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.

  • Article
  • Published:

ACUTE LYMPHOBLASTIC LEUKEMIA

Effective targeting of NAMPT in patient-derived xenograft models of high-risk pediatric acute lymphoblastic leukemia

Abstract

The prognosis for children diagnosed with high-risk acute lymphoblastic leukemia (ALL) remains suboptimal, and more potent and less toxic treatments are urgently needed. We investigated the efficacy of a novel nicotinamide phosphoribosyltransferase inhibitor, OT-82, against a panel of patient-derived xenografts (PDXs) established from high-risk and poor outcome pediatric ALL cases. OT-82 was well-tolerated and demonstrated impressive single agent in vivo efficacy, achieving significant leukemia growth delay in 95% (20/21) and disease regression in 86% (18/21) of PDXs. In addition, OT-82 enhanced the efficacy of the established drugs cytarabine and dasatinib and, as a single agent, showed similar efficacy as an induction-type regimen combining three drugs used to treat pediatric ALL. OT-82 exerted its antileukemic action by depleting NAD+ and ATP, inhibiting the NAD+-requiring DNA damage repair enzyme PARP-1, increasing mitochondrial ROS levels and inducing DNA damage, culminating in apoptosis induction. OT-82 sensitivity was associated with the occurrence of mutations in major DNA damage response genes, while OT-82 resistance was characterized by high expression levels of CD38. In conclusion, our study provides evidence that OT-82, as a single agent, and in combination with established drugs, is a promising new therapeutic strategy for a broad spectrum of high-risk pediatric ALL for which improved therapies are urgently needed.

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

Fig. 1: OT-82 potently decreases the viability of leukemia cell lines and pediatric ALL PDX cells.
Fig. 2: OT-82 decreases intracellular NAD+ levels culminating in apoptosis induction in leukemia cells.
Fig. 3: OT-82 induces regressions in vivo in a broad panel of pediatric ALL PDX models.
Fig. 4: OT-82 reduces NAD+, ATP and serum visfatin levels and inhibits PARP-1 in ALL PDXs in vivo.
Fig. 5: OT-82 potentiates currently used therapies for pediatric high-risk ALL.
Fig. 6: Determinants of response to OT-82.

Similar content being viewed by others

References

  1. Hunger SP, Mullighan CG. Redefining ALL classification: toward detecting high-risk ALL and implementing precision medicine. Blood. 2015;125:3977–87.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Pui CH, Carroll WL, Meshinchi S, Arceci RJ. Biology, risk stratification, and therapy of pediatric acute leukemias: an update. J Clin Oncol. 2011;29:551–65.

    Article  PubMed  Google Scholar 

  3. Hunger SP, Lu X, Devidas M, Camitta BM, Gaynon PS, Winick NJ, et al. Improved survival for children and adolescents with acute lymphoblastic leukemia between 1990 and 2005: a report from the children's oncology group. J Clin Oncol. 2012;30:1663–9.

    Article  PubMed  PubMed Central  Google Scholar 

  4. Arico M, Schrappe M, Hunger SP, Carroll WL, Conter V, Galimberti S, et al. Clinical outcome of children with newly diagnosed Philadelphia chromosome-positive acute lymphoblastic leukemia treated between 1995 and 2005. J Clin Oncol. 2010;28:4755–61.

    Article  PubMed  PubMed Central  Google Scholar 

  5. Pui CH, Chessells JM, Camitta B, Baruchel A, Biondi A, Boyett JM, et al. Clinical heterogeneity in childhood acute lymphoblastic leukemia with 11q23 rearrangements. Leukemia. 2003;17:700–6.

    Article  CAS  PubMed  Google Scholar 

  6. Ko RH, Ji L, Barnette P, Bostrom B, Hutchinson R, Raetz E, et al. Outcome of patients treated for relapsed or refractory acute lymphoblastic leukemia: a Therapeutic Advances in Childhood Leukemia Consortium study. J Clin Oncol. 2010;28:648–54.

    Article  PubMed  Google Scholar 

  7. Smith MA, Seibel NL, Altekruse SF, Ries LA, Melbert DL, O'Leary M, et al. Outcomes for children and adolescents with cancer: challenges for the twenty-first century. J Clin Oncol. 2010;28:2625–34.

    Article  PubMed  PubMed Central  Google Scholar 

  8. Smith M, Arthur D, Camitta B, Carroll AJ, Crist W, Gaynon P, et al. Uniform approach to risk classification and treatment assignment for children with acute lymphoblastic leukemia. J Clin Oncol. 1996;14:18–24.

    Article  CAS  PubMed  Google Scholar 

  9. Linabery AM, Ross JA. Trends in childhood cancer incidence in the U.S. (1992–2004). Cancer. 2008;112:416–32.

    Article  PubMed  Google Scholar 

  10. Ness KK, Armenian SH, Kadan-Lottick N, Gurney JG. Adverse effects of treatment in childhood acute lymphoblastic leukemia: general overview and implications for long-term cardiac health. Expert Rev Hematol. 2011;4:185–97.

    Article  PubMed  PubMed Central  Google Scholar 

  11. Vander Heiden MG. Targeting cancer metabolism: a therapeutic window opens. Nat Rev Drug Discov. 2011;10:671–84.

    Article  CAS  Google Scholar 

  12. Garten A, Schuster S, Penke M, Gorski T, de Giorgis T, Kiess W. Physiological and pathophysiological roles of NAMPT and NAD metabolism. Nat Rev Endocrinol. 2015;11:535–46.

    Article  CAS  PubMed  Google Scholar 

  13. Sampath D, Zabka TS, Misner DL, O'Brien T, Dragovich PS. Inhibition of nicotinamide phosphoribosyltransferase (NAMPT) as a therapeutic strategy in cancer. Pharm Ther. 2015;151:16–31.

    Article  CAS  Google Scholar 

  14. Zhao G, Green CF, Hui YH, Prieto L, Shepard R, Dong S, et al. Discovery of a highly selective NAMPT inhibitor that demonstrates robust efficacy and improved retinal toxicity with nicotinic acid coadministration. Mol Cancer Ther. 2017;16:2677–88.

    Article  CAS  PubMed  Google Scholar 

  15. Takao S, Chien W, Madan V, Lin DC, Ding LW, Sun QY, et al. Targeting the vulnerability to NAD(+) depletion in B-cell acute lymphoblastic leukemia. Leukemia. 2018;32:616–25.

    Article  CAS  PubMed  Google Scholar 

  16. Matheny CJ, Wei MC, Bassik MC, Donnelly AJ, Kampmann M, Iwasaki M, et al. Next-generation NAMPT inhibitors identified by sequential high-throughput phenotypic chemical and functional genomic screens. Chem Biol. 2013;20:1352–63.

    Article  CAS  PubMed  Google Scholar 

  17. Dong G, Chen W, Wang X, Yang X, Xu T, Wang P, et al. Small molecule inhibitors simultaneously targeting cancer metabolism and epigenetics: discovery of novel nicotinamide phosphoribosyltransferase (NAMPT) and histone deacetylase (HDAC) dual inhibitors. J Med Chem. 2017;60:7965–83.

    Article  CAS  PubMed  Google Scholar 

  18. Palacios DS, Meredith E, Kawanami T, Adams C, Chen X, Darsigny V, et al. Structure based design of nicotinamide phosphoribosyltransferase (NAMPT) inhibitors from a phenotypic screen. Bioorg Med Chem Lett. 2018;28:365–70.

    Article  CAS  PubMed  Google Scholar 

  19. Estoppey D, Hewett JW, Guy CT, Harrington E, Thomas JR, Schirle M, et al. Identification of a novel NAMPT inhibitor by CRISPR/Cas9 chemogenomic profiling in mammalian cells. Sci Rep. 2017;7:42728.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Korotchkina L, Kazyulkin D, Komarov PG, Polinsky A, Andrianova EL, Joshi S, et al. OT-82, a novel anticancer drug candidate that targets the strong dependence of hematological malignancies on NAD biosynthesis. Leukemia. 2019. https://doi.org/10.1038/s41375-019-0692-5

  21. Houghton PJ, Morton CL, Tucker C, Payne D, Favours E, Cole C, et al. The pediatric preclinical testing program: description of models and early testing results. Pediatr Blood Cancer. 2007;49:928–40.

    Article  PubMed  Google Scholar 

  22. Liem NL, Papa RA, Milross CG, Schmid MA, Tajbakhsh M, Choi S, et al. Characterization of childhood acute lymphoblastic leukemia xenograft models for the preclinical evaluation of new therapies. Blood. 2004;103:3905–14.

    Article  CAS  PubMed  Google Scholar 

  23. Jones L, Richmond J, Evans K, Carol H, Jing D, Kurmasheva RT, et al. Bioluminescence imaging enhances analysis of drug responses in a patient-derived xenograft model of pediatric ALL. Clin Cancer Res. 2017;23:3744–55.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Somers K, Chudakova DA, Middlemiss SM, Wen VW, Clifton M, Kwek A, et al. CCI-007, a novel small molecule with cytotoxic activity against infant leukemia with MLL rearrangements. Oncotarget. 2016;7:46067–87.

    Article  PubMed  PubMed Central  Google Scholar 

  25. Khaw SL, Suryani S, Evans K, Richmond J, Robbins A, Kurmasheva RT, et al. Venetoclax responses of pediatric ALL xenografts reveal sensitivity of MLL-rearranged leukemia. Blood. 2016;128:1382–95.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  26. Somers K, Wen VW, Middlemiss SMC, Osborne B, Forgham H, Jung M, et al. A novel small molecule that kills a subset of MLL-rearranged leukemia cells by inducing mitochondrial dysfunction. Oncogene. 2019;38:3824–42.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Somers K, Kosciolek A, Bongers A, El-Ayoubi A, Karsa M, Mayoh C, et al. Potent antileukemic activity of curaxin CBL0137 against MLL-rearranged leukemia. Int J Cancer. 2019. Jul 20. https://doi.org/10.1002/ijc.32582.

  28. Kees UR, Ford J, Watson M, Murch A, Ringner M, Walker RL, et al. Gene expression profiles in a panel of childhood leukemia cell lines mirror critical features of the disease. Mol Cancer Ther. 2003;2:671–7.

    CAS  PubMed  Google Scholar 

  29. Lock RB, Liem N, Farnsworth ML, Milross CG, Xue C, Tajbakhsh M, et al. The nonobese diabetic/severe combined immunodeficient (NOD/SCID) mouse model of childhood acute lymphoblastic leukemia reveals intrinsic differences in biologic characteristics at diagnosis and relapse. Blood. 2002;99:4100–8.

    Article  CAS  PubMed  Google Scholar 

  30. Watson M, Roulston A, Belec L, Billot X, Marcellus R, Bedard D, et al. The small molecule GMX1778 is a potent inhibitor of NAD+ biosynthesis: strategy for enhanced therapy in nicotinic acid phosphoribosyltransferase 1-deficient tumors. Mol Cell Biol. 2009;29:5872–88.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Chan M, Gravel M, Bramoulle A, Bridon G, Avizonis D, Shore GC, et al. Synergy between the NAMPT inhibitor GMX1777(8) and pemetrexed in non-small cell lung cancer cells is mediated by PARP activation and enhanced NAD consumption. Cancer Res. 2014;74:5948–54.

    Article  CAS  PubMed  Google Scholar 

  32. Xiao Y, Elkins K, Durieux JK, Lee L, Oeh J, Yang LX, et al. Dependence of tumor cell lines and patient-derived tumors on the NAD salvage pathway renders them sensitive to NAMPT inhibition with GNE-618. Neoplasia. 2013;15:1151–60.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  33. Xiao Y, Kwong M, Daemen A, Belvin M, Liang X, Hatzivassiliou G, et al. Metabolic Response to NAD Depletion across Cell Lines Is Highly Variable. PLoS ONE. 2016;11:e0164166.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  34. Gehrke I, Bouchard ED, Beiggi S, Poeppl AG, Johnston JB, Gibson SB, et al. On-target effect of FK866, a nicotinamide phosphoribosyl transferase inhibitor, by apoptosis-mediated death in chronic lymphocytic leukemia cells. Clin Cancer Res. 2014;20:4861–72.

    Article  CAS  PubMed  Google Scholar 

  35. Cerna D, Li H, Flaherty S, Takebe N, Coleman CN, Yoo SS. Inhibition of nicotinamide phosphoribosyltransferase (NAMPT) activity by small molecule GMX1778 regulates reactive oxygen species (ROS)-mediated cytotoxicity in a p53- and nicotinic acid phosphoribosyltransferase1 (NAPRT1)-dependent manner. J Biol Chem. 2012;287:22408–17.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Coustan-Smith E, Mullighan CG, Onciu M, Behm FG, Raimondi SC, Pei D, et al. Early T-cell precursor leukaemia: a subtype of very high-risk acute lymphoblastic leukaemia. Lancet Oncol. 2009;10:147–56.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. van der Veer A, Waanders E, Pieters R, Willemse ME, Van Reijmersdal SV, Russell LJ, et al. Independent prognostic value of BCR-ABL1-like signature and IKZF1 deletion, but not high CRLF2 expression, in children with B-cell precursor ALL. Blood. 2013;122:2622–9.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  38. Szymanska B, Wilczynska-Kalak U, Kang MH, Liem NL, Carol H, Boehm I, et al. Pharmacokinetic modeling of an induction regimen for in vivo combined testing of novel drugs against pediatric acute lymphoblastic leukemia xenografts. PLoS ONE. 2012;7:e33894.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Kotecha RS, Gottardo NG, Kees UR, Cole CH. The evolution of clinical trials for infant acute lymphoblastic leukemia. Blood Cancer J. 2014;4:e200.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Grolla AA, Travelli C, Genazzani AA, Sethi JK. Extracellular nicotinamide phosphoribosyltransferase, a new cancer metabokine. Br J Pharmacol. 2016;173:2182–94.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Moore Z, Chakrabarti G, Luo X, Ali A, Hu Z, Fattah FJ, et al. NAMPT inhibition sensitizes pancreatic adenocarcinoma cells to tumor-selective, PAR-independent metabolic catastrophe and cell death induced by beta-lapachone. Cell Death Dis. 2015;6:e1599.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Bi TQ, Che XM, Liao XH, Zhang DJ, Long HL, Li HJ, et al. Overexpression of Nampt in gastric cancer and chemopotentiating effects of the Nampt inhibitor FK866 in combination with fluorouracil. Oncol Rep. 2011;26:1251–7.

    CAS  PubMed  Google Scholar 

  43. Cruickshank MN, Ford J, Cheung LC, Heng J, Singh S, Wells J, et al. Systematic chemical and molecular profiling of MLL-rearranged infant acute lymphoblastic leukemia reveals efficacy of romidepsin. Leukemia. 2017;31:40–50.

    Article  CAS  PubMed  Google Scholar 

  44. Leoni V, Biondi A. Tyrosine kinase inhibitors in BCR-ABL positive acute lymphoblastic leukemia. Haematologica. 2015;100:295–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Bajrami I, Kigozi A, Van Weverwijk A, Brough R, Frankum J, Lord CJ, et al. Synthetic lethality of PARP and NAMPT inhibition in triple-negative breast cancer cells. EMBO Mol Med. 2012;4:1087–96.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Cea M, Soncini D, Fruscione F, Raffaghello L, Garuti A, Emionite L, et al. Synergistic interactions between HDAC and sirtuin inhibitors in human leukemia cells. PLoS ONE. 2011;6:e22739.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Zoppoli G, Cea M, Soncini D, Fruscione F, Rudner J, Moran E, et al. Potent synergistic interaction between the Nampt inhibitor APO866 and the apoptosis activator TRAIL in human leukemia cells. Exp Hematol. 2010;38:979–88.

    Article  CAS  PubMed  Google Scholar 

  48. Nahimana A, Aubry D, Breton CS, Majjigapu SR, Sordat B, Vogel P, et al. The anti-lymphoma activity of APO866, an inhibitor of nicotinamide adenine dinucleotide biosynthesis, is potentialized when used in combination with anti-CD20 antibody. Leuk Lymphoma. 2014;55:2141–50.

    Article  CAS  PubMed  Google Scholar 

  49. Tateishi K, Wakimoto H, Iafrate AJ, Tanaka S, Loebel F, Lelic N, et al. Extreme vulnerability of IDH1 mutant cancers to NAD+ depletion. Cancer Cell. 2015;28:773–84.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Lee J, Kim H, Lee JE, Shin SJ, Oh S, Kwon G, et al. Selective cytotoxicity of the NAMPT inhibitor FK866 toward gastric cancer cells with markers of the epithelial-mesenchymal transition, due to loss of NAPRT. Gastroenterology. 2018;155:799–814 e13.

    Article  CAS  PubMed  Google Scholar 

  51. Shames DS, Elkins K, Walter K, Holcomb T, Du P, Mohl D, et al. Loss of NAPRT1 expression by tumor-specific promoter methylation provides a novel predictive biomarker for NAMPT inhibitors. Clin Cancer Res. 2013;19:6912–23.

    Article  CAS  PubMed  Google Scholar 

  52. Piacente F, Caffa I, Ravera S, Sociali G, Passalacqua M, Vellone VG, et al. Nicotinic acid phosphoribosyltransferase regulates cancer cell metabolism, susceptibility to NAMPT inhibitors, and DNA repair. Cancer Res. 2017;77:3857–69.

    Article  CAS  PubMed  Google Scholar 

  53. Konstantinopoulos PA, Spentzos D, Karlan BY, Taniguchi T, Fountzilas E, Francoeur N, et al. Gene expression profile of BRCAness that correlates with responsiveness to chemotherapy and with outcome in patients with epithelial ovarian cancer. J Clin Oncol. 2010;28:3555–61.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Minchom A, Aversa C, Lopez J. Dancing with the DNA damage response: next-generation anti-cancer therapeutic strategies. Ther Adv Med Oncol. 2018;10:1758835918786658.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  55. Peek CB, Affinati AH, Ramsey KM, Kuo HY, Yu W, Sena LA, et al. Circadian clock NAD+ cycle drives mitochondrial oxidative metabolism in mice. Science. 2013;342:1243417.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  56. Chini CC, Guerrico AM, Nin V, Camacho-Pereira J, Escande C, Barbosa MT, et al. Targeting of NAD metabolism in pancreatic cancer cells: potential novel therapy for pancreatic tumors. Clin Cancer Res. 2014;20:120–30.

    Article  CAS  PubMed  Google Scholar 

  57. Chini EN. CD38 as a regulator of cellular NAD: a novel potential pharmacological target for metabolic conditions. Curr Pharm Des. 2009;15:57–63.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. Lee HC. Structure and enzymatic functions of human CD38. Mol Med. 2006;12:317–23.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Lord CJ, Ashworth A. The DNA damage response and cancer therapy. Nature. 2012;481:287–94.

    Article  CAS  PubMed  Google Scholar 

  60. Di Stefano G, Manerba M, Vettraino M. NAD metabolism and functions: a common therapeutic target for neoplastic, metabolic and neurodegenerative diseases. Curr Top Med Chem. 2013;13:2918–29.

    Article  PubMed  CAS  Google Scholar 

  61. Canto C, Menzies KJ, Auwerx J. NAD(+) metabolism and the control of energy homeostasis: a balancing Act between mitochondria and the nucleus. Cell Metab. 2015;22:31–53.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. Zabka TS, Singh J, Dhawan P, Liederer BM, Oeh J, Kauss MA, et al. Retinal toxicity, in vivo and in vitro, associated with inhibition of nicotinamide phosphoribosyltransferase. Toxicol Sci. 2015;144:163–72.

    Article  CAS  PubMed  Google Scholar 

  63. Misner DL, Kauss MA, Singh J, Uppal H, Bruening-Wright A, Liederer BM, et al. Cardiotoxicity associated with nicotinamide phosphoribosyltransferase inhibitors in rodents and in rat and human-derived cells lines. Cardiovasc Toxicol. 2017;17:307–18.

    Article  CAS  PubMed  Google Scholar 

  64. Jones L, Carol H, Evans K, Richmond J, Houghton PJ, Smith MA, et al. A review of new agents evaluated against pediatric acute lymphoblastic leukemia by the pediatric preclinical testing program. Leukemia. 2016;30:2133–41.

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

This research was supported by grants from the National Cancer Institute (CA199222 and CA199000), The National Health and Medical Research Council of Australia (NHMRC Fellowships APP1059804 and APP1157871), Cancer Australia and Kids’ Cancer Project (Priority-driven Collaborative Cancer Research Scheme APP1164865), Anthony Rothe Memorial Trust, Cancer Council NSW (PG16-01), Tenix Foundation, ISG Foundation, the Children’s Leukemia and Cancer Research Foundation (Perth), and Australian Postgraduate Awards from the Australian Government Department of Education and Training. Children’s Cancer Institute is affiliated with the UNSW Sydney and the Sydney Children’s Hospital Network. The authors would like to thank Raymond Yung and Lisa McDermott (CCI) for their help with experiments.

Author information

Authors and Affiliations

Authors

Contributions

KS, KE, LC, AK, MK, TP, AB, AE, HF, LJ, SM, OC, LK, and MG conducted the experiments. KS, KE, LC, MK, HF, LJ, LK, BT, and SWE analyzed the data. CM performed the analysis of the RNA sequencing and SNP data. URK provided guidance and access to the cell lines and patient material used in the study. KS, KE, LJ, AG, OC, MDN, MH, RBL, and MJH conceived the project and designed the experiments. MAS provided support with study design. KS wrote the manuscript under the guidance of MJH, MDN, MH, and RBL who critically reviewed the manuscript. KE assisted in manuscript preparation. All authors reviewed the manuscript.

Corresponding author

Correspondence to Michelle J. Henderson.

Ethics declarations

Conflict of interest

AVG is a consultant of and OC, MG, and LK are employed by Oncotartis, Inc. which developed and holds the IP on OT-82.

Additional information

Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Supplementary information

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Somers, K., Evans, K., Cheung, L. et al. Effective targeting of NAMPT in patient-derived xenograft models of high-risk pediatric acute lymphoblastic leukemia. Leukemia 34, 1524–1539 (2020). https://doi.org/10.1038/s41375-019-0683-6

Download citation

  • Received:

  • Revised:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41375-019-0683-6

This article is cited by

Search

Quick links