Familial renal cancer: molecular genetics and surgical management

Glen W Barrisford, Eric A Singer, Inger L Rosner, W Marston Linehan, Gennady Bratslavsky, Glen W Barrisford, Eric A Singer, Inger L Rosner, W Marston Linehan, Gennady Bratslavsky

Abstract

Familial renal cancer (FRC) is a heterogeneous disorder comprised of a variety of subtypes. Each subtype is known to have unique histologic features, genetic alterations, and response to therapy. Through the study of families affected by hereditary forms of kidney cancer, insights into the genetic basis of this disease have been identified. This has resulted in the elucidation of a number of kidney cancer gene pathways. Study of these pathways has led to the development of novel targeted molecular treatments for patients affected by systemic disease. As a result, the treatments for families affected by von Hippel-Lindau (VHL), hereditary papillary renal carcinoma (HPRC), hereditary leiomyomatosis renal cell carcinoma (HLRCC), and Birt-Hogg-Dubé (BHD) are rapidly changing. We review the genetics and contemporary surgical management of familial forms of kidney cancer.

Figures

Figure 1
Figure 1
Histopathology of the most common malignant renal neoplasms. (a) Clear cell; (b) papillary type 1; (c) papillary type 2; and (d) chromophobe. (From Linehan et al. [10], with permission.)
Figure 2
Figure 2
The VHL complex targets HIF-1α and HIF-2α for ubiquitin-mediated degradation. In clear cell RCC, an alteration in the VHL gene in the α or β domain disrupts HIF degradation. HIF overaccumulates leading to increased transcription of downstream genes. (a) VHL alteration; (b) VHL/HIF pathway molecular targeting; and (c) VHL/HIF downstream molecular targeting. (From Linehan and Zbar [23], with permission.)
Figure 3
Figure 3
Alterations in the intracellular tyrosine kinase domain of the MET proto-oncogene are found in patients with HPRC. These mutations result in the activation of the MET pathway. (From Linehan et al. [10], with permission.)
Figure 4
Figure 4
In a normoxic environment, HIF is hydroxylated by HPH allowing the VHL complex to initiate ubiquitin-mediated breakdown in the proteosome. In HLRCC, FH alteration results in a buildup of fumarate. Fumarate competitively inhibits HPH allowing a rise in HIF levels and subsequent transcription of downstream genes. (From Pfaffenroth and Linehan [59], with permission.)
Figure 5
Figure 5
FLCN pathway. (a) FLCN is the gene associated with BHD. Normal FCLN protein complexes with FNIP1, FNIP2, and AMPK. This complex is phosphorylated by a rapamycin-sensitive kinase (mTORC1). (b) When FLCN is altered, it fails to complex and allows activation of AKT, mTORC1, and mTORC2. (From Hasumi et al. [69], with permission.)

References

    1. Jemal A, Bray F, Center MM, Ferlay J, Ward E, Forman D. Global cancer statistics. CA: Cancer Journal for Clinicians. 2011;61(2):69–90.
    1. Jemal A, Siegel R, Xu J, Ward E. Cancer statistics, 2010. CA: Cancer Journal for Clinicians. 2010;60(5):277–300.
    1. Altekruse S, Kosary C, Krapcho M, et al. Seer cancer statistics review, 1975–2007, National Cancer Institute. 2009, .
    1. Hollingsworth JM, Miller DC, Daignault S, Hollenbeck BK. Rising incidence of small renal masses: a need to reassess treatment effect. Journal of the National Cancer Institute. 2006;98(18):1331–1334.
    1. Chow WH, Devesa SS, Warren JL, Fraumeni JF. Rising incidence of renal cell cancer in the United States. Journal of the American Medical Association. 1999;281(17):1628–1631.
    1. Yagoda A, Petrylak D, Thompson S. Cytotoxic chemotherapy for advanced renal cell carcinoma. Urologic Clinics of North America. 1993;20(2):303–321.
    1. Nelson EC, Evans CP, Lara PN. Renal cell carcinoma: current status and emerging therapies. Cancer Treatment Reviews. 2007;33(3):299–313.
    1. Uchida K, Miyao N, Masumori N, et al. Recurrence of renal cell carcinoma more than 5 years after nephrectomy. International Journal of Urology. 2002;9(1):19–23.
    1. Kovacs G, Akhtar M, Beckwith BJ, et al. The Heidelberg classification of renal cell tumours. The Journal of pathology. 1997;183(2):131–133.
    1. Linehan WM, Walther MM, Zbar B. The genetic basis of cancer of the kidney. Journal of Urology. 2003;170(6):2163–2172.
    1. Kovacs G, Emanuel A, Neumann HPH, Kung HF. Cytogenetics of renal cell carcinomas associated with von Hippel-Lindau disease. Genes Chromosomes and Cancer. 1991;3(4):256–262.
    1. Linehan WM, Vasselli J, Srinivasan R, et al. Genetic basis of cancer of the kidney: disease-specific approaches to therapy. Clinical Cancer Research. 2004;10(18, part 2):6282S–6289S.
    1. Rosner I, Bratslavsky G, Pinto PA, Linehan WM. The clinical implications of the genetics of renal cell carcinoma. Urologic Oncology. 2009;27(2):131–136.
    1. Coleman JA, Russo P. Hereditary and familial kidney cancer. Current Opinion in Urology. 2009;19(5):478–485.
    1. Rini BI, Campbell SC, Rathmell WK. Renal cell carcinoma. Current Opinion in Oncology. 2006;18(3):289–296.
    1. Zbar B, Glenn G, Merino M, et al. Familial renal carcinoma: clinical evaluation, clinical subtypes and risk of renal carcinoma development. Journal of Urology. 2007;177(2):461–465.
    1. Robson ME, Storm CD, Weitzel J, Wollins DS, Offit K. American Society of Clinical Oncology Policy Statement update: genetic and genomic testing for cancer susceptibility. Journal of Clinical Oncology. 2010;28(5):893–901.
    1. Linehan WM, Bratslavsky G, Pinto PA, et al. Molecular diagnosis and therapy of kidney cancer. Annual Review of Medicine. 2010;61:329–343.
    1. Singer EA, Gupta GN, Srinivasan R. Update on targeted therapies for clear cell renal cell carcinoma. Current Opinion in Oncology. 2011;23(3):283–289.
    1. Di Lorenzo G, Autorino R, Sternberg CN. Metastatic renal cell carcinoma: recent advances in the targeted therapy era. European Urology. 2009;56(6):959–971.
    1. Rini BI. Metastatic renal cell carcinoma: many treatment options, one patient. Journal of Clinical Oncology. 2009;27(19):3225–3234.
    1. Singer EA, Bratslavsky G, Linehan WM, Srinivasan R. Targeted therapies for non-clear renal cell carcinoma. Targeted Oncology. 2010;5(2):119–129.
    1. Linehan WM, Zbar B. Focus on kidney cancer. Cancer Cell. 2004;6(3):223–228.
    1. Walther MM, Lubensky IA, Venzon D, Zbar B, Linehan WM, Marshall FF. Prevalence of microscopic lesions in grossly normal renal parenchyma from patients with Von Hippel-Lindau disease, sporadic renal cell carcinoma and no renal disease: clinical implications. Journal of Urology. 1995;154(6):2010–2015.
    1. Maher ER, Iselius L, Yates JRW, et al. Von Hippel-Lindau disease: a genetic study. Journal of Medical Genetics. 1991;28(7):443–447.
    1. Glenn G, Choyke P, Zbar B, Linehan WM. Problems in Urologic Surgery: Benign and Malignant Tumors of the Kidney. Philadelphia, Pa, USA: J. B. Lippincott; 1990. Von hippel-lindau disease: clinical review and molecular genetics; pp. 312–330.
    1. Tory K, Brauch H, Linehan M, et al. Specific genetic change in tumors associated with von Hippel-Lindau disease. Journal of the National Cancer Institute. 1989;81(14):1097–1101.
    1. Hosoe S, Brauch H, Latif F, et al. Localization of the von Hippel-Lindau disease gene to a small region of chromosome 3. Genomics. 1990;8(4):634–640.
    1. Latif F, Tory K, Gnarra J, et al. Identification of the von Hippel-Lindau disease tumor suppressor gene. Science. 1993;260(5112):1317–1320.
    1. Zbar B, Brauch H, Talmadge C, Linehau M. Loss of alleles of loci on the short arm of chromosome 3 in renal cell carcinoma. Nature. 1987;327(6124):721–724.
    1. Stolle C, Glenn G, Zbar B, et al. Improved detection of germline mutations in the von Hippel-Lindau disease tumor suppressor gene. Human Mutation. 1998;12(6):417–423.
    1. Gnarra JR, Tory K, Weng Y, et al. Mutations of the VHL tumour suppressor gene in renal carcinoma. Nature Genetics. 1994;7(1):85–90.
    1. Kai RO, Woodward ER, Killick P, Lim C, Macdonald F, Maher ER. Genotype-phenotype correlations in von Hippel-Lindau disease. Human Mutation. 2007;28(2):143–149.
    1. Gallou C, Chauveau D, Richard S, et al. Genotype-phenotype correlation in von Hippel-Lindau families with renal lesions. Human Mutation. 2004;24(3):215–224.
    1. Hartmut PH, Neumann PH, Zbar B. Renal cysts, renal cancer and von Hippel-Lindau disease. Kidney International. 1997;51(1):16–26.
    1. Chen F, Kishida T, Yao M, et al. Germline mutations in the von Hippel-Lindau disease tumor suppressor gene: correlations with phenotype. Human Mutation. 1995;5(1):66–75.
    1. Maranchie JK, Afonso A, Albert PS, et al. Solid renal tumor severity in von Hippel Lindau disease is related to germline deletion length and location. Human Mutation. 2004;23(1):40–46.
    1. Duan DR, Pause A, Burgess WH, et al. Inhibition of transcription elongation by the VHL tumor suppressor protein. Science. 1995;269(5229):1402–1406.
    1. Kibel A, Iliopoulos O, DeCaprio JA, Kaelin WG. Binding of the von Hippel-Lindau tumor suppressor protein to Elongin B and C. Science. 1995;269(5229):1444–1446.
    1. Pause A, Lee S, Worrell RA, et al. The von Hippel-Lindau tumor-suppressor gene product forms a stable complex with human CUL-2, a member of the Cdc53 family of proteins. Proceedings of the National Academy of Sciences of the United States of America. 1997;94(6):2156–2161.
    1. Brugarolas J, Kaelin WG. Dysregulation of HIF and VEGF is a unifying feature of the familial hamartoma syndromes. Cancer Cell. 2004;6(1):7–10.
    1. Iliopoulos O, Levy AP, Jiang C, Kaelin WG, Goldberg MA. Negative regulation of hypoxia-inducible genes by the von Hippel-Lindau protein. Proceedings of the National Academy of Sciences of the United States of America. 1996;93(20):10595–10599.
    1. Singer EA, Bratslavsky G, Middelton L, Srinivasan R, Linehan WM. Impact of genetics on the diagnosis and treatment of renal cancer. Current Urology Reports. 2011;12(1):47–55.
    1. Kaelin WG. The von Hippel-Lindau tumor suppressor protein and clear cell renal carcinoma. Clinical Cancer Research. 2007;13(2, part 2):680s–684s.
    1. Linehan WM. Editorial: Kidney cancer—a unique opportunity for the development of disease specific therapy. Journal of Urology. 2002;168(6):2411–2412.
    1. Schmidt L, Junker K, Weirich G, et al. Two North American families with hereditary papillary renal carcinoma and identical novel mutations in the MET proto-oncogene. Cancer Research. 1998;58(8):1719–1722.
    1. Zbar B, Tory K, Merino M, et al. Hereditary papillary renal cell carcinoma. Journal of Urology. 1994;151(3):561–566.
    1. Schmidt L, Duh F-M, Chen F, et al. Germline and somatic mutations in the tyrosine kinase domain of the MET proto-oncogene in papillary renal carcinomas. Nature Genetics. 1997;16(1):68–73.
    1. 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. Journal of Urology. 2004;172(4):1256–1261.
    1. Zbar B, Glenn G, Lubensky I, et al. Hereditary papillary renal cell carcinoma: clinical studies in 10 families. Journal of Urology. 1995;153(3, part 2):907–912.
    1. Choyke PL, Walther MM, Glenn GM, et al. Imaging features of hereditary papillary renal cancers. Journal of Computer Assisted Tomography. 1997;21(5):737–741.
    1. Bottaro DP, Rubin JS, Faletto DL, et al. Identification of the hepatocyte growth factor receptor as the c-met proto-oncogene product. Science. 1991;251(4995):802–804.
    1. Zhang YW, Vande Woude GF. HGF/SF-Met signaling in the control of branching morphogenesis and invasion. Journal of Cellular Biochemistry. 2003;88(2):408–417.
    1. Schmidt L, Junker K, Nakaigawa N, et al. Novel mutations of the MET proto-oncogene in papillary renal carcinomas. Oncogene. 1999;18(14):2343–2350.
    1. Choi JS, Kim MK, Seo JW, et al. MET expression in sporadic renal cell carcinomas. Journal of Korean Medical Science. 2006;21(4):672–677.
    1. Dharmawardana PG, Giubellino A, Bottaro DP. Hereditary papillary renal carcinoma type I. Current Molecular Medicine. 2004;4(8):855–868.
    1. Zhuang Z, Park WS, Pack S, et al. Trisomy 7-harbouring non-random duplication of the mutant MET allele in hereditary papillary renal carcinomas. Nature Genetics. 1998;20(1):66–69.
    1. Srinivasan R, Linehan WM, Vaishampayan U, et al. A phase ii study of two dosing regimens of gsk 1363089 (gsk089), a dual met/vegfr2 inhibitor, in patients (pts) with papillary renal carcinoma (prc) Journal of Clinical Oncology. 2009;27(15S, abstract 5103)
    1. Pfaffenroth EC, Linehan WM. Genetic basis for kidney cancer: opportunity for disease-specific approaches to therapy. Expert Opinion on Biological Therapy. 2008;8(6):779–790.
    1. Launonen V, Vierimaa O, Kiuru M, et al. Inherited susceptibility to uterine leiomyomas and renal cell cancer. Proceedings of the National Academy of Sciences of the United States of America. 2001;98(6):3387–3392.
    1. Merino MJ, Torres-Cabala C, Pinto P, Marston Linehan W. The morphologic spectrum of kidney tumors in hereditary leiomyomatosis and renal cell carcinoma (HLRCC) syndrome. American Journal of Surgical Pathology. 2007;31(10):1578–1585.
    1. Grubb RL, Franks ME, Toro J, et al. Hereditary leiomyomatosis and renal cell cancer: a syndrome associated with an aggressive form of inherited renal cancer. Journal of Urology. 2007;177(6):2074–2080.
    1. Linehan WM, Pinto PA, Srinivasan R, et al. Identification of the genes for kidney cancer: opportunity for disease-specific targeted therapeutics. Clinical Cancer Research. 2007;13(2, part 2):671s–679s.
    1. Tomlinson IPM, Alam NA, Rowan AJ, et al. Germline mutations in FH predispose to dominantly inherited uterine fibroids, skin leiomyomata and papillary renal cell cancer the multiple leiomyoma consortium. Nature Genetics. 2002;30(4):406–410.
    1. Isaacs JS, Yun JJ, Mole DR, et al. HIF overexpression correlates with biallelic loss of fumarate hydratase in renal cancer: novel role of fumarate in regulation of HIF stability. Cancer Cell. 2005;8(2):143–153.
    1. Toro JR, Nickerson ML, Wei MH, et al. Mutations in the fumarate hydratase gene cause hereditary leiomyomatosis and renal cell cancer in families in North America. American Journal of Human Genetics. 2003;73(1):95–106.
    1. Gordon MS, Hussey M, Nagle RB, et al. Phase II study of erlotinib in patients with locally advanced or metastatic papillary histology renal cell cancer: SWOG S0317. Journal of Clinical Oncology. 2009;27(34):5788–5793.
    1. NIH. A phase ii study of bevacizumab and erlotinib in subjects with advanced hereditary leiomyomatosis and renal cell cancer (hlrcc) or sporadic papillary renal cell cancer. 2011, .
    1. Hasumi Y, Baba M, Ajima R, et al. Homozygous loss of BHD causes early embryonic lethality and kidney tumor development with activation of mTORC1 and mTORC2. Proceedings of the National Academy of Sciences of the United States of America. 2009;106(44):18722–18727.
    1. Zbar B, Alvord WG, Glenn G, et al. Risk of renal and colonic neoplasms and spontaneous pneumothorax pneurnothorax in the Birt-Hogg-Dubé syndrome. Cancer Epidemiology Biomarkers and Prevention. 2002;11(4):393–400.
    1. Pavlovich CP, Walther MM, Eyler RA, et al. Renal tumors in the Birt-Hogg-Dubé syndrome. American Journal of Surgical Pathology. 2002;26(12):1542–1552.
    1. Schmidt LS, Warren MB, Nickerson ML, et al. Birt-Hogg-Dubé syndrome, a genodermatosis associated with spontaneous pneumothorax and kidney neoplasia, maps to chromosome 17p11.2. American Journal of Human Genetics. 2001;69(4):876–882.
    1. Nickerson ML, Warren MB, Toro JR, et al. Mutations in a novel gene lead to kidney tumors, lung wall defects, and benign tumors of the hair follicle in patients with the Birt-Hogg-Dubé syndrome. Cancer Cell. 2002;2(2):157–164.
    1. Schmidt LS, Nickerson ML, Warren MB, et al. Germline BHD-mutation spectrum and phenotype analysis of a large cohort of families with Birt-Hogg-Dubé syndrome. American Journal of Human Genetics. 2005;76(6):1023–1033.
    1. Baba M, Hong SB, Sharma N, et al. Folliculin encoded by the BHD gene interacts with a binding protein, FNIP1, and AMPK, and is involved in AMPK and mTOR signaling. Proceedings of the National Academy of Sciences of the United States of America. 2006;103(42):15552–15557.
    1. Vocke CD, Yang Y, Pavlovich CP, et al. High frequency of somatic frameshift BHD gene mutations in Birt-Hogg-Dubé-associated renal tumors. Journal of the National Cancer Institute. 2005;97(12):931–935.
    1. Hasumi H, Baba M, Hong SB, et al. Identification and characterization of a novel folliculin-interacting protein FNIP2. Gene. 2008;415(1-2):60–67.
    1. Baba M, Furihata M, Hong SB, et al. Kidney-targeted Birt-Hogg-Dubé gene inactivation in a mouse model: Erk1/2 and Akt-mTOR activation, cell hyperproliferation, and polycystic kidneys. Journal of the National Cancer Institute. 2008;100(2):140–154.
    1. 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. Journal of Urology. 1999;161(5):1475–1479.
    1. Herring JC, Enquist EG, Chernoff A, Linehan WM, Choyke PL, Walther MM. Parenchymal sparing surgery in patients with hereditary renal cell carcinoma: 10-year experience. Journal of Urology. 2001;165(3):777–781.
    1. Boris R, Proano M, Linehan WM, Pinto PA, Bratslavsky G. Initial experience with robot assisted partial nephrectomy for multiple renal masses. Journal of Urology. 2009;182(4):1280–1286.
    1. Singer EA, Bratslavsky G. Management of locally recurrent kidney cancer. Current Urology Reports. 2010;11(1):15–21.
    1. Walther MM, Choyke PL, Weiss G, et al. Parenchymal sparing surgery in patients with hereditary renal cell carcinoma. Journal of Urology. 1995;153(3, part 2):913–916.
    1. Belldegrun A, Tsui KH, DeKernion JB, Smith RB. Efficacy of nephron-sparing surgery for renal cell carcinoma: analysis based on the new 1997 tumor-node-metastasis staging system. Journal of Clinical Oncology. 1999;17(9):2868–2875.
    1. Hafez KS, Fergany AF, Novick AC. Nephron sparing surgery for localized renal cell carcinoma: impact of tumor size on patient survival, tumor recurrence and TNM staging. Journal of Urology. 1999;162(6):1930–1933.
    1. Kunkle DA, Egleston BL, Uzzo RG. Excise, ablate or observe: the small renal mass dilemma—a meta-analysis and review. Journal of Urology. 2008;179(4):1227–1234.
    1. Liu NW, Khurana K, Sudarshan S, Pinto PA, Linehan WM, Bratslavsky G. Repeat partial nephrectomy on the solitary kidney: surgical, functional and oncological outcomes. Journal of Urology. 2010;183(5):1719–1724.
    1. Johnson A, Sudarshan S, Liu J, Linehan WM, Pinto PA, Bratslavsky G. Feasibility and outcomes of repeat partial nephrectomy. Journal of Urology. 2008;180(1):89–93.
    1. Pasticier G, Timsit MO, Badet L, et al. Nephron-sparing surgery for renal cell carcinoma: detailed analysis of complications over a 15-year period. European Urology. 2006;49(3):485–490.
    1. Ray ER, Turney BW, Singh R, Chandra A, Cranston DW, O’Brien TS. Open partial nephrectomy: outcomes from two UK centres. BJU International. 2006;97(6):1211–1215.
    1. Yang JC, Sherry RM, Steinberg SM, et al. Randomized study of high-dose and low-dose interleukin-2 in patients with metastatic renal cancer. Journal of Clinical Oncology. 2003;21(16):3127–3132.
    1. Alt AL, Boorjian SA, Lohse CM, Costello BA, Leibovich BC, Blute ML. Survival after complete surgical resection of multiple metastases from renal cell carcinoma. Cancer. 2011;117(13):2873–2882.
    1. Crispen PL, Breau RH, Allmer C, et al. Lymph node dissection at the time of radical nephrectomy for high-risk clear cell renal cell carcinoma: indications and recommendations for surgical templates. European Urology. 2011;59(1):18–23.

Source: PubMed

3
Prenumerera