONE 4, e7633CrossRefGoogle ScholarPubMed
46Degnin, C.R., Laederich, M.B. and Horton, W.A. (2011) Ligand activation leads to regulated intramembrane proteolysis (RIP) of Fibroblast Growth Factor Receptor 3. Molecular Biology of the Cell 22, 3861-3873Google Scholar
47Lyu, J., Yamamoto, V. and Lu, W. (2008) Cleavage of the Wnt receptor Ryk regulates neuronal differentiation during cortical neurogenesis. Developmental Cell 15, 773-780Google Scholar
48Carpenter, G. and Liao, H.J. (2009) Trafficking of receptor tyrosine kinases to the nucleus. Experimental Cell Research 315, 1556-1566Google Scholar
49Parkhurst, C.N., Zampieri, N. and Chao, M.V. (2010) Nuclear localization of the p75 neurotrophin receptor intracellular domain. Journal of Biological Chemistry 285, 5361-5368Google Scholar
50Ben-Zvi, T. et al. (2006) Suppressors of cytokine signaling (SOCS) 1 and SOCS3 interact with and modulate fibroblast growth factor receptor signaling. Journal of Cell Science 119(Pt 2), 380-387CrossRefGoogle ScholarPubMed
51de Frutos, C.A. et al. (2007) Snail1 is a transcriptional effector of FGFR3 signaling during chondrogenesis and achondroplasias. Developmental Cell 13, 872-883CrossRefGoogle ScholarPubMed
52Potter, L.R., Abbey-Hosch, S. and Dickey, D.M. (2006) Natriuretic peptides, their receptors, and cyclic guanosine monophosphate-dependent signaling functions. Endocrine Reviews 27, 47-72Google Scholar
53Yasoda, A. et al. (2004) Overexpression of CNP in chondrocytes rescues achondroplasia through a MAPK-dependent pathway. Nature Medicine 10, 80-86Google Scholar
54Pejchalova, K., Krejci, P. and Wilcox, W.R. (2007) C-natriuretic peptide: an important regulator of cartilage. Molecular Genetics and Metabolism 92, 210-215Google Scholar
55Sellitti, D.F., Koles, N. and Mendonca, M.C. (2011) Regulation of C-type natriuretic peptide expression. Peptides 32, 1964-1971Google Scholar
56Bartels, C.F. et al. (2004) Mutations in the transmembrane natriuretic peptide receptor NPR-B impair skeletal growth and cause acromesomelic dysplasia, type Maroteaux. American Journal of Human Genetics 75, 27-34Google Scholar
57Yasoda, A. et al. (1998) Natriuretic peptide regulation of endochondral ossification. Evidence for possible roles of the C-type natriuretic peptide/guanylyl cyclase-B pathway. Journal of Biological Chemistry 273, 11695-11700Google Scholar
58Miyazawa, T. et al. (2002) Cyclic GMP-dependent protein kinase II plays a critical role in C-type natriuretic peptide-mediated endochondral ossification. Endocrinology 143, 3604-3610Google Scholar
59Krejci, P. et al. (2005) Interaction of fibroblast growth factor and C-natriuretic peptide signaling in regulation of chondrocyte proliferation and extracellular matrix homeostasis. Journal of Cell Science 118(Pt 21), 5089-5100Google Scholar
60Chusho, H. et al. (2001) Dwarfism and early death in mice lacking C-type natriuretic peptide. Proceedings of the National Academy of Sciences of the United States of America 98, 4016-4021Google Scholar
61Horton, W.A. (2006) Skeletal development. In Cell Signaling and Growth Factors in Development (Unsicker, K. and Krieglstein, K., eds), pp. 619-640, Wiley-VCH VerlagGmbH & Co., WeinheimGoogle Scholar
62Laederich, M.B. and Horton, W.A. (2010) Achondroplasia: pathogenesis and implications for future treatment. Current Opinion in Pediatrics 22, 516-523Google Scholar
63Webster, M.K. and Donoghue, D.J. (1996) Constitutive activation of fibroblast growth factor receptor 3 by the transmembrane domain point mutation found in achondroplasia. EMBO Journal 15, 520-527CrossRefGoogle ScholarPubMed
64You, M., Li, E. and Hristova, K. (2006) The achondroplasia mutation does not alter the dimerization energetics of the fibroblast growth factor receptor 3 transmembrane domain. Biochemistry 45, 5551-5556CrossRefGoogle Scholar
65Monsonego-Ornan, E. et al. (2000) The transmembrane mutation G380R in fibroblast growth factor receptor 3 uncouples ligand-mediated receptor activation from down-regulation. Molecular and Cellular Biology 20, 516-522CrossRefGoogle ScholarPubMed
66Cho, J.Y. et al. (2004) Defective lysosomal targeting of activated fibroblast growth factor receptor 3 in achondroplasia. Proceedings of the National Academy of Sciences of the United States of America 101, 609-614Google Scholar
67Guo, C. et al. (2008) Sprouty 2 disturbs FGFR3 degradation in thanatophoric dysplasia type II: a severe form of human achondroplasia. Cellular Signalling 20, 1471-1477CrossRefGoogle ScholarPubMed
68Degnin, C.R., Laederich, M.B. and Horton, W.A. (2011) Ligand activation leads to regulated intramembrane proteolysis of fibroblast growth factor receptor 3. Molecular Biology of the Cell 22, 3861-3873Google Scholar
69Kim, Y.S. et al. (2009) Update on Hsp90 inhibitors in clinical trial. Current Topics in Medicinal Chemistry 9, 1479-1492CrossRefGoogle Scholar
70Trepel, J. et al. (2010) Targeting the dynamic HSP90 complex in cancer. Nature Reviews. Cancer 10, 537-549Google Scholar
71Horton, W.A. (2003) Skeletal development: insights from targeting the mouse genome. Lancet 362, 560-569Google Scholar
72Kronenberg, H.M. (2003) Developmental regulation of the growth plate. Nature 423, 332-336Google Scholar
73Dailey, L. et al. (2005) Mechanisms underlying differential responses to FGF signaling. Cytokine and Growth Factor Reviews 16, 233-247Google Scholar
74Murakami, S. et al. (2000) Up-regulation of the chondrogenic Sox9 gene by fibroblast growth factors is mediated by the mitogen-activated protein kinase pathway. Proceedings of the National Academy of Sciences of the United States of America 97, 1113-1118Google Scholar
75Hartmann, J.T. et al. (2009) Tyrosine kinase inhibitors – a review on pharmacology, metabolism and side effects. Current Drug Metabolism 10, 470-481Google Scholar
76Collins, I. and Workman, P. (2006) Design and development of signal transduction inhibitors for cancer treatment:experience and challenges with kinase targets. Current Signal Transduction Therapy 1, 13-23Google Scholar
77Aviezer, D., Golembo, M. and Yayon, A. (2003) Fibroblast growth factor receptor-3 as a therapeutic target for Achondroplasia–genetic short limbed dwarfism. Current Drug Targets 4, 353-365Google Scholar
78Novartis (2011) Dovitinib (TKI258) and BGJ398. In http://www.novartisoncology.us/research/pipeline/tki258.jspGoogle Scholar
79Hall, P.S. and Cameron, D.A. (2009) Current perspective – trastuzumab. European Journal of Cancer 45, 12-18Google Scholar
80Rauchenberger, R. et al. (2003) Human combinatorial Fab library yielding specific and functional antibodies against the human fibroblast growth factor receptor 3. Journal of Biological Chemistry 278, 38194-38205CrossRefGoogle ScholarPubMed
81Cappellen, D. et al. (1999) Frequent activating mutations of FGFR3 in human bladder and cervix carcinomas. Nature Genetics 23, 18-20CrossRefGoogle ScholarPubMed
82Martinez-Torrecuadrada, J. et al. (2005) Targeting the extracellular domain of fibroblast growth factor receptor 3 with human single-chain Fv antibodies inhibits bladder carcinoma cell line proliferation. Clinical Cancer Research 11, 6280-6290Google Scholar
83Qing, J. et al. (2009) Antibody-based targeting of FGFR3 in bladder carcinoma and t(4;14)-positive multiple myeloma in mice. Journal of Clinical Investigation 119, 1216-1229Google Scholar
84Hadari, Y. and Schlessinger, J. (2009) FGFR3-targeted mAb therapy for bladder cancer and multiple myeloma. Journal of Clinical Investigation 119, 1077-1079Google Scholar
85Suda, M. et al. (1998) Skeletal overgrowth in transgenic mice that overexpress brain natriuretic peptide. Proceedings of the National Academy of Sciences of the United States of America 95, 2337-2342Google Scholar
86Kake, T. et al. (2009) Chronically elevated plasma C-type natriuretic peptide level stimulates skeletal growth in transgenic mice. American Journal of Physiology. Endocrinology and Metabolism 297, E1339-E1348Google Scholar
87Yasoda, A. et al. (2009) Systemic administration of C-type natriuretic peptide as a novel therapeutic strategy for skeletal dysplasias. Endocrinology 150, 3138-3144Google Scholar
88Hunt, P.J. et al. (1994) Bioactivity and metabolism of C-type natriuretic peptide in normal man. Journal of Clinical Endocrinology and Metabolism 78, 1428-1435Google ScholarPubMed
89Lorget, F. et al. BMN 111, a CNP analogue promotes skeletal growth and rescues dwarfism in two transgenic mouse models of Fgfr3-related chondrodysplasia. International Congress of Human Genetics (2011; Montreal)Google Scholar
90Olney, R.C. et al. (2006) Heterozygous mutations in natriuretic peptide receptor-B (NPR2) are associated with short stature. Journal of Clinical Endocrinology and Metabolism 91, 1229-1232Google Scholar
91Bocciardi, R. et al. (2007) Overexpression of the C-type natriuretic peptide (CNP) is associated with overgrowth and bone anomalies in an individual with balanced t(2;7) translocation. Human Mutation 28, 724-731Google Scholar
92Suga, S. et al. (1992) Receptor selectivity of natriuretic peptide family, atrial natriuretic peptide, brain natriuretic peptide, and C-type natriuretic peptide. Endocrinology 130, 229-239Google Scholar
93Davidson, B.L. and McCray, P.B. Jr (2011) Current prospects for RNA interference-based therapies. Nature Reviews. Genetics 12, 329-340Google Scholar
94Boudreau, R.L., Rodriguez-Lebron, E. and Davidson, B.L. (2011) RNAi medicine for the brain: progresses and challenges. Human Molecular Genetics 20(R1), R21-R27Google Scholar
95Lochmatter, D. and Mullis, P.E. (2011) RNA interference in mammalian cell systems. Hormone Research in Paediatrics 75, 63-69Google Scholar
96Hartl, F.U., Bracher, A. and Hayer-Hartl, M. (2011) Molecular chaperones in protein folding and proteostasis. Nature 475, 324-332Google Scholar
97Laederich, M.B. et al. (2011) Fibroblast growth factor receptor 3 (FGFR3) is a strong heat shock protein 90 (Hsp90) client: implications for therapeutic manipulation. Journal of Biological Chemistry 286, 19597-19604Google Scholar
98Calamia, V. et al. (2011) Hsp90beta inhibition modulates nitric oxide production and nitric oxide-induced apoptosis in human chondrocytes. BMC Musculoskeletal Disorders 12, 237Google Scholar
99Herzog, A. et al. (2011) Hsp90 and angiogenesis in bone disorders–lessons from the avian growth plate. American Journal of Physiology. Regulatory, Integrative and Comparative Physiology 301, R140-R147Google Scholar
100Pandit, S.G. et al. (2002) The fibroblast growth factor receptor, FGFR3, forms gradients of intact and degraded protein across the growth plate of developing bovine ribs. Biochemical Journal 361(Pt 2), 231-241CrossRefGoogle ScholarPubMed
101Wolfe, M.S. (2009) gamma-Secretase in biology and medicine. Seminars in Cell and Developmental Biology 20, 219-224Google Scholar
102Mulla, H. (2010) Understanding developmental pharmacodynamics: importance for drug development and clinical practice. Paediatric Drugs 12, 223-233Google Scholar
103Goldberg, M., Langer, R. and Jia, X. (2007) Nanostructured materials for applications in drug delivery and tissue engineering. Journal of Biomaterials Science. Polymer Edition 18, 241-268Google Scholar
104Alexis, F. et al. (2008) Factors affecting the clearance and biodistribution of polymeric nanoparticles. Molecular Pharmaceutics 5, 505-515Google Scholar
105Rothenfluh, D.A. et al. (2008) Biofunctional polymer nanoparticles for intra-articular targeting and retention in cartilage. Nature Materials 7, 248-254CrossRefGoogle ScholarPubMed

Further reading, resources and contacts

Little People of America (LPA), a support group for patients and parents of patients with achondroplasia and other forms of dwarfism, provides much information about the clinical management of achondroplasia as well as social and medical support for patients and their families: