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.

  • Leading Article
  • Published:

Synergistic effect of targeting mTOR by rapamycin and depleting ATP by inhibition of glycolysis in lymphoma and leukemia cells

Abstract

The mammalian target of rapamycin (mTOR) pathway plays important roles in regulating nutrient metabolism and promoting the growth and survival of cancer cells, which exhibit increased glycolysis for ATP generation. In this study, we tested the hypothesis that inhibition of the mTOR pathway and glycolysis would synergistically impact the energy metabolism in cancer cells and may serve as an effective therapeutic strategy to kill malignant cells. Using human lymphoma cells and leukemia cells, we demonstrated that the combination of rapamycin, an mTOR inhibitor, with a glycolytic inhibitor produced synergistic cytotoxic effect, as evidenced by apoptosis and cell growth inhibition assays. Mechanistic studies showed that inhibition of the mTOR pathway by rapamycin alone sufficiently suppressed the phosphorylation of the downstream molecules p70S6K and 4E-BP-1, but only caused a moderate cytostatic effect. Combination of mTOR inhibition and blockage of glycolysis synergistically suppressed glucose uptake and severely depleted cellular ATP pools, leading to significant enhancement of cell killing. In contrast, combination of rapamycin and ara-C did not increase cytotoxicity in vitro. Our findings suggest that targeting mTOR pathway in combination with inhibition of glycolysis may be an effective therapeutic strategy for hematological malignancies. This mechanism-based drug combination warrants further investigation in preclinical and clinical settings.

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
Figure 2
Figure 3
Figure 4
Figure 5
Figure 6

References

  1. Warburg O . The Metabolism of Tumors. London: Constable Co. Ltd, 1930.

    Google Scholar 

  2. Warburg O . On the origin of cancer cells. Science 1956; 123: 309–314.

    Article  CAS  Google Scholar 

  3. Zu XL, Guppy M . Cancer metabolism: facts, fantasy, and fiction. Biochem Biophys Res Commun 2004; 313: 459–465.

    Article  CAS  Google Scholar 

  4. Xu RH, Pelicano H, Zhou Y, Carew JS, Feng L, Bhalla KN et al. Inhibition of glycolysis in cancer cells: a novel strategy to overcome drug resistance associated with mitochondrial respiratory defect and hypoxia. Cancer Res 2005; 65: 613–621.

    CAS  PubMed  Google Scholar 

  5. Geschwind JF, Georgiades CS, Ko YH, Pedersen PL . Recently elucidated energy catabolism pathways provide opportunities for novel treatments in hepatocellular carcinoma. Expert Rev Anticancer Ther 2004; 4: 449–457.

    Article  CAS  Google Scholar 

  6. Maher JC, Krishan A, Lampidis T . Greater cell cycle inhibition and cytotoxicity induced by 2-deoxy-D-glucose in tumor cells treated under hypoxic vs aerobic condition. Cancer Chemother Pharmacol 2004; 53: 116–122.

    Article  CAS  Google Scholar 

  7. Maschek G, Savaraj N, Priebe W, Braunschweiger P, Hamilton K, Tidmarsh GF et al. 2-Deoxy-D-glucose increases the efficacy of Adriamycin and paclitaxel in human osteosarcoma and non-small cell lung cancers in vivo. Cancer Res 2004; 64: 31–34.

    Article  CAS  Google Scholar 

  8. Ko YH, Pedersen PL, Geschwind JF . Glucose catabolism in the rabbit VX2 model for liver cancer. Cancer Lett 2001; 173: 83–91.

    Article  CAS  Google Scholar 

  9. Dennis PB, Jaeschke A, Saitoh M, Fowler B, Kozma SC, Thomas G . Mammalian TOR: a homeostatic ATP sensor. Science 2001; 294: 1102–1105.

    Article  CAS  Google Scholar 

  10. Bjornsti MA, Houghton PJ . The TOR pathway: a target for cancer therapy. Nat Rev Cancer 2004; 4: 335–348.

    Article  CAS  Google Scholar 

  11. Hay N, Sonenberg N . Upstream and downstream of mTOR. Genes Dev 2004; 18: 1926–1945.

    Article  CAS  Google Scholar 

  12. Schmelzle T, Hall MN . TOR, a central controller of cell growth. Cell 2000; 103: 253–262.

    Article  CAS  Google Scholar 

  13. Raymond E, Alexandre J, Faivre S, Vera K, Materman E, Boni J et al. Safety and pharmacokinetics of escalated doses of weekly intravenous infusion of CCI-779, a novel mTOR inhibitor, in patients with cancer. J Clin Oncol 2004; 22: 2336–2347.

    Article  CAS  Google Scholar 

  14. Panwalkar A, Verstovsek S, Giles FJ . Mammalian target of rapamycin inhibition as therapy for hematologic malignancies. Cancer 2004; 100: 657–666.

    Article  CAS  Google Scholar 

  15. Dancey JE . Inhibitors of the mammalian target of rapamycin. Expert Opin Investig Drugs 2005; 14: 313–328.

    Article  CAS  Google Scholar 

  16. Ko YH, Smith BL, Wang Y, Pomper MG, Rini DA, Torbenson MS et al. Advanced cancers: eradication in all cases using 3-bromopyruvate therapy to deplete ATP. Biochem Biophys Res Commun 2004; 324: 269–275.

    Article  CAS  Google Scholar 

  17. Seymour JF, Huang P, Plunkett W, Gandhi V . Influence of fludarabine on pharmacokinetics and pharmacodynamics of cytarabine: implications for a continuous infusion schedule. Clin Cancer Res 1996; 2: 653–658.

    CAS  PubMed  Google Scholar 

  18. Chou TC, Hayball MP . CalcuSyn: window software for dose effect analysis. Cambridge, United Kingdom: Biosoft.

  19. Easton JB, Houghton PJ . Therapeutic potential of target of rapamycin inhibitors. Expert Opin Ther Targets 2004; 8: 551–564.

    Article  CAS  Google Scholar 

  20. Huang L, Shu M, Dilling J, Easton F, Harwood H, Ichijo P et al. Sustained activation of the JNK cascade and rapamycin-induced apoptosis are suppressed by p53/p21Cip1. Mol Cell 2003; 11: 1491–1501.

    Article  CAS  Google Scholar 

  21. Edinger AL, Thompson CB . Akt maintains cell size and survival by increasing mTOR-dependent nutrient uptake. Mol Biol Cell 2002; 13: 2276–2288.

    Article  CAS  Google Scholar 

  22. Peterson RT, Desai BN, Hardwick JS, Schreiber SL . Protein phosphatase 2A interacts with the 70-kDa S6 kinase and is activated by inhibition of FKBP12-rapamycinassociated protein. Proc Natl Acad Sci USA 1999; 96: 4438–4442.

    Article  CAS  Google Scholar 

Download references

Acknowledgements

This study was supported in part by Grants CA85563, CA100428, CA109041, and CA100632 from the National Cancer Institute, the National Institutes of Health.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to P Huang.

Rights and permissions

Reprints and permissions

About this article

Cite this article

Xu, RH., Pelicano, H., Zhang, H. et al. Synergistic effect of targeting mTOR by rapamycin and depleting ATP by inhibition of glycolysis in lymphoma and leukemia cells. Leukemia 19, 2153–2158 (2005). https://doi.org/10.1038/sj.leu.2403968

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/sj.leu.2403968

Keywords

This article is cited by

Search

Quick links