Pharmacogenetics in Neuroblastoma: What Can Already Be Clinically Implemented and What Is Coming Next?
Abstract
:1. Introduction
2. Pharmacogenetic Variants for Clinical Implementation
2.1. Drug Regulatory Agencies
2.2. PharmGKB Clinical Annotations
2.3. Clinical Implementation Guidelines
3. Pharmacogenetic Variants under Investigation Regarding NB Therapy
4. Further Consideration for a Perspective on NB PGx Translational Research
4.1. The Role of Ontogeny in NB Pharmacogenetics
4.2. What Else Do We Need to Take into Account? Research Integrating Epigenomics and Metabolomics
5. Conclusions
Author Contributions
Funding
Data Availability Statement
Acknowledgments
Conflicts of Interest
References
- Tolbert, V.P.; Coggins, G.E.; Maris, J.M. Genetic susceptibility to neuroblastoma. Curr. Opin. Genet. Dev. 2017, 42, 81–90. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Hero, B.; Simon, T.; Spitz, R.; Ernestus, K.; Gnekow, A.K.; Scheel-Walter, H.G.; Schwabe, D.; Schilling, F.H.; Benz-Bohm, G.; Berthold, F. Localized infant neuroblastomas often show spontaneous regression: Results of the prospective trials NB95-S and NB97. J. Clin. Oncol. 2008, 26, 1504–1510. [Google Scholar] [CrossRef] [PubMed]
- Park, J.R.; Bagatell, R.; Cohn, S.L.; Pearson, A.D.; Villablanca, J.G.; Berthold, F.; Burchill, S.; Boubaker, A.; McHugh, K.; Nuchtern, J.G.; et al. Revisions to the international neuroblastoma response criteria: A consensus statement from the National Cancer Institute clinical trials planning meeting. J. Clin. Oncol. 2017, 35, 2580–2587. [Google Scholar] [CrossRef]
- Morgenstern, D.A.; Pötschger, U.; Moreno, L.; Papadakis, V.; Owens, C.; Ash, S.; Pasqualini, C.; Luksch, R.; Garaventa, A.; Canete, A.; et al. Risk stratification of high-risk metastatic neuroblastoma: A report from the HR-NBL-1/SIOPEN study. Pediatr. Blood Cancer 2018, 65, e27363. [Google Scholar] [CrossRef] [PubMed]
- Clemens, E.; van der Kooi, A.L.F.; Broer, L.; van Dulmen-den Broeder, E.; Visscher, H.; Kremer, L.; Tissing, W.; Loonen, J.; Ronckers, C.M.; Pluijm, S.M.F.; et al. The influence of genetic variation on late toxicities in childhood cancer survivors: A review. Crit. Rev. Oncol. Hematol. 2018, 126, 154–167. [Google Scholar] [CrossRef]
- Ross, C.J.D.; Visscher, H.; Rod Rassekh, S.; Castro-Pastrana, L.I.; Shereck, E.; Carleton, B.; Hayden, M.R. Pharmacogenomics of serious adverse drug reactions in pediatric oncology. J. Popul. Ther. Clin. Pharmacol. 2011, 18, 134–151. [Google Scholar]
- Conyers, R.; Devaraja, S.; Elliott, D. Systematic review of pharmacogenomics and adverse drug reactions in paediatric oncology patients. Pediatr. Blood Cancer 2018, 65, e26937. [Google Scholar] [CrossRef] [Green Version]
- Whirl-Carrillo, M.; McDonagh, E.M.; Hebert, J.M.; Gong, L.; Sangkuhl, K.; Thorn, C.F.; Altman, R.B.; Klein, T.E. Pharmacogenomics knowledge for personalized medicine. Clin. Pharmacol. Ther. 2012, 92, 414–417. [Google Scholar] [CrossRef]
- U S Food and Drug Administration Home Page. Available online: https://www.fda.gov/ (accessed on 25 June 2021).
- European Medicines Agency. Available online: https://www.ema.europa.eu/ (accessed on 25 June 2021).
- Pharmaceuticals and Medical Devices Agency. Available online: http://www.pmda.go.jp/english/ (accessed on 25 June 2021).
- Drugs and Health Products—Canada. Available online: https://www.canada.ca/en/health-canada/services/drugs-health-products.html (accessed on 25 June 2021).
- Clinical Phramacogenetics Implementation Consortium. Available online: https://cpicpgx.org (accessed on 25 June 2021).
- Dutch Pharmacogenetics Working Group. Available online: https://knmp.nl (accessed on 25 June 2021).
- García-Alfonso, P.; Saiz-Rodríguez, M.; Mondéjar, R.; Salazar Blanco, J.; Páez, D.; Borobia Pérez, A.; Safont Aguilera, M.J.; García García, I.; Colomer, R.; García González, X.; et al. Consensus of experts from the Spanish Pharmacogenetics and Pharmacogenomics Society and the Spanish Society of Medical Oncology for the genotyping of DPYD in cancer patients who are candidates for treatment with fluoropyrimidines. Clin. Transl. Oncol. 2021. (submitted, under review). [Google Scholar]
- Lee, J.W.; Pussegoda, K.; Rassekh, S.R.; Monzon, J.G.; Liu, G.; Hwang, S.; Bhavsar, A.P.; Pritchard, S.; Ross, C.J.; Amstutz, U.; et al. Clinical Practice Recommendations for the Management and Prevention of Cisplatin-Induced Hearing Loss Using Pharmacogenetic Markers. Ther. Drug Monit. 2016, 38, 423–431. [Google Scholar] [CrossRef] [Green Version]
- Aminkeng, F.; Ross, C.J.; Rassekh, S.R.; Hwang, S.; Rieder, M.J.; Bhavsar, A.P.; Smith, A.; Sanatani, S.; Gelmon, K.A.; Bernstein, D.; et al. Recommendations for genetic testing to reduce the incidence of anthracycline-induced cardiotoxicity. Br. J. Clin. Pharmacol. 2016, 82, 683–695. [Google Scholar] [CrossRef] [Green Version]
- Huezo-Diaz Curtis, P.; Uppugunduri, C.R.S.; Muthukumaran, J.; Rezgui, M.A.; Peters, C.; Bader, P.; Duval, M.; Bittencourt, H.; Krajinovic, M.; Ansari, M. Association of CTH variant with sinusoidal obstruction syndrome in children receiving intravenous busulfan and cyclophosphamide before hematopoietic stem cell transplantation. Pharm. J. 2018, 18, 64–69. [Google Scholar] [CrossRef] [Green Version]
- Uppugunduri, C.R.; Rezgui, M.A.; Diaz, P.H.; Tyagi, A.K.; Rousseau, J.; Daali, Y.; Duval, M.; Bittencourt, H.; Krajinovic, M.; Ansari, M. The association of cytochrome P450 genetic polymorphisms with sulfolane formation and the efficacy of a busulfan-based conditioning regimen in pediatric patients undergoing hematopoietic stem cell transplantation. Pharm. J. 2014, 14, 263–271. [Google Scholar] [CrossRef]
- Ten Brink, M.H.; van Bavel, T.; Swen, J.J.; van der Straaten, T.; Bredius, R.G.; Lankester, A.C.; Zwaveling, J.; Guchelaar, H.J. Effect of genetic variants GSTA1 and CYP39A1 and age on busulfan clearance in pediatric patients undergoing hematopoietic stem cell transplantation. Pharmacogenomics 2013, 14, 1683–1690. [Google Scholar] [CrossRef] [Green Version]
- Yee, S.W.; Mefford, J.A.; Singh, N.; Percival, M.E.; Stecula, A.; Yang, K.; Witte, J.S.; Takahashi, A.; Kubo, M.; Matsuda, K.; et al. Impact of polymorphisms in drug pathway genes on disease-free survival in adults with acute myeloid leukemia. J. Hum. Genet. 2013, 58, 353–361. [Google Scholar] [CrossRef]
- Xu, X.; Duan, L.; Zhou, B.; Ma, R.; Zhou, H.; Liu, Z. Genetic polymorphism of copper transporter protein 1 is related to platinum resistance in Chinese non-small cell lung carcinoma patients. Clin. Exp. Pharmacol. Physiol. 2012, 39, 786–792. [Google Scholar] [CrossRef]
- Xu, J.L.; Wang, Z.W.; Hu, L.M.; Yin, Z.Q.; Huang, M.D.; Hu, Z.B.; Shen, H.B.; Shu, Y.Q. Genetic variants in the PI3K/PTEN/AKT/mTOR pathway predict platinum-based chemotherapy response of advanced non-small cell lung cancers in a Chinese population. Asian Pac. J. Cancer Prev. 2012, 13, 2157–2162. [Google Scholar] [CrossRef] [PubMed]
- Xu, X.; Han, L.; Yang, H.; Duan, L.; Zhou, B.; Zhao, Y.; Qu, J.; Ma, R.; Zhou, H.; Liu, Z. The A/G allele of eIF3a rs3740556 predicts platinum-based chemotherapy resistance in lung cancer patients. Lung Cancer 2013, 79, 65–72. [Google Scholar] [CrossRef] [PubMed]
- Patiño-García, A.; Zalacaín, M.; Marrodán, L.; San-Julián, M.; Sierrasesúmaga, L. Methotrexate in pediatric osteosarcoma: Response and toxicity in relation to genetic polymorphisms and dihydrofolate reductase and reduced folate carrier 1 expression. J. Pediatr. 2009, 154, 688–693. [Google Scholar] [CrossRef] [PubMed]
- Pu, X.; Hildebrandt, M.A.; Lu, C.; Lin, J.; Stewart, D.J.; Ye, Y.; Gu, J.; Spitz, M.R.; Wu, X. PI3K/PTEN/AKT/mTOR pathway genetic variation predicts toxicity and distant progression in lung cancer patients receiving platinum-based chemotherapy. Lung Cancer 2011, 71, 82–88. [Google Scholar] [CrossRef] [Green Version]
- Caronia, D.; Patiño-Garcia, A.; Peréz-Martínez, A.; Pita, G.; Moreno, L.T.; Zalacain-Díez, M.; Molina, B.; Colmenero, I.; Sierrasesúmaga, L.; Benítez, J.; et al. Effect of ABCB1 and ABCC3 polymorphisms on osteosarcoma survival after chemotherapy: A pharmacogenetic study. PLoS ONE 2011, 6, e26091. [Google Scholar] [CrossRef] [Green Version]
- Low, S.K.; Kiyotani, K.; Mushiroda, T.; Daigo, Y.; Nakamura, Y.; Zembutsu, H. Association study of genetic polymorphism in ABCC4 with cyclophosphamide-induced adverse drug reactions in breast cancer patients. J. Hum. Genet. 2009, 54, 564–571. [Google Scholar] [CrossRef] [Green Version]
- Shu, W.; Guan, S.; Yang, X.; Liang, L.; Li, J.; Chen, Z.; Zhang, Y.; Chen, L.; Wang, X.; Huang, M. Genetic markers in CYP2C19 and CYP2B6 for prediction of cyclophosphamide’s 4-hydroxylation, efficacy and side effects in Chinese patients with systemic lupus erythematosus. Br. J. Clin. Pharmacol. 2016, 81, 327–340. [Google Scholar] [CrossRef] [Green Version]
- Rocha, V.; Porcher, R.; Fernandes, J.F.; Filion, A.; Bittencourt, H.; Silva, W., Jr.; Vilela, G.; Zanette, D.L.; Ferry, C.; Larghero, J.; et al. Association of drug metabolism gene polymorphisms with toxicities, graft-versus-host disease and survival after HLA-identical sibling hematopoietic stem cell transplantation for patients with leukemia. Leukemia 2009, 23, 545–556. [Google Scholar] [CrossRef]
- Bray, J.; Sludden, J.; Griffin, M.J.; Cole, M.; Verrill, M.; Jamieson, D.; Boddy, A.V. Influence of pharmacogenetics on response and toxicity in breast cancer patients treated with doxorubicin and cyclophosphamide. Br. J. Cancer 2010, 102, 1003–1009. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Su, H.I.; Sammel, M.D.; Velders, L.; Horn, M.; Stankiewicz, C.; Matro, J.; Gracia, C.R.; Green, J.; DeMichele, A. Association of cyclophosphamide drug-metabolizing enzyme polymorphisms and chemotherapy-related ovarian failure in breast cancer survivors. Fertil. Steril. 2010, 94, 645–654. [Google Scholar] [CrossRef] [Green Version]
- Chen, R.L.; Reynolds, C.P.; Seeger, R.C. Neutrophils are cytotoxic and growth-inhibiting for neuroblastoma cells with an anti-GD2 antibody but, without cytotoxicity, can be growth-stimulating. Cancer Immunol. Immunother. 2000, 48, 603–612. [Google Scholar] [CrossRef] [PubMed]
- Greenwood, K.L.; Foster, J.H. The safety of dinutuximab for the treatment of pediatric patients with high-risk neuroblastoma. Expert Opin. Drug Saf. 2018, 17, 1257–1262. [Google Scholar] [CrossRef] [PubMed]
- Lal, S.; Wong, Z.W.; Sandanaraj, E.; Xiang, X.; Siang Ang, P.C.; Lee, E.J.D.; Chowbay, B. Influence of ABCB1 and ABCG2 polymorphisms on doxorubicin disposition in Asian breast cancer patients. Cancer Sci. 2008, 99, 816–823. [Google Scholar] [CrossRef] [PubMed]
- Wojnowski, L.; Kulle, B.; Schirmer, M.; Schlüter, G.; Schmidt, A.; Rosenberger, A.; Vonhof, S.; Bickeböller, H.; Toliat, M.R.; Suk, E.K.; et al. NAD(P)H oxidase and multidrug resistance protein genetic polymorphisms are associated with doxorubicin-induced cardiotoxicity. Circulation 2005, 112, 3754–3762. [Google Scholar] [CrossRef] [Green Version]
- Lal, S.; Sandanaraj, E.; Wong, Z.W.; Ang, P.C.; Wong, N.S.; Lee, E.J.; Chowbay, B. CBR1 and CBR3 pharmacogenetics and their influence on doxorubicin disposition in Asian breast cancer patients. Cancer Sci. 2008, 99, 2045–2054. [Google Scholar] [CrossRef]
- Fan, L.; Goh, B.C.; Wong, C.I.; Sukri, N.; Lim, S.E.; Tan, S.H.; Guo, J.Y.; Lim, R.; Yap, H.L.; Khoo, Y.M.; et al. Genotype of human carbonyl reductase CBR3 correlates with doxorubicin disposition and toxicity. Pharmacogenet. Genom. 2008, 18, 621–631. [Google Scholar] [CrossRef]
- Megías-Vericat, J.E.; Montesinos, P.; Herrero, M.J.; Moscardó, F.; Bosó, V.; Rojas, L.; Martínez-Cuadrón, D.; Rodríguez-Veiga, R.; Sendra, L.; Cervera, J.; et al. Impact of NADPH oxidase functional polymorphisms in acute myeloid leukemia induction chemotherapy. Pharm. J. 2018, 18, 301–307. [Google Scholar] [CrossRef]
- Gelderblom, H.; Blay, J.Y.; Seddon, B.M.; Leahy, M.; Ray-Coquard, I.; Sleijfer, S.; Kerst, J.M.; Rutkowski, P.; Bauer, S.; Ouali, M.; et al. Brostallicin versus doxorubicin as first-line chemotherapy in patients with advanced or metastatic soft tissue sarcoma: An European Organisation for Research and Treatment of Cancer Soft Tissue and Bone Sarcoma Group randomised phase II and pharmacogenetic study. Eur. J. Cancer 2014, 50, 388–396. [Google Scholar] [CrossRef]
- Windsor, R.E.; Strauss, S.J.; Kallis, C.; Wood, N.E.; Whelan, J.S. Germline genetic polymorphisms may influence chemotherapy response and disease outcome in osteosarcoma: A pilot study. Cancer 2012, 118, 1856–1867. [Google Scholar] [CrossRef] [PubMed]
- Relling, M.V.; Nemec, J.; Schuetz, E.G.; Schuetz, J.D.; Gonzalez, F.J.; Korzekwa, K.R. O-demethylation of epipodophyllotoxins is catalyzed by human cytochrome P450 3A4. Mol. Pharmacol. 1994, 45, 352–358. [Google Scholar] [PubMed]
- Zelcer, N.; Saeki, T.; Reid, G.; Beijnen, J.H.; Borst, P. Characterization of drug transport by the human multidrug resistance protein 3 (ABCC3). J. Biol. Chem. 2001, 276, 46400–46407. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Huang, R.S.; Duan, S.; Bleibel, W.K.; Kistner, E.O.; Zhang, W.; Clark, T.A.; Chen, T.X.; Schweitzer, A.C.; Blume, J.E.; Cox, N.J.; et al. A genome-wide approach to identify genetic variants that contribute to etoposide-induced cytotoxicity. Proc. Natl. Acad. Sci. USA 2007, 104, 9758–9763. [Google Scholar] [CrossRef] [Green Version]
- Shen, Y.; Baker, E.; Callen, D.F.; Sutherland, G.R.; Willson, T.A.; Rakar, S.; Gough, N.M. Localization of the human GM-CSF receptor beta chain gene (CSF2RB) to chromosome 22q12.2-->q13.1. Cytogenet. Cell Genet. 1992, 61, 175–177. [Google Scholar] [CrossRef]
- Ladenstein, R.; Pötschger, U.; Valteau-Couanet, D.; Luksch, R.; Castel, V.; Yaniv, I.; Laureys, G.; Brock, P.; Michon, J.M.; Owens, C.; et al. Interleukin 2 with anti-GD2 antibody ch14.18/CHO (dinutuximab beta) in patients with high-risk neuroblastoma (HR-NBL1/SIOPEN): A multicentre, randomised, phase 3 trial. Lancet Oncol. 2018, 19, 1617–1629. [Google Scholar] [CrossRef]
- Yamane, B.H.; Hank, J.A.; Albertini, M.R.; Sondel, P.M. The development of antibody-IL-2 based immunotherapy with hu14.18-IL2 (EMD-273063) in melanoma and neuroblastoma. Expert Opin. Investig. Drugs 2009, 18, 991–1000. [Google Scholar] [CrossRef]
- Shusterman, S.; Naranjo, A.; van Ryn, C.; Hank, J.A.; Parisi, M.T.; Shulkin, B.L.; Servaes, S.; London, W.B.; Shimada, H.; Gan, J.; et al. Antitumor Activity and Tolerability of hu14.18-IL2 with GMCSF and Isotretinoin in Recurrent or Refractory Neuroblastoma: A Children’s Oncology Group phase II study. Clin. Cancer Res. 2019, 25, 6044–6051. [Google Scholar] [CrossRef] [Green Version]
- Harkey, M.A.; Czerwinski, M.; Slattery, J.; Kiem, H.P. Overexpression of glutathione-S-transferase, MGSTII, confers resistance to busulfan and melphalan. Cancer Investig. 2005, 23, 19–25. [Google Scholar] [CrossRef]
- Hodges, L.M.; Markova, S.M.; Chinn, L.W.; Gow, J.M.; Kroetz, D.L.; Klein, T.E.; Altman, R.B. Very important pharmacogene summary: ABCB1 (MDR1, P-glycoprotein). Pharmacogenet. Genom. 2011, 21, 152–161. [Google Scholar] [CrossRef] [Green Version]
- Pu, X.; Wang, L.; Chang, J.Y.; Hildebrandt, M.A.T.; Ye, Y.; Lu, C.; Skinner, H.D.; Niu, N.; Jenkins, G.D.; Komaki, R.; et al. Inflammation-related genetic variants predict toxicity following definitive radiotherapy for lung cancer. Clin. Pharmacol. Ther. 2014, 96, 609–615. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Fachal, L.; Gómez-Caamaño, A.; Barnett, G.C.; Peleteiro, P.; Carballo, A.M.; Calvo-Crespo, P.; Kerns, S.L.; Sánchez-García, M.; Lobato-Busto, R.; Dorling, L.; et al. A three-stage genome-wide association study identifies a susceptibility locus for late radiotherapy toxicity at 2q24.1. Nat. Genet. 2014, 46, 891–894. [Google Scholar] [CrossRef] [PubMed]
- Roberts, J.K.; Birg, A.V.; Lin, T.; Daryani, V.M.; Panetta, J.C.; Broniscer, A.; Robinson, G.W.; Gajjar, A.J.; Stewart, C.F. Population pharmacokinetics of oral topotecan in infants and very young children with brain tumors demonstrates a role of ABCG2 rs4148157 on the absorption rate constant. Drug Metab. Dispos. 2016, 44, 1116–1122. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ceppi, F.; Langlois-Pelletier, C.; Gagné, V.; Rousseau, J.; Ciolino, C.; De Lorenzo, S.; Kevin, K.M.; Cijov, D.; Sallan, S.E.; Silverman, L.B.; et al. Polymorphisms of the vincristine pathway and response to treatment in children with childhood acute lymphoblastic leukemia. Pharmacogenomics 2014, 15, 1105–1116. [Google Scholar] [CrossRef] [Green Version]
- Egbelakin, A.; Ferguson, M.J.; MacGill, E.A.; Lehmann, A.S.; Topletz, A.R.; Quinney, S.K.; Li, L.; McCammack, K.C.; Hall, S.D.; Renbarger, J.L. Increased risk of vincristine neurotoxicity associated with low CYP3A5 expression genotype in children with acute lymphoblastic leukemia. Pediatr. Blood Cancer 2011, 56, 361–367. [Google Scholar] [CrossRef] [Green Version]
- Khabour, O.F.; Alzoubi, K.H.; Firoz, A.S.; Al-Awad, R.M.M. Association between leptin gene rs7799039 polymorphism and lipid profile changes induced by isotretinoin treatment in acne patients. Ther. Clin. Risk Manag. 2018, 14, 949–954. [Google Scholar] [CrossRef] [Green Version]
- Lee, J.J.; Wu, X.; Hildebrandt, M.A.T.; Yang, H.; Khuri, F.R.; Kim, E.; Gu, J.; Ye, Y.; Lotan, R.; Spitz, M.R.; et al. Global assessment of genetic variation influencing response to retinoid chemoprevention in head and neck cancer patients. Cancer Prev. Res. 2011, 4, 185–193. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Olivera, G.G.; Yáñez, Y.; Gargallo, P.; Sendra, L.; Aliño, S.F.; Segura, V.; Sanz, M.Á.; Cañete, A.; Castel, V.; De Mora, J.F.; et al. MTHFR and VDR polymorphisms improve the prognostic value of MYCN status on overall survival in neuroblastoma patients. Int. J. Mol. Sci. 2020, 21, 2714. [Google Scholar] [CrossRef]
- Nordgard, S.H.; Alnæs, G.I.G.; Hihn, B.; Lingjærde, O.C.; Liestøl, K.; Tsalenko, A.; Sørlie, T.; Lønning, P.E.; Børresen-Dale, A.L.; Kristensen, V.N. Pathway based analysis of SNPs with relevance to 5-FU therapy: Relation to intratumoral mRNA expression and survival. Int. J. Cancer 2008, 123, 577–585. [Google Scholar] [CrossRef] [PubMed]
- Findlay, J.M.; Middleton, M.R.; Tomlinson, I. A systematic review and meta-analysis of somatic and germline DNA sequence biomarkers of esophageal cancer survival, therapy response and stage. Ann. Oncol. 2015, 26, 624–644. [Google Scholar] [CrossRef] [PubMed]
- Rubio-San-Simón, A.; Verdú-Amorós, J.; Hladun, R.; Juan-Ribelles, A.; Molero, M.; Guerra-García, P.; Pérez-Martínez, A.; Castañeda, A.; Cañete, A.; de Rojas, T.; et al. Challenges in early phase clinical trials for childhood cancer during the COVID-19 pandemic: A report from the new agents group of the Spanish Society of Paediatric Haematology and Oncology (SEHOP). Clin. Transl. Oncol. 2021, 23, 183–189. [Google Scholar] [CrossRef] [PubMed]
- Pritchard-Jones, K.; Dixon-Woods, M.; Naafs-Wilstra, M.; Valsecchi, M.G. Improving recruitment to clinical trials for cancer in childhood. Lancet Oncol. 2008, 9, 392–399. [Google Scholar] [CrossRef]
- Mlakar, V.; Curtis, P.H.D.; Uppugunduri, C.R.S.; Krajinovic, M.; Ansari, M. Pharmacogenomics in pediatric oncology: Review of gene—Drug associations for clinical use. Int. J. Mol. Sci. 2016, 17, 1502. [Google Scholar] [CrossRef]
- Lin, J.H.; Yamazaki, M. Role of P-glycoprotein in pharmacokinetics: Clinical implications. Clin. Pharmacokinet. 2003, 42, 59–98. [Google Scholar] [CrossRef]
- Haag, C.K.B. Ontogeny and Pharmacogenetics: Determinants of Age-Associated Differences in Drng Clearance during Human Development; Erasmus University Rotterdam: Rotterdam, The Netherlands, 2002; ISBN 9067340057. [Google Scholar]
- Fernandez, E.; Perez, R.; Hernandez, A.; Tejada, P.; Arteta, M.; Ramos, J.T. Factors and mechanisms for pharmacokinetic differences between pediatrie population and adults. Pharmaceutics 2011, 3, 53–72. [Google Scholar] [CrossRef] [Green Version]
- Rao, S.; Uppugunduri, C.; Ansari, M. Commentary: A Myriad Aberrations on Information of Ontogeny of Drug Metabolizing Enzymes in the Pediatric Population: An Obstacle for Personalizing Drug Therapy in the Pediatric Population. Drug Metab. Lett. 2016, 10, 72–74. [Google Scholar] [CrossRef]
- Katsila, T.; Patrinos, G.P. The Implications of Metabotypes for Rationalizing Therapeutics in Infants and Children. Front. Pediatr. 2015, 3, 68. [Google Scholar] [CrossRef] [Green Version]
- Kim, I.W.; Han, N.; Burckart, G.J.; Oh, J.M. Epigenetic changes in gene expression for drug-metabolizing enzymes and transporters. Pharmacotherapy 2014, 34, 140–150. [Google Scholar] [CrossRef] [PubMed]
- Ingelman-Sundberg, M.; Zhong, X.B.; Hankinson, O.; Beedanagari, S.; Yu, A.M.; Peng, L.; Osawa, Y. Potential role of epigenetic mechanisms in the regulation of drug metabolism and transport. Drug Metab. Dispos. 2013, 41, 1725–1731. [Google Scholar] [CrossRef] [PubMed] [Green Version]
Drug | Gene | SNP | Reference Genotype | Risk Genotype | ClinAnnot: Level | Recommendation for the Risk Genotype | Guideline/ Drug Label |
---|---|---|---|---|---|---|---|
Busulfan | −/FDA: Actionable ABL1, BCR (in ALL) | ||||||
Carboplatin | ERCC1 | rs11615 | GG | AA, AG | 2B: E,T | Moderate risk of inefficacy and toxicity | −/− |
GSTP1 | rs1695 | GG | AA, AG | 2A: T | Moderate risk of toxicity | ||
MTHFR | rs1801133 | AA | AG, GG | 2A: E | Moderate risk of inefficacy | ||
XRCC1 | rs25487 | CC | CT, TT | 2B: E | Moderate risk of inefficacy | ||
ERCC1 | rs3212986 | AA | AC, CC | 2B: T | Moderate risk of toxicity | ||
NQO1 | rs1800566 | GG | AA, AG | 2A: E | Moderate risk of inefficacy | ||
Cycloph. | TP53 | rs1042522 | CC | CG, GG | 2B: E,T | Moderate risk of inefficacy and toxicity | −/− |
SOD2 | rs4880 | AA | AG, GG | 2B: E | Moderate risk of inefficacy | ||
GSTP1 | rs1695 | AA,AG | GG | 2A: E,T | Moderate risk of inefficacy and toxicity | ||
Cisplatin | TP53 | rs1042522 | CC | CG, GG | 2B: E,T | Moderate risk of inefficacy and toxicity | CPNDS: TPMT/ FDA: Informative TPMT |
MTHFR | rs1801133 | AA | AG, GG | 2A: E | Moderate risk of inefficacy | ||
GSTP1 | rs1695 | AA | AG, GG | 2B: T | Moderate risk of toxicity | ||
GSTP1 | rs1695 | GG | AG, AA | 2A: E | Moderate risk of inefficacy | ||
XPC | rs2228001 | TT | GT, GG | 1B: T | High risk of toxicity | ||
XRCC1 | rs25487 | CC | CT, TT | 2B: E | Moderate risk of inefficacy | ||
ERCC1 | rs3212986 | AA | AC, CC | 2B: T | Moderate risk of toxicity | ||
ERCC1 | rs11615 | GG | AA, AG | 2B: E,T | Moderate risk of inefficacy and toxicity | ||
Doxorubicin | NQO1 | rs1800566 | GG | AA, AG | 2A: E | Moderate risk of inefficacy | CPNDS: RARG, SLC28A3, UGT1A6/− |
Etoposide | DYNC2H1 | rs716274 | AA | AG, GG | 2B: T | Moderate risk of toxicity | −/− |
Vincristine | CEP72 | rs924607 | CC,CT | TT | 2B: T | Moderate risk of toxicity | −/FDA: Required ABL1, BCR (in ALL) |
Gene | SNP | Reference Genotype | Risk Genotype | CPNDS Recommendations |
---|---|---|---|---|
CISPLATIN | ||||
TPMT | rs1800462 | CC | CG, GG | The consortium recommends testing these variants due to their relation with ototoxicity. +++ Physicians are encouraged to consider the use of otoprotectors if the patient’s tumor type is one for which otoprotectors can be effective without adversely affecting antitumor activity. Alternative treatments may be prescribed when they have demonstrated equal efficacy, manageable and acceptable toxicity, less ototoxicity, and are considered options within the current standards of care. Increase monitoring in high-risk patients. Should be encouraged to receive more frequent follow-up audiometric hearing tests after treatment has ended. |
rs1800460 | CC | CT, TT | ||
rs1142345 | TT | CT, CC | ||
DOXORUBICIN | ||||
RARG | rs2229774 | GG | AG, AA | The consortium recommends testing of these variants due to their relation with cardiotoxicity. + Increase frequency of monitoring, even with serial yearly echocardiographic monitoring and follow-up as recommended by COG guidelines; aggressive screening and management of cardiovascular risk factors, if the patient is considered at high risk. ++ Prescribe dexrazoxane. +++ Use liposomal encapsulated anthracycline preparations; use of continuous inclusion or slower inclusion rates; use of less cardiotoxic types of anthracyclines; use of other cardioprotective agents; prescribe alternative chemotherapy regiments for certain tumor types where alternative regiments have been shown to be equally effective. |
SLC28A3 | rs7853758 | AA, AG | GG | |
UGT1A6 | rs17863783 | GG | GT, TT |
Drug. | Gene | SNP | Hypothetic Effect | References |
---|---|---|---|---|
Busulfan | CTH | rs1021737 | Pediatric patients with the TT genotype (receiving hematopoietic stem cell transplantation) may have an increased risk for sinusoidal obstruction syndrome (SOS) when treated with cyclophosphamide and busulfan as compared to patients with the GG or GT genotypes. | Huezo-Diaz Curtis P., 2018 (Ref. [18]) |
CYP2C9 | rs1799853 | Pediatric patients with the CT and TT genotypes (undergoing hematopoietic stem cell transplant) may have decreased metabolism of busulfan as compared to patients with the CC genotype. | Uppugunduri CR., 2014 (Ref. [19]) | |
CYP2C19 | rs12248560 | Pediatric patients with the CC genotype (undergoing transplantation) may have decreased metabolism of busulfan as compared to patients with the CT or TT genotypes. | ||
GSTA1 | rs3957357 | Pediatric patients with the AG and GG genotypes (who are undergoing hematopoietic stem cell transplantation) may have decreased clearance of busulfan as compared to patients with the AA genotype. | Ten Brink MH., 2013 (Ref. [20]) | |
GSTM1 | rs3754446 | Patients with the AA and AC genotypes (and acute myeloid leukemia) may have decreased clearance of busulfan as compared to patients with the CC genotype. | Yee SW., 2013 (Ref. [21]) | |
Carboplatin Cisplatin | AKT1 | rs2494752 | Patients with the GG and AA genotypes who are treated with carboplatin or cisplatin may have decreased risk of progression of the disease as compared to patients with the AG genotype. | Xu X., 2012 (Ref. [22]) |
rs1130214 | Patients with the CC genotype (and lung cancer) who are treated with carboplatin or cisplatin may have a higher risk of distant disease progression as compared to patients with the AC or AA genotype. | Xu JL., 2012; (Ref. [23]) | ||
EIF3A | rs3740556 | Patients with the GG genotype (and lung cancer) may have a poorer response when treated with platinum-based chemotherapy as compared to patients with the AA or AG genotype. | Xu X., 2013 (Ref. [24]) | |
MTR | rs1805087 | Pediatric patients with the GG genotype and cancer may have an increased risk for drug toxicity and an increased response to treatment with cisplatin or carboplatin as compared to patients with the AA or AG genotypes. | Patiño A., 2009 (Ref. [25]) | |
PIK3CA | rs2699887 | Patients with the CC genotype (and non-small-cell lung cancer) may have an increased risk for toxicity when treated with platinum-based chemotherapy as compared to patients with the TT genotype. | Pu X., 2011 (Ref. [26]) | |
PTEN | rs2299939 | Patients with the AA genotype (and non-small-cell lung cancer) may have an increased risk for toxicity when treated with platinum-based chemotherapy as compared to patients with the AC or CC genotype. | ||
SLC31A1 | rs7851395 | Patients with the AA genotype may have increased overall survival when treated with carboplatin or cisplatin (in people with Non-Small-Cell Lung Carcinoma) as compared to patients with genotypes AG or GG. | Xu X., 2012 (Ref. [22]) | |
Unknown (Intronic) | rs2498804 | Patients with the CC genotype (and non-small-cell lung cancer) may have an increased risk of distant disease progression when treated with platinum-based chemotherapy as compared to patients with the AA or AC genotypes. | Pu X., 2011 (Ref. [26]) | |
Cyclophosph. | ABCB1 | rs1045642 | Allele G is associated with an increased risk of death when treated with cyclophosphamide in combination with other drugs, (in patients with osteosarcoma) as compared to allele A. | Caronia D., 2011 (Ref. [27]) |
ABCC4 | rs9561778 | Patients with the TT or GT genotypes (and breast cancer) who are treated with cyclophosphamide may have an increased risk of neutropenia/leukopenia and gastrointestinal toxicity, as compared to patients with the GG genotype. | Low SK., 2009 (Ref. [28]) | |
CYP2B6 | rs7254579 | Patients (with lupus) and the CC or CT genotypes may have decreased metabolism of cyclophosphamide, resulting in decreased concentrations of active cyclophosphamide metabolite as compared to patients with TT genotype. | Su W., 2016 (Ref. [29]) | |
rs4802101 | Patients with the CC genotype may have decreased metabolism of cyclophosphamide, resulting in decreased concentrations of active cyclophosphamide metabolites and decreased risk of gastrointestinal toxicity, or leukopenia, as compared to patients with the CT or TT genotypes. | |||
rs8192709 | (Recipients of HLA-identical hematopoietic stem cell transplantation) with the TT or CT genotypes (and leukemia) may have an increased risk for hemorrhagic cystitis when treated with cyclophosphamide compared to patients with the CC genotype. | Rocha V., 2009 (Ref. [30]) | ||
rs2279343 | Patients with the GG or AG genotypes (who have received a hematopoietic stem cell transplant) and are treated with cyclophosphamide may have an increased risk for oral mucositis as compared to patients with the AA genotype. | |||
rs3745274 | (Leukemia patients who are) recipients (of HLA-identical hematopoietic stem cell transplantation) from donors with the GG genotype may have an increased risk of developing veno-occlusive disease of the liver when treated with cyclophosphamide as compared to donor cells with the GT or TT genotype.Patients with the GG or GT genotypes (and Breast Cancer) who are treated with cyclophosphamide and doxorubicin may be more likely to require a reduction in dose as compared to patients with the TT genotype. | Bray J., 2010 (Ref. [31]) | ||
CYP2C19 | rs4244285 | Patients with the GG genotype (and Systemic Lupus Erythematosus) who are treated with cyclophosphamide may have increased metabolism of cyclophosphamide, leading to higher concentrations of the active metabolite and an increased risk of toxicity (ovarian, gastrointestinal, or hematological) as compared to patients with the AA and AG genotype. | Su W., 2016 (Ref. [29]) | |
CYP3A4 | rs2740574 | Premenopausal patients with the TT genotype (and breast cancer) who are treated with cyclophosphamide may have a shorter period before chemotherapy-induced ovarian failure compared to patients with the CC or CT genotype. | Su HI., 2010 (Ref. [32]) | |
Dinutuximab | SNPs reducing or impairing the expression of GD2 could impede Dinutuximab efficacy. | Chen RL., 2000 Greenwood KL., 2018 (Refs. [33,34]) | ||
Doxorubicin | ABCB1 | rs1045642 rs2032582 rs1128503 | Patients harboring the CC-GG-CC genotypes had significantly lower peak plasma concentrations of doxorubicinol compared to patients who had TT-TT-TT genotypes. | Lal S., 2008 (Ref. [35]) |
ABCC1 | rs45511401 | Patients with the TT or GT genotypes (and non-Hodgkin lymphoma) who are treated with doxorubicin may have an increased risk for cardiotoxicity as compared to patients with the GG genotype. | Wojnowski L., 2005 (Ref. [36]) | |
ABCC2 | rs8187710 | Patients with the AA or AG genotypes (and non-Hodgkin lymphoma) who are treated with doxorubicin may have an increased risk of cardiotoxicity as compared to patients with the GG genotype. | ||
rs17222723 | Patients with the AA or AT genotypes (and non-Hodgkin lymphoma) who are treated with doxorubicin may have an increased risk of cardiotoxicity as compared to patients with the TT genotype. | |||
CBR1 | rs9024 | Patients with the GG genotype may have increased clearance of doxorubicin and decreased exposure to doxorubicin compared to patients with the AG genotype. | Lal S., 2008 (Ref. [37]) | |
CBR3 | rs8133052 | Patients with the AA genotype (and breast cancer) who are treated with doxorubicin may have decreased metabolism of doxorubicin and may have greater tumor reduction, but may also have increased severity of neutropenia as compared to patients with the GG genotype. | Fan L., 2008 (Ref. [38]) | |
CYBA | rs4673 | Cancer patients with the AA or AG genotypes who are treated with doxorubicin may have an increased risk for cardiotoxicity as compared to patients with the GG genotype. | Megías-Vericat JE., 2018 and Wojnowski L., 2005 (Refs. [36,39]) | |
GSTA1 | rs3957357 | Patients with the GG and AG genotypes (and soft tissue sarcoma) may have a shorter progression-free survival time when treated with doxorubicin as compared to patients with the AA genotype. | Gelderblom H., 2014 (Ref. [40]) | |
GSTP1 | rs1695 | Patients with osteosarcoma and the GG or AG genotypes may be at an increased risk of developing leukopenia when treated with doxorubicin as compared to patients with the AA genotype. | Windsor RE., 2012 (Ref. [41]) | |
RAC2 | rs13058338 | Cancer patients with the TT or AT genotypes who are treated with doxorubicin or idarubicin may have an increased risk for drug toxicity as compared to patients with the AA genotype. | Megías-Vericat JE., 2018 and Wojnowski L., 2005 (Refs. [36,39]) | |
Etoposide | All these relevant pharmacogenes ABCB1, ABCC3, CYP3A4, CYP3A5, GSTP1, UGT1A1, are known to be relevant in etoposide pharmacokinetics, thus studies validating their main SNPs regarding the drug’s toxicity and efficacy are needed. | Relling M.V., 1994; Zelcer N., 2001; Huang RS.,2007 (Refs. [42,43,44]) | ||
GM-CSF | Human GM-CSF receptor beta chain gene could be a good candidate to investigate the role of SNPs that could interfere the activation of the receptor by the drug. | Shen Y., 1992 (Ref. [45]) | ||
Interleukin2 | SNPs in the gene coding for IL2 receptor could be very informative to assess the response to this treatment. | Ladenstein R., 2018; YamaneB.H., 2009; Shusterman S., 2019 (Refs. [46,47,48]) | ||
Melphalan | ABCB1 and GSTP1 are relevant pharmacogenes that seem to be involved in melphalan pharmacokinetics. Exploring their main SNPs could be of interest. | Karkey M.A., 2005 and Hodges L.M., 2011 (Refs. [49,50]) | ||
Radiotherapy | CDK1 | rs10711 | Patients with the GG or GT genotypes may have increased risk of pneumonitis when treated with radiotherapy (lung cancer) as compared to patients with genotype TT. | Pu X., 2014 (Ref. [51]) |
PRKCE | rs11125035 | Patients with the AA genotype may have increased risk of esophagitis when treated with radiotherapy as compared to patients with genotype TT or AT. | Pu X., 2014 (Ref. [51]) | |
TANC1 | rs10497203 rs264631 rs264651 rs6432512 rs264588 rs264663 rs7582141 | Patients with the CC or AC/CG or GG/GG/CT or TT/AA or AC/CT or TT/GT or TT genotypes, respectively in the referred SNPs (left column) (and prostate cancer) who are treated with radiotherapy may have an increased risk of late stage toxicity as compared to patients with the other possible genotypes. | Fachal L., 2014 (Ref. [52]) | |
Topotecan | ABCG2 | rs4148157 | Pediatric patients with the GG genotype (and brain tumors) may have decreased absorption and lower concentrations of topotecan compared to patients with the AA and AG genotypes. | Roberts J.K.,2016 (Ref. [53]) |
Vincristine | ABCB1 | rs1045642 | Pediatric patients with the AA or AG genotypes (and acute lymphoblastic leukemia) who are treated with vincristine may have a decreased likelihood of event-free survival as compared to patients with the GG genotype. | Ceppi F.,2014 (Ref. [54]) |
rs4728709 | Pediatric patients with the GG genotype (and acute lymphoblastic leukemia) who are treated with vincristine may have an increased risk of grade 1–2 neurotoxicity as compared to patients with the AA or AG genotypes. | Ceppi F.,2014 (Ref. [54]) | ||
CYP3A5 seems to be the main metabolizing enzyme for vincristine, thus analyzing its main SNPs could be of interest | Egbelakin A.,2011 (Ref. [55]) | |||
Isotretinoin | LEP | rs7799039 | These SNPs seem to correlate with the lipid disorders caused by the drug. | Khabour O.F.,2018 (Ref. [56]) |
Other SNPs in relevant genes related to the mechanism of action of the drug could shed light for decreasing adverse side effects and increasing efficacy: RXRA, JAK2, CDC25C | Lee J.J,2011 (Ref. [57]) |
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations. |
© 2021 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (https://creativecommons.org/licenses/by/4.0/).
Share and Cite
Olivera, G.G.; Urtasun, A.; Sendra, L.; Aliño, S.F.; Yáñez, Y.; Segura, V.; Gargallo, P.; Berlanga, P.; Castel, V.; Cañete, A.; et al. Pharmacogenetics in Neuroblastoma: What Can Already Be Clinically Implemented and What Is Coming Next? Int. J. Mol. Sci. 2021, 22, 9815. https://doi.org/10.3390/ijms22189815
Olivera GG, Urtasun A, Sendra L, Aliño SF, Yáñez Y, Segura V, Gargallo P, Berlanga P, Castel V, Cañete A, et al. Pharmacogenetics in Neuroblastoma: What Can Already Be Clinically Implemented and What Is Coming Next? International Journal of Molecular Sciences. 2021; 22(18):9815. https://doi.org/10.3390/ijms22189815
Chicago/Turabian StyleOlivera, Gladys G., Andrea Urtasun, Luis Sendra, Salvador F. Aliño, Yania Yáñez, Vanessa Segura, Pablo Gargallo, Pablo Berlanga, Victoria Castel, Adela Cañete, and et al. 2021. "Pharmacogenetics in Neuroblastoma: What Can Already Be Clinically Implemented and What Is Coming Next?" International Journal of Molecular Sciences 22, no. 18: 9815. https://doi.org/10.3390/ijms22189815
APA StyleOlivera, G. G., Urtasun, A., Sendra, L., Aliño, S. F., Yáñez, Y., Segura, V., Gargallo, P., Berlanga, P., Castel, V., Cañete, A., & Herrero, M. J. (2021). Pharmacogenetics in Neuroblastoma: What Can Already Be Clinically Implemented and What Is Coming Next? International Journal of Molecular Sciences, 22(18), 9815. https://doi.org/10.3390/ijms22189815