Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Article
  • Published:

A somatic activating KRAS variant identified in an affected lesion of a patient with Gorham–Stout disease

Abstract

Gorham–Stout disease (GSD), a rare disorder of unknown etiology, is characterized by massive osteolysis that is associated with proliferation and dilation of lymphatic vessels. Variants in cancer-associated genes have been described in complex lymphatic anomalies. To explore the pathogenesis of GSD, we performed the amplicon-based deep sequencing on 50 cancer-related genes to assay affected tissues from the six patients with GSD. In one patient, a somatic activating KRAS c.182A > G variant (p.Q61R) was detected in 1% of the tissue sample. Conversely, the mutant allele was not detected in uninvolved normal skin and blood samples. Histopathology of the patient’s tissue sample showed proliferation of abnormal lymphatic and blood vascular endothelial cells, osteoclasts, and activated macrophages. The activating KRAS variant is a known ‘hotspot’ variant, frequently identified in several types of human cancer. This is the first report of identifying a pathogenic variant in a patient with GSD. This finding may set the stage for elucidation of pathophysiology and the development of novel therapies for GSD.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Fig. 1
Fig. 2

Similar content being viewed by others

References

  1. Trenor CC 3rd, Chaudry G. Complex lymphatic anomalies. Semin Pediatr Surg. 2014;23:186–90.

    Article  Google Scholar 

  2. Dellinger MT, Garg N, Olsen BR. Viewpoints on vessels and vanishing bones in Gorham–Stout disease. Bone. 2014;63:47–52.

    Article  Google Scholar 

  3. Manevitz-Mendelson E, Leichner GS, Barel O, Davidi-Avrahami I, Ziv-Strasser L, Eyal E, et al. Somatic NRAS mutation in patient with generalized lymphatic anomaly. Angiogenesis. 2018;21:287–98.

    Article  CAS  Google Scholar 

  4. Barclay SF, Inman KW, Luks VL, McIntyre JB, Al-Ibraheemi A, Church AJ, et al. A somatic activating NRAS variant associated with kaposiform lymphangiomatosis. Genet Med. 2019;21:1517–24.

    Article  CAS  Google Scholar 

  5. Ozeki M, Aoki Y, Nozawa A, Yasue S, Endo S, Hori Y, et al. Detection of NRAS mutation in cell-free DNA biological fluids from patients with kaposiform lymphangiomatosis. Orphanet J Rare Dis. 2019;14:215.

    Article  Google Scholar 

  6. Rodriguez-Laguna L, Agra N, Ibañez K, Oliva-Molina G, Gordo G, Khurana N, et al. Somatic activating mutations in PIK3CA cause generalized lymphatic anomaly. J Exp Med. 2019;216:407–18.

    Article  CAS  Google Scholar 

  7. Muñoz-Maldonado C, Zimmer Y, Medová M. A comparative analysis of individual RAS mutations in cancer biology. Front Oncol. 2019;9:1088.

    Article  Google Scholar 

  8. Samuels Y, Ericson K. Oncogenic PI3K and its role in cancer. Curr Opin Oncol. 2006;18:77–82.

    Article  CAS  Google Scholar 

  9. Heffez L, Doku HC, Carter BL, Feeney JE. Perspectives on massive osteolysis. Report of a case and review of the literature. Oral Surg Oral Med Oral Pathol. 1983;55:331–43.

    Article  CAS  Google Scholar 

  10. Nozawa A, Ozeki M, Hori T, Kato H, Ohe N, Fukao T. Fatal progression of Gorham–Stout Disease with skull base osteomyelitis and lateral medullary syndrome. Intern Med. 2019;58:1929–33.

    Article  Google Scholar 

  11. Aoki Y, Niihori T, Inoue S, Matsubara Y. Recent advances in RASopathies. J Hum Genet. 2016;61:33–39.

    Article  CAS  Google Scholar 

  12. Nguyen HL, Boon LM, Vikkula M. Vascular anomalies caused by abnormal signaling within endothelial cells: targets for novel therapies. Semin Intervent Radio. 2017;34:233–8.

    Article  Google Scholar 

  13. Li QF, Decker-Rockefeller B, Bajaj A, Pumiglia K. Activation of Ras in the vascular endothelium induces brain vascular malformations and hemorrhagic stroke. Cell Rep. 2018;24:2869–82.

    Article  CAS  Google Scholar 

  14. Nikolaev SI, Vetiska S, Bonilla X, Boudreau E, Jauhiainen S, Rezai Jahromi B, et al. Somatic activating KRAS mutations in arteriovenous malformations of the brain. N Engl J Med. 2018;378:250–61.

    Article  CAS  Google Scholar 

  15. Priemer DS, Vortmeyer AO, Zhang S, Chang HY, Curless KL, Cheng L. Activating KRAS mutations in arteriovenous malformations of the brain: frequency and clinicopathologic correlation. Hum Pathol. 2019;89:33–39.

    Article  CAS  Google Scholar 

  16. Siegel DH, Cottrell CE, Streicher JL, Schilter KF, Basel DG, Baselga E, et al. Analyzing the genetic spectrum of vascular anomalies with overgrowth via cancer genomics. J Investig Dermatol. 2018;138:957–67.

    Article  CAS  Google Scholar 

  17. Ozeki M, Fukao T. Generalized lymphatic anomaly and Gorham–Stout disease: overview and recent insights. Adv Wound Care. 2019;8:230–45.

    Article  Google Scholar 

  18. Colucci S, Taraboletti G, Primo L, Viale A, Roca C, Valdembri D, et al. Gorham–Stout syndrome: a monocyte-mediated cytokine propelled disease. J Bone Min Res. 2006;21:207–18.

    Article  CAS  Google Scholar 

  19. Rak J, Mitsuhashi Y, Bayko L, Filmus J, Shirasawa S, Sasazuki T, et al. Mutant ras oncogenes upregulate VEGF/VPF expression: implications for induction and inhibition of tumor angiogenesis. Cancer Res. 1995;55:4575–80.

    CAS  PubMed  Google Scholar 

  20. Zhang X, Gaspard JP, Chung DC. Regulation of vascular endothelial growth factor by the Wnt and K-ras pathways in colonic neoplasia. Cancer Res. 2001;61:6050–4.

    CAS  PubMed  Google Scholar 

  21. Ancrile B, Lim KH, Counter CM. Oncogenic Ras-induced secretion of IL6 is required for tumorigenesis. Genes Dev. 2007;21:1714–9.

    Article  CAS  Google Scholar 

  22. Dias Carvalho P, Guimarães CF, Cardoso AP, Mendonça S, Costa ÂM, Oliveira MJ, et al. KRAS oncogenic signaling extends beyond cancer cells to orchestrate the microenvironment. Cancer Res. 2018;78:7–14.

    Article  CAS  Google Scholar 

  23. Franco-Barrera MJ, Zavala-Cerna MG, Aguilar-Portillo G, Sánchez-Gomez DB, Torres-Bugarin O, Franco-Barrera MA, et al. Gorham–Stout disease: a clinical case report and immunological mechanisms in bone erosion. Clin Rev Allergy Immunol. 2017;52:125–32.

    Article  CAS  Google Scholar 

  24. Illeez OG, Ozkan K, Ozkan FU, Bostan AB, Akpinar F, Bilgic B, et al. Zoledronic acid: treatment option for Gorham–Stout disease. Orthopade. 2018;47:1032–5.

    Article  Google Scholar 

  25. Adams DM, Trenor CC 3rd, Hammill AM, Vinks AA, Patel MN, Chaudry G, et al. Efficacy and safety of sirolimus in the treatment of complicated vascular anomalies. Pediatrics. 2016;137:e20153257.

    Article  Google Scholar 

Download references

Acknowledgements

We thank Dr. Saori Endo, Dr. Shiho Yasue, Dr. Akihito Nagano, and Dr. Toshiyuki Fukao of Gifu University, Dr. Yasuyuki Shibuya of Nagoya City University Hospital, Dr. Koji Yokoyama of Japanese Red Cross Wakayama Medical Center, Dr. Masataka Takahashi of the University of Tokyo Hospital, and Dr. Mitsuharu Fukazawa of Tagawa Municipal Hospital for their cooperation in the data analyses. We thank Dr. Koki Nagai, Dr. Taiki Abe, Ms. Yoko Tateda in Tohoku University for providing technical assistance of NGS analysis. We also acknowledge the technical support of the Biomedical Research Core of Tohoku University Graduate School of Medicine. We would like to thank Editage (www.editage.com) for English language editing.

Funding

The present study was supported partly by the Clinical Research-Clinical Trial Promotion Research Project (18lk0201055h0003), Practical Research Project for Rare/Intractable Diseases (18ek0109277h0002 and 18ek0109278h0002) from Japan’s Agency for Medical Research and Development (AMED), and a research grant from the Naito Foundation.

Author information

Authors and Affiliations

Authors

Contributions

AN, MO, and YA conceived the study and participated in its design. AN, TN, and YA performed the NGS and genetic analysis of the patients. NS and TM performed the pathological analysis. AN, MO, and YA wrote the paper. All authors read and approved the final paper.

Corresponding authors

Correspondence to Michio Ozeki or Yoko Aoki.

Ethics declarations

Conflict of interest

The authors declare that they have no conflict of interest.

Ethical approval

This study was approved by the Ethics Committees of the Gifu University School of Medicine and Tohoku University School of Medicine.

Informed consent

Informed consent was obtained from the patients or their legal guardians for their inclusion in the study and subsequent publication.

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

Nozawa, A., Ozeki, M., Niihori, T. et al. A somatic activating KRAS variant identified in an affected lesion of a patient with Gorham–Stout disease. J Hum Genet 65, 995–1001 (2020). https://doi.org/10.1038/s10038-020-0794-y

Download citation

  • Received:

  • Revised:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s10038-020-0794-y

This article is cited by

Search

Quick links