You are on page 1of 15

International Journal of Rheumatic Diseases 2017; 20: 526–540

ORIGINAL ARTICLE

Lack of association between MTHFR A1298C polymorphism


and outcome of methotrexate treatment in rheumatoid
arthritis patients: evidence from a systematic review and
meta-analysis
Hongqiong FAN,1 Yanhui LI,2 Li ZHANG,3 Yuying LI1 and Wei LI1
1
Cancer Center and Departments of 2Cardiology and Echocardiography and 3Nephrology, First Hospital of Jilin University,
Changchun, China

Abstract
Objectives: The aim of this study was to evaluate the association of methylene tetrahydrofolate reductase (MTHFR)
gene polymorphism A1298C and methotrexate (MTX) outcome in rheumatoid arthritis (RA) patients.
Methods: We conducted a meta-analysis of the relevant published literature through to May 2016. Odds ratios
(ORs) and 95% confidence intervals (CIs) were calculated using fixed- and random-effect models.
Results: A total of 1325 cases (10 studies) of MTX efficacy and 2777 cases (18 studies) of MTX toxicity in RA
patients were analyzed. Pooled results showed that MTHFR gene A1298C polymorphism was not significantly
related to MTX toxicity or efficacy in RA patients. However, subgroup analysis indicated a significant association
between MTHFR gene A1298C polymorphism and decreased MTX efficacy in the South Asian population (CC
vs. CA + AA: OR = 0.45, 95% CI = 0.23–0.89, P = 0.021). Also, MTHFR gene A1298C polymorphism in the
partial folate supplementation group showed a relationship with decreased MTX efficacy (CC vs. CA + AA:
OR = 0.43, 95% CI = 0.20–0.92, P = 0.029) and toxicity (CC vs. CA + AA: OR = 0.40, 95% CI = 0.17–0.96,
P = 0.04; CC vs. AA: OR = 0.38, 95% CI = 0.16–0.94, P = 0.035).
Conclusions: Overall, our meta-analysis suggested no significant effect of MTHFR gene A1298C polymorphism
on MTX outcome in RA patients. However, due to several limitations of our meta-analysis, the results should be
interpreted cautiously and require further confirmation using high-quality studies.
Key words: meta-analysis, MTHFR, rheumatoid arthritis, SNPs.

BACKGROUND development of therapeutic interventions for RA treat-


ment in the past decade, including biological and tar-
Rheumatoid arthritis (RA) is a chronic inflammatory
geted synthetic disease-modifying anti-rheumatic drugs
joint disease that can cause cartilage and bone damage
(DMARDs), has transformed articular and systemic
and lead to disability.1 As one of the most prevalent
outcomes.1,3 Among these therapeutics, low-dose
chronic inflammatory diseases, RA places a substantial
methotrexate (MTX) (5–25 mg/week), a conventional
burden on both the individuals and society.2 The
synthetic DMARD, is still the ideal and most widely
used DMARD in clinical practice, due to its well-estab-
Correspondence: Yuying Li MD and Wei Li MD, PhD, Cancer lished efficacy and low cost.4,5 However, still 40–60%
Center, First Hospital of Jilin University, 71 Xinmin Street, of RA patients fail to achieve a satisfactory response to
Changchun 10032, Jilin Province, China. MTX and around 15–30% of them even develop
Emails: lyying1001@foxmail.com and drweili@outlook.com
adverse drug effects.6 Thus, the MTX treatment
Hongqiong Fan and Yanhui Li are the co-first authors.

© 2017 Asia Pacific League of Associations for Rheumatology and John Wiley & Sons Australia, Ltd
MTHFR polymorphism and RA

outcomes in RA are not uniform and indicate individ- a statistical association between decreased MTX toxicity
ual-specific variability. Therefore, it is important to and MTHFR A1298C polymorphism in RA patients.
identify predictors of response in the early stage of the Regarding efficacy, three different meta-analyses15,18,19
disease, so that physicians can individualize treatment observed similar results with relatively small numbers
plans. of subjects (692, 743 and 1040, respectively). In the
MTX is an anti-folate drug with anti-inflammatory past 3 years, seven additional studies20–26 have been
and anti-proliferative effects.7 However, the exact mech- conducted to detect the association between MTHFR
anism of its actions and predictors of its treatment A1298C polymorphism and MTX response, and thus, it
response have not been fully characterized. Recently, is necessary to perform a larger updated meta-analysis
several of studies have attempted to identify associa- to confirm this association based on all the studies out
tions between the MTX clinical response in RA and there in the literature.
genetic modifications involved in the MTX action mech-
anism, such as adenosine triphosphate-binding cassette
MATERIALS AND METHODS
(ABC) and reduced folate carrier 1 (RFC-1) involving
MTX membrane transport pathway, and 5,10-methyle- Study selection
netetrahydrofolate reductase (MTHFR), 5-aminoimida- A systematic literature search was conducted to identify
zole-4-carboxamide ribonucleotide formyltransferase/ all the clinical studies evaluating MTHFR A1298C poly-
IMP cyclohydrolase (ATIC), thymidylate synthetase morphism and MTX outcome in RA patients using
(TYMS), g-folypolyglutamate synthetase (FPGS), glu- PubMed, Embase and Cochrane databases, up until
tamyl hydrolase (GGH), dihydrofolate reductase May 2016. The following terms were used to identify
(DHFR) and ADORA2A involving MTX intracellular the relevant studies: rheumatoid arthritis, RA, MTHFR
pathways.8–11 Among these, the MTHFR gene located and methylenetetrahydrofolate reductase. These search
on chromosome 1 (1p36.6) is one of the most critical terms were combined using the Boolean operators
enzymes involved in folate metabolism and DNA syn- ‘AND’ and ‘OR’ in several combinations, without
thesis, which irreversibly catalyzes the conversion of restrictions. All the eligible studies were retrieved, and
5,10-methylenetetrahydrofolate to 5-methyltetrahydro- their bibliographies were checked for additional rele-
folate.12 As MTX indirectly inhibits MTHFR and influ- vant articles independently by two investigators.
ences the regeneration of reduced folate,10 it is
plausible that MTHFR single nucleotide polymorphisms Inclusion and exclusion criteria
(SNPs) might be related to the MTX response in RA The inclusion criteria for the relevant studies was as fol-
patients. lows: (i) study should be focused on the relationship
To date, more than 80 polymorphisms have been between MTHFR A1298C polymorphism and outcome
described in the MTHFR gene, but functional data are (toxicity and/or efficacy) of MTX treatment in RA; (ii)
available for only a few of these variants. The A1298C should be a clinical study; (iii) should use a case–con-
polymorphism (rs1801131) is one of the most preva- trol or cohort design; and (iv) should provide the avail-
lent and extensively studied genotypes and causes a glu- able genotype data, including AA, CA, CC or CA + CC
tamine to alanine replacement at codon 429 of the for MTHFR A1298C polymorphism. However, studies
MTHFR protein, subsequently decreasing the enzymatic were excluded if they were: (i) unpublished data, case
activity.13 reports, conference articles and reviews, (ii) did not
Several meta-analyses have studied associations have available genotype frequencies; (iii) included
between MTHFR A1298C polymorphism and RA out- duplicate data. Disagreements about the study selection
comes, including five meta-analyses of MTX toxicity14–18 were resolved by discussion and consensus among all
and three of its efficacy,15,18,19 through February 2013. the authors.
However, the inconsistent results and relatively small
sample sizes of these meta-analyses, limit their power Data extraction
for identifying the relationship. For instance, the meta- Two investigators reviewed all enrolled studies and
analyses by Fisher et al.,14 Owen et al.15 and Song independently extracted the data. The data extraction
et al.16 regarding toxicity suggested no significant rela- form included the following items: first author, title,
tionship between MTHFR A1298C polymorphism and date of publication, country, ethnicity, gender, mean
MTX toxicity in RA patients. However, the meta-analysis age or age range, disease duration, treatment protocol,
by Spyridopoulou et al.17 demonstrated the presence of dose of MTX, MTX time, folate supplementation rate,

International Journal of Rheumatic Diseases 2017; 20: 526–540 527


H. Fan et al.

dose of folic acid supplementation, follow-up period, RA patients, were found to be eligible for further
measure of MTX toxicity and/or efficacy, and genotypes analysis according to our inclusion criteria (Fig. 1).
of MTHFR A1298C SNP. The main characteristics of these 22 included studies,
published between 2002 and 2015, are summarized
Study outcomes in Table 1.
The two endpoints of efficacy and toxicity of MTX in RA The association between MTHFR A1298C SNP and
patients were studied. The efficacy was evaluated by MTX treatment efficacy in RA was reported in four stud-
improvement in the Disease Activity Score of 28 joints ies,22–24,36 while 12 studies20,21,26,30,32–35,37–40 reported
(DDAS28) based on European League Against Rheuma- an association with toxicity and six studies15,25,27–29,31
tism (EULAR) criteria, MTX dose, American College of provided data for both efficacy and toxicity. Overall, the
Rheumatology (ACR)20 at 6 months guidelines, and data from 10 studies22–25,27–29,31,36 (1325 RA patients)
other rules designed by authors from different laborato- was analyzed for determining the correlation between
ries. The MTX toxicity was evaluated based on all the A1298C polymorphism and efficacy, and 18 stud-
reported MTX-related adverse effects or discontinuation ies15,20,21,25–35,37–40 (2777 RA patients) were included
of MTX due to adverse effects. for the toxicity analysis.
Regarding the details of the studies, eight studies had
Statistical analysis small sample sizes (fewer than 100
The results are reported as odds ratios (ORs) with 95% cases)9,23,24,28,30,31,34,35 whereas 14 studies had large
confidence intervals (CIs). Pooled ORs were calculated sample sizes (more than 100 cases).15,20–22,25–
27,29,32,33,36–38,40
for the following genetic models: dominant model In addition, the studies included
(CC + CA vs. AA); recessive model (CC vs. CA + AA); patients of different ethnicities, with 1115,20,24–
26,30,31,33,34,36,40
homozygous model (CC vs. AA); heterozygous model studying Caucasian populations,
(CA vs. AA); and allele comparison (C vs. A). A P-value five28,29,32,37,38 East Asian populations, two22,23 South
of < 0.05 by Z-test was considered statistically Asian populations, one39 a Jewish population, one35 a
significant. Latin American population and two21,27 mixed-ethni-
Heterogeneity between the studies was assessed using city populations. Nineteen studies20–28,30,31,33–40 pro-
Q test (a P-value of < 0.1 represented statistical signifi- vided the full distribution of AA, CA and CC genotypes,
cance) and I2 test (values of 25%, 50% and 75% repre- and three studies15,29,32 provided the frequencies of AA
sented mild, moderate and severe heterogeneity, and CA + CC genotypes. Based on the criteria used for
respectively). If the effect appeared to be homogeneous toxicity evaluation, 14 studies20,26–35,37–39 analyzed
(P > 0.1, I2 < 50%), the fixed-effect method was used adverse effects of MTX, whereas three studies21,25,40
for analysis; otherwise the random-effect model was analyzed toxicity related to MTX discontinuation. More-
applied. The heterogeneity was also tested by subgroup over, two studies15,24 enrolled patients treated with
analyses, based on ethnicity, folate supplementation, MTX only, whereas 16 studies20,22,25–33,36–40 enrolled
mono-MTX therapy, measure of efficacy and measure of patients treated with mono-MTX or combined therapy
toxicity. (MTX concomitant with DMARDs/NSAIDs/glucocorti-
Sensitivity analyses were performed to identify the coid). Among these 16 studies, two studies25,30 pro-
potential influence of each study on the pooled OR by vided information about genotype frequency of MTHFR
sequentially omitting one individual study at a time. A1298C polymorphism and efficacy, and one30 study
Potential publication bias was assessed by the Egger’s reported toxicity in mono-MTX-treated RA patients. In
weighted linear regression test and Begg’s rank correla- terms of the frequency of patients receiving folic acid,
tion test. A P-value of < 0.05 was considered to indicate 100% of patients in 11 studies15,20,24–27,30,33,34,36,40
statistically significant publication bias. received folic acid, whereas no patients received folic
All statistical analyses were performed using Stata acid in five studies.22,31,32,37,39 Six studies21,23,28,29,35,38
12.0 (StataCorp, College Station, TX, USA) software. provided no information regarding folic acid supple-
mentation. Furthermore, studies were also stratified by
measure of efficacy, with two studies24,25 following
RESULTS
DDAS28/EULAR criteria, one29 following MTX dose,
Study characteristics three28,31,36 following ACR20 at 6 months, and
The initial search identified 116 articles. Among four15,22,23,27 following different laboratory and clinical
these, a total of 22 studies15,20–40 encompassing 3346 judgments.

528 International Journal of Rheumatic Diseases 2017; 20: 526–540


MTHFR polymorphism and RA

Figure 1 Flow chart depicting the screening and selection strategy of relevant studies.

MTHFR A1298C polymorphism and MTX evidence of significant heterogeneity between these
outcome in RA studies.
Efficacy Furthermore, subgroup analyses based on ethnicity,
Ten studies15,22–25,27–29,31,36 involving 1325 RA cases showed that MTHFR A1298C SNP was significantly
addressed the association between the MTHFR A1298C associated with decreased efficacy among the South
SNP and efficacy of MTX in RA patients. Eight stud- Asian population (recessive model [CC vs. CA + AA]:
ies22–25,27,28,31,36 provided frequencies of the full geno- OR = 0.45, 95% CI = 0.23–0.89, P = 0.021; Fig. 2a)
type distributions of AA, CA and CC, whereas two and increased efficacy in the East Asian population
studies15,29 provided frequencies of AA and CA + CC (heterozygous model [CA vs. AA]: OR = 3.37, 95%
genotypes. Thus, the pooled ORs for the dominant CI = 1.14–9.99, P = 0.028; allele comparison [C vs. A]:
model were calculated from 10 studies, while for the OR = 3.02, 95% CI = 1.10–8.31, P = 0.032). However,
other four genetic models, eight studies were analyzed. no associations were observed in Caucasian and mixed-
The analysis of pooled results found no statistical asso- ethnicity populations. In addition, there was no
ciation between A1298C SNP and MTX efficacy, under evidence of significant heterogeneity between studies
any genetic model (Table 2). Moreover, there was no performed in Caucasian and South Asian populations,

International Journal of Rheumatic Diseases 2017; 20: 526–540 529


Table 1 Characteristics of studies included in the meta-analysis

530
First Year Country Ethnicity No. of Female Age, years, Folate Outcome measure Therapy
author subjects (%) range supplementation, method
Toxicity Efficacy
H. Fan et al.

%
Berkun 2004 Israel Jewish 93 82.8 58.7  13.7 58.8 Any adverse EULAR criteria† Mixed
effects
Caliz 2012 Spain Caucasian 468 74 49  13.4 100.0 Discontinuation NA Mixed
of MTX
Chaabane 2015 Tunisia Caucasian 141 79.4 52.8  12.48 100.0 Any adverse NA Mixed
effects
Choe 2012 Korea East Asian 167 95.2 53.9  10.4 NA Any adverse NA Mixed
effects
Davis 2014 America Mixed 319 7.96 68.75  1.89 NA Discontinuation NA NA
of MTX
Ghodke- 2015 India South 322 86 43.8  10.4 84 Any adverse ACR50 at Mixed
Puranik Asian effects† 12 months
Iqbal 2015 Pakistan South 51 86.6 42.87  13.5 NA NA ≥ 50% or NA
Asian reduction in
ESR, Richie index,
number of swollen
joints and
duration of
morning stiffness
at 6 months
Kumagai 2003 Japan East Asian 115 82.6 AE: 59.7  9.8 Non 27.8 Any adverse CRP/MTX dose Mixed
AE: 60.0  11.7 effects (6 mg/week)†
Kurzawski 2007 Poland Caucasian 174 74.1 54.8  11.1 100 NA ACR20 at Mixed
6 months
Mena 2011 Mexico Latin 70 NA NA NA MTX-related liver NA NA
American dysfunction
Owen 2013 British Caucasian 309 NA NA 100 Discontinuation CPR, ESR, and Mono
of MTX clinician MTX
judgments
Plaza-Plaza 2012 Spain Caucasian 53 81.1 54.7  14.37 100 Any adverse NA NA
effects
Salazar 2014 Spain Caucasian 124 81.5 55.62  1.297 100 Discontinuation DDAS28/EULAR Mono
of MTX† criteria MTX
Soukup 2015 Czech Caucasian 120 73.3 58.5  12.6 100 Discontinuation DDAS28/EULAR Mixed‡
of MTX criteria (mono
MTX for
efficacy)

International Journal of Rheumatic Diseases 2017; 20: 526–540


Table 1 (continued)
First Year Country Ethnicity No. of Female Age, years, Folate Outcome measure Therapy
author subjects (%) range supplementation, method
Toxicity Efficacy
%
Stamp 2010 New Caucasian 191 72.7 60.5 (18–84) 100 Any adverse DAS28 ≤ 3.2 Mixed
Zealand effects
Swierkot 2015 Poland Caucasian 240 82 Responders 100 Any adverse DDAS28/ Mixed
54  10; effects EULAR
Non-responders: criteria†
50  11
Taniguchi 2007 Japan East Asian 156 CC: 83.3%, CC 56.1  12.2; CC 22.7; Any adverse MTX dose Mixed
CT + TT: CT + TT 55.8  12.1 CT + TT 40 effects (6 mg/week) at
87.8% 1 year and CRP

International Journal of Rheumatic Diseases 2017; 20: 526–540


Taraborelli 2009 Italy Caucasian 79 81 50  13.9 91 Any adverse ACR20 at Mixed
effects 6 months
Tasbas 2011 Turkey Caucasian 64 82.8 48.7  12.5 100 Any adverse NA Mixed‡
effects
Urano 2002 Japan East Asian 106 87.7 56.7  10.2 NA Any adverse MTX dose Mixed
effects (5 mg)
Wessels 2006 Netherlands Mixed 203 68.8 54.6  13.3 100 Any adverse DAS44 ≤ 2.4 Mixed
affects† at 6 months
Xiao 2010 China East Asian 95 74.2 49.6  14.1 NA Any adverse ACR20 Mixed
effects at 6 months

No genotype data. ‡Provide genotype data for mono-MTX treated RA. DDAS28, improvement in Disease Activity Score; EULAR, European League Against Rheumatism; ACR, American
College of Rheumatology; MTX, methotrexate; NA, not applicable.
MTHFR polymorphism and RA

531
H. Fan et al.

Table 2 Meta-analysis summary of pooled ORs and heterogeneity tests assessing the association of MTHFR A1298C polymor-
phism with methotrexate (MTX) efficacy in rheumatoid arthritis (RA) patients
Genetic models Sub-type No. of Test of association Test of Effect model
studies heterogeneity

OR 95% CI P Ph (%) I2 (%)


Dominant model Overall 10 1.13 0.89–1.44 0.302 0.042 48.5 F
CC + CA versus AA Partial folate supplementation 2 0.99 0.56–1.77 0.985 0.49 0 F
100% folate supplementation 5 0.91 0.68–1.22 0.543 0.419 0 F
Combined therapy 7 1.28 0.82–2.02 0.277 0.031 56.9 R
Mono MTX 3 0.9 0.58–1.38 0.186 0.464 0 F
South Asian 2 1.39 0.71–2.72 0.343 0.258 21.9 F
Caucasian 4 1 0.66–1.51 0.995 0.469 0 F
East Asian 3 2.03 0.82–5.06 0.127 0.022 73.8 R
Mixed-ethnicity 1 0.66 0.38–1.16 0.151 NA NA R
ACR20 at 6 months 3 1.5 0.70–3.22 0.294 0.114 53.9 R
DDAS28/EULAR criteria 2 0.82 0.44–1.55 0.548 0.365 0 F
Recessive model Overall 8 0.86 0.58–1.28 0.469 0.144 35.6 F
CC versus CA + AA Partial folate supplementation 2 0.43 0.20–0.92 0.029 0.479 0 F
100% folate supplementation 4 1.33 0.78–2.27 0.297 0.154 42.9 F
Combined therapy 6 0.95 0.62–1.47 0.835 0.1 45.8 F
Mono MTX 2 1.12 0.07–18.12 0.935 0.083 66.8 R
South Asian 2 0.45 0.23–0.89 0.021 0.504 0 F
Caucasian 4 1.46 0.77–2.78 0.252 0.161 41.8 F
Mixed-ethnicity 1 1.03 0.43–2.47 0.949 NA NA R
East Asian 1 1.54 0.06–38.82 0.794 NA NA R
ACR20 at 6 months 3 1.62 0.74–3.54 0.223 0.789 0 F
DDAS28/EULAR criteria 2 1.22 0.09–16.69 0.88 0.041 76.1 R
Homozygous model Overall 8 1.04 0.66–1.64 0.868 0.263 20.9 F
CC versus AA Partial folate supplementation 2 0.58 0.24–1.40 0.227 0.777 0 R
100% folate supplementation 4 1.16 0.45–2.96 0.759 0.097 52.5 R
Combined therapy 6 1.11 0.68–1.82 0.686 0.292 18.7 F
Mono MTX 2 0.84 0.07–10.85 0.896 0.121 58.4 R
South Asian 2 0.73 0.31–1.71 0.465 0.224 32.5 F
Caucasian 4 1.46 0.73–2.94 0.288 0.129 47.1 F
Mixed-ethnicity 1 0.81 0.32–2.04 0.65 NA NA R
East Asian 1 2.12 0.08–54.08 0.649 NA NA R
ACR20 at 6 months 3 1.79 0.76–4.22 0.182 0.666 0 F
DDAS28/EULAR criteria 2 1.08 0.07–15.75 0.956 0.044 75.4 R
Heterozygous model Overall 8 1.09 0.81–1.47 0.561 0.081 44.6 F
CA versus AA Partial folate supplementation 2 1.25 0.69–2.28 0.458 0.214 35.2 F
100% folate supplementation 4 0.82 0.56–1.20 0.306 0.506 0 F
Combined therapy 6 1.07 0.79–1.46 0.657 0.081 49 F
Mono MTX 2 0.56 0.24–1.32 0.187 0.74 0 F
South Asian 2 1.96 0.97–3.96 0.062 0.346 0 F
Caucasian 4 0.94 0.60–1.45 0.764 0.739 0 F
Mixed-ethnicity 1 0.63 0.35–1.14 0.127 NA NA R
East Asian 1 3.37 1.14–9.99 0.028 NA NA R
ACR20 at 6 months 3 1.43 0.67–3.04 0.355 0.132 50.6 R
DDAS28/EULAR criteria 2 0.78 0.40–1.52 0.466 0.783 0 F
Allele comparison Overall 8 0.99 0.81–1.21 0.951 0.094 42.7 F
C versus A Partial folate supplementation 2 0.77 0.52–1.14 0.18 0.849 0 F
100% folate supplementation 4 1 0.77–1.29 0.997 0.123 48.1 F
Combined therapy 6 1.03 0.83–1.27 0.814 0.081 49.1 F

532 International Journal of Rheumatic Diseases 2017; 20: 526–540


MTHFR polymorphism and RA

Table 2 (continued)
Genetic models Sub-type No. of Test of association Test of Effect model
studies heterogeneity

OR 95% CI P Ph (%) I2 (%)


Mono MTX 2 0.75 0.42–1.35 0.34 0.235 29 F
South Asian 2 0.83 0.56–1.23 0.36 0.382 0 F
Caucasian 4 1.09 0.80–1.47 0.589 0.194 36.4 F
Mixed-ethnicity 1 0.81 0.53–1.22 0.307 NA NA R
East Asian 1 3.02 1.10–8.31 0.032 NA NA R
ACR20 at 6 months 3 1.4 0.78–2.51 0.265 0.128 51.3 R
DDAS28/EULAR criteria 2 0.85 0.37–1.95 0.694 0.1 63.1 R
Data are highlighted in bold if P-value is significant (< 0.05). Ph, P-value of Q test for heterogeneity; NA, not applicable; DDAS28, improvement in
Disease Activity Score; EULAR, European League Against Rheumatism; ACR, American College of Rheumatology; F, fixed-effect model; R, random-
effect model.

whereas the heterogeneity was not assessed for studies on folate supplementation revealed that A1298C SNP
on other ethnic groups due to the small number of was statistically associated with decreased MTX toxicity
studies. Next, subgroup analyses based on folate sup- in the partial folate supplementation group (recessive
plementation showed that MTHFR A1298C SNP was model [CC vs. CA + AA]: OR = 0.40, 95% CI = 0.17–
significantly associated with decreased efficacy in the 0.96, P = 0.04; homozygous model [CC vs. AA]:
partial folate supplementation group (recessive model OR = 0.38, 95% CI = 0.16–0.94, P = 0.035), but not
[CC vs. CA + AA]: OR = 0.43, 95% CI = 0.20–0.92, in the 100% folate supplementation group. Also, we
P = 0.029), but not in the 100% folate supplementa- did not observe any evidence of significant heterogene-
tion group (Fig. 2b). Again, no significant heterogeneity ity between the studies (CC vs. CA + AA: I2 = 39.2%,
was observed between these studies. Furthermore, sub- P = 0.193; CA vs. AA: I2 = 25.3%, P = 0.262). How-
group analyses based on therapy method (mono-MTX ever, stratification based on the treatment method indi-
and combined therapy) and measure of efficacy cated that MTHFR A1298C SNP had a trend of
(DDAS28/EULAR criteria, ACR20 at 6 months) showed association with increased MTX toxicity in the mono-
possible alteration in efficacy, but the data were not sig- MTX group but decreased MTX toxicity in the combined
nificant in any genetic model. The findings of these dif- therapy method group. However, these results did not
ferent meta-analysis and heterogeneity tests are reach significance. Also, there was no evidence of signif-
summarized in Table 2. icant heterogeneity between the studies in these two
subgroups.
Toxicity Furthermore, subgroup analyses based on ethnicity
There were 18 studies analyzed to assess the relation- revealed that A1298C SNP was significantly associated
ship between MTHFR A1298C SNP and MTX treatment with increased toxicity among the Latin American pop-
toxicity. Fifteen studies20,21,25–28,30,31,33–35,37–40 pro- ulation (allele comparison [C vs. A]: OR = 2.75, 95%
vided frequencies of the full genotype distributions of CI = 1.12–6.76, P = 0.027) and mixed ethnicity popu-
AA, CA and CC, whereas three studies15,29,32 provided lation (dominant model [CC + CA vs. AA]: OR = 2.29,
frequencies of AA and CA + CC. The pooled ORs calcu- 95% CI = 1.46–3.61, P < 0.001; heterozygous model
lated for the dominant model were from 18 studies, [CA vs. AA]: OR = 2.24, 95% CI = 1.39–3.60,
and the other four genetic models were analyzed based P = 0.001). In contrast, A1298C SNP was associated
on 15 studies. with decreased toxicity among the Jewish population
The pooled results showed that MTHFR A1298C SNP (recessive model [CC vs. CA + AA]: OR = 0.20, 95%
was not significantly associated with MTX toxicity in CI = 0.05–0.74, P = 0.015; homozygous model [CC vs.
RA, and there was significant heterogeneity between the AA]: OR = 0.19, 95% CI = 0.05–0.73, P = 0.015; allele
studies under dominant and heterozygous models and comparison [C vs. A]: OR = 0.35, 95% CI = 0.18–0.70,
allele comparison (CC + CA vs. AA: I2 = 53.4%, P = 0.003). There was no evidence of significant hetero-
P = 0.004; CA vs. AA: I2 = 53.8%, P = 0.007; C vs. A: geneity between these studies. Further stratification by
I2 = 65.2%, P < 0.001). Next, subgroup analyses based

International Journal of Rheumatic Diseases 2017; 20: 526–540 533


H. Fan et al.

Figure 2 Forest plot of association between the MTHFR A1298C polymorphism and methotrexate (MTX) efficacy in RA patients:
odds ratios (ORs) with 95% confidence intervals (CIs). (a) Subgroup analysis based on different ethnicities: recessive model, CC
versus CA + AA; (b) subgroup analysis based on folate supplementation: recessive model, CC versus CA + AA.

534 International Journal of Rheumatic Diseases 2017; 20: 526–540


MTHFR polymorphism and RA

measure of toxicity indicated that the MTHFR 1298 CC MTHFR A1298C polymorphism was not associated
genotype was associated with increased discontinuation with the efficacy or toxicity of MTX in RA patients.
of MTX in RA patients previously treated with MTX (re- The main reason for this discrepancy between biolog-
cessive model [CC vs. CA + AA]: OR = 1.8, 95% ical and epidemiological findings remains unclear but
CI = 1.07–3.04, P = 0.027). However, MTHFR A1298C may partially be explained as follows. First, several
was associated with decreased adverse effects, but this additional genetic mutations, such as MTHFR C677T,17
did not reach significance. All the results of these meta- ATIC C347G,27 RFC-1 G80A43 and TYMS (50 untrans-
analyses and heterogeneity tests are summarized in lated region repeat element),44 in the coding sequences
Table 3. of key enzymes in the MTX cellular pathway have been
identified, and they likely all affect the efficacy or toxic-
Sensitivity analysis and publication bias ity of MTX. Specifically, Ongaro et al. pointed toward
Sensitivity analysis was performed in all genetic models the existence of linkage disequilibrium between the
for efficacy and toxicity. These results indicated that MTHFR 677T and MTHFR 1298C alleles.45 Soukup
pooled ORs were not significantly affected by the omis- et al. identified a synergistic effect of 677CT and
sion of the individual studies. Therefore, the present 1298AC genotypes on the MTX efficacy in Czech RA
results appeared to be statistically reliable. In addition, patients.25 Choe et al. suggested that the 677C/1298A
the Begg’s rank correlation test and Egger’s linear regres- haplotype was associated with overall MTX toxicity in
sion test did not provide any statistical evidence of pub- Korean RA patients.32 In summary, it appears that link-
lication bias for either the efficacy or toxicity analyses age disequilibrium and other genetic variations can
(Fig. 3a,b). modify the relationship between MTHFR A1298C SNP
and the outcome of MTX treatment in RA patients. In
addition, MTX response in RA is not only determined
DISCUSSION
by genetic factors, but also by clinical factors. Recently
We analyzed the association between MTHFR published studies have identified several clinical factors
A1298C SNP and clinical response to MTX in RA as predictors of MTX outcome in RA, including gender,
patients in our meta-analysis. The results from five age, smoking, disease duration, history of failed
previously published meta-analyses were inconsistent, DMARDs and baseline disease activity.46–49 These non-
likely due to the relatively small sample sizes, which genetic factors may have some influence on the rela-
limited the power to identify this relationship. Thus, tionship between the genetic factors and MTX clinical
in our meta-analysis, we included additional recently outcome in RA patients. We wanted to address these
published studies, including four studies22–25 (448 issues by subgroup analyses but were unable to conduct
RA cases) of MTX efficacy and four studies20,21,25,26 a fully stratified analysis with these variables of non-
(749 RA cases) of MTX toxicity, to undertake the genetic or genetic factors due to a lack of sufficient data
comprehensive meta-analysis of the association, if from the available studies. Therefore, further investiga-
any, and draw a more concrete conclusion. The tion of the polymorphism in the context of specific
pooled results of the current meta-analysis showed non-genetic or genetic factors is required to elucidate
that MTHFR gene A1298C polymorphism was not the effect of MTHFR A1298C SNP on clinical outcome
significantly related to the toxicity and efficacy of following MTX treatment in RA patients.
MTX in RA patients. Our efficacy results were consis- Additionally, our results from the subgroup analyses
tent with the results published by Owen et al.,15 Lee based on ethnicity showed that MTHFR A1298C poly-
et al.18 and Morgan et al.19 and our toxicity results morphism was related with MTX outcome among dif-
were consistent with the results published by Owen ferent populations. However, the power of the
et al.,15, Lee et al.18 and Fisher et al.14 subgroup analysis to produce statistically significant
The MTHFR A1298C polymorphism, first discovered results was limited due to the low number of studies
in 1998, leads to a decrease in MTHFR enzyme activity, enrolled in the analysis (less than three). Furthermore,
which can be further indirectly inhibited by MTX. The the A1298C polymorphism varied between ethnic
homozygous variant carries about 60% of the wild-type groups, as the observed frequency of the 1298C allele
function.13 The influence of MTHFR A1298C polymor- was 34% in Caucasian patients, 21% in Japanese
phism on MTX efficacy and toxicity has been explored patients and 9% in African patients.50 Thus, these
in other diseases, such as acute lymphoblastic leuke- results emphasize the need for future studies to confirm
mia.41,42 However, our meta-analysis revealed that the relationship among different populations.

International Journal of Rheumatic Diseases 2017; 20: 526–540 535


H. Fan et al.

Table 3 Meta-analysis summary of pooled ORs and heterogeneity tests assessing the association of MTHFR A1298C polymor-
phism and methotrexate (MTX) toxicity in rheumatoid arthritis (RA) patients
Genetic models Sub-type No. of Test of association Test of Effect
studies heterogeneity model

OR 95% CI P Ph (%) I2 (%)


Dominant model Overall 18 0.94 0.72–1.22 0.64 0.004 53.4 R
CC + CA versus AA Not 100% folate supplement 4 0.75 0.50–1.14 0.182 0.39 0.4 F
100% folate supplement 8 0.95 0.77–1.19 0.677 0.248 21.9 F
Not mono MTX 14 0.85 0.70–1.03 0.089 0.297 14.3 F
Mono MTX 2 1.7 0.57–5.10 0.341 0.153 51 R
Jewish 1 0.44 0.18–1.06 0.057 NA NA F
Caucasian 9 0.87 0.69–1.09 0.221 0.411 2.9 F
East Asian 5 0.76 0.53–1.08 0.13 0.53 0 F
Mixed-ethnicity 2 2.29 1.46–3.61 <0.001 0.167 47.6 F
Latin American 1 3.58 0.98–13.04 0.053 NA NA F
Any adverse affects 14 0.9 0.73–1.10 0.296 0.097 34.8 F
Discontinuation of MTX 4 1.12 0.54–2.33 0.758 0.002 79.7 R
Recessive model Overall 15 1.14 0.82–1.60 0.436 0.128 30.2 F
CC versus CA + AA Partial folate supplementation 3 0.4 0.17–0.96 0.04 0.193 39.2 F
100% folate supplement 8 1.16 0.73–1.83 0.538 0.565 0 F
Not mono MTX 12 0.93 0.63–1.37 0.701 0.287 16 F
Mono MTX 1 2.57 0.24–28.09 0.439 NA NA F
Jewish 1 0.2 0.05–0.74 0.015 NA NA F
Caucasian 8 1.34 0.80–2.22 0.265 0.656 0 F
East Asian 3 0.89 0.27–2.97 0.856 0.492 0 F
Mixed-ethnicity 2 1.39 0.46–4.22 0.561 0.066 70.4 R
Latin American 1 5 0.63–39.39 0.126 NA NA F
Any adverse affects 12 0.84 0.54–1.31 0.452 0.286 16.1 F
Discontinuation of MTX 3 1.8 1.07–3.04 0.027 0.316 13.1 F
Homozygous model Overall 15 1.21 0.85–1.73 0.282 0.023 46.9 F
CC versus AA Partial folate supplementation 3 0.38 0.16–0.94 0.035 0.262 25.3 F
100% folate supplement 8 1.16 0.72–1.87 0.544 0.52 0 F
Not mono MTX 12 0.91 0.61–1.37 0.658 0.328 11.9 F
Mono MTX 1 4.8 0.38–59.89 0.223 NA NA R
Jewish 1 0.19 0.05–0.73 0.015 NA NA F
Caucasian 8 1.18 0.70–1.98 0.528 0.518 0 F
East Asian 3 0.8 0.23–2.73 0.716 0.589 0 F
Mixed-ethnicity 2 2.2 0.59–8.24 0.242 0.051 73.7 R
Latin American 1 8.75 0.95–80.26 0.055 NA NA F
Any adverse affects 12 0.88 0.55–1.39 0.572 0.19 25.8 F
Discontinuation of MTX 3 1.64 0.54–5.02 0.386 0.037 69.8 R
Heterozygous model Overall 15 0.92 0.68–1.25 0.601 0.007 53.8 R
CA versus AA Partial folate supplementation 3 0.82 0.47–1.42 0.469 0.469 0 F
100% folate supplement 8 0.9 0.71–1.15 0.4 0.197 29 F
Not mono MTX 12 0.84 0.68–1.03 0.097 0.218 23 F
Mono MTX 1 3.6 0.71–18.25 0.122 NA NA R
Jewish 1 0.85 0.30–2.44 0.76 NA NA F
Caucasian 8 0.79 0.61–1.01 0.063 0.506 0 F
East Asian 3 0.65 0.41–1.03 0.064 0.208 36.3 F
Mixed-ethnicity 2 2.24 1.39–3.60 0.001 0.357 0 F
Latin American 1 3.06 0.80–11.76 0.103 NA NA F
Any adverse affects 12 0.91 0.73–1.14 0.409 0.109 35.1 F
Discontinuation of MTX 3 1 0.34–2.96 0.995 0.001 84.7 R

536 International Journal of Rheumatic Diseases 2017; 20: 526–540


MTHFR polymorphism and RA

Table 3 (continued)
Genetic models Sub-type No. of Test of association Test of Effect
studies heterogeneity model

OR 95% CI P Ph (%) I2 (%)


Allele comparison Overall 15 0.94 0.72–1.21 0.613 <0.001 65.2 R
C versus A Partial folate supplementation 3 0.62 0.34–1.13 0.119 0.124 52.1 R
100% folate supplement 8 0.98 0.81–1.17 0.805 0.32 14.1 F
Not mono MTX 12 0.88 0.75–1.03 0.124 0.124 33.3 F
Mono MTX 1 2.63 0.84–8.20 0.095 NA NA R
Jewish 1 0.35 0.18–0.70 0.003 NA NA F
Caucasian 8 0.92 0.76–1.12 0.434 0.368 8 F
East Asian 3 0.73 0.50–1.07 0.108 0.679 0 F
Mixed-ethnicity 2 1.62 0.88–3.00 0.123 0.041 76.1 R
Latin American 1 2.75 1.12–6.76 0.027 NA NA F
Any adverse affects 12 0.88 0.68–1.15 0.347 0.019 51.8 R
Discontinuation of MTX 3 1.12 0.55–2.31 0.752 0.001 84.8 R
Data are highlighted in bold if P-value is significant (< 0.05). Ph, P-value of Q test for heterogeneity; NA, not applicable; F, fixed-effect model; R,
random-effect model.

Another challenge in undertaking this meta-analysis


was the use of inconsistent criteria for the evaluation of
MTX response and toxicity among the enrolled studies.
In the current meta-analysis, there were numbers of
measures for efficacy, including DDAS28/EULAR crite-
ria, ACR20 and MTX dose. For MTX toxicity, different
measures, including discontinuation of MTX and types
of adverse effects categorized according to different toxi-
city criteria, were used in different studies. Furthermore,
the time point of evaluation ranged from 4 weeks to
12 months in the enrolled studies, which could also
influence the evaluation results. Taken together, these
unstandardized outcome criteria can variably impact
the results. Subgroup analysis indicated the altered rela-
tionship between MTHFR A1298C SNP and MTX out-
come in RA, based on measures of toxicity and efficacy,
but the small number of studies for each outcome made
it difficult to confirm the validity of these results.
In addition, different treatment protocols were used
between studies, including mono-MTX, MTX in combi-
nation with DMARDs/NSAIDs/glucocorticoids, and
MTX in combination with folic acid. We know that the
combination of MTX with DMARDs/NSAIDs/glucocor-
ticoids improves the response to treatment, but it also
results in induction of more adverse effects than mono-
MTX treatment. Similarly, folate supplementation has
also been suggested in clinical practice to reduce the risk
of MTX adverse effects.51 This suggests that the combi-
Figure 3 Assessment of publication bias using funnel plot for
nation of drugs may confound the results for the associ-
the association between the MTHFR A1298C polymorphism
and (a) methotrexate (MTX) efficacy (recessive model, CC vs. ation between MTHFR SNP and clinical outcome. To
CA + AA) and (b) MTX toxicity (recessive model, CC vs. get a detailed understanding of this issue, we tried to
CA + AA). perform a subtype analysis based on treatment

International Journal of Rheumatic Diseases 2017; 20: 526–540 537


H. Fan et al.

protocol. However, the results were inconclusive for 3 Smolen JS, Aletaha D (2015) Rheumatoid arthritis therapy
multiple reasons. First, patients were treated with reappraisal: strategies, opportunities and challenges. Nat
mono-MTX treatment in fewer than three studies. Sec- Rev Rheumatol 11, 276–89.
ond, the dose and time of folate supplementation were 4 Kremer JM (2004) Toward a better understanding of
methotrexate. Arthritis Rheum 50, 1370–82.
different between different studies. Also, there were dif-
5 Smolen JS, Landewe R, Breedveld FC et al. (2014) EULAR
ferences in dietary folate intake between studies per-
recommendations for the management of rheumatoid
formed in different populations.52 Thus, further arthritis with synthetic and biological disease-modifying
research is needed to confirm the association between antirheumatic drugs: 2013 update. Ann Rheum Dis 73,
A1298C SNP and outcome of RA patients with mono- 492–509.
MTX therapy. 6 Kooloos WM, Huizinga TW, Guchelaar HJ et al. (2010)
Pharmacogenetics in treatment of rheumatoid arthritis.
Curr Pharm Des 16, 164–75.
CONCLUSION 7 Cronstein BN (2005) Low-dose methotrexate: a mainstay
In summary, the overall results of our meta-analysis in the treatment of rheumatoid arthritis. Pharmacol Rev 57,
showed no association between MTHFR A1298C poly- 163–72.
8 Zhu H, Deng FY, Mo XB et al. (2014) Pharmacogenetics
morphism and outcome in RA patients receiving MTX.
and pharmacogenomics for rheumatoid arthritis respon-
However, multiple factors might have confounded the
siveness to methotrexate treatment: the 2013 update. Phar-
results and limited the power of the current meta-analy- macogenomics 15, 551–66.
sis. Thus, larger, well-designed, prospective studies 9 Zhang LL, Yang S, Wei W et al. (2014) Genetic polymor-
which take into account the described genetic and non- phisms affect efficacy and adverse drug reactions of
genetic factors, will be important to improve our under- DMARDs in rheumatoid arthritis. Pharmacogenet Geomics
standing of the association between MTHFR A1298C 24, 531–8.
SNP and MTX outcome in RA patients. 10 Owen SA, Hider SL, Martin P et al. (2013) Genetic poly-
morphisms in key methotrexate pathway genes are associ-
ated with response to treatment in rheumatoid arthritis
ETHICS APPROVAL patients. Pharmacogenomics J 13, 227–34.
11 Lima A, Bernardes M, Azevedo R et al. (2016) Moving
This article does not contain any studies with animals
toward personalized medicine in rheumatoid arthritis:
or human participants performed directly by any of its
SNPs in methotrexate intracellular pathways are associated
authors. Authors of the included studies have declared with methotrexate therapeutic outcome. Pharmacogenomics
in their published articles that institutional review 17, 1649–74.
boards or ethics committee at each participating site 12 Blount BC, Mack MM, Wehr CM et al. (1997) Folate
approved their protocols. deficiency causes uracil misincorporation into human
DNA and chromosome breakage: implications for cancer
and neuronal damage. Proc Natl Acad Sci USA 94,
INFORMED CONSENT 3290–5.
The authors of this article did not directly involve any 13 van der Put NM, Gabreels F, Stevens EM et al. (1998) A
human subjects; however, the individuals have declared second common mutation in the methylenetetrahydrofo-
late reductase gene: an additional risk factor for neural-
obtaining informed consent from the patients.
tube defects? Am J Hum Genet 62, 1044–51.
14 Fisher MC, Cronstein BN (2009) Metaanalysis of
CONFLICT OF INTEREST methylenetetrahydrofolate reductase (MTHFR) polymor-
phisms affecting methotrexate toxicity. J Rheumatol 36,
The authors declare that they have no conflict of inter- 539–45.
est. 15 Owen SA, Lunt M, Bowes J et al. (2013) MTHFR gene
polymorphisms and outcome of methotrexate treatment
in patients with rheumatoid arthritis: analysis of key poly-
REFERENCES morphisms and meta-analysis of C677T and A1298C
1 Smolen JS, Aletaha D, McInnes IB (2016) Rheumatoid polymorphisms. Pharmacogenomics J 13, 137–47.
arthritis. Lancet 388, 2023–38. 16 Song GG, Bae SC, Lee YH (2014) Association of the
2 Cross M, Smith E, Hoy D et al. (2014) The global burden MTHFR C677T and A1298C polymorphisms with
of rheumatoid arthritis: estimates from the global burden methotrexate toxicity in rheumatoid arthritis: a meta-ana-
of disease 2010 study. Ann Rheum Dis 73, 1316–22. lysis. Clin Rheumatol 33, 1715–24.

538 International Journal of Rheumatic Diseases 2017; 20: 526–540


MTHFR polymorphism and RA

17 Spyridopoulou KP, Dimou NL, Hamodrakas SJ et al. 29 Urano W, Taniguchi A, Yamanaka H et al. (2002) Poly-
(2012) Methylene tetrahydrofolate reductase gene morphisms in the methylenetetrahydrofolate reductase
polymorphisms and their association with methotrexate gene were associated with both the efficacy and the toxic-
toxicity: a meta-analysis. Pharmacogenet Geomics 22, ity of methotrexate used for the treatment of rheumatoid
117–33. arthritis, as evidenced by single locus and haplotype analy-
18 Lee YH, Song GG (2010) Associations between the C677T ses. Pharmacogenetics 12, 183–90.
and A1298C polymorphisms of MTHFR and the efficacy 30 Tasbas O, Borman P, Gurhan Karabulut H et al. (2011)
and toxicity of methotrexate in rheumatoid arthritis: a The frequency of A1298C and C677T polymorphisms of
meta-analysis. Clin Drug Investig 30, 101–8. the methylentetrahydrofolate gene in Turkish patients
19 Morgan MD, Al-Shaarawy N, Martin S et al. (2014) with rheumatoid arthritis: relationship with methotrexate
MTHFR functional genetic variation and methotrexate toxicity. Open Rheumatol J 5, 30–5.
treatment response in rheumatoid arthritis: a meta-analy- 31 Taraborelli M, Andreoli L, Archetti S et al. (2009)
sis. Pharmacogenomics 15, 467–75. Methylenetetrahydrofolate reductase polymorphisms and
20 Chaabane S, Marzouk S, Akrout R et al. (2015) Genetic methotrexate: no association with response to therapy nor
determinants of methotrexate toxicity in tunisian patients with drug-related adverse events in an Italian population
with rheumatoid arthritis: a study of polymorphisms of rheumatic patients. Clin Exp Rheumatol 27, 499–502.
involved in the MTX metabolic pathway. Eur J Drug Metab 32 Taniguchi A, Urano W, Tanaka E et al. (2007) Validation
Pharmacokinet 41, 385–93. of the associations between single nucleotide polymor-
21 Davis LA, Polk B, Mann A et al. (2014) Folic acid pathway phisms or haplotypes and responses to disease-modifying
single nucleotide polymorphisms associated with antirheumatic drugs in patients with rheumatoid arthritis:
methotrexate significant adverse events in United States a proposal for prospective pharmacogenomic study in
veterans with rheumatoid arthritis. Clin Exp Rheumatol 32, clinical practice. Pharmacogenet Geomics 17, 383–90.
324–32. 33 Stamp LK, Chapman PT, O’Donnell JL et al. (2010) Poly-
22 Ghodke-Puranik Y, Puranik AS, Shintre P et al. (2015) morphisms within the folate pathway predict folate con-
Folate metabolic pathway single nucleotide polymor- centrations but are not associated with disease activity in
phisms: a predictive pharmacogenetic marker of rheumatoid arthritis patients on methotrexate. Pharmaco-
methotrexate response in Indian (Asian) patients with genet Geomics 20, 367–76.
rheumatoid arthritis. Pharmacogenomics 16, 2019–34. 34 Plaza-Plaza JC, Aguilera M, Canadas-Garre M et al. (2012)
23 Iqbal MPAA, Mehboobali N, Iqbal K (2015) Lack of asso- Pharmacogenetic polymorphisms contributing to toxicity
ciation between MTHFR gene polymorphisms and induced by methotrexate in the southern Spanish popula-
response to methotrexate treatment in Pakistani patients tion with rheumatoid arthritis. OMICS 16, 589–95.
with rheumatoid arthritis. Pak J Pharm Sci 28, 4. 35 Mena JP, Salazar-Paramo M, Gonzalez-Lopez L et al.
24 Salazar J, Moya P, Altes A et al. (2014) Polymorphisms in (2011) Polymorphisms C677T and A1298C in the
genes involved in the mechanism of action of methotrex- MTHFR gene in Mexican patients with rheumatoid arthri-
ate: are they associated with outcome in rheumatoid tis treated with methotrexate: implication with elevation
arthritis patients? Pharmacogenomics 15, 1079–90. of transaminases. Pharmacogenomics J 11, 287–91.
25 Soukup T, Dosedel M, Pavek P et al. (2015) The impact of 36 Kurzawski M, Pawlik A, Safranow K et al. (2007) 677C>T
C677T and A1298C MTHFR polymorphisms on and 1298A>C MTHFR polymorphisms affect methotrexate
methotrexate therapeutic response in East Bohemian treatment outcome in rheumatoid arthritis. Pharmacoge-
region rheumatoid arthritis patients. Rheumatol Int 35, nomics 8, 1551–9.
1149–61. 37 Kumagai K, Hiyama K, Oyama T et al. (2003) Polymor-
26 Swierkot J, Slezak R, Karpinski P et al. (2015) Associations phisms in the thymidylate synthase and methylenetetrahy-
between single-nucleotide polymorphisms of RFC-1, drofolate reductase genes and sensitivity to the low-dose
GGH, MTHFR, TYMS, and TCII genes and the efficacy and methotrexate therapy in patients with rheumatoid arthri-
toxicity of methotrexate treatment in patients with tis. Int J Mol Med 11, 593–600.
rheumatoid arthritis. Pol Arch Med Wewn 125, 152–61. 38 Choe JY, Lee H, Jung HY et al. (2012) Methylenetetrahy-
27 Wessels JA, de Vries-Bouwstra JK, Heijmans BT et al. drofolate reductase polymorphisms, C677T and A1298C,
(2006) Efficacy and toxicity of methotrexate in early are associated with methotrexate-related toxicities in Kor-
rheumatoid arthritis are associated with single-nucleotide ean patients with rheumatoid arthritis. Rheumatol Int 32,
polymorphisms in genes coding for folate pathway 1837–42.
enzymes. Arthritis Rheum 54, 1087–95. 39 Berkun Y, Levartovsky D, Rubinow A et al. (2004)
28 Xiao H, Xu J, Zhou X et al. (2010) Associations between Methotrexate related adverse effects in patients with
the genetic polymorphisms of MTHFR and outcomes of rheumatoid arthritis are associated with the A1298C poly-
methotrexate treatment in rheumatoid arthritis. Clin Exp morphism of the MTHFR gene. Ann Rheum Dis 63, 1227–
Rheumatol 28, 728–33. 31.

International Journal of Rheumatic Diseases 2017; 20: 526–540 539


H. Fan et al.

40 Caliz R, del Amo J, Balsa A et al. (2012) The C677T poly- the importance of disease duration. Arthritis Rheum 43,
morphism in the MTHFR gene is associated with the toxic- 22–9.
ity of methotrexate in a Spanish rheumatoid arthritis 47 Hoekstra M, van Ede AE, Haagsma CJ et al. (2003) Factors
population. Scand J Rheumatol 41, 10–4. associated with toxicity, final dose, and efficacy of
41 Eissa DS, Ahmed TM (2013) C677T and A1298C poly- methotrexate in patients with rheumatoid arthritis. Ann
morphisms of the methylenetetrahydrofolate reductase Rheum Dis 62, 423–6.
gene: effect on methotrexate-related toxicity in adult acute 48 Hider SL, Silman AJ, Thomson W et al. (2009) Can clini-
lymphoblastic leukaemia. Blood Coagul Fibrinolysis 24, cal factors at presentation be used to predict outcome of
181–8. treatment with methotrexate in patients with early inflam-
42 Yang L, Hu X, Xu L (2012) Impact of methylenetetrahy- matory polyarthritis? Ann Rheum Dis 68, 57–62.
drofolate reductase (MTHFR) polymorphisms on 49 van Ede AE, Laan RF, Rood MJ et al. (2001) Effect of folic
methotrexate-induced toxicities in acute lymphoblastic or folinic acid supplementation on the toxicity and effi-
leukemia: a meta-analysis. Tumour Biol 33, 1445–54. cacy of methotrexate in rheumatoid arthritis: a forty-eight
43 Hayashi H, Tazoe Y, Tsuboi S et al. (2013) A single week, multicenter, randomized, double-blind, placebo-
nucleotide polymorphism of reduced folate carrier 1 pre- controlled study. Arthritis Rheum 44, 1515–24.
dicts methotrexate efficacy in Japanese patients with 50 Rosenberg N, Murata M, Ikeda Y et al. (2002) The fre-
rheumatoid arthritis. Drug Metab Pharmacokinet 28, 164–8. quent 5,10-methylenetetrahydrofolate reductase C677T
44 Szekanecz Z, Mesko B, Poliska S et al. (2013) Pharmacoge- polymorphism is associated with a common haplotype in
netics and pharmacogenomics in rheumatology. Immunol whites, Japanese, and Africans. Am J Hum Genet 70, 758–
Res 56, 325–33. 62.
45 Ongaro A, De Mattei M, Della Porta MG et al. (2009) 51 Harten P (2005) Reducing toxicity of methotrexate with
Gene polymorphisms in folate metabolizing enzymes in folic acid. Z Rheumatol 64, 353–8.
adult acute lymphoblastic leukemia: effects on methotrex- 52 Donin AS, Nightingale CM, Owen CG et al. (2010) Nutri-
ate-related toxicity and survival. Haematologica 94, 1391– tional composition of the diets of South Asian, black Afri-
8. can-Caribbean and white European children in the United
46 Anderson JJ, Wells G, Verhoeven AC et al. (2000) Factors Kingdom: the Child Heart and Health Study in England
predicting response to treatment in rheumatoid arthritis: (CHASE). Br J Nutr 104, 276–85.

540 International Journal of Rheumatic Diseases 2017; 20: 526–540

You might also like