Home Life Sciences The influence of MTHFR genetic polymorphisms on methotrexate therapy in pediatric acute lymphoblastic leukemia
Article Open Access

The influence of MTHFR genetic polymorphisms on methotrexate therapy in pediatric acute lymphoblastic leukemia

  • Yaqing Shen , Zhujun Wang , Fen Zhou EMAIL logo and Runming Jin EMAIL logo
Published/Copyright: November 6, 2021

Abstract

MTHFR is a crucial enzyme in folate metabolism. This study aimed to determine the relationship between MTHFR genetic polymorphism and elimination and toxicities of methotrexate (MTX). To do that, the study enrolled 145 patients diagnosed with acute lymphoblastic leukemia, who received chemotherapy following the Chinese Children’s Cancer Group Acute Lymphoblastic Leukemia (CCCG-ALL)-2015 protocol (clinical trial number: ChiCTR-IPR-14005706). We analyzed the effects of MTHFR C677T and A1298C polymorphisms on MTX elimination and toxicities. Patients with the MTHFR C677T TT genotype could tolerate a significantly higher MTX dose than those with the CC/CT genotype. However, patients with C677T TT genotypes had an increased risk of hypokalemia (1.369 to CC and 1.409 to CT types). The MTX infusion rate in patients with the MTHFR A1298C AC genotype was slightly lower than that in those with CC or AA genotypes. Patients with the A1298C AA genotype had a 1.405-fold higher risk of hepatotoxicity than those with the AC genotype (P > 0.05). There was no significant difference between the prevalence of other toxicities among MTHFR C677T or A1298C genotypes (P > 0.05). Neither MTHFR C677T nor A1298C polymorphisms were significantly associated with delayed MTX clearance. To conclude, MTHFR polymorphisms were not good predictors of MTX-related toxicities.

1 Introduction

Methotrexate (MTX) is a crucial agent in treating pediatric acute lymphoblastic leukemia (ALL). It interferes with folate metabolism [1,2]. Upon entering cells through the reduced folate carrier, MTX competitively inhibits dihydrofolate reductase, which leads to reduced conversion of dihydrofolate to tetrahydrofolate. A deficiency of tetrahydrofolate inhibits the synthesis of DNA, RNA, and protein and exerts the antitumor effect of MTX [2,3] (Figure 1).

Figure 1 
               Simplified scheme of folate metabolism pathway targets of MTX. MTX competitively inhibits DHFR after entering cells through the RFC1. Abbreviations: MTX, methotrexate; MTXPG, methotrexate polyglutamated forms; RFC1, reduced folate carrier; ABCs, ABC family transporters; SLCs, SLC family transporters; DHF, dihydrofolate; THF, tetrahydrofolate; DHFR, dihydrofolate reductase; MTHFR, methylenetetrahydrofolate reductase; dTMP, deoxythymidine monophosphate; dUMP, deoxyuridine monophosphate; TS, Thymidylate synthase; 5-CH-THF, 5-methyltetrahydrofolate; 5,10-CH2-THF, 5,10-methylenetetrahydrofolate.
Figure 1

Simplified scheme of folate metabolism pathway targets of MTX. MTX competitively inhibits DHFR after entering cells through the RFC1. Abbreviations: MTX, methotrexate; MTXPG, methotrexate polyglutamated forms; RFC1, reduced folate carrier; ABCs, ABC family transporters; SLCs, SLC family transporters; DHF, dihydrofolate; THF, tetrahydrofolate; DHFR, dihydrofolate reductase; MTHFR, methylenetetrahydrofolate reductase; dTMP, deoxythymidine monophosphate; dUMP, deoxyuridine monophosphate; TS, Thymidylate synthase; 5-CH-THF, 5-methyltetrahydrofolate; 5,10-CH2-THF, 5,10-methylenetetrahydrofolate.

High-dose methotrexate (HD-MTX) chemotherapy is defined as MTX >500 mg/m2 and is part of the regimen for pediatric ALL [1,2]. High-intensity chemotherapy can improve the overall prognosis of childhood ALL, but can also cause severe toxicities. Therefore, MTX-induced toxicities are a major cause of treatment interruption and lead to an enhanced risk of relapse and even death [4].

The large interindividual and intraindividual variability in MTX pharmacokinetics hampers the prediction of the toxicities caused by MTX treatment [4,5]. In recent years, several studies have suggested that single nucleotide polymorphisms (SNPs) of the key enzymes and transporters in folic-acid metabolism are associated with MTX metabolism and the toxicities caused by MTX [2,3,6].

Methylenetetrahydrofolate reductase (MTHFR) is a key enzyme of the folic-acid metabolic pathway: it reduces 5,10-methyltetrahydrofolate to 5-methyltetrahydrofolate. Under the catalysis of thymidylate synthetase, 5,10-methyltetrahydrofolate changes dUMP to dTMP and participates in DNA synthesis. 5-methyltetrahydrofolate is a single-carbon provider involved in a wide range of methylation reactions in vivo [4,7] (shown in Figure 1). MTX cannot inhibit MTHFR directly, but changes in MTHFR activity affect intracellular folate distribution and methylation of nucleic acids, which may influence the efficacy and toxicities of MTX [4,8,9].

Two genetic polymorphisms can reduce the enzymatic activity of MTHFR. The first genetic polymorphism is rs1801133 (C677T), where the cytosine (C) of the 677th nucleotide mutates to thymine (T) and causes alanine to be replaced by valine [4]. Studies have suggested that the mutant CT genotype and TT genotype have 60 and 30% of enzymatic activity, respectively, compared with that of the wild genotype [10,11]. The other genetic polymorphism is rs1801131 (A1298C), where the adenine (A) of the 1298th nucleotide mutates to C and causes glutamic acid to be replaced by alanine [4]. It has been reported that patients with the CC genotype had 60–68% of enzymatic activity compared with that of the AA type [4,11]. Studies suggested that ALL patients with mutant types (CT/TT) of MTHFR C677T were more sensitive to HD-MTX chemotherapy and were at a higher risk of toxicities compared with individuals with the wild-type (CC) [12,13,14]. Additional studies have shown that the MTHFR A1298C AA genotype was associated with an increased risk of adverse events [4,15]. It has been suggested that the MTX dose should be reduced in patients with high-risk genotypes [6].

Based on previous research reports, MTHFR genetic polymorphism was recommended as one of the predictors of MTX-related toxicity in our hospital in recent years. However, in clinical practice, we found that MTHFR genetic polymorphism seems unlikely to accurately identify patients at risk from serious MTX-related toxicity. Therefore, in this pharmacogenetic study, we investigated the relationship between MTHFR C677T or MTHFR A1298C polymorphisms and the elimination and toxicities of MTX in Chinese children suffering from ALL. Herein, we aimed to determine the practicality of detecting MTHFR genetic polymorphisms.

2 Methods

2.1 Inclusion criteria and patients

The inclusion criteria were patients: (i) who are aged 0–14 years; (ii) who are diagnosed with ALL as the primary disease; (iii) who qualified for treatment according to the Chinese Children’s Cancer Group Acute Lymphoblastic Leukemia (CCCG-ALL)-2015 protocol (clinical trial number: ChiCTR-IPR-14005706); (iv) who were not suffering from another type of malignancy; (iv) who had completed at least one cycle of HD-MTX chemotherapy; (vi) from whom venous blood was collected and tested for MTHFR C677T and MTHFR A1298C polymorphisms; (vii) for whom the chemotherapy data and pharmacokinetic parameters of MTX were known.

A total of 145 patients from Union Hospital from November 2016 to September 2020 fulfilled the inclusion criteria and formed the study cohort.

  1. Informed consent: Informed consent has been obtained from all the parents or guardians of children included in this study.

  2. Ethical approval: The research related to human use has been complied with all the relevant national regulations, institutional policies, and in accordance with the tenets of the Helsinki Declaration and has been approved by the Human Research Ethics Committee of Union Hospital Affiliated to Tongji Medical College (Huazhong University of Science and Technology, Wuhan, China). Ethical permission number: S106-2015.

2.2 Treatment protocol of HD-MTX

Consolidation chemotherapy of CCCG-ALL-2015 consisted of four cycles of HD-MTX every 2 weeks and 6-mercaptopurine once a day given via the oral route. The recommended MTX dose was 3 g/m2 for low-risk (LR) patients and 5 g/m2 for intermediate/high-risk (IR/HR) patients. The MTX dose was reduced according to the creatinine clearance rate upon first exposure to HD-MTX and then adjusted based on the previous MTX concentration at 44 h (C 44h). Ten percent of the full MTX dose was transfused (i.v.) within 30 min, and the remaining amount was pumped continuously for 23.5 h. Patients underwent prehydration at 200 mL/m2/h for 2–4 h or 100 mL/m2/h for > 12 h before infusion and then hydration at 3,000 mL/m2/day for 3 days. Sodium bicarbonate (5%) was given at 100 mL/m2 for 3 days from the day of MTX chemotherapy to maintain urine pH between 7 and 8. Leucovorin rescue was initiated 42 h from the beginning of the MTX infusion at a basic dose of 15 mg/m2/6 h and adjusted according to the MTX concentration. The MTX concentration was monitored every 24 h until it reached < 0.2 µmol/L.

2.3 MTHFR genotyping

Venous blood was collected and tested for MTHFR C677T and A1298C polymorphisms before MTX injection. MTHFR genotyping was performed in the Department of Pharmacy, Union Hospital Affiliated to Tongji Medical College of Huazhong University of Science and Technology, using the PCR-RFLP technique.

2.4 Data collection and definitions

We prospectively collected demographic information, details of HD-MTX chemotherapy (actual dose of MTX, MTX concentration, leucovorin dose), MTHFR polymorphisms, and MTX-related toxicities (e.g., myelosuppression, hepatotoxicity, acute kidney injury, mucositis, neurotoxicity). Patients were followed from the day of diagnosis of ALL to Aug 2021. In order to eliminate the bias caused by MTX dose adjustment and different recommended MTX doses (3 g/m2 for LR patients and 5 g/m2 for IR/HR patients), “MTX infusion rate (=actual MTX dose/recommended dose)” was used instead of “actual MTX dose”. Myelosuppression was represented by the number of days with absolute neutrophil count (ANC) < 1.0 × 109/L. “Delay of MTX elimination” was defined as C 44h > 1.0 µmol/L or C 68h ≥ 0.2 µmol/L. Other toxicities were assessed based on the National Cancer Institute Common Toxicity Criteria v5.0 [16].

2.5 Statistical analyses

Continuous data are described as the median and range or interquartile range. Categorical data are presented as n (%). The Hardy–Weinberg equilibrium was tested for the genetic balance of MTHFR using the chi-square test. Relationships between MTHFR polymorphisms and the pharmacokinetics or toxicity of MTX were analyzed by the chi-square test (categorical data) or nonparametric test (continuous data). The risk factors for toxicities and delay in MTX elimination were analyzed by binary logistic regression. Relapse-free survival (RFS) probabilities were estimated using the Kaplan–Meier method together with the Log-rank test. P < 0.05 was considered significant. Statistical analyses were undertaken using SPSS v25.0 (IBM, Armonk, NY, USA).

3 Results

3.1 Demographic data of patients and genotype distribution

Of the 145 participants (88 male and 57 female) who formed our study, 130 had B-ALL, 15 had T-ALL; 52 were in the LR group; and 93 were in the IR/HR group. The median age was 5.0 years (range, 0.4–14.0 years). They received 576 cycles of HD-MTX chemotherapy.

For MTHFR C677T, the wild-type (CC) was the most common one, accounting for 40.0%, and the heterozygous mutant (CT) and homozygous mutant (TT) accounted for 36.6 and 23.4%, respectively. In the case of MTHFR A1298C, 65.5, 29.0, and 5.5% of patients had wild-type (AA), heterozygous mutant (AC), and homozygous mutant (CC), respectively. The Hardy–Weinberg equilibrium test indicated that the gene frequency in the sample population was in accordance with the genetic balance law and that the sample population was representative (P > 0.05) (Table 1).

Table 1

Relationship between MTHFR polymorphisms and MTX chemotherapy or MTX concentrations

Characteristic C677T P A1298C P
CC CT TT AA AC CC
N (%) 58 (40.0%) 53 (36.6%) 34 (23.4%) 0.75* 95 (65.5%) 42 (29.0%) 8 (5.5%) 0.99*
Age (years) 5.2 (3.4–8.0) 4.3 (3.0–9.0) 6.0 (4.0–9.1) 0.412 6.0 (3.7–9.1) 4.5 (3.0–7.2) 4.3 (1.5–7.0) 0.145
Sex 0.101 0.279
  Male 41 27 20 56 25 7
  Female 17 26 14 39 17 1
Risk group 0.936 0.653
  Low 20 19 13 33 17 2
  Intermediate/high 38 34 21 62 25 6
MTX infusion rate 0.96 (0.7–1.0) 1.0 (0.7–1.0) 1.0 (0.8–1.0) 0.026 1.0 (0.8–1.0) 0.85 (0.7–1.0) 1.0 (0.8–1.0) 0.008
Leucovorin dose (mg/m2) 75 (75–135) 75 (75–195) 75 (75–221) 0.22 75 (75–195) 75 (75–146) 75 (75–75) 0.057
C 44h (μmol/L) 0.61 (0.33–3.38) 0.74 (0.47–1.95) 0.7 (0.37–2.91) 0.822 0.58 (0.39–1.22) 0.51 (0.36–1.01) 0.51 (0.42–0.82) 0.266
  ≤1.0 37 (63.8%) 31 (58.5%) 20 (58.8%) 57 (60.0%) 24 (57.1%) 7 (87.5%)
  >1.0 21 (36.2%) 22 (41.5%) 14 (41.2%) 38 (40.0%) 18 (42.9%) 1 (12.5%)
C 68h (μmol/L) 0.2 (0.1–0.54) 0.21 (0.14–0.55) 0.23 (0.1–0.61) 0.663 0.19 (0.11–0.35) 0.16 (0.10–0.30) 0.18 (0.1–0.25) 0.351
  <0.2 30 (51.7%) 23 (43.4%) 17 (50.0%) 45 (47.4%) 20 (47.6%) 5 (62.5%)
  ≥0.2 28 (48.3%) 30 (56.6%) 17 (50.0%) 50 (52.6%) 22 (52.4%) 3 (37.5%)

*Hardy–Weinberg test.

C 44h, MTX concentration at 44 h; C 68h, MTX concentration at 68 h.

3.2 MTX chemotherapy and MTX elimination

There was no significant difference in the distribution of sex, age, immunotyping, or risk group among all the MTHFR C677T and MTHFR A1298C genotypes (P > 0.05) (Table 1). For MTX chemotherapy and MTX elimination, only the MTX infusion rate (actual MTX dose/recommended dose) was significantly associated with MTHFR polymorphisms (P < 0.05). The median MTX infusion rate of MTHFR C677T CC/CT/TT genotypes was 0.96 (IQR, 0.7–1.0), 1.0 (IQR, 0.7–1.0), and 1.0 (IQR, 0.8–1.0), respectively. The subsequent pairwise nonparametric test showed that the MTX infusion rate for the TT genotype was significantly higher than that of CC and CT genotypes (P = 0.011 and P = 0.019), whereas there was no significant difference between the CC and CT genotypes (P = 0.838).

With regard to MTHFR A1298C, the median MTX infusion rate of the AA/AC/CC genotypes was 1.0 (IQR, 0.8–1.0), 0.85 (IQR, 0.7–1.0), and 1.0 (IQR, 0.8–1.0), respectively. The subsequent pairwise nonparametric test showed that patients with the AC genotype had a higher MTX infusion rate than that of patients with the AA genotype or CC genotype (P AC/AA < 0.001, P AC/CC = 0.001, P AA/CC = 0.111).

Delayed elimination of MTX at 44 and 68 h was noted in 39.3 and 51.7% of cycles, respectively. Neither MTHFR C677T nor MTHFR A1298C polymorphisms were associated with the leucovorin dose or delay of MTX clearance when evaluated by the chi-square test or nonparametric test.

3.3 HD-MTX-induced toxicities

MTX-related toxicities were noted in 80.2% (461/576) cycles and were mainly of grade 1/2. Grade-3/4 toxicity was found in 18.6% (107/576) cycles, of which infection accounted for 60.7%. Myelosuppression was the most common complication, followed by hepatotoxicity (Table 2). Further, 20.1% of cycles had ANC ≤ 1.0 × 109/L lasting >7 days, whereas hepatotoxicity occurred in 37.1% of cycles (5.2% of which were graded 3/4).

Table 2

MTX-related toxicities

Toxicity n %
Myelosuppression
  1–7 days 244 42.4
  >7 days 116 20.1
Length of chemotherapy delay
  1–7 days 17 3.0
  >7 days 8 1.4
Hepatotoxicity grade
  1/2 184 31.9
  3/4 30 5.2
Hypokalemia 89 15.4
Hyperkalemia 12 2.1
Grade of gastrointestinal toxicity
  1/2 32 5.6
  3/4 9 1.6
Infection (grade 3/4) 65 11.3
Nephrotoxicity grade
  1/2 4 0.7
  3/4 5 0.9
Mucositis grade
  1/2 26 4.5
  3/4 7 1.2
Neurotoxicity grade
  1/2 1 0.2
  3/4 9 1.6

3.4 Risk factors for grade-3/4 toxicities and delay in MTX elimination

Predictors with a significant difference in the univariate analysis or identified clinically were added in the binary logistic regression model. Nine predictors were included (Table 3). Being female, IR/HR, and C 44h > 1.0 μmol/L were risk factors for grade-3/4 toxicity (P < 0.05). Age ≥5 years, male sex, IR/HR, and MTX infusion rate ≥0.8 were risk factors for delayed elimination of MTX. Neither MTHFR C677T nor MTHFR A1298C was associated with the prevalence of severe adverse events or MTX elimination (P > 0.05).

Table 3

Risk factors for grade-3/4 toxicities and delay in MTX elimination according to binary logistic regression

Characteristic* Toxicity** C 44h C 68h
P/OR P/OR P/OR
Age (<5 vs ≥5 years) 0.781/0.936 0.021/1.616 0.009/1.638
Sex (female vs male) 0.016/0.558 0.005/1.834 0.002/1.832
Immunophenotype (B-ALL vs T-ALL) 0.766/0.895 0.573/0.837 0.544/1.202
Risk group (LR vs IR/HR) 0.045/1.727 0.000/3.014 0.000/2.324
MTX infusion rate (<0.8 vs ≥0.8) 0.318/1.274 0.001/2.059 0.000/1.993
C 44h (≤1.0 vs >1.0 μmol/L) 0.002/3.017
C 68h (<0.2 vs ≥0.2 μmol/L) 0.900/1.045
C677T
  CC 0.599 0.087 0.118
  CT 0.833/0.944 0.057/1.367 0.095/1.255
  TT 0.328/0.731 0.164/1.480 0.261/1.334
A1298C
  AA 0.177 0.586 0.925
  AC 0.191/0.693 0.976/1.007 0.737/1.076
  CC 0.300/1.639 0.312/0.602 0.892/0.945

*The first entry is the reference group.

**Grade-3/4 toxicities.

C 44h: MTX concentration at 44 h; C 68h, MTX concentration at 68 h.

3.5 Relationship between MTHFR polymorphisms and MTX-related toxicity

The chi-square test was used to detect the difference in the prevalence of MTX-related toxicities among the genotypes tested (Table 4). The MTHFR C677T polymorphism was found to be significantly associated with the prevalence of hypokalemia. Patients with the TT genotype were at a higher risk of hypokalemia than those with the CC type or CT type (P TT/CC = 0.008, ORTT/CC = 1.369, 95% CI: 1.045–1.792; P TT/CT = 0.007, ORTT/CT = 1.409, 95% CI: 1.053–1.884), whereas there was no significant difference between patients with the CC genotype or CT genotype (P = 0.923). None of the other toxicities were significantly associated with the MTHFR C677T polymorphism (P > 0.05).

Table 4

Relationship between MTHFR polymorphisms and MTX-related toxicity

Toxicity C677T P A1298C P
CC (n = 230) CT (n = 212) TT (n = 134) AA (n = 376) AC (n = 168) CC (n = 32)
Myelosuppression 0.623 0.230
  1–7 days 103 86 55 158 69 17
  >7 days 43 49 24 68 41 7
Chemotherapy delay (days) 11 12 2 0.258 14 8 3 0.306
Hepatotoxicity 93 71 50 0.564 153 50 11 0.048***
Hypokalemia 29 27 32 0.009** 66 19 4 0.158
Hyperkalemia 8 3 1 0.147 7 3 2 0.236
Gastrointestinal toxicity 18 11 12 0.358 30 11 0 0.228
Infection 26 25 15 0.178 44 15 7 0.105
Nephrotoxicity 4 1 4 0.428 7 2 0 0.645
Mucositis 9 14 10 0.294 20 11 2 0.843
Neurotoxicity 3 7 0 0.059 7 3 0 0.740

*Including toxicities graded 1/2/3/4.

** P TT/CC = 0.008, ORTT/CC = 1.369, 95% CI: 1.045–1.792; P TT/CT = 0.007, ORTT/CT = 1.409, 95% CI: 1.053–1.884.

*** P AA/AC = 0.015, ORAA/AC = 1.405, 95% CI: 1.060–1.862.

The chi-square test suggested that the MTHFR A1298C polymorphism was associated with an increased risk of hepatotoxicity (P = 0.048). Patients with the AA genotype had a 1.405-fold higher risk of hepatotoxicity than those with the AC genotype. There was no significant relationship between the MTHFR A1298C polymorphism and myelosuppression, chemotherapy delay, neurotoxicity, hyperkalemia, hypokalemia, infection, mucositis, or nephrotoxicity (P > 0.05).

3.6 MTHFR polymorphisms with treatment outcome

As of Aug 2021, the median follow-up time was 31.3 months (range: 10–58). One hundred and seventeen patients survived without events, 14 patients relapsed (1 in the testes, 9 in bone marrow, 1 in central nervous system, 3 combined), 11 patients were lost to follow-up, 1 patient abandoned because of serious adverse events, and 2 patients died. No one died from HD-MTX treatment. To detect the influence of MTHFR polymorphisms on treatment outcomes of pediatric ALL patients, we did both univariate and multivariate analyses. No significant correlation between MTHFR C677T or A1298C and RFS was found. The survival curves drawn by the Kaplan-Meier method are shown in Figure 2.

Figure 2 
                  MTHFR polymorphisms and relapse-free survival. (a) MTHFR C677T and (b) MTHFR A1298C.
Figure 2

MTHFR polymorphisms and relapse-free survival. (a) MTHFR C677T and (b) MTHFR A1298C.

4 Discussion

MTX is a crucial therapeutic agent for the successful treatment of ALL [1]. Since its first clinical trial in 1953 using a mini-dosage, the overall prognosis of childhood ALL has ameliorated [1]. Higher MTX dose increases the cure rate of childhood ALL [17]. However, intensive chemotherapy will increase the incidence of MTX-related toxicity, leading to chemotherapy delay, long-term sequelae, or even death [6]. Even with adequate hydration and aggressive monitoring of MTX concentration when receiving HD-MTX chemotherapy, some patients will experience severe adverse events [6]. It is still a major clinical challenge to identify a predictor of the occurrence of MTX-related toxicity. MTHFR genetic polymorphism is one of the predictors studied. However, the conclusions drawn have been controversial.

Our prospective study enrolled 145 children diagnosed with ALL and treated following the CCCG-ALL-2015 protocol in China. We wished to investigate whether the MTHFR C677T and MTHFR A1298C polymorphisms were correlated with MTX elimination or MTX-related toxicities in the Chinese group.

In the present study, the rate of homozygous mutation of MTHFR C677T and MTHFR A1298C was 23.4 and 5%, whereas the heterozygous mutation rate was 36.6 and 29.0%, respectively. The Hardy–Weinberg equilibrium test indicated that the gene frequency in the sample population was in accordance with the genetic balance law and that the sample population was representative.

In our research, although the C677T polymorphism was not associated directly with a delay in MTX clearance, patients with the TT genotype had a higher MTX infusion rate. This finding suggested that they could tolerate a higher MTX dose and were less likely to experience dose reduction, which was based on a high C 44h previously. This result was similar to data documented by Cwiklinska and colleagues [12], which suggested that patients with the TT genotype achieved significantly lower steady state MTX concentrations. However, Chae et al. [18] postulated that, in patients with the C677T genotype, the MTX dose was adjusted more frequently than in those with CC or CT genotypes. Faganel and coworkers [19] indicated that the MTX clearance in individuals with the TT genotype decreased to 73.8%. Suthandiram et al. [20], Yanagimachi and colleagues [21], EL-Khodary and coworkers [22], Kantar and collaborators [23], and Esmaili et al. [24] came to similar conclusions. Studies have shown that MTHFR was not associated with MTX clearance [14,25,26,27].

Apart from the observation that patients with the TT genotype were at a higher risk of hypokalemia, there was no significant relationship between the MTHFR C677T polymorphism and the prevalence of other MTX-related toxicities. To date, research on the relationship between hypokalemia and MTHFR polymorphisms has not been published. MTX is mainly eliminated by renal excretion. MTX and its metabolites can precipitate and crystallize in renal tubules, leading to acute kidney injury [6]. This may be the cause of abnormal serum potassium. Moreover, insufficient volume and urine acidity are major risk factors for acute kidney injury, so hydration and urine alkalinity are necessary to mitigate kidney damage during HDMTX therapy [6]. Increased potassium excretion due to rapid hydration may also be the cause of hypokalemia. MTX dose is another risk factor for nephrotoxicity [1]. In our research, the results suggested that patients with the TT genotype received a higher MTX dose. Therefore, we hypothesized that although there was no significant relationship between the TT genotype and the prevalence of nephrotoxicity in our study, it would ultimately lead to increased nephrotoxicity and affect potassium metabolism.

Whether the MTHFR C677T polymorphism is related to toxicities is controversial. However, studies have suggested that ALL patients with MTHFR C677T mutant types (CT/TT) were more sensitive to HD-MTX chemotherapy and at a higher risk of toxicities compared with individuals with the wild-type mutant (CC) [12,13,14,22,28,29]. For instance, a prospective study involving 74 cases indicated that the T allele was associated with an increased risk of hematopoietic toxicity. Other studies discovered that mucositis, diarrhea, anemia, hepatotoxicity, or leukemia were more frequent in patients with the mutant genotype than in those with the wild-type [18,19,25,30,31,32]. However, the types of toxicities related to the MTHFR C677T polymorphism differed in those studies. Moreover, some studies found no relationship between MTHFR C677T- and MTX-related toxicities or drew the opposite conclusion [23,33,34,35].

With regard to the MTHFR A1298C polymorphism, we noticed that patients with the AA genotype carried a 1.405-fold higher risk of hepatotoxicity than those with the AC genotype. There was no relationship between the MTHFR polymorphism and myelosuppression, chemotherapy delay, neurotoxicity, hyperkalemia, hypokalemia, infection, mucositis, or nephrotoxicity. The delay in MTX elimination was irrelevant to the MTHFR A1298C gene polymorphism. It has been reported that the C allele was a protective factor of MTX-related toxicities (e.g., hepatotoxicity, neutropenia, mucositis) [19,24,27]. However, studies conducted by Yousef et al. [25], Moulik and colleagues [32], and Eissa and Ahmed [27] suggested that the MTHFR A1298C CC genotype was associated with an increased risk of cytopenia. Other studies concluded that MTHFR A1298C was not related to MTX-related toxicity or delay in MTX elimination [20,30,34,36].

The controversies mentioned above may have been caused by various confounding factors in the research process: population, chemotherapy protocol, sample size, toxicity-assessment method, as well as large interindividual and intraindividual variability in MTX pharmacokinetics [5]. Several factors can affect MTX clearance and the prevalence of MTX-related toxicities, such as age, sex, and MTX dose, as reported in our study. Renal function, amount of hydration, urine pH, dose, and timing of leucovorin administration have also been reported to be the main factors in MTX-related toxicities and a delay in MTX clearance [6]. Therefore, MTHFR polymorphisms are not the main factor leading to toxicity or delay in MTX clearance in the Chinese group, although there may be some relationship between them.

Since altered MTX pharmacokinetics may affect treatment efficacy, we investigated the effect of MTHFR polymorphisms on patients’ RFS. However, there was no significant correlation between MTHFR C677T or A1298C and RFS. He et al. [37] published a systematic review including six studies on the relationship between MTHFR polymorphisms and pediatric ALL outcome in 2014. Two studies showed that there was a higher relapse risk in individuals with the MTHFR 677TT genotype. Nevertheless, Umerez et al. [38] performed a new literature review from 2013 to 2016. Eight studies have analyzed the association between MTHFR C677T polymorphism and pediatric ALL outcome, but none showed a significant association. With regard to MTHFR A1298C, two studies showed significant results but with the opposite effect. All these findings support that MTHFR polymorphisms might not be good outcome predictors for childhood ALL.

5 Conclusion

There was some correlation between MTHFR C677T or MTHFR A1298C polymorphisms and MTX-related toxicities. However, MTHFR polymorphisms were not good predictors of toxicities or a delay in MTX clearance. The role of MTHFR polymorphisms in guiding consolidation chemotherapy is limited.


Yaqing Shen and Zhujun Wang contributed equally to this work.

tel: +86-27-85726762
tel: +86-13971078982

Acknowledgments

We are grateful for support from the Department of Pharmacy, Union Hospital Affiliated to Tongji Medical College, Huazhong University of Science and Technology.

  1. Funding information: The present study was supported by the National Natural Science Foundation of China (81700147, 82070172).

  2. Author contributions: All authors contributed to the article and approved the submitted version. Y.S. and F.Z. designed the research study. Y.S. and Z.W. collected the data and wrote the paper. R.J. and F.Z. guided the writing and critically revised the manuscript. The authors applied the SDC approach for the sequence of authors.

  3. Conflict of interest: The authors state no conflict of interest.

  4. Data availability statement: The datasets generated and/or analyzed during the current study are available from the corresponding author on reasonable request.

References

[1] Wijaya J, Gose T, Schuetz JD. Using pharmacology to squeeze the life out of childhood leukemia, and potential strategies to achieve breakthroughs in medulloblastoma treatment. Pharmacol Rev. 2020;72(3):668–91.10.1124/pr.118.016824Search in Google Scholar

[2] Bleyer WA. The clinical pharmacology of methotrexate: new applications of an old drug. Cancer. 1978;41(1):36–51.10.1002/1097-0142(197801)41:1<36::AID-CNCR2820410108>3.0.CO;2-ISearch in Google Scholar

[3] Giletti A, Esperon P. Genetic markers in methotrexate treatments. Pharmacogenomics J. 2018;18(6):689–703.10.1038/s41397-018-0047-zSearch in Google Scholar

[4] Zahra FT, Nahid NA, Islam MR, Al-Mamun MMA, Apu MNH, Nahar Z, et al. Pharmacogenetic variants in MTHFR gene are significant predictors of methotrexate toxicities in Bangladeshi patients with acute lymphoblastic leukemia. Clin Lymphoma Myeloma Leuk. 2020;20(2):E58–65.10.1016/j.clml.2019.11.020Search in Google Scholar

[5] Evans WE, Crom WR, Abromowitch M, Dodge R, Look AT, Bowman WP, et al. Clinical pharmacodynamics of high-dose methotrexate in acute lymphocytic leukemia. Identification of a relation between concentration and effect. N Engl J Med. 1986;314(8):471–7.10.1056/NEJM198602203140803Search in Google Scholar

[6] Howard SC, McCormick J, Pui CH, Buddington RK, Harvey RD. Preventing and managing toxicities of high-dose methotrexate. Oncologist. 2016;21(12):1471–82.10.1634/theoncologist.2015-0164Search in Google Scholar

[7] Koppen IJ, Hermans FJ, Kaspers GJ. Folate related gene polymorphisms and susceptibility to develop childhood acute lymphoblastic leukaemia. Br J Haematol. 2010;148(1):3–14.10.1111/j.1365-2141.2009.07898.xSearch in Google Scholar

[8] Dulucq S, St-Onge G, Gagné V, Ansari M, Sinnett D, Labuda D, et al. DNA variants in the dihydrofolate reductase gene and outcome in childhood ALL. Blood. 2008;111(7):3692–700.10.1182/blood-2007-09-110593Search in Google Scholar

[9] 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(2):717–20.10.1182/blood-2004-12-4941Search in Google Scholar

[10] Schwahn B, Rozen R. Polymorphisms in the methylenetetrahydrofolate reductase gene: clinical consequences. Am J Pharmacogenomics. 2001;1(3):189–201.10.2165/00129785-200101030-00004Search in Google Scholar

[11] 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(3):169–72.10.1006/mgme.1998.2714Search in Google Scholar PubMed

[12] Cwiklinska M, Czogala M, Kwiecinska K, Madetko-Talowska A, Szafarz M, Pawinska K, et al. Polymorphisms of SLC19A1 80 G > A, MTHFR 677 C > T, and Tandem TS repeats influence pharmacokinetics, acute liver toxicity, and vomiting in children with acute lymphoblastic leukemia treated with high doses of methotrexate. Front Pediatr. 2020;8:307.10.3389/fped.2020.00307Search in Google Scholar PubMed PubMed Central

[13] 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(5):291–300.10.1007/s00277-005-0072-2Search in Google Scholar PubMed

[14] Shimasaki N, Mori T, Samejinia R, Sato R, Shimada IK, 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(2):64–8.10.1097/01.mph.0000198269.61948.90Search in Google Scholar PubMed

[15] Yao P, He X, Zhang R, Tong R, Xiao H. The influence of MTHFR genetic polymorphisms on adverse reactions after methotrexate in patients with hematological malignancies: a meta-analysis. Hematology. 2019;24(1):10–9.10.1080/10245332.2018.1500750Search in Google Scholar PubMed

[16] Institute NC, Institute NC. Common terminology criteria for adverse events (CTCAE) version 5.0. National Cancer Institute; 2017 Nov 27. Available from: https://ctep.cancer.gov/protocolDevelopment/electronic_applications/docs/CTCAE_v5_Quick_Reference_8.5x11.pdf.Search in Google Scholar

[17] Abromowitch M, Ochs J, Pui CH, Kalwinsky D, Rivera GK, Fairclough D, et al. High-dose methotrexate improves clinical outcome in children with acute lymphoblastic leukemia: St. Jude total therapy study X. Med Pediatr Oncol. 1988;16(5):297–303.10.1002/mpo.2950160502Search in Google Scholar PubMed

[18] Chae H, Kim M, Choi SH, Kim SK, Lee JW, Chung NG, et al. Influence of plasma methotrexate level and MTHFR genotype in Korean paediatric patients with acute lymphoblastic leukaemia. J Chemother. 2020;32(5):251–9.10.1080/1120009X.2020.1764280Search in Google Scholar PubMed

[19] Faganel Kotnik B, Grabnar I, Bohanec Grabar P, Dolzan V, Jazbec J. Association of genetic polymorphism in the folate metabolic pathway with methotrexate pharmacokinetics and toxicity in childhood acute lymphoblastic leukaemia and malignant lymphoma. Eur J Clin Pharmacol. 2011;67(10):993–1006.10.1007/s00228-011-1046-zSearch in Google Scholar PubMed

[20] Suthandiram S, Gan GG, Zain SM, Bee PC, Lian LH, Chang KM, et al. Effect of polymorphisms within methotrexate pathway genes on methotrexate toxicity and plasma levels in adults with hematological malignancies. Pharmacogenomics. 2014;15(11):1479–94.10.2217/pgs.14.97Search in Google Scholar PubMed

[21] Yanagimachi M, Goto H, Kaneko T, Naruto T, Sasaki K, Takeuchi M, et al. Influence of pre-hydration and pharmacogenetics on plasma methotrexate concentration and renal dysfunction following high-dose methotrexate therapy. Int J Hematol. 2013;98(6):702–7.10.1007/s12185-013-1464-zSearch in Google Scholar PubMed

[22] EL-Khodary NM, EL-Haggar SM, Eid MA, Ebeid EN. Study of the pharmacokinetic and pharmacogenetic contribution to the toxicity of high-dose methotrexate in children with acute lymphoblastic leukemia. Med Oncol. 2012;29(3):2053–62.10.1007/s12032-011-9997-6Search in Google Scholar PubMed

[23] Kantar M, Kosova B, Cetingul N, Gumus S, Toroslu E, Zafer N, et al. Methylenetetrahydrofolate reductase C677T and A1298C gene polymorphisms and therapy-related toxicity in children treated for acute lymphoblastic leukemia and non-Hodgkin lymphoma. Leuk Lymphoma. 2009;50(6):912–7.10.1080/10428190902893819Search in Google Scholar PubMed

[24] Esmaili MA, Kazemi A, Faranoush M, Mellstedt H, Zaker F, Safa M, et al. Polymorphisms within methotrexate pathway genes: Relationship between plasma methotrexate levels, toxicity experienced and outcome in pediatric acute lymphoblastic leukemia. Iran J Basic Med Sci. 2020;23(6):800–9.Search in Google Scholar

[25] Yousef AM, Farhad R, Alshamaseen D, Alsheikh A, Zawiah M, Kadi T. Folate pathway genetic polymorphisms modulate methotrexate-induced toxicity in childhood acute lymphoblastic leukemia. Cancer Chemother Pharmacol. 2019;83(4):755–62.10.1007/s00280-019-03776-8Search in Google Scholar PubMed

[26] Wang SM, Sun LL, Zeng WX, Wu WS, Zhang GL. Influence of genetic polymorphisms of FPGS, GGH, and MTHFR on serum methotrexate levels in Chinese children with acute lymphoblastic leukemia. Cancer Chemother Pharmacol. 2014;74(2):283–9.10.1007/s00280-014-2507-8Search in Google Scholar PubMed

[27] Eissa DS, Ahmed TM. C677T and A1298C polymorphisms of the methylenetetrahydrofolate reductase gene: effect on methotrexate-related toxicity in adult acute lymphoblastic leukaemia. Blood Coagul Fibrinolysis. 2013;24(2):181–8.10.1097/MBC.0b013e32835b249dSearch in Google Scholar PubMed

[28] Yang L, Hu X, Xu L. Impact of methylenetetrahydrofolate reductase (MTHFR) polymorphisms on methotrexate-induced toxicities in acute lymphoblastic leukemia: a meta-analysis. Tumour Biol. 2012;33(5):1445–54.10.1007/s13277-012-0395-2Search in Google Scholar PubMed

[29] 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(1):231–4.10.1182/blood.V98.1.231Search in Google Scholar

[30] Zhu C, Liu YW, Wang SZ, Li XL, Nie XL, Yu XT, et al. Associations between the C677T and A1298C polymorphisms of MTHFR and the toxicity of methotrexate in childhood malignancies: a meta-analysis. Pharmacogenomics J. 2018;18(3):450–9.10.1038/tpj.2017.34Search in Google Scholar PubMed

[31] Mahmoud LB, Mdhaffar M, Frikha R, Ghozzi H, Hakim A, Sahnoun Z, et al. Use of MTHFR C677T polymorphism and plasma pharmacokinetics to predict methotrexate toxicity in patients with acute lymphoblastic leukemia. Adv Clin Exp Med. 2018;27(8):1061–8.10.17219/acem/69802Search in Google Scholar PubMed

[32] Roy Moulik N, Kumar A, Agrawal S, Awasthi S, Mahdi AA, Kumar A. Role of folate status and methylenetetrahydrofolate reductase genotype on the toxicity and outcome of induction chemotherapy in children with acute lymphoblastic leukemia. Leuk Lymphoma. 2015;56(5):1379–84.10.3109/10428194.2014.947608Search in Google Scholar PubMed

[33] Ramirez-Pacheco A, Moreno-Guerrero S, Alamillo I, Medina-Sanson A, Lopez B, Moreno-Galvan M. Mexican childhood acute lymphoblastic leukemia: a pilot study of the MDR1 and MTHFR gene polymorphisms and their associations with clinical outcomes. Genet Test Mol Biomarkers. 2016;20(10):597–602.10.1089/gtmb.2015.0287Search in Google Scholar PubMed

[34] Erculj N, Kotnik BF, Debeljak M, Jazbec J, Dolzan V. Influence of folate pathway polymorphisms on high-dose methotrexate-related toxicity and survival in childhood acute lymphoblastic leukemia. Leuk Lymphoma. 2012;53(6):1096–104.10.3109/10428194.2011.639880Search in Google Scholar PubMed

[35] 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(16):3084–91.10.1200/JCO.2003.07.056Search in Google Scholar PubMed

[36] Lopez-Lopez E, Martin-Guerrero I, Ballesteros J, Garcia-Orad A. A systematic review and meta-analysis of MTHFR polymorphisms in methotrexate toxicity prediction in pediatric acute lymphoblastic leukemia. Pharmacogenomics J. 2013;13(6):498–506.10.1038/tpj.2012.44Search in Google Scholar PubMed

[37] He HR, Liu P, He GH, Dong WH, Wang MY, Dong YL, et al. Association between reduced folate carrier G80A polymorphism and methotrexate toxicity in childhood acute lymphoblastic leukemia: a meta-analysis. Leuk Lymphoma. 2014;55(12):2793–800.10.3109/10428194.2014.898761Search in Google Scholar PubMed

[38] Umerez M, Gutierrez-Camino Á, Muñoz-Maldonado C, Martin-Guerrero I, Garcia-Orad A. MTHFR polymorphisms in childhood acute lymphoblastic leukemia: influence on methotrexate therapy. Pharmgenomics Pers Med. 2017;10:69–78.10.2147/PGPM.S107047Search in Google Scholar PubMed PubMed Central

Received: 2021-07-13
Revised: 2021-09-15
Accepted: 2021-10-11
Published Online: 2021-11-06

© 2021 Yaqing Shen et al., published by De Gruyter

This work is licensed under the Creative Commons Attribution 4.0 International License.

Articles in the same Issue

  1. Biomedical Sciences
  2. Research progress on the mechanism of orexin in pain regulation in different brain regions
  3. Adriamycin-resistant cells are significantly less fit than adriamycin-sensitive cells in cervical cancer
  4. Exogenous spermidine affects polyamine metabolism in the mouse hypothalamus
  5. Iris metastasis of diffuse large B-cell lymphoma misdiagnosed as primary angle-closure glaucoma: A case report and review of the literature
  6. LncRNA PVT1 promotes cervical cancer progression by sponging miR-503 to upregulate ARL2 expression
  7. Two new inflammatory markers related to the CURB-65 score for disease severity in patients with community-acquired pneumonia: The hypersensitive C-reactive protein to albumin ratio and fibrinogen to albumin ratio
  8. Circ_0091579 enhances the malignancy of hepatocellular carcinoma via miR-1287/PDK2 axis
  9. Silencing XIST mitigated lipopolysaccharide (LPS)-induced inflammatory injury in human lung fibroblast WI-38 cells through modulating miR-30b-5p/CCL16 axis and TLR4/NF-κB signaling pathway
  10. Protocatechuic acid attenuates cerebral aneurysm formation and progression by inhibiting TNF-alpha/Nrf-2/NF-kB-mediated inflammatory mechanisms in experimental rats
  11. ABCB1 polymorphism in clopidogrel-treated Montenegrin patients
  12. Metabolic profiling of fatty acids in Tripterygium wilfordii multiglucoside- and triptolide-induced liver-injured rats
  13. miR-338-3p inhibits cell growth, invasion, and EMT process in neuroblastoma through targeting MMP-2
  14. Verification of neuroprotective effects of alpha-lipoic acid on chronic neuropathic pain in a chronic constriction injury rat model
  15. Circ_WWC3 overexpression decelerates the progression of osteosarcoma by regulating miR-421/PDE7B axis
  16. Knockdown of TUG1 rescues cardiomyocyte hypertrophy through targeting the miR-497/MEF2C axis
  17. MiR-146b-3p protects against AR42J cell injury in cerulein-induced acute pancreatitis model through targeting Anxa2
  18. miR-299-3p suppresses cell progression and induces apoptosis by downregulating PAX3 in gastric cancer
  19. Diabetes and COVID-19
  20. Discovery of novel potential KIT inhibitors for the treatment of gastrointestinal stromal tumor
  21. TEAD4 is a novel independent predictor of prognosis in LGG patients with IDH mutation
  22. circTLK1 facilitates the proliferation and metastasis of renal cell carcinoma by regulating miR-495-3p/CBL axis
  23. microRNA-9-5p protects liver sinusoidal endothelial cell against oxygen glucose deprivation/reperfusion injury
  24. Long noncoding RNA TUG1 regulates degradation of chondrocyte extracellular matrix via miR-320c/MMP-13 axis in osteoarthritis
  25. Duodenal adenocarcinoma with skin metastasis as initial manifestation: A case report
  26. Effects of Loofah cylindrica extract on learning and memory ability, brain tissue morphology, and immune function of aging mice
  27. Recombinant Bacteroides fragilis enterotoxin-1 (rBFT-1) promotes proliferation of colorectal cancer via CCL3-related molecular pathways
  28. Blocking circ_UBR4 suppressed proliferation, migration, and cell cycle progression of human vascular smooth muscle cells in atherosclerosis
  29. Gene therapy in PIDs, hemoglobin, ocular, neurodegenerative, and hemophilia B disorders
  30. Downregulation of circ_0037655 impedes glioma formation and metastasis via the regulation of miR-1229-3p/ITGB8 axis
  31. Vitamin D deficiency and cardiovascular risk in type 2 diabetes population
  32. Circ_0013359 facilitates the tumorigenicity of melanoma by regulating miR-136-5p/RAB9A axis
  33. Mechanisms of circular RNA circ_0066147 on pancreatic cancer progression
  34. lncRNA myocardial infarction-associated transcript (MIAT) knockdown alleviates LPS-induced chondrocytes inflammatory injury via regulating miR-488-3p/sex determining region Y-related HMG-box 11 (SOX11) axis
  35. Identification of circRNA circ-CSPP1 as a potent driver of colorectal cancer by directly targeting the miR-431/LASP1 axis
  36. Hyperhomocysteinemia exacerbates ischemia-reperfusion injury-induced acute kidney injury by mediating oxidative stress, DNA damage, JNK pathway, and apoptosis
  37. Potential prognostic markers and significant lncRNA–mRNA co-expression pairs in laryngeal squamous cell carcinoma
  38. Gamma irradiation-mediated inactivation of enveloped viruses with conservation of genome integrity: Potential application for SARS-CoV-2 inactivated vaccine development
  39. ADHFE1 is a correlative factor of patient survival in cancer
  40. The association of transcription factor Prox1 with the proliferation, migration, and invasion of lung cancer
  41. Is there a relationship between the prevalence of autoimmune thyroid disease and diabetic kidney disease?
  42. Immunoregulatory function of Dictyophora echinovolvata spore polysaccharides in immunocompromised mice induced by cyclophosphamide
  43. T cell epitopes of SARS-CoV-2 spike protein and conserved surface protein of Plasmodium malariae share sequence homology
  44. Anti-obesity effect and mechanism of mesenchymal stem cells influence on obese mice
  45. Long noncoding RNA HULC contributes to paclitaxel resistance in ovarian cancer via miR-137/ITGB8 axis
  46. Glucocorticoids protect HEI-OC1 cells from tunicamycin-induced cell damage via inhibiting endoplasmic reticulum stress
  47. Prognostic value of the neutrophil-to-lymphocyte ratio in acute organophosphorus pesticide poisoning
  48. Gastroprotective effects of diosgenin against HCl/ethanol-induced gastric mucosal injury through suppression of NF-κβ and myeloperoxidase activities
  49. Silencing of LINC00707 suppresses cell proliferation, migration, and invasion of osteosarcoma cells by modulating miR-338-3p/AHSA1 axis
  50. Successful extracorporeal membrane oxygenation resuscitation of patient with cardiogenic shock induced by phaeochromocytoma crisis mimicking hyperthyroidism: A case report
  51. Effects of miR-185-5p on replication of hepatitis C virus
  52. Lidocaine has antitumor effect on hepatocellular carcinoma via the circ_DYNC1H1/miR-520a-3p/USP14 axis
  53. Primary localized cutaneous nodular amyloidosis presenting as lymphatic malformation: A case report
  54. Multimodal magnetic resonance imaging analysis in the characteristics of Wilson’s disease: A case report and literature review
  55. Therapeutic potential of anticoagulant therapy in association with cytokine storm inhibition in severe cases of COVID-19: A case report
  56. Neoadjuvant immunotherapy combined with chemotherapy for locally advanced squamous cell lung carcinoma: A case report and literature review
  57. Rufinamide (RUF) suppresses inflammation and maintains the integrity of the blood–brain barrier during kainic acid-induced brain damage
  58. Inhibition of ADAM10 ameliorates doxorubicin-induced cardiac remodeling by suppressing N-cadherin cleavage
  59. Invasive ductal carcinoma and small lymphocytic lymphoma/chronic lymphocytic leukemia manifesting as a collision breast tumor: A case report and literature review
  60. Clonal diversity of the B cell receptor repertoire in patients with coronary in-stent restenosis and type 2 diabetes
  61. CTLA-4 promotes lymphoma progression through tumor stem cell enrichment and immunosuppression
  62. WDR74 promotes proliferation and metastasis in colorectal cancer cells through regulating the Wnt/β-catenin signaling pathway
  63. Down-regulation of IGHG1 enhances Protoporphyrin IX accumulation and inhibits hemin biosynthesis in colorectal cancer by suppressing the MEK-FECH axis
  64. Curcumin suppresses the progression of gastric cancer by regulating circ_0056618/miR-194-5p axis
  65. Scutellarin-induced A549 cell apoptosis depends on activation of the transforming growth factor-β1/smad2/ROS/caspase-3 pathway
  66. lncRNA NEAT1 regulates CYP1A2 and influences steroid-induced necrosis
  67. A two-microRNA signature predicts the progression of male thyroid cancer
  68. Isolation of microglia from retinas of chronic ocular hypertensive rats
  69. Changes of immune cells in patients with hepatocellular carcinoma treated by radiofrequency ablation and hepatectomy, a pilot study
  70. Calcineurin Aβ gene knockdown inhibits transient outward potassium current ion channel remodeling in hypertrophic ventricular myocyte
  71. Aberrant expression of PI3K/AKT signaling is involved in apoptosis resistance of hepatocellular carcinoma
  72. Clinical significance of activated Wnt/β-catenin signaling in apoptosis inhibition of oral cancer
  73. circ_CHFR regulates ox-LDL-mediated cell proliferation, apoptosis, and EndoMT by miR-15a-5p/EGFR axis in human brain microvessel endothelial cells
  74. Resveratrol pretreatment mitigates LPS-induced acute lung injury by regulating conventional dendritic cells’ maturation and function
  75. Ubiquitin-conjugating enzyme E2T promotes tumor stem cell characteristics and migration of cervical cancer cells by regulating the GRP78/FAK pathway
  76. Carriage of HLA-DRB1*11 and 1*12 alleles and risk factors in patients with breast cancer in Burkina Faso
  77. Protective effect of Lactobacillus-containing probiotics on intestinal mucosa of rats experiencing traumatic hemorrhagic shock
  78. Glucocorticoids induce osteonecrosis of the femoral head through the Hippo signaling pathway
  79. Endothelial cell-derived SSAO can increase MLC20 phosphorylation in VSMCs
  80. Downregulation of STOX1 is a novel prognostic biomarker for glioma patients
  81. miR-378a-3p regulates glioma cell chemosensitivity to cisplatin through IGF1R
  82. The molecular mechanisms underlying arecoline-induced cardiac fibrosis in rats
  83. TGF-β1-overexpressing mesenchymal stem cells reciprocally regulate Th17/Treg cells by regulating the expression of IFN-γ
  84. The influence of MTHFR genetic polymorphisms on methotrexate therapy in pediatric acute lymphoblastic leukemia
  85. Red blood cell distribution width-standard deviation but not red blood cell distribution width-coefficient of variation as a potential index for the diagnosis of iron-deficiency anemia in mid-pregnancy women
  86. Small cell neuroendocrine carcinoma expressing alpha fetoprotein in the endometrium
  87. Superoxide dismutase and the sigma1 receptor as key elements of the antioxidant system in human gastrointestinal tract cancers
  88. Molecular characterization and phylogenetic studies of Echinococcus granulosus and Taenia multiceps coenurus cysts in slaughtered sheep in Saudi Arabia
  89. ITGB5 mutation discovered in a Chinese family with blepharophimosis-ptosis-epicanthus inversus syndrome
  90. ACTB and GAPDH appear at multiple SDS-PAGE positions, thus not suitable as reference genes for determining protein loading in techniques like Western blotting
  91. Facilitation of mouse skin-derived precursor growth and yield by optimizing plating density
  92. 3,4-Dihydroxyphenylethanol ameliorates lipopolysaccharide-induced septic cardiac injury in a murine model
  93. Downregulation of PITX2 inhibits the proliferation and migration of liver cancer cells and induces cell apoptosis
  94. Expression of CDK9 in endometrial cancer tissues and its effect on the proliferation of HEC-1B
  95. Novel predictor of the occurrence of DKA in T1DM patients without infection: A combination of neutrophil/lymphocyte ratio and white blood cells
  96. Investigation of molecular regulation mechanism under the pathophysiology of subarachnoid hemorrhage
  97. miR-25-3p protects renal tubular epithelial cells from apoptosis induced by renal IRI by targeting DKK3
  98. Bioengineering and Biotechnology
  99. Green fabrication of Co and Co3O4 nanoparticles and their biomedical applications: A review
  100. Agriculture
  101. Effects of inorganic and organic selenium sources on the growth performance of broilers in China: A meta-analysis
  102. Crop-livestock integration practices, knowledge, and attitudes among smallholder farmers: Hedging against climate change-induced shocks in semi-arid Zimbabwe
  103. Food Science and Nutrition
  104. Effect of food processing on the antioxidant activity of flavones from Polygonatum odoratum (Mill.) Druce
  105. Vitamin D and iodine status was associated with the risk and complication of type 2 diabetes mellitus in China
  106. Diversity of microbiota in Slovak summer ewes’ cheese “Bryndza”
  107. Comparison between voltammetric detection methods for abalone-flavoring liquid
  108. Composition of low-molecular-weight glutenin subunits in common wheat (Triticum aestivum L.) and their effects on the rheological properties of dough
  109. Application of culture, PCR, and PacBio sequencing for determination of microbial composition of milk from subclinical mastitis dairy cows of smallholder farms
  110. Investigating microplastics and potentially toxic elements contamination in canned Tuna, Salmon, and Sardine fishes from Taif markets, KSA
  111. From bench to bar side: Evaluating the red wine storage lesion
  112. Establishment of an iodine model for prevention of iodine-excess-induced thyroid dysfunction in pregnant women
  113. Plant Sciences
  114. Characterization of GMPP from Dendrobium huoshanense yielding GDP-D-mannose
  115. Comparative analysis of the SPL gene family in five Rosaceae species: Fragaria vesca, Malus domestica, Prunus persica, Rubus occidentalis, and Pyrus pyrifolia
  116. Identification of leaf rust resistance genes Lr34 and Lr46 in common wheat (Triticum aestivum L. ssp. aestivum) lines of different origin using multiplex PCR
  117. Investigation of bioactivities of Taxus chinensis, Taxus cuspidata, and Taxus × media by gas chromatography-mass spectrometry
  118. Morphological structures and histochemistry of roots and shoots in Myricaria laxiflora (Tamaricaceae)
  119. Transcriptome analysis of resistance mechanism to potato wart disease
  120. In silico analysis of glycosyltransferase 2 family genes in duckweed (Spirodela polyrhiza) and its role in salt stress tolerance
  121. Comparative study on growth traits and ions regulation of zoysiagrasses under varied salinity treatments
  122. Role of MS1 homolog Ntms1 gene of tobacco infertility
  123. Biological characteristics and fungicide sensitivity of Pyricularia variabilis
  124. In silico/computational analysis of mevalonate pyrophosphate decarboxylase gene families in Campanulids
  125. Identification of novel drought-responsive miRNA regulatory network of drought stress response in common vetch (Vicia sativa)
  126. How photoautotrophy, photomixotrophy, and ventilation affect the stomata and fluorescence emission of pistachios rootstock?
  127. Apoplastic histochemical features of plant root walls that may facilitate ion uptake and retention
  128. Ecology and Environmental Sciences
  129. The impact of sewage sludge on the fungal communities in the rhizosphere and roots of barley and on barley yield
  130. Domestication of wild animals may provide a springboard for rapid variation of coronavirus
  131. Response of benthic invertebrate assemblages to seasonal and habitat condition in the Wewe River, Ashanti region (Ghana)
  132. Molecular record for the first authentication of Isaria cicadae from Vietnam
  133. Twig biomass allocation of Betula platyphylla in different habitats in Wudalianchi Volcano, northeast China
  134. Animal Sciences
  135. Supplementation of probiotics in water beneficial growth performance, carcass traits, immune function, and antioxidant capacity in broiler chickens
  136. Predators of the giant pine scale, Marchalina hellenica (Gennadius 1883; Hemiptera: Marchalinidae), out of its natural range in Turkey
  137. Honey in wound healing: An updated review
  138. NONMMUT140591.1 may serve as a ceRNA to regulate Gata5 in UT-B knockout-induced cardiac conduction block
  139. Radiotherapy for the treatment of pulmonary hydatidosis in sheep
  140. Retraction
  141. Retraction of “Long non-coding RNA TUG1 knockdown hinders the tumorigenesis of multiple myeloma by regulating microRNA-34a-5p/NOTCH1 signaling pathway”
  142. Special Issue on Reuse of Agro-Industrial By-Products
  143. An effect of positional isomerism of benzoic acid derivatives on antibacterial activity against Escherichia coli
  144. Special Issue on Computing and Artificial Techniques for Life Science Applications - Part II
  145. Relationship of Gensini score with retinal vessel diameter and arteriovenous ratio in senile CHD
  146. Effects of different enantiomers of amlodipine on lipid profiles and vasomotor factors in atherosclerotic rabbits
  147. Establishment of the New Zealand white rabbit animal model of fatty keratopathy associated with corneal neovascularization
  148. lncRNA MALAT1/miR-143 axis is a potential biomarker for in-stent restenosis and is involved in the multiplication of vascular smooth muscle cells
Downloaded on 10.1.2026 from https://www.degruyterbrill.com/document/doi/10.1515/biol-2021-0121/html
Scroll to top button