Skip to main content
Top
Published in: BMC Cancer 1/2015

Open Access 01-12-2015 | Research article

Effect of sulfasalazine on human neuroblastoma: analysis of sepiapterin reductase (SPR) as a new therapeutic target

Authors: Lisette P. Yco, Dirk Geerts, Gabor Mocz, Jan Koster, André S. Bachmann

Published in: BMC Cancer | Issue 1/2015

Login to get access

Abstract

Background

Neuroblastoma (NB) is an aggressive childhood malignancy in children up to 5 years of age. High-stage tumors frequently relapse even after aggressive multimodal treatment, and then show therapy resistance, typically resulting in patient death. New molecular-targeted compounds that effectively suppress tumor growth and prevent relapse with more efficacy are urgently needed. We and others previously showed that polyamines (PA) like spermidine and spermine are essential for NB tumorigenesis and that DFMO, an inhibitor of the key PA synthesis gene product ODC, is effective both in vitro and in vivo, securing its evaluation in NB clinical trials. To find additional compounds interfering with PA biosynthesis, we tested sulfasalazine (SSZ), an FDA-approved salicylate-based anti-inflammatory and immune-modulatory drug, recently identified to inhibit sepiapterin reductase (SPR). We earlier presented evidence for a physical interaction between ODC and SPR and we showed that RNAi-mediated knockdown of SPR expression significantly reduced native ODC enzyme activity and impeded NB cell proliferation.

Methods

Human NB mRNA expression datasets in the public domain were analyzed using the R2 platform. Cell viability, isobologram, and combination index analyses as a result of SSZ treatment with our without DFMO were carried out in NB cell cultures. Molecular protein-ligand docking was achieved using the GRAMM algorithm. Statistical analyses were performed with the Kruskal-Wallis test, 2log Pearson test, and Student’s t test.

Results

In this study, we show the clinical relevance of SPR in human NB tumors. We found that high SPR expression is significantly correlated to unfavorable NB characteristics like high age at diagnosis, MYCN amplification, and high INSS stage. SSZ inhibits the growth of NB cells in vitro, presumably due to the inhibition of SPR as predicted by computational docking of SSZ into SPR. Importantly, the combination of SSZ with DFMO produces synergistic antiproliferative effects in vitro.

Conclusions

The results suggest the use of SSZ in combination with DFMO for further experiments, and possible prioritization as a novel therapy for the treatment of NB patients.
Literature
1.
go back to reference Brodeur GM. Neuroblastoma: biological insights into a clinical enigma. Nat Rev Cancer. 2003;3(3):203–16.CrossRefPubMed Brodeur GM. Neuroblastoma: biological insights into a clinical enigma. Nat Rev Cancer. 2003;3(3):203–16.CrossRefPubMed
4.
5.
go back to reference Park JR, Eggert A, Caron H. Neuroblastoma: biology, prognosis, and treatment. Hematol Oncol Clin North Am. 2010;24(1):65–86.CrossRefPubMed Park JR, Eggert A, Caron H. Neuroblastoma: biology, prognosis, and treatment. Hematol Oncol Clin North Am. 2010;24(1):65–86.CrossRefPubMed
6.
go back to reference Schwab M, Westermann F, Hero B, Berthold F. Neuroblastoma: biology and molecular and chromosomal pathology. Lancet Oncol. 2003;4(8):472–80.CrossRefPubMed Schwab M, Westermann F, Hero B, Berthold F. Neuroblastoma: biology and molecular and chromosomal pathology. Lancet Oncol. 2003;4(8):472–80.CrossRefPubMed
7.
go back to reference Baker DL, Schmidt ML, Cohn SL, Maris JM, London WB, Buxton A, et al. Outcome after reduced chemotherapy for intermediate-risk neuroblastoma. N Engl J Med. 2010;363(14):1313–23.CrossRefPubMedPubMedCentral Baker DL, Schmidt ML, Cohn SL, Maris JM, London WB, Buxton A, et al. Outcome after reduced chemotherapy for intermediate-risk neuroblastoma. N Engl J Med. 2010;363(14):1313–23.CrossRefPubMedPubMedCentral
8.
go back to reference Cohn SL, Pearson AD, London WB, Monclair T, Ambros PF, Brodeur GM, et al. The International Neuroblastoma Risk Group (INRG) classification system: an INRG Task Force report. J Clin Oncol. 2009;27(2):289–97.CrossRefPubMedPubMedCentral Cohn SL, Pearson AD, London WB, Monclair T, Ambros PF, Brodeur GM, et al. The International Neuroblastoma Risk Group (INRG) classification system: an INRG Task Force report. J Clin Oncol. 2009;27(2):289–97.CrossRefPubMedPubMedCentral
9.
go back to reference Kreissman SG, Seeger RC, Matthay KK, London WB, Sposto R, Grupp SA, et al. Purged versus non-purged peripheral blood stem-cell transplantation for high-risk neuroblastoma (COG A3973): a randomised phase 3 trial. Lancet Oncol. 2013;14(10):999–1008.CrossRefPubMedPubMedCentral Kreissman SG, Seeger RC, Matthay KK, London WB, Sposto R, Grupp SA, et al. Purged versus non-purged peripheral blood stem-cell transplantation for high-risk neuroblastoma (COG A3973): a randomised phase 3 trial. Lancet Oncol. 2013;14(10):999–1008.CrossRefPubMedPubMedCentral
10.
go back to reference Strother DR, London WB, Schmidt ML, Brodeur GM, Shimada H, Thorner P, et al. Outcome after surgery alone or with restricted use of chemotherapy for patients with low-risk neuroblastoma: results of Children’s Oncology Group study P9641. J Clin Oncol. 2012;30(15):1842–8.CrossRefPubMedPubMedCentral Strother DR, London WB, Schmidt ML, Brodeur GM, Shimada H, Thorner P, et al. Outcome after surgery alone or with restricted use of chemotherapy for patients with low-risk neuroblastoma: results of Children’s Oncology Group study P9641. J Clin Oncol. 2012;30(15):1842–8.CrossRefPubMedPubMedCentral
11.
go back to reference Canete A, Gerrard M, Rubie H, Castel V, Di Cataldo A, Munzer C, et al. Poor survival for infants with MYCN-amplified metastatic neuroblastoma despite intensified treatment: the International Society of Paediatric Oncology European Neuroblastoma Experience. J Clin Oncol. 2009;27(7):1014–9.CrossRefPubMed Canete A, Gerrard M, Rubie H, Castel V, Di Cataldo A, Munzer C, et al. Poor survival for infants with MYCN-amplified metastatic neuroblastoma despite intensified treatment: the International Society of Paediatric Oncology European Neuroblastoma Experience. J Clin Oncol. 2009;27(7):1014–9.CrossRefPubMed
12.
go back to reference Kushner BH, Kramer K, LaQuaglia MP, Modak S, Yataghene K, Cheung NK. Reduction from seven to five cycles of intensive induction chemotherapy in children with high-risk neuroblastoma. J Clin Oncol. 2004;22(24):4888–92.CrossRefPubMed Kushner BH, Kramer K, LaQuaglia MP, Modak S, Yataghene K, Cheung NK. Reduction from seven to five cycles of intensive induction chemotherapy in children with high-risk neuroblastoma. J Clin Oncol. 2004;22(24):4888–92.CrossRefPubMed
13.
go back to reference Bachmann AS. The role of polyamines in human cancer: prospects for drug combination therapies. Hawaii Med J. 2004;63(12):371–4.PubMed Bachmann AS. The role of polyamines in human cancer: prospects for drug combination therapies. Hawaii Med J. 2004;63(12):371–4.PubMed
14.
go back to reference Casero Jr RA, Marton LJ. Targeting polyamine metabolism and function in cancer and other hyperproliferative diseases. Nat Rev Drug Discov. 2007;6(5):373–90.CrossRefPubMed Casero Jr RA, Marton LJ. Targeting polyamine metabolism and function in cancer and other hyperproliferative diseases. Nat Rev Drug Discov. 2007;6(5):373–90.CrossRefPubMed
15.
go back to reference Pegg AE. Polyamine metabolism and its importance in neoplastic growth and a target for chemotherapy. Cancer Res. 1988;48(4):759–74.PubMed Pegg AE. Polyamine metabolism and its importance in neoplastic growth and a target for chemotherapy. Cancer Res. 1988;48(4):759–74.PubMed
16.
17.
go back to reference Gerner EW, Meyskens Jr FL. Polyamines and cancer: old molecules, new understanding. Nat Rev Cancer. 2004;4(10):781–92.CrossRefPubMed Gerner EW, Meyskens Jr FL. Polyamines and cancer: old molecules, new understanding. Nat Rev Cancer. 2004;4(10):781–92.CrossRefPubMed
18.
go back to reference 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 Res. 2008;68(23):9825–31.CrossRefPubMedPubMedCentral 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 Res. 2008;68(23):9825–31.CrossRefPubMedPubMedCentral
19.
go back to reference 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.CrossRefPubMed 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.CrossRefPubMed
20.
go back to reference Koomoa DL, Borsics T, Feith DJ, Coleman CC, Wallick CJ, Gamper I, et al. Inhibition of S-adenosylmethionine decarboxylase by inhibitor SAM486A connects polyamine metabolism with p53-Mdm2-Akt/protein kinase B regulation and apoptosis in neuroblastoma. Mol Cancer Ther. 2009;8(7):2067–75.CrossRefPubMedPubMedCentral Koomoa DL, Borsics T, Feith DJ, Coleman CC, Wallick CJ, Gamper I, et al. Inhibition of S-adenosylmethionine decarboxylase by inhibitor SAM486A connects polyamine metabolism with p53-Mdm2-Akt/protein kinase B regulation and apoptosis in neuroblastoma. Mol Cancer Ther. 2009;8(7):2067–75.CrossRefPubMedPubMedCentral
21.
go back to reference 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.PubMedPubMedCentral 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.PubMedPubMedCentral
22.
go back to reference Bandino A, Geerts D, Koster J, Bachmann AS. Deoxyhypusine synthase (DHPS) inhibitor GC7 induces p21/Rb-mediated inhibition of tumor cell growth and DHPS expression correlates with poor prognosis in neuroblastoma patients. Cell Oncol. 2014;37(6):387–98. Bandino A, Geerts D, Koster J, Bachmann AS. Deoxyhypusine synthase (DHPS) inhibitor GC7 induces p21/Rb-mediated inhibition of tumor cell growth and DHPS expression correlates with poor prognosis in neuroblastoma patients. Cell Oncol. 2014;37(6):387–98.
23.
go back to reference 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 Res. 2008;68(23):9735–45.CrossRefPubMedPubMedCentral 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 Res. 2008;68(23):9735–45.CrossRefPubMedPubMedCentral
24.
go back to reference Rounbehler RJ, Li W, Hall MA, Yang C, Fallahi M, Cleveland JL. Targeting ornithine decarboxylase impairs development of MYCN-amplified neuroblastoma. Cancer Res. 2009;69(2):547–53.CrossRefPubMedPubMedCentral Rounbehler RJ, Li W, Hall MA, Yang C, Fallahi M, Cleveland JL. Targeting ornithine decarboxylase impairs development of MYCN-amplified neuroblastoma. Cancer Res. 2009;69(2):547–53.CrossRefPubMedPubMedCentral
25.
go back to reference Sholler G, Currier E, Koomoa DL, Bachmann AS. Synergistic inhibition of neuroblastoma tumor development by targeting ornithine decarboxylase and topoisomerase II. In: 14th Advances in Neuroblastoma Research (ANR) Conference. Stockholm, Sweden, June 21–24; 2010: POT74. Sholler G, Currier E, Koomoa DL, Bachmann AS. Synergistic inhibition of neuroblastoma tumor development by targeting ornithine decarboxylase and topoisomerase II. In: 14th Advances in Neuroblastoma Research (ANR) Conference. Stockholm, Sweden, June 21–24; 2010: POT74.
26.
go back to reference Saulnier Sholler GL, Gerner EW, Bergendahl G, MacArthur MW, VanderWerff A, Ashikaga T, Bond JP, Ferguson W, Roberts W, Wada RK et al.: A phase I trial of DFMO targeting polyamine addiction in patients with relapsed/refractory neuroblastoma. PloS One 2015, In Press. Saulnier Sholler GL, Gerner EW, Bergendahl G, MacArthur MW, VanderWerff A, Ashikaga T, Bond JP, Ferguson W, Roberts W, Wada RK et al.: A phase I trial of DFMO targeting polyamine addiction in patients with relapsed/refractory neuroblastoma. PloS One 2015, In Press.
27.
go back to reference Bachmann AS, Geerts D, Sholler G: Neuroblastoma: Ornithine decarboxylase and polyamines are novel targets for therapeutic intervention. In: Pediatric Cancer, Neuroblastoma: Diagnosis, Therapy, and Prognosis. Volume 1, edn. Edited by Hayat MA: Springer, Heidelberg, Germany. 2012;91–103. Bachmann AS, Geerts D, Sholler G: Neuroblastoma: Ornithine decarboxylase and polyamines are novel targets for therapeutic intervention. In: Pediatric Cancer, Neuroblastoma: Diagnosis, Therapy, and Prognosis. Volume 1, edn. Edited by Hayat MA: Springer, Heidelberg, Germany. 2012;91–103.
28.
go back to reference Bachmann AS, Levin VA. Clinical applications of polyamine-based therapeutics. In: Polyamine Drug Discovery. edn. Edited by Woster PM, Casero RA, Jr.: Royal Society of Chemistry Publishing, Cambridge, UK. 2012;257–276. Bachmann AS, Levin VA. Clinical applications of polyamine-based therapeutics. In: Polyamine Drug Discovery. edn. Edited by Woster PM, Casero RA, Jr.: Royal Society of Chemistry Publishing, Cambridge, UK. 2012;257–276.
29.
go back to reference Samal K, Zhao P, Kendzicky A, Yco LP, McClung H, Gerner E, et al. AMXT-1501, a novel polyamine transport inhibitor, synergizes with DFMO in inhibiting neuroblastoma cell proliferation by targeting both ornithine decarboxylase and polyamine transport. Int J Cancer. 2013;133(6):1323–33.CrossRefPubMed Samal K, Zhao P, Kendzicky A, Yco LP, McClung H, Gerner E, et al. AMXT-1501, a novel polyamine transport inhibitor, synergizes with DFMO in inhibiting neuroblastoma cell proliferation by targeting both ornithine decarboxylase and polyamine transport. Int J Cancer. 2013;133(6):1323–33.CrossRefPubMed
30.
go back to reference Chidley C, Haruki H, Pedersen MG, Muller E, Johnsson K. A yeast-based screen reveals that sulfasalazine inhibits tetrahydrobiopterin biosynthesis. Nat Chem Biol. 2011;7(6):375–83.CrossRefPubMed Chidley C, Haruki H, Pedersen MG, Muller E, Johnsson K. A yeast-based screen reveals that sulfasalazine inhibits tetrahydrobiopterin biosynthesis. Nat Chem Biol. 2011;7(6):375–83.CrossRefPubMed
31.
go back to reference Costigan M, Latremoliere A, Woolf CJ. Analgesia by inhibiting tetrahydrobiopterin synthesis. Curr Opin Pharmacol. 2012;12(1):92–9.CrossRefPubMed Costigan M, Latremoliere A, Woolf CJ. Analgesia by inhibiting tetrahydrobiopterin synthesis. Curr Opin Pharmacol. 2012;12(1):92–9.CrossRefPubMed
32.
go back to reference Lange I, Geerts D, Feith DJ, Mocz G, Koster J, Bachmann AS. Novel interaction of ornithine decarboxylase with sepiapterin reductase regulates neuroblastoma cell proliferation. J Mol Biol. 2014;426(2):332–46.CrossRefPubMed Lange I, Geerts D, Feith DJ, Mocz G, Koster J, Bachmann AS. Novel interaction of ornithine decarboxylase with sepiapterin reductase regulates neuroblastoma cell proliferation. J Mol Biol. 2014;426(2):332–46.CrossRefPubMed
33.
go back to reference Halgren TA. Merck molecular force field. I. Basis, form, scope, parameterization, and performance of MMFF94. J Comp Chem. 1996;17(5/6):490–519.CrossRef Halgren TA. Merck molecular force field. I. Basis, form, scope, parameterization, and performance of MMFF94. J Comp Chem. 1996;17(5/6):490–519.CrossRef
34.
go back to reference Sillitoe I, Cuff AL, Dessailly BH, Dawson NL, Furnham N, Lee D, et al. New functional families (FunFams) in CATH to improve the mapping of conserved functional sites to 3D structures. Nucleic Acids Res. 2013;41(Database issue):D490–498.CrossRefPubMed Sillitoe I, Cuff AL, Dessailly BH, Dawson NL, Furnham N, Lee D, et al. New functional families (FunFams) in CATH to improve the mapping of conserved functional sites to 3D structures. Nucleic Acids Res. 2013;41(Database issue):D490–498.CrossRefPubMed
36.
go back to reference van Rossum MA, Fiselier TJ, Franssen MJ, Zwinderman AH, ten Cate R, van Suijlekom-Smit LW, et al. Sulfasalazine in the treatment of juvenile chronic arthritis: a randomized, double-blind, placebo-controlled, multicenter study. Dutch Juvenile Chronic Arthritis Study Group. Arthritis Rheum. 1998;41(5):808–16.CrossRefPubMed van Rossum MA, Fiselier TJ, Franssen MJ, Zwinderman AH, ten Cate R, van Suijlekom-Smit LW, et al. Sulfasalazine in the treatment of juvenile chronic arthritis: a randomized, double-blind, placebo-controlled, multicenter study. Dutch Juvenile Chronic Arthritis Study Group. Arthritis Rheum. 1998;41(5):808–16.CrossRefPubMed
37.
38.
go back to reference Zhao L, Au JL, Wientjes MG. Comparison of methods for evaluating drug-drug interaction. Front Biosci. 2010;2:241–9. Zhao L, Au JL, Wientjes MG. Comparison of methods for evaluating drug-drug interaction. Front Biosci. 2010;2:241–9.
39.
go back to reference Chou TC. Theoretical basis, experimental design, and computerized simulation of synergism and antagonism in drug combination studies. Pharmacol Rev. 2006;58(3):621–81.CrossRefPubMed Chou TC. Theoretical basis, experimental design, and computerized simulation of synergism and antagonism in drug combination studies. Pharmacol Rev. 2006;58(3):621–81.CrossRefPubMed
40.
go back to reference Chou TC. Drug combination studies and their synergy quantification using the Chou-Talalay method. Cancer Res. 2010;70(2):440–6.CrossRefPubMed Chou TC. Drug combination studies and their synergy quantification using the Chou-Talalay method. Cancer Res. 2010;70(2):440–6.CrossRefPubMed
41.
go back to reference Berman HM, Battistuz T, Bhat TN, Bluhm WF, Bourne PE, Burkhardt K, et al. The Protein Data Bank. Acta Crystallogr D Biol Crystallogr. 2002;58(Pt 6 No 1):899–907.CrossRefPubMed Berman HM, Battistuz T, Bhat TN, Bluhm WF, Bourne PE, Burkhardt K, et al. The Protein Data Bank. Acta Crystallogr D Biol Crystallogr. 2002;58(Pt 6 No 1):899–907.CrossRefPubMed
42.
go back to reference Vakser IA. Long-distance potentials: an approach to the multiple-minima problem in ligand-receptor interaction. Protein Eng. 1996;9(1):37–41.CrossRefPubMed Vakser IA. Long-distance potentials: an approach to the multiple-minima problem in ligand-receptor interaction. Protein Eng. 1996;9(1):37–41.CrossRefPubMed
43.
go back to reference Katchalski-Katzir E, Shariv I, Eisenstein M, Friesem AA, Aflalo C, Vakser IA. Molecular surface recognition: determination of geometric fit between proteins and their ligands by correlation techniques. Proc Natl Acad Sci U S A. 1992;89(6):2195–9.CrossRefPubMedPubMedCentral Katchalski-Katzir E, Shariv I, Eisenstein M, Friesem AA, Aflalo C, Vakser IA. Molecular surface recognition: determination of geometric fit between proteins and their ligands by correlation techniques. Proc Natl Acad Sci U S A. 1992;89(6):2195–9.CrossRefPubMedPubMedCentral
44.
45.
go back to reference Vakser IA. Low-resolution docking: prediction of complexes for underdetermined structures. Biopolymers. 1996;39(3):455–64.CrossRefPubMed Vakser IA. Low-resolution docking: prediction of complexes for underdetermined structures. Biopolymers. 1996;39(3):455–64.CrossRefPubMed
46.
go back to reference Revet I, Huizenga G, Chan A, Koster J, Volckmann R, van Sluis P, et al. The MSX1 homeobox transcription factor is a downstream target of PHOX2B and activates the Delta-Notch pathway in neuroblastoma. Exp Cell Res. 2008;314(4):707–19.CrossRefPubMed Revet I, Huizenga G, Chan A, Koster J, Volckmann R, van Sluis P, et al. The MSX1 homeobox transcription factor is a downstream target of PHOX2B and activates the Delta-Notch pathway in neuroblastoma. Exp Cell Res. 2008;314(4):707–19.CrossRefPubMed
Metadata
Title
Effect of sulfasalazine on human neuroblastoma: analysis of sepiapterin reductase (SPR) as a new therapeutic target
Authors
Lisette P. Yco
Dirk Geerts
Gabor Mocz
Jan Koster
André S. Bachmann
Publication date
01-12-2015
Publisher
BioMed Central
Published in
BMC Cancer / Issue 1/2015
Electronic ISSN: 1471-2407
DOI
https://doi.org/10.1186/s12885-015-1447-y

Other articles of this Issue 1/2015

BMC Cancer 1/2015 Go to the issue
Webinar | 19-02-2024 | 17:30 (CET)

Keynote webinar | Spotlight on antibody–drug conjugates in cancer

Antibody–drug conjugates (ADCs) are novel agents that have shown promise across multiple tumor types. Explore the current landscape of ADCs in breast and lung cancer with our experts, and gain insights into the mechanism of action, key clinical trials data, existing challenges, and future directions.

Dr. Véronique Diéras
Prof. Fabrice Barlesi
Developed by: Springer Medicine