#PAGE_PARAMS# #ADS_HEAD_SCRIPTS# #MICRODATA#

Hereditary renal cell carcinoma syndromes


Authors: Jiří Kolář 1;  Tomáš Pitra 1;  Kristýna Pivovarčíková 2;  Radka Jaklová 3;  Tomáš Zavoral 3;  Ivan Trávníček 1;  Hana Sedláčková 1;  Kristýna Procházková 4;  Tomáš Vaneček 2;  Ondřej Hes 2;  Milan Hora 1
Authors‘ workplace: Urologická klinika LF UK a FN Plzeň 1;  Šiklův ústav patologie LF UK a FN Plzeň 2;  Ústav lékařské genetiky LF UK a FN Plzeň 3;  Chirurgická klinika LF UK a FN Plzeň 4
Published in: Ces Urol 2020; 24(1): 26-41
Category: Review article

Overview

In recent years, there has been an increase in the number of defined hereditary syndromes, some of which may predispose to kidney cancer. Hereditary renal cell carcinoma syndromes are often suggested by early age of onset, family history of kidney cancer, multifocal or bilateral lesions and the presence of other extrarenal lesions typical for the respective syndrome. The most common encountered syn‑ dromes are von Hippel Lindau, Birt-Hogg-Dubé, hereditary papillary renal cell carcinoma, hereditary leiomyomatosis and renal cell carcinoma, succinate dehydrogenase deficient renal cell carcinoma, tuberous sclerosis complex and Cowden syndrome. Related to hereditary renal cancer syndromes, germline mutations in tumor suppressor genes (VHL, BAP1, PTEN, TSC1, TSC2) and protooncogenes (MET) are described. Different histological subtypes are found among hereditary kidney cancer syndromes. The degree of kidney cancer penetrance, tumor aggressiveness, metastatic potential widely vary and the treatment and surveillance of kidney cancer is different among hereditary syndromes.

Keywords:

Genetics – germline mutation – hereditary syndrome – Renal cell carcinoma – surveillance


Sources

1. Haas NB, Nathanson KL. Hereditary kidney cancer syndromes. Adv Chronic Kidney, DiS. 2014; 21(1): 81–90.

2. Leung C, Pan S, Shuch B. Management of renal cell carcinoma in young patients and patients with hereditary syndromes. Curr Opin Urol. 2016; 26(5): 396–404.

3. Huang KL, Mashl RJ, Wu Y, et al. Pathogenic Germline Variants in 10,389 Adult Cancers. Cell. 2018; 173(2): 355–70.e14.

4. Shuch B, Vourganti S, Ricketts CJ, et al. Defining early‑onset kidney cancer: implications for germline and somatic mutation testing and clinical management. J Clin Oncol. 2014; 32(5): 431–437.

5. Menko FH, Maher ER. Diagnosis and Management of Hereditary Renal Cell Cancer. Recent Results Cancer Res. 2016; 205: 85–104.

6. Gossage L, Eisen T, Maher ER. VHL, the story of a tumour suppressor gene. Nat Rev Cancer 2015; 15(1): 55–64.

7. Bader HL, Hsu T. Systemic VHL gene functions and the VHL disease. FEBS Lett. 2012; 586(11): 1562–1569.

8. Richard S, Graff J, Lindau J, Resche F. Von Hippel‑Lindau disease. Lancet. 2004; 363(9416): 1231–1234.

9. Ürge T, Hora M, Toufarová P, et al. Renální karcinom u nemocných s morbus von Hippel-Lindau. Ces Urol 2007; 11(2): 93–97.

10. Nielsen SM, Rhodes L, Blanco I, et al. Von Hippel‑Lindau Disease: Genetics and Role of Genetic Counseling in a Multiple Neoplasia Syndrome. J Clin Oncol. 2016;34(18): 2172–2181.

11. Lattouf JB, Pautler SE, Reaume MN, et al. Structured assessment and followup for patients with hereditary kidney tumour syndromes. Can Urol Assoc J. 2016; 10(7–8):E214–E22.

12. Maddock IR, Moran A, Maher ER, et al. A genetic register for von Hippel‑Lindau disease. J Med Genet1996; 33(2): 120–127.

13. Ong KR, Woodward ER, Killick P, et al. Genotype‑phenotype correlations in von Hippel‑Lindau disease.Hum Mutat. 2007; 28(2): 143–149.

14. Deml KF, Schildhaus HU, Compérat E, et al. Clear cell papillary renal cell carcinoma and renal angiomyoadenomatous tumor: two variants of a morphologic,immunohistochemical, and genetic distinctentity of renal cell carcinoma. Am J Surg Pathol. 2015; 39(7): 889–901.

15. Maher ER, Neumann HP, Richard S. Von Hippel‑Lindau disease: a clinical and scientific review. Eur JHum Genet. 2011; 19(6): 617–623.

16. Walther MM, Choyke PL, Glenn G, et al. Renal cancer in families with hereditary renal cancer: prospective analysis of a tumor size threshold for renal parenchymal sparing surgery. J Urol. 1999; 161(5): 1475–1479.

17. Duffey BG, Choyke PL, Glenn G, et al. The relationship between renal tumor size and metastases inpatients with von Hippel‑Lindau disease. J Urol. 2004; 172(1): 63–65.

18. Guo J, Tretiakova MS, Troxell ML, et al. Tuberous sclerosis‑associated renal cell carcinoma: a clinicopathologic study of 57 separate carcinomas in 18 patients. Am J Surg Pathol. 2014; 38(11): 1457–1467.

19. Vrtel R, Fillipová H, Vodicka R, et al. Tuberous sclerosis. Klin Onkol. 2009; 22(Suppl): S50–53.

20. Northrup H, Krueger DA, Group ITSCC. Tuberous sclerosis complex diagnostic criteria update: recommendations of the 2012 Iinternational Tuberous Sclerosis Complex Consensus Conference. PediatrNeurol. 2013; 49(4): 243–254.

21. Roach ES, Sparagana SP. Diagnosis of tuberous sclerosis complex. J Child Neurol. 2004; 19(9): 643–649.

22. Dixon BP, Hulbert JC, Bissler JJ. Tuberous sclerosis complex renal disease. Nephron Exp Nephrol. 2011;118(1): e15–20.

23. Tello R, Blickman JG, Buonomo C, Herrin J. Meta analysis of the relationship between tuberous sclerosis complex and renal cell carcinoma. Eur J Radiol. 1998; 27(2): 131–138.

24. Moch H, Ohashi R, Gandhi JS, Amin MB. Morphological clues to the appropriate recognition of hereditary renal neoplasms. Semin Diagn Pathol. 2018; 35(3): 184–192.

25. Palsgrove DN, Li Y, Pratilas CA, et al. Eosinophilic Solid and Cystic (ESC) Renal Cell Carcinomas HarborTSC Mutations: Molecular Analysis Supports an Expanding Clinicopathologic Spectrum. Am J Surg Pathol.2018; 42(9): 1166–1181.

26. Trpkov K, Hes O. New and emerging renal entities: a perspective post‑WHO 2016 classification. Histopathology 2019; 74(1): 31–59.

27. Trpkov K, Abou‑Ouf H, Hes O, et al. Eosinophilic Solid and Cystic Renal Cell Carcinoma (ESC RCC):Further Morphologic and Molecular Characterization of ESC RCC as a Distinct Entity. Am J Surg Pathol.2017; 41(10): 1299–308.

28. Carlo MI, Hakimi AA, Stewart GD, et al. Familial Kidney Cancer: Implications of New Syndromes and Molecular Insights. Eur Urol. 2019; 76(6): 754–764.

29. Dabora SL, Franz DN, Ashwal S, et al. Multicenter phase 2 trial of sirolimus for tuberous sclerosis: kidney angiomyolipomas and other tumors regress and VEGF- D levels decrease. PLoS One. 2011; 6(9): e23379.

30. El‑Hashemite N, Walker V, Zhang H, Kwiatkowski DJ. Loss of Tsc1 or Tsc2 induces vascular endothelial growth factor production through mammalian target of rapamycin. Cancer Res. 2003; 63(17): 5173–5177.

31. Chen YB, Mirsadraei L, Jayakumaran G, et al. Somatic Mutations of TSC2 or MTOR Characterize a Morphologically Distinct Subset of Sporadic Renal Cell Carcinoma With Eosinophilic and Vacuolated Cytoplasm. Am J Surg Pathol. 2019; 43(1): 121–131.

32. Franz DN, Budde K, Kingswood JC, et al. Effect of everolimus on skin lesions in patients treated for subependymal giant cell astrocytoma and renal angiomyolipoma: final 4-year results from the randomized EXIST-1 and EXIST-2 studies. J Eur Acad Dermatol Venereol. 2018; 32(10): 1796–1803.

33. Bissler JJ, Kingswood JC, Radzikowska E, et al. Everolimus long‑term use in patients with tuberous sclerosis complex: Four‑year update of the EXIST-2 study. PLoS One. 2017; 12(8): e0180939.

34. Bissler JJ, Kingswood JC, Radzikowska E, et al. Everolimus for angiomyolipoma associated with tuberous sclerosis complex or sporadic lymphangioleiomyomatosis (EXIST-2): a multicentre, randomised, double‑blind, placebo‑controlled trial. Lancet 2013; 381(9869): 817–824.

35. Staehler M, Sauter M, Helck A, et al. Nephron‑sparing resection of angiomyolipoma after sirolimus pretreatment in patients with tuberous sclerosis. Int Urol Nephrol. 2012; 44(6): 1657–1661.

36. Ürge T, Pitra T, Chudáček Z, et al. Nové trendy v léčbě renalního angiomyolipomu. Ces Urol. 2015; 19(2): 106–117.

37. Moch H, Cubilla AL, Humphrey PA, Reuter VE, Ulbright TM. The 2016 WHO Classification of Tumours of the Urinary System and Male Genital Organs‑Part A: Renal, Penile, and Testicular Tumours. Eur Urol.2016; 70(1): 93–105.

38. Hora M, Ürge T, Kalusová K, et al. Novelizovaná klasifikace nádorů ledvin 2013 (International Society of Urological Pathology Vancouver Classification of Renal Neoplasia). Ces Urol 2014; 18(1): 9–20.

39. Schmidt LS, Linehan WM. Hereditary leiomyomatosis and renal cell carcinoma. Int J Nephrol Reno‑vasc, DiS. 2014; 7: 253–260.

40. Trpkov K, Hes O, Agaimy A, et al. Fumarate Hydratase‑deficient Renal Cell Carcinoma Is Strongly Correlated With Fumarate Hydratase Mutation and Hereditary Leiomyomatosis and Renal Cell Carcinoma Syndrome. Am J Surg Pathol. 2016; 40(7): 865–875.

41. Pivovarcikova K, Martinek P, Grossmann P, et al. Fumarate hydratase deficient renal cell carcinoma: Chromosomal numerical aberration analysis of 12 cases. Ann Diagn Pathol. 2019; 39: 63–68.

42. Kuroda N, Ohe C, Kato I, et al. Review of hereditary leiomyomatosis renal cell carcinoma with focus on clinical and pathobiological aspects of renal tumors. Pol J Pathol. 2017; 68(4): 284–290.

43. Plevová P, Hladíková A, Tesařová M. Hereditary leiomyomatosis and renal cell cancer – HLRCC/multiple cutaneous and uterine leimomyomatosis – MCUL. Klin Onkol. 2012; 25 Suppl: S55–58.

44. Przybycin CG, Magi‑Galluzzi C, McKenney JK. Hereditary syndromes with associated renal neoplasia: a practical guide to histologic recognition in renal tumor resection specimens. Adv Anat Pathol. 2013; 20(4): 245–263.

45. Choyke PL, Walther MM, Glenn GM, et al. Imaging features of hereditary papillary renal cancers.J Comput Assist Tomogr. 1997; 21(5): 737–741.

46. Choueiri TK, Vaishampayan U, Rosenberg JE, et al. Phase II and biomarker study of the dual MET/ VEGFR2 inhibitor foretinib in patients with papillary renal cell carcinoma. J Clin Oncol. 2013; 31(2): 181–186.

47. Schmidt LS, Nickerson ML, Angeloni D, et al. Early onset hereditary papillary renal carcinoma: germline missense mutations in the tyrosine kinase domain of the met proto‑oncogene. J Urol. 2004; 172(4 Pt1): 1256–1261.

48. Menko FH, van Steensel MA, Giraud S, et al. Birt‑Hogg‑Dubé syndrome: diagnosis and management.Lancet Oncol. 2009; 10(12): 1199–1206.

49. Hartman TR, Nicolas E, Klein‑Szanto A, et al. The role of the Birt‑Hogg‑Dubé protein in mTOR activation and renal tumorigenesis. Oncogene 2009; 28(13): 1594–1604.

50. Maher ER. Hereditary renal cell carcinoma syndromes: diagnosis, surveillance and management. WorldJ Urol. 2018; 36(12): 1891–1898.

51. Křepelová A, Puchmajerová A, Vasovčák P, Chocholatý M. Birt‑Hogg‑Dubé syndrome. Klin Onkol.2012; 25(Suppl): S18–20.

52. Pavlovich CP, Walther MM, Eyler RA, et al. Renal tumors in the Birt‑Hogg‑Dubé syndrome. Am J SurgPathol. 2002; 26(12): 1542–1552.

53. He H, Trpkov K, Martinek P, et al. „High‑grade oncocytic renal tumor“: morphologic, immunohistochemical, and molecular genetic study of 14 cases. Virchows Arch. 2018; 473(6): 725–738.

54. Stamatakis L, Metwalli AR, Middelton LA, Marston Linehan W. Diagnosis and management of BHD‑associated kidney cancer. Fam Cancer 2013; 12(3): 397–402.

55. Musil Z, Vícha A, Zelinka T, et al. Hereditary pheochromocytoma and paraganglioma. Klin Onkol.2012; 25 Suppl: S21–26.

56. Ricketts C, Woodward ER, Killick P, et al. Germline SDHB mutations and familial renal cell carcinoma.J Natl Cancer Inst. 2008; 100(17): 1260–1262.

57. Kuroda N, Yorita K, Nagasaki M, et al. Review of succinate dehydrogenase‑deficient renal cell carcinoma with focus on clinical and pathobiological aspects. Pol J Pathol. 2016; 67(1): 3–7.

58. Gill AJ, Hes O, Papathomas T, et al. Succinate dehydrogenase (SDH)-deficient renal carcinoma: a morphologically distinct entity: a clinicopathologic series of 36 tumors from 27 patients. Am J Surg Pathol.2014; 38(12): 1588–1602.

59. Rednam SP, Erez A, Druker H, et al. Von Hippel‑Lindau and Hereditary Pheochromocytoma/Paraganglioma Syndromes: Clinical Features, Genetics, and Surveillance Recommendations in Childhood. ClinCancer Res. 2017; 23(12): e68–e75.

60. Mester JL, Zhou M, Prescott N, Eng C. Papillary renal cell carcinoma is associated with PTEN hamartoma tumor syndrome. Urology. 2012; 79(5): 1187.e1–7.

61. Farley MN, Schmidt LS, Mester JL, et al. A novel germline mutation in BAP1 predisposes to familialclear‑cell renal cell carcinoma. Mol Cancer Res. 2013; 11(9): 1061–1071.

62. Popova T, Hebert L, Jacquemin V, et al. Germline BAP1 mutations predispose to renal cell carcinomas.Am J Hum Genet. 2013; 92(6): 974–980.

63. Chen JD, Morrison C, Zhang C, et al. Hyperparathyroidism‑jaw tumour syndrome. J Intern Med. 2003;253(6): 634–642.

64. Hora M, Hes O, Eret V, et al. Translokační renální karcinom Xp11.2 typu ASPL/TFE3. Ces Urol 2008;12(3): 186–193.

Labels
Paediatric urologist Nephrology Urology
Login
Forgotten password

Enter the email address that you registered with. We will send you instructions on how to set a new password.

Login

Don‘t have an account?  Create new account

#ADS_BOTTOM_SCRIPTS#