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.

  • Original Article
  • Published:

Targeting glutamine metabolism in multiple myeloma enhances BIM binding to BCL-2 eliciting synthetic lethality to venetoclax

Abstract

Multiple myeloma (MM) is a plasma cell malignancy that is largely incurable due to development of resistance to therapy-elicited cell death. Nutrients are intricately connected to maintenance of cellular viability in part by inhibition of apoptosis. We were interested to determine if examination of metabolic regulation of BCL-2 proteins may provide insight on alternative routes to engage apoptosis. MM cells are reliant on glucose and glutamine and withdrawal of either nutrient is associated with varying levels of apoptosis. We and others have demonstrated that glucose maintains levels of key resistance-promoting BCL-2 family member, myeloid cell leukemic factor 1 (MCL-1). Cells continuing to survive in the absence of glucose or glutamine were found to maintain expression of MCL-1 but importantly induce pro-apoptotic BIM expression. One potential mechanism for continued survival despite induction of BIM could be due to binding and sequestration of BIM to alternate pro-survival BCL-2 members. Our investigation revealed that cells surviving glutamine withdrawal in particular, enhance expression and binding of BIM to BCL-2, consequently sensitizing these cells to the BH3 mimetic venetoclax. Glutamine deprivation-driven sensitization to venetoclax can be reversed by metabolic supplementation with TCA cycle intermediate α-ketoglutarate. Inhibition of glucose metabolism with the GLUT4 inhibitor ritonavir elicits variable cytotoxicity in MM that is marginally enhanced with venetoclax treatment, however, targeting glutamine metabolism with 6-diazo-5-oxo-l-norleucine uniformly sensitized MM cell lines and relapse/refractory patient samples to venetoclax. Our studies reveal a potent therapeutic strategy of metabolically driven synthetic lethality involving targeting glutamine metabolism for sensitization to venetoclax in MM.

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

Similar content being viewed by others

References

  1. Palumbo A, Anderson K . Multiple myeloma. N Engl J Med 2011; 364: 1046–1060.

    Article  CAS  Google Scholar 

  2. Barlogie B, Mitchell A, van Rhee F, Epstein J, Morgan GJ, Crowley J . Curing myeloma at last: defining criteria and providing the evidence. Blood 2014; 124: 3043–3051.

    Article  CAS  Google Scholar 

  3. Danial NN, Korsmeyer SJ . Cell death: critical control points. Cell 2004; 116: 205–219.

    Article  CAS  Google Scholar 

  4. Cheng EH, Wei MC, Weiler S, Flavell RA, Mak TW, Lindsten T et al. BCL-2, BCL-X(L) sequester BH3 domain-only molecules preventing BAX- and BAK-mediated mitochondrial apoptosis. Mol Cell 2001; 8: 705–711.

    Article  CAS  Google Scholar 

  5. Ni Chonghaile T, Letai A . Mimicking the BH3 domain to kill cancer cells. Oncogene 2008; 27 (Suppl 1): S149–S157.

    Article  Google Scholar 

  6. Podar K, Gouill SL, Zhang J, Opferman JT, Zorn E, Tai YT et al. A pivotal role for Mcl-1 in Bortezomib-induced apoptosis. Oncogene 2008; 27: 721–731.

    Article  CAS  Google Scholar 

  7. Mills JR, Hippo Y, Robert F, Chen SM, Malina A, Lin CJ et al. mTORC1 promotes survival through translational control of Mcl-1. Proc Natl Acad Sci USA 2008; 105: 10853–10858.

    Article  CAS  Google Scholar 

  8. Eguchi T, Itadani H, Shimomura T, Kawanishi N, Hirai H, Kotani H . Expression levels of p18INK4C modify the cellular efficacy of cyclin-dependent kinase inhibitors via regulation of Mcl-1 expression in tumor cell lines. Mol Cancer Ther 2009; 8: 1460–1472.

    Article  CAS  Google Scholar 

  9. Nguyen M, Marcellus RC, Roulston A, Watson M, Serfass L, Murthy Madiraju SR et al. Small molecule obatoclax (GX15-070) antagonizes MCL-1 and overcomes MCL-1-mediated resistance to apoptosis. Proc Natl Acad Sci USA 2007; 104: 19512–19517.

    Article  CAS  Google Scholar 

  10. Pradelli LA, Beneteau M, Chauvin C, Jacquin MA, Marchetti S, Munoz-Pinedo C et al. Glycolysis inhibition sensitizes tumor cells to death receptors-induced apoptosis by AMP kinase activation leading to Mcl-1 block in translation. Oncogene 2009; 29: 1641–1652.

    Article  Google Scholar 

  11. Liu Z, Xu J, He J, Zheng Y, Li H, Lu Y et al. A critical role of autocrine sonic hedgehog signaling in human CD138+ myeloma cell survival and drug resistance. Blood 2014; 124: 2061–2071.

    Article  CAS  Google Scholar 

  12. Pepper C, Hoy T, Bentley DP . Bcl-2/Bax ratios in chronic lymphocytic leukaemia and their correlation with in vitro apoptosis and clinical resistance. Br J Cancer 1997; 76: 935–938.

    Article  CAS  Google Scholar 

  13. Ni Chonghaile T, Sarosiek KA, Vo TT, Ryan JA, Tammareddi A, Moore Vdel G et al. Pretreatment mitochondrial priming correlates with clinical response to cytotoxic chemotherapy. Science 2011; 334: 1129–1133.

    Article  Google Scholar 

  14. Bartel TB, Haessler J, Brown TL, Shaughnessy JD Jr., van Rhee F, Anaissie E et al. F18-fluorodeoxyglucose positron emission tomography in the context of other imaging techniques and prognostic factors in multiple myeloma. Blood 2009; 114: 2068–2076.

    Article  CAS  Google Scholar 

  15. Roberts RS, Hsu HW, Lin KD, Yang TJ . Amino acid metabolism of myeloma cells in culture. J Cell Sci 1976; 21: 609–615.

    CAS  PubMed  Google Scholar 

  16. Dalva-Aydemir S, Bajpai R, Martinez M, Adekola KU, Kandela I, Wei C et al. Targeting the metabolic plasticity of multiple myeloma with FDA-approved ritonavir and metformin. Clin Cancer Res 2015; 21: 1161–1171.

    Article  CAS  Google Scholar 

  17. Graham NA, Tahmasian M, Kohli B, Komisopoulou E, Zhu M, Vivanco I et al. Glucose deprivation activates a metabolic and signaling amplification loop leading to cell death. Mol Syst Biol 2012; 8: 589.

    Article  Google Scholar 

  18. Coloff JL, Mason EF, Altman BJ, Gerriets VA, Liu T, Nichols AN et al. Akt requires glucose metabolism to suppress puma expression and prevent apoptosis of leukemic T cells. J Biol Chem 2011; 286: 5921–5933.

    Article  CAS  Google Scholar 

  19. Zhao Y, Altman BJ, Coloff JL, Herman CE, Jacobs SR, Wieman HL et al. Glycogen synthase kinase 3alpha and 3beta mediate a glucose-sensitive antiapoptotic signaling pathway to stabilize Mcl-1. Mol Cell Biol 2007; 27: 4328–4339.

    Article  CAS  Google Scholar 

  20. Danial NN, Gramm CF, Scorrano L, Zhang CY, Krauss S, Ranger AM et al. BAD and glucokinase reside in a mitochondrial complex that integrates glycolysis and apoptosis. Nature 2003; 424: 952–956.

    Article  CAS  Google Scholar 

  21. Rathmell JC, Fox CJ, Plas DR, Hammerman PS, Cinalli RM, Thompson CB . Akt-directed glucose metabolism can prevent Bax conformation change and promote growth factor-independent survival. Mol Cell Biol 2003; 23: 7315–7328.

    Article  CAS  Google Scholar 

  22. Zhang B, Gojo I, Fenton RG . Myeloid cell factor-1 is a critical survival factor for multiple myeloma. Blood 2002; 99: 1885–1893.

    Article  CAS  Google Scholar 

  23. Derenne S, Monia B, Dean NM, Taylor JK, Rapp MJ, Harousseau JL et al. Antisense strategy shows that Mcl-1 rather than Bcl-2 or Bcl-x(L) is an essential survival protein of human myeloma cells. Blood 2002; 100: 194–199.

    Article  CAS  Google Scholar 

  24. Morales AA, Kurtoglu M, Matulis SM, Liu J, Siefker D, Gutman DM et al. Distribution of Bim determines Mcl-1 dependence or codependence with Bcl-xL/Bcl-2 in Mcl-1-expressing myeloma cells. Blood 2011; 118: 1329–1339.

    Article  CAS  Google Scholar 

  25. McBrayer SK, Cheng JC, Singhal S, Krett NL, Rosen ST, Shanmugam M . Multiple myeloma exhibits novel dependence on GLUT4, GLUT8, and GLUT11: implications for glucose transporter-directed therapy. Blood 2012; 119: 4686–4697.

    Article  CAS  Google Scholar 

  26. Souers AJ, Leverson JD, Boghaert ER, Ackler SL, Catron ND, Chen J et al. ABT-199, a potent and selective BCL-2 inhibitor, achieves antitumor activity while sparing platelets. Nat Med 2013; 19: 202–208.

    Article  CAS  Google Scholar 

  27. Wensveen FM, van Gisbergen KP, Derks IA, Gerlach C, Schumacher TN, van Lier RA et al. Apoptosis threshold set by Noxa and Mcl-1 after T cell activation regulates competitive selection of high-affinity clones. Immunity 2010; 32: 754–765.

    Article  CAS  Google Scholar 

  28. Nakajima W, Hicks MA, Tanaka N, Krystal GW, Harada H . Noxa determines localization and stability of MCL-1 and consequently ABT-737 sensitivity in small cell lung cancer. Cell Death Dis 2014; 5: e1052.

    Article  CAS  Google Scholar 

  29. Vyas AK, Koster JC, Tzekov A, Hruz PW . Effects of the HIV protease inhibitor ritonavir on GLUT4 knock-out mice. J Biol Chem 2010; 285: 36395–36400.

    Article  CAS  Google Scholar 

  30. Shapiro RA, Clark VM, Curthoys NP . Inactivation of rat renal phosphate-dependent glutaminase with 6-diazo-5-oxo-L-norleucine. Evidence for interaction at the glutamine binding site. J Biol Chem 1979; 254: 2835–2838.

    CAS  PubMed  Google Scholar 

  31. Brown G, Singer A, Proudfoot M, Skarina T, Kim Y, Chang C et al. Functional and structural characterization of four glutaminases from Escherichia coli and Bacillus subtilis. Biochemistry 2008; 47: 5724–5735.

    Article  CAS  Google Scholar 

  32. Letai A, Bassik MC, Walensky LD, Sorcinelli MD, Weiler S, Korsmeyer SJ . Distinct BH3 domains either sensitize or activate mitochondrial apoptosis, serving as prototype cancer therapeutics. Cancer Cell 2002; 2: 183–192.

    Article  CAS  Google Scholar 

  33. Lessene G, Czabotar PE, Colman PM . BCL-2 family antagonists for cancer therapy. Nat Rev Drug Discov 2008; 7: 989–1000.

    Article  CAS  Google Scholar 

  34. van Delft MF, Wei AH, Mason KD, Vandenberg CJ, Chen L, Czabotar PE et al. The BH3 mimetic ABT-737 targets selective Bcl-2 proteins and efficiently induces apoptosis via Bak/Bax if Mcl-1 is neutralized. Cancer Cell 2006; 10: 389–399.

    Article  CAS  Google Scholar 

  35. Wilson WH, O'Connor OA, Czuczman MS, LaCasce AS, Gerecitano JF, Leonard JP et al. Navitoclax, a targeted high-affinity inhibitor of BCL-2, in lymphoid malignancies: a phase 1 dose-escalation study of safety, pharmacokinetics, pharmacodynamics, and antitumour activity. Lancet Oncol 2010; 11: 1149–1159.

    Article  CAS  Google Scholar 

  36. Schoenwaelder SM, Jarman KE, Gardiner EE, Hua M, Qiao J, White MJ et al. Bcl-xL-inhibitory BH3 mimetics can induce a transient thrombocytopathy that undermines the hemostatic function of platelets. Blood 2011; 118: 1663–1674.

    Article  CAS  Google Scholar 

  37. Touzeau C, Dousset C, Le Gouill S, Sampath D, Leverson JD, Souers AJ et al. The Bcl-2 specific BH3 mimetic ABT-199: a promising targeted therapy for t(11;14) multiple myeloma. Leukemia 2014; 28: 210–212.

    Article  CAS  Google Scholar 

  38. Olsen RR, Mary-Sinclair MN, Yin Z, Freeman KW . Antagonizing Bcl-2 family members sensitizes neuroblastoma and Ewing's sarcoma to an inhibitor of glutamine metabolism. PLoS One 2015; 10: e0116998.

    Article  Google Scholar 

  39. Leverson JD, Zhang H, Chen J, Tahir SK, Phillips DC, Xue J et al. Potent and selective small-molecule MCL-1 inhibitors demonstrate on-target cancer cell killing activity as single agents and in combination with ABT-263 (navitoclax). Cell Death Dis 2015; 6: e1590.

    Article  CAS  Google Scholar 

  40. Thomas RL, Roberts DJ, Kubli DA, Lee Y, Quinsay MN, Owens JB et al. Loss of MCL-1 leads to impaired autophagy and rapid development of heart failure. Genes Dev 2013; 27: 1365–1377.

    Article  CAS  Google Scholar 

  41. Tuffy LP, Concannon CG, D'Orsi B, King MA, Woods I, Huber HJ et al. Characterization of Puma-dependent and Puma-independent neuronal cell death pathways following prolonged proteasomal inhibition. Mol Cell Biol 2010; 30: 5484–5501.

    Article  CAS  Google Scholar 

  42. Puthalakath H, Huang DC, O'Reilly LA, King SM, Strasser A . The proapoptotic activity of the Bcl-2 family member Bim is regulated by interaction with the dynein motor complex. Mol Cell 1999; 3: 287–296.

    Article  CAS  Google Scholar 

  43. Lei K, Davis RJ . JNK phosphorylation of Bim-related members of the Bcl2 family induces Bax-dependent apoptosis. Proc Natl Acad Sci USA 2003; 100: 2432–2437.

    CAS  Google Scholar 

  44. Son J, Lyssiotis CA, Ying H, Wang X, Hua S, Ligorio M et al. Glutamine supports pancreatic cancer growth through a KRAS-regulated metabolic pathway. Nature 2013; 496: 101–105.

    Article  CAS  Google Scholar 

  45. Fabrizio Accardi MC, Bolzoni M, Storti P, Todoerti K, Agnelli L, Ferrari M et al. Ammonium production and glutamine-addiction of myeloma cells: new attractive targets in multiple myeloma. Blood 2014; 124: 2067.

    Google Scholar 

  46. Gross MI, Demo SD, Dennison JB, Chen L, Chernov-Rogan T, Goyal B et al. Antitumor activity of the glutaminase inhibitor CB-839 in triple-negative breast cancer. Mol Cancer Ther 2014; 13: 890–901.

    Article  CAS  Google Scholar 

  47. Prideaux SM, Conway O'Brien E, Chevassut TJ . The genetic architecture of multiple myeloma. Adv Hematol 2014; 2014: 864058.

    Article  Google Scholar 

  48. Shanmugam M, McBrayer SK, Qian J, Raikoff K, Avram MJ, Singhal S et al. Targeting glucose consumption and autophagy in myeloma with the novel nucleoside analogue 8-aminoadenosine. J Biol Chem 2009; 284: 26816–26830.

    Article  CAS  Google Scholar 

  49. Vo TT, Ryan J, Carrasco R, Neuberg D, Rossi DJ, Stone RM et al. Relative mitochondrial priming of myeloblasts and normal HSCs determines chemotherapeutic success in AML. Cell 2012; 151: 344–355.

    Article  CAS  Google Scholar 

  50. Boise LH, Minn AJ, Noel PJ, June CH, Accavitti MA, Lindsten T et al. CD28 costimulation can promote T cell survival by enhancing the expression of Bcl-XL. Immunity 1995; 3: 87–98.

    Article  CAS  Google Scholar 

  51. Mishra RK, Wei C, Hresko RC, Bajpai R, Heitmeier M, Matulis SM et al. In silico modeling-based identification of glucose transporter 4 (GLUT4)-selective inhibitors for cancer therapy. J Biol Chem 2015; 290: 14441–14453.

    Article  CAS  Google Scholar 

Download references

Acknowledgements

This research was supported by the American Cancer Society Research scholar grant (RSG-11-254-01-CSM) to MS. LHB is a Georgia Research Alliance distinguished cancer scientist and also received support from the TJ Martell Foundation and P30CA138292.

Author contributions

RB and MS conceived the research with conceptual advice from LHB; RB, SMM, CW and MS performed experimentation and data analysis with advice from LHB. HEVH, AKN and SL oversaw collection of myeloma patient samples and provided conceptual advice. RB and MS wrote the manuscript and MS supervised the project.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to M Shanmugam.

Ethics declarations

Competing interests

LHB is a consultant for Onyx Pharmaceuticals and Novartis; AKN is consultant for Onyx Pharmaceuticals and Spectrum pharmaceuticals; SL is a consultant for Millennium, Onyx Pharmaceuticals, Novartis, BMS, Janssen and Celgene. The remaining authors declare no conflict of interest.

Additional information

Supplementary Information accompanies this paper on the Oncogene website

Supplementary information

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Bajpai, R., Matulis, S., Wei, C. et al. Targeting glutamine metabolism in multiple myeloma enhances BIM binding to BCL-2 eliciting synthetic lethality to venetoclax. Oncogene 35, 3955–3964 (2016). https://doi.org/10.1038/onc.2015.464

Download citation

  • Received:

  • Revised:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/onc.2015.464

This article is cited by

Search

Quick links