Outcomes of methotrexate therapy for psoriasis and relationship to genetic polymorphisms

R B Warren, R L Smith, E Campalani, S Eyre, C H Smith, J N W N Barker, J Worthington, C E M Griffiths, R B Warren, R L Smith, E Campalani, S Eyre, C H Smith, J N W N Barker, J Worthington, C E M Griffiths

Abstract

Background: The use of methotrexate is limited by interindividual variability in response. Previous studies in patients with either rheumatoid arthritis or psoriasis suggest that genetic variation across the methotrexate metabolic pathway might enable prediction of both efficacy and toxicity of the drug.

Objectives: To assess if single nucleotide polymorphisms (SNPs) across four genes that are relevant to methotrexate metabolism [folypolyglutamate synthase (FPGS), gamma-glutamyl hydrolase (GGH), methylenetetrahydrofolate reductase (MTHFR) and 5-aminoimidazole-4-carboxamide ribonucleotide transformylase (ATIC)] are related to treatment outcomes in patients with psoriasis.

Methods: DNA was collected from 374 patients with psoriasis who had been treated with methotrexate. Data were available on individual outcomes to therapy, namely efficacy and toxicity. Haplotype-tagging SNPs (r(2) > 0.8) for the four genes with a minor allele frequency of > 5% were selected from the HAPMAP phase II data. Genotyping was undertaken using the MassARRAY spectrometric method (Sequenom).

Results: There were no significant associations detected between clinical outcomes in patients with psoriasis treated with methotrexate and SNPs in the four genes investigated.

Conclusions: Genetic variation in four key genes relevant to the intracellular metabolism of methotrexate does not appear to predict response to methotrexate therapy in patients with psoriasis.

Figures

Fig 1
Fig 1
Illustration of some of the key enzymes involved in the metabolism of methotrexate (MTX). MTX is transported into the cell via the solute carrier family 19, member 1 (SLC19A1). It can be actively transported out of the cell by the ATP-binding cassette transporters including ATP-binding cassette, subfamily C (CFTR/MRP), member 1–4 (ABCC1-C4) and ATP-binding cassette, subfamily G, member 2 (ABCG2). Within the cell it undergoes polyglutamation (activation) under the enzymic control of folylpolyglutamate synthase (FPGS). This is a dynamic process where glutamate residues can be removed by gamma-glutamyl hydrolase (GGH). In the polyglutamated form MTX inhibits aminoimidazole-4-carboxamide ribonucleotide transformylase (ATIC), which probably accounts for some of its anti-inflammatory effects via an intracellular rise in adenosine. Inhibition of the folate pathway may not be as important to its mechanism of action in psoriasis, but this pathway includes the enzyme 5,10-methylenetetrahydrofolate reductase (MTHFR) which has been subject to a number of MTX pharmacogenetic studies in the past. MTHFR catalyses the conversion of 5,10-methylenetetrahydrofolate 5,10-CH2-THF to 5-methyltetrahydofolate (5-CH3-THF), which is a cosubstrate for homocysteine remethylation. The polyglutamated form of MTX also inhibits thymidylate synthase (TYMS), which converts deoxyuridylate (dUMP) to deoxythymidylate (dTMP) in the de novo pyrimidine biosynthetic pathway. Genes chosen for this investigation are highlighted in red.

References

    1. van Ede AE, Laan RF, Blom HJ, 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;44:2525–30.
    1. Urano W, Taniguchi A, Yamanaka H, et al. Polymorphisms in the methylenetetrahydrofolate reductase gene were associated with both the efficacy and the toxicity of methotrexate used for the treatment of rheumatoid arthritis, as evidenced by single locus and haplotype analyses. Pharmacogenetics. 2002;12:183–90.
    1. Campalani E, Arenas M, Marinaki AM, et al. Polymorphisms in folate, pyrimidine, and purine metabolism are associated with efficacy and toxicity of methotrexate in psoriasis. J Invest Dermatol. 2007;127:1860–7.
    1. Berkun Y, Levartovsky D, Rubinow A, et al. Methotrexate related adverse effects in patients with rheumatoid arthritis are associated with the A1298C polymorphism of the MTHFR gene. Ann Rheum Dis. 2004;63:1227–31.
    1. Wessels JA, de Vries-Bouwstra JK, Heijmans BT, 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;54:1087–95.
    1. Wessels JA, Kooloos WM, De JR, 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;54:2830–9.
    1. Chiusolo P, Reddiconto G, Casorelli I, et al. Preponderance of methylenetetrahydrofolate reductase C677T homozygosity among leukemia patients intolerant to methotrexate. Ann Oncol. 2002;13:1915–18.
    1. Dervieux T, Greenstein N, Kremer J. Pharmacogenomic and metabolic biomarkers in the folate pathway and their association with methotrexate effects during dosage escalation in rheumatoid arthritis. Arthritis Rheum. 2006;54:3095–103.
    1. Dervieux T, Kremer J, Lein DO, et al. Contribution of common polymorphisms in reduced folate carrier and gamma-glutamylhydrolase to methotrexate polyglutamate levels in patients with rheumatoid arthritis. Pharmacogenetics. 2004;14:733–9.
    1. Kumagai K, Hiyama K, Oyama T, et al. Polymorphisms in the thymidylate synthase and methylenetetrahydrofolate reductase genes and sensitivity to the low-dose methotrexate therapy in patients with rheumatoid arthritis. Int J Mol Med. 2003;11:593–600.
    1. Hughes LB, Beasley TM, Patel H, 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;65:1213–18.
    1. Weisman MH, Furst DE, Park GS, et al. Risk genotypes in folate-dependent enzymes and their association with methotrexate-related side effects in rheumatoid arthritis. Arthritis Rheum. 2006;54:607–12.
    1. Hider SL, Bruce IN, Thompson CJ. The pharmacogenetics of methotrexate. Rheumatology. 2007;46:1520–4.
    1. Fredriksson T, Pettersson U. Severe psoriasis––oral therapy with a new retinoid. Dermatologica. 1978;157:238–44.
    1. Purcell S, Neale B, Todd-Brown K, et al. PLINK: a tool set for whole-genome association and population-based linkage analyses. Am J Hum Genet. 2007;81:559–75.

Source: PubMed

Подписаться