We studied whether common polymorphisms in genes involved in folate metabolism affect methotrexate (MTX) sensitivity. Ex vivo MTX sensitivity of lymphoblasts obtained from pediatric patients with acute lymphoblastic leukemia (ALL; n = 157) was determined by the in situ thymidylate synthase inhibition assay after either continuous (21 hours; TSI50, cont) or short-term (3 hours; TSI50, short) MTX exposure. DNA was isolated from lymphoblasts obtained from cytospin slides. Polymorphisms in methylenetetrahydrofolate reductase (MTHFR 677C>T, MTHFR 1298A>C), methionine synthase (MTR 2756A>G), methionine synthase reductase (MTRR 66A>G), methylenetetrahydrofolate dehydrogenase (MTHFD1 1958G>A), serine hydroxymethyl transferase (SHMT1 1420C>T), thymidylate synthase (TS 2R3R), and the reduced folate carrier (RFC 80G>A) were detected by polymerase chain reaction-restriction fragment length polymorphism (PCR-RFLP) or real-time PCR. Patients with the MTHFR 1298AC variant or the MTRR 66 G-allele showed decreased in vitro MTX sensitivity measured under both test conditions. SHMT1 1420TT homozygotes only showed decreased MTX sensitivity in the TSI50, cont. In conclusion, polymorphisms in the folate-related genes MTHFR, MTRR, and SHMT1 are related to MTX resistance in pediatric patients with ALL. (Blood. 2005;106:717-720)

Reduced tetrahydrofolates (THFs) serve as cofactors that carry one-carbon moieties for (1) the synthesis of de novo purines and thymidylate (dTMP) and (2) remethylation of homocysteine to methionine (Figure 1). The antifolate methotrexate (MTX) and its active polyglutamate forms inhibit RNA and DNA synthesis in cells by inhibiting dihydrofolate reductase (DHFR) and thymidylate synthase (TS), respectively, 2 crucial enzymes in pyrimidine synthesis and folate recirculation (Figure 1). MTX is an important chemotherapeutic drug in the treatment of acute lymphoblastic leukemia (ALL) and other malignancies. Recent clinical and in vitro studies indicate that common polymorphisms in genes coding for enzymes involved in folate metabolism affect the sensitivity of patients to folate-based chemotherapeutic drugs such as MTX.1-10  This may be a direct effect or indirect due to shifts in intracellular folate distribution.9,11  We investigated whether common polymorphisms in folate-related genes influence (ex vivo) MTX sensitivity.

From newly diagnosed Western European pediatric patients with ALL, peripheral blood or bone marrow mononuclear cells were isolated with Ficoll Isopaque (> 80% leukemic cell enrichment) and MTX sensitivity was previously measured with the in situ thymidylate synthase inhibition assay (TSI).12,13  In short, leukemia cells were incubated with different concentrations of MTX for 21 hours (TSIcont) or for 3 hours followed by an 18-hour drug-free period (TSIshort), after which the degree of inhibition of TS was measured using [5-3H]-2′-deoxycytidine as substrate.12  TSI data were expressed as the concentration of MTX needed to inhibit 50% of the control TS activity (TSI50). Due to limited cell numbers, of all 157 patients included in this study, the TSI50, cont was measured in 86 patients and the TSI50, short in 120 patients. DNA was obtained from unstained cytospin slides of 157 patients. Informed consent from each patient was obtained according to local procedures in accordance with the Declaration of Helsinki. Genomic DNA was isolated using the QIAmp DNA Mini Blood Kit (Qiagen, Vealo, Netherlands). PCR-RFLP was used to detect polymorphisms in methylenetetrahydrofolate reductase (MTHFR 677C>T, MTHFR 1298A>C),14,15  methionine synthase (MTR 2756A>G),16  serine hydroxymethyl transferase (SHMT1 1420C>T),17  methylenetetrahydrofolate dehydrogenase (MTHFD1 1958G>A),18  and the reduced folate carrier (RFC 80G>A).19  Double (2R2R) or triple (3R3R) 28-bp tandem repeats in the promoter region of the thymidylate synthase gene (TS 2R3R) were visualized on agarose gel directly after the PCR reaction.20  The polymorphism in the methionine synthase reductase gene (MTRR 66A>G) was determined with real-time PCR using hybridization probes and Lightcycler Technology (Roche, Mannheim, Germany). Because TSI data were not normally distributed, ln (natural logarithm)-transformed TSI values were used as outcome measure in univariate and multivariate regression analysis.

For each genetic variant, the median (50th percentile) and interquartile range (25th-75th percentile) for the TSI50, cont and the TSI50, short is presented in Tables 1 and 2. Univariate regression analysis revealed MTHFR 1298AC (P = .01), MTRR 66AG (P = .08), and MTRR 66GG (P = .09) as potential determinants of TSI50, cont; MTHFR 1298AC (P = .03), MTRR 66AG (P < .001), and MTRR 66GG (P = .02) were determinants of TSI50, short. Multiple regression analysis identified MTHFR 1298AC (P = .06), MTRR 66AG (P = .08), and SHMT1 1420TT (P = .04) as determinants of TSI50, cont; determinants of TSI50, short were MTHFR 1298AC (P = .01), MTRR 66AG (P = .001), and MTRR 66GG (P = .03).

Table 1.

Univariate analysis of the effect of polymorphisms in folate-related genes on in vitro MTX sensitivity (TSI50) of childhood ALL



Univariate analysis
TSI50,cont, μM
TSI50,short, μM
Polymorphism
n
Median [IQR]
n
Median [IQR]
MTHFR 677C > T     
CC   47   0.071 [0.041-0.195]   68   0.562 [0.156-4.978]  
CT   31   0.063 [0.038-0.155]   39   0.540 [0.156-3.500]  
TT   8   0.090 [0.037-0.168]   13   1.481 [1.037-3.250]  
MTHFR 1298A > C     
AA   36   0.052 [0.030-0.124]   51   0.600 [0.156-1.488]  
AC   36   0.107 [0.055-0.225]  49   0.904 [0.156-8.656] 
CC   14   0.082 [0.034-0.194]   20   0.479 [0.156-3.449]  
MTR 2756 A > G     
AA   54   0.103 [0.035-0.180]   77   0.600 [0.156-6.059]  
AG   26   0.052 [0.038-0.117]   36   0.661 [0.156-1.795]  
GG   6   0.098 [0.056-0.204]   7   0.906 [0.156-1.460]  
MTRR 66A > G     
AA   23   0.054 [0.038-0.088]   29   0.156 [0.156-0.560]  
AG   37   0.104 [0.038-0.191]§  54   1.260 [0.169-7.658] 
GG   26   0.112 [0.038-0.190]  36   0.984 [0.156-4.802] 
MTHFD1 1958G > A     
GG   26   0.120 [0.033-0.196]   35   0.830 [0.156-2.840]  
GA   48   0.065 [0.041-0.164]   67   0.762 [0.156-5.310]  
AA   12   0.053 [0.036-0.174]   18   0.560 [0.156-2.653]  
SHMT1 1420 C > T     
CC   49   0.063 [0.037-0.161]   74   0.661 [0.156-4.096]  
CT   30   0.067 [0.037-0.182]   35   0.600 [0.156-3.500]  
TT   7   0.133 [0.088-0.235]   11   1.131 [0.410-5.614]  
TS 2R3R     
3R3R   25   0.059 [0.039-0.192]   42   0.951 [0.156-5.255]  
3R2R   42   0.069 [0.035-0.132]   52   0.398 [0.156-2.963]  
2R2R   19   0.151 [0.050-0.181]   25   1.042 [0.178-5.384]  
RFC 80G > A     
GG   30   0.058 [0.034-0.173]   43   0.580 [0.156-1.858]  
GA   43   0.071 [0.036-0.188]   54   0.508 [0.156-3.620]  
AA
 
12
 
0.080 [0.044-0.163]
 
20
 
1.110 [0.190-9.315]
 


Univariate analysis
TSI50,cont, μM
TSI50,short, μM
Polymorphism
n
Median [IQR]
n
Median [IQR]
MTHFR 677C > T     
CC   47   0.071 [0.041-0.195]   68   0.562 [0.156-4.978]  
CT   31   0.063 [0.038-0.155]   39   0.540 [0.156-3.500]  
TT   8   0.090 [0.037-0.168]   13   1.481 [1.037-3.250]  
MTHFR 1298A > C     
AA   36   0.052 [0.030-0.124]   51   0.600 [0.156-1.488]  
AC   36   0.107 [0.055-0.225]  49   0.904 [0.156-8.656] 
CC   14   0.082 [0.034-0.194]   20   0.479 [0.156-3.449]  
MTR 2756 A > G     
AA   54   0.103 [0.035-0.180]   77   0.600 [0.156-6.059]  
AG   26   0.052 [0.038-0.117]   36   0.661 [0.156-1.795]  
GG   6   0.098 [0.056-0.204]   7   0.906 [0.156-1.460]  
MTRR 66A > G     
AA   23   0.054 [0.038-0.088]   29   0.156 [0.156-0.560]  
AG   37   0.104 [0.038-0.191]§  54   1.260 [0.169-7.658] 
GG   26   0.112 [0.038-0.190]  36   0.984 [0.156-4.802] 
MTHFD1 1958G > A     
GG   26   0.120 [0.033-0.196]   35   0.830 [0.156-2.840]  
GA   48   0.065 [0.041-0.164]   67   0.762 [0.156-5.310]  
AA   12   0.053 [0.036-0.174]   18   0.560 [0.156-2.653]  
SHMT1 1420 C > T     
CC   49   0.063 [0.037-0.161]   74   0.661 [0.156-4.096]  
CT   30   0.067 [0.037-0.182]   35   0.600 [0.156-3.500]  
TT   7   0.133 [0.088-0.235]   11   1.131 [0.410-5.614]  
TS 2R3R     
3R3R   25   0.059 [0.039-0.192]   42   0.951 [0.156-5.255]  
3R2R   42   0.069 [0.035-0.132]   52   0.398 [0.156-2.963]  
2R2R   19   0.151 [0.050-0.181]   25   1.042 [0.178-5.384]  
RFC 80G > A     
GG   30   0.058 [0.034-0.173]   43   0.580 [0.156-1.858]  
GA   43   0.071 [0.036-0.188]   54   0.508 [0.156-3.620]  
AA
 
12
 
0.080 [0.044-0.163]
 
20
 
1.110 [0.190-9.315]
 

IQR indicates interquartile range.

P < .01.

P < .05.

§

P = .08.

P < .001.

P = .09.

Table 2.

Multivariate analysis of the effect of polymorphisms in folate-related genes on in vitro MTX sensitivity (TSI50) of childhood ALL



Multivariate analysis
Polymorphism
TSI50,cont, Stand β
TSI50,short, Stand β
MTHFR 677C > T   
CT vs CC   −0.033   −0.043  
TT vs CC   0.044   0.067  
MTHFR 1298A > C   
AC vs AA   0.264*  0.281 
CC vs AA   0.064   0.074  
MTR 2756 A > G   
AG vs AA   −0.100   −0.047  
GG vs AA   −0.005   −0.019  
MTRR 66A > G   
AG vs AA   0.272§  0.399 
GG vs AA   0.204   0.272 
MTHFD1 1958G > A   
GA vs GG   0.012   0.137  
AA vs GG   −0.060   −0.003  
SHMT1 1420 C > T   
CT vs CC   −0.002   0.026  
TT vs CC   0.259  0.096  
TS 2R3R   
3R2R vs 3R3R   −0.088   −0.058  
2R2R vs 3R3R   −0.034   0.012  
RFC 80G > A   
GA vs GG   0.043   −0.003  
AA vs GG
 
0.009
 
0.057
 


Multivariate analysis
Polymorphism
TSI50,cont, Stand β
TSI50,short, Stand β
MTHFR 677C > T   
CT vs CC   −0.033   −0.043  
TT vs CC   0.044   0.067  
MTHFR 1298A > C   
AC vs AA   0.264*  0.281 
CC vs AA   0.064   0.074  
MTR 2756 A > G   
AG vs AA   −0.100   −0.047  
GG vs AA   −0.005   −0.019  
MTRR 66A > G   
AG vs AA   0.272§  0.399 
GG vs AA   0.204   0.272 
MTHFD1 1958G > A   
GA vs GG   0.012   0.137  
AA vs GG   −0.060   −0.003  
SHMT1 1420 C > T   
CT vs CC   −0.002   0.026  
TT vs CC   0.259  0.096  
TS 2R3R   
3R2R vs 3R3R   −0.088   −0.058  
2R2R vs 3R3R   −0.034   0.012  
RFC 80G > A   
GA vs GG   0.043   −0.003  
AA vs GG
 
0.009
 
0.057
 

The standardized regression coefficient of each independent variable is given as standardized β (Stand β) in multivariate analysis. Because independent variables may have different units, the strength of the regression coefficients (β) may not be comparable and therefore the coefficients are standardized to their means (Stand β) for comparison reasons. In univariate regression, the strength of the association between each independent variable and the dependent variable (outcome measure) is looked at without taking into account the other independent variables; in contrast, in multiple linear regression, the strength of each independent variable is adjusted by the other independent variables in the model. No significant interactions were observed in multiple regression analysis for MTHFR 677C>T*MTHFR 1298A>C, MS2756 A>G*MTRR 66A>G, MTHFR 677C>T*MTHFD1 1958G>A, and MTHFR 677C>T*MTHFD1 1958G>A.

*

P = 006.

P < .01.

P < .05.

§

P = 0.08.

P < .001.

Figure 1.

Simplified scheme depicting intracellular metabolism of folates. For clarity reasons, the reaction catalysed by methylenetetrahydrofolate dehydrogenase/methenyltetrahydrofolate cyclohydrolase/formyltetrahydrofolate synthetase (MTHFD1) is not shown; MTHFD1 converts CH2-THF in 3 steps to THF and provides formyl-THF (CHO-THF) for purine synthesis. THF indicates tetrahydrofolate; DHF, dihydrofolate; CH2-THF, methylene-THF; CH3-THF, methyl-THF; SAM, S-adenosylmethionine; SAH, S-adenosylhomocysteine; dTMP, deoxy thymidine monophosphate; dUMP, deoxy uridine monophosphate; TS, thymidylate synthase; MTHFR, methylenetetrahydrofolate reductase; MS, methionine synthase; MTRR, methionine synthase reductase; SHMT, serine-hydroxymethyltransferase; DHFR, dihydrofolate reductase; RFC, reduced folate carrier; MTX-PG, methotrexate-polyglutamate; and NADP, nicotinamide adenine dinucleotide.

Figure 1.

Simplified scheme depicting intracellular metabolism of folates. For clarity reasons, the reaction catalysed by methylenetetrahydrofolate dehydrogenase/methenyltetrahydrofolate cyclohydrolase/formyltetrahydrofolate synthetase (MTHFD1) is not shown; MTHFD1 converts CH2-THF in 3 steps to THF and provides formyl-THF (CHO-THF) for purine synthesis. THF indicates tetrahydrofolate; DHF, dihydrofolate; CH2-THF, methylene-THF; CH3-THF, methyl-THF; SAM, S-adenosylmethionine; SAH, S-adenosylhomocysteine; dTMP, deoxy thymidine monophosphate; dUMP, deoxy uridine monophosphate; TS, thymidylate synthase; MTHFR, methylenetetrahydrofolate reductase; MS, methionine synthase; MTRR, methionine synthase reductase; SHMT, serine-hydroxymethyltransferase; DHFR, dihydrofolate reductase; RFC, reduced folate carrier; MTX-PG, methotrexate-polyglutamate; and NADP, nicotinamide adenine dinucleotide.

Close modal

This is the first report describing the influence on a large population of several genetic polymorphisms in folate-related genes on ex vivo MTX sensitivity. Our results show that patients with the MTRR 66 G-allele and the MTHFR 1298AC variant are less sensitive to MTX in both the TSI50, cont and the TSI50, short. Interestingly, both polymorphisms were also related to hematologic toxicity (manuscript in preparation). The MTHFR 677C>T polymorphism was not related to sensitivity in this study. In fact, published data on the role of MTHFR 677C>T are quite contradictory. Patients carrying the MTHFR 677C>T polymorphism are at a higher risk of developing MTX-related toxicity,2,4,8  whereas in an earlier small pilot study (n = 6), greater MTX sensitivity was observed in the 2 MTHFR 677TT patients.6  The MTHFR 677T1298A haplotype or the MTHFD1 1958A variant, in interaction with the TS 3R repeat, resulted in a decreased event- and disease-free survival in pediatric patients with ALL treated with MTX.10  Sohn et al9  transfected human colon and breast cancer cells with either wild-type or mutant 677T human MTHFR cDNA. Only mutant cells had decreased MTHFR activity, changed intracellular folate distribution, increased TS activity, and decreased chemosensitivity toward MTX. Our study with clinical samples of childhood ALL could not confirm the influence of the MTHFR 677C>T polymorphism on ex vivo MTX sensitivity, although a trend toward decreased sensitivity was observed. Some of the discrepancies might be explained by the different conditions used for the in vitro assays. In our assay, high folate is present in the culture medium, possibly counteracting changed folate redistribution due to polymorphisms in folate genes. The clinical results are possibly explained by changed folate distribution. We have no explanation why only the heterozygous MTHFR 1298AC variant showed decreased MTX sensitivity whereas the homozygous (CC) variant did not.

Also, in other diseases in which MTX is being used, genetic polymorphisms play a role. MTHFR 1298A>C and not MTHFR 677C>T modulated the efficacy of MTX treatment in rheumatoid arthritis patients, whereas MTHFR 677C>T was related to toxicity.3,5  Both TS 2R/3R7,21  and RFC 80G>A21  polymorphisms influenced efficacy and not toxicity of low-dose MTX therapy in patients with rheumatoid arthritis, whereas both MTHFR polymorphisms showed no effect.7 

Our study is the first to report on the MTRR 66A>G and SHMT1 1420C>T polymorphisms as pharmacogenetic determinants of MTX treatment efficacy. Further studies are warranted to evaluate the effect of these polymorphisms on enzyme activity and on intracellular folate redistribution. However, increased risk of neural tube defect pregnancy outcome and cardiovascular disease have been associated with the MTRR 66A>G polymorphism.22 SHMT1 1420CC individuals show increased homocysteine concentrations and lower folate status22  and exhibit an increased ALL risk.17  TS competes with SHMT and MTHFR for methylenetetrahydrofolate.23 MTHFR 677C>T and MTHFR 1298A>C reduce in vitro enzyme activity.14,15 MTHFR 677C>T directs methylenetetrahydrofolate toward purine and pyrimidine synthesis at the expense of methyltetrahydrofolate, and increases catalytic activity of TS.9,11  SHMT mediates the competition between TS and MTHFR for one-carbon units: increased SHMT expression favors DNA synthesis and inhibits homocysteine remethylation and S-adenosylmethionine (SAM) synthesis.24  MTRR maintains adequate levels of methyl-cobalamin, the activated cofactor of MS, and thus maintains MS in its active state. Low MTRR activity prevents MTR-mediated remethylation of homocysteine and, because the MTHFR reaction is irreversible, results in trapping of methyl-tetrahydrofolate with concomitant inhibition of purine and thymidylate biosynthesis.23  We have no explanation why SHMT1 TT patients only showed increased MTX resistance in the TSI50, cont.

In contrast to the TSI50, short, borderline significances were observed for the TSI50, cont in our study. This may be due to the lower number of patients in which the TSI50, cont was measured (n = 86) compared with the TSI50, short (n = 120) with concomitant lower statistical power. More likely, however, accumulation of long-chain MTX polyglutamates is higher after continuous incubations (TSI50, cont), resulting in an increased inhibition capacity of TS. As a consequence, TSI50, cont is less sensitive to changes in folate redistribution due to polymorphisms in other folate-related enzymes. The TSI correlates with conventional cytotoxic assays in leukemic cell lines and the TSI50, short gives a better reflection of clinical MTX sensitivity than the TSI50, cont.12 

In conclusion, we show that the common polymorphisms in the folate-related genes MTHFR, MTRR, and SHMT1 are related to in vitro MTX sensitivity in pediatric patients with ALL. To date, the MTX dose applied is based mostly on the body surface area, while the patient's cellular folate status or pharmacogenetic background25  is not yet routinely taken into account. Identifying predictors of MTX sensitivity may lead to the development of individualized treatment strategies with improved efficacy and reduced toxicity. In some countries, but not The Netherlands, the TPMT polymorphism is used to dose adjust the initial dose of 6-mercaptopurine. We feel that in a similar way polymorphisms in folate-related genes such as MTRR could be used to adjust the initial (not maintenance) MTX dose once it is firmly established that these polymorphisms influence efficacy and/or toxicity. In 500 healthy white subjects, we established high allele frequencies of MTRR 66A>G (55%), MTHFR 1298A>C (31%), and SHMT1 1420C>T (32%). Athough ethnic and geographic variations exist in these polymorphisms, high allele frequencies have also been reported in other ethnic groups and thus, they may significantly contribute to sensitivity and toxicity associated with MTX treatment.

Prepublished online as Blood First Edition Paper, March 29, 2005; DOI 10.1182/blood-2004-12-4941.

The publication costs of this article were defrayed in part by page charge payment. Therefore, and solely to indicate this fact, this article is hereby marked “advertisement” in accordance with 18 U.S.C. section 1734.

1
Jansen G, Mauritz R, Drori S, et al. A structurally altered human reduced folate carrier with increased folic acid transport mediates a novel mechanism of antifolate resistance.
J Biol Chem.
1998
;
273
:
30189
-30198.
2
Ulrich CM, Yasui Y, Storb R, et al. Pharmacogenetics of methotrexate: toxicity among marrow transplantation patients varies with the methylenetetrahydrofolate reductase C677T polymorphism.
Blood.
2001
;
98
:
231
-234.
3
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
-2530.
4
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
-1918.
5
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
-190.
6
Taub JW, Matherly LH, Ravindranath Y, Kaspers GJ, Rots MG, Zantwijk CH. Polymorphisms in methylenetetrahydrofolate reductase and methotrexate sensitivity in childhood acute lymphoblastic leukemia.
Leukemia.
2002
;
16
:
764
-765.
7
Kumagai K, Hiyama K, Oyama T, Maeda H, Kohno N. 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.
8
Toffoli G, Russo A, Innocenti F, et al. Effect of methylenetetrahydrofolate reductase 677C—>T polymorphism on toxicity and homocysteine plasma level after chronic methotrexate treatment of ovarian cancer patients.
Int J Cancer.
2003
;
103
:
294
-299.
9
Sohn KJ, Croxford R, Yates Z, Lucock M, Kim YI. Effect of the methylenetetrahydrofolate reductase C677T polymorphism on chemosensitivity of colon and breast cancer cells to 5-fluorouracil and methotrexate.
J Natl Cancer Inst.
2004
;
96
:
134
-144.
10
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.
11
Bagley PJ, Selhub J. A common mutation in the methylenetetrahydrofolate reductase gene is associated with an accumulation of formylated tetrahydrofolates in red blood cells.
Proc Natl Acad Sci U S A.
1998
;
95
:
13217
-13220.
12
Rots MG, Pieters R, Kaspers GJ, et al. Differential methotrexate resistance in childhood T-versus common/preB-acute lymphoblastic leukemia can be measured by an in situ thymidylate synthase inhibition assay, but not by the MTT assay.
Blood.
1999
;
93
:
1067
-1074.
13
Rots MG, Pieters R, Peters GJ, et al. Circumvention of methotrexate resistance in childhood leukemia subtypes by rationally designed antifolates.
Blood.
1999
;
94
:
3121
-3128.
14
Frosst P, Blom HJ, Milos R, et al. A candidate genetic risk factor for vascular disease: a common mutation in methylenetetrahydrofolate reductase.
Nat Genet.
1995
;
10
:
111
-113.
15
Weisberg I, Tran P, Christensen B, Sibani S, Rozen R. A second genetic polymorphism in methylenetetrahydrofolate reductase (MTHFR) associated with decreased enzyme activity.
Mol Genet Metab.
1998
;
64
:
169
-172.
16
Leclerc D, Campeau E, Goyette P, et al. Human methionine synthase: cDNA cloning and identification of mutations in patients of the cblG complementation group of folate/cobalamin disorders.
Hum Mol Genet.
1996
;
5
:
1867
-1874.
17
Skibola CF, Smith MT, Hubbard A, et al. Polymorphisms in the thymidylate synthase and serine hydroxymethyltransferase genes and risk of adult acute lymphocytic leukemia.
Blood.
2002
;
99
:
3786
-3791.
18
Hol FA, van der Put NM, Geurds MP, et al. Molecular genetic analysis of the gene encoding the trifunctional enzyme MTHFD (methylenetetrahydrofolate-dehydrogenase, methenyltetrahydrofolate-cyclohydrolase, formyltetrahydrofolate synthetase) in patients with neural tube defects.
Clin Genet.
1998
;
53
:
119
-125.
19
Chango A, Emery-Fillon N, de Courcy GP, et al. A polymorphism (80G->A) in the reduced folate carrier gene and its associations with folate status and homocysteinemia.
Mol Genet Metab.
2000
;
70
:
310
-315.
20
Horie N, Aiba H, Oguro K, Hojo H, Takeishi K. Functional analysis and DNA polymorphism of the tandemly repeated sequences in the 5′-terminal regulatory region of the human gene for thymidylate synthase.
Cell Struct Funct.
1995
;
20
:
191
-197.
21
Dervieux T, Furst D, Lein DO, 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.
22
Lievers KJ, Kluijtmans LA, Blom HJ. Genetics of hyperhomocysteinaemia in cardiovascular disease.
Ann Clin Biochem.
2003
;
40
:
46
-59.
23
Stover PJ. Physiology of folate and vitamin B12 in health and disease.
Nutr Rev.
2004
;
62
:
S3
-12; discussion S13.
24
Herbig K, Chiang EP, Lee LR, Hills J, Shane B, Stover PJ. Cytoplasmic serine hydroxymethyltransferase mediates competition between folate-dependent deoxyribonucleotide and S-adenosylmethionine biosyntheses.
J Biol Chem.
2002
;
277
:
38381
-38389.
25
Evans WE, McLeod HL. Pharmacogenomics—drug disposition, drug targets, and side effects.
N Engl J Med.
2003
;
348
:
538
-549.
Sign in via your Institution