Methotrexate is a folate analog that is used in the treatment of cancers (e.g. acute lymphoblastic leukemia, non-Hodgkin lymphoma, osteosarcoma, and colon cancer) and autoimmune diseases (e.g. rheumatoid arthritis, Crohn’s disease, and psoriasis). In the treatment of autoimmune diseases, methotrexate is usually administrated orally or subcutaneously, whereas in the cancer treatment, it can be given orally, intramuscularly, as intrathecal injections, or as intravenous infusions (up to 12 g/m2) [1-3]. The pharmacokinetics and pharmacodynamics of methotrexate show large interpatient variability regardless of the route of administration or disease being treated [4-6]. The goal of this study is to provide an introduction to methotrexate pharmacogenomics, showing the candidate genes in the PharmGKB methotrexate pathway (Fig. 1), important variants (Tables 1 and 2), discussing key knowledge, and pointing to more in-depth resources.
Table 1. Well-studied genes and variants involved in methotrexate pharmacogenomics.
Gene | Variant (rs#) | Phenotype (population) (PMID) | No association (population) (PMID) |
---|---|---|---|
MTHFR | 677C > T (rs1801133) |
C toxicity/adverse events (pediatric ALL, CML) (16870553 [7]) (16920564 [8]) T toxicity/adverse events (pediatric ALL; NHL; CML; RA, Black; PsA; RA, White; RA, Asian; JIA, White; pediatric ALL, Asian) (12453860 [9], 17488658 [10], 11418485 [11], 18381794 [12], 20472929 [13], 17512587 [14], 16501586 [15], 15051775 [16], 11710708 [17], 20863444 [18], 20595278 [19], 18458567 [20]) C efficacy (pediatric ALL; NHL; CML) (14647408 [21]; 15781665 [22], 17488658 [10], 17512587 [14], 15385937 [23]) |
Toxicity (pediatric ALL; pediatric ALL, Asian; RA, Black, White; RA, Hispanic; Psoriasis; NHL) (12915598 [24], 16439441 [25], 19016697 [26], 20514079 [27], 18368069 [28], 15713801 [29], 16462575 [30], 15781665 [22], 16463153 [31]) Efficacy (psoriasis; NHL; pediatric ALL) (19016697 [26], 16463153 [31], 19827168 [32], 16019535 [33]) |
MTHFR | 1298A>C (rs1801131) |
A toxicity (RA, White; pediatric ALL)(16439441 [25], 18368069 [28], 17323057 [34]) C toxicity/adverse events (NHL; CML; RA; RA Hispanic; CML) (17488658 [10], 16501586 [15], 16572443 [35], 20514079 [27], 16920564 [8]) A efficacy (RA, White; CML, White) (16572443 [35], 15569990 [36]) C efficacy (RA, Asian; pediatric ALL; CML) (20863444 [18], 14647408 [21], 15385937 [23]) |
Toxicity (RA, Black; RA, White, Black; Psoriasis, PsA, pediatric ALL) (16439441 [25], 18381794 [12], 19016697 [26], 20472929 [13], 16870553 [7], 15713801 [29], 15781665 [22], 17512587 [14]) Efficacy (Psoriasis; pediatric ALL) (19016697 [26], 15781665 [22], 17512587 [14], 16019535 [33]) |
SLC19A1 | 80G>A (rs1051266) |
A toxicity/adverse events (pediatric ALL; ALL; pediatric ALL, Asian) (12411325 [37], (17264302 [38], 17180579 [39], 20335220 [40]) G toxicity (pediatric ALL, Asian) (16462575 [30]) A efficacy (RA; RA, Asian; ALL) (17325736 [41], 18322994 [42], 20335220 [40], 19827168 [32]) G efficacy (pediatric ALL) (12411325 [37]) |
Toxicity (pediatric ALL, PsA) (12915598 [24], 20472929 [13], 18368069 [28], 16870553 [7], 15713801 [29]) |
TYMS | 28 bp repeat (rs34743033) |
3R/2R toxicity (CML) (16920564 [8]) 3R/3R reduced efficiacy (pediatric ALL) (11937185 [43], 12972956 [44], 15713801 [29], 14647408 [21], 18096764 [45]) |
Efficacy (RA, White) (20386493 [46], 12651279 [47]) Toxicity (pediatric ALL, CML) (16870553 [7], 16501586 [15]) |
ALL, acute lymphoblastic leukemia; CML, chronic myelogenous leukemia; NHL, non-Hodgkin’s lymphoma; PMID, PubMed identifier; PsA psoriatic arthritis; RA, rheumatoid arthritis.
Table 2.
Gene | Studied variants (rs) | PMIDs |
---|---|---|
ABCB1 | 3435C>T (rs1045642), 1236 C>T (rs1128503) |
19093297 [48], 20386493a[46], 18381794 [12], 20136356 [49] |
ABCC1 | Arg723Gln (rs4148356), rs3784862, Ala1337Thr (rs28364006), 4002 G>A (rs2230671), IVS 14+115C>T, IVS 18-30C>G (rs2074087) |
16041243 [50], 18381794a[12], 20386493 [46], 18256692 [51] |
ABCC2 | 1249 G>A (rs2273697), IVS23+56C>T (rs4148396) |
18381794 [12], 20386493 [46] |
ABCC3 | 11585759b[52], 19996279b[53] | |
ABCC4 | 934A>C (rs2274407), −1393T>C |
19515727 [54] |
ABCG2 | 914C>A (rs2231142), rs17731538 |
20386493 [46], 12208758b[55], 12874005b[56] |
ADA | Val178Val (rs244076), IVS 2 C/G (rs1799880) |
19902562 [57] |
ADORA1 | rs16851020, rs3766553, rs3766554, rs17530497, rs4423076, rs1845466, rs3753475, rs10800901, rs10920574, rs7549561, rs6427993, rs6701725, rs12123037, rs1494487, rs3898276 |
9214432b[58], 18256692a[51] |
ADORA2A | 1976T>C (rs5751876), rs5760410, rs2298383, rs3761422, rs2267076, rs2236624, rs9624472, rs4822489 |
18539621 [59], 19902562 [57] |
ADORA3 | 19790066 [60], 17922624 [61] | |
AMPD1 | 34C>T (rs17602729) | 16947783 [62], 19902562a[57], 20386493 [46] |
ATIC | 347C>G (rs2372536) rs4673993 |
19902562a[57], 16947783 [62], 19016697a[26], 20386493a[46], 19193698 [63] |
CBS | 844ins68 | 14647408a[21], 16013960 [64] |
CCND1 | 870A>G (rs9344) | 12972956 [44], 16870553 [7], 18096764 [45] |
DHFR | 35289A>G (rs1232027), −473T>C (rs1650697), 308G > A (rs1105525), rs7387 |
20472929 [13], 19861437 [65], 18096764 [45], 19902562 [57], 20136356a[49] |
FOLH1 | 1561C>T | 12204797a[66] |
FOLR1 | 19493312b[67], 19093297a[48] | |
FPGS | rs1544105 | 19902562 [57], 19093297a[48] |
GART | 15142881b[68] | |
GGH | 452C>T (rs11545078) −401T>C (rs3758149) 16T>C (rs1800909) |
20386493a[46], 16999998 [69], 19827168 [32], 15564880 [70], 17286537 [71] |
GSTM1 | Null | 17180579 [39], 15713801 [29] |
HPRT1 | 12429535b[72] | |
HLA-G | 14bp in/del | 16906016 [73], 17626975 [74], 19088262a[75], 20136356a[49] |
IMPDH2 | +787C>T | 20136356a[49] |
ITPA | 94C>A (rs41320251), IVS2 +21A>C (rs7270101) |
16947783 [62], 17530705 [76], 19193698a[63], 20136356a[49] |
MTHFD1 | 1958G>A (rs2236225) | 14647408 [21], 15797993a[77], 17530705 [76], 20386493a[46] |
MTHFR | 1793G>A (rs2274976), rs2066462 |
20863444 [18] |
MTHFS | 3801490b[78] | |
MTR | 2756A>G (rs1805087) | 20386493a[46], 19159907 [79], 18322994 [42], 17855192 [80], 17611986 [81] |
MTRR | 66A>G (rs1801394) | 20386493a[46], 15797993 [77], 18368069 [28] |
NOS3 | 894G>T (rs1799983) | 14647408 [21], 16013960 [64] |
NP | 12429535b[72] | |
PPAT | 14529544b[82] | |
SHMT1 | 1420C>T (rs1979277) | 20386493 [46], 18368069 [28], 15797993 [77] |
SLC22A11 | 11327718b[83] | |
SLC22A6 | 20460822b[84] | |
SLC22A8 | 20460822b[84] | |
SLC46A1 | 19403800b[85] | |
SLCO1B1 | rs11045879, rs4149081 |
19901119 [4] |
SLCO1A2 | Glu172Asp (rs11568563), Arg168Cys (rs11568564), Asn277DEL (rs72559749) |
16702441b[86] |
TGFB1 | 869T>C (rs1982073) | 20136356a[49] |
TLR4 | 896A>G (rs4986790) | 20136356 [49] |
TYMS | 1494del (TTAAAG) | 18381794 [12], 16130010 [87] |
Annotations are available in full at http://www.pharmgkb.org/do/serve?objCls=Drug&objId=PA450428&tabType=tabGenetics#tabview=tab2.
PMID, PubMed identifier.
No association.
In-vitro study, (a) folate only, not methotrexate.
The interindividual variability in methotrexate pharmacokinetics can be explained partially by genetic variations in membrane transporter proteins with an affinity for methotrexate [4,89]. In the gastrointestinal tract, methotrexate is absorbed through active transport mediated by the reduced folate carrier (SLC19A1) and possibly also by the proton-coupled folate transporter SLC46A1 (HCP1, PCFT) at the apical membrane of enterocytes [90]. Furthermore, the bioavailability of methotrexate after oral dosing may be affected by ABC transporters, which can move methotrexate out of the enterocytes and back into the intestinal tract (ABCC2, ABCB1, and ABCG2) or into the blood (ABCC1 and ABCC3) [91-93]. Systemic clearance of methotrexate happens primarily through renal glomerular filtration and active secretion over the proximal tubular cells. Several renal transporter proteins have an affinity for methotrexate (SLC22A6, SLC22A8, SLC19A1, ABCG2, ABCC2, and ABCC4), and single nucleotide polymorphisms (SNPs) in ABCC2 have been associated with delayed methotrexate clearance [53,89,94-98]. Hepatic uptake of methotrexate involves the SLCO1B1 and SLCO1B3 transporters, in which SNPs have recently been found to explain up to 10% of the interpatient variability in the clearance of high-dose methotrexate [4,99-101]. Most of the methotrexate in hepatocytes reenters the blood circulation by transporter proteins (ABCC3 and ABCC4) in the basolateral membrane, and only a small portion of the methotrexate is excreted into the bile duct by the ABCC2 and ABCB1 transporters [102-105]. Inside the hepatocytes, aldehyde oxidase can convert methotrexate to 7-hydroxymethotrexate, a metabolite that is eliminated by the same route as methotrexate. At the blood–brain barrier, ABC transporters with affinities for methotrexate are located in endothelial cells, but the effect of facilitated transport of methotrexate out of the cerebrospinal fluid remains to be unclear [106].
The pharmacodynamic profile of methotrexate can, to a large extent, be explained by its interactions with enzymes in the folate pathway. The effects on methotrexate response of variations in these genes have been extensively studied in cancer treatments [107]. At extracellular methotrexate concentrations below 20 μmol/l, methotrexate enters cancer cells primarily through the reduced folate carrier (SLC19A1) [108,109], whereas efflux across the cell membrane is mediated by various ABC transporters; variations in these genes are known mechanisms of drug resistance in cancer cells [110]. Inside the cells, methotrexate is converted to active methotrexate polyglutamates (MTXPGs) by folylpolyglutamate synthetase, which adds glutamate residues to methotrexate [111]. The primary action of methotrexate is inhibition of the enzyme dihydrofolate reductase (DHFR), which converts dihydrofolate to tetrahydrofolate (THF)[112]. Tetrahydrofolate is essential for de novo purine synthesis, and in the biologically active form, 5-methyl-tetrahydrofolate, is an important cofactor in one-carbon metabolism. The effect of methotrexate depends on the function and expression of several other enzymes in the folate pathway, including methylenetetrahydrofolate dehydrogenase (MTHFD1), 5,10-methylenetetrahydrofolate reductase (MTHFR), and thymidylate synthetase (TYMS). Compared with methotrexate, the active metabolites, MTXPGs induce stronger inhibition of the target enzymes (i.e. TYMS and DHFR) and further inhibit key enzymes such as phosphoribosylglycinamide formyltransferase, phosphoribosylglycinamide synthetase, phosphoribosylaminoimidazole synthetase and 5-aminoimidazole-4-carboxamide ribonucleotide formyltransferase/IMP cyclohydrolase (ATIC) in the de novo purine synthesis pathway. MTXPG inhibition results in decreased protein and DNA methylation in addition to impaired DNA formation and repair. MTXPG levels are sustained inside the cells for a longer time than those of methotrexate; degradation of MTXPGs to methotrexate depends on the activity of the lysosomal enzyme γ-glutamyl hydrolase, which catalyzes the removal of polyglutamates [113-115]. MTXPGs have been investigated in relation to clinical outcomes in rheumatoid arthritis (RA) and juvenile idiopathic arthritis (JIA). Specifically, higher concentrations of long-chain MTXPGs have been associated with favorable outcomes in RA [116] and risk of gastrointestinal and hepatic toxicity in JIA [117].
Gene expression and genetic variation in candidate genes have been studied extensively in relation to many methotrexate response measures, including MTXPG accumulation (γ-glutamyl hydrolase, folylpolyglutamate synthetase, and SLC19A1) [108,118,119], reduction in tumor size (SLC19A1 and DHFR) [120], toxicity (MTHFR, and TYMS) [11,15], and relapse (DHFR, TYMS, MTHFR, DHFR, and SLC19A1) [29,37,40,45,54,87,121]. Conflicting results among studies suggest that the effects of genetic variation are therapy dependent and probably reflect the route of administration, dose, and duration of methotrexate treatment [37,40]. Although these studies have contributed to our understanding of the effects of Methotrexate and the molecular mechanisms involved in drug resistance, no genetic variant has yet been prospectively evaluated as a predictor of an outcome in a clinical trial.
Genome-wide studies have linked genes outside the folate pathway to the pharmacokinetics and effects of methotrexate; many of these genes have not been analyzed earlier in studies using the candidate approach. A recent study analyzed the association between MTXPG accumulation and genetic variations such as leukemic cell gene expression, somatic copy number variation, and SNPs [122]. Six genes on chromosome 18 (FHOD3, IMPA2, ME2, SLC39A6, SMAD2, and SMAD4) and one on chromosome 10 (RASSF4) were found to be associated with in-vivo intracellular accumulation of MTXPG in leukemic cells in all three categories of genetic variation. In another genome-wide study of patients with acute lymphoblastic leukemia, in-vivo response to MTX was found to be significantly associated with the expression of genes in the nucleotide pathway (e.g. TYMS), but also with genes involved in cell proliferation and apoptosis, and DNA repair and replication in the leukemic cells [123]. Finally, a genome-wide association study that assessed the link between inherited genomic variation and initial treatment response in patients with acute lymphoblastic leukemia showed 14 SNPs significantly associated with both treatment response and methotrexate clearance or MTXPG accumulation in leukemic cells [124]; early treatment response assessed by eradication of leukemic cells is strongly associated with cure rates and is therefore considered an important clinical phenotype.
No genome-wide association studies have yet been carried out in patients with rheumatoid arthritis, but inherited variations in most genes from the folate pathway have been examined in relation to methotrexate treatment response and toxicity [12,35]. However, to see a clinically relevant effect of genetic variants in the folate, purine, and pyrimidine pathways, it seems crucial to study gene–gene interactions; it has been suggested that the effects of individual SNPs are enhanced when they occur in combination with other common SNPs in these pathways [1]. Combinations of SNPs in the genes ATIC and adenosine receptor 2a have been associated with differential MTXPG concentrations in JIA [117]. The anti-inflammatory effect of methotrexate is further thought to be mediated through interaction with the adenosine biosynthesis pathway [125]. MTXPGs inhibit the enzyme ATIC, which after a cascade of events leads to the accumulation of the anti-inflammatory molecule adenosine; SNPs in genes from the adenosine biosynthesis pathway (i.e. ATIC, inosine triphosphatase, and adenosine monophosphate deaminase 1) have been found to predict the efficacy of methotrexate treatment for RA and JIA [1,62,76,126].
Regardless of disease, it seems clear that future studies should continue to examine the combined effect of variations in multiple genes to characterize the extent of genomic determinants on variation in the pharmacokinetics and pharmacodynamics of methotrexate.
Acknowledgements
The authors thank Fen Liu for assistance with the graphics. This work is supported by the NIH/NIGMS (U01GM61374).
References
- 1.Dervieux T, Wessels JA, Van der ST, Penrod N, Moore JH, Guchelaar HJ, et al. Gene–gene interactions in folate and adenosine biosynthesis pathways affect methotrexate efficacy and tolerability in rheumatoid arthritis. Pharmacogenet Genomics. 2009;19:935–944. doi: 10.1097/FPC.0b013e32833315d1. [DOI] [PubMed] [Google Scholar]
- 2.Pui CH, Campana D, Pei D, Bowman WP, Sandlund JT, Kaste SC, et al. Treating childhood acute lymphoblastic leukemia without cranial irradiation. N Engl J Med. 2009;360:2730–2741. doi: 10.1056/NEJMoa0900386. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Widemann BC, Balis FM, Kempf-Bielack B, Bielack S, Pratt CB, Ferrari S, et al. High-dose methotrexate-induced nephrotoxicity in patients with osteosarcoma. Cancer. 2004;100:2222–2232. doi: 10.1002/cncr.20255. [DOI] [PubMed] [Google Scholar]
- 4.Trevino LR, Shimasaki N, Yang W, Panetta JC, Cheng C, Pei D, et al. Germ line genetic variation in an organic anion transporter polypeptide associated with methotrexate pharmacokinetics and clinical effects. J Clin Oncol. 2009;27:5972–5978. doi: 10.1200/JCO.2008.20.4156. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Buitenkamp TD, Mathot RAA, de Haas V, Pieters R, Zwaan CM. Methotrexate-induced side effects are not due to differences in pharmacokinetics in children with Down syndrome and acute lymphoblastic leukemia. Haematologica. 2010;95:1106–1113. doi: 10.3324/haematol.2009.019778. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Tetef ML, Margolin KA, Doroshow JH, Akman S, Leong LA, Morgan RJ, Jr, et al. Pharmacokinetics and toxicity of high-dose intravenous methotrexate in the treatment of leptomeningeal carcinomatosis. Cancer Chemother Pharmacol. 2000;46:19–26. doi: 10.1007/s002800000118. [DOI] [PubMed] [Google Scholar]
- 7.Costea I, Moghrabi A, Laverdiere C, Graziani A, Krajinovic M. Folate cycle gene variants and chemotherapy toxicity in pediatric patients with acute lymphoblastic leukemia. Haematologica. 2006;91:1113–1116. [PubMed] [Google Scholar]
- 8.Robien K, Bigler J, Yasui Y, Potter JD, Martin P, Storb R, et al. Methylenetetrahydrofolate reductase and thymidylate synthase genotypes and risk of acute graft-versus-host disease following hematopoietic cell transplantation for chronic myelogenous leukemia. Biol Blood Marrow Transplant. 2006;12:973–980. doi: 10.1016/j.bbmt.2006.05.016. [DOI] [PubMed] [Google Scholar]
- 9.Chiusolo P, Reddiconto G, Casorelli I, Laurenti L, Sora F, Mele L, et al. Preponderance of methylenetetrahydrofolate reductase C677T homozygosity among leukemia patients intolerant to methotrexate. Ann Oncol. 2002;13:1915–1918. doi: 10.1093/annonc/mdf322. [DOI] [PubMed] [Google Scholar]
- 10.Gemmati D, Ongaro A, Tognazzo S, Catozzi L, Federici F, Mauro E, et al. Methylenetetrahydrofolate reductase C677T and A1298C gene variants in adult non-Hodgkin’s lymphoma patients: association with toxicity and survival. Haematologica. 2007;92:478–485. doi: 10.3324/haematol.10587. [DOI] [PubMed] [Google Scholar]
- 11.Ulrich CM, Yasui Y, Storb R, Schubert MM, Wagner JL, Bigler J, et al. Pharmacogenetics of methotrexate: toxicity among marrow transplantation patients varies with the methylenetetrahydrofolate reductase C677T polymorphism. Blood. 2001;98:231–234. doi: 10.1182/blood.v98.1.231. [DOI] [PubMed] [Google Scholar]
- 12.Ranganathan P, Culverhouse R, Marsh S, Mody A, Scott-Horton TJ, Brasington R, et al. Methotrexate (MTX) pathway gene polymorphisms and their effects on MTX toxicity in Caucasian and African American patients with rheumatoid arthritis. J Rheumatol. 2008;35:572–579. [PubMed] [Google Scholar]
- 13.Chandran V, Siannis F, Rahman P, Pellett FJ, Farewell VT, Gladman DD. Folate pathway enzyme gene polymorphisms and the efficacy and toxicity of methotrexate in psoriatic arthritis. J Rheumatol. 2010;37:1508–1512. doi: 10.3899/jrheum.091311. [DOI] [PubMed] [Google Scholar]
- 14.Chiusolo P, Reddiconto G, Farina G, Mannocci A, Fiorini A, Palladino M, et al. MTHFR polymorphisms’ influence on outcome and toxicity in acute lymphoblastic leukemia patients. Leuk Res. 2007;31:1669–1674. doi: 10.1016/j.leukres.2007.03.028. [DOI] [PubMed] [Google Scholar]
- 15.Robien K, Schubert MM, Chay T, Bigler J, Storb R, Yasui Y, et al. Methylenetetrahydrofolate reductase and thymidylate synthase genotypes modify oral mucositis severity following hematopoietic stem cell transplantation. Bone Marrow Transplant. 2006;37:799–800. doi: 10.1038/sj.bmt.1705330. [DOI] [PubMed] [Google Scholar]
- 16.Robien K, Schubert MM, Bruemmer B, Lloid ME, Potter JD, Ulrich CM. Predictors of oral mucositis in patients receiving hematopoietic cell transplants for chronic myelogenous leukemia. J Clin Oncol. 2004;22:1268–1275. doi: 10.1200/JCO.2004.05.147. [DOI] [PubMed] [Google Scholar]
- 17.Van Ede AE, Laan RF, Blom HJ, Huizinga TW, Haagsma CJ, Giesendorf BA, et al. The C677T mutation in the methylenetetrahydrofolate reductase gene: a genetic risk factor for methotrexate-related elevation of liver enzymes in rheumatoid arthritis patients. Arthritis Rheum. 2001;44:2525–2530. doi: 10.1002/1529-0131(200111)44:11<2525::aid-art432>3.0.co;2-b. [DOI] [PubMed] [Google Scholar]
- 18.Xiao H, Xu J, Zhou X, Stankovich J, Pan F, Zhang Z, et al. Associations between the genetic polymorphisms of MTHFR and outcomes of methotrexate treatment in rheumatoid arthritis. Clin Exp Rheumatol. 2010;28:728–733. [PubMed] [Google Scholar]
- 19.Tukova J, Chladek J, Hroch M, Nemcova D, Hoza J, Dolezalova P. 677TT genotype is associated with elevated risk of methotrexate (MTX) toxicity in juvenile idiopathic arthritis: treatment outcome, erythrocyte concentrations of MTX and folates, and MTHFR polymorphisms. J Rheumatol. 2010;37:2180–2186. doi: 10.3899/jrheum.091427. [DOI] [PubMed] [Google Scholar]
- 20.Shimasaki N, Mori T, Torii C, Sato R, Shimada H, Tanigawara Y, et al. Influence of MTHFR and RFC1 polymorphisms on toxicities during maintenance chemotherapy for childhood acute lymphoblastic leukemia or lymphoma. J Pediatr Hematol Oncol. 2008;30:347–352. doi: 10.1097/MPH.0b013e318165b25d. [DOI] [PubMed] [Google Scholar]
- 21.Krajinovic M, Lemieux-Blanchard E, Chiasson S, Primeau M, Costea I, Moghrabi A. Role of polymorphisms in MTHFR and MTHFD1 genes in the outcome of childhood acute lymphoblastic leukemia. Pharmacogenomics J. 2004;4:66–72. doi: 10.1038/sj.tpj.6500224. [DOI] [PubMed] [Google Scholar]
- 22.Aplenc R, Thompson J, Han P, La M, Zhao H, Lange B, et al. Methylenetetrahydrofolate reductase polymorphisms and therapy response in pediatric acute lymphoblastic leukemia. Cancer Res. 2005;65:2482–2487. doi: 10.1158/0008-5472.CAN-04-2606. [DOI] [PubMed] [Google Scholar]
- 23.Nuckel H, Frey UH, Durig J, Duhrsen U, Siffert W. Methylenetetrahydrofolate reductase (MTHFR) gene 677C>T and 1298A>C polymorphisms are associated with differential apoptosis of leukemic B cells in vitro and disease progression in chronic lymphocytic leukemia. Leukemia. 2004;18:1816–1823. doi: 10.1038/sj.leu.2403484. [DOI] [PubMed] [Google Scholar]
- 24.Kishi S, Griener J, Cheng C, Das S, Cook EH, Pei D, et al. Homocysteine, pharmacogenetics, and neurotoxicity in children with leukemia. J Clin Oncol. 2003;21:3084–3091. doi: 10.1200/JCO.2003.07.056. [DOI] [PubMed] [Google Scholar]
- 25.Hughes LB, Beasley TM, Patel H, Tiwari HK, Morgan SL, Baggott JE, et al. Racial or ethnic differences in allele frequencies of single-nucleotide polymorphisms in the methylenetetrahydrofolate reductase gene and their influence on response to methotrexate in rheumatoid arthritis. Ann Rheum Dis. 2006;65:1213–1218. doi: 10.1136/ard.2005.046797. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Warren RB, Smith RL, Campalani E, Eyre S, Smith CH, Barker JN, et al. Outcomes of methotrexate therapy for psoriasis and relationship to genetic polymorphisms. Br J Dermatol. 2009;160:438–441. doi: 10.1111/j.1365-2133.2008.08898.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Mena JP, Salazar-Paramo M, Gonzalez-Lopez L, Gamez-Nava JI, Sandoval-Ramirez L, Sanchez JD, et al. Polymorphisms C677T and A1298C in the MTHFR gene in Mexican patients with rheumatoid arthritis treated with methotrexate: implication with elevation of transaminases. Pharmacogenomics J. 2010 doi: 10.1038/tpj.2010.32. Epub ahead of print. [DOI] [PubMed] [Google Scholar]
- 28.Huang L, Tissing WJ, de Jonge R, Van Zelst BD, Pieters R. Polymorphisms in folate-related genes: association with side effects of high-dose methotrexate in childhood acute lymphoblastic leukemia. Leukemia. 2008;22:1798–1800. doi: 10.1038/leu.2008.66. [DOI] [PubMed] [Google Scholar]
- 29.Rocha JC, Cheng C, Liu W, Kishi S, Das S, Cook EH, et al. Pharmacogenetics of outcome in children with acute lymphoblastic leukemia. Blood. 2005;105:4752–4758. doi: 10.1182/blood-2004-11-4544. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Shimasaki N, Mori T, Samejima H, Sato R, Shimada H, Yahagi N, et al. Effects of methylenetetrahydrofolate reductase and reduced folate carrier 1 polymorphisms on high-dose methotrexate-induced toxicities in children with acute lymphoblastic leukemia or lymphoma. J Pediatr Hematol Oncol. 2006;28:64–68. doi: 10.1097/01.mph.0000198269.61948.90. [DOI] [PubMed] [Google Scholar]
- 31.Seidemann K, Book M, Zimmermann M, Meyer U, Welte K, Stanulla M, et al. MTHFR 677 (C–>T) polymorphism is not relevant for prognosis or therapy-associated toxicity in pediatric NHL: results from 484 patients of multicenter trial NHL-BFM 95. Ann Hematol. 2006;85:291–300. doi: 10.1007/s00277-005-0072-2. [DOI] [PubMed] [Google Scholar]
- 32.Hayashi H, Fujimaki C, Daimon T, Tsuboi S, Matsuyama T, Itoh K. Genetic polymorphisms in folate pathway enzymes as a possible marker for predicting the outcome of methotrexate therapy in Japanese patients with rheumatoid arthritis. J Clin Pharm Ther. 2009;34:355–361. doi: 10.1111/j.1365-2710.2009.01046.x. [DOI] [PubMed] [Google Scholar]
- 33.Jazbec J, Kitanovski L, Aplenc R, Debeljak M, Dolzan V. No evidence of association of methylenetetrahydrofolate reductase polymorphism with occurrence of second neoplasms after treatment of childhood leukemia. Leuk Lymphoma. 2005;46:893–897. doi: 10.1080/10428190500086428. [DOI] [PubMed] [Google Scholar]
- 34.Pakakasama S, Kanchanakamhaeng K, Kajanachumpol S, Udomsubpayakul U, Sirachainan N, Thithapandha A, et al. Genetic polymorphisms of folate metabolic enzymes and toxicities of high-dose methotrexate in children with acute lymphoblastic leukemia. Ann Hematol. 2007;86:609–611. doi: 10.1007/s00277-007-0274-x. [DOI] [PubMed] [Google Scholar]
- 35.Wessels JAM, de Vries-Bouwstra JK, Heijmans BT, Slagboom PE, Goekoop-Ruiterman YPM, Allaart CF, et al. Efficacy and toxicity of methotrexate in early rheumatoid arthritis are associated with single-nucleotide polymorphisms in genes coding for folate pathway enzymes. Arthritis Rheum. 2006;54:1087–1095. doi: 10.1002/art.21726. [DOI] [PubMed] [Google Scholar]
- 36.Robien K, Ulrich CM, Bigler J, Yasui Y, Gooley T, Bruemmer B, et al. Methylenetetrahydrofolate reductase genotype affects risk of relapse after hematopoietic cell transplantation for chronic myelogenous leukemia. Clin Cancer Res. 2004;10:7592–7598. doi: 10.1158/1078-0432.CCR-04-1057. [DOI] [PubMed] [Google Scholar]
- 37.Laverdiere C, Chiasson S, Costea I, Moghrabi A, Krajinovic M. Polymorphism G80A in the reduced folate carrier gene and its relationship to methotrexate plasma levels and outcome of childhood acute lymphoblastic leukemia. Blood. 2002;100:3832–3834. doi: 10.1182/blood.V100.10.3832. [DOI] [PubMed] [Google Scholar]
- 38.Kishi S, Cheng C, French D, Pei D, Das S, Cook EH, et al. Ancestry and pharmacogenetics of antileukemic drug toxicity. Blood. 2007;109:4151–4157. doi: 10.1182/blood-2006-10-054528. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Imanishi H, Okamura N, Yagi M, Noro Y, Moriya Y, Nakamura T, et al. Genetic polymorphisms associated with adverse events and elimination of methotrexate in childhood acute lymphoblastic leukemia and malignant lymphoma. J Hum Genet. 2007;52:166–171. doi: 10.1007/s10038-006-0096-z. [DOI] [PubMed] [Google Scholar]
- 40.Gregers J, Christensen IJ, Dalhoff K, Lausen B, Schroeder H, Rosthoej S, et al. The association of reduced folate carrier 80G>A polymorphism to outcome in childhood acute lymphoblastic leukemia interacts with chromosome 21 copy number. Blood. 2010;115:4671–4677. doi: 10.1182/blood-2010-01-256958. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Drozdzik M, Rudas T, Pawlik A, Gornik W, Kurzawski M, Herczynska M. Reduced folate carrier-1 80G>A polymorphism affects methotrexate treatment outcome in rheumatoid arthritis. Pharmacogenomics J. 2007;7:404–407. doi: 10.1038/sj.tpj.6500438. [DOI] [PubMed] [Google Scholar]
- 42.James HM, Gillis D, Hissaria P, Lester S, Somogyi AA, Cleland LG, et al. Common polymorphisms in the folate pathway predict efficacy of combination regimens containing methotrexate and sulfasalazine in early rheumatoid arthritis. J Rheumatol. 2008;35:562–571. [PubMed] [Google Scholar]
- 43.Krajinovic M, Costea I, Chiasson S. Polymorphism of the thymidylate synthase gene and outcome of acute lymphoblastic leukaemia. Lancet. 2002;359:1033–1034. doi: 10.1016/S0140-6736(02)08065-0. [DOI] [PubMed] [Google Scholar]
- 44.Costea I, Moghrabi A, Krajinovic M. The influence of cyclin D1 (CCND1) 870A>G polymorphism and CCND1-thymidylate synthase (TS) gene–gene interaction on the outcome of childhood acute lymphoblastic leukaemia. Pharmacogenetics. 2003;13:577–580. doi: 10.1097/00008571-200309000-00006. [DOI] [PubMed] [Google Scholar]
- 45.Dulucq S, St-Onge G, Gagne V, Ansari M, Sinnett D, Labuda D, et al. DNA variants in the dihydrofolate reductase gene and outcome in childhood ALL. Blood. 2008;111:3692–3700. doi: 10.1182/blood-2007-09-110593. [DOI] [PubMed] [Google Scholar]
- 46.Stamp LK, Chapman PT, O’Donnell JL, Zhang M, James J, Frampton C, et al. Polymorphisms within the folate pathway predict folate concentrations but are not associated with disease activity in rheumatoid arthritis patients on methotrexate. Pharmacogenet Genomics. 2010;20:367–376. doi: 10.1097/FPC.0b013e3283398a71. [DOI] [PubMed] [Google Scholar]
- 47.Lauten M, Asgedom G, Welte K, Schrappe M, Stanulla M. Thymidylate synthase gene polymorphism and its association with relapse in childhood B-cell precursor acute lymphoblastic leukemia. Haematologica. 2003;88:353–354. [PubMed] [Google Scholar]
- 48.Sharma S, Das M, Kumar A, Marwaha V, Shankar S, Aneja R, et al. Interaction of genes from influx-metabolism-efflux pathway and their influence on methotrexate efficacy in rheumatoid arthritis patients among Indians. Pharmacogenet Genomics. 2008;18:1041–1049. doi: 10.1097/fpc.0b013e328311a8fd. [DOI] [PubMed] [Google Scholar]
- 49.Kooloos WM, Wessels JA, Van der Straaten T, Allaart CF, Huizinga TW, Guchelaar HJ. Functional polymorphisms and methotrexate treatment outcome in recent-onset rheumatoid arthritis. Pharmacogenomics. 2010;11:163–175. doi: 10.2217/pgs.09.139. [DOI] [PubMed] [Google Scholar]
- 50.Letourneau IJ, Deeley RG, Cole SP. Functional characterization of nonsynonymous single nucleotide polymorphisms in the gene encoding human multidrug resistance protein 1 (MRP1/ABCC1) Pharmacogenet Genomics. 2005;15:647–657. doi: 10.1097/01.fpc.0000173484.51807.48. [DOI] [PubMed] [Google Scholar]
- 51.Warren RB, Smith RL, Campalani E, Eyre S, Smith CH, Barker JN, et al. Genetic variation in efflux transporters influences outcome to methotrexate therapy in patients with psoriasis. J Invest Dermatol. 2008;128:1925–1929. doi: 10.1038/jid.2008.16. [DOI] [PubMed] [Google Scholar]
- 52.Zeng H, Chen ZS, Belinsky MG, Rea PA, Kruh GD. Transport of methotrexate (MTX) and folates by multidrug resistance protein (MRP) 3 and MRP1: effect of polyglutamylation on MTX transport. Cancer Res. 2001;61:7225–7232. [PubMed] [Google Scholar]
- 53.Vlaming ML, Van Esch A, Pala Z, Wagenaar E, Van de Wetering K, Van Tellingen O, et al. Abcc2 (Mrp2), Abcc3 (Mrp3), and Abcg2 (Bcrp1) are the main determinants for rapid elimination of methotrexate and its toxic metabolite 7-hydroxymethotrexate in vivo. Mol Cancer Ther. 2009;8:3350–3359. doi: 10.1158/1535-7163.MCT-09-0668. [DOI] [PubMed] [Google Scholar]
- 54.Ansari M, Sauty G, Labuda M, Gagne V, Laverdiere C, Moghrabi A, et al. Polymorphisms in multidrug resistance-associated protein gene 4 is associated with outcome in childhood acute lymphoblastic leukemia. Blood. 2009;114:1383–1386. doi: 10.1182/blood-2008-11-191098. [DOI] [PubMed] [Google Scholar]
- 55.Volk EL, Farley KM, Wu Y, Li F, Robey RW, Schneider E. Overexpression of wild-type breast cancer resistance protein mediates methotrexate resistance. Cancer Res. 2002;62:5035–5040. [PubMed] [Google Scholar]
- 56.Chen ZS, Robey RW, Belinsky MG, Shchaveleva I, Ren XQ, Sugimoto Y, et al. Transport of methotrexate, methotrexate polyglutamates, and 17beta-estradiol 17-(beta-D-glucuronide) by ABCG2: effects of acquired mutations at R482 on methotrexate transport. Cancer Res. 2003;63:4048–4054. [PubMed] [Google Scholar]
- 57.Sharma S, Das M, Kumar A, Marwaha V, Shankar S, Singh P, et al. Purine biosynthetic pathway genes and methotrexate response in rheumatoid arthritis patients among north Indians. Pharmacogenet Genomics. 2009;19:823–828. doi: 10.1097/fpc.0b013e328331b53e. [DOI] [PubMed] [Google Scholar]
- 58.Merrill JT, Shen C, Schreibman D, Coffey D, Zakharenko O, Fisher R, et al. Adenosine A1 receptor promotion of multinucleated giant cell formation by human monocytes: a mechanism for methotrexate-induced nodulosis in rheumatoid arthritis. Arthritis Rheum. 1997;40:1308–1315. doi: 10.1002/1529-0131(199707)40:7<1308::AID-ART16>3.0.CO;2-M. [DOI] [PubMed] [Google Scholar]
- 59.Hider SL, Thomson W, Mack LF, Armstrong DJ, Shadforth M, Bruce IN. Polymorphisms within the adenosine receptor 2a gene are associated with adverse events in RA patients treated with MTX. Rheumatology (Oxford) 2008;47:1156–1159. doi: 10.1093/rheumatology/ken182. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Varani K, Massara A, Vincenzi F, Tosi A, Padovan M, Trotta F, et al. Normalization of A2A and A3 adenosine receptor upregulation in rheumatoid arthritis patients by treatment with anti-tumor necrosis factor alpha but not methotrexate. Arthritis Rheum. 2009;60:2880–2891. doi: 10.1002/art.24794. [DOI] [PubMed] [Google Scholar]
- 61.Bar-Yehuda S, Silverman MH, Kerns WD, Ochaion A, Cohen S, Fishman P. The anti-inflammatory effect of A3 adenosine receptor agonists: a novel targeted therapy for rheumatoid arthritis. Expert Opin Investig Drugs. 2007;16:1601–1613. doi: 10.1517/13543784.16.10.1601. [DOI] [PubMed] [Google Scholar]
- 62.Wessels JAM, Kooloos WM, De Jonge R, de Vries-Bouwstra JK, Allaart CF, Linssen A, et al. Relationship between genetic variants in the adenosine pathway and outcome of methotrexate treatment in patients with recent-onset rheumatoid arthritis. Arthritis Rheum. 2006;54:2830–2839. doi: 10.1002/art.22032. [DOI] [PubMed] [Google Scholar]
- 63.Lee YC, Cui J, Costenbader KH, Shadick NA, Weinblatt ME, Karlson EW. Investigation of candidate polymorphisms and disease activity in rheumatoid arthritis patients on methotrexate. Rheumatology (Oxford) 2009;48:613–617. doi: 10.1093/rheumatology/ken513. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Krajinovic M, Robaey P, Chiasson S, Lemieux-Blanchard E, Rouillard M, Primeau M, et al. Polymorphisms of genes controlling homocysteine levels and IQ score following the treatment for childhood ALL. Pharmacogenomics. 2005;6:293–302. doi: 10.1517/14622416.6.3.293. [DOI] [PubMed] [Google Scholar]
- 65.Al-Shakfa F, Dulucq S, Brukner I, Milacic I, Ansari M, Beaulieu P, et al. DNA variants in region for noncoding interfering transcript of dihydrofolate reductase gene and outcome in childhood acute lymphoblastic leukemia. Clin Cancer Res. 2009;15:6931–6938. doi: 10.1158/1078-0432.CCR-09-0641. [DOI] [PubMed] [Google Scholar]
- 66.Lievers KJ, Kluijtmans LA, Boers GH, Verhoef P, den Heijer M, Trijbels FJ, et al. Influence of a glutamate carboxypeptidase II (GCPII) polymorphism (1561C–>T) on plasma homocysteine, folate and vitamin B(12) levels and its relationship to cardiovascular disease risk. Atherosclerosis. 2002;164:269–273. doi: 10.1016/s0021-9150(02)00065-5. [DOI] [PubMed] [Google Scholar]
- 67.Sanchez-del-Campo L, Montenegro MF, Cabezas-Herrera J, Rodriguez-Lopez JN. The critical role of alpha-folate receptor in the resistance of melanoma to methotrexate. Pigment Cell Melanoma Res. 2009;22:588–600. doi: 10.1111/j.1755-148X.2009.00586.x. [DOI] [PubMed] [Google Scholar]
- 68.Zaza G, Yang W, Kager L, Cheok M, Downing J, Pui CH, et al. Acute lymphoblastic leukemia with TEL-AML1 fusion has lower expression of genes involved in purine metabolism and lower de novo purine synthesis. Blood. 2004;104:1435–1441. doi: 10.1182/blood-2003-12-4306. [DOI] [PubMed] [Google Scholar]
- 69.Garcia-Bournissen F, Moghrabi A, Krajinovic M. Therapeutic responses in childhood acute lymphoblastic leukemia (ALL) and haplotypes of gamma glutamyl hydrolase (GGH) gene. Leuk Res. 2007;31:1023–1025. doi: 10.1016/j.leukres.2006.08.007. [DOI] [PubMed] [Google Scholar]
- 70.Dervieux T, Kremer J, Lein DO, Capps R, Barham R, Meyer G, et al. Contribution of common polymorphisms in reduced folate carrier and gamma-glutamylhydrolase to methotrexate polyglutamate levels in patients with rheumatoid arthritis. Pharmacogenetics. 2004;14:733–739. doi: 10.1097/00008571-200411000-00004. [DOI] [PubMed] [Google Scholar]
- 71.Van der Straaten RJ, Wessels JA, de Vries-Bouwstra JK, Goekoop-Ruiterman YP, Allaart CF, Bogaartz J, et al. Exploratory analysis of four polymorphisms in human GGH and FPGS genes and their effect in methotrexate-treated rheumatoid arthritis patients. Pharmacogenomics. 2007;8:141–150. doi: 10.2217/14622416.8.2.141. [DOI] [PubMed] [Google Scholar]
- 72.Van Ede AE, Laan RF, De Abreu RA, Stegeman AB, Van de Putte LB. Purine enzymes in patients with rheumatoid arthritis treated with methotrexate. Ann Rheum Dis. 2002;61:1060–1064. doi: 10.1136/ard.61.12.1060. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Rizzo R, Rubini M, Govoni M, Padovan M, Melchiorri L, Stignani M, et al. HLA-G 14-bp polymorphism regulates the methotrexate response in rheumatoid arthritis. Pharmacogenet Genomics. 2006;16:615–623. doi: 10.1097/01.fpc.0000230115.41828.3a. [DOI] [PubMed] [Google Scholar]
- 74.Baricordi OR, Govoni M, Rizzo R, Trotta F. In rheumatoid arthritis, a polymorphism in the HLA-G gene concurs in the clinical response to methotrexate treatment. Ann Rheum Dis. 2007;66:1125–1126. doi: 10.1136/ard.2006.064022. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Stamp LK, O’Donnell JL, Chapman PT, Barclay ML, Kennedy MA, Frampton CM, et al. Lack of association between HLA-G 14 bp insertion/deletion polymorphism and response to long-term therapy with methotrexate response in rheumatoid arthritis. Ann Rheum Dis. 2009;68:154–155. doi: 10.1136/ard.2008.089383. [DOI] [PubMed] [Google Scholar]
- 76.Wessels JAM, Van der Kooij SM, le Cessie S, Kievit W, Barerra P, Allaart CF, et al. A clinical pharmacogenetic model to predict the efficacy of methotrexate monotherapy in recent-onset rheumatoid arthritis. Arthritis Rheum. 2007;56:1765–1775. doi: 10.1002/art.22640. [DOI] [PubMed] [Google Scholar]
- 77.De Jonge R, Hooijberg JH, Van Zelst BD, Jansen G, Van Zantwijk CH, Kaspers GJ, et al. Effect of polymorphisms in folate-related genes on in-vitro methotrexate sensitivity in pediatric acute lymphoblastic leukemia. Blood. 2005;106:717–720. doi: 10.1182/blood-2004-12-4941. [DOI] [PubMed] [Google Scholar]
- 78.Bertrand R, MacKenzie RE, Jolivet J. Human liver methenyltetrahydrofolate synthetase: improved purification and increased affinity for folate polyglutamate substrates. Biochim Biophys Acta. 1987;911:154–161. doi: 10.1016/0167-4838(87)90004-5. [DOI] [PubMed] [Google Scholar]
- 79.Patino-Garcia A, Zalacain M, Marrodan L, San-Julian M, Sierrasesumaga 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. doi: 10.1016/j.jpeds.2008.11.030. [DOI] [PubMed] [Google Scholar]
- 80.Linnebank M, Malessa S, Moskau S, Semmler A, Pels H, Klockgether T, et al. Acute methotrexate-induced encephalopathy–causal relation to homozygous allelic state for MTR c.2756A>G (D919G)? J Chemother. 2007;19:455–457. doi: 10.1179/joc.2007.19.4.455. [DOI] [PubMed] [Google Scholar]
- 81.Berkun Y, Abou Atta I, Rubinow A, Orbach H, Levartovsky D, Aamar S, et al. 2756GG genotype of methionine synthase reductase gene is more prevalent in rheumatoid arthritis patients treated with methotrexate and is associated with methotrexate-induced nodulosis. J Rheumatol. 2007;34:1664–1669. [PubMed] [Google Scholar]
- 82.McGuire JJ. Anticancer antifolates: current status and future directions. Curr Pharm Des. 2003;9:2593–2613. doi: 10.2174/1381612033453712. [DOI] [PubMed] [Google Scholar]
- 83.Sun W, Wu RR, Van Poelje PD, Erion MD. Isolation of a family of organic anion transporters from human liver and kidney. Biochem Biophys Res Commun. 2001;283:417–422. doi: 10.1006/bbrc.2001.4774. [DOI] [PubMed] [Google Scholar]
- 84.Uwai Y, Iwamoto K. Transport of aminopterin by human organic anion transporters hOAT1 and hOAT3: comparison with methotrexate. Drug Metab Pharmacokinet. 2010;25:163–169. doi: 10.2133/dmpk.25.163. [DOI] [PubMed] [Google Scholar]
- 85.Unal ES, Zhao R, Goldman ID. Role of the glutamate 185 residue in proton translocation mediated by the proton-coupled folate transporter SLC46A1. Am J Physiol Cell Physiol. 2009;297:C66–C74. doi: 10.1152/ajpcell.00096.2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Badagnani I, Castro RA, Taylor TR, Brett CM, Huang CC, Stryke D, et al. Interaction of methotrexate with organic anion transporting polypeptide 1A2 and its genetic variants. J Pharmacol Exp Ther. 2006;318:521–529. doi: 10.1124/jpet.106.104364. [DOI] [PubMed] [Google Scholar]
- 87.Krajinovic M, Costea I, Primeau M, Dulucq S, Moghrabi A. Combining several polymorphisms of thymidylate synthase gene for pharmacogenetic analysis. Pharmacogenomics J. 2005;5:374–380. doi: 10.1038/sj.tpj.6500332. [DOI] [PubMed] [Google Scholar]
- 88.Goode EL, Potter JD, Bigler J, Ulrich CM. Methionine synthase D919G polymorphism, folate metabolism, and colorectal adenoma risk. Cancer Epidemiol Biomarkers Prev. 2004;13:157–162. doi: 10.1158/1055-9965.epi-03-0097. [DOI] [PubMed] [Google Scholar]
- 89.Rau T, Erney B, Gores R, Eschenhagen T, Beck J, Langer T. High-dose methotrexate in pediatric acute lymphoblastic leukemia: impact of ABCC2 polymorphisms on plasma concentrations. Clin Pharmacol Ther. 2006;80:468–476. doi: 10.1016/j.clpt.2006.08.012. [DOI] [PubMed] [Google Scholar]
- 90.Qiu A, Jansen M, Sakaris A, Min SH, Chattopadhyay S, Tsai E, et al. Identification of an intestinal folate transporter and the molecular basis for hereditary folate malabsorption. Cell. 2006;127:917–928. doi: 10.1016/j.cell.2006.09.041. [DOI] [PubMed] [Google Scholar]
- 91.Gradhand U, Kim RB. Pharmacogenomics of MRP transporters (ABCC1-5) and BCRP (ABCG2) Drug Metab Rev. 2008;40:317–354. doi: 10.1080/03602530801952617. [DOI] [PubMed] [Google Scholar]
- 92.Kruh GD, Belinsky MG, Gallo JM, Lee K. Physiological and pharmacological functions of Mrp2, Mrp3 and Mrp4 as determined from recent studies on gene-disrupted mice. Cancer Metastasis Rev. 2007;26:5–14. doi: 10.1007/s10555-007-9039-1. [DOI] [PubMed] [Google Scholar]
- 93.Chan L, Lowes S, Hirst BH. The ABCs of drug transport in intestine and liver: efflux proteins limiting drug absorption and bioavailability. Eur J Pharm Sci. 2004;21:25–51. doi: 10.1016/j.ejps.2003.07.003. [DOI] [PubMed] [Google Scholar]
- 94.El-Sheikh AA, Van den Heuvel JJ, Koenderink JB, Russel FG. Interaction of nonsteroidal anti-inflammatory drugs with multidrug resistance protein (MRP) 2/A. J Pharmacol Exp Ther. 2007;320:229–235. doi: 10.1124/jpet.106.110379. [DOI] [PubMed] [Google Scholar]
- 95.Hulot JS, Villard E, Maguy A, Morel V, Mir L, Tostivint I, et al. A mutation in the drug transporter gene ABCC2 associated with impaired methotrexate elimination. Pharmacogenet Genomics. 2005;15:277–285. doi: 10.1097/01213011-200505000-00002. [DOI] [PubMed] [Google Scholar]
- 96.Uwai Y, Taniguchi R, Motohashi H, Saito H, Okuda M, Inui K. Methotrexate-loxoprofen interaction: involvement of human organic anion transporters hOAT1 and hOAT3. Drug Metab Pharmacokinet. 2004;19:369–374. doi: 10.2133/dmpk.19.369. [DOI] [PubMed] [Google Scholar]
- 97.Van Aubel RA, Smeets PH, Peters JG, Bindels RJ, Russel FG. The MRP4/ABCC4 gene encodes a novel apical organic anion transporter in human kidney proximal tubules: putative efflux pump for urinary cAMP and cGMP. J Am Soc Nephrol. 2002;13:595–603. doi: 10.1681/ASN.V133595. [DOI] [PubMed] [Google Scholar]
- 98.Vanwert AL, Sweet DH. Impaired clearance of methotrexate in organic anion transporter 3 (Slc22a8) knockout mice: a gender-specific impact of reduced folates. Pharm Res. 2008;25:453–462. doi: 10.1007/s11095-007-9407-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99.Abe T, Kakyo M, Tokui T, Nakagomi R, Nishio T, Nakai D, et al. Identification of a novel gene family encoding human liver-specific organic anion transporter LST-1. J Biol Chem. 1999;274:17159–17163. doi: 10.1074/jbc.274.24.17159. [DOI] [PubMed] [Google Scholar]
- 100.Abe T, Unno M, Onogawa T, Tokui T, Kondo TN, Nakagomi R, et al. LST-2, a human liver-specific organic anion transporter, determines methotrexate sensitivity in gastrointestinal cancers. Gastroenterology. 2001;120:1689–1699. doi: 10.1053/gast.2001.24804. [DOI] [PubMed] [Google Scholar]
- 101.Van de SE, Van der Kruijssen CM, Wagenaar E, Burggraaff JE, Mesman E, Kenworthy KE, et al. Methotrexate pharmacokinetics in transgenic mice with liver-specific expression of human OATP1B1 (SLCO1B1) Drug Metab Dispos. 2009;37:277–281. doi: 10.1124/dmd.108.024315. [DOI] [PubMed] [Google Scholar]
- 102.Kitamura Y, Hirouchi M, Kusuhara H, Schuetz JD, Sugiyama Y. Increasing systemic exposure of methotrexate by active efflux mediated by multidrug resistance-associated protein 3 (Mrp3/Abcc3) J Pharmacol Exp Ther. 2008;27:465–473. doi: 10.1124/jpet.108.140475. [DOI] [PubMed] [Google Scholar]
- 103.Vlaming ML, Mohrmann K, Wagenaar E, de Waart DR, Elferink RP, Lagas JS, et al. Carcinogen and anticancer drug transport by Mrp2 in vivo: studies using Mrp2 (Abcc2) knockout mice. J Pharmacol Exp Ther. 2006;318:319–327. doi: 10.1124/jpet.106.101774. [DOI] [PubMed] [Google Scholar]
- 104.Vlaming ML, Pala Z, Van EA, Wagenaar E, Van TO, de Waart DR, et al. Impact of Abcc2 (Mrp2) and Abcc3 (Mrp3) on the in-vivo elimination of methotrexate and its main toxic metabolite 7-hydroxymethotrexate. Clin Cancer Res. 2008;14:8152–8160. doi: 10.1158/1078-0432.CCR-08-1609. [DOI] [PubMed] [Google Scholar]
- 105.Vlaming ML, Pala Z, Van EA, Wagenaar E, de Waart DR, Van de WK, et al. Functionally overlapping roles of Abcg2 (Bcrp1) and Abcc2 (Mrp2) in the elimination of methotrexate and its main toxic metabolite 7-hydroxymethotrexate in vivo. Clin Cancer Res. 2009;15:3084–3093. doi: 10.1158/1078-0432.CCR-08-2940. [DOI] [PubMed] [Google Scholar]
- 106.Lîscher W, Potschka H. Role of drug efflux transporters in the brain for drug disposition and treatment of brain diseases. Prog Neurobiol. 2005;76:22–76. doi: 10.1016/j.pneurobio.2005.04.006. [DOI] [PubMed] [Google Scholar]
- 107.Schmiegelow K. Advances in individual prediction of methotrexate toxicity: a review. Br J Haematol. 2009;146:489–503. doi: 10.1111/j.1365-2141.2009.07765.x. [DOI] [PubMed] [Google Scholar]
- 108.Belkov VM, Krynetski EY, Schuetz JD, Yanishevski Y, Masson E, Mathew S, et al. Reduced folate carrier expression in acute lymphoblastic leukemia: a mechanism for ploidy but not lineage differences in methotrexate accumulation. Blood. 1999;93:1643–1650. [PubMed] [Google Scholar]
- 109.Zhang L, Taub JW, Williamson M, Wong SC, Hukku B, Pullen J, et al. Reduced folate carrier gene expression in childhood acute lymphoblastic leukemia: relationship to immunophenotype and ploidy. Clin Cancer Res. 1998;4:2169–2177. [PubMed] [Google Scholar]
- 110.Hooijberg JH, de Vries NA, Kaspers GJ, Pieters R, Jansen G, Peters GJ. Multidrug resistance proteins and folate supplementation: therapeutic implications for antifolates and other classes of drugs in cancer treatment. Cancer Chemother Pharmacol. 2006;58:1–12. doi: 10.1007/s00280-005-0141-1. [DOI] [PubMed] [Google Scholar]
- 111.Barredo JC, Synold TW, Laver J, Relling MV, Pui CH, Priest DG, et al. Differences in constitutive and post-methotrexate folylpolyglutamate synthetase activity in B-lineage and T-lineage leukemia. Blood. 1994;84:564–569. [PubMed] [Google Scholar]
- 112.Gorlick R, Goker E, Trippett T, Waltham M, Banerjee D, Bertino JR. Intrinsic and acquired resistance to methotrexate in acute leukemia. New Eng J Med. 1996;335:1041. doi: 10.1056/NEJM199610033351408. [DOI] [PubMed] [Google Scholar]
- 113.Panetta JC, Wall A, Pui CH, Relling MV, Evans WE. Methotrexate intracellular disposition in acute lymphoblastic leukemia: a mathematical model of gamma-glutamyl hydrolase activity. Clin Cancer Res. 2002;8:2423–2429. [PubMed] [Google Scholar]
- 114.Cheng Q, Yang W, Raimondi SC, Pui CH, Relling MV, Evans WE. Karyotypic abnormalities create discordance of germ line genotype and cancer cell phenotypes. Nat Genet. 2005;37:878–882. doi: 10.1038/ng1612. [DOI] [PubMed] [Google Scholar]
- 115.Cheng Q, Cheng C, Crews KR, Ribeiro RC, Pui CH, Relling MV, et al. Epigenetic regulation of human gamma-glutamyl hydrolase activity in acute lymphoblastic leukemia cells. Am J Hum Genet. 2006;79:264–274. doi: 10.1086/505645. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116.Dervieux T, Furst D, Lein DO, Capps R, Smith K, Walsh M, et al. Polyglutamation of methotrexate with common polymorphisms in reduced folate carrier, aminoimidazole carboxamide ribonucleotide transformylase, and thymidylate synthase are associated with methotrexate effects in rheumatoid arthritis. Arthritis Rheum. 2004;50:2766–2774. doi: 10.1002/art.20460. [DOI] [PubMed] [Google Scholar]
- 117.Becker ML, Gaedigk R, van Haandel L, Thomas B, Lasky A, Hoeltzel M, et al. The effect of genotype on methotrexate polyglutamate variability in juvenile idiopathic arthritis and association with drug response. Arthritis Rheum. 2010 doi: 10.1002/art.30080. Epub ahead of print. [DOI] [PubMed] [Google Scholar]
- 118.Cheng Q, Wu B, Kager L, Panetta JC, Zheng J, Pui CH, et al. A substrate specific functional polymorphism of human [gamma]-glutamyl hydrolase alters catalytic activity and methotrexate polyglutamate accumulation in acute lymphoblastic leukaemia cells. Pharmacogenet Genomics. 2004;14:557. doi: 10.1097/01.fpc.0000114761.78957.7e. [DOI] [PubMed] [Google Scholar]
- 119.Kager L, Cheok M, Yang W, Zaza G, Cheng Q, Panetta JC, et al. Folate pathway gene expression differs in subtypes of acute lymphoblastic leukemia and influences methotrexate pharmacodynamics. J Clin Invest. 2005;115:110–117. doi: 10.1172/JCI22477. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 120.Guo W, Healey JH, Meyers PA, Ladanyi M, Huvos AG, Bertino JR, et al. Mechanisms of methotrexate resistance in osteosarcoma. Clin Cancer Res. 1999;5:621. [PubMed] [Google Scholar]
- 121.Krajinovic M, Lemieux-Blanchard E, Chiasson S, Primeau M, Costea I, Moghrabi A. Role of polymorphisms in MTHFR and MTHFD1 genes in the outcome of childhood acute lymphoblastic leukemia. Pharmacogenomics J. 2003;4:66–72. doi: 10.1038/sj.tpj.6500224. [DOI] [PubMed] [Google Scholar]
- 122.French D, Yang W, Cheng C, Raimondi SC, Mullighan CG, Downing JR, et al. Acquired variation outweighs inherited variation in whole genome analysis of methotrexate polyglutamate accumulation in leukemia. Blood. 2009;113:4512–4520. doi: 10.1182/blood-2008-07-172106. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 123.Sorich MJ, Pottier N, Pei D, Yang W, Kager L, Stocco G, et al. In-vivo response to methotrexate forecasts outcome of acute lymphoblastic leukemia and has a distinct gene expression profile. PloS Med. 2008;5:e83. doi: 10.1371/journal.pmed.0050083. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 124.Yang JJ, Cheng C, Yang W, Pei D, Cao X, Fan Y, et al. Genome-wide interrogation of germ line genetic variation associated with treatment response in childhood acute lymphoblastic leukemia. JAMA. 2009;301:393–403. doi: 10.1001/jama.2009.7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 125.Chan ES, Cronstein BN. Molecular action of methotrexate in inflammatory diseases. Arthritis Res. 2002;4:266–273. doi: 10.1186/ar419. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 126.Hinks A, Moncrieffe H, Martin P, Lal SD, Ursu S, Kassoumeri L, et al. Genetic polymorphisms in key methotrexate pathway genes associated with response to MTX treatment in juvenile idiopathic arthritis. Arthritis Rheum. 2009;60(10 Suppl):S233. [Google Scholar]