Skip to main content

Advertisement

Log in

Fibrous Dysplasia of Bone and McCune–Albright Syndrome: A Bench to Bedside Review

  • Review
  • Published:
Calcified Tissue International Aims and scope Submit manuscript

Abstract

Fibrous dysplasia is an uncommon mosaic disorder in which bone is replaced by structurally unsound fibro-osseous tissue. It is caused by the sporadic post-zygotic activating mutations in GNAS, resulting in dysregulated GαS-protein signaling in affected tissues. This manifests on a broad clinical spectrum ranging from insignificant solitary lesions to severe disease with deformities, fractures, functional impairment, and pain. Fibrous dysplasia may present in isolation or in association with hyperfunctioning endocrinopathies and café-au-lait macules, known as McCune–Albright Syndrome. This review summarizes the current understanding of pathophysiology in fibrous dysplasia, describes key pre-clinical and clinical investigations, and details the current approach to diagnosis and management.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Fig. 1
Fig. 2
Fig. 3
Fig. 4
Fig. 5
Fig. 6
Fig. 7
Fig. 8

Similar content being viewed by others

References

  1. El-Mofty SK (2014) Fibro-osseous lesions of the craniofacial skeleton: an update. Head Neck Pathol 8(4):432–444

    Article  PubMed  PubMed Central  Google Scholar 

  2. Yang L et al (2017) Prevalence of different forms and involved bones of craniofacial fibrous dysplasia. J Craniofac Surg 28(1):21–25

    Article  PubMed  Google Scholar 

  3. Weinstein LS et al (1991) Activating mutations of the stimulatory G protein in the McCune-Albright syndrome. N Engl J Med 325(24):1688–1695

    Article  CAS  PubMed  Google Scholar 

  4. Michienzi S et al (2007) GNAS transcripts in skeletal progenitors: evidence for random asymmetric allelic expression of Gs alpha. Hum Mol Genet 16(16):1921–1930

    Article  CAS  PubMed  Google Scholar 

  5. Hilger D, Masureel M, Kobilka BK (2018) Structure and dynamics of GPCR signaling complexes. Nat Struct Mol Biol 25(1):4–12

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Lumbroso S et al (2004) Activating gsalpha mutations: analysis of 113 patients with signs of McCune-Albright syndrome–a European Collaborative Study. J Clin Endocrinol Metab 89(5):2107–2113

    Article  CAS  PubMed  Google Scholar 

  7. Bianco P et al (2000) Mutations of the GNAS1 gene, stromal cell dysfunction, and osteomalacic changes in non-McCune-Albright fibrous dysplasia of bone. J Bone Miner Res 15(1):120–128

    Article  CAS  PubMed  Google Scholar 

  8. Idowu BD et al (2007) A sensitive mutation-specific screening technique for GNAS1 mutations in cases of fibrous dysplasia: the first report of a codon 227 mutation in bone. Histopathology 50(6):691–704

    Article  CAS  PubMed  Google Scholar 

  9. Graziano MP, Gilman AG (1989) Synthesis in Escherichia coli of GTPase-deficient mutants of Gs alpha. J Biol Chem 264(26):15475–15482

    CAS  PubMed  Google Scholar 

  10. Landis CA et al (1989) GTPase inhibiting mutations activate the alpha chain of Gs and stimulate adenylyl cyclase in human pituitary tumours. Nature 340(6236):692–696

    Article  CAS  PubMed  Google Scholar 

  11. Masters SB et al (1989) Mutations in the GTP-binding site of GS alpha alter stimulation of adenylyl cyclase. J Biol Chem 264(26):15467–15474

    CAS  PubMed  Google Scholar 

  12. Hu Q, Shokat KM (2018) Disease-causing mutations in the G protein Galphas subvert the roles of GDP and GTP. Cell 173(5):1254–1264

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Celi FS et al (2008) The role of type 1 and type 2 5’-deiodinase in the pathophysiology of the 3,5,3’-triiodothyronine toxicosis of McCune-Albright syndrome. J Clin Endocrinol Metab 93(6):2383–2389

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Happle R (1986) The McCune-Albright syndrome: a lethal gene surviving by mosaicism. Clin Genet 29(4):321–324

    Article  CAS  PubMed  Google Scholar 

  15. Riminucci M et al (2006) Fibrous dysplasia as a stem cell disease. J Bone Miner Res 21(Suppl 2):P125–P131

    Article  CAS  PubMed  Google Scholar 

  16. Riminucci M et al (1997) Fibrous dysplasia of bone in the McCune-Albright syndrome: abnormalities in bone formation. Am J Pathol 151(6):1587–1600

    CAS  PubMed  PubMed Central  Google Scholar 

  17. Riminucci M et al (1999) The histopathology of fibrous dysplasia of bone in patients with activating mutations of the Gs alpha gene: site-specific patterns and recurrent histological hallmarks. J Pathol 187(2):249–258

    Article  CAS  PubMed  Google Scholar 

  18. Riminucci M et al (2003) Osteoclastogenesis in fibrous dysplasia of bone: in situ and in vitro analysis of IL-6 expression. Bone 33(3):434–442

    Article  CAS  PubMed  Google Scholar 

  19. Collins MT et al (2001) Renal phosphate wasting in fibrous dysplasia of bone is part of a generalized renal tubular dysfunction similar to that seen in tumor-induced osteomalacia. J Bone Miner Res 16(5):806–813

    Article  CAS  PubMed  Google Scholar 

  20. Riminucci M et al (2003) FGF-23 in fibrous dysplasia of bone and its relationship to renal phosphate wasting. J Clin Invest 112(5):683–692

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Shimada T et al (2004) FGF-23 is a potent regulator of vitamin D metabolism and phosphate homeostasis. J Bone Miner Res 19(3):429–435

    Article  CAS  PubMed  Google Scholar 

  22. Shimada T et al (2001) Cloning and characterization of FGF23 as a causative factor of tumor-induced osteomalacia. Proc Natl Acad Sci USA 98(11):6500–6505

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Bhattacharyya N et al (2012) Mechanism of FGF23 processing in fibrous dysplasia. J Bone Miner Res 27(5):1132–1141

    Article  CAS  PubMed  Google Scholar 

  24. Kuznetsov SA et al (2008) Age-dependent demise of GNAS-mutated skeletal stem cells and “normalization” of fibrous dysplasia of bone. J Bone Miner Res 23(11):1731–1740

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Bianco P et al (1998) Reproduction of human fibrous dysplasia of bone in immunocompromised mice by transplanted mosaics of normal and Gsalpha-mutated skeletal progenitor cells. J Clin Invest 101(8):1737–1744

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Hsiao EC et al (2008) Osteoblast expression of an engineered Gs-coupled receptor dramatically increases bone mass. Proc Natl Acad Sci USA 105(4):1209–1214

    Article  PubMed  PubMed Central  Google Scholar 

  27. Hsiao EC et al (2010) Gs G protein-coupled receptor signaling in osteoblasts elicits age-dependent effects on bone formation. J Bone Miner Res 25(3):584–593

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Saggio I et al (2014) Constitutive expression of Gsalpha(R201C) in mice produces a heritable, direct replica of human fibrous dysplasia bone pathology and demonstrates its natural history. J Bone Miner Res 29(11):2357–2368

    Article  CAS  PubMed  Google Scholar 

  29. Karaca A et al (2018) Constitutive stimulatory G protein activity in limb mesenchyme impairs bone growth. Bone 110:230–237

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Khan SK et al (2018) Induced Gnas(R201H) expression from the endogenous Gnas locus causes fibrous dysplasia by up-regulating Wnt/beta-catenin signaling. Proc Natl Acad Sci USA 115(3):E418–e418

    Article  CAS  PubMed  Google Scholar 

  31. Zhao X et al (2018) Expression of an active Galphas mutant in skeletal stem cells is sufficient and necessary for fibrous dysplasia initiation and maintenance. Proc Natl Acad Sci U S A 115(3):E428–E437

    Article  CAS  PubMed  Google Scholar 

  32. Hart ES et al (2007) Onset, progression, and plateau of skeletal lesions in fibrous dysplasia and the relationship to functional outcome. J Bone Miner Res 22(9):1468–1474

    Article  PubMed  Google Scholar 

  33. Kushchayeva YS et al (2018) Fibrous dysplasia for radiologists: beyond ground glass bone matrix. Insights Imaging 9(6):1035–1056

    Article  PubMed  PubMed Central  Google Scholar 

  34. Leet AI, Collins MT (2007) Current approach to fibrous dysplasia of bone and McCune-Albright syndrome. J Child Orthop 1(1):3–17

    Article  PubMed  PubMed Central  Google Scholar 

  35. Leet AI et al (2004) Fracture incidence in polyostotic fibrous dysplasia and the McCune-Albright syndrome. J Bone Miner Res 19(4):571–577

    Article  PubMed  Google Scholar 

  36. Ippolito E et al (2014) Radiographic classification of coronal plane femoral deformities in polyostotic fibrous dysplasia. Clin Orthop Relat Res 472(5):1558–1567

    Article  PubMed  Google Scholar 

  37. Leet AI et al (2004) Fibrous dysplasia in the spine: prevalence of lesions and association with scoliosis. 86(3):531–537

    Google Scholar 

  38. Mancini F et al (2009) Scoliosis and spine involvement in fibrous dysplasia of bone. Eur Spine J 18(2):196–202

    Article  PubMed  PubMed Central  Google Scholar 

  39. Berglund JA et al (2018) Scoliosis in Fibrous Dysplasia/McCune-Albright Syndrome: Factors Associated With Curve Progression and Effects of Bisphosphonates. J Bone Miner Res 33(9):1641–1648

    Article  CAS  PubMed  Google Scholar 

  40. Lee JS et al (2002) Normal vision despite narrowing of the optic canal in fibrous dysplasia. N Engl J Med 347(21):1670–1676

    Article  PubMed  Google Scholar 

  41. Burke AB, Collins MT, Boyce AM (2017) Fibrous dysplasia of bone: craniofacial and dental implications. Oral Dis 23(6):697–708

    Article  CAS  PubMed  Google Scholar 

  42. Chapurlat RD, Orcel P (2008) Fibrous dysplasia of bone and McCune–Albright syndrome. Best Pract Res Clin Rheumatol. 22(1):55–69

    Article  CAS  PubMed  Google Scholar 

  43. Becelli R et al (2002) Surgical treatment of fibrous dysplasia of the cranio-maxillo-facial area. Review of the literature and personal experience form 1984 to 1999. Minerva Stomatol 51(7-8):293–300

    CAS  PubMed  Google Scholar 

  44. Boyce AM et al (2013) Optic neuropathy in McCune-Albright syndrome: effects of early diagnosis and treatment of growth hormone excess. J Clin Endocrinol Metab 98(1):E126–E134

    Article  CAS  PubMed  Google Scholar 

  45. Amit M et al (2011) Surgery versus watchful waiting in patients with craniofacial fibrous dysplasia–a meta-analysis. PLoS One 6(9):e25179

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Boyce AM et al (2018) Association of hearing loss and otologic outcomes with fibrous dysplasia. JAMA Otolaryngol Head Neck Surg 144(2):102–107

    Article  PubMed  Google Scholar 

  47. Pan KS et al (2018) Chiari I malformation and basilar invagination in fibrous dysplasia: prevalence, mechanisms, and clinical implications. J Bone Miner Res. 33(11):1990–1998

    Article  CAS  PubMed  Google Scholar 

  48. Berglund JA et al (2018) Scoliosis in fibrous dysplasia/McCune-Albright syndrome: factors associated with curve progression and effects of bisphosphonates. J Bone Miner Res. 33(9):1641–1648

    Article  CAS  PubMed  Google Scholar 

  49. Kelly MH, Brillante B, Collins MT (2008) Pain in fibrous dysplasia of bone: age-related changes and the anatomical distribution of skeletal lesions. Osteoporos Int 19(1):57–63

    Article  CAS  PubMed  Google Scholar 

  50. Dumitrescu CE, Collins MT (2008) McCune-Albright syndrome. Orphanet J Rare Dis 3:12

    Article  PubMed  PubMed Central  Google Scholar 

  51. McCune DJ (1936) Osteitis fibrosa cystica; the case of a nine year old girl who also exhibits precocious puberty, multiple pigmentation of the skin and hyperthyroidism. Am J Dis Child. 52:743–744

    Google Scholar 

  52. Collins MT, Singer FR, Eugster E (2012) McCune-Albright syndrome and the extraskeletal manifestations of fibrous dysplasia. Orphanet J Rare Dis 7(Suppl 1):S4

    Article  PubMed  PubMed Central  Google Scholar 

  53. Boyce AM et al (2012) Characterization and management of testicular pathology in McCune-Albright syndrome. J Clin Endocrinol Metab 97(9):E1782–E1790

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Tessaris D et al (2012) Thyroid abnormalities in children and adolescents with McCune-Albright syndrome. Horm Res Paediatr 78(3):151–157

    Article  CAS  PubMed  Google Scholar 

  55. Salenave S et al (2014) Acromegaly and McCune-Albright syndrome. J Clin Endocrinol Metab 99(6):1955–1969

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Tessaris D et al (2018) Growth hormone-Insulin-like growth factor 1 axis hyperactivity on bone fibrous dysplasia in McCune-Albright Syndrome. Clin Endocrinol (Oxf) 89(1):56–64

    Article  CAS  Google Scholar 

  57. Boyce AM et al (2012) Optic neuropathy in McCune-Albright syndrome: effects of early diagnosis and treatment of growth hormone excess. J Clin Endocrinol Metab. 98(1):E126–E134

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. Boyce AM et al (2017) Association of hearing loss and otologic outcomes with fibrous dysplasia. JAMA Otolaryngol Head Neck Surg. 144(2):102–107

    Article  PubMed Central  Google Scholar 

  59. Brown RJ, Kelly MH, Collins MT (2010) Cushing syndrome in the McCune-Albright syndrome. J Clin Endocrinol Metab 95(4):1508–1515

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Carney JA, Young WF, Stratakis CA (2011) Primary bimorphic adrenocortical disease: cause of hypercortisolism in McCune-Albright syndrome. Am J Surg Pathol 35(9):1311–1326

    Article  PubMed  PubMed Central  Google Scholar 

  61. Ruggieri P et al (1994) Malignancies in fibrous dysplasia. Cancer 73(5):1411–1424

    Article  CAS  PubMed  Google Scholar 

  62. Majoor BC et al (2018) Increased risk of breast cancer at a young age in women with fibrous dysplasia. J Bone Miner Res 33(1):84–90

    Article  CAS  PubMed  Google Scholar 

  63. Collins MT et al (2003) Thyroid carcinoma in the McCune-Albright syndrome: contributory role of activating Gs alpha mutations. J Clin Endocrinol Metab 88(9):4413–4417

    Article  CAS  PubMed  Google Scholar 

  64. Gaujoux S et al (2014) Hepatobiliary and Pancreatic neoplasms in patients with McCune-Albright syndrome. J Clin Endocrinol Metab 99(1):E97–E101

    Article  PubMed  Google Scholar 

  65. Parvanescu A et al (2014) Lessons from McCune-Albright syndrome-associated intraductal papillary mucinous neoplasms: : GNAS-activating mutations in pancreatic carcinogenesis. JAMA Surg 149(8):858–862

    Article  PubMed  Google Scholar 

  66. Paul SM et al (2014) Disease severity and functional factors associated with walking performance in polyostotic fibrous dysplasia. Bone 60:41–47

    Article  PubMed  Google Scholar 

  67. de Castro LF et al (2018) Activation of RANK/RANKL/OPG pathway is involved in the pathophysiology of fibrous dysplasia and associated with disease burden. J Bone Miner Res. 34(2):290–294

    Article  CAS  PubMed  Google Scholar 

  68. Chapurlat RD et al (2004) Treatment of fibrous dysplasia of bone with intravenous pamidronate: long-term effectiveness and evaluation of predictors of response to treatment. Bone 35(1):235–242

    Article  CAS  PubMed  Google Scholar 

  69. Plotkin H et al (2003) Effect of pamidronate treatment in children with polyostotic fibrous dysplasia of bone. J Clin Endocrinol Metab 88(10):4569–4575

    Article  CAS  PubMed  Google Scholar 

  70. Majoor BC et al (2017) Outcome of long-term bisphosphonate therapy in McCune-Albright syndrome and polyostotic fibrous dysplasia. J Bone Miner Res 32(2):264–276

    Article  CAS  PubMed  Google Scholar 

  71. Parisi MS, Oliveri B, Mautalen CA (2003) Effect of intravenous pamidronate on bone markers and local bone mineral density in fibrous dysplasia. Bone 33(4):582–588

    Article  CAS  PubMed  Google Scholar 

  72. Matarazzo P et al (2002) Pamidronate treatment in bone fibrous dysplasia in children and adolescents with McCune-Albright syndrome. J Pediatr Endocrinol Metab 15(Suppl 3):929–937

    CAS  PubMed  Google Scholar 

  73. Florenzano P et al (2019) Age-related changes and effects of bisphosphonates on bone turnover and disease progression in fibrous dysplasia of bone. J Bone Miner Res. https://doi.org/10.1002/jbmr.3649

    Article  PubMed  Google Scholar 

  74. Boyce AM et al (2014) A randomized, double blind, placebo-controlled trial of alendronate treatment for fibrous dysplasia of bone. J Clin Endocrinol Metab 99(11):4133–4140

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  75. Boyce, A.M., et al., Fibrous Dysplasia/McCune-Albright Syndrome, in GeneReviews((R)), M.P. Adam, et al., Editors. 1993, University of Washington, Seattle University of Washington, Seattle. GeneReviews is a registered trademark of the University of Washington, Seattle. All rights reserved.: Seattle (WA).

  76. Metwally T et al (2016) Fibrous dysplasia and medication-related osteonecrosis of the jaw. J Oral Maxillofac Surg 74(10):1983–1999

    Article  PubMed  PubMed Central  Google Scholar 

  77. Boyce AM et al (2012) Denosumab treatment for fibrous dysplasia. J Bone Miner Res 27(7):1462–1470

    Article  CAS  PubMed  Google Scholar 

  78. Ganda K, Seibel MJ (2014) Rapid biochemical response to denosumab in fibrous dysplasia of bone: report of two cases. Osteoporos Int 25(2):777–782

    Article  CAS  PubMed  Google Scholar 

  79. Benhamou J, Gensburger D, Chapurlat R (2014) Transient improvement of severe pain from fibrous dysplasia of bone with denosumab treatment. Joint Bone Spine 81(6):549–550

    Article  PubMed  Google Scholar 

  80. Stanton RP et al (2012) The surgical management of fibrous dysplasia of bone. Orphanet J Rare Dis 7(Suppl 1):S1

    Article  PubMed  PubMed Central  Google Scholar 

  81. Majoor BC et al (2017) What is the role of allogeneic cortical strut grafts in the treatment of fibrous dysplasia of the proximal femur? Clin Orthop Relat Res 475(3):786–795

    Article  PubMed  Google Scholar 

  82. Leet AI et al (2016) Bone-grafting in polyostotic fibrous dysplasia. J Bone Joint Surg Am 98(3):211–219

    Article  PubMed  PubMed Central  Google Scholar 

  83. Boyce AM et al (2016) Surgical management of polyostotic craniofacial fibrous dysplasia: long-term outcomes and predictors for postoperative regrowth. Plast Reconstr Surg 137(6):1833–1839

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  84. Estrada A et al (2016) Long-term outcomes of letrozole treatment for precocious puberty in girls with McCune-Albright syndrome. Eur J Endocrinol 175(5):477–483

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  85. Eugster EA et al (2003) Tamoxifen treatment for precocious puberty in McCune–Albright syndrome: a multicenter trial. J Pediatr 143(1):60–66

    Article  CAS  PubMed  Google Scholar 

  86. Salenave S et al (2014) Acromegaly and McCune–Albright syndrome. J Clin Endocrinol Metab 99(6):1955–1969

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  87. Vortmeyer AO et al (2012) Somatic GNAS mutation causes widespread and diffuse pituitary disease in acromegalic patients with McCune–Albright syndrome. J Clin Endocrinol Metab 97(7):2404–2413

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  88. Liu F et al (2011) A case of McCune–Albright syndrome associated with pituitary GH adenoma: therapeutic process and autopsy. J Pediatr Endocrinol Metab 24(5–6):283–287

    PubMed  Google Scholar 

  89. Hansen MR, Moffat JC (2003) Osteosarcoma of the skull base after radiation therapy in a PATIENT with McCune–Albright syndrome: case report. Skull Base 13(2):79–83

    Article  PubMed  PubMed Central  Google Scholar 

  90. Carpenter TO et al (2011) A clinician’s guide to X-linked hypophosphatemia. J Bone Miner Res 26(7):1381–1388

    Article  PubMed  Google Scholar 

  91. Chapurlat RD et al (2012) Pathophysiology and medical treatment of pain in fibrous dysplasia of bone. Orphanet J Rare Dis 7(Suppl 1):S3

    Article  PubMed  PubMed Central  Google Scholar 

  92. de Boysson H et al (2015) Tocilizumab in the treatment of a polyostotic variant of fibrous dysplasia of bone. Rheumatology 54(9):1747–1749

    Article  PubMed  Google Scholar 

  93. Bhattacharyya N et al (2014) A high throughput screening assay system for the identification of small molecule inhibitors of gsp. PLoS ONE 9(3):e90766

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Conflict of interest

The NIDCR receives funding from Amgen, Inc for collaborative research projects related to fibrous dysplasia.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Alison M. Boyce.

Additional information

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Hartley, I., Zhadina, M., Collins, M.T. et al. Fibrous Dysplasia of Bone and McCune–Albright Syndrome: A Bench to Bedside Review. Calcif Tissue Int 104, 517–529 (2019). https://doi.org/10.1007/s00223-019-00550-z

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s00223-019-00550-z

Keywords

Navigation