Hostname: page-component-76fb5796d-25wd4 Total loading time: 0 Render date: 2024-04-28T01:22:49.569Z Has data issue: false hasContentIssue false

Supplementation with (6S)-5-methyltetrahydrofolic acid appears as effective as folic acid in maintaining maternal folate status while reducing unmetabolised folic acid in maternal plasma: a randomised trial of pregnant women in Canada

Published online by Cambridge University Press:  10 August 2023

Kelsey M. Cochrane
Affiliation:
Food, Nutrition, and Health, Faculty of Land and Food Systems, The University of British Columbia, Vancouver, Canada BC Children’s Hospital Research Institute, Healthy Starts, Vancouver, Canada
Rajavel Elango
Affiliation:
BC Children’s Hospital Research Institute, Healthy Starts, Vancouver, Canada Population and Public Health, Faculty of Medicine, The University of British Columbia, Vancouver, Canada
Angela M. Devlin
Affiliation:
BC Children’s Hospital Research Institute, Healthy Starts, Vancouver, Canada Paediatrics, Faculty of Medicine, The University of British Columbia, Vancouver, Canada
Chantal Mayer
Affiliation:
Obstetrics and Gynaecology, Faculty of Medicine, The University of British Columbia, Vancouver, Canada
Jennifer A. Hutcheon
Affiliation:
BC Children’s Hospital Research Institute, Healthy Starts, Vancouver, Canada Obstetrics and Gynaecology, Faculty of Medicine, The University of British Columbia, Vancouver, Canada
Crystal D. Karakochuk*
Affiliation:
Food, Nutrition, and Health, Faculty of Land and Food Systems, The University of British Columbia, Vancouver, Canada BC Children’s Hospital Research Institute, Healthy Starts, Vancouver, Canada
*
*Corresponding author: Crystal D. Karakochuk, email crystal.karakochuk@ubc.ca
Rights & Permissions [Opens in a new window]

Abstract

Folic acid supplementation is recommended during pregnancy to support healthy fetal development; (6S)-5-methyltetrahydrofolic acid ((6S)-5-MTHF) is available in some commercial prenatal vitamins as an alternative to folic acid, but its effect on blood folate status during pregnancy is unknown. To address this, we randomised sixty pregnant individuals at 8–21 weeks’ gestation to 0·6 mg/d folic acid or (6S)-5-MTHF × 16 weeks. Fasting blood specimens were collected at baseline and after 16 weeks (endline). Erythrocyte and serum folate were quantified via microbiological assay (as globally recommended) and plasma unmetabolised folic acid (UMFA) via LC-MS/MS. Differences in biochemical folate markers between groups were explored using multivariable linear/quantile regression, adjusting for baseline concentrations, dietary folate intake and gestational weeks. At endline (n 54), the mean values and standard deviations (or median, inter-quartile range) of erythrocyte folate, serum folate and plasma UMFA (nmol/l) in those supplemented with (6S)-5-MTHF v. folic acid, respectively, were 1826 (sd 471) and 1998 (sd 421); 70 (sd 13) and 78 (sd 17); 0·5 (0·4, 0·8) and 1·3 (0·9, 2·1). In regression analyses, erythrocyte and serum folate did not differ by treatment group; however, concentrations of plasma UMFA in pregnancy were 0·6 nmol/l higher (95 % CI 0·2, 1·1) in those supplementing with folic acid as compared with (6S)-5-MTHF. In conclusion, supplementation with (6S)-5-MTHF may reduce plasma UMFA by ∼50 % as compared with supplementation with folic acid, the biological relevance of which is unclear. As folate is currently available for purchase in both forms, the impact of circulating maternal UMFA on perinatal outcomes needs to be determined.

Type
Research Article
Creative Commons
Creative Common License - CCCreative Common License - BY
This is an Open Access article, distributed under the terms of the Creative Commons Attribution licence (http://creativecommons.org/licenses/by/4.0/), which permits unrestricted re-use, distribution and reproduction, provided the original article is properly cited.
Copyright
© Crown Copyright - The University of British Columbia, 2023. Published by Cambridge University Press on behalf of The Nutrition Society

Folate is an essential, water-soluble B-vitamin that is present in numerous chemically related forms (Fig. 1)(Reference Shane1). Naturally occurring folates are mainly reduced and polyglutamated, with 5-methyltetrahydrofolate (5-MTHF) being the predominant form in food and systemic circulation(Reference Shane1,Reference Lucock2) . Reduced folates serve as methyl donors in one carbon metabolism; thus, folate is critical during periods of growth, such as pregnancy, as it supports numerous physiological processes including cellular proliferation, re-methylation of homocysteine to methionine, nucleic acid synthesis and methylation of DNA, RNA, proteins and phospholipids(Reference Ducker and Rabinowitz3Reference Guéant, Namour and Guéant-Rodriguez5).

Fig. 1. Chemical structure of folic acid and (6S)-5-methyltetrahydrofolic acid. (a) Folic acid; a synthetic analogue of the folate parent structure. Folic acid is fully oxidised, increasing its stability in supplements and fortified foods, and requires reduction via dihydrofolate reductase for co-enzymatic function. (b) Ca salt of (6S)-5-methyltetrahydrofolic acid (Metafolin®); a synthetic analogue of natural 5-methyltetrahydrofolate.

To reduce the risk of neural tube defects (NTD), North American public health agencies advise all individuals of reproductive age to consume 0·4 mg/d folic acid, starting preconceptionally and continued throughout pregnancy and lactation(69). However, prenatal vitamins in these regions generally contain 0·6–1·0 mg folic acid(Reference Lamers, Macfarlane and O’Connor10Reference Bailey, Pac and Iii12) and concerns have been raised regarding excessive intake in pregnancy(Reference Lamers, Macfarlane and O’Connor10,Reference Maruvada, Stover and Mason13) . Folic acid is a synthetic, oxidised form of folate (Fig. 1(a))(Reference Shane1,Reference Lucock2) ; as such, folic acid requires reduction by dihydrofolate reductase for use in one carbon metabolism(Reference Pietrzik, Bailey and Shane14). However, unmetabolised folic acid (UMFA) is detected in biological fluids upon folic acid intake, suggesting that the capacity of intestinal dihydrofolate reductase to reduce folic acid in humans may be limited(Reference Patanwala, King and Barrett15,Reference Bailey and Ayling16) . The biological and clinical relevance of UMFA on human health remains unclear(Reference Pannia, Hammoud and Simonian17).

An increasing number of prenatal vitamins contain (6S)-5-methyltetrahydrofolic acid (Fig. 1(b); (6S)-5-MTHF)) as an alternative to folic acid(Reference Saldanha, Dwyer and Haggans11,18) . In 2019, 32 % of prescription and 25 % of non-prescription prenatal vitamins commercially available in the USA (as per the NIH Dietary Supplement Label and DailyMed databases) contained (6S)-5-MTHF; prior to 2015, none was listed(Reference Saldanha, Dwyer and Haggans11). Supplements in Canada which are marketed for use in pregnancy may contain folic acid or (6S)-5-MTHF, so long as a minimum dose of 0·4 mg is provided(18). A possible metabolic advantage of (6S)-5-MTHF is that it does not require reduction via dihydrofolate reductase, as it is already in an active form(Reference Pietrzik, Bailey and Shane14). However, there is no clear messaging from public health agencies regarding the difference of supplemental folate forms for pregnant individuals, as the effect of (6S)-5-MTHF on folate status during pregnancy has never been evaluated, and only folic acid has been shown to reduce the risk of NTD in randomised controlled trials(Reference Viswanathan, Treiman and Kish-Doto19Reference Czeizel and Dudás21).

While supplementation with (6S)-5-MTHF has proven more effective than folic acid in non-pregnant women(Reference Bailey and Ayling22Reference Houghton, Sherwood and Pawlosky25), pregnancy is a unique physiological state associated with numerous metabolic changes, including increased haemodilution, urinary excretion and micronutrient needs to support growth of maternal and fetal tissue(Reference Tamura and Picciando26,Reference Cochrane, Williams and Elango27) . Maternal plasma supplies folate to the fetus and uteroplacental organs to accommodate growth within these compartments(Reference Tamura and Picciando26). Placental folate receptors are established in early pregnancy(Reference Solanky, Requena Jimenez and D’Souza28), and folate accumulates in the intervillous space at a concentration three times that of maternal plasma for transportation of folate to the fetus against a concentration gradient(Reference Tamura and Picciando26,Reference Antony29) ; further, in ex-vivo experiments, the transport of folate from maternal to fetal perfusate has been found to be non-saturable(Reference Tamura and Picciando26,Reference Bisseling, Steegers and van den Heuvel30) . As (6S)-5-MTHF supplements are currently widely consumed by pregnant individuals(Reference Saldanha, Dwyer and Haggans11), despite a lack of evidence in pregnancy, it is imperative to ensure that (6S)-5-MTHF can maintain folate status to the same extent as folic acid during pregnancy.

This study aimed to generate estimates of folate status, including erythrocyte folate, serum folate and plasma UMFA following supplementation with 0·6 mg/d (6S)-5-MTHF or folic acid × 16 weeks of pregnancy. These estimates are a critical first step in understanding any differences between commercially available folate forms.

Methods

Study participants and design

The protocol for this two-armed randomised double-blind trial has previously been published(Reference Cochrane, Mayer and Devlin31). In brief, individuals aged 19–42 years with singleton pregnancies in Vancouver, Canada were recruited via printed posters and social media advertisements; full details on the recruitment methods are published elsewhere(Reference Cochrane, Hutcheon and Karakochuk32). The study took place between September 2019 and September 2021. Exclusion criteria included medical conditions, medications and behavioural factors (current smoking, alcohol consumption or recreational drug use) associated with altered folate status(Reference Douglas Wilson8), a pre-pregnancy BMI ≥ 30 kg/m2 and being medium to high risk for development of an NTD-affected pregnancy(Reference Douglas Wilson8). The trial is registered at ClinicalTrials.gov (NCT04022135). The study supplements were approved for clinical trial use by the natural and non-prescription health products directorate of Health Canada (Submission No. 244456). This study was conducted according to the guidelines laid down in the Declaration of Helsinki, and all procedures involving human subjects were approved by the UBC Clinical Research Ethics Board (H18-02635). Written informed consent was obtained from all subjects.

Eligible participants were scheduled for a baseline visit between 8 and 21 weeks’ gestation, with instructions to continue any folate supplementation until the day before this visit. At baseline, written informed consent was obtained (including confirmation of eligibility via a checklist), weight and height were measured and demographic information and usual intake of dietary folate equivalents (DFE; as per a validated FFQ; NutritionQuest(Reference Clifford, Noceti and Block-Joy33)) were collected. The Block Folic Acid/DFE Screener was used; this FFQ includes twenty-one items identified as the top dietary folate contributors as per National Health and nutrition examination survey (NHANES) 1999–2000. DFE are calculated based on the respondent’s reported frequency of consumption of each item, a default portion size (per the respondent’s age and sex) and the folate content listed in the US Department of Agriculture Food and Nutrient Database for Dietary Studies(Reference Clifford, Noceti and Block-Joy33). A conversion factor of 1·7 is used to convert µg supplemental folate (from supplements and folic acid fortified foods) to 1 µg DFE; 1 µg folate from natural food sources is equivalent to 1 µg DFE(Reference Clifford, Noceti and Block-Joy33,Reference Bailey, Stover and McNulty34) .

Participants were randomised to 0·6 mg/d folic acid or an equimolar dose (0·625 mg/d) of Metafolin® (Ca salt of (6S)-5-MTHF), based on molecular weights of 441·4 and 459·5 g/mol for folic acid and Metafolin®, respectively; a prenatal vitamin (NPN: 80025456) with folic acid removed was also provided to all participants. The randomisation sequence was produced by an independent statistician using permuted, equal blocks of four. Blinded allocations (‘A’ or ‘B’) for each study ID were kept in individual envelopes prepared by a research volunteer. Participants were assigned a unique study ID, and at baseline visits, the allocated supplements for that study ID were provided. Participants and the research team remained blinded to folate group allocations until final statistical analyses were complete. Participants were given all supplements needed for the 16-week intervention period and were instructed to begin supplementation immediately following baseline visits. Participants were contacted at the study midpoint (8 weeks after the baseline visit) to touch base and answer any questions about the trial.

Following 16 weeks, participants were scheduled for a second visit (endline). Participants had the option to continue supplementation for the postpartum study phase, to explore the effect on folate status after delivery (∼1 week postpartum); new written informed consent was obtained, and more study supplements were provided to those interested. Capsule counts were completed at endline and postpartum visits to assess adherence to daily supplementation. Participants were also given a supplement diary to record daily intake or reasons for missing a dose.

Blood collection and processing

Venous blood specimens were collected at baseline, endline and postpartum visits in a 4 ml serum tube, 6 ml EDTA tube and 2 ml EDTA tube (Becton Dickinson). At baseline and endline visits, participants were instructed to fast for 3 h to mitigate the influence of recent folic acid intake on plasma UMFA(Reference Bailey and Ayling16,Reference Sweeney, McPartlin and Weir35) . Blood specimen collections at postpartum visits were non-fasting to reduce participant burden, with instruction to consume the folate supplement 2 h prior to the visit (to standardise any peak in plasma UMFA following intake); erythrocyte folate is less sensitive to recent intake(Reference Bailey, Stover and McNulty34). After collection, tubes were shielded from light, inverted gently and transported to the laboratory for immediate processing (within 2 h of collection).

Whole blood (0·3 ml) was removed from the 6 ml EDTA tube and was diluted 1/11 with a 3 ml 1 % ascorbic acid solution, followed by incubation at 37°C for 30 min(Reference Pirkle36). Remaining whole blood in the 6 ml EDTA tube was centrifuged at 3000 rpm for 15 min at 4°C; plasma was separated into aliquots. Remaining contents in the 6 ml EDTA tube were processed for peripheral blood mononuclear cell isolation (SepMateTM STEMCELL Technologies). The 2 ml EDTA tube was used for a complete blood count, including haematocrit (L/L) determination via an automated haematology analyser (Sysmex XNL-550, Sysmex Corp.). Serum tubes were left at room temperature to clot for minimally 30 min and then centrifuged at 3000 rpm for 10 min at 4°C; serum was separated into aliquots. All specimens were frozen immediately after processing at –80°C until analysis.

Biochemical analyses

Total serum and whole blood folate (nmol/l) were determined as per the microbiological assay (Bevital AS) with chloramphenicol-resistant Lactobacillus rhamnosus and folic acid calibration, using previously described methods(Reference Zhang, Sternberg and Pfeiffer37). The inter-assay CV ranged from 3·1 to 5·6 % based on three quality controls that were analysed in duplicate. Erythrocyte folate (nmol/l) was subsequently calculated as recommended(Reference Pirkle36):

$$\begin{gathered}\!\!\!\!\!\!\!\!\!\!\!\!\!\!\!\!\!\!\!\!\!\!\!\!\!\!\!\!\!\!\!\!\!\!\!\!\!\!\!\!\!\!\!\!\!\!\!\!\!\!\!\!\!\!\!\!\!\!\!\!\!\!\!\!\!\!\!\!\!\!\!\!\!\!\!\!\!\!\!\!\!\!\!\!\!\!\!\!\!\!\!\!\!\!\!\!\!\!\!\!\!\!\!\!\!\!\!\!\!\!\!\!\!\!\!\!\!\!\!\!\!\!\!\!\!\!\!\!\!\!\!\!\!\!\!\!\!\!\!\!\!\!\!\!\!\!\!\!\!\!\!\!\!\!\!\!\!\!\!\!\!\!\!\!\!\!\!\!\!\!\!\!\!\!\!\!\!\!\!\!\!\!\!\!\!\!\!\!\!\!\!\!\!\!\!\!{\text{Erythrocyte }}{\mkern 1mu} {\text{folate}}{\mkern 1mu} \hfill \\ = \frac{{\left( {Whole\;blood\;haemolysate\;folate{\mkern 1mu} \times {\mkern 1mu} 11} \right)\; - \;Serum\;folate\;\left( {1 - Haematocrit/100} \right)}}{{Haematocrit/100}} \hfill \\ \end{gathered}$$

Plasma UMFA (nmol/l) was determined via LC-MS/MS as previously described(Reference Pfeiffer, Fazili and McCoy38Reference Page, Robichaud and Arbuckle42). The inter-assay CV for plasma UMFA was 8 %, as determined using internal quality control samples. Exploratory outcomes included other methyl nutrients and MTHFR 677 C > T variant genotyping. Plasma vitamin B12 (pmol/l) was determined via an immunoanalyser (Abbott Architect i1000; CV = 3·3 %). Plasma pyridoxal phosphate (PLP; nmol/l; CV = 8 %), total homocysteine (µmol/l; CV = 1·7 %), cysteine (µmol/l; CV = 1·4 %), methionine (µmol/l; CV = 1·1 %), free choline (µmol/l; CV = 5·4 %) and betaine (µmol/l; CV = 2·7 %) were determined using high-performance LC-MS/MS(Reference Innis and Hasman43,Reference Dominguez-Salas, Moore and Cole44) . Genotyping of MTHFR 677 C > T variant (rs1801133) was determined via pyrosequencing using PyromarkTM Q96 MD Pyrosequencer (Qiagen), as previously described(Reference Clifford, Jones and MacIsaac45). Plasma vitamin B12, PLP, total homocysteine, cysteine, methionine, free choline and betaine were quantified at baseline and endline visits only.

Statistical analyses

Participant demographic data and biochemical outcomes were summarised descriptively. Folate status in each group (erythrocyte folate, serum folate and plasma UMFA) was summarised using mean values and standard deviations or medians with inter-quartile ranges (IQR) if not normally distributed. The median (IQR) within-person change in folate status (erythrocyte folate, serum folate and plasma UMFA) from baseline to endline was calculated. The change in concentrations of erythrocyte folate, serum folate and plasma UMFA in each intervention group was further explored by evaluating differences in endline concentrations, adjusting for baseline concentrations, dietary folate intake and weeks gestation at supplement initiation, using multivariable linear regression (or multivariable quantile regression for non-normally distributed outcomes). Quantile regression was chosen over data transformations as it requires fewer assumptions about the underlying distribution and leads to more straightforward biological interpretations of the model estimates(Reference Cook and Manning46). Differences in postpartum erythrocyte folate were explored using quantile regression, adjusting for endline concentrations, dietary folate intake and total weeks supplementing. Postpartum concentrations of serum folate and plasma UMFA were not explored in adjusted models given their limited interpretability due to the non-fasting state. Covariates were chosen a priori; however, in a post-hoc protocol change, we opted not to adjust for exploratory biomarkers (other methyl nutrients, MTHFR genotype) as no clinically meaningful differences in biochemical markers between groups were observed, and due to few participants with the homozygous (TT) polymorphism (n 2 in the folic acid group). All analyses were completed on an intention-to-treat basis. Due to high adherence to the study protocol throughout the trial (median (IQR) adherence to daily supplementation = 98 % (96 %, 100 %), as per capsule counts), per-protocol analyses were deemed unnecessary. A P value < 0·05 was considered statistically significant.

Sample size considerations

The sample size was based on the number of participants generally recognised as sufficient for conducting a pilot study in clinical research (n 50)(Reference Sim and Lewis47), with the aim of generating estimates that can be used to inform a definitive trial. To account for an estimated 20 % attrition, we aimed to recruit a total of sixty participants to the current study.

Results

Overall, sixty participants completed baseline visits, fifty-four completed endline visits (retention rate of 90 %) and thirty-seven provided a postpartum blood specimen (retention rate of 69 %). One participant delivered prematurely and could not participate in the endline visit; however, they provided a postpartum blood specimen. See Fig. 2 for the participant flow diagram. Adherence to daily supplementation as per capsule counts was high, with rates of daily supplementation in 93 % (n 50) of participants ≥ 90 %. At postpartum visits, 92 % (n 34) of participants had rates of daily adherence ≥ 90 %. Per the supplement diary, primary reasons for a missed dose included: fatigue, forgetting, illness (gastrointestinal distress), being on vacation and lack of intake while admitted to hospital for delivery.

Fig. 2. Participant flow diagram. The folic acid group includes n 19 at the postpartum visit; this includes n 18 from endline (n 7 chose to not participate in the postpartum blood draw) and n 1 who delivered prematurely (thus, did not participate in the endline visit), but provided a postpartum blood specimen.

Participant characteristics are presented in Table 1. Participants appeared similar between groups and were ∼33 years of age, highly educated (n 58 (97 %) with post-secondary education) and predominantly nulliparous (73 %). All participants reported a pre-pregnancy BMI < 30 km/m2; median (IQR) gestational weight gain from baseline to endline was 0·44 kg/week (0·36–0·56) and 0·46 kg/week (0·39–0·56) in the (6S)-5-MTHF and folic acid groups, respectively (missing n 3 endline weights). The intervention period from baseline to endline was 16 weeks (± 6 days); however, due to COVID-19 pandemic countermeasures, endline visits for n 5 (n 2 in the (6S)-5-MTHF group and n 3 in the folic acid group) were moved earlier or delayed; the min/max intervention period was 13/17 weeks in the (6S)-5-MTHF group and 14/20 weeks in the folic acid group. The postpartum visit was planned for ∼1 week after delivery; however, this varied slightly based on participant availability; the median (IQR) days postpartum in the (6S)-5-MTHF and folic acid groups, respectively, was day 9 (7–11 d) and day 8 (7–10 d); overall, weeks of supplementing from baseline to postpartum were 24 (sd 4) weeks.

Table 1. Participant characteristics of pregnant individuals supplemented with (6S)-5-MTHF or folic acid (Vancouver, Canada, 2019–2021) (Numbers and percentages; mean values and standard deviations; medians and inter-quartile ranges)

(6S)-5-MTHF, (6S)-5-methyltetrahydrofolic acid; DFE, dietary folate equivalents.

* Missing n 2 household income in the folic acid group (participants chose not to disclose).

As determined by the FFQ.

Supplementation prior to study enrolment; all participants reported folate supplementation prior to study enrolment, which was discontinued and replaced with the study vitamins at baseline.

§ Lacto-ovo-vegetarian (n 1), pescatarian (n 1), vegan (n 1), gluten-free (n 3), other (n 2).

Baseline biochemical outcomes are presented in Table 2; outcomes at endline and postpartum visits are presented in Table 3 (erythrocyte folate, serum folate and plasma UMFA) and online Supplementary Table 1 (exploratory outcomes: vitamin B12, PLP, total homocysteine, cysteine, methionine, free choline and betaine). While cut-offs during pregnancy are not established, median concentrations of vitamin B12 and PLP remained above cut-offs for deficiency in non-pregnant women (vitamin B12 < 148 pmol/l and PLP < 20 nmol/l), and total homocysteine remained < 13 µmol/l (cut-off for hyperhomocysteinaemia) in all participants, at all visits(Reference Visentin, Masih and Plumptre48,Reference Plumptre, Masih and Sohn49) . No participants had erythrocyte folate (< 305 nmol/l) or serum folate (< 7 nmol/l) indicative of deficiency at any point(Reference Pfeiffer, Sternberg and Hamner50). Further, erythrocyte folate remained > 906 nmol/l in all participants throughout the study (a cut-off associated with maximal risk reduction of NTD(Reference Lamers, Macfarlane and O’Connor10,Reference Daly, Kirke and Molloy51) ). Plasma UMFA was detectable in all participants at all timepoints.

Table 2. Baseline biochemical outcomes in pregnant individuals supplemented with (6S)-5-MTHF or folic acid (Vancouver, Canada, 2019–2021) (Medians and inter-quartile ranges; numbers and percentages)

(6S)-5-MTHF, (6S)-5-methyltetrahydrofolic acid; UMFA, unmetabolised folic acid.

Results are median (IQR) unless otherwise noted. Missing n 1 in the (6S)-5-MTHF group for the assessment of some exploratory plasma outcomes due to insufficient blood sample (n 29 for pyridoxal phosphate, total homocysteine, cysteine, methionine, choline, betaine).

Table 3. Effect of supplemental folate form on late pregnancy and postpartum erythrocyte folate, serum folate and plasma UMFA among pregnant individuals supplemented with (6S)-5-MTHF or folic acid (Vancouver, Canada, 2019–2021) (Mean values and standard deviation; 95 % confidence intervals)

UMFA, unmetabolised folic acid; (6S)-5-MTHF, (6S)-5-methyltetrahydrofolic acid; IQR, inter-quartile range.

n 54 participants included for all endline analyses (crude and adjusted); n 36 participants included for crude estimates of postpartum erythrocyte folate and serum folate (missing n 1 due to insufficient blood sample); n 35 participants included for adjusted estimates of postpartum erythrocyte folate (missing n 2 due to (1) insufficient blood sample and (2) missed endline visit due to premature delivery, participated in postpartum visit only); n 37 participants included for crude estimates of postpartum plasma UMFA.

* Endline outcomes adjusted for: folate form ((6S)-5-MTHF as the reference group), baseline values (nmol/l), weeks of gestation at baseline and dietary folate intake (mg dietary folate equivalents/d). Postpartum erythrocyte folate adjusted for: folate form ((6S)-5-MTHF as the reference group), endline values (nmol/l), total weeks supplementing and dietary folate intake (mg dietary folate equivalents/d).

We note a high degree of inter-individual variability of within-person change in folate status during pregnancy; the median (IQR) within-person change from baseline to endline in erythrocyte and serum folate, respectively, in those supplemented with (6S)-5-MTHF was 88 nmol/l (26, 346) and 2·5 nmol/l (–9·8, 15), and in those supplemented with folic acid was 368 nmol/l (–81, 549) and 0·8 nmol/l (–15, 15). In those supplemented with (6S)-5-MTHF, plasma UMFA decreased by a median (IQR) of 0·5 nmol/l (0·9, 0·2), representing a ∼50 % decrease at endline. Conversely, in those supplemented with folic acid, plasma UMFA remained mostly consistent from baseline to endline (median (IQR) within-person change = 0·03 nmol/l (–0·2, 0·9)).

Crude and adjusted differences between intervention groups in erythrocyte folate, serum folate and plasma UMFA at endline and postpartum visits are presented in Table 3 (see online Supplementary Table 2 for full model outputs). Concentrations of erythrocyte and serum folate at endline and postpartum were not clinically different between intervention groups, as both groups were well-above cut-offs for deficiency or NTD risk reduction, with a high degree of overlap and variability in crude concentrations (± ∼450 nmol/l for erythrocyte folate and ± ∼15 nmol/l for serum folate, at endline); differences in erythrocyte and serum folate were also not statistically significant, as the 95 % CI for all crude and adjusted differences crossed 0. Conversely, there appeared to be a meaningful difference in plasma UMFA at endline and postpartum, as there was no overlap in the IQR of crude estimates, and concentrations were significantly higher at endline (adjusted difference = 0·6 nmol/l, 95 % CI 0·2, 1·1) and postpartum (crude difference = 12 nmol/l, 95 % CI 6·1, 19) in those supplemented with folic acid as compared with (6S)-5-MTHF.

Discussion

In this investigation of sixty folate-replete, low-risk, singleton pregnancies, supplementation with 0·625 mg/d (6S)-5-MTHF and an equimolar dose of folic acid (0·6 mg/d) appeared similarly effective in maintaining folate status (erythrocyte and serum folate) in pregnancy and after delivery. The most meaningful difference between supplemental folate forms was the effect on plasma UMFA, as supplementation with folic acid resulted in significantly higher circulating concentrations of UMFA as compared with (6S)-5-MTHF.

Investigations in non-pregnant(Reference Bailey and Ayling22Reference Lamers, Prinz-langenohl and Bramswig24) and lactating(Reference Houghton, Sherwood and Pawlosky25) women have reported that (6S)-5-MTHF is more effective than folic acid in raising erythrocyte and/or serum folate concentrations. Bailey et al. (Reference Bailey and Ayling22) conducted a pharmacokinetic investigation of 7·5 mg/d (6S)-5-MTHF or folic acid × 3 d, followed by 0·4 mg/d × 2 weeks; those supplemented with (6S)-5-MTHF more quickly achieved the target cut-off (serum total folate >50 nmol/l), but by day 12, there were no differences between groups(Reference Bailey and Ayling22). Henderson et al. (Reference Henderson, Aleliunas and Loh23) and Lamers et al. (Reference Lamers, Prinz-langenohl and Bramswig24) investigated (6S)-5-MTHF and folic acid supplementation at 1·0 mg/d × 12 weeks(Reference Henderson, Aleliunas and Loh23) and 0·4 mg/d × 24 weeks(Reference Lamers, Prinz-langenohl and Bramswig24) in non-pregnant women of childbearing age; both reported greater increases in erythrocyte folate concentrations following (6S)-5-MTHF supplementation and an equal or increased effect on plasma folate concentrations. Similarly, Houghton et al. (Reference Houghton, Sherwood and Pawlosky25) reported significantly higher erythrocyte folate concentrations in those supplementing with (6S)-5-MTHF v. folic acid at 0·4 mg/d × 16 weeks during lactation. While we did not investigate pharmacokinetic action, we propose that the lack of differences in erythrocyte and serum folate found between groups in the current study may be attributed to changes in folate handling during pregnancy(Reference Tamura and Picciando26); perhaps any increased effect of (6S)-5-MTHF on folate status as compared with folic acid was nullified due to the abundant transfer of folate from maternal plasma to the placenta and fetus, or due to increased folate catabolism and excretion, particularly in later pregnancy(Reference McPartlin, Weir and Halligan52,Reference Higgins, Quinlivin and McPartlin53) .

While the difference in plasma concentrations of UMFA between groups was modest after fasting, there did appear to be a meaningful effect driven by supplemental folate form. All participants reported supplementation with folate prior to study enrolment; as per the type of folate or brand of prenatal vitamin, >90 % consumed folic acid. At baseline visits, plasma UMFA was ∼1 nmol/l in both groups; we did not observe continued increases of UMFA in the folic acid group (UMFA remained at ∼1 nmol/l at endline); however, concentrations in the (6S)-5-MTHF group decreased by ∼50 %. Fasting state and folic acid supplementation have previously been identified as the strongest predictors of circulating UMFA(Reference Pfeiffer, Sternberg and Fazili54,Reference Pfeiffer, Sternberg and Fazili55) . We do suspect a high degree of UMFA clearance following the (minimum) 3 h fast, given comparison with non-fasting postpartum UMFA values (see Table 3). However, as per NHANES 2007–2008(Reference Pfeiffer, Sternberg and Fazili55), plasma UMFA concentrations remain significantly higher after fasting ≥ 8 h in folic acid supplement users (∼1 nmol/l) v. non-supplement users (∼0·7 nmol/l). Overall, it seems that a significant reduction of UMFA in plasma can be effectively achieved by supplementation with (6S)-5-MTHF. Ultimately, it is noted that while plasma UMFA is different between groups, after fasting, concentrations are relatively low overall. Thus, the biological relevance of this difference is uncertain.

Excess folic acid intake has been associated with various adverse health outcomes for both the mother and child(Reference Lamers, Macfarlane and O’Connor10,Reference Maruvada, Stover and Mason13,Reference Pannia, Hammoud and Simonian17) . Clinical concerns related to offspring health include neurodevelopmental disorders(Reference Husebye, Wendel and Gilhus56Reference Egorova, Myte and Schneede63), allergic diseases(Reference McGowan, Hong and Selhub64Reference Molloy, Collier and Saffery68), metabolic outcomes(Reference Pannia, Hammoud and Simonian17,Reference Henderson, Tai and Aleliunas69Reference Xie, Liu and Retnakaran72) and poor fetal growth (small-for-gestational-age birth)(Reference Pastor-Valero, Navarrete-Muoz and Rebagliato73,Reference Li, Li and Ye74) . However, results in human studies are mixed, with some reporting a harmful association(Reference Raghavan, Selhub and Paul58,Reference Egorova, Myte and Schneede63Reference Whitrow, Moore and Rumbold65,Reference Pastor-Valero, Navarrete-Muoz and Rebagliato73) , null findings(Reference Husebye, Wendel and Gilhus56,Reference Wang, Wei and Wang59,Reference Best, Green and Sulistyoningrum66Reference Molloy, Collier and Saffery68,Reference Crider, Wang and Ling75) or a protective effect(Reference Caffrey, McNulty and Rollins57,Reference Levine, Kodesh and Viktorin60Reference Schmidt, Tancredi and Ozonoff62,Reference Li, Li and Ye74) of maternal folic acid supplementation. Further, the effects of folic acid may vary by dose; for example, folic acid supplementation at recommended doses (0·4 mg/d) is associated with a reduced risk of small-for-gestational-age(Reference Li, Li and Ye74), whereas intakes > 1·0 mg have shown an increased risk(Reference Pastor-Valero, Navarrete-Muoz and Rebagliato73). It is proposed that UMFA demonstrates a dose–response relation with adverse outcomes, whereby higher UMFA is associated with greater risk(Reference Maruvada, Stover and Mason13,Reference McGee, Bainbridge and Fontaine-Bisson76) .

Interpretation of findings is further complicated by a lack of clarity as to whether concerns are related specifically to folic acid, or more generally, a ‘high’ folate status. While a cut-off for ‘high’ erythrocyte folate is not established, 1360 nmol/l was reflective of the 97th percentile as per NHANES 1999–2004(Reference Pfeiffer, Johnson and Jain77,Reference Colapinto, O’Connor and Tremblay78) . Overall, it is very difficult for pregnant individuals in North America to consume only 0·4 mg/d folic acid, due to folic acid food fortification and as the vast majority of prenatal vitamins contain 0·6–1·0 mg folic acid(Reference Lamers, Macfarlane and O’Connor10,Reference Saldanha, Dwyer and Haggans11,Reference Patti, Braun and Arbuckle79Reference Dubois, Diasparra and Bedard81) ; this has contributed to very high folate levels (erythrocyte folate ∼1500–2500 nmol/l) among pregnant and lactating individuals(Reference Lamers, Macfarlane and O’Connor10,Reference Plumptre, Masih and Ly82Reference Stamm, March and Karakochuk84) ; similarly high erythrocyte and serum folate concentrations were evident in the current study. Those consuming folic acid from fortified foods and prenatal vitamins are very likely to exceed the tolerable upper intake level of 1·0 mg/d folic acid(Reference Bailey, Pac and Iii12,Reference Patti, Braun and Arbuckle79Reference Dubois, Diasparra and Bedard81,Reference Ledowsky, Mahimbo and Scarf85) .

Other considerations regarding the use of (6S)-5-MTHF v. folic acid include the lack of an established conversion factor of (6S)-5-MTHF to DFE, stability differences and cost. North American supplement manufacturers are permitted to use the folic acid factor of 1·7 to convert µg (6S)-5-MTHF to DFE(Reference Saldanha, Dwyer and Haggans11,Reference Pannia, Hammoud and Simonian17) . Appropriateness of this is unclear, given potential differences in bioavailability as observed in non-pregnant populations(Reference Turck, Bohn and Castenmiller86); however, findings of the current trial suggest that bioavailability of folate forms during pregnancy may differ from non-pregnant individuals, in that a greater effect of (6S)-5-MTHF on folate status was not observed at a 0·6 mg dose. Ca salt of (6S)-5-MTHF (Metafolin®) is stable in its crystalline form for use in supplements(Reference Obeid, Schön and Pietrzik87,88) . As a food fortificant in white flour, stability of (6S)-5-MTHF can be improved via microencapsulation technology, alone or in combination with sodium ascorbate(Reference Liu, Green and Wong89,Reference Green, Liu and Dadgar90) ; however, large-scale implementation of these techniques has had limited success in practice(Reference Kitts and Liu91) and only folic acid is currently used in North American food fortification programmes(Reference Ami, Bernstein and Boucher92). Finally, given the increased cost of (6S)-5-MTHF v. folic acid, compelling evidence of its improved safety as compared with folic acid is likely required before it would be considered for use in population-wide food fortification programmes.

Ultimately, a definitive trial is required to confirm that erythrocyte and serum folate are similarly maintained following supplementation with (6S)-5-MTHF and folic acid during pregnancy; while we found no evidence for a clinically important difference, the upper end of the 95 % CI for the adjusted difference in erythrocyte folate between groups at endline was 400 nmol/l, favouring a stronger response following folic acid supplementation. Interpretation of this is limited as a non-inferiority margin was not pre-specified in our protocol, as this was a pilot trial. Overall, we hypothesise that the 95 % CI would tighten following assessment in a larger sample size, suggesting a similar effect of both forms at a 0·6 mg dose, given that overall there was a high degree of variability in folate estimates, 95 % CIs were wide and crossed 0, and a stronger response to folic acid contradicts the body of evidence on dose–response of folic acid v. (6S)-5-MTHF(Reference Turck, Bohn and Castenmiller86). Both biochemical (e.g. folate status, plasma UMFA) and clinical outcomes should be evaluated in a definitive trial; we suggest assessment of fetal growth and neurodevelopment as outcomes of interest(Reference Lamers, Macfarlane and O’Connor10,Reference Maruvada, Stover and Mason13) .

Limitations include the small sample size, as this was a pilot trial which aimed to generate estimates for a definitive trial. While a 3 h fast was selected to mitigate the effect of folic acid on plasma UMFA(Reference Bailey and Ayling16,Reference Sweeney, McPartlin and Weir35) , total serum folate concentrations may still have been influenced by recent intake within this time frame(Reference Bailey, Stover and McNulty34,Reference Pfeiffer, Sternberg and Fazili55) . Estimation of dietary folate intake was only assessed once (at baseline); thus, its accuracy in later pregnancy and after delivery is uncertain. Dietary folate assessment via an FFQ can be useful when assessing a single nutrient over a longer period, as it captures food sources that may be consumed only occasionally and thus may be missed via other methods(Reference Park, Dodd and Kipnis93). However, previous research indicates that FFQ may overestimate folate intake(Reference Park, Dodd and Kipnis93,Reference Green, Allen and O’Connor94) . We further note that the FFQ used was an American tool; while dietary patterns of those in Canada v. the USA may differ, green salads, orange juice, bread products and cold cereals are reported as top folate contributors for pregnant and lactating Canadians(Reference Masih, Plumptre and Ly80,Reference Sherwood, Houghton and Tarasuk95) , all of which were included on the folate screener. Both countries fortify foods with folic acid at similar rates (0·14 and 0·15 mg per 100 g white flour and cereals in the USA and Canada, respectively), aiming to provide ∼0·1 mg/d(Reference Crider, Bailey and Berry96). However, DFE of folate supplementation prior to the trial as per the FFQ are likely underestimated; the median (IQR) mg DFE from supplements was 0·7 (0·7, 0·7 mg DFE), assuming a folic acid dose of 0·4 mg (×1·7 = 0·7 mg DFE). While the dose of folate provided in the study (0·6 mg/d) was chosen to improve generalisability, given that it matches folic acid content in leading Canadian brands (Materna®), over-the-counter prenatal vitamins contain varying quantities of folate (often 1 mg in Canada). Finally, timing of supplement initiation varied widely (8–21 weeks). While folate recommendations in Canada and the USA do not differ based on gestational weeks, we note a potential limitation when comparing folate status of those across different stages of pregnancy.

In conclusion, supplementation with (6S)-5-MTHF resulted in similar erythrocyte and serum folate concentrations as folic acid during pregnancy and after delivery, while reducing maternal plasma UMFA. More research is needed to confirm whether there is any risk associated with folic acid supplementation in pregnancy (particularly at doses > 0·4 mg) or the presence of plasma UMFA. This should be followed by the establishment of high-risk cut-offs for maternal folate status, which can be incorporated into perinatal clinical practice guidelines, and re-formulation of prenatal vitamins available in North America (perhaps by lowering the dose to 0·4 mg), to support achievement of optimal maternal folate status. Results of this study should be confirmed in a definitive trial; however, the current findings are very timely and of interest immediately, as both folate forms are currently widely available and consumed in pregnancy at a dose of 0·6 mg/d or greater.

Acknowledgements

We thank our research assistant, Dahlia Parolin, for her assistance with double data entry. Erythrocyte and serum folate were quantified by microbiological assay at Bevital AS (Bergen, Norway) and plasma UMFA was quantified at the University of British Columbia (Vancouver, Canada). Plasma vitamin B12, PLP, total homocysteine, cysteine, methionine, free choline and betaine were performed at the Analytical Core for Metabolomics and Nutrition at the BC Children’s Hospital Research Institute, Core Technologies and Services (Vancouver, Canada; Roger Dyer). Genotyping of MTHFR 677 C > T variant (rs1801133) was determined in the Kobor Lab at the BC Children’s Hospital Research Institute (Vancouver, Canada).

This work was supported by a Healthy Starts Catalyst Grant, BC Children’s Hospital Research Institute (C. D. K., J. A. H., R. E., C. M.). K. M. C. was supported by Frederick Banting and Charles Best Canada Graduate Scholarship Doctoral Award from the Canadian Institute of Health Research. C. D. K. was supported by a Michael Smith Foundation for Health Research Scholar Award and holds a Canada Research Chair in Micronutrients and Human Health. J. A. H. holds a Canada Research Chair in Perinatal Population Health. A. M. D. was supported by an Investigator Grant from BC Children’s Hospital Research Institute. Natural Factors® Canada manufactured the study supplements and donated the raw materials for folic acid and prenatal vitamin compounding; Merck & Cie (Schaffhausen, Switzerland) donated the (6S)-5-methyltetrahydrofolic acid (Metafolin®). Funders had no role in the design, analysis or writing of this article.

Study concept: R. E., C. M., J. A. H. and C. D. K. Finalisation of the study protocol: K. M. C., R. E., A. M. D., C. M., J. A. H. and C. D. K. Data collection, analysis and drafting of the manuscript: K. M. C. Manuscript revisions and final writing: K. M. C., R. E., A. M. D., C. M., J. A. H. and C. D. K. C. D. K. has primary responsibility for final content; all authors have read and approved the final manuscript.

There are no conflicts of interest.

Supplementary material

For supplementary materials referred to in this article, please visit https://doi.org/10.1017/S0007114523001733

References

Shane, B (2008) Folate and vitamin B12 metabolism: overview and interaction with riboflavin, vitamin B6, and polymorphisms. Food Nutr Bull 29, S516.Google Scholar
Lucock, M (2000) Folic acid: nutritional biochemistry, molecular biology, and role in disease processes. Mol Genet Metab 71, 121138.Google Scholar
Ducker, G & Rabinowitz, J (2017) One-carbon metabolism in liver health and disease. Cell Metab 25, 2749.Google Scholar
Tibbetts, AS & Appling, DR (2010) Compartmentalization of mammalian folate-mediated one-carbon metabolism. Annu Rev Nutr 30, 5781.Google Scholar
Guéant, JL, Namour, F, Guéant-Rodriguez, RM, et al. (2013) Folate and fetal programming: a play in epigenomics? Trends Endocrinol Metab 24, 279289.Google Scholar
Lamers, Y (2011) Folate recommendations for pregnancy, lactation, and infancy. Ann Nutr Metab 59, 3237.Google Scholar
Douglas Wilson, R (2015) SOGC Clinical Practice Guideline: pre-conception folic acid and multivitamin supplementation for the primary and secondary prevention of neural tube defects and other folic acid-sensitive congenital anomalies. J Obs Gynaecol Can 37, 534549.Google Scholar
Centers for Disease Control and Prevention & National Center on Birth Defects and Developmental Disabilities (2022) Folic Acid Recommendations (Internet). Atlanta, GA, USA. https://www.cdc.gov/ncbddd/folicacid/recommendations.html (accessed January 2023).Google Scholar
Lamers, Y, Macfarlane, AJ, O’Connor, DL, et al. (2018) Periconceptional intake of folic acid among low-risk women in Canada: summary of a workshop aiming to align prenatal folic acid supplement composition with current expert guidelines. Am J Clin Nutr 108, 13571368.Google Scholar
Saldanha, LG, Dwyer, JT, Haggans, CJ, et al. (2020) Perspective: time to resolve confusion on folate amounts, units, and forms in prenatal supplements. Adv Nutr 11, 753759.Google Scholar
Bailey, RL, Pac, SG, Iii, VLF, et al. (2019) Estimation of total usual dietary intakes of pregnant women in the United States. JAMA Netw Open 2, e195967.Google Scholar
Maruvada, P, Stover, PJ, Mason, JB, et al. (2020) Knowledge gaps in understanding the metabolic and clinical effects of excess folates/folic acid: a summary, and perspectives, from an NIH workshop. Am J Clin Nutr 112, 13901403.Google Scholar
Pietrzik, K, Bailey, L & Shane, B (2010) Folic acid and L-5-methyltetrahydrofolate: comparison of clinical pharmacokinetics and pharmacodynamics. Clin Pharmacokinet 49, 535548.Google Scholar
Patanwala, I, King, MJ, Barrett, DA, et al. (2014) Folic acid handling by the human gut: implications for food fortification and supplementation. Am J Clin Nutr 100, 593599.CrossRefGoogle ScholarPubMed
Bailey, SW & Ayling, JE (2009) The extremely slow and variable activity of dihydrofolate reductase in human liver and its implications for high folic acid intake. PNAS 106, 1542415429.CrossRefGoogle ScholarPubMed
Pannia, E, Hammoud, R, Simonian, R, et al. (2022) Folate dose and form during pregnancy may program maternal and fetal health and disease risk. Nutr Rev 80, 21872197.Google Scholar
Health Canada (2022) Multi-Vitamin/Mineral Supplements Monograph (Internet). Ottawa, ON, Canada. pp. 1–71. https://webprod.hc-sc.gc.ca/nhpid-bdipsn/atReq.do?atid=multi_vitmin_suppl (accessed December 2022).Google Scholar
Viswanathan, M, Treiman, KA, Kish-Doto, J, et al. (2017) Folic acid supplementation for the prevention of neural tube defects an updated evidence report and systematic review for the US preventive services task force. JAMA 317, 190203.Google Scholar
MRC Vitamin Study Research Group (1991) Prevention of neural tube defects: results of the Medical Research Council Vitamin Study. Lancet 338, 131137.CrossRefGoogle Scholar
Czeizel, AE & Dudás, I (1992) Prevention of the first occurrence of neural-tube defects by periconceptional vitamin supplementation. N Engl J Med 327, 18321835.Google Scholar
Bailey, SW & Ayling, JE (2018) The pharmacokinetic advantage of 5-methyltetrahydrofolate for minimization of the risk for birth defects. Sci Rep 8, 4096.Google Scholar
Henderson, AM, Aleliunas, RE, Loh, SP, et al. (2018) L-5-methyltetrahydrofolate supplementation increases blood folate concentrations to a greater extent than folic acid supplementation in Malaysian women. J Nutr 148, 855890.Google Scholar
Lamers, Y, Prinz-langenohl, R, Bramswig, S, et al. (2006) Red blood cell folate concentrations increase more after supplementation with (6S)-5-methyltetrahydrofolate than with folic acid in women of childbearing age. Am J Clin Nutr 84, 156161.Google Scholar
Houghton, LA, Sherwood, KL, Pawlosky, R, et al. (2006) (6S)-5-Methyltetrahydrofolate is at least as effective as folic acid in preventing a decline in blood folate concentrations during lactation. Am J Clin Nutr 83, 842850.Google Scholar
Tamura, T & Picciando, MF (2006) Folate and human reproduction. Am J Clin Nutr 83, 9931016.Google Scholar
Cochrane, KM, Williams, BA, Elango, R, et al. (2022) Pregnancy-induced alterations of 1-carbon metabolism and significance for maternal nutrition requirements. Nutr Rev 80, 19852001.Google Scholar
Solanky, N, Requena Jimenez, A, D’Souza, SW, et al. (2010) Expression of folate transporters in human placenta and implications for homocysteine metabolism. Placenta 31, 134143.Google Scholar
Antony, AC (1996) Folate receptors. Ann Rev Nutr 16, 501521.Google Scholar
Bisseling, TM, Steegers, EAP, van den Heuvel, JJM, et al. (2004) Placental folate transport and binding are not impaired in pregnancies complicated by fetal growth restriction. Placenta 25, 588593.Google Scholar
Cochrane, KM, Mayer, C, Devlin, AM, et al. (2020) Is natural (6S)-5-methyltetrahydrofolic acid as effective as synthetic folic acid in increasing serum and red blood cell folate concentrations during pregnancy? A proof-of-concept pilot study. Trials 21, 380.Google Scholar
Cochrane, KM, Hutcheon, JA & Karakochuk, CD (2022) Strategies for improving recruitment of pregnant women to clinical research: an evaluation of social media v. traditional offline methods. Digit Heal 8, 110.Google Scholar
Clifford, AJ, Noceti, EM, Block-Joy, A, et al. (2005) Erythrocyte folate and its response to folic acid supplementation is assay dependent in women. J Nutr 135, 137143.Google Scholar
Bailey, LB, Stover, PJ, McNulty, H, et al. (2015) Biomarkers of nutrition for development-Folate review. J Nutr 145, 1636S1680S.Google Scholar
Sweeney, MR, McPartlin, J, Weir, DG, et al. (2006) Postprandial serum folic acid response to multiple doses of folic acid in fortified bread. Br J Nutr 95, 145151.Google Scholar
Pirkle, J (2014) CDC Laboratory Procedure Manual, Total Folate, Serum/Whole Blood Microbiological Assay. https://wwwn.cdc.gov/nchs/data/nhanes/2013-2014/labmethods/FOLATE_H_MET.pdf (accessed August 2023).Google Scholar
Zhang, M, Sternberg, MR & Pfeiffer, CM (2018) Harmonizing the calibrator and microorganism used in the folate microbiological assay increases the comparability of serum and whole-blood folate results in a CDC round-robin study. J Nutr 148, 807817.Google Scholar
Pfeiffer, C, Fazili, Z, McCoy, L, et al. (2004) Determination of folate vitamers in human serum by stable-isotope-dilution tandem mass spectrometry and comparison with radioassay and microbiologic assay. Clin Chem 50, 423432.Google Scholar
Fazili, Z & Pfeiffer, CM (2013) Accounting for an isobaric interference allows correct determination of folate vitamers in serum by isotope dilution liquid chromatography-tandem MS. J Nutr 143, 108113.Google Scholar
Stamm, RA, Fazili, Z & Pfeiffer, CM (2018) Addition of exogenous γ-glutamyl hydrolase eliminates the need for lengthy incubation of whole-blood lysate for quantitation of folate vitamers by high-performance liquid chromatography-tandem mass spectrometry. Curr Dev Nutr 2, nx003.Google Scholar
Fazili, Z, Sternberg, MR, Paladugula, N, et al. (2017) Two international round-robin studies showed good comparability of 5-methyltetrahydrofolate but poor comparability of folic acid measured in serum by different high-performance liquid chromatography-tandem mass spectrometry methods. J Nutr 147, 18151825.Google Scholar
Page, R, Robichaud, A, Arbuckle, TE, et al. (2017) Total folate and unmetabolized folic acid in the breast milk of a cross-section of Canadian women. Am J Clin Nutr 105, 11011109.Google Scholar
Innis, SM & Hasman, D (2006) Evidence of choline depletion and reduced betaine and dimethylglycine with increased homocysteine in plasma of children with cystic fibrosis. J Nutr 136, 22262231.Google Scholar
Dominguez-Salas, P, Moore, SE, Cole, D, et al. (2013) DNA methylation potential: dietary intake and blood concentrations of one-carbon metabolites and cofactors in rural African women. Am J Clin Nutr 97, 12171227.Google Scholar
Clifford, RL, Jones, MJ, MacIsaac, JL, et al. (2017) Inhalation of diesel exhaust and allergen alters human bronchial epithelium DNA methylation. J Allergy Clin Immunol 139, 112121.Google Scholar
Cook, B & Manning, W (2013) Thinking beyond the mean: a practical guide for using quantile regression methods for health services research. Shanghai Arch Psychiatry 25, 5559.Google Scholar
Sim, J & Lewis, M (2012) The size of a pilot study for a clinical trial should be calculated in relation to considerations of precision and efficiency. J Clin Epidemiol 65, 301308.Google Scholar
Visentin, CE, Masih, SP, Plumptre, L, et al. (2016) Low serum vitamin B-12 concentrations are prevalent in a cohort of pregnant Canadian women. J Nutr 146, 10351042.Google Scholar
Plumptre, L, Masih, SP, Sohn, KJ, et al. (2018) Suboptimal maternal and cord plasma pyridoxal 5′ phosphate concentrations are uncommon in a cohort of Canadian pregnant women and newborn infants. Matern Child Nutr 14, e12467.Google Scholar
Pfeiffer, CM, Sternberg, MR, Hamner, HC, et al. (2016) Applying inappropriate cutoffs leads to misinterpretation of folate status in the US population. Am J Clin Nutr 104, 16071615.Google Scholar
Daly, LE, Kirke, P, Molloy, A, et al. (1995) Folate levels and neural tube defects. JAMA 274, 16981702.Google Scholar
McPartlin, J, Weir, D, Halligan, A, et al. (1993) Accelerated folate breakdown in pregnancy. Lancet 341, 148149.CrossRefGoogle ScholarPubMed
Higgins, J, Quinlivin, E, McPartlin, J, et al. (2001) The relationship between increased folate catabolism and the increased requirement for folate in pregnancy. Br J Obstet Gynaecol 108, 772773.Google Scholar
Pfeiffer, CM, Sternberg, MR, Fazili, Z, et al. (2015) Folate status and concentrations of serum folate forms in the US population: national Health and Nutrition Examination Survey 2011–2012. Br J Nutr 113, 19651977.Google Scholar
Pfeiffer, CM, Sternberg, MR, Fazili, Z, et al. (2015) Unmetabolized folic acid is detected in nearly all serum samples from US children, adolescents, and adults. J Nutr 145, 520531.Google Scholar
Husebye, ESN, Wendel, AWK, Gilhus, NE, et al. (2022) Plasma unmetabolized folic acid in pregnancy and risk of autistic traits and language impairment in antiseizure medication-exposed children of women with epilepsy. Am J Clin Nutr 115, 14321440.Google Scholar
Caffrey, A, McNulty, H, Rollins, M, et al. (2021) Effects of maternal folic acid supplementation during the second and third trimesters of pregnancy on neurocognitive development in the child: an 11-year follow-up from a randomised controlled trial. BMC Med 19, 73.Google Scholar
Raghavan, R, Selhub, J, Paul, L, et al. (2020) A prospective birth cohort study on cord blood folate subtypes and risk of autism spectrum disorder. Am J Clin Nutr 112, 13041317.Google Scholar
Wang, S, Wei, J, Wang, D, et al. (2021) The association between folic acid supplementation, maternal folate during pregnancy and intelligence development in infants: a prospective cohort study. Food Sci Hum Wellness 10, 197204.Google Scholar
Levine, SZ, Kodesh, A, Viktorin, A, et al. (2018) Association of maternal use of folic acid and multivitamin supplements in the periods before and during pregnancy with the risk of autism spectrum disorder in offspring. JAMA Psychiatry 75, 176184.Google Scholar
Surén, P, Roth, C, Bresnahan, M, et al. (2013) Association between maternal use of folic acid supplements and risk of autism in children. JAMA 309, 570577.Google Scholar
Schmidt, RJ, Tancredi, DJ, Ozonoff, S, et al. (2012) Maternal periconceptional folic acid intake and risk of autism spectrum disorders and developmental delay in the CHARGE (CHildhood Autism Risks from Genetics and Environment) case-control study. Am J Clin Nutr 96, 8089.Google Scholar
Egorova, O, Myte, R, Schneede, J, et al. (2020) Maternal blood folate status during early pregnancy and occurrence of autism spectrum disorder in offspring: a study of 62 serum biomarkers. Mol Autism 11, 7.Google Scholar
McGowan, E, Hong, X, Selhub, J, et al. (2020) Association between folate metabolites and the development of food allergy in children. J Allergy Clin Immunol Pr 8, 132140.Google Scholar
Whitrow, MJ, Moore, VM, Rumbold, AR, et al. (2009) Effect of supplemental folic acid in pregnancy on childhood asthma: a prospective birth cohort study. Am J Epidemiol 170, 14861493.Google Scholar
Best, KP, Green, TJ, Sulistyoningrum, DC, et al. (2021) Maternal late-pregnancy serum unmetabolized folic acid concentrations are not associated with infant allergic disease: a prospective cohort study. J Nutr 151, 15531560.Google Scholar
Magdelijns, FJH, Mommers, M, Penders, J, et al. (2011) Folic acid use in pregnancy and the development of atopy, asthma, and lung function in childhood. Pediatrics 128, e13544.Google Scholar
Molloy, J, Collier, F, Saffery, R, et al. (2020) Folate levels in pregnancy and offspring food allergy and eczema. Pediatr Allergy Immunol 31, 3846.Google Scholar
Henderson, AM, Tai, DC, Aleliunas, RE, et al. (2018) Maternal folic acid supplementation with vitamin B12 deficiency during pregnancy and lactation affects the metabolic health of adult female offspring but is dependent on offspring diet. FASEB J 32, 50395050.Google Scholar
Krishnaveni, GV, Veena, SR, Yajnik, C, et al. (2014) Association between maternal folate concentrations during pregnancy and insulin resistance in Indian children. Diabetologia 57, 110121.Google Scholar
Yajnik, CS, Deshpande, SS, Jackson, AA, et al. (2008) Vitamin B12 and folate concentrations during pregnancy and insulin resistance in the offspring: the Pune Maternal Nutrition Study. Diabetologia 51, 2938.Google Scholar
Xie, RH, Liu, YJ, Retnakaran, R, et al. (2016) Maternal folate status and obesity/insulin resistance in the offspring: a systematic review. Int J Obes 40, 19.Google Scholar
Pastor-Valero, M, Navarrete-Muoz, EM, Rebagliato, M, et al. (2011) Periconceptional folic acid supplementation and anthropometric measures at birth in a cohort of pregnant women in Valencia, Spain. Br J Nutr 105, 13521360.Google Scholar
Li, N, Li, Z, Ye, R, et al. (2017) Impact of periconceptional folic acid supplementation on low birth weight and small-for-gestational-age infants in China: a large prospective cohort study. J Pediatr 187, 105110.Google Scholar
Crider, KS, Wang, A, Ling, H, et al. (2022) Supplementation and DNA methylation patterns in adolescent offspring. J Nutr 00, 18.Google Scholar
McGee, M, Bainbridge, S & Fontaine-Bisson, B (2018) A crucial role for maternal dietary methyl donor intake in epigenetic programming and fetal growth outcomes. Nutr Rev 76, 469478.Google Scholar
Pfeiffer, CM, Johnson, CL, Jain, RB, et al. (2007) Trends in blood folate and vitamin B-12 concentrations in the United States, 1988–2004. Am J Clin Nutr 86, 718727.Google Scholar
Colapinto, CK, O’Connor, DL & Tremblay, MS (2011) Folate status of the population in the Canadian Health Measures Survey. CMAJ 183, E100E106.Google Scholar
Patti, MA, Braun, JM, Arbuckle, TE, et al. (2022) Associations between folic acid supplement use and folate status biomarkers in the first and third trimesters of pregnancy in the Maternal – Infant Research on Environmental Chemicals (MIREC) Pregnancy Cohort Study. Am J Clin Nutr 116, 18521863.Google Scholar
Masih, SP, Plumptre, L, Ly, A, et al. (2015) Pregnant Canadian women achieve recommended intakes of one-carbon nutrients through prenatal supplementation but the supplement composition, including choline, requires reconsideration. J Nutr 145, 18241834.Google Scholar
Dubois, L, Diasparra, M, Bedard, B, et al. (2017) Adequacy of nutritional intake from food and supplements in a cohort of pregnant women in Québec, Canada: the 3D Cohort Study (Design, Develop, Discover). Am J Clin Nutr 106, 541548.Google Scholar
Plumptre, L, Masih, SP, Ly, A, et al. (2015) High concentrations of folate and unmetabolized folic acid in a cohort of pregnant Canadian women and umbilical cord blood. Am J Clin Nutr 102, 848857.Google Scholar
Fayyaz, F, Wang, F, Jacobs, RL, et al. (2014) vitamin B12, and vitamin B6 status of a group of high socioeconomic status women in the Alberta Pregnancy Outcomes and Nutrition (APrON) cohort. Appl Physiol Nutr Metab 39, 14021408.Google Scholar
Stamm, RA, March, KM, Karakochuk, CD, et al. (2018) Lactating Canadian women consuming 1000 mcg folic acid daily have high circulating serum folic acid above a threshold concentration of serum total folate. J Nutr 148, 16.Google Scholar
Ledowsky, C, Mahimbo, A, Scarf, V, et al. (2022) Women taking a folic acid supplement in countries with mandatory food fortification programs may be exceeding the upper tolerable limit of folic acid: a systematic review. Nutrients 14, 2715.Google Scholar
Turck, D, Bohn, T, Castenmiller, J, et al. (2022) Conversion of calcium-L-methylfolate and (6S)-5-methyltetrahydrofolic acid glucosamine salt into dietary folate equivalents. EFSA J 20, 7452.Google Scholar
Obeid, R, Schön, C, Pietrzik, K, et al. (2020) Pharmacokinetics of sodium and calcium salts of (6S)-5-methyltetrahydrofolic acid compared to folic acid and indirect comparison of the two salts. Nutrients 12, 3623.Google Scholar
EFSA (European Food Safety Authority) (2004) Opinion of the scientific panel on food additives, flavourings, processing aids and materials in contact with food on a request from the commission related to Calcium L-Methylfolate. EFSA J 135, 120.Google Scholar
Liu, Y, Green, TJ, Wong, P, et al. (2013) Microencapsulation of L-5-methyltetrahydrofolic acid with ascorbate improves stability in baked bread products. J Agric Food Chem 61, 247254.Google Scholar
Green, TJ, Liu, Y, Dadgar, S, et al. (2013) Wheat rolls fortified with microencapsulated L-5-methyltetrahydrofolic acid or equimolar folic acid increase blood folate concentrations to a similar extent in healthy men and women. J Nutr 143, 867871.Google Scholar
Kitts, DD & Liu, Y (2015) Encapsulation strategies to stabilize a natural folate, L-5-methyltetrahydrofolic acid, for food fortification practices. In Nanotechnology and Functional Foods: Effective Delivery of Bioactive Ingredients, 1st ed, pp. 142157 [C Sabliov, H Chen & RY Yada, editors]. New Jersey, USA: John Wiley & Sons, Ltd.Google Scholar
Ami, N, Bernstein, M, Boucher, F, et al. (2016) Folate and neural tube defects: the role of supplements and food fortification. Paediatr Child Heal 21, 145149.Google Scholar
Park, Y, Dodd, KW, Kipnis, V, et al. (2018) Comparison of self-reported dietary intakes from the automated self-administered 24-h recall, 4-d food records, and food-frequency questionnaires against recovery biomarkers. Am J Clin Nutr 107, 8093.Google Scholar
Green, TJ, Allen, OB & O’Connor, DL (1998) A three-day weighed food record and a semiquantitative food-frequency questionnaire are valid measures for assessing the folate and vitamin B-12 intakes of women aged 16–19 years. J Nutr 128, 16651671.Google Scholar
Sherwood, KL, Houghton, LA, Tarasuk, V, et al. (2006) One-third of pregnant and lactating women may not be meeting their folate requirements from diet alone based on mandated levels of folic acid fortification. J Nutr 136, 28202826.Google Scholar
Crider, KS, Bailey, LB & Berry, RJ (2011) Folic acid food fortification-its history, effect, concerns, and future directions. Nutrients 3, 370384.Google Scholar
Figure 0

Fig. 1. Chemical structure of folic acid and (6S)-5-methyltetrahydrofolic acid. (a) Folic acid; a synthetic analogue of the folate parent structure. Folic acid is fully oxidised, increasing its stability in supplements and fortified foods, and requires reduction via dihydrofolate reductase for co-enzymatic function. (b) Ca salt of (6S)-5-methyltetrahydrofolic acid (Metafolin®); a synthetic analogue of natural 5-methyltetrahydrofolate.

Figure 1

Fig. 2. Participant flow diagram. The folic acid group includes n 19 at the postpartum visit; this includes n 18 from endline (n 7 chose to not participate in the postpartum blood draw) and n 1 who delivered prematurely (thus, did not participate in the endline visit), but provided a postpartum blood specimen.

Figure 2

Table 1. Participant characteristics of pregnant individuals supplemented with (6S)-5-MTHF or folic acid (Vancouver, Canada, 2019–2021) (Numbers and percentages; mean values and standard deviations; medians and inter-quartile ranges)

Figure 3

Table 2. Baseline biochemical outcomes in pregnant individuals supplemented with (6S)-5-MTHF or folic acid (Vancouver, Canada, 2019–2021) (Medians and inter-quartile ranges; numbers and percentages)

Figure 4

Table 3. Effect of supplemental folate form on late pregnancy and postpartum erythrocyte folate, serum folate and plasma UMFA among pregnant individuals supplemented with (6S)-5-MTHF or folic acid (Vancouver, Canada, 2019–2021) (Mean values and standard deviation; 95 % confidence intervals)

Supplementary material: File

Cochrane et al. supplementary material

Table S1

Download Cochrane et al. supplementary material(File)
File 17.6 KB
Supplementary material: File

Cochrane et al. supplementary material

Table S2

Download Cochrane et al. supplementary material(File)
File 18.8 KB