Review
The metabolic basis of kidney cancer

https://doi.org/10.1016/j.semcancer.2012.06.002Get rights and content

Abstract

Kidney cancer is not a single disease; it is made up of a number of different types of cancer that occur in the kidney. Each of these different types of kidney cancer can have a different histology, have a different clinical course, can respond differently to therapy and is caused by a different gene. Kidney cancer is essentially a metabolic disease; each of the known genes for kidney cancer, VHL, MET, FLCN, TSC1, TSC2, TFE3, TFEB, MITF, fumarate hydratase (FH), succinate dehydrogenase B (SDHB), succinate dehydrogenase D (SDHD), and PTEN genes is involved in the cells ability to sense oxygen, iron, nutrients or energy. Understanding the metabolic basis of kidney cancer will hopefully provide the foundation for the development of effective forms of therapy for this disease.

Section snippets

An introduction to kidney cancer

It is now known that kidney cancer is not a single uniform disease; it is in fact a number of different and specific types of cancers that can occur within the kidney. Each of these different types of kidney cancer can be characterized by differing histologies, different clinical courses, differing responses to a number of varied therapies and association with alterations to different tumor suppressor genes or oncogenes. Currently there are at least twelve different genes associated with the

Hereditary kidney cancer

Much of what we know about the genetic basis of kidney cancer was learned from the study of inherited forms of kidney cancer. There are a number of familial forms of kidney cancer, including von Hippel–Lindau (VHL), Hereditary Papillary Renal Carcinoma (HPRC), Birt–Hogg–Dubé (BHD), Hereditary Leiomyomatosis Renal Cell Carcinoma (HLRCC), Succinate Dehydrogenase Renal Cell Carcinoma (SDH-RCC), Tuberous Sclerosis (TS), and Cowden's Disease (Fig. 1) [1], [2]. All these syndromes are associated with

Von Hippel–Lindau (VHL): clear cell kidney cancer

Von Hippel–Lindau (VHL) is a hereditary kidney cancer syndrome in which affected individuals are at risk for the development of tumors in a number of organs, including the kidneys [3]. It represents a well studied form of inherited cancer risk syndrome, which has additionally provided invaluable insight into the study of non-familial, sporadic kidney cancer.

Hereditary papillary renal carcinoma (HPRC): type 1 papillary kidney cancer

Hereditary papillary renal carcinoma (HPRC) is an autosomal dominant hereditary cancer syndrome in which affected individuals are at risk for the development of bilateral, multifocal type 1 papillary kidney cancer (Fig. 3) [33], [34]. It is estimated that patients affected with HPRC are at risk for the development of up to 1100 tumors per kidney throughout their lifetime [35].

Clinical management, like in patients with VHL-associated kidney tumors, involves active surveillance until the largest

Birt–Hogg–Dubé (BHD): chromophobe kidney cancer

Birt–Hogg–Dubé is an autosomal dominant, hereditary cancer syndrome in which affected individuals are at risk for the development of cutaneous fibrofolliculomas [41], pulmonary cysts [42] and kidney cancer [43]. BHD-associated kidney cancers can be multifocal, bilateral and they can metastasize. Patients affected with BHD are at risk for the development of chromophobe, hybrid oncocytic, and clear cell kidney cancer and oncocytoma (Fig. 4) [44]. Like VHL and HPRC, BHD-associated kidney cancers

MiT transcription factor associated kidney cancer

The MiT family of transcription factors includes TFE3, TFEB, MITF, and TFEC, a family of transcription factors that share a highly homologous basic-helix-loop-helix-leucine zipper DNA binding and dimerization domain. These proteins can produce both hetero- and homodimers and bind identical DNA response elements, suggesting that they may have common downstream targets and a degree of functional redundancy. In tumors, these genes are mainly over expressed due to somatic translocations that create

Tuberous sclerosis complex: regulation of the mTOR pathway

The tuberous sclerosis complex (TSC) is an autosomal dominant disorder in which affected individuals are at risk for the development of manifestations involving multiple organs, including cutaneous angiofribroma, pulmonary lymphangiomyomatosis and renal tumors [68]. Although angiomyolipoma is the most common type of renal tumor found in TSC patients, other types of tumors have been identified, including clear cell kidney cancer [69].

Cowden syndrome: PTEN and the regulation of the mTOR pathway

Cowden syndrome is an autosomal dominant disorder that results from germline mutation of the PTEN gene, in which affected individuals are at risk for manifestations in a number of organs, including tumors of the breast, thyroid, endometrium and kidney [73], [74], [75]. The protein product of the PTEN gene, PTEN, is a phosphatase that catalyzes the conversion of PIP3 (phosphatidylinositol 3,4,5 triphosphate) to PIP2 (phosphatidylinositol 4,5 biphosphate). In response to growth factor receptor

Tricarboxylic acid mutation kidney cancers: exemplifying the Warburg effect

In the 1920s Otto Warburg proposed that a basic characteristic of cancer would be that it is characterized by aerobic glycolysis [77], [78]. Subsequently, this has been shown to be true to a greater and lesser extent in nearly all general cancer types, including kidney cancer. There are two types of inherited kidney cancer that are unique examples of the Warburg effect in cancer driven by specific gene mutation; fumarate hydratase-deficient kidney cancer and succinate dehydrogenase deficient

Conclusion – kidney cancer is a metabolic disease

Kidney cancer is fundamentally a metabolic disease. The known genes for kidney cancer, VHL, MET, FLCN, MITF, TFE3, TFEB, TSC1, TSC2, PTEN, FH, SDHB and SDHD are involved in the cell's ability to sense oxygen, iron, nutrients, and, particularly in the TCA cycle enzymes, energy (Fig. 7). Although significant progress has been made targeting, we still have a long way to go. Most patients treated with the approved drugs targeting the VHL gene pathway, such as sunitinib, sorafenib, bevacuzimab,

Acknowledgements

This research was supported by the Intramural Research Program of the NIH, National Cancer Institute, Center for Cancer Research.

The authors thanks Georgia Shaw for the outstanding editorial and graphics support.

References (94)

  • I.A. Lubensky et al.

    Hereditary and sporadic papillary renal carcinomas with c-met mutations share a distinct morphological phenotype

    American Journal of Pathology

    (1999)
  • D.K. Ornstein et al.

    Prevalence of microscopic tumors in normal appearing renal parenchyma of patients with hereditary papillary renal cancer

    Journal of Urology

    (2000)
  • L.S. Schmidt 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)
  • M.L. Nickerson 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)
  • H. Hasumi et al.

    Identification and characterization of a novel folliculin-interacting protein FNIP2

    Gene

    (2008)
  • G.G. Malouf et al.

    Transcription factor E3 and transcription factor EB renal cell carcinomas: clinical features, biological behavior and prognostic factors

    Journal of Urology

    (2011)
  • P. Argani et al.

    A distinctive pediatric renal neoplasm characterized by epithelioid morphology, basement membrane production, focal HMB45 immunoreactivity, and t(6;11)(p21.1;q12) chromosome translocation

    American Journal of Pathology

    (2001)
  • G.G. Malouf et al.

    Targeted agents in metastatic Xp11 translocation/TFE3 gene fusion renal cell carcinoma (RCC): a report from the Juvenile RCC Network

    Annals of Oncology

    (2010)
  • J.B. Brugarolas et al.

    TSC2 regulates VEGF through mTOR-dependent and -independent pathways

    Cancer Cell

    (2003)
  • J. Brugarolas et al.

    Dysregulation of HIF and VEGF is a unifying feature of the familial hamartoma syndromes

    Cancer Cell

    (2004)
  • S. Gustafson et al.

    Cowden syndrome

    Seminars in Oncology

    (2007)
  • B. Zbar et al.

    Hereditary papillary renal cell carcinoma: clinical studies in 10 families

    Journal of Urology

    (1995)
  • J.R. Toro 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)
  • R.L. Grubb et al.

    Hereditary leiomyomatosis and renal cell cancer: a syndrome associated with an aggressive form of inherited renal cancer

    Journal of Urology

    (2007)
  • J.S. Isaacs 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)
  • W.H. Tong et al.

    The glycolytic shift in fumarate-hydratase-deficient kidney cancer lowers AMPK levels, increases metabolic propensities and lowers cellular iron levels

    Cancer Cell

    (2011)
  • Y. Yang et al.

    UOK 262 cell line, fumarate hydratase deficient (FH-/FH-) hereditary leiomyomatosis renal cell carcinoma: in vitro and in vivo model of an aberrant energy metabolic pathway in human cancer

    Cancer Genetics and Cytogenetics

    (2010)
  • S. Vanharanta et al.

    Early-onset renal cell carcinoma as a novel extraparaganglial component of SDHB-associated heritable paraganglioma

    American Journal of Human Genetics

    (2004)
  • W.M. Linehan et al.

    The genetic basis of kidney cancer: a metabolic disease

    Nature Reviews Urology

    (2010)
  • W.T. Wong et al.

    Genotype-phenotype correlation in von Hippel–Lindau disease with retinal angiomatosis

    Archives of Ophthalmology

    (2007)
  • M.R. Filling-Katz et al.

    Central nervous system involvement in von Hippel–Lindau disease

    Neurology

    (1991)
  • D. Choo et al.

    Endolymphatic sac tumors in von Hippel–Lindau disease

    Journal of Neurosurgery

    (2004)
  • J.A. Blansfield et al.

    Clinical, genetic and radiographic analysis of 108 patients with von Hippel–Lindau disease (VHL) manifested by pancreatic neuroendocrine tumors (PNETs)

    Surgery

    (2007)
  • M.M. Walther et al.

    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)
  • F. Latif et al.

    Identification of the von Hippel–Lindau disease tumor suppressor gene

    Science

    (1993)
  • C. Stolle et al.

    Improved detection of germline mutations in the von Hippel–Lindau disease tumor suppressor gene

    Human Mutation

    (1998)
  • M.L. Nickerson et al.

    Improved identification of von Hippel–Lindau gene alterations in clear cell renal tumors

    Clinical Cancer Research

    (2008)
  • L.E. Moore et al.

    Von Hippel–Lindau (VHL) inactivation in sporadic clear cell renal cancer: Associations with germline VHL polymorphisms and etiologic risk factors

    PLoS Genetics

    (2011)
  • D.R. Duan et al.

    Inhibition of transcription elongation by the VHL tumor suppressor protein

    Science

    (1995)
  • A. Pause 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)
  • A. Kibel et al.

    Binding of the von Hippel–Lindau tumor suppressor protein to elongin B and C

    Science

    (1995)
  • T. Kamura et al.

    Rbx1, a component of the VHL tumor suppressor complex and SCF ubiquitin ligase

    Science

    (1999)
  • P.H. Maxwell et al.

    The tumour suppressor protein VHL targets hypoxia-inducible factors for oxygen-dependent proteolysis

    Nature

    (1999)
  • M. Ohh et al.

    Ubiquitination of hypoxia-inducible factor requires direct binding to the beta-domain of the von Hippel–Lindau protein

    Nature Cell Biology

    (2000)
  • P. Jaakkola et al.

    Targeting of HIF-alpha to the von Hippel–Lindau ubiquitylation complex by O2-regulated prolyl hydroxylation

    Science

    (2001)
  • J.C. Yang et al.

    A randomized trial of bevacizumab, an anti-vascular endothelial growth factor antibody, for metastatic renal cancer

    New England Journal of Medicine

    (2003)
  • R.J. Motzer et al.

    Sunitinib versus interferon alfa in metastatic renal-cell carcinoma

    New England Journal of Medicine

    (2007)
  • Cited by (119)

    • Adult genitourinary cancer: Renal and testicular

      2023, Penn Clinical Manual of Urology, Third Edition
    • Hypoxia-inducible factor pathway genes predict survival in metastatic clear cell renal cell carcinoma

      2022, Urologic Oncology: Seminars and Original Investigations
      Citation Excerpt :

      Non-canonical HIF regulation involves oxygen-independent mechanisms. These mechanisms ultimately increase HRGs by suppressing PHD [4] or altering HIF stability [5] Examples of these mechanisms include the Warburg effect and deficiency in enzymes like succinate dehydrogenase and fumarate hydratase that result in accumulation of metabolites that inhibit PHD. RACK1 (receptor of activated protein C kinase) and HSP90 (heat shock protein 90) are other examples of proteins that bind and modulate HIF1α stability [6].

    View all citing articles on Scopus
    View full text