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.

  • Case Study
  • Published:

Exploring a glycolytic inhibitor for the treatment of an FH-deficient type-2 papillary RCC

Abstract

Background. A 24-year-old woman presented with a 45 cm complex cystic renal mass, which was resected. The tumor was a type-2 papillary renal cell carcinoma (pRCC-2), and several nodules remained. The patient was treated with mammalian target of rapamycin complex 1 (mTORC1) inhibitors, but after 5 months the tumor had progressed. Genetic testing of the patient revealed a novel heterozygous germline mutation in the gene encoding fumarate hydratase (FH), an enzyme of the tricarboxylic acid (TCA) cycle. As the tumor exhibited loss of heterozygosity for FH and markedly reduced FH activity, and in the absence of other established therapies, treatment with the glycolytic inhibitor 2DG (2-deoxy-D-glucose) was explored.

Investigations. CT, histology, immunohistochemistry, genetic studies, 2-deoxy-2-(18F)fluoro-D-glucose (18FDG)-PET/CT, FH enzymatic assays, reconstitution experiments and in vitro studies of the effects of 2DG on FH-deficient tumor cells.

Diagnosis. pRCC-2 arising in a patient with a novel germline FH mutation and de novo hereditary leiomyomatosis and renal cell cancer (HLRCC) syndrome progressing after mTORC1 inhibitor therapy.

Management. Surgical resection of the renal mass, treatment with mTORC1 inhibitors followed by 2DG. Unfortunately, 2DG was not effective, and the patient died several weeks later.

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

Figure 1: CT and 18FDG-PET/CT imaging throughout treatment.
Figure 2: Representative photomicrographs of tumor sections, showing fibrovascular papillae lined by stratified, large, pleomorphic cells with eosinophilic cytoplasm and large, prominent nucleoli.
Figure 3: FH gene sequencing and enzymatic activity analyses.
Figure 4: Characterization of the mutant FHAsp341Asn protein.
Figure 5: The effect of 2DG on the proliferation of UOK262 cells.
Figure 6: The effect of 2DG treatment on ECAR, OCR and ATP levels in UOK262 cells and two control cell lines: 786-O and the extensively characterized C2C12 myoblast cell line.
Figure 7: Western blot of UOK262 cells treated with 1 mM 2DG for the indicated periods of time in the presence of compound C (an AMPK inhibitor) or DMSO (vehicle).

References

  1. Hudes, G. et al. Temsirolimus, interferon alfa, or both for advanced renal-cell carcinoma. N. Engl. J. Med. 356, 2271–2281 (2007).

    Article  CAS  PubMed  Google Scholar 

  2. Motzer, R. J. et al. Efficacy of everolimus in advanced renal cell carcinoma: a double-blind, randomised, placebo-controlled phase III trial. Lancet 372, 449–456 (2008).

    Article  CAS  PubMed  Google Scholar 

  3. Tomlinson, I. P. et al. Germline mutations in FH predispose to dominantly inherited uterine fibroids, skin leiomyomata and papillary renal cell cancer. Nat. Genet. 30, 406–410 (2002).

    Article  CAS  PubMed  Google Scholar 

  4. Toro, J. R. et al. Mutations in the fumarate hydratase gene cause hereditary leiomyomatosis and renal cell cancer in families in North America. Am. J. Hum. Genet. 73, 95–106 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Gottlieb, E. & Tomlinson, I. P. Mitochondrial tumour suppressors: a genetic and biochemical update. Nat. Rev. Cancer 5, 857–866 (2005).

    Article  CAS  PubMed  Google Scholar 

  6. Lehtonen, H. J. et al. Increased risk of cancer in patients with fumarate hydratase germline mutation. J. Med. Genet. 43, 523–526 (2006).

    Article  CAS  PubMed  Google Scholar 

  7. Grubb, R. L. 3rd et al. Hereditary leiomyomatosis and renal cell cancer: a syndrome associated with an aggressive form of inherited renal cancer. J. Urol. 177, 2074–2079 (2007).

    Article  CAS  PubMed  Google Scholar 

  8. Bayley, J. P., Launonen, V. & Tomlinson, I. P. The FH mutation database: an online database of fumarate hydratase mutations involved in the MCUL (HLRCC) tumor syndrome and congenital fumarase deficiency. BMC Med. Genet. 9, 20 (2008).

    Article  PubMed  PubMed Central  Google Scholar 

  9. Weaver, T. M., Levitt, D. G., Donnelly, M. I., Stevens, P. P. & Banaszak, L. J. The multisubunit active site of fumarase C from Escherichia coli. Nat. Struct. Biol. 2, 654–662 (1995).

    Article  CAS  PubMed  Google Scholar 

  10. Isaacs, J. S. 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 8, 143–153 (2005).

    Article  CAS  PubMed  Google Scholar 

  11. Yang, Y. 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 Genet. Cytogenet. 196, 45–55 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. O'Flaherty, L. et al. Dysregulation of hypoxia pathways in fumarate hydratase-deficient cells is independent of defective mitochondrial metabolism. Hum. Mol. Genet. 19, 3844–3851 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Kiuru, M. et al. Few FH mutations in sporadic counterparts of tumor types observed in hereditary leiomyomatosis and renal cell cancer families. Cancer Res. 62, 4554–4557 (2002).

    CAS  PubMed  Google Scholar 

  14. Morris, M. R. et al. Molecular genetic analysis of FIH-1, FH, and SDHB candidate tumour suppressor genes in renal cell carcinoma. J. Clin. Pathol. 57, 706–711 (2004).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Sudarshan, S., Pinto, P. A., Neckers, L. & Linehan, W. M. Mechanisms of disease: hereditary leiomyomatosis and renal cell cancer—a distinct form of hereditary kidney cancer. Nat. Clin. Pract. Urol. 4, 104–110 (2007).

    Article  CAS  PubMed  Google Scholar 

  16. Choueiri, T. K. et al. Efficacy of sunitinib and sorafenib in metastatic papillary and chromophobe renal cell carcinoma. J. Clin. Oncol. 26, 127–131 (2008).

    Article  CAS  PubMed  Google Scholar 

  17. Plimack, E. R. et al. Sunitinib in papillary renal cell carcinoma (pRCC): results from a single-arm phase II study. J. Clin. Oncol. 28, 4604 (2010).

    Article  Google Scholar 

  18. Pollard, P. J. et al. Accumulation of Krebs cycle intermediates and over-expression of HIF1α in tumours which result from germline FH and SDH mutations. Hum. Mol. Genet. 14, 2231–2239 (2005).

    Article  CAS  PubMed  Google Scholar 

  19. Sudarshan, S. et al. Fumarate hydratase deficiency in renal cancer induces glycolytic addiction and hypoxia-inducible transcription factor 1α stabilization by glucose-dependent generation of reactive oxygen species. Mol. Cell Biol. 29, 4080–4090 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Pelicano, H., Martin, D. S., Xu, R. H. & Huang, P. Glycolysis inhibition for anticancer treatment. Oncogene 25, 4633–4646 (2006).

    Article  CAS  PubMed  Google Scholar 

  21. Stein, M. et al. Targeting tumor metabolism with 2-deoxyglucose in patients with castrate-resistant prostate cancer and advanced malignancies. Prostate 70, 1388–1394 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Raez, L. E. et al. Responses to the combination of the glycolytic inhibitor 2-deoxy-glucose (2DG) and docetaxel (DC) in patients with lung and head and neck (H/N) carcinomas. J. Clin. Oncol. 25, 14025 (2007).

    Google Scholar 

  23. Alam, N. A. et al. Genetic and functional analyses of FH mutations in multiple cutaneous and uterine leiomyomatosis, hereditary leiomyomatosis and renal cancer, and fumarate hydratase deficiency. Hum. Mol. Genet. 12, 1241–1252 (2003).

    Article  CAS  PubMed  Google Scholar 

  24. Fong, P. C. et al. Inhibition of poly(ADP-ribose) polymerase in tumors from BRCA mutation carriers. N. Engl. J. Med. 361, 123–134 (2009).

    Article  CAS  PubMed  Google Scholar 

  25. Farmer, H. et al. Targeting the DNA repair defect in BRCA mutant cells as a therapeutic strategy. Nature 434, 917–921 (2005).

    Article  CAS  PubMed  Google Scholar 

  26. Linehan, W. M. et al. Hereditary kidney cancer: unique opportunity for disease-based therapy. Cancer 115, 2252–2261 (2009).

    Article  CAS  PubMed  Google Scholar 

  27. Vander Heiden, M. G., Cantley, L. C. & Thompson, C. B. Understanding the Warburg effect: the metabolic requirements of cell proliferation. Science 324, 1029–1033 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Mankoff, D. A. et al. Tumor-specific positron emission tomography imaging in patients: [18F] fluorodeoxyglucose and beyond. Clin. Cancer Res. 13, 3460–3469 (2007).

    Article  CAS  PubMed  Google Scholar 

  29. Inoki, K., Zhu, T. & Guan, K. L. TSC2 mediates cellular energy response to control cell growth and survival. Cell 115, 577–590 (2003).

    Article  CAS  PubMed  Google Scholar 

  30. Kimura, N. et al. A possible linkage between AMP-activated protein kinase (AMPK) and mammalian target of rapamycin (mTOR) signalling pathway. Genes Cells 8, 65–79 (2003).

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

This manuscript is dedicated to the memory of the patient and to her family whose request it was that the findings be reported so that others may potentially benefit. pBabe–FH–flag was a gift from Dr. Sunil Sudarshan (University of Texas Health Science Center, San Antonio, TX) and UOK262 cells were a gift from Dr. Youfeng Yang and Dr. W. Marston Linehan (National Cancer Institute, Bethesda, MD). We are grateful to Dr. Samuel Peña-Llopis, Sharanya Sivanand and Jessica Gillen for tissue collection and processing, Dr. Virginia Langmuir and Stew Kroll at Threshold Pharmaceuticals for providing the drug and for advice, Capt. Frank H. Cross Jr at the FDA, the Simmons Cancer Center pharmacy and nursing staff, and the Brugarolas lab. T. A. T. Tran was supported by a T32CA124334 grant, and R. J. DeBerardinis was supported by NIH (DK072565) and CPRIT (HIRP100437) grants. This research was supported by the following grants to J. Brugarolas: Clinical Scientist Development Award from the Doris Duke Charitable Foundation (2007062), K08NS051843, RO1CA129387 and CPRIT (RP101075). J. Brugarolas is a Virginia Murchison Linthicum Scholar in Medical Research at UT Southwestern.

Author information

Authors and Affiliations

Authors

Contributions

T. Yamasaki designed and carried out most experiments. T. A. T. Tran was responsible for the site-directed mutagenesis and the native gel electrophoresis. O. K. Oz evaluated the PET scans, X. Zhang performed the crystallographic analyses, and R. J. DeBerardinis was involved in discussions. G. V. Raj and R. E. Schwarz contributed to the care of the patient and were responsible for the initial and subsequent surgeries, respectively. J. Brugarolas was the patient's primary oncologist, directed the project, and wrote the manuscript. T. Yamasaki and all the authors reviewed and made suggestions to the manuscript.

Corresponding author

Correspondence to James Brugarolas.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Rights and permissions

Reprints and permissions

About this article

Cite this article

Yamasaki, T., Tran, T., Oz, O. et al. Exploring a glycolytic inhibitor for the treatment of an FH-deficient type-2 papillary RCC. Nat Rev Urol 8, 165–171 (2011). https://doi.org/10.1038/nrurol.2010.234

Download citation

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nrurol.2010.234

This article is cited by

Search

Quick links

Nature Briefing: Cancer

Sign up for the Nature Briefing: Cancer newsletter — what matters in cancer research, free to your inbox weekly.

Get what matters in cancer research, free to your inbox weekly. Sign up for Nature Briefing: Cancer