A Phase I Trial of DFMO Targeting Polyamine Addiction in Patients with Relapsed/Refractory Neuroblastoma

Giselle L Saulnier Sholler, Eugene W Gerner, Genevieve Bergendahl, Robert B MacArthur, Alyssa VanderWerff, Takamaru Ashikaga, Jeffrey P Bond, William Ferguson, William Roberts, Randal K Wada, Don Eslin, Jacqueline M Kraveka, Joel Kaplan, Deanna Mitchell, Nehal S Parikh, Kathleen Neville, Leonard Sender, Timothy Higgins, Masao Kawakita, Kyoko Hiramatsu, Shun-Suke Moriya, André S Bachmann, Giselle L Saulnier Sholler, Eugene W Gerner, Genevieve Bergendahl, Robert B MacArthur, Alyssa VanderWerff, Takamaru Ashikaga, Jeffrey P Bond, William Ferguson, William Roberts, Randal K Wada, Don Eslin, Jacqueline M Kraveka, Joel Kaplan, Deanna Mitchell, Nehal S Parikh, Kathleen Neville, Leonard Sender, Timothy Higgins, Masao Kawakita, Kyoko Hiramatsu, Shun-Suke Moriya, André S Bachmann

Abstract

Background: Neuroblastoma (NB) is the most common cancer in infancy and most frequent cause of death from extracranial solid tumors in children. Ornithine decarboxylase (ODC) expression is an independent indicator of poor prognosis in NB patients. This study investigated safety, response, pharmacokinetics, genetic and metabolic factors associated with ODC in a clinical trial of the ODC inhibitor difluoromethylornithine (DFMO) ± etoposide for patients with relapsed or refractory NB.

Methods and findings: Twenty-one patients participated in a phase I study of daily oral DFMO alone for three weeks, followed by additional three-week cycles of DFMO plus daily oral etoposide. No dose limiting toxicities (DLTs) were identified in patients taking doses of DFMO between 500-1500 mg/m2 orally twice a day. DFMO pharmacokinetics, single nucleotide polymorphisms (SNPs) in the ODC gene and urinary levels of substrates for the tissue polyamine exporter were measured. Urinary polyamine levels varied among patients at baseline. Patients with the minor T-allele at rs2302616 of the ODC gene had higher baseline levels (p=0.02) of, and larger decreases in, total urinary polyamines during the first cycle of DFMO therapy (p=0.003) and had median progression free survival (PFS) that was over three times longer, compared to patients with the major G allele at this locus although this last result was not statistically significant (p=0.07). Six of 18 evaluable patients were progression free during the trial period with three patients continuing progression free at 663, 1559 and 1573 days after initiating treatment. Median progression-free survival was less among patients having increased urinary polyamines, especially diacetylspermine, although this result was not statistically significant (p=0.056).

Conclusions: DFMO doses of 500-1500 mg/m2/day are safe and well tolerated in children with relapsed NB. Children with the minor T allele at rs2302616 of the ODC gene with relapsed or refractory NB had higher levels of urinary polyamine markers and responded better to therapy containing DFMO, compared to those with the major G allele at this locus. These findings suggest that this patient subset may display dependence on polyamines and be uniquely susceptible to therapies targeting this pathway.

Trial registration: Clinicaltrials.gov NCT#01059071.

Trial registration: ClinicalTrials.gov NCT01059071.

Conflict of interest statement

Competing Interests: The authors of this manuscript have the following competing interests: E.W.G. is a paid employee of Cancer Prevention Pharmaceuticals (CPP). He has an ownership interest in, and serves on the board of, CPP. This does not alter the authors' adherence to PLOS ONE policies on sharing data and materials.

Figures

Fig 1. NMTRC002 CONSORT Flow Diagram- modified…
Fig 1. NMTRC002 CONSORT Flow Diagram- modified for non-randomized trial design.
Fig 2. Flowchart of NMTRC 002—Safety Study…
Fig 2. Flowchart of NMTRC 002—Safety Study for Refractory or Relapsed Neuroblastoma With DFMO Alone and in Combination With Etoposide.
Fig 3. Serum DFMO concentration versus time…
Fig 3. Serum DFMO concentration versus time measurements for three patients receiving 750 mg/m2 PO BID during cycle 1 of therapy.
Fig 4. Rationale for DFMO- and specific…
Fig 4. Rationale for DFMO- and specific genetic and metabolic markers of DFMO effect, in neuroblastoma
. ODC transcription is influenced by specific genetic variability, including the SNPs rs2302615 [19, 22] and rs2302616 [24]. The DFMO target ODC decarboxylates ornithine to form the diamine putrescine, which is then metabolized into longer chain amines. Spermidine is a substrate for two acetyltransferases that monoacetylate this amine at either the N1 or N8 positions. Spermine is a substrate for one of these transferases (SAT1), which diacetylates this amine. Putrescine, the monoacetylspermidines and diacetylspermine are all substrates for the solute carrier transporter SLC3A2/Y+LAT, which exports these amines.
Fig 5. Progression free survival (PFS) and…
Fig 5. Progression free survival (PFS) and overall survival (OS) rates in patients enrolled in NMTRC 002 (N = 21).
The number of patients shown at risk for disease progression (PFS) or death (OS) is shown in the figure.

References

    1. Maris JM. Recent advances in neuroblastoma. The New England journal of medicine. 2010;362(23):2202–11. 10.1056/NEJMra0804577
    1. Park JR, Bagatell R, London WB, Maris JM, Cohn SL, Mattay KK, et al. Children's Oncology Group's 2013 blueprint for research: neuroblastoma. Pediatric blood & cancer. 2013;60(6):985–93.
    1. Yu AL, Gilman AL, Ozkaynak MF, London WB, Kreissman SG, Chen HX, et al. Anti-GD2 antibody with GM-CSF, interleukin-2, and isotretinoin for neuroblastoma. The New England journal of medicine. 2010;363(14):1324–34. 10.1056/NEJMoa0911123
    1. Modak S, Cheung NK. Neuroblastoma: Therapeutic strategies for a clinical enigma. Cancer treatment reviews. 2010;36(4):307–17. 10.1016/j.ctrv.2010.02.006
    1. Bachmann AS. The role of polyamines in human cancer: prospects for drug combination therapies. Hawaii medical journal. 2004;63(12):371–4.
    1. Wallick CJ, Gamper I, Thorne M, Feith DJ, Takasaki KY, Wilson SM, et al. Key role for p27Kip1, retinoblastoma protein Rb, and MYCN in polyamine inhibitor-induced G1 cell cycle arrest in MYCN-amplified human neuroblastoma cells. Oncogene. 2005;24(36):5606–18.
    1. Koomoa DL, Yco LP, Borsics T, Wallick CJ, Bachmann AS. Ornithine Decarboxylase Inhibition by {alpha}-Difluoromethylornithine Activates Opposing Signaling Pathways via Phosphorylation of Both Akt/Protein Kinase B and p27Kip1 in Neuroblastoma. Cancer research. 2008;68(23):9825–31. 10.1158/0008-5472.CAN-08-1865
    1. Koomoa DL, Geerts D, Lange I, Koster J, Pegg AE, Feith DJ, et al. DFMO/eflornithine inhibits migration and invasion downstream of MYCN and involves p27Kip1 activity in neuroblastoma. Int J Oncol. 2013;42(4):1219–28. 10.3892/ijo.2013.1835
    1. Hogarty MD, Norris MD, Davis K, Liu X, Evageliou NF, Hayes CS, et al. ODC1 is a critical determinant of MYCN oncogenesis and a therapeutic target in neuroblastoma. Cancer research. 2008;68(23):9735–45. 10.1158/0008-5472.CAN-07-6866
    1. Rounbehler RJ, Li W, Hall MA, Yang C, Fallahi M, Cleveland JL. Targeting ornithine decarboxylase impairs development of MYCN-amplified neuroblastoma. Cancer research. 2009;69(2):547–53. 10.1158/0008-5472.CAN-08-2968
    1. Geerts D, Koster J, Albert D, Koomoa DL, Feith DJ, Pegg AE, et al. The polyamine metabolism genes ornithine decarboxylase and antizyme 2 predict aggressive behavior in neuroblastomas with and without MYCN amplification. International journal of cancer Journal international du cancer. 2010;126(9):2012–24. 10.1002/ijc.25074
    1. Auvinen M, Paasinen A, Andersson LC, Holtta E. Ornithine decarboxylase activity is critical for cell transformation. Nature. 1992;360(6402):355–8.
    1. Auvinen M, Paasinen-Sohns A, Hirai H, Andersson LC, Holtta E. Ornithine decarboxylase- and ras-induced cell transformations: reversal by protein tyrosine kinase inhibitors and role of pp130CAS. Molecular and cellular biology. 1995;15(12):6513–25.
    1. Auvinen M, Jarvinen K, Hotti A, Okkeri J, Laitinen J, Janne OA, et al. Transcriptional regulation of the ornithine decarboxylase gene by c-Myc/Max/Mad network and retinoblastoma protein interacting with c-Myc. The international journal of biochemistry & cell biology. 2003;35(4):496–521.
    1. Lutz W, Stohr M, Schurmann J, Wenzel A, Lohr A, Schwab M. Conditional expression of N-myc in human neuroblastoma cells increases expression of alpha-prothymosin and ornithine decarboxylase and accelerates progression into S-phase early after mitogenic stimulation of quiescent cells. Oncogene. 1996;13(4):803–12.
    1. Ben-Yosef T, Yanuka O, Halle D, Benvenisty N. Involvement of Myc targets in c-myc and N-myc induced human tumors. Oncogene. 1998;17(2):165–71.
    1. Lu X, Pearson A, Lunec J. The MYCN oncoprotein as a drug development target. Cancer letters. 2003;197(1–2):125–30.
    1. Bachmann AS, Geerts D, Sholler G. Neuroblastoma: Ornithine decarboxylase and polyamines are novel targets for therapeutic intervention In: Hayat MA, editor. Pediatric Cancer, Neuroblastoma: Diagnosis, Therapy, and Prognosis. 1: Springer; 2012. p. 91–103.
    1. Martinez ME, O'Brien TG, Fultz KE, Babbar N, Yerushalmi H, Qu N, et al. Pronounced reduction in adenoma recurrence associated with aspirin use and a polymorphism in the ornithine decarboxylase gene. Proceedings of the National Academy of Sciences of the United States of America. 2003;100(13):7859–64.
    1. Visvanathan K, Helzlsouer KJ, Boorman DW, Strickland PT, Hoffman SC, Comstock GW, et al. Association among an ornithine decarboxylase polymorphism, androgen receptor gene (CAG) repeat length and prostate cancer risk. J Urol. 2004;171(2 Pt 1):652–5.
    1. Brown I, Halliday S, Greig H, Heys SD, Wallace HM, Schofield AC. Genetic polymorphism in ornithine decarboxylase and risk of breast cancer. Fam Cancer. 2009;8(4):307–11. 10.1007/s10689-009-9237-9
    1. Zell JA, Ziogas A, Ignatenko N, Honda J, Qu N, Bobbs AS, et al. Associations of a polymorphism in the ornithine decarboxylase gene with colorectal cancer survival. Clin Cancer Res. 2009;15(19):6208–16. 10.1158/1078-0432.CCR-09-0592
    1. Norris MDG, Purgato S, Valli E, Rihani A, Van Maerken T, Vandesompele J, et al. The ornithine decarboxylase G317A polymorphism is prognostic of outcome in primary neuroblastoma and differentially affects promoter binding by the MYCN oncogene. Advances in Neuroblastoma Research. 2014;Abstract OR059:140.
    1. Garcia-Huidobro JM, Brooks T, Gerner EW. Functional consequence of genetic variability in a G-quadruplex structure in the ornithine decarboxylase (odc1) gene. Nucleic Acids Research. 2014;submitted.
    1. Garcia-Huidobro JT, Wertheim BC, Gerner EW. Intron 1 polymorphisms cooperate to modulate odc1 transcriptional activity and risk of colorectal adenomas. Cancer research. 2014;submitted.
    1. Casero RA Jr., Marton LJ. Targeting polyamine metabolism and function in cancer and other hyperproliferative diseases. Nature reviews Drug discovery. 2007;6(5):373–90.
    1. Pegg AE, Feith DJ. Polyamines and neoplastic growth. Biochemical Society transactions. 2007;35(Pt 2):295–9.
    1. Priotto G, Kasparian S, Mutombo W, Ngouama D, Ghorashian S, Arnold U, et al. Nifurtimox-eflornithine combination therapy for second-stage African Trypanosoma brucei gambiense trypanosomiasis: a multicentre, randomised, phase III, non-inferiority trial. Lancet. 2009;374(9683):56–64. 10.1016/S0140-6736(09)61117-X
    1. Alirol E, Schrumpf D, Amici Heradi J, Riedel A, de Patoul C, Quere M, et al. Nifurtimox-eflornithine combination therapy for second-stage gambiense human African trypanosomiasis: Medecins Sans Frontieres experience in the Democratic Republic of the Congo. Clinical infectious diseases: an official publication of the Infectious Diseases Society of America. 2013;56(2):195–203.
    1. Blume-Peytavi U, Hahn S. Medical treatment of hirsutism. Dermatologic therapy. 2008;21(5):329–39. 10.1111/j.1529-8019.2008.00215.x
    1. Kushner BH, Kramer K, Cheung NK. Oral etoposide for refractory and relapsed neuroblastoma. Journal of clinical oncology: official journal of the American Society of Clinical Oncology. 1999;17(10):3221–5.
    1. Dorr RT, Liddil JD, Gerner EW. Modulation of etoposide cytotoxicity and DNA strand scission in L1210 and 8226 cells by polyamines. Cancer research. 1986;46(8):3891–5.
    1. Thompson PA, Wertheim BC, Zell JA, Chen WP, McLaren CE, LaFleur BJ, et al. Levels of rectal mucosal polyamines and prostaglandin E2 predict ability of DFMO and sulindac to prevent colorectal adenoma. Gastroenterology. 2010;139(3):797–805, e1 10.1053/j.gastro.2010.06.005
    1. Kawakita M, Hiramatsu K, Yanagiya M, Doi Y, Kosaka M. Determination of N(1),N(1)(2)-diacetylspermine in urine: a novel tumor marker. Methods in molecular biology. 2011;720:367–78. 10.1007/978-1-61779-034-8_23
    1. Kablfleisch RL. The Statistical Analysis of Failure Time Data. New York: Wiley; 1980.
    1. Team RC. R: A language and environment for statistical computing. Vienna, Austria2014. Available: .
    1. T T. _A Package for Survival Analysis in S_. R package version 2.37–7 ed2014.
    1. Therneau TM, Grambsch PM. Modeling survival data: extending the Cox model. New York: Springer; 2000. xiii, 350 p. p.
    1. Harrington TR.. A class of rank test procedures for censored survival data. Biometrika 1982;69:553–66.
    1. Davison AC, Hinkley DV. Bootstrap methods and their application. Cambridge; New York, NY, USA: Cambridge University Press; 1997. x, 582 p. p.
    1. Ripley ACaB. boot: Bootstrap R (S-Plus) Functions. 2015.
    1. Xie X, Gillies RJ, Gerner EW. Characterization of a diamine exporter in Chinese hamster ovary cells and identification of specific polyamine substrates. J Biol Chem. 1997;272(33):20484–9.
    1. Uemura T, Yerushalmi HF, Tsaprailis G, Stringer DE, Pastorian KE, Hawel L 3rd, et al. Identification and characterization of a diamine exporter in colon epithelial cells. J Biol Chem. 2008;283(39):26428–35. 10.1074/jbc.M804714200
    1. Uemura T, Stringer DE, Blohm-Mangone KA, Gerner EW. Polyamine transport is mediated by both endocytic and solute carrier transport mechanisms in the gastrointestinal tract. Am J Physiol Gastrointest Liver Physiol. 2010;299(2):G517–22. 10.1152/ajpgi.00169.2010
    1. Gerner EW, Meyskens FL Jr. Polyamines and cancer: old molecules, new understanding. Nature Reviews Cancer. 2004;4(10):781–92.
    1. Meyskens FL Jr., McLaren CE, Pelot D, Fujikawa-Brooks S, Carpenter PM, Hawk E, et al. Difluoromethylornithine plus sulindac for the prevention of sporadic colorectal adenomas: a randomized placebo-controlled, double-blind trial. Cancer prevention research. 2008;1(1):32–8. 10.1158/1940-6207.CAPR-08-0042
    1. Bailey HH, Kim K, Verma AK, Sielaff K, Larson PO, Snow S, et al. A randomized, double-blind, placebo-controlled phase 3 skin cancer prevention study of {alpha}-difluoromethylornithine in subjects with previous history of skin cancer. Cancer prevention research. 2010;3(1):35–47. 10.1158/1940-6207.CAPR-09-0096
    1. Kareva I, Waxman DJ, Klement GL. Metronomic chemotherapy: An attractive alternative to maximum tolerated dose therapy that can activate anti-tumor immunity and minimize therapeutic resistance. Cancer letters. 2014.
    1. Robison NJ, Campigotto F, Chi SN, Manley PE, Turner CD, Zimmerman MA, et al. A phase II trial of a multi-agent oral antiangiogenic (metronomic) regimen in children with recurrent or progressive cancer. Pediatric blood & cancer. 2014;61(4):636–42.
    1. Evageliou NF, Hogarty MD. Disrupting polyamine homeostasis as a therapeutic strategy for neuroblastoma. Clin Cancer Res. 2009;15(19):5956–61. 10.1158/1078-0432.CCR-08-3213
    1. Gamble LD, Hogarty MD, Liu X, Ziegler DS, Marshall G, Norris MD, et al. Polyamine pathway inhibition as a novel therapeutic approach to treating neuroblastoma. Frontiers in oncology. 2012;2:162 10.3389/fonc.2012.00162
    1. Weinstein IB, Joe AK. Mechanisms of disease: Oncogene addiction—a rationale for molecular targeting in cancer therapy. Nature clinical practice Oncology. 2006;3(8):448–57.
    1. Weinstein IB, Joe A. Oncogene addiction. Cancer research. 2008;68(9):3077–80; discussion 80. 10.1158/0008-5472.CAN-07-3293
    1. Choi PS, Li Y, Felsher DW. Addiction to multiple oncogenes can be exploited to prevent the emergence of therapeutic resistance. Proceedings of the National Academy of Sciences of the United States of America. 2014.
    1. Pugh TJ, Morozova O, Attiyeh EF, Asgharzadeh S, Wei JS, Auclair D, et al. The genetic landscape of high-risk neuroblastoma. Nature genetics. 2013;45(3):279–84. 10.1038/ng.2529
    1. Samal K, Zhao P, Kendzicky A, Yco LP, McClung H, Gerner EW, et al. AMXT-15, a novel polyamine transport inhibitor, synergizes with DFMO in inhibiting cell proliferation by targeting both ornithine decarboxylase and polyamine transport. International Journal of Cancer. 2013;accepted.
    1. Lange I, Geerts D, Feith DJ, Mocz G, Koster J, Bachmann AS. Novel interaction of ornithine decarboxylase with sepiapterin reductase regulates neuroblastoma cell proliferation. Journal of molecular biology. 2014;426(2):332–46. 10.1016/j.jmb.2013.09.037
    1. Pendyala L, Creaven PJ, Porter CW. Urinary and erythrocyte polyamines during the evaluation of oral alpha-difluoromethylornithine in a phase I chemoprevention clinical trial. Cancer epidemiology, biomarkers & prevention: a publication of the American Association for Cancer Research, cosponsored by the American Society of Preventive Oncology. 1993;2(3):235–41.
    1. Carbone PP, Douglas JA, Thomas J, Tutsch K, Pomplun M, Hamielec M, et al. Bioavailability study of oral liquid and tablet forms of alpha-difluoromethylornithine. Clinical Cancer Research. 2000;6(10):3850–4.
    1. Witherspoon M, Chen Q, Kopelovich L, Gross SS, Lipkin SM. Unbiased metabolite profiling indicates that a diminished thymidine pool is the underlying mechanism of colon cancer chemoprevention by alpha-difluoromethylornithine. Cancer discovery. 2013;3(9):1072–81. 10.1158/-12-0305
    1. Babbar N, Murray-Stewart T, Casero RA Jr. Inflammation and polyamine catabolism: the good, the bad and the ugly. Biochemical Society transactions. 2007;35(Pt 2):300–4.
    1. Soda K. The mechanisms by which polyamines accelerate tumor spread. Journal of experimental & clinical cancer research: CR. 2011;30:95.
    1. Hayes CS, Burns MR, Gilmour SK. Polyamine blockade promotes antitumor immunity. Oncoimmunology. 2014;3(1):e27360
    1. Russell DH, Levy CC. Polyamine accumulation and biosynthesis in a mouse L1210 leukemia. Cancer research. 1971;31(3):248–51.
    1. Hiramatsu K, Takahashi K, Yamaguchi T, Matsumoto H, Miyamoto H, Tanaka S, et al. N(1),N(12)-Diacetylspermine as a sensitive and specific novel marker for early- and late-stage colorectal and breast cancers. Clinical Cancer Research. 2005;11(8):2986–90.
    1. Sholler GL. A Phase 1 Trial of DFMO as a Single Agent and in Combination with Etoposide in Patients with Refractory or Recurrent Neuroblastoma. Proc AACR. 2013.

Source: PubMed

3
Suscribir