Novel and replicated clinical and genetic risk factors for toxicity from high‐dose methotrexate in pediatric acute lymphoblastic leukemia

Methotrexate (MTX) is a key component of treatment for high‐risk pediatric acute lymphoblastic leukemia (ALL) but may cause acute kidney injury and prolonged hospitalization due to delayed clearance. The purpose of this study is to identify clinical and genetic factors that may predict which children are at risk for creatinine increase and prolonged MTX clearance.

[1]  F. Wen,et al.  Risk prediction for delayed clearance of high-dose methotrexate in pediatric hematological malignancies by machine learning , 2021, International Journal of Hematology.

[2]  M. Scheurer,et al.  Novel risk factors for glucarpidase use in pediatric acute lymphoblastic leukemia: Hispanic ethnicity, age, and the ABCC4 gene , 2021, Pediatric blood & cancer.

[3]  M. Kay Tidy Data and 'Geoms' for Bayesian Models [R package tidybayes version 2.3.1] , 2020 .

[4]  M. Scheurer,et al.  Hispanic ethnicity is associated with prolonged clearance of high dose methotrexate and severe nephrotoxicity in children and adolescents with acute lymphoblastic leukemia , 2020, Leukemia & lymphoma.

[5]  A. Gafter-Gvili,et al.  Risk factors for high‐dose methotrexate associated acute kidney injury in patients with hematological malignancies , 2020, Hematological oncology.

[6]  R. Viney,et al.  Financial toxicity of childhood cancer and changes to parents’ employment after treatment completion , 2020, Pediatric blood & cancer.

[7]  A. Vinks,et al.  MTXPK.org: A Clinical Decision Support Tool Evaluating High‐Dose Methotrexate Pharmacokinetics to Inform Post‐Infusion Care and Use of Glucarpidase , 2020, medRxiv.

[8]  Matthew Kay tidybayes: Tidy Data and Geoms for Bayesian Models , 2020 .

[9]  A. Siryk,et al.  Impact of Ethnicity on Toxicities Associated with Dose Escalating Methotrexate in Pediatric Patients with Acute Lymphoblastic Leukemia , 2019, Blood.

[10]  S. Medellín-Garibay,et al.  Population pharmacokinetics of methotrexate in Mexican pediatric patients with acute lymphoblastic leukemia , 2019, Cancer Chemotherapy and Pharmacology.

[11]  G. Michel,et al.  The impact of childhood cancer on parents' socio‐economic situation—A systematic review , 2019, Psycho-oncology.

[12]  S. Hilsenbeck,et al.  A prospective study of a simple algorithm to individually dose high-dose methotrexate for children with leukemia at risk for methotrexate toxicities , 2018, Cancer Chemotherapy and Pharmacology.

[13]  Austin L. Brown,et al.  Disparities in Neurotoxicity Risk and Outcomes among Pediatric Acute Lymphoblastic Leukemia Patients , 2018, Clinical Cancer Research.

[14]  Tao-tao Liu,et al.  Identification of Risk Factors in High-Dose Methotrexate-Induced Acute Kidney Injury in Childhood Acute Lymphoblastic Leukemia , 2018, Chemotherapy.

[15]  Paul-Christian Bürkner,et al.  brms: An R Package for Bayesian Multilevel Models Using Stan , 2017 .

[16]  K. Schmiegelow,et al.  Delayed elimination of high‐dose methotrexate and use of carboxypeptidase G2 in pediatric patients during treatment for acute lymphoblastic leukemia , 2017, Pediatric blood & cancer.

[17]  Aki Vehtari,et al.  Practical Bayesian model evaluation using leave-one-out cross-validation and WAIC , 2015, Statistics and Computing.

[18]  S. Cole,et al.  Genetic variation underlying renal uric acid excretion in Hispanic children: the Viva La Familia Study , 2017, BMC Medical Genetics.

[19]  S. Devlin,et al.  Hypoalbuminemia is significantly associated with increased clearance time of high dose methotrexate in patients being treated for lymphoma or leukemia , 2016, Annals of Hematology.

[20]  C. Pui,et al.  Preventing and Managing Toxicities of High-Dose Methotrexate , 2016, The oncologist.

[21]  G. Phillips,et al.  Evaluation of incidence and risk factors for high-dose methotrexate-induced nephrotoxicity , 2016, Journal of oncology pharmacy practice : official publication of the International Society of Oncology Pharmacy Practitioners.

[22]  R Core Team,et al.  R: A language and environment for statistical computing. , 2014 .

[23]  L. Almasy,et al.  Genome-wide association analysis confirms and extends the association of SLC2A9 with serum uric acid levels to Mexican Americans , 2013, Front. Genet..

[24]  M. Loh,et al.  Genome-wide study of methotrexate clearance replicates SLCO1B1. , 2013, Blood.

[25]  G. Kovács,et al.  Comparison of pharmacokinetics and toxicity after high-dose methotrexate treatments in children with acute lymphoblastic leukemia , 2013, Anti-cancer drugs.

[26]  E. Lopez-Lopez,et al.  Polymorphisms in the methotrexate transport pathway: a new tool for MTX plasma level prediction in pediatric acute lymphoblastic leukemia , 2013, Pharmacogenetics and genomics.

[27]  R. Altman,et al.  Pharmacogenomics Knowledge for Personalized Medicine , 2012, Clinical pharmacology and therapeutics.

[28]  Xiaomin Lu,et al.  Plasma Methotrexate, Red Blood Cell Methotrexate, and Red Blood Cell Folate Values and Outcome in Children With Precursor B-acute Lymphoblastic Leukemia: A Report From the Children's Oncology Group , 2012, Journal of pediatric hematology/oncology.

[29]  G. Rosner,et al.  Rare versus common variants in pharmacogenetics: SLCO1B1 variation and methotrexate disposition. , 2012, Genome research.

[30]  Joseph O. Deasy,et al.  Common Terminology Criteria for Adverse Events (CTCAE) v4.0 Based Hybrid Patient and Physician Questionnaire for Head and Neck (HN) Radiotherapy Symptom Reporting , 2011 .

[31]  Russ B Altman,et al.  PharmGKB summary: methotrexate pathway. , 2011, Pharmacogenetics and genomics.

[32]  Cedric E. Ginestet ggplot2: Elegant Graphics for Data Analysis , 2011 .

[33]  R. Altman,et al.  SLC19A1 pharmacogenomics summary , 2010, Pharmacogenetics and genomics.

[34]  I. Christensen,et al.  The association of reduced folate carrier 80G>A polymorphism to outcome in childhood acute lymphoblastic leukemia interacts with chromosome 21 copy number. , 2010, Blood.

[35]  R. Mei,et al.  A genomewide admixture mapping panel for Hispanic/Latino populations. , 2007, American journal of human genetics.

[36]  E. Cook,et al.  Ancestry and pharmacogenetics of antileukemic drug toxicity. , 2007, Blood.

[37]  Ching-Hon Pui,et al.  Body mass index does not influence pharmacokinetics or outcome of treatment in children with acute lymphoblastic leukemia. , 2006, Blood.

[38]  P. Adamson,et al.  Understanding and managing methotrexate nephrotoxicity. , 2006, The oncologist.

[39]  J. Lukas,et al.  Population Pharmacokinetics of High-Dose Methotrexate in Children with Acute Lymphoblastic Leukaemia , 2006, Clinical pharmacokinetics.

[40]  J. Downing,et al.  Folate pathway gene expression differs in subtypes of acute lymphoblastic leukemia and influences methotrexate pharmacodynamics. , 2005, The Journal of clinical investigation.

[41]  K. Ness,et al.  Survival variability by race and ethnicity in childhood acute lymphoblastic leukemia. , 2003, JAMA.

[42]  M. Kuwano,et al.  Analysis of methotrexate and folate transport by multidrug resistance protein 4 (ABCC4): MRP4 is a component of the methotrexate efflux system. , 2002, Cancer research.

[43]  D. Murry,et al.  Impact of nutrition on pharmacokinetics of anti‐neoplastic agents , 1998, International journal of cancer. Supplement = Journal international du cancer. Supplement.

[44]  J. Daly,et al.  Effects of nutritional depletion and repletion on plasma methotrexate pharmacokinetics , 1984, Cancer.

[45]  V. Grossie,et al.  Effect of malnutrition on methotrexate toxicity and tissue levels of dihydrofolate reductase in the rat. , 1982, Cancer treatment reports.