Do pharmacokinetic polymorphisms explain treatment failure in high-risk patients with neuroblastoma?

Francesco Bellanti, Bertil Kågedal, Oscar Della Pasqua, Francesco Bellanti, Bertil Kågedal, Oscar Della Pasqua

Abstract

Purpose: Neuroblastoma is the most common extracranial solid tumour in childhood. It accounts for 15% of all paediatric oncology deaths. In the last few decades, improvement in treatment outcome for high-risk patients has not occurred, with an overall survival rate <30-40%. Many reasons may account for such a low survival rate. The aim of this review is to evaluate whether pharmacogenetic factors can explain treatment failure in neuroblastoma.

Methods: A literature search based on PubMed's database Medical Subject Headings (MeSH) was performed to retrieve all pertinent publications on current treatment options and new classes of drugs under investigation. One hundred and fifty-eight articles wer reviewed, and relevant data were extracted and summarised.

Results and conclusions: Few of the large number of polymorphisms identified thus far showed an effect on pharmacokinetics that could be considered clinically relevant. Despite their clinical relevance, none of the single nucleotide polymorphisms (SNPs) investigated can explain treatment failure. These findings seem to reflect the clinical context in which anti-tumour drugs are used, i.e. in combination with multimodal therapy. In addition, many pharmacogenetic studies did not assess (differences in) drug exposure, which could contribute to explaining pharmacogenetic associations. Furthermore, it remains unclear whether the significant activity of new drugs on different neuroblastoma cell lines translates into clinical efficacy, irrespective of resistance or myelocytomatosis viral related oncogene, neuroblastoma derived (MYCN) amplification. Elucidation of the clinical role of pharmacogenetic factors in the treatment of neuroblastoma demands an integrated pharmacokinetic-pharmacodynamic approach to the analysis of treatment response data.

References

    1. Maris JM, Hogarty MD, Bagatell R, et al. Neuroblastoma. Lancet. 2007;369:2106–2120.
    1. Maris JM. The biologic basis for neuroblastoma heterogeneity and risk stratification. Curr Opin Pediatr. 2005;17:7–13.
    1. Park JR, Eggert A, Caron H. Neuroblastoma: biology, prognosis, and treatment. Pediatr Clin N Am. 2008;55:97–120.
    1. Schramm A, Schulte JH, Klein-Hitpass L, et al. Prediction of clinical outcome and biological characterization of neuroblastoma by expression profiling. Oncogene. 2005;24:7902–7912.
    1. Mora J, Gerald WL, Cheung NK. Evolving significance of prognostic markers associated with new treatment strategies in neuroblastoma. Cancer Lett. 2003;197:119–124.
    1. Castel V, Grau E, Noguera R, et al. Molecular biology of neuroblastoma. Clin Transl Oncol. 2007;9:478–483.
    1. Fischer M, Spitz R, Oberthur A, et al. Risk estimation of neuroblastoma patients using molecular markers. Klin Pädiatr. 2008;220:137–146.
    1. Krekels EH, van den Anker JN, Baiardi P, et al. Pharmacogenetics and paediatric drug development: issues and consequences to labelling and dosing recommendations. Expert Opin Pharmacother. 2007;8:1787–1799.
    1. Pruthi S, Boughey JC, Brandt KR, et al. A multidisciplinary approach to the management of breast cancer, part 2: therapeutic considerations. Mayo Clin Proc. 2007;82:1131–1140.
    1. Yan L, Hsu K, Beckman RA. Antibody-based therapy for solid tumors. Cancer J. 2008;14:178–183.
    1. Gardiner SJ, Begg EJ. Pharmacogenetics, drug-metabolizing enzymes, and clinical practice. Pharmacol Rev. 2006;58:521–590.
    1. Hedgecoe A, Martin P. The drugs don't work: expectations and the shaping of pharmacogenetics. Soc Stud Sci. 2003;33:327–364.
    1. Groninger E, Meeuwsen-de Boer T, Koopmans P, et al. Vincristine pharmacokinetics and response to vincristine monotherapy in an up-front window study of the Dutch Childhood Leukaemia Study Group (DCLSG) Eur J Cancer. 2005;41:98–103.
    1. Gidding CE, Meeuwsen-de Boer GJ, Koopmans P, et al. Vincristine pharmacokinetics after repetitive dosing in children. Cancer Chemother Pharmacol. 1999;44:203–209.
    1. Frost BM, Lonnerholm G, Koopmans P, et al. Vincristine in childhood leukaemia: no pharmacokinetic rationale for dose reduction in adolescents. Acta Paediatr. 2003;92:551–557.
    1. De Bernardi B, Nicolas B, Boni L, et al. Disseminated neuroblastoma in children older than one year at diagnosis: comparable results with three consecutive high-dose protocols adopted by the Italian Co-Operative Group for Neuroblastoma. J Clin Oncol. 2003;21:1592–1601.
    1. Groninger E, Meeuwsen-de Boar T, Koopmans P, et al. Pharmacokinetics of vincristine monotherapy in childhood acute lymphoblastic leukemia. Pediatr Res. 2002;52:113–118.
    1. Lonnerholm G, Frost BM, Abrahamsson J, et al. Vincristine pharmacokinetics is related to clinical outcome in children with standard risk acute lymphoblastic leukemia. Br J Haematol. 2008;142:616–621.
    1. Kushner BH, Kramer K, LaQuaglia MP, et al. Reduction from seven to five cycles of intensive induction chemotherapy in children with high-risk neuroblastoma. J Clin Oncol. 2004;22:4888–4892.
    1. Coze C, Hartmann O, Michon J, et al. NB87 induction protocol for stage 4 neuroblastoma in children over 1 year of age: a report from the French Society of Pediatric Oncology. J Clin Oncol. 1997;15:3433–3440.
    1. Garaventa A, Luksch R, Biasotti S, et al. A phase II study of topotecan with vincristine and doxorubicin in children with recurrent/refractory neuroblastoma. Cancer. 2003;98:2488–2494.
    1. Valteau-Couanet D, Michon J, Boneu A, et al. Results of induction chemotherapy in children older than 1 year with a stage 4 neuroblastoma treated with the NB 97 French Society of Pediatric Oncology (SFOP) protocol. J Clin Oncol. 2005;23:532–540.
    1. Lanvers-Kaminsky C, Krefeld B, Dinnesen AG, et al. Continuous or repeated prolonged cisplatin infusions in children: a prospective study on ototoxicity, platinum concentrations, and standard serum parameters. Pediatr Blood Cancer. 2006;47:183–193.
    1. Mastrangelo R, Tornesello A, Lasorella A, et al. Optimal use of the 131-I-metaiodobenzylguanidine and cisplatin combination in advanced neuroblastoma. J Neurooncol. 1997;31:153–158.
    1. Souid AK, Dubowy RL, Blaney SM, et al. Phase I clinical and pharmacologic study of weekly cisplatin and irinotecan combined with amifostine for refractory solid tumors. Clin Cancer Res. 2003;9:703–710.
    1. Kushner BH, Budnick A, Kramer K, et al. Ototoxicity from high-dose use of platinum compounds in patients with neuroblastoma. Cancer. 2006;107:417–422.
    1. Kaneko M, Tsuchida Y, Mugishima H, et al. Intensified chemotherapy increases the survival rates in patients with stage 4 neuroblastoma with MYCN amplification. J Pediatr Hematol Oncol. 2002;24:613–621.
    1. Nath CE, Shaw PJ, Montgomery K, et al. Melphalan pharmacokinetics in children with malignant disease: influence of body weight, renal function, carboplatin therapy and total body irradiation. Br J Clin Pharmacol. 2005;59:314–324.
    1. Nath CE, Shaw PJ, Montgomery K, et al. Population pharmacokinetics of melphalan in paediatric blood or marrow transplant recipients. Br J Clin Pharmacol. 2007;64:151–164.
    1. Vassal G, Tranchand B, Valteau-Couanet D, et al. Pharmacodynamics of tandem high-dose melphalan with peripheral blood stem cell transplantation in children with neuroblastoma and medulloblastoma. Bone Marrow Transplant. 2001;27:471–477.
    1. Nieto Y, Vaughan WP. Pharmacokinetics of high-dose chemotherapy. Bone Marrow Transplant. 2004;33:259–269.
    1. Canete A, Gerrard M, Rubie H, 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;27:1014–1019.
    1. Diaz MA, Vicent MG, Madero L. High-dose busulfan/melphalan as conditioning for autologous PBPC transplantation in pediatric patients with solid tumors. Bone Marrow Transplant. 1999;24:1157–1159.
    1. Hartmann O, Valteau-Couanet D, Benhamou E, et al. Stage IV neuroblastoma in patients over 1 year of age at diagnosis: consolidation of poor responders with combined busulfan, cyclophosphamide and melphalan followed by in vitro mafosfamide-purged autologous bone marrow transplantation. Eur J Cancer. 1997;33:2126–2129.
    1. Pritchard J, Cotterill SJ, Germond SM, et al. High dose melphalan in the treatment of advanced neuroblastoma: results of a randomised trial (ENSG-1) by the European Neuroblastoma Study Group. Pediatr Blood Cancer. 2005;44:348–357.
    1. Eksborg S, Soderhall S, Frostvik-Stolt M, et al. Plasma pharmacokinetics of etoposide (VP-16) after i.v. administration to children. Anticancer Drugs. 2000;11:237–241.
    1. Kato Y, Nishimura S, Sakura N, et al. Pharmacokinetics of etoposide with intravenous drug administration in children and adolescents. Pediatr Int. 2003;45:74–79.
    1. Wurthwein G, Klingebiel T, Krumpelmann S, et al. Population pharmacokinetics of high-dose etoposide in children receiving different conditioning regimens. Anticancer Drugs. 2002;13:101–110.
    1. Davidson A, Gowing R, Lowis S, et al. Phase II study of 21 day schedule oral etoposide in children. New agents group of the United Kingdom children's cancer study group (UKCCSG) Eur J Cancer. 1997;33:1816–1822.
    1. Lowis SP, Pearson AD, Newell DR, et al. Etoposide pharmacokinetics in children: the development and prospective validation of a dosing equation. Cancer Res. 1993;53:4881–4889.
    1. Mross K, Reifke J, Bewermeier P, et al. The pharmacokinetics and toxicity of two application schedules with high-dose VP-16 in patients receiving an allogeneic bone marrow transplantation. Ann Oncol. 1996;7:83–88.
    1. Palle J, Britt-Marie F, Goran G, et al. Etoposide pharmacokinetics in children treated for acute myeloid leukemia. Anticancer Drugs. 2006;17:1087–1094.
    1. Kushner BH, Kramer K, Cheung NK. Oral etoposide for refractory and relapsed neuroblastoma. J Clin Oncol. 1999;17:3221–3225.
    1. Simon T, Langler A, Harnischmacher U, et al. Topotecan, cyclophosphamide, and etoposide (TCE) in the treatment of high-risk neuroblastoma. Results of a phase-II trial. J Cancer Res Clin Oncol. 2007;133:653–661.
    1. Simon T, Langler A, Berthold F, et al. Topotecan and etoposide in the treatment of relapsed high-risk neuroblastoma: results of a phase 2 trial. J Pediatr Hematol Oncol. 2007;29:101–106.
    1. Frost BM, Eksborg S, Bjork O, et al. Pharmacokinetics of doxorubicin in children with acute lymphoblastic leukemia: multi-institutional collaborative study. Med Pediatr Oncol. 2002;38:329–337.
    1. Hempel G, Flege S, Wurthwein G, et al. Peak plasma concentrations of doxorubicin in children with acute lymphoblastic leukemia or non-Hodgkin lymphoma. Cancer Chemother Pharmacol. 2002;49:133–141.
    1. Palle J, Frost BM, Peterson C, et al. Doxorubicin pharmacokinetics is correlated to the effect of induction therapy in children with acute myeloid leukemia. Anticancer Drugs. 2006;17:385–392.
    1. Mugishima H, Matsunaga T, Yagi K, et al. Phase I study of irinotecan in pediatric patients with malignant solid tumors. J Pediatr Hematol Oncol. 2002;24:94–100.
    1. Vassal G, Doz F, Frappaz D, et al. A phase I study of irinotecan as a 3-week schedule in children with refractory or recurrent solid tumors. J Clin Oncol. 2003;21:3844–3852.
    1. Rodriguez-Galindo C, Crews KR, Stewart CF, et al. Phase I study of the combination of topotecan and irinotecan in children with refractory solid tumors. Cancer Chemother Pharmacol. 2006;57:15–24.
    1. Bomgaars L, Kerr J, Berg S, et al. A phase I study of irinotecan administered on a weekly schedule in pediatric patients. Pediatr Blood Cancer. 2006;46:50–55.
    1. Levy AS, Meyers PA, Wexler LH, et al. Phase 1 and pharmacokinetic study of concurrent carboplatin and irinotecan in subjects aged 1 to 21 years with refractory solid tumors. Cancer. 2009;115:207–216.
    1. Gore L, Chawla S, Petrilli A, et al. Aprepitant in adolescent patients for prevention of chemotherapy-induced nausea and vomiting: a randomized, double-blind, placebo-controlled study of efficacy and tolerability. Pediatr Blood Cancer. 2009;52:242–247.
    1. Weigel BJ, Blaney SM, Reid JM, et al. A phase I study of 17-allylaminogeldanamycin in relapsed/refractory pediatric patients with solid tumors: a children's oncology group study. Clin Cancer Res. 2007;13:1789–1793.
    1. Bagatell R, Gore L, Egorin MJ, et al. Phase I pharmacokinetic and pharmacodynamic study of 17-N-allylamino-17-demethoxygeldanamycin in pediatric patients with recurrent or refractory solid tumors: a pediatric oncology experimental therapeutics investigators consortium study. Clin Cancer Res. 2007;13:1783–1788.
    1. Bond M, Bernstein ML, Pappo A, et al. A phase II study of imatinib mesylate in children with refractory or relapsed solid tumors: a children's oncology group study. Pediatr Blood Cancer. 2008;50:254–258.
    1. Marangon E, Citterio M, Sala F, et al. Pharmacokinetic profile of imatinib mesylate and N-desmethyl-imatinib (CGP 74588) in children with newly diagnosed Ph + acute leukemias. Cancer Chemother Pharmacol. 2009;63:563–566.
    1. Pollack IF, Jakacki RI, Blaney SM, et al. Phase I trial of imatinib in children with newly diagnosed brainstem and recurrent malignant gliomas: a pediatric brain tumor consortium report. Neuro Oncol. 2007;9:145–160.
    1. Dennison JB, Jones DR, Renbarger JL, et al. Effect of CYP3A5 expression on vincristine metabolism with human liver microsomes. J Pharmacol Exp Ther. 2007;321:553–563.
    1. Plasschaert SL, Groninger E, Boezen M, et al. Influence of functional polymorphisms of the MDR1 gene on vincristine pharmacokinetics in childhood acute lymphoblastic leukemia. Clin Pharmacol Ther. 2004;76:220–229.
    1. Schaefer M, Roots I, Gerloff T. In-vitro transport characteristics discriminate wild-type ABCB1 (MDR1) from ALA893SER and ALA893THR polymorphisms. Pharmacogenet Genomics. 2006;16:855–861.
    1. Filipski KK, Loos WJ, Verweij J, et al. Interaction of Cisplatin with the human organic cation transporter 2. Clin Cancer Res. 2008;14:3875–3880.
    1. Oldenburg J, Fossa SD, Ikdahl T. Genetic variants associated with cisplatin-induced ototoxicity. Pharmacogenomics. 2008;9:1521–1530.
    1. Rabik CA, Dolan ME. Molecular mechanisms of resistance and toxicity associated with platinating agents. Cancer Treat Rev. 2007;33:9–23.
    1. Zhang L, Plon SE, Nuchtern JG, et al. Cyclin D and cisplatin cytotoxicity in primary neuroblastoma cell lines. Anticancer Drugs. 2004;15:883–888.
    1. de Cremoux P, Jourdan-Da-Silva N, Couturier J, et al. Role of chemotherapy resistance genes in outcome of neuroblastoma. Pediatr Blood Cancer. 2007;48:311–317.
    1. Renbarger JL, McCammack KC, Rouse CE, et al. Effect of race on vincristine-associated neurotoxicity in pediatric acute lymphoblastic leukemia patients. Pediatr Blood Cancer. 2008;50:769–771.
    1. Dennison JB, Kulanthaivel P, Barbuch RJ, et al. Selective metabolism of vincristine in vitro by CYP3A5. Drug Metab Dispos. 2006;34:1317–1327.
    1. Woodahl EL, Crouthamel MH, Bui T, et al. (2009) MDR1 (ABCB1) G1199A (Ser400Asn) polymorphism alters transepithelial permeability and sensitivity to anticancer agents. Cancer Chemother Pharmacol
    1. Yang Z, Wu D, Bui T, et al. A novel human multidrug resistance gene MDR1 variant G571A (G191R) modulates cancer drug resistance and efflux transport. J Pharmacol Exp Ther. 2008;327:474–481.
    1. Oldenburg J, Kraggerud SM, Cvancarova M, et al. Cisplatin-induced long-term hearing impairment is associated with specific glutathione s-transferase genotypes in testicular cancer survivors. J Clin Oncol. 2007;25:708–714.
    1. del Amo EM, Urtti A, Yliperttula M. Pharmacokinetic role of L-type amino acid transporters LAT1 and LAT2. Eur J Pharm Sci. 2008;35:161–174.
    1. Kuhne A, Kaiser R, Schirmer M, et al. Genetic polymorphisms in the amino acid transporters LAT1 and LAT2 in relation to the pharmacokinetics and side effects of melphalan. Pharmacogenet Genomics. 2007;17:505–517.
    1. van Schaik RH. CYP450 pharmacogenetics for personalizing cancer therapy. Drug Resist Updat. 2008;11:77–98.
    1. Kishi S, Yang W, Boureau B, et al. Effects of prednisone and genetic polymorphisms on etoposide disposition in children with acute lymphoblastic leukemia. Blood. 2004;103:67–72.
    1. Sohn JW, Lee SY, Lee SJ, et al. MDR1 polymorphisms predict the response to etoposide-cisplatin combination chemotherapy in small cell lung cancer. Jpn J Clin Oncol. 2006;36:137–141.
    1. Lal S, Sandanaraj E, Wong ZW, et al. CBR1 and CBR3 pharmacogenetics and their influence on doxorubicin disposition in Asian breast cancer patients. Cancer Sci. 2008;99:2045–2054.
    1. Gonzalez-Covarrubias V, Zhang J, Kalabus JL, et al. Pharmacogenetics of human carbonyl reductase 1 (CBR1) in livers from black and white donors. Drug Metab Dispos. 2009;37:400–407.
    1. Wojnowski L, Kulle B, Schirmer M, et al. NAD(P)H oxidase and multidrug resistance protein genetic polymorphisms are associated with doxorubicin-induced cardiotoxicity. Circulation. 2005;112:3754–3762.
    1. Bains OS, Karkling MJ, Grigliatti TA, et al. (2009) Two non-synonymous single nucleotide polymorphisms of human carbonyl reductase 1 demonstrate reduced in vitro metabolism of daunorubicin and doxorubicin. Drug Metab Dispos
    1. Crouthamel MH, Wu D, Yang Z, et al. A novel MDR1 G1199T variant alters drug resistance and efflux transport activity of P-glycoprotein in recombinant Hek cells. J Pharm Sci. 2006;95:2767–2777.
    1. Lal S, Wong ZW, Jada SR, et al. Novel SLC22A16 polymorphisms and influence on doxorubicin pharmacokinetics in Asian breast cancer patients. Pharmacogenomics. 2007;8:567–575.
    1. Mossallam GI. Abdel Hamid TM, Samra MA: Glutathione S-transferase GSTM1 and GSTT1 polymorphisms in adult acute myeloid leukemia; its impact on toxicity and response to chemotherapy. J Egypt Natl Canc Inst. 2006;18:264–273.
    1. Smith NF, Figg WD, Sparreboom A. Pharmacogenetics of irinotecan metabolism and transport: an update. Toxicol In Vitro. 2006;20:163–175.
    1. Araki K, Fujita K, Ando Y, et al. Pharmacogenetic impact of polymorphisms in the coding region of the UGT1A1 gene on SN-38 glucuronidation in Japanese patients with cancer. Cancer Sci. 2006;97:1255–1259.
    1. Cote JF, Kirzin S, Kramar A, et al. UGT1A1 polymorphism can predict hematologic toxicity in patients treated with irinotecan. Clin Cancer Res. 2007;13:3269–3275.
    1. Jada SR, Lim R, Wong CI, et al. Role of UGT1A1*6, UGT1A1*28 and ABCG2 c.421 C>A polymorphisms in irinotecan-induced neutropenia in Asian cancer patients. Cancer Sci. 2007;98:1461–1467.
    1. Minami H, Sai K, Saeki M, et al. Irinotecan pharmacokinetics/pharmacodynamics and UGT1A genetic polymorphisms in Japanese: roles of UGT1A1*6 and *28. Pharmacogenet Genomics. 2007;17:497–504.
    1. de Jong FA, Scott-Horton TJ, Kroetz DL, et al. Irinotecan-induced diarrhea: functional significance of the polymorphic ABCC2 transporter protein. Clin Pharmacol Ther. 2007;81:42–49.
    1. Han JY, Lim HS, Yoo YK, et al. Associations of ABCB1, ABCC2, and ABCG2 polymorphisms with irinotecan-pharmacokinetics and clinical outcome in patients with advanced non-small cell lung cancer. Cancer. 2007;110:138–147.
    1. Sai K, Saito Y, Fukushima-Uesaka H, et al. Impact of CYP3A4 haplotypes on irinotecan pharmacokinetics in Japanese cancer patients. Cancer Chemother Pharmacol. 2008;62:529–537.
    1. Bomgaars LR, Bernstein M, Krailo M, et al. Phase II trial of irinotecan in children with refractory solid tumors: a children's oncology group study. J Clin Oncol. 2007;25:4622–4627.
    1. Gaspar N, Sharp SY, Pacey S, et al. Acquired resistance to 17-allylamino-17-demethoxygeldanamycin (17-AAG, tanespimycin) in glioblastoma cells. Cancer Res. 2009;69:1966–1975.
    1. Goetz MP, Toft D, Reid J, et al. Phase I trial of 17-allylamino-17-demethoxygeldanamycin in patients with advanced cancer. J Clin Oncol. 2005;23:1078–1087.
    1. Guo W, Reigan P, Siegel D, et al. Formation of 17-allylamino-demethoxygeldanamycin (17-AAG) hydroquinone by NAD(P)H:quinone oxidoreductase 1: role of 17-AAG hydroquinone in heat shock protein 90 inhibition. Cancer Res. 2005;65:10006–10015.
    1. Kelland LR, Sharp SY, Rogers PM, et al. DT-Diaphorase expression and tumor cell sensitivity to 17-allylamino, 17-demethoxygeldanamycin, an inhibitor of heat shock protein 90. J Natl Cancer Inst. 1999;91:1940–1949.
    1. Gardner ER, Burger H, van Schaik RH, et al. Association of enzyme and transporter genotypes with the pharmacokinetics of imatinib. Clin Pharmacol Ther. 2006;80:192–201.
    1. Gurney H, Wong M, Balleine RL, et al. Imatinib disposition and ABCB1 (MDR1, P-glycoprotein) genotype. Clin Pharmacol Ther. 2007;82:33–40.
    1. Ando Y, Saka H, Ando M, et al. Polymorphisms of UDP-glucuronosyltransferase gene and irinotecan toxicity: a pharmacogenetic analysis. Cancer Res. 2000;60:6921–6926.
    1. Ando Y, Ueoka H, Sugiyama T, et al. Polymorphisms of UDP-glucuronosyltransferase and pharmacokinetics of irinotecan. Ther Drug Monit. 2002;24:111–116.
    1. Carlini LE, Meropol NJ, Bever J, et al. UGT1A7 and UGT1A9 polymorphisms predict response and toxicity in colorectal cancer patients treated with capecitabine/irinotecan. Clin Cancer Res. 2005;11:1226–1236.
    1. Charasson V, Bellott R, Meynard D, et al. Pharmacogenetics of human carboxylesterase 2, an enzyme involved in the activation of irinotecan into SN-38. Clin Pharmacol Ther. 2004;76:528–535.
    1. de Jong FA, Marsh S, Mathijssen RH, et al. ABCG2 pharmacogenetics: ethnic differences in allele frequency and assessment of influence on irinotecan disposition. Clin Cancer Res. 2004;10:5889–5894.
    1. Font A, Sanchez JM, Taron M, et al. Weekly regimen of irinotecan/docetaxel in previously treated non-small cell lung cancer patients and correlation with uridine diphosphate glucuronosyltransferase 1A1 (UGT1A1) polymorphism. Investig New Drugs. 2003;21:435–443.
    1. Gagne JF, Montminy V, Belanger P, et al. Common human UGT1A polymorphisms and the altered metabolism of irinotecan active metabolite 7-ethyl-10-hydroxycamptothecin (SN-38) Mol Pharmacol. 2002;62:608–617.
    1. Innocenti F, Grimsley C, Das S, et al. Haplotype structure of the UDP-glucuronosyltransferase 1A1 promoter in different ethnic groups. Pharmacogenetics. 2002;12:725–733.
    1. Innocenti F, Undevia SD, Iyer L, et al. Genetic variants in the UDP-glucuronosyltransferase 1A1 gene predict the risk of severe neutropenia of irinotecan. J Clin Oncol. 2004;22:1382–1388.
    1. Iyer L, Hall D, Das S, et al. Phenotype-genotype correlation of in vitro SN-38 (active metabolite of irinotecan) and bilirubin glucuronidation in human liver tissue with UGT1A1 promoter polymorphism. Clin Pharmacol Ther. 1999;65:576–582.
    1. Jinno H, Tanaka-Kagawa T, Hanioka N, et al. Glucuronidation of 7-ethyl-10-hydroxycamptothecin (SN-38), an active metabolite of irinotecan (CPT-11), by human UGT1A1 variants, G71R, P229Q, and Y486D. Drug Metab Dispos. 2003;31:108–113.
    1. Jinno H, Saeki M, Saito Y, et al. Functional characterization of human UDP-glucuronosyltransferase 1A9 variant, D256N, found in Japanese cancer patients. J Pharmacol Exp Ther. 2003;306:688–693.
    1. Kaniwa N, Kurose K, Jinno H, et al. Racial variability in haplotype frequencies of UGT1A1 and glucuronidation activity of a novel single nucleotide polymorphism 686 C>T (P229L) found in an African-American. Drug Metab Dispos. 2005;33:458–465.
    1. Marcuello E, Altes A, Menoyo A, et al. UGT1A1 gene variations and irinotecan treatment in patients with metastatic colorectal cancer. Br J Cancer. 2004;91:678–682.
    1. Mathijssen RH, Marsh S, Karlsson MO, et al. Irinotecan pathway genotype analysis to predict pharmacokinetics. Clin Cancer Res. 2003;9:3246–3253.
    1. Mathijssen RH, de Jong FA, van Schaik RH, et al. Prediction of irinotecan pharmacokinetics by use of cytochrome P450 3A4 phenotyping probes. J Natl Cancer Inst. 2004;96:1585–1592.
    1. Paoluzzi L, Singh AS, Price DK, et al. Influence of genetic variants in UGT1A1 and UGT1A9 on the in vivo glucuronidation of SN-38. J Clin Pharmacol. 2004;44:854–860.
    1. Rouits E, Boisdron-Celle M, Dumont A, et al. Relevance of different UGT1A1 polymorphisms in irinotecan-induced toxicity: a molecular and clinical study of 75 patients. Clin Cancer Res. 2004;10:5151–5159.
    1. Sai K, Kaniwa N, Itoda M, et al. Haplotype analysis of ABCB1/MDR1 blocks in a Japanese population reveals genotype-dependent renal clearance of irinotecan. Pharmacogenetics. 2003;13:741–757.
    1. Sai K, Saeki M, Saito Y, et al. UGT1A1 haplotypes associated with reduced glucuronidation and increased serum bilirubin in irinotecan-administered Japanese patients with cancer. Clin Pharmacol Ther. 2004;75:501–515.
    1. Soepenberg O, Dumez H, Verweij J, et al. Phase I pharmacokinetic, food effect, and pharmacogenetic study of oral irinotecan given as semisolid matrix capsules in patients with solid tumors. Clin Cancer Res. 2005;11:1504–1511.
    1. Villeneuve L, Girard H, Fortier LC, et al. Novel functional polymorphisms in the UGT1A7 and UGT1A9 glucuronidating enzymes in Caucasian and African-American subjects and their impact on the metabolism of 7-ethyl-10-hydroxycamptothecin and flavopiridol anticancer drugs. J Pharmacol Exp Ther. 2003;307:117–128.
    1. Wu MH, Chen P, Wu X, et al. Determination and analysis of single nucleotide polymorphisms and haplotype structure of the human carboxylesterase 2 gene. Pharmacogenetics. 2004;14:595–605.
    1. Zhou Q, Sparreboom A, Tan EH, et al. Pharmacogenetic profiling across the irinotecan pathway in Asian patients with cancer. Br J Clin Pharmacol. 2005;59:415–424.
    1. Kang J, Kamal A, Burrows FJ, et al. Inhibition of neuroblastoma xenograft growth by Hsp90 inhibitors. Anticancer Res. 2006;26:1903–1908.
    1. Jayanthan A, Fowler J, Hawkins L, et al. Effects of Hsp90 inhibition in neuroblastoma: analysis of drug sensitivity, target modulation and the influence of bone marrow microenvironment. J Exp Ther Oncol. 2008;7:183–193.
    1. Munoz M, Rosso M (2009) The NK-1 receptor antagonist aprepitant as a broad spectrum antitumor drug. Invest New Drugs
    1. Huang F, Greer A, Hurlburt W, et al. The mechanisms of differential sensitivity to an insulin-like growth factor-1 receptor inhibitor (BMS-536924) and rationale for combining with EGFR/HER2 inhibitors. Cancer Res. 2009;69:161–170.
    1. Wallick CJ, Gamper I, Thorne M, 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;24:5606–5618.
    1. Hogarty MD, Norris MD, Davis K, et al. ODC1 is a critical determinant of MYCN oncogenesis and a therapeutic target in neuroblastoma. Cancer Res. 2008;68:9735–9745.
    1. Koomoa DL, Yco LP, Borsics T, et al. 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;68:9825–9831.
    1. Rounbehler RJ, Li W, Hall MA, et al. Targeting ornithine decarboxylase impairs development of MYCN-amplified neuroblastoma. Cancer Res. 2009;69:547–553.
    1. Beppu K, Jaboine J, Merchant MS, et al. Effect of imatinib mesylate on neuroblastoma tumorigenesis and vascular endothelial growth factor expression. J Natl Cancer Inst. 2004;96:46–55.
    1. Rossler J, Zambrzycka I, Lagodny J, et al. Effect of STI-571 (imatinib mesylate) in combination with retinoic acid and gamma-irradiation on viability of neuroblastoma cells. Biochem Biophys Res Commun. 2006;342:1405–1412.
    1. Palmberg E, Johnsen JI, Paulsson J, et al. Metronomic scheduling of imatinib abrogates clonogenicity of neuroblastoma cells and enhances their susceptibility to selected chemotherapeutic drugs in vitro and in vivo. Int J Cancer. 2009;124:1227–1234.
    1. Vitali R, Cesi V, Nicotra MR, et al. c-Kit is preferentially expressed in MYCN-amplified neuroblastoma and its effect on cell proliferation is inhibited in vitro by STI-571. Int J Cancer. 2003;106:147–152.
    1. Petain A, Kattygnarath D, Azard J, et al. Population pharmacokinetics and pharmacogenetics of imatinib in children and adults. Clin Cancer Res. 2008;14:7102–7109.
    1. Van Maerken T, Speleman F, Vermeulen J, et al. Small-molecule MDM2 antagonists as a new therapy concept for neuroblastoma. Cancer Res. 2006;66:9646–9655.
    1. Ribas J, Boix J, Meijer L. (R)-roscovitine (CYC202, Seliciclib) sensitizes SH-SY5Y neuroblastoma cells to nutlin-3-induced apoptosis. Exp Cell Res. 2006;312:2394–2400.
    1. Becker K, Marchenko ND, Maurice M, et al. Hyperubiquitylation of wild-type p53 contributes to cytoplasmic sequestration in neuroblastoma. Cell Death Differ. 2007;14:1350–1360.
    1. Michaelis M, Rothweiler F, Klassert D, et al. Reversal of P-glycoprotein-mediated multidrug resistance by the murine double minute 2 antagonist nutlin-3. Cancer Res. 2009;69:416–421.
    1. Barbieri E, Mehta P, Chen Z, et al. MDM2 inhibition sensitizes neuroblastoma to chemotherapy-induced apoptotic cell death. Mol Cancer Ther. 2006;5:2358–2365.
    1. Aghi M, Martuza RL. Oncolytic viral therapies - the clinical experience. Oncogene. 2005;24:7802–7816.
    1. Shen Y, Nemunaitis J. Herpes simplex virus 1 (HSV-1) for cancer treatment. Cancer Gene Ther. 2006;13:975–992.
    1. Geoerger B, van Beusechem VW, Opolon P, et al. Expression of p53, or targeting towards EGFR, enhances the oncolytic potency of conditionally replicative adenovirus against neuroblastoma. J Gene Med. 2005;7:584–594.
    1. Toyoda H, Yin J, Mueller S, et al. Oncolytic treatment and cure of neuroblastoma by a novel attenuated poliovirus in a novel poliovirus-susceptible animal model. Cancer Res. 2007;67:2857–2864.
    1. Mahller YY, Williams JP, Baird WH, et al. Neuroblastoma cell lines contain pluripotent tumor initiating cells that are susceptible to a targeted oncolytic virus. PLoS ONE. 2009;4:e4235.
    1. Ino Y, Saeki Y, Fukuhara H, et al. Triple combination of oncolytic herpes simplex virus-1 vectors armed with interleukin-12, interleukin-18, or soluble B7-1 results in enhanced antitumor efficacy. Clin Cancer Res. 2006;12:643–652.
    1. Mahller YY, Vaikunth SS, Ripberger MC, et al. Tissue inhibitor of metalloproteinase-3 via oncolytic herpesvirus inhibits tumor growth and vascular progenitors. Cancer Res. 2008;68:1170–1179.
    1. Parikh NS, Currier MA, Mahller YY, et al. Oncolytic herpes simplex virus mutants are more efficacious than wild-type adenovirus Type 5 for the treatment of high-risk neuroblastomas in preclinical models. Pediatr Blood Cancer. 2005;44:469–478.
    1. Li H, Dutuor A, Tao L, et al. Virotherapy with a type 2 herpes simplex virus-derived oncolytic virus induces potent antitumor immunity against neuroblastoma. Clin Cancer Res. 2007;13:316–322.
    1. Thomas SK, Messam CA, Spengler BA, et al. Nestin is a potential mediator of malignancy in human neuroblastoma cells. J Biol Chem. 2004;279:27994–27999.
    1. Korja M, Finne J, Salmi TT, et al. Chromogenic in situ hybridization-detected hotspot MYCN amplification associates with Ki-67 expression and inversely with nestin expression in neuroblastomas. Mod Pathol. 2005;18:1599–1605.
    1. Breimer DD. PK/PD modelling and beyond: impact on drug development. Pharm Res. 2008;25:2720–2722.
    1. Zhang L, Sinha V, Forgue ST, et al. Model-based drug development: the road to quantitative pharmacology. J Pharmacokinet Pharmacodyn. 2006;33:369–393.
    1. Derendorf H, Lesko LJ, Chaikin P, et al. Pharmacokinetic/pharmacodynamic modeling in drug research and development. J Clin Pharmacol. 2000;40:1399–1418.
    1. Miller R, Ewy W, Corrigan BW, et al. How modeling and simulation have enhanced decision making in new drug development. J Pharmacokinet Pharmacodyn. 2005;32:185–197.
    1. Rajman I. PK/PD modelling and simulations: utility in drug development. Drug Discov Today. 2008;13:341–346.

Source: PubMed

3
Subscribe