Folinic acid

Folinic acid

Folinic acid also known as LeucovorinLevofolinic acid or 5-formyltetrahydrofolate (5-formylTHF) is a stable form of folate, which means it is similar to the vitamin Folate (vitamin B9). Folinic acid (Leucovorin) is used in combination with certain chemotherapy drugs (anti-cancer drugs) like 5-fluorouracil (5-FU) to enhance their ability to kill cancer cells or as antidote to lessen the toxic effects of certain drugs such as methotrexate that interfere with folate metabolism by inhibiting dihydrofolate reductase (DHFR) 1, 2. Dihydrofolate reductase (DHFR) is an enzyme that converts dihydrofolate (DHF) to tetrahydrofolate (THF) using NADPH as a cofactor 3. This process is essential for synthesizing purines and thymidylate, which are vital for DNA synthesis and other cellular processes. The human DHFR enzyme is encoded by the DHFR gene and is a key target for antifolate drugs like methotrexate, used to treat cancers and other diseases by inhibiting cell proliferation. Folinic acid (Leucovorin) is also used to treat folate-deficiency anemia 4.

Often, doctors use the terms “folic acid” and “folinic acid” interchangeably, but they are not the same 5, 6. Folic acid is a synthetic (man-made) oxidized version of the water soluble vitamin Folate or vitamin B9 that requires multiple enzymatic steps for activation (see Figure 4 below). Folate is a generic term referring to both natural “folates” found in many foods and particularly in leafy green vegetables and “folic acid“, the man-made oxidized folate that is used in dietary supplements and added to certain foods (fortified foods) 7, 8.

Other synthetic versions of folate include Folinic acid (Leucovorin) also known as 5-formyltetrahydrofolate (5-formylTHF) is a stable form of folate and Levomefolic acid also known as 5-methyltetrahydrofolate (5-MTHF) is a biologically active form of folate. Folic acid has no biological activity unless it is converted into folates 9Folic acid is the most common form of Vitamin B9 and is a synthetic oxidized version of folate that must be metabolized by your body before it can be used (see Figure 4 below). Folic acid is converted by your body first to dihydrofolate (DHF), then tetrahydrofolate (THF) and finally to the biologically active form, levomefolic acid also known as 5-methyltetrahydrofolate (5-MTHF). Folinic acid, also known as leucovorin or 5-formyltetrahydrofolate (5-formylTHF), is a formyl derivative of tetrahydrofolate (THF). Folinic acid exists in two isomeric forms, and only the 6S-isomer [(6S)5-formylTHF also known as Isovorin (calcium levofolinate) or Fusilev (levoleucovorin calcium)] is converted to tetrahydrofolate (THF) and then levomefolic acid (5-MTHF). As a result, unless folinic acid is provided as purified 6S-isomer it does not provide a 1:1 molar equivalent source of folate relative to folic acid. Levomefolic acid also known as 5-methyltetrahydrofolate (5-MTHF) is the biologically active form of folate and the form found in your blood. Levomefolic acid (5-methyltetrahydrofolate [5-MTHF]) does not require enzymatic conversion and can be used directly by your body. The relationship between these forms of folate is summarized in Figure 4 below. Specifically, it demonstrates the point at which these ingredients enter the metabolic pathway.

Folate functions as a coenzyme (a molecule that binds to an enzyme and is essential for its activity but is not permanently altered by the reaction) or cosubstrate in single-carbon transfers in the synthesis of nucleic acids (DNA and RNA) and metabolism of amino acids 10, 11, 12.

  • Nucleic acid metabolism. Folate coenzymes play a vital role in DNA metabolism through two different pathways: (1) The synthesis of DNA from its precursors (thymidine and purines) is dependent on folate coenzymes. (2) A folate coenzyme is required for the synthesis of methionine from homocysteine and the methionine is required for the synthesis of S-adenosylmethionine (SAM) 13. Another folate-dependent reaction, the methylation of deoxyuridylate to thymidylate in the formation of DNA, is required for proper cell division 7. S-adenosyl-methionine (SAMe) is a methyl group (-CH3 or one-carbon unit) donor used in most biological methylation reactions, including the methylation of a number of sites within DNA, RNA, proteins, and phospholipids. The methylation of DNA plays a role in controlling gene expression and is critical during cell differentiation. Defects in DNA methylation have been linked to the development of cancer.
  • Amino acid metabolism. Folate coenzymes are required for the metabolism of several important amino acids (the fundamental building blocks of proteins), namely methionine, cysteine, serine, glycine, and histidine. The synthesis of methionine from homocysteine is catalyzed by methionine synthase, an enzyme that requires not only folate as 5-methyltetrahydrofolate (5-MTHF) but also vitamin B12 (cobalamin). Therefore, folate and/or vitamin B12 deficiency can result in decreased synthesis of methionine and an accumulation of homocysteine. Elevated blood concentrations of homocysteine have been considered for many years to be a risk factor for some chronic diseases, including cardiovascular disease and dementia.

Isolated folate deficiency is uncommon 7. Folate deficiency usually coexists with other nutrient deficiencies because of its strong association with poor diet, alcoholism, and malabsorptive disorders 14. The primary clinical sign of folate deficiency or vitamin B12 deficiency is megaloblastic anemia or macrocytic anemia, which is characterized by large abnormally nucleated immature red blood cells 10, 14. Rapidly dividing cells like those derived from bone marrow are most vulnerable to the effects of folate deficiency since DNA synthesis and cell division are dependent on folate coenzymes. When folate supply to the rapidly dividing cells of the bone marrow is inadequate, blood cell division is reduced, resulting in fewer but larger red blood cells. Neutrophils, a type of white blood cell, become hypersegmented, a change that can be found by examining a blood sample microscopically. Because normal red blood cells have a lifetime in the circulation of approximately four months (120 days), it can take months for folate-deficient individuals to develop the characteristic megaloblastic anemia. Progression of megaloblastic anemia leads to a decreased oxygen carrying capacity of the blood and may ultimately result in symptoms of fatigue, weakness, and shortness of breath 9. Folate deficiency can also produce soreness in and shallow ulcerations on the tongue and oral mucosa; changes in skin, hair, or fingernail pigmentation; gastrointestinal symptoms; and elevated blood concentrations of homocysteine 10, 14, 15. Individuals in the early stages of folate deficiency may not show obvious symptoms, but blood concentrations of homocysteine may increase. Yet, the concentration of circulating homocysteine is not a specific indicator of folate status, as elevated homocysteine can be the result of vitamin B12 and other B-vitamin deficiencies, lifestyle factors, and kidney disease. Subclinical folate deficiency is typically detected by measurement of folate concentrations in serum/plasma or in red blood cells.

It is important to point out that megaloblastic anemia resulting from folate deficiency is identical to the megaloblastic anemia resulting from vitamin B12 deficiency, and further clinical testing is required to diagnose the true cause of megaloblastic anemia. Megaloblastic anemia symptoms include weakness, fatigue, difficulty concentrating, irritability, headache, heart palpitations, and shortness of breath 9, 14.

Women with insufficient folate intakes are at increased risk of giving birth to infants with neural tube defects that result in malformations of the spine (spina bifida), skull, and brain (anencephaly) 9. The prevalence rate of spina bifida and anencephaly (the two most common types of neural tube defects) in the United States is 5.5 to 6.5 per 10,000 births 16. Inadequate maternal folate status has also been associated with low infant birth weight, preterm delivery, and fetal growth retardation 10, 17.

Because of folate’s role in the synthesis of DNA and other critical cell components, folate is especially important during phases of rapid cell growth 18. Although the mechanism has not been fully established, clinical trial evidence shows that adequate periconceptional (the critical period of time from before conception to early pregnancy, generally encompassing a few weeks before and a few weeks after conception) folic acid consumption by women prevents a substantial proportion of neural tube defects 19, 20, 21, 22, 23.

Since 1998, when mandatory folic acid fortification began in the United States where vitamins, minerals, and other essential nutrients were added to food products during processing to improve its nutritional quality and provide a public health benefit, such as preventing or correcting nutrient deficiencies, neural tube defect rates have declined by 28% 16. However, significant racial and ethnic disparities persist. Neural tube defect prevalence rates are highest among Hispanic women and lowest among non-Hispanic black women. Factors that might contribute to these disparities include differences in dietary and supplement-taking practices 24 as well as factors other than folate status such as maternal diabetes, obesity, and intake of other nutrients (e.g., vitamin B12), which are also believed to affect the risk of neural tube defects 23, 25, 26, 27. In addition, women with genetic mutation in 677C>T MTHFR gene, which is more common in Hispanics than Caucasians, Asians, and African Americans, might have an increased risk of neural tube defects 10, 12, 28, 29. Another consideration is the fact that the data on neural tube defect prevalence rates were collected before 2016, when FDA approved the voluntary addition of folic acid to corn masa flour, an ingredient commonly consumed by Hispanic populations 30. Whether this policy change has affected the disparities in neural tube defect rates between Hispanic women and other populations is not yet known.

During pregnancy, demands for folate increase because of its role in nucleic acid synthesis 17. To meet this need, the the Food and Nutrition Board at the National Academies of Sciences, Engineering, and Medicine increased the folate Recommended Dietary Allowance (RDA) from 400 micrograms (400 mcg) dietary folate equivalent (DFE)/day for nonpregnant women to 600 micrograms (600 mcg) dietary folate equivalent (DFE)/day during pregnancy 9. This level of folate intake might be difficult for some women to achieve through diet alone. The American College of Obstetricians and Gynecologists recommends a prenatal vitamin supplement for most pregnant women to ensure that they obtain adequate amounts of folic acid and other nutrients 31, 32.

All women of reproductive age (15–45 years) should take folic acid supplementation. For average-risk women, supplementation with 600 micrograms of folic acid (1000 mcg DFE) per day is adequate 31. Because it’s hard to get this much folic acid from food alone, you should take a daily prenatal vitamin with at least 600 micrograms (1000 mcg DFE) starting at least 1 month before pregnancy and during the first 12 weeks of pregnancy. Women at increased risk of neural tube defects, including women with a prior pregnancy with an neural tube defect or women with seizure disorders, should be counseled to take 4 milligrams (4 mg) of folic acid daily for at least 3 months before pregnancy and for the first 3 months of pregnancy 33. Because of the risk of vitamin A toxicity, women who need additional folic acid should not take additional prenatal vitamins; instead, women at higher risk of neural tube defects should be prescribed additional folic acid supplements. Most prenatal multivitamins contain adequate amounts of folic acid for average-risk-women 34. Prenatal vitamins use also is associated with a lower risk of miscarriage 35. Moderate caffeine consumption (less than 200 mg per day) does not appear to be a major contributing factor in miscarriage or preterm birth 60.

Dietary Folate Equivalent (DFE) is defined as 7:

  • 1 mcg dietary folate equivalent (DFE) = 1 mcg food folate
  • 1 mcg dietary folate equivalent (DFE) = 0.6 mcg folic acid from fortified foods or dietary supplements consumed with foods (1,000 mcg folic acid from fortified foods or dietary supplements is equivalent to 1,667 mcg DFE because 0.6 mcg folic acid = 1 mcg DFE)
  • 1 mcg dietary folate equivalent (DFE) = 0.5 mcg folic acid from dietary supplements taken on an empty stomach

Factors for converting mcg dietary folate equivalent (DFE) to mcg for supplemental folate in the form of 5-MTHF have not been formally established 7.

Food folates are in the potent tetrahydrofolate (THF) form and usually have additional glutamate residues, making them polyglutamates 10. Folic acid, the synthetic folate, is the fully oxidized monoglutamate form of folate (vitamin B9) that is used in fortified foods and most dietary supplements 7. Some dietary supplements also contain folate in the monoglutamyl form, 5-MTHF also known as L-5-MTHF, 5-methyl-folate, L-methylfolate, and methylfolate 7.

Heating during cooking destroys folic acid 36. Folate is absorbed in the jejunum (the 2nd part of your small intestine) by active and passive transport mechanisms across the intestinal wall 36. Folate or folic acid is a water-soluble type of vitamin B. This means it is not stored in the fat tissues of the body. Leftover amounts of the vitamin leave the body through the urine. The body has about 1,000-20,000 mcg of folate stores, and adults need about 400 mcg/day to replenish the daily losses. Folate deficiency may take 8-16 weeks to become evident 36.

Folate is poorly stored, and folate deficiency can develop in weeks to months in persons with folate-deficient diets. Most of the serum folate is present in the inactive 5-methyltetrahydrofolate (5-methyl THF) form 36. Upon entering cells, 5-methyltetrahydrofolate (5-methyl THF) demethylates to tetrahydrofolate (THF), the biologically active form of folate that is involved in folate-dependent enzymatic reactions. Cobalamin (vitamin B12) serves as a co-factor for this demethylation to occur, and in the absence of vitamin B12, folate is “trapped” inside cells as 5-methyltetrahydrofolate (5-methyl THF). Tetrahydrofolate (THF) is involved in the formation of many coenzymes in metabolic systems, particularly for purine and pyrimidine synthesis, nucleoprotein synthesis, and maintenance in red blood cells formation 37. The deficiency of folate, as a result, leads to impairment of cell division, accumulation of toxic metabolites, and impartment of methylation reactions required for regulation of gene expression, resulting in megaloblastic anemia, which is characterized by large, abnormally nucleated red blood cells 14, 10.

The total body content of folate is estimated to be 10 to 30 mg; about half of this amount is stored in the liver and the remainder in blood and body tissues. A serum folate concentration is commonly used to assess folate status, with a value above 3 nanograms (ng)/mL indicating adequacy 38. This indicator, however, is sensitive to recent dietary intake, so it might not reflect long-term status. Erythrocyte folate concentration provides a longer-term measure of folate intakes, so when day-to-day folate intakes are variable—such as in people who are ill and whose folate intake has recently declined—it might be a better indicator of tissue folate stores than serum folate concentration 39. An erythrocyte folate concentration above 140 ng/mL indicates adequate folate status 39, although some researchers have suggested that higher values are optimal for preventing neural tube defects 40.

A combination of serum or red blood cell concentration and indicators of metabolic function can also be used to assess folate status. Plasma homocysteine concentration is a commonly used functional indicator of folate status because homocysteine levels rise when the body cannot convert homocysteine to methionine due to a 5-methyl-THF deficiency. Homocysteine levels, however, are not a highly specific indicator of folate status because they can be influenced by other factors, including kidney dysfunction and deficiencies of vitamin B12 and other micronutrients 41. The most commonly used cutoff value for elevated homocysteine is 16 micromoles/L, although slightly lower values of 12 to 14 micromoles/L have also been used 39.

Folate-deficiency anemia is most common during pregnancy. Other causes of folate-deficiency anemia include alcoholism and certain medicines to treat seizures, anxiety, or arthritis.

The symptoms of folate-deficiency anemia include:

  • Fatigue
  • Headache
  • Pale skin
  • Sore mouth and tongue

The latest research reveals the following about folic acid deficiency 36:

  • There may be a link between elevated homocysteine (a marker for an increased risk for arteriosclerosis) and folate deficiency.
  • A lowering of the risk of stroke but not adverse cardiac event when hyperhomocysteinemia is corrected with folic acid
  • Reduction in the incidence of neural tube defects with folic acid supplementation during pregnancy.
  • Lack of folic acid during pregnancy may increase the risk of diabetes-associated congenital disabilities and autism.
  • Maternal folic acid during pregnancy may lower the risk of childhood leukemia.
  • Folic acid supplementation may increase the risk of cancer.

If you have folate-deficiency anemia, your doctor may recommend taking folic acid vitamins and eating more foods with folate.

Folic acid is available in multivitamins and prenatal vitamins, supplements containing other B-complex vitamins, and supplements containing only folic acid. Common doses range from 680 to 1,360 mcg dietary folate equivalent (DFE) (400 to 800 mcg folic acid) in supplements for adults and 340 to 680 mcg DFE (200 to 400 mcg folic acid) in children’s multivitamins 42. About 85% of supplemental folic acid, when taken with food, is bioavailable 11, 14. When consumed without food, nearly 100% of supplemental folic acid is bioavailable 7.

Figure 1. Folinic acid (Leucovorin)

Leucovorin
[Source 43 ]

Figure 2. Folic acid

Folic acid

Footnotes: Folic acid (pteroylglutamic acid) is a complex chemical compound whose molecule contains three components: a pteridine derivative (2-amino-4-hydroxy-6-methylpteridine), p-aminobenzoic acid (PABA) and glutamic acid 44.

[Source 45 ]

Figure 3. Folic acid biologically active derivatives

Folic acid biologically active derivatives

Footnotes: (A) dihydrofolate (DHF); (B) tetrahydrofolate (THF); (C) 5-methyltetrahydrofolate (5-methylTHF)

[Source 45 ]

Figure 4. Folic acid metabolism

folic acid metabolic pathway
Folic acid metabolism

Footnotes: This diagram shows the process by which folate and folic acid is used for DNA methylation (a biochemical reaction resulting in the addition of a methyl group [-CH3] to another molecule). The methylenetetrahydrofolate reductase (MTHFR) 677C/T genetic mutation reduces enzyme activity 46 and may help to divert the available methyl (-CH3) groups from the DNA methylation pathway toward the DNA synthesis pathway 47, 48, 49. The pathway is complex and highly regulated with feedback loops and interactions not shown in the schematic. Gene names for enzymes are in italics and cofactors (compounds that are essential for the activity of an enzyme) are in parentheses.

Abbreviations: DHF = dihydrofolate; DHFR = dihydrofolate reductase; DNMT = DNA methyltransferase; dTMP = thymidylate; dUMP = deoxyuridine monophosphate; MS = methionine synthase; MTHFR = methylenetetrahydrofolate reductase; SAH = S-adenosylhomocysteine; SAM = S-adenosylmethionine; SHMT = serine hydroxymethyltransferase; THF = tetrahydrofolate; TS = thymidylate synthase.

[Source 50 ]

Figure 5. Folate functions

Folate functions

Footnotes: The only function of folate coenzymes (a molecule that binds to an enzyme and is essential for its activity but is not permanently altered by the reaction) in the body appears to be in mediating the transfer of one-carbon (-CH3 methyl group) units 51. Folate coenzymes act as acceptors and donors of one-carbon units in a variety of reactions critical to the metabolism of nucleic acids and amino acids 52.

[Source 53 ]

Figure 6. Folate and methionine cycle

Folate and methionine cycle

Footnote: Folate and methionine cycle. The Folate cycle begins with the conversion of dietary folate (vitamin B9) into dihydrofolate (DHF) then tetrahydrofolate (THF) by dihydrofolate reductase (DHFR). Next, tetrahydrofolate (THF) is converted to 5, 10-methylene-THF by serine hydroxymethyltransferase (SHMT) before being reduced into 5-methyl-THF (5-mTHF) by methylenetetrahydrofolate reductase (MTHFR). As part of the methionine cycle, 5-methyl-THF (5-mTHF) donates its carbon group to convert homocysteine (Hcy) to methionine (Met), which is catalyzed by methionine synthase (MS) and requires vitamin B12 as a cofactor, hence initiating Methionine cycle. In turn, methionine (Met) is used by methionine adenosyltransferase (MAT) to generate S-Adenosyl-Methionine (SAM) – the principal donor of methyl groups for DNA and proteins methylation. Thus, SAM is used by different methyltransferases, resulting in S-adenosylhomocysteine after its demethylation. Finally, S-adenosylhomocysteine hydrolase (SAHH) mediates deadenylation of S-adenosylhomocysteine to hcysteine, enclosing the methionine cycle. Homocysteine can be used by cystathionine synthase (CBS), which converts it to cystathionine. In turn, cystathionine is a substrate for cystathionine gamma-lyase (CTH), which uses it for synthesis of cysteine. Cysteine is required for the synthesis of proteins as well as for generation of taurine and glutathione, the latter is one of the critical molecules for redox homeostasis.

Abbreviations: Ado = adenosine; ATP = adeno-sine triphosphate; vitamin B6, vitamin B9, vitamin B12; Cbs = cystathionine beta synthase; DHF = dihydrofolate; DHFR = dihydrofolate reductase; Cys = cysteine; GSH = glutathione; Hcy = homocysteine; MAT = methionine adenosyltransferase; Met = methionine; 5–methylTHF = 5, 10–methyleneTHF; MTHFR = methylenetetrahydrofolate reductase or 5,10-methylene-trahydrofolate reductase; MS = methionine synthase, SAM = S-adenosyl-methionine; SAH = S-adenosylhomocysteine; SHMT = hydroxymethyltransferase; THF = tetrahydrofolate.

[Source 54 ]

Figure 7. Homocysteine metabolism

Homocysteine metabolism

Footnote: Schematic representation of pathways of homocysteine metabolism. The metabolism of homocysteine, an intermediate in the metabolism of sulfur-containing amino acids, provides an example of the interrelationships among nutrients necessary for optimal physiological function and health. Healthy individuals utilize two different pathways to metabolize homocysteine (Figure 3). One pathway (methionine synthase) synthesizes methionine from homocysteine and is dependent on both folate and vitamin B12 as cofactors. The other pathway converts homocysteine to another amino acid, cysteine, and requires two vitamin B6-dependent enzymes. Thus, the concentration of homocysteine in the blood is regulated by three B-vitamins: folate (vitamin B9), vitamin B12 (cobalamin), and vitamin B6 (pyridoxine) 55. In some individuals, riboflavin (vitamin B2) is also involved in the regulation of homocysteine concentrations.

Abbreviations: DHFR = dihydrofolate reductase; THF = tetrahydrofolate; SHMT = serinehydroxymethyltransferase; MTHF = methylenetetrahydrofolate; MTHFR = 5,10-methylene-THF reductase; ATP = adenosine triphosphate; MAT = methionine adenosyltransferase; ADP = adenosine diphosphate; SAM = S-adenosylmethionine; SAH = S-adenosylhomocysteine; BHMT = betaine-Hcy S-methyltransferase; CBS = cystathionine beta-synthase; CSE = cystathionase; GSH = glutathione; H2S = hydrogen sulphide

[Source 56 ]

What is the difference between Folinic acid and Folic acid?

Folic acid is a synthetic (man-made) oxidized version of the water soluble vitamin Folate (vitamin B9) or Folacin that requires multiple enzymatic steps for activation (see Figure 4 above). Folate is a generic term referring to both natural “folates” found in many foods and particularly in leafy green vegetables and “folic acid“, the man-made oxidized folate that is used in dietary supplements and added to certain foods (fortified foods) 7, 8. Other synthetic versions of folate include Folinic acid (Leucovorin) also known as 5-formyltetrahydrofolate (5-formylTHF) is a stable form of folate and Levomefolic acid also known as 5-methyltetrahydrofolate (5-MTHF) a biologically active form of folate. Folic acid has no biological activity unless it is converted into tetrahydrofolate (THF) 9.

Folic acid is the most common form of Vitamin B9 and is a synthetic oxidized version of folate that must be metabolized by your body before it can be used. Folic acid is converted by your body first to dihydrofolate (DHF), then tetrahydrofolate (THF) and finally to the biologically active form, 5-methyltetrahydrofolate (5-MTHF) also known as levomefolic acid.

Folinic acid, also known as leucovorin or 5-formyltetrahydrofolate (5-formylTHF), is a man-made molecule, a formyl derivative of tetrahydrofolate (THF) that is readily converted to 5-10 MTHF (5,10-methylenetetrahydrofolate) and 5-methyltetrahydrofolate (5-MTHF) a biologically active form of folate without requiring the action of the enzyme methylenetetrahydrofolate reductase (MTHFR) (see Figure 4 above).

Folinic acid exists in two isomeric forms, and only the 6S-isomer [(6S)5-formylTHF also known as Isovorin (calcium levofolinate) or Fusilev (levoleucovorin calcium)] is converted to tetrahydrofolate (THF) and then levomefolic acid (5-MTHF) the biologically active form of folate. As a result, unless folinic acid is provided as purified 6S-isomer [(6S)5-formylTHF also known as Isovorin (calcium levofolinate) or Fusilev (levoleucovorin calcium)] it does not provide a 1:1 molar equivalent source of folate relative to folic acid.

Levomefolic acid also known as 5-methyltetrahydrofolate (5-MTHF) is the biologically active form of folate and the form found in your blood. Levomefolic acid (5-methyltetrahydrofolate [5-MTHF]) does not require enzymatic conversion and can be used directly by your body. The relationship between these forms of folate is summarized in Figure 4 above. Specifically, it demonstrates the point at which these ingredients enter the metabolic pathway.

Your body needs folate to make DNA, other genetic material, several amino acids, as well as in methylation reactions (a biochemical reaction resulting in the addition of a methyl group [-CH3] to another molecule). Folic acid and folate also help your body make healthy new red blood cells. Red blood cells carry oxygen to all the parts of your body. If your body does not make enough red blood cells, you can develop anemia. Anemia happens when your blood cannot carry enough oxygen to your body, which makes you pale, tired, or weak. Also, if you do not get enough folic acid, you could develop a type of anemia called folate-deficiency anemia 4.

Folinic acid mechanism of action

Folates (naturally occurring vitamin B9) and folic acid (synthetic folate) are not biologically active and must be converted into tetrahydrofolate (THF) through dihydrofolate reductase (DHFR). Folinic acid (Leucovorin or 5-formyltetrahydrofolate [5-formylTHF]) does not require dihydrofolate reductase (DHFR) for conversion into tetrahydrofolate (THF) 5. Tetrahydrofolate (THF) and its derivatives then participate in thymidylate and purine synthesis as they are essential in carrying out one-carbon transfer reactions in the body 57. These reactions are essential in the generation of nucleic acids, the regulation of gene expression, and the overall stability of the genome 57. Serine, methionine, histidine, and glycine metabolism also depend on such substrates and reactions. Therefore, folinic acid ultimately plays a crucial role in normal metabolism and gene regulation 5. Furthermore, folinic acid is essential for synthesizing methionine from homocysteine, as this is a methylation reaction 5.

When consumed, food folates are hydrolyzed to the monoglutamate form in the gut prior to absorption by active transport across the intestinal mucosa 39, 10. Passive diffusion also occurs when pharmacological doses of folic acid (the man made folate) are consumed 39. Before entering the bloodstream, the monoglutamate form is reduced to tetrahydrofolate (THF) and converted to either methyl or formyl forms. The main form of folate in plasma is 5-methyl-THF (5-MTHF). Folic acid can also be found in the blood unaltered known as unmetabolized folic acid, but whether this form has any biological activity or can be used as a biomarker of status is not known 58.

Some dietary supplements also contain folate in the monoglutamyl form, 5-methylenetetrahydrofolate or 5-methyl-folate (5-MTHF), also known as L-5-MTHF, L-methylfolate, and methylfolate 7. For some people with an methylenetetrahydrofolate reductase polymorphism (MTHFR polymorphism), supplementation with 5-methylenetetrahydrofolate (5-MTHF) might be more beneficial than with folic acid 5, 59. The bioavailability of 5-methylenetetrahydrofolate (5-MTHF) in supplements is the same as or greater than that of folic acid 60, 61, 62, 28. However, conversion factors between mcg and mcg dietary folate equivalent (DFE) for 5-MTHF (5-methyl-folate) have not been formally established.

Folate and its coenzymes require transporters to cross cell membranes. Folate transporters include the reduced folate carrier (RFC), the proton-coupled folate transporter (PCFT), and the folate receptor proteins, folate receptor alpha (FRα) and folate receptor beta (FRβ). Folate homeostasis is supported by the ubiquitous distribution of folate transporters, although abundance and importance vary among tissues 63. Proton-coupled folate transporter (PCFT) plays a major role in folate intestinal transport since mutations affecting the SLC46A1 gene encoding PCFT cause hereditary folate malabsorption. Defective PCFT (proton-coupled folate transporter) also leads to impaired folate transport into the brain. Folate receptor alpha (FRα) and reduced folate carrier (RFC) are also critical for folate transport across the blood-brain barrier when extracellular folate is either low or high, respectively. Folate is essential for the proper development of the embryo and the fetus. The placenta is known to concentrate folate to the fetal circulation, leading to higher folate concentrations in the fetus compared to those found in the pregnant woman. All three types of receptors have been associated with folate transport across the placenta during pregnancy 64.

Pharmacokinetics

  • Absorption: Folinic acid is rapidly and almost completely absorbed following oral administration. Bioavailability is dose-dependent 65.
  • Distribution: IV folinic acid administration results in a greater volume of distribution than oral, with oral dosing showing a lower maximum blood concentration primarily due to first-pass metabolism 65.
  • Metabolism: Folinic acid is metabolized in the liver and gastrointestinal tract and has an active metabolite. The average half-life is approximately 6 hours.
  • Elimination: Excretion is 80% to 90% in urine and 5% to 8% in the feces.

Folinic acid uses

Indications for folate supplementation include preventing several diseases, such as ulcerative colitis, neural tube defects, and cognitive dysfunction in older patients 5. Doctors consider folinic acid or leucovorin (5-formyltetrahydrofolate [5-formylTHF]) supplementation superior to folic acid supplementation because folinic acid can reach higher concentrations in plasma and function in the face of defective folate metabolism 5, 66.

The 2 main roles of folinic acid in clinical practice are to counteract the effects of folic acid antagonists (antifolates) and enhance the impact of fluoropyrimidines 65. The former function is possible because folinic acid can enter cells through the reduced folate carrier and subsequently be converted to tetrahydrofolate despite the presence of methotrexate, thereby “rescuing” these cells from methotrexate toxicity 67.

Most folic acid antagonists (antifolates) share a similar mechanism of action that includes the inhibition of dihydrofolate reductase (DHFR), the enzyme responsible for generating the functional tetrahydrofolate molecule. Folinic acid does not require dihydrofolate reductase (DHFR) to convert into its active derivatives. In this setting, folinic acid is an antidote that rescues these cells from the chemotherapeutic toxicities of folate antagonists such as methotrexate 57.

While folinic acid acts as antidote to the adverse effects of methotrexate, folinic acid also functions to enhance the anti-cancer effects of the drug fluorouracil (5FU). In the cell, fluorouracil (5FU) converts to fluoro-deoxy uridylic acid, a molecule that inhibits thymidylate synthase. Thymidylate synthase is an enzyme that is important in DNA repair and replication. The functional derivative of folinic acid, 5-10 MTHF (5,10-methylenetetrahydrofolate), stabilizes the bound fluoro-deoxy uridylic acid to thymidylate synthase 2. This interaction yields a ternary complex known as the thymidylate synthase 5-fluorodeoxyuridine monophosphate-methylenetetrahydrofolate complex, which inhibits thymidylate synthase 2. Increased cellular amounts of folinic acid derivatives lead to increased stability of the aforementioned inhibitory complex, which leads to a depletion of thymidylate synthesis and disrupts DNA synthesis and repair 65, 66.

Folinic Acid FDA-Approved Indications

  • Folinic acid enhances the cytotoxic effects and effectiveness of the cancer drug fluorouracil (5FU) in the cell by stabilizing its binding to its target enzyme 68. This finding has revolutionized the treatment of several cancers, most notably colorectal cancer, which, according to the American Cancer Society, represents the third leading cause of cancer-related death in both sexes 69. The combination of folinic acid with fluorouracil (5FU) is an FDA-approved indication for the palliative treatment of colorectal cancer. Specifically, patients with advanced, nonresectable adenocarcinoma of the colon who are receiving treatment with folinic acid and 5-FU display more prolonged progression-free survival and response rates 68. High-dose folinic acid + fluorouracil (5FU) regimens can also be used as adjuvant therapy for resectable colorectal cancer. Folinic acid aids in producing a significant increase in disease-free survival compared to patients who receive treatment with surgical resection alone 70
  • Folinic acid (leucovorin) is helpful as an antidote to folic acid antagonists or antifolates (ie, methotrexate, trimethoprim [an antibiotic] and pyrimethamine [an antimalarial drug]), which are class of drugs that inhibit the use of folic acid (folate) by cells to make DNA, particularly by blocking the enzyme dihydrofolate reductase (DHFR) and may kill cancer cells 71, 67. Frequently referred to as “leucovorin rescue” or “folinic acid rescue therapy” relates to folinic acid administration with the chemotherapy drug methotrexate. Methotrexate works by inhibiting dihydrofolate reductase (DHFR), shutting down folate metabolism and killing fast-dividing cancer cells. Giving folinic acid bypasses this blockade, “rescuing” healthy cells from the toxic effects of methotrexate therapy and reducing side effects. Methotrexate has a variety of indications in the treatment of various cancers and autoimmune disorders. Therefore, all conditions requiring methotrexate therapy, especially in high dosages, benefit from folinic acid as rescue therapy 67. As an antidote, folinic acid limits bone marrow suppression, gastrointestinal toxicity, kidney toxicity, and neurotoxicity that can result secondary to high dosages of methotrexate and other folic acid antagonists 67. Study results show that administering folinic acid after weekly doses of methotrexate reduces the incidence of transaminitis (an elevated level of liver enzymes called transaminases (specifically AST and ALT in the blood, indicating liver injury or inflammation), gastrointestinal complications, and stomatitis (oral ulcers) 72. Folinic acid supplementation during methotrexate therapy is so essential that chemotherapy protocols using methotrexate also include detailed recommendations regarding folinic acid rescue administration 67. For reducing toxicity and counteracting the effects of high-dose methotrexate, folinic acid is FDA-approved in both adults and children.
  • The intravenous formulation of folinic acid (leucovorin calcium) is also FDA-approved for use in adults and children to treat megaloblastic anemia in patients who have normal vitamin B12 levels and in whom oral therapy is not possible 73.
  • Folinic acid is also approved to treat folate-deficiency-associated megaloblastic anemia.

Off-Label Non-FDA-approved uses for folinic acid

  • Treating Breast Cancer: Similar to its use in colorectal cancer, folinic acid has also been shown to potentiate the effects of fluorouracil (5FU) in breast cancer. This regimen is currently non-FDA approved, partly because there is insufficient evidence that folinic acid can increase the therapeutic index of fluorouracil (5FU) in breast cancer and because many such clinical trials are ongoing 2.
  • Regimens including folinic acid and fluorouracil (5FU) also have non-FDA-approved indications for treating unresectable/advanced gallbladder and biliary tree carcinoma, gastric cancer, squamous cell carcinoma of the head and neck, and resectable pancreatic cancer 74, 75, 76, 77.
  • Combination chemotherapy regimens that include folinic acid have been effective in various non-Hodgkin lymphomas. Methotrexate and folinic acid rescue, combined with other chemotherapy agents (ie, doxorubicin, cyclophosphamide), have shown a complete response rate of 84% in treating diffuse large cell lymphoma 78.
  • For patients who require prophylactic therapy for toxoplasmosis and who are unable to tolerate sulfamethoxazole/trimethoprim, folinic acid is recommended in combination with clindamycin and pyrimethamine as an alternative. Folinic acid, in conjunction with pyrimethamine and sulfadoxine, is another option 79.
  • For patients requiring prophylactic treatment for Pneumocystis jirovecii pneumonia and who cannot tolerate sulfamethoxazole/trimethoprim, an alternative therapy can be used that includes folinic acid in combination with pyrimethamine and dapsone, among other combinations 80.
  • Management of Mitochondrial Disorders: Folinic acid is used in some cases to support cellular energy production pathways.
  • Specialized Supplementation: Folinic acid is increasingly being used in supplements targeting individuals with the MTHFR gene mutation and in Cerebral Folate Deficiency and Autism Spectrum Disorder, especially in those patients that are positive for folate receptor autoantibodies.
  • Serum plasma levels of homocysteine and folate derivatives have an inverse relationship. In particular, the methionine cycle, which uses homocysteine as a substrate, is sensitive to folate deficiency. Therefore, plasma homocysteine levels are markedly increased when cells have functional depletion of folinic acid 5. In patients with hyperhomocysteinemia, folinic acid has been found to reduce the plasma level of homocysteine, particularly in patients on hemodialysis 81. This finding suggests that folinic acid intake may reduce the risk of cardiovascular disease. Furthermore, folinic acid levels can be an indirect indicator of homocysteine levels 5.
  • Patients with repeated nitrous oxide exposure or requiring treatment with long-term nitrous oxide may benefit from folinic acid prophylaxis to prevent bone marrow suppression 82.

Folinic acid in Pregnancy

Everyone needs folic acid to be healthy. But folic acid is especially important for women of reproductive age (15–45 years):

Before and during pregnancy, folic acid protects your unborn child against serious birth defects called neural tube defects that result in malformations of the spine (spina bifida), skull, and brain (anencephaly) 9. The prevalence rate of spina bifida and anencephaly (the two most common types of neural tube defects) in the United States is 5.5 to 6.5 per 10,000 births 16. Inadequate maternal folate status has also been associated with low infant birth weight, preterm delivery, and fetal growth retardation 10, 17. These birth defects happen in the first few weeks of pregnancy, often before a woman knows she is pregnant. Folic acid might also help prevent other types of birth defects and early pregnancy loss (miscarriage). Since about half of all pregnancies in the United States are unplanned 83, experts recommend all women get enough folic acid via supplements even if you are not trying to get pregnant. For average-risk women, supplementation with 600 micrograms (600 mcg) of folic acid per day is adequate 31. Because it’s hard to get this much folic acid from food alone, you should take a daily prenatal vitamin with at least 600 micrograms (600 mcg) folic acid starting at least 1 month before pregnancy and during the first 12 weeks of pregnancy. Women at increased risk of neural tube defects, including women with a prior pregnancy with an neural tube defect or women with seizure disorders, should be counseled to take 4 milligrams (4 mg) of folic acid daily for at least 3 months before pregnancy and for the first 3 months of pregnancy 33. Because of the risk of vitamin A toxicity, women who need additional folic acid should not take additional prenatal vitamins; instead, women at higher risk of neural tube defects should be prescribed additional folic acid supplements. Most prenatal multivitamins contain adequate amounts of folic acid for average-risk-women 34. Prenatal vitamins use also is associated with a lower risk of miscarriage 35. Moderate caffeine consumption (less than 200 mg per day) does not appear to be a major contributing factor in miscarriage or preterm birth 60.

Not getting enough folic acid can lead to a type of anemia called folate-deficiency anemia. Folate-deficiency anemia is more common in women of childbearing age than in men.

Preterm birth, congenital heart defects, and other congenital anomalies

According to observational studies, folic acid supplementation might increase mean gestational age and lower the risk of preterm birth 10, 84. In addition, folic acid in combination with a multivitamin supplement helps minimize the risk of congenital heart defects, possibly because heart tissue development depends on cells that require large amounts of folate 10, 85.

The authors of a large population-based cohort study of about 98% of all births in Canada from 1990 to 2011 concluded that folic acid fortification of foods was associated with an 11% reduction in the rate of nonchromosomal congenital heart defects 86. In a population-based case-control study in Atlanta involving 3,987 infants, congenital heart defects were 24% less common in the infants of women who took multivitamins containing folic acid during the periconceptional period than in the infants of women who did not 87. A case-control study in California in 866 infants had similar results 88. However, it is not possible to determine whether the findings from these studies could be attributed to components of multivitamins other than folic acid.

Studies have also found associations between the use of folic acid in combination with multivitamin supplements and reduced occurrence at birth of urinary tract anomalies, oral facial clefts, limb defects, and hydrocephalus, but the results of these studies have been inconsistent 85.

Additional research is needed to fully understand the extent to which maternal consumption of folic acid might affect the risk of these adverse birth outcomes. However, folic acid’s established role in preventing neural tube defects and possibly other birth defects underscores its importance during the periconceptional period.

Metabolic diseases

Folinic acid (leucovorin), a tetrahydrofolic acid derivative, is used in the clinical management of rare inborn errors that affect folate transport or metabolism 89. Such conditions are of autosomal recessive inheritance, meaning only individuals receiving two copies of the mutated gene (one from each parent) develop the disease. Folinic acid can be administered orally, intravenously, or intramuscularly.

Hereditary folate malabsorption

Hereditary folate malabsorption is caused by mutations in the SLC46A1 gene that provides instructions for making a protein called the proton-coupled folate transporter (PCFT) 90, 91. The proton-coupled folate transporter (PCFT) protein is found within the membrane of cells, where it helps transport folates into the cell. PCFT (proton-coupled folate transporter) is primarily found in cells that line the walls of the small intestine. These cells have fingerlike projections called microvilli that absorb nutrients from food as it passes through the intestine. Based on their appearance, groups of these microvilli are known collectively as the brush border. PCFT (proton-coupled folate transporter) is involved in transporting folates from food across the brush border membrane so they can be used by the body 92. PCFT (proton-coupled folate transporter) is also found in the brain, where it is involved in the transport of folates between the brain and the surrounding fluid (cerebrospinal fluid) 92. Patients with hereditary folate malabsorption due to SLC46A1 gene mutations present with low to undetectable concentrations of folate in their serum and cerebrospinal fluid (CSF); anemia (often macrocytic), low platelets count (thrombocytopenia), and/or low number of all blood cells (pancytopenia); impaired immune responses that increase susceptibility to infections; and a general failure to thrive 93, 90. Neurologic symptoms, including developmental delays, cognitive disorders, and seizures, have also been observed 94, 90. Early treatment with intramuscular or high-dose oral folinic acid (leucovorin or 5-formyltetrahydrofolate [5-formylTHF]) or, preferably, the active isomer of 5-formyltetrahydrofolate (calcium levofolinate [Isovorin] or levoleucovorin calcium [Fusilev]) readily corrects the systemic folate deficiency and, if the dose is sufficient, can achieve cerebrospinal fluid (CSF) folate levels that prevent or mitigate the neurologic consequences of hereditary folate malabsorption 90, 95, 96. The active, physiologic, isomer of 5-formyltetrahydrofolate [5-formylTHF] is (6S)5-formylTHF also known as Isovorin (calcium levofolinate) or Fusilev (levoleucovorin calcium); it is available for intramuscular administration. The biologic impact of the active isomer is twice that of folinic acid (leucovorin or 5-formyltetrahydrofolate [5-formylTHF]) when the dose is the same 90. Isovorin (calcium levofolinate) or Fusilev (levoleucovorin calcium) is the preferred form of folate for treating hereditary folate malabsorption, if it is available and cost is not a limiting factor. The active isomer should be utilized especially when there is refractory neurologic disease. Complete reversal of the systemic consequences of folate deficiency is easily achieved. While correction of the neurologic consequences is more difficult, favorable neurologic outcomes are possible when adequate treatment is initiated promptly after birth 97. Severe neurologic and cognitive complications, including seizures, are invariably due to delay in diagnosis and implementation of treatment to achieve adequate cerebrospinal fluid (CSF) folate levels.

Dosing is aimed at achieving CSF folate trough concentrations as close as possible to the normal range for the age of the affected individual (infants and children have higher CSF folate levels than adults) 90. Because hereditary folate malabsorption is rare, controlled studies to establish optimal treatment have not been possible. The oral dose of folinic acid (leucovorin or 5-formyltetrahydrofolate [5-formylTHF]) required to overcome the loss of the proton-coupled folate transporter (PCFT)-mediated intestinal folate absorption appears to vary among individuals 90. The dose required to obviate the neurologic consequences is much higher than that needed to correct the systemic folate deficiency. The dose should be guided by its effect on trough CSF folate concentrations. The endpoint is CSF folate concentrations as close as possible to the normal range for the affected individual’s age.

The reported oral dose of folinic acid (leucovorin or 5-formyltetrahydrofolate [5-formylTHF]) associated with a “good” outcome is approximately 150-200 mg daily 98. Much higher doses have been used as well 90. A reasonable starting oral dose of folinic acid (leucovorin or 5-formyltetrahydrofolate [5-formylTHF]) in an infant could be 50 mg or 10-15 mg/kg given daily as a single dose, with subsequent dosing dependent on correction of the systemic signs of the disorder and achievement of an adequate cerebrospinal fluid (CSF) folate level 90.

Note: Normal CSF folate is ~100 nmol/L for infants to age two years, decreasing to ~75 nmol/L by age five years and to ~65 nmol/L by age 19 years 99.

The intramuscular dose required to achieve adequate serum and cerebrospinal fluid (CSF) folate levels is much lower than the oral dose. With intramuscular injections of approximately 1 mg/day of folinic acid (leucovorin or 5-formyltetrahydrofolate [5-formylTHF]), the anemia, immunologic, and gastrointestinal manifestations will fully resolve. However, the endpoint for treatment is based on achieving an adequate cerebrospinal fluid (CSF) folate level to mitigate the neurologic consequences of hereditary folate malabsorption, which will require much higher folate doses. It would appear that the maximum achievable cerebrospinal fluid (CSF) folate levels are in the range of 40-50 nmol/L 100, 97, 101, 102. For instance, with a folinic acid (leucovorin or 5-formyltetrahydrofolate [5-formylTHF]) oral dose of 28 mg/kg/day versus a parenteral dose of 2 mg/kg/day (both racemic), the CSF folate levels were 16 nmol/L and 43 nmol/L, while the blood levels were ~800 nmol/L and ~2,000 nmol/L, respectively 101, 95.

Cerebral Folate Deficiency (CFD)

Cerebral Folate Deficiency (CFD) also called cerebral folate transport deficiency or neurodegeneration due to cerebral folate transport deficiency is a neurological syndrome in which development is usually normal in the first year of life, but at approximately 2 years of age, affected children start to lose mental and motor skills (psychomotor regression) 103, 104, 105, 106, 107, 108. Mutations in the FOLR1 gene cause cerebral folate transport deficiency. The FOLR1 gene provides instructions for making a protein called folate receptor alpha (FRA). The folate receptor alpha (FRA) protein is found within the cell membrane where it attaches (binds) to folate, allowing folate (vitamin B9) to be brought into the cell. Folate receptor alpha (FRA) is produced in largest amounts in the brain, specifically in an area of the brain called the choroid plexus. This region releases cerebrospinal fluid (CSF), which surrounds and protects the brain and spinal cord. Folate receptor alpha (FRA) is thought to play a major role in bringing folate from the bloodstream into brain cells. It transports folate across the choroid plexus and into the CSF (cerebrospinal fluid), ultimately reaching the brain. In the brain, folate is needed for making myelin and chemical messengers called neurotransmitters. Both of these substances play essential roles in transmitting signals in the nervous system. Additionally, folate is involved in the production and repair of DNA, regulation of gene activity (expression), and protein production.

FOLR1 gene mutations result in a lack of folate receptor alpha (FRA) protein or malfunctioning of folate receptor alpha (FRA) protein. As a result, folate from the bloodstream cannot be transported into the cerebrospinal fluid (CSF). Without folate, many processes in the brain are impaired, leading to the neurological problems typical of cerebral folate transport deficiency.

Even though there may be normal folate levels in the serum and red blood cells, evaluation of the cerebrospinal fluid (CSF) shows a decreased level of 5-methyltetrahydrofolate (5MTHF) 107. The brain may appear normal on an MRI, but in some affected children, a loss of white matter in the brain (leukodystrophy) may be seen 107. Frontotemporal atrophy and impairment of the protective layer that surrounds nerve fibers in the brain and spinal cord (subcortical demyelination) can be seen as early as 18 months 107.

The symptoms of cerebral folate deficiency may begin as early as four to six months of age with irritability and sleep problems (insomnia) 107. Delays in development may be noted including slow head growth, low muscle tone (hypotonia), ataxia, loss of voluntary movement (dyskinesia), constant contracted muscles (spasticity), speech complications, and epilepsy 107. Additional signs may involve visual disturbances, hearing loss and autistic features 107.

Some early symptoms of cerebral folate deficiency are intellectual disability, speech difficulties, and development of recurrent seizures in a third of affected children. Motor issues such as tremors and lack of muscle control or coordination of voluntary movements (ataxia) can become severe.

Neurological abnormalities, along with visual and hearing impairments, have been described in children with cerebral folate deficiency. Autism spectrum disorder (ASD) is present in some cases. Cerebral folate deficiency has also been described in adults presenting with neurological symptoms 109.

Cerebral folate deficiency condition can be treated with folinic acid (leucovorin calcium). Oral treatment with folinic acid (leucovorin) has been shown to improve symptoms and stabilize the level of 5-methyltetrahydrofolate (5MTHF) in the cerebrospinal fluid (CSF). Folinic acid can enter the brain and normalize the level of folate coenzymes and has been shown to normalize folate concentrations and improve various social interactions in cerebral folate deficiency condition, including mood, behavior, and verbal communication in children with autism spectrum disorder (ASD) 103, 110, 111.

The overall outcome of cerebral folate deficiency seems to depend on the age at which treatment is initiated, the earlier the treatment, the better outcome. Supplementation with folic acid is not recommended because it is associated with adverse effects such as producing epileptic seizures. No serious adverse effects have been recorded during leucovorin calcium treatment. A milk-free diet in combination with leucovorin has been reported to improve symptoms, especially when used in the early stages of the disease.

Dihydrofolate reductase (DHFR) deficiency

Dihydrofolate reductase (DHFR) is the NADPH-dependent enzyme that catalyzes the conversion of dihydrofolate (DHF) to tetrahydrofolate (THF), an essential step in the synthesis of precursors of DNA, including glycine and purines, and the conversion of deoxyuridine monophosphate (dUMP) to deoxythymidine monophosphate (dTMP) (see Figure 4 above) 112, 113. Dihydrofolate reductase (DHFR) is also required to convert folic acid (a synthetic folate [vitamin B9] not found in nature) to dihydrofolate (DHF). Dihydrofolate reductase (DHFR) deficiency is characterized by megaloblastic anemia and cerebral folate deficiency causing intractable seizures and mental deficits 114, 115, 116, 116, 117, 118. Although folinic acid treatment can alleviate the symptoms of DHFR deficiency, early diagnosis is essential to prevent irreversible brain damage and improve clinical outcomes 114, 115.

Autism spectrum disorder

Autism spectrum disorder (ASD) also known as autism, is a lifelong neurodevelopmental disorder that affects how you behave and interact with the world 119, 120, 121, 122. People with autism have difficulty with communication and interacting with other people as well as repetitive behaviors and limited interests. The classification and diagnosis of autism spectrum disorder (ASD) was changed in 2013 to include conditions previously known as autistic disorder, Asperger’s syndrome, and pervasive developmental disorder not otherwise specified 123. However, The term ‘Asperger’s syndrome’ is no longer used. Health professionals used to think of autism as a condition that ranged from high functioning to severe. People who were diagnosed with Asperger’s syndrome would now be diagnosed with autism spectrum disorder. “Spectrum” means that autism affects people differently. There is a wide range of symptoms and levels of support needed by people with autism. People with autism will all have different experiences and need different levels of support. Along with some challenges, a person with autism will also have a range of strengths. People with autism are “neurodivergent”. This means their brains work in ways that are different from what is ‘typical’ or common, but it is not necessarily a problem. The term neurodivergent also describes people with many other conditions, such as dyslexia and attention deficit hyperactivity disorder (ADHD).

The main symptoms related to autism fall into 2 broad areas:

  • difficulty with social interactions and communication
  • limited and repetitive behaviours and interests

The features of autism usually start in infancy, but they may not be noticeable until the age of 18 months or older. Sometimes autism is only noticed much later in life.

The causes of autism spectrum disorder are not clear, but genetic and environmental factors (including infections) and prenatal exposure to certain drugs, pollutants, and pesticides are believed to play a role 124, 125, 126, 127, 128, 129, 130, 131, 132, 133.

Emerging evidence suggests that folic acid supplementation before or around the time of conception might reduce the risk of autism spectrum disorder or reduce the potentially increased risk of autism spectrum disorder from prenatal exposure to certain drugs and toxic chemicals. The mechanism of these potential benefits is unknown, but it might be related to folic acid’s role in DNA methylation, which, in turn, can affect neurodevelopment 134, 135, 136.

Some, but not all, observational studies have shown associations between maternal use of folic acid and/or multivitamin supplements before and/or during pregnancy and lower risk of autism spectrum disorder in the women’s offspring. For example, the prospective Norwegian Mother and Child Cohort Study that included 85,176 children age 3.3 to 10.2 years found that children of mothers who took up to 400 mcg per day folic acid during all or part of the time from 4 weeks before to 8 weeks after the start of pregnancy were 39% less likely to have autistic disorder than those whose mothers did not take the supplements 137. The results showed no significant associations, however, between folic acid supplementation and Asperger’s syndrome or pervasive developmental disorder not otherwise specified. In a U.S. population-based, case-control study of 837 children, those born to mothers who consumed a mean of 600 mcg folic acid per day or more from supplements and fortified breakfast cereals during the first month of pregnancy had a 38% lower risk of autism spectrum disorder than those of mothers who consumed less than 600 mcg per day 138. This association was strongest for mothers and children with the genetic mutation involving 677C>T MTHFR gene (a common genetic variant where a change occurs in the MTHFR (methylenetetrahydrofolate reductase) gene, which produces an enzyme critical for processing folate and homocysteine). Similarly, a 2018 case-control cohort study of 45,300 Israeli children demonstrated a significantly decreased risk of autism spectrum disorder in children of mothers who took folic acid and/or multivitamin supplements before and/or during pregnancy 139. In contrast, a longitudinal, population-based cohort of 35,059 pregnant Danish women and their children found no association between periconceptional folic acid or multivitamin use and autism spectrum disorder 140.

Periconceptional use of folic acid might reduce the potentially increased risk of autism spectrum disorder in children exposed to certain drugs and neurotoxins in utero 131, 132, 133. An analysis of data from the Norwegian Mother and Child Cohort Study, which included 104,946 children, found that children exposed to antiepileptic drugs (known to reduce folate in the body) in utero were 5.9 to 7.9 times more likely to have autistic traits at age 18 and 36 months if their mothers did not take folic acid periconceptionally than if they did 131. In addition, the severity of autistic traits was inversely associated with both maternal plasma folate concentrations and folic acid doses. Similarly, in a U.S. study of 712 children, mothers exposed to any indoor pesticide during pregnancy who had folic acid intakes of 800 mcg or more per day during the first month of pregnancy were 1.7 times more likely to have a child with autism spectrum disorder than women with the same folic acid intakes who were not exposed to indoor pesticides 132. The risk of autism spectrum disorder was even higher (2.5 times) if the women were exposed to indoor pesticides and had daily folic acid intakes of less than 800 mcg, suggesting that folic acid might attenuate the potentially increased risk of autism spectrum disorder from pesticide exposure.

Overall, the evidence to date suggests a possible inverse association between mothers’ periconceptional folic acid intakes and risk of autism spectrum disorder in their offspring. However, most, if not all, of the currently available data are observational, and confounding weakens the ability to demonstrate causal inference. Additional research and validation in other studies are needed before firm conclusions can be drawn.

Cancer

Several epidemiological studies have suggested an inverse association between folate intakes and status and the risk of colorectal, lung, pancreatic, esophageal, stomach, cervical, ovarian, breast, bladder, and other cancers 10, 141, 142, 143. Research has not established the precise nature of folate’s effect on carcinogenesis, but scientists hypothesize that folate might influence cancer development through its role in one-carbon metabolism and subsequent effects on DNA replication and cell division 143, 144. Evidence also indicates that folate might play a dual role in cancer initiation and progression 145. That is, folate might suppress some types of cancer during the early stages of development, whereas high doses of folic acid taken after preneoplastic lesions have been established might promote cancer development and progression.

Results from clinical trials involving folic acid supplementation have been mixed. In addition, most trials have included other B-vitamins (frequently at doses well above RDA levels) and sometimes other nutrients, making it difficult to disentangle the effects, if any, of folic acid alone. For example, in a trial in France, 2,501 people with a history of cardiovascular disease received daily supplements of 560 mcg folic acid, 3 mg vitamin B6, and 20 mcg vitamin B12 for 5 years 146. The researchers found no association between B-vitamin supplementation and cancer outcomes. In a combined analysis of two trials in Norway (where foods are not fortified with folic acid), supplementation with 800 mcg/day folic acid plus 400 mcg/day vitamin B12 for a median of 39 months in 3,411 people with ischemic heart disease increased cancer incidence rates by 21% and cancer mortality rates by 38% compared with no supplementation 147. Findings from these Norwegian trials have raised concerns about folic acid supplementation’s potential to raise cancer risk.

The most thorough research has focused on folate’s effect on the development of colorectal cancer and its precursor, adenoma 143, 148. Several epidemiological studies have found inverse associations between high dietary folate intakes and the risk of colorectal adenoma and cancer 149, 150, 151, 152. For example, in the NIH-AARP Diet and Health Study, a cohort study of more than 525,000 people age 50 to 71 years in the United States, individuals with total folate intakes of 900 mcg/day or higher had a 30% lower risk of colorectal cancer than those with intakes lower than 200 mcg/day 150. Other studies, however, have found no significant associations between dietary folate intakes 153, 154 or circulating folate concentrations 155, 156 and colorectal cancer risk.

Several clinical trials have examined whether supplemental folic acid (sometimes in combination with other B-vitamins) reduces the risk of colorectal adenoma in individuals with or without a history of adenoma. In the Women’s Antioxidant and Folic Acid Cardiovascular Study, which included 1,470 older women at high risk of cardiovascular disease, daily supplementation with 2,500 mcg folic acid, 50 mg vitamin B6, and 1,000 mcg vitamin B12 did not affect rates of colorectal adenoma during 7.3 years of intervention and about 2 years of postintervention follow-up 157. A pooled analysis of three large clinical trials (one in Canada, one in both the United States and Canada, and one in both the United Kingdom and Denmark) found that folic acid supplementation for up to 3.5 years neither increased nor decreased adenoma recurrence rates in people with a history of adenoma 158. However, in one of the studies included in the analysis, folic acid supplementation (1,000 mcg/day) significantly increased the risks of having three or more adenomas and of noncolorectal cancers, although it had no effect on colorectal cancer risk 159.

Folic acid supplementation also had no effect on the risk of all cancer types combined in the pooled analysis of three clinical trials cited above 158. Similarly, a meta-analysis of 13 randomized trials showed no statistically significant effects of folic acid supplementation (median daily dose of 2,000 mcg) over an average treatment period of 5.2 years on overall cancer incidence or the incidence of colorectal, lung, breast, prostate, or other cancers 160.

Some research has found associations between folic acid supplementation and increased cancer risk. In a randomized clinical trial investigating osteoporotic fracture incidence in 2,919 participants age 65 years or older with elevated homocysteine levels, those who received 400 mcg folic acid plus 500 mcg vitamin B12 and 600 International Units (IU) vitamin D3 for 2 years reported a significantly higher cancer incidence, especially of colorectal and other gastrointestinal cancers, than those who received only 600 IU vitamin D3 161. In addition, a 2018 prospective study found that folic acid intake from fortified foods and supplements was positively associated with a risk of cancer recurrence among 619 patients with non–muscle-invasive bladder cancer, whereas natural folate intakes showed no significant association 162. Higher plasma folate concentrations have also been associated with an increased risk of breast cancer in women with a BRCA1 or BRCA2 mutation 163. A secondary analysis of the study by Cole and colleagues 159 found that folic acid supplementation significantly increased the risk of prostate cancer 164. Subsequent research has shown an association between increased cancer cell proliferation and higher serum folate concentrations in men with prostate cancer 165. A meta-analysis of six randomized controlled trials that included a total of 25,738 men found that the risk of prostate cancer was 24% higher in men receiving folic acid supplements than those taking a placebo 166.

The mixed findings from clinical trials, combined with evidence from laboratory and animal studies indicating that high folate status promotes tumor progression, suggest that folate might play dual roles in cancer risk, depending on the dosage and timing of the exposure. Modest doses of folic acid taken before preneoplastic lesions are established might suppress cancer development in healthy tissues, whereas high doses taken after the establishment of preneoplastic lesions might promote cancer development and progression 148, 167, 168, 20, 169. This hypothesis is supported by a 2011 prospective study that found an inverse association between folate intake and risk of colorectal cancer only during early preadenoma stages 170.

A 2015 expert panel convened by the National Toxicology Program and the National Institutes of Health Office of Dietary Supplements concluded that folic acid supplements do not reduce cancer risk in people with adequate baseline folate status 171. The panel also determined that the consistent findings from human studies that supplemental folic acid has an adverse effect on cancer growth justify additional research on the effects of folic acid supplementation on cancer risk 171. Several important questions about these effects remain, including the dose and timing of folic acid supplementation that might exert tumor-promoting effects and whether this effect is specific to synthetic folic acid or other forms of folate 145.

Overall, the evidence to date indicates that adequate dietary folate intake might reduce the risk of some forms of cancer. However, the effects of supplemental folic acid on cancer risk are unclear, especially among individuals with a history of colorectal adenomas or other forms of cancer. More research is needed to fully understand how dietary folate and supplemental folic acid affect cancer risk and whether their effects differ by timing of exposure.

Cardiovascular disease and stroke

An elevated homocysteine level has been associated with an increased risk of cardiovascular disease 10. Folate and other B vitamins are involved in homocysteine metabolism, and researchers have hypothesized that these micronutrients reduce cardiovascular disease risk by lowering homocysteine levels 172, 10.

Folic acid and vitamin B12 (cobalamin) supplements lower homocysteine levels. However, these supplements do not actually decrease the risk of cardiovascular disease, although they appear to provide protection from stroke 173, 174, 172, 175, 176, 177, 178, 179, 180, 181. For example, in 5,442 U.S. women age 42 or older who were at high risk of cardiovascular disease, daily supplements containing 2,500 mcg folic acid, 1 mg vitamin B12, and 50 mg vitamin B6 for 7.3 years did not reduce the risk of major cardiovascular events 176. In a substudy of 300 participants, the supplementation also had no significant effects on biomarkers of vascular inflammation 182, but it did lower homocysteine levels by a mean of 18.5% 176. Another clinical trial included 5,522 patients age 55 years or older with vascular disease or diabetes from various countries (including the United States and Canada) that had a folic acid fortification program and some that did not 175. Patients received 2,500 mcg folic acid plus 50 mg vitamin B6 and 1 mg vitamin B12 or placebo for an average of 5 years. Compared with placebo, treatment with B vitamins significantly decreased homocysteine levels but did not reduce the risk of death from cardiovascular causes or myocardial infarction. Supplementation did, however, significantly reduce the risk of stroke by 25%.

In a large trial in regions of China without folic acid fortification among 20,702 adults with hypertension but no history of stroke or myocardial infarction, supplementation with 800 mcg folic acid plus 10 mg enalapril (used to treat high blood pressure) for a median of 4.5 years significantly reduced the risk of stroke by 21% compared with enalapril alone 179. The effect was more pronounced in participants with the lowest baseline levels of plasma folate. An analysis of 10,789 participants from this trial found that folic acid supplementation significantly reduced the risk of stroke by 73% among those who had a low platelet count and an elevated homocysteine level (increasing their risk of stroke) but had no significant effect on participants with a high platelet count and low homocysteine level 183. These findings suggest that folic acid supplementation might primarily benefit those with insufficient folate levels, which are less common in countries, such as the United States, with folic acid fortification 184.

The authors of a 2012 meta-analysis of 19 randomized controlled trials that included 47,921 participants concluded that B-vitamin supplementation has no effect on the risk of cardiovascular disease, myocardial infarction, coronary heart disease, or cardiovascular death, although it does reduce the risk of stroke by 12% 172. Likewise, the authors of the third update of a Cochrane Review of the effects of homocysteine-lowering interventions on cardiovascular events concluded that folic acid supplementation alone or in combination with vitamin B6 and vitamin B12 does not affect the risk of myocardial infarction or death from any cause, but it does reduce the risk of stroke 185. Three other meta-analyses have also found that folic acid is effective for preventing stroke, especially in populations exposed to no or partial folic acid fortification 181, 186, 187.

Overall, the available evidence suggests that supplementation with folic acid alone or in combination with other B-vitamins reduces the risk of stroke, especially in populations with low folate status, but does not affect other cardiovascular endpoints.

Dementia, cognitive function, and Alzheimer’s disease

Most observational studies conducted to date have shown positive associations between elevated homocysteine levels and the incidence of both Alzheimer’s disease and dementia 188, 189, 190, 191, 167, 15, 192. Scientists hypothesize that elevated homocysteine levels might have a negative effect on the brain via numerous mechanisms, including cerebrovascular ischemia leading to neuronal cell death, activation of tau kinases leading to tangle deposition, and inhibition of methylation reactions 191. Some, but not all, observational studies have also found correlations between low serum folate concentrations and both poor cognitive function and higher risk of dementia and Alzheimer’s disease 191167189188193.

Despite this evidence, most clinical trial research has not shown that folic acid supplementation affects cognitive function or the development of dementia or Alzheimer’s disease, even though supplementation lowers homocysteine levels. In one randomized, double-blind, placebo-controlled trial in the Netherlands, 195 people age 70 years or older with no or moderate cognitive impairment received 400 mcg folic acid plus 1 mg vitamin B12; 1 mg vitamin B12; or placebo for 24 weeks 194. Treatment with folic acid plus vitamin B12 reduced homocysteine concentrations by 36% but did not improve cognitive function. In another clinical trial in older adults (mean age 74.1 years) with elevated homocysteine levels, supplementation with 400 mcg folic acid plus 500 mcg vitamin B12 and 600 IU vitamin D3 for 2 years lowered homocysteine levels but did not affect cognitive performance compared with 600 IU vitamin D3 alone 195.

As part of the Women’s Antioxidant and Folic Acid Cardiovascular Study, 2,009 U.S. women age 65 years or older at high risk of cardiovascular disease were randomly assigned to receive daily supplements containing 2,500 mcg folic acid plus 1 mg vitamin B12 and 50 mg vitamin B6 or placebo 196. After an average of 1.2 years, B-vitamin supplementation did not affect mean cognitive change from baseline compared with placebo. However, in a subset of women with a low baseline dietary intake of B vitamins, supplementation significantly slowed the rate of cognitive decline. In a trial that included 340 individuals in the United States with mild-to-moderate Alzheimer’s disease, daily supplements of 5,000 mcg folic acid plus 1 mg vitamin B12 and 25 mg vitamin B6 for 18 months did not slow cognitive decline compared with placebo 197.

A secondary analysis of a study in Australia (which did not have mandatory folic acid fortification at the time of the study) found that daily supplementation with 400 mcg folic acid plus 100 mcg vitamin B12 for 2 years improved some measures of cognitive function, particularly memory, in 900 adults age 60 to 74 years who had depressive symptoms 198. Another meta-analysis included 11 randomized controlled trials in more than 20,000 older adults (mean age 60–82 years) that administered 400 to 2,500 mcg folic acid plus 20–1,000 mcg vitamin B12 in 10 trials and 3–50 mg vitamin B6 in 8 trials for 0.3 to 7.1 years. The supplementation significantly lowered homocysteine levels but did not affect cognitive aging, global cognitive function, or specific cognitive domains (including memory, speed, and executive function) 199.

Several large reviews have evaluated the effect of B vitamins on cognitive function. Most of the authors concluded that supplementation with folic acid alone or in combination with vitamins B12 or B6 does not appear to improve cognitive function in individuals with or without cognitive impairment 200, 201, 202, 203. Some noted, however, that when researchers took baseline homocysteine and B-vitamin status into account, B-vitamin supplementation slowed cognitive decline in individuals at high risk of cognitive decline 191, 192. For example, one trial in the Netherlands administered either 800 mcg folic acid or placebo daily for 3 years to 818 participants age 50–70 years with elevated homocysteine levels (13 micromol/L or higher) and normal vitamin B12 levels 204. Folic acid supplementation reduced homocysteine concentrations by 26% and significantly improved global cognitive function, memory, and information processing speed compared with placebo, but it did not affect sensorimotor speed, complex speed, or word fluency.

Additional clinical trials are needed to better understand the effects of folic acid supplementation on cognitive function and cognitive decline.

Depression

Low folate status has been linked to depression and poor response to antidepressants in some, but not all, studies. The possible mechanisms are unclear but might be related to folate’s role in methylation reactions in the brain, neurotransmitter synthesis, and homocysteine metabolism 205, 206. However, secondary factors linked to depression, such as unhealthy eating patterns and alcohol use disorder, might also contribute to the observed association between low folate status and depression 207.

In an ethnically diverse population study of 2,948 people age 15 to 39 years in the United States, serum and erythrocyte folate concentrations were significantly lower in individuals with major depression than in those who had never been depressed 207. An analysis of 2005–2006 NHANES data found that higher serum concentrations of folate were associated with a lower prevalence of depression in 2,791 adults age 20 or older 205. The association was statistically significant in females, but not in males. However, another analysis showed no associations between folate intakes from both food and dietary supplements and depression among 1,368 healthy Canadians age 67–84 years 206. Results from a study of 52 men and women with major depressive disorder showed that only 1 of 14 participants with low serum folate levels responded to antidepressant treatment compared with 17 of 38 with normal folate levels 208.

A few studies have examined whether folate status affects the risk of depression during pregnancy or after childbirth. A systematic review of these studies had mixed results 209. One study included in the review among 709 women in Singapore found that compared with women with higher plasma folate concentrations (mean 40.4 nmol/L [17.8 ng/mL]) at 26–28 weeks’ gestation, those with lower plasma folate concentrations (mean 27.3 nmol/L [12.0 ng/mL]) had a significantly higher risk of depression during pregnancy but not after giving birth 210. Another study of 2,856 women in the United Kingdom found no significant associations between red blood cell folate levels or folate intakes from food and dietary supplements before or during pregnancy and postpartum depressive symptoms 211. More recently, a cohort study of 1,592 Chinese women found a lower prevalence of postpartum depression in women who took folic acid supplements for more than 6 months during pregnancy than in those who took them for less time 212.

Studies have had mixed results on whether folic acid supplementation might be a helpful adjuvant treatment for depression when used with traditional antidepressant medications. In a clinical trial in the United Kingdom, 127 patients with major depression were randomly assigned to receive either 500 mcg folic acid or placebo in addition to 20 mg of fluoxetine daily for 10 weeks 213. Although the effects in men were not statistically significant, women who received fluoxetine plus folic acid had a significantly greater improvement in depressive symptoms than those who received fluoxetine plus placebo. Another clinical trial in the United Kingdom randomized 475 adults with moderate to severe depression who were taking antidepressant medications to either 5,000 mcg folic acid or placebo daily for 12 weeks in addition to their antidepressants 214. Measures of depression did not improve in participants taking folic acid compared with those taking placebo. The authors of a systematic review and meta-analysis of four trials of folic acid (<5,000 mcg/day in two trials; 5,000 mcg/day in two trials) in combination with fluoxetine or other antidepressants in patients with major depressive disorder concluded that less than 5,000 mcg/day folic acid might be beneficial as an adjunct to serotonin reuptake inhibitor (SSRI) therapy 215. The authors noted, however, that this conclusion was based on low-quality evidence. Another meta-analysis of four clinical trials found that 500–10,000 mcg folic acid per day for 6–12 weeks as an adjunctive treatment did not significantly affect measures of depression compared with placebo 216.

Other studies have examined the effects of 5-MTHF supplementation as an adjuvant treatment to antidepressants, and results suggest that it might have more promise than folic acid 215, 216, 217, 218. In a clinical trial in 148 adults with major depressive disorder, supplementation with 7,500 mcg/day 5-MTHF for 30 days followed by 15,000 mcg/day for another 30 days, both in conjunction with SSRI treatment, did not improve measures of depression compared with SSRI treatment plus placebo 219. However, in a subsequent trial with the same study design in 75 adults, supplementation with 15,000 mcg/day 5-MTHF plus SSRI treatment for the full 60 days did significantly improve depression compared with SSRI treatment plus placebo 219.

The authors of a systematic review and meta-analysis of three trials of 5-MTHF (<15,000 mcg/day in one trial, and 15,000 mcg/day in two trials) in combination with fluoxetine or other antidepressants, concluded that 15,000 mcg/day 5-MTHF might be an effective adjunct to SSRI therapy in patients with major depressive disorder, although they noted that this conclusion was based on low-quality evidence 215. In addition, evidence-based guidelines from the British Association for Psychopharmacology 217 and the Canadian Network for Mood and Anxiety Treatments 218 state that 5-MTHF might be effective as an adjunct to SSRI treatment for depressive disorders.

Additional research is needed to fully understand the association between folate status and depression. Although limited evidence suggests that supplementation with certain forms and doses of folate might be a helpful adjuvant treatment for depressive disorders, more research is needed to confirm these findings. In addition, many of the doses of folate used in studies of depression exceed the Tolerable Upper Intake Level (maximum daily intake unlikely to cause adverse health effects) and should be taken only under medical supervision.

Folinic acid dosage

Folinic acid (leucovorin) is available in 5, 10, 15, and 25 mg tablets. Injectable folinic acid (leucovorin) formulations are 10 and 20 mg/mL solutions. Folinic acid is usually compounded with calcium, so it should not be administered intravenously at rates >160 mg/min 2. Folinic acid is also not to be administered intrathecally 220.

Due to the wide range of folinic acid indications and administration guidelines, specific protocols are available that clinicians should follow; the dosing regimens below are general and are not a replacement for checking the facility protocols. In general, folinic acid is compounded with leucovorin calcium (especially for FDA-approved indications) and can be administered intramuscularly, intravenously, or orally 2. The timing, dosage, and route of folinic acid administration depend on the desired outcome for the particular indication 65.

Adult Dose

For high-dose methotrexate leucovorin rescue, the folinic acid (leucovorin) dosage is typically 15 mg orally, IM, or IV every 6 hours for 10 doses, starting 24 hours after initiating methotrexate. The maximum oral dose is 25 mg; IM or IV doses may be higher.

  • In patients with delayed methotrexate administration, the dosage is 15 mg orally, IM, or IV every 6 hours. Continue until the methotrexate level <0.05 micromolar 67.

For leucovorin rescue for methotrexate overdose, the dosage is typically 10 mg/m² orally, IM, or IV every 6 hours, starting ASAP after overdose or within 24 hours if delayed methotrexate elimination.

For folate antagonist overdose:

  • For pemetrexed: 50 mg/m2/dose IV every 6 hours for 8 days. Start with 100 mg/m2/dose IV for the first dose.
  • For trimethoprim or pyrimethamine: Give 5 to 15 mg orally, IM, or IV daily until the restoration of hematopoiesis.

For colorectal cancer combination therapy with 5-FU, clinicians should follow specific administration schedules as individual protocols vary. In general, when used for combination therapy, folinic acid is administered as an IV bolus or short IV infusions (minutes to hours) 66. Dosing may require adjustments based on toxicity.

  • With 5-FU 370 mg/m²/day: Leucovorin 200 mg/m²/day administered IV on days 1 to 5 of a 28-day cycle. Give 2 cycles, then continue 28-day cycles or extend to 35-day cycles.
  • With 5-FU 425 mg/m²/day: Leucovorin 20 mg/m2/day administered IV on days 1 to 5 of a 28-day cycle. Give 2 cycles, then continue 28-day cycles or extend to 35-day cycles.

Folate-deficiency associated megaloblastic anemia:

  • Dosing is <1 mg IV daily; maximum dose 1 mg daily.

Children Dose

As with adult dosing recommendations, individual protocols require close adherence, as guidelines differ depending on the desired outcome and particular indication. However, there are similar generalities in both patient populations. For both methotrexate toxicity and combination therapy with 5-FU, the previously mentioned adult administration recommendations are similar for the pediatric patient population 66.

For high-dose methotrexate leucovorin rescue:

  • In patients with standard methotrexate administration, the dosage is 10 mg/m²/dose, orally, IM, or IV every 6 hours for 10 doses, starting 24 hours after initiating methotrexate. The maximum oral dose is 25 mg; IM or IV doses may be higher.
  • In patients with delayed methotrexate administration, the dosage is mg/m²/dose orally, IM, or IV every 6 hours. Continue until the methotrexate level <0.05 μM 67.

Leucovorin rescue for methotrexate overdose:

  • Dosage is 10 mg/m²/dose orally, IM, or IV every 6 hours, starting ASAP after overdose or within 24 hours if delayed methotrexate elimination.

For folate antagonist overdose:

  • For trimethoprim or pyrimethamine: Give 5 to 15 mg orally, IM, or IV daily until the restoration of hematopoiesis.

Folate-deficiency-associated megaloblastic anemia:

  • Dosing is <1 mg IV daily; maximum dose 1 mg daily.

Special Patient Populations

  • Liver impairment: Dosing in hepatic impairment is undefined.
  • Kidney impairment: Dosing for patients with renal impairment, including those on dialysis, is undefined.
  • Pregnant women: Folinic acid/leucovorin may be used during pregnancy; human data shows no known risk of fetal harm.
  • Breastfeeding considerations: Folinic acid can be used by breastfeeding women. No human data is available, but based on the drug’s properties, infant harm is not expected.
  • Older patients: No data specific to leucovorin prohibits or limits its use in older patients.

Folinic acid side effects

Along with its needed effects, folinic acid may cause some unwanted effects. Although not all of these side effects may occur, if they do occur they may need medical attention.

Check with your doctor immediately if any of the following side effects occur.

Rare side effects:

  • Skin rash, hives, or itching
  • Wheezing

Check with your doctor as soon as possible if any of the following side effects occur:

Rare side effects reported with use in treatment of cancer:

  • Convulsions (seizures)

Other side effects not listed may also occur in some patients. If you notice any other effects, check with your healthcare professional.

Drug interactions

When nonsteroidal anti-inflammatory drugs (NSAIDs), such as aspirin or ibuprofen, are taken in very large therapeutic dosages (i.e., to treat severe arthritis), they may interfere with folate metabolism. In contrast, routine use of nonsteroidal anti-inflammatory drugs (NSAIDs) has not been found to adversely affect folate status. The anticonvulsant (antiepileptic), phenytoin, has been shown to inhibit the intestinal absorption of folate, and several studies have associated decreased folate status with long-term use of the anticonvulsants, phenytoin, phenobarbital, and primidone 221. However, few studies controlled for differences in dietary folate intake between anticonvulsant users and nonusers. Also, taking folic acid at the same time as the cholesterol-lowering agents, cholestyramine and colestipol, may decrease the absorption of folic acid 222. Methotrexate is a folic acid antagonist used to treat a number of diseases, including cancer, rheumatoid arthritis, and psoriasis. Some of the side effects of methotrexate are similar to those of severe folate deficiency, and supplementation with folic or folinic acid is used to reduce antifolate toxicity. Other antifolate molecules currently used in cancer therapy include aminopterin, pemetrexed, pralatrexate, and raltitrexed 6, 223. Furthermore, a number of other medications have been shown to have antifolate activity, including trimethoprim (an antibiotic), pyrimethamine (an antimalarial), triamterene (a blood pressure medication), and sulfasalazine (a treatment for ulcerative colitis). Early studies of oral contraceptives (birth control pills) containing high doses of estrogen indicated adverse effects on folate status; however, this finding has not been supported in more recent studies that used low-dose oral contraceptives and controlled for dietary folate 224.

Health Risks from Excessive Folinic Acid

No adverse effects have been associated with the consumption of excess folate from food. Concerns regarding folate safety are limited to synthetic folic acid intake e.g. folinic acid. Some health experts have been concerned that high intakes of folate supplements might mask vitamin B12 deficiency until its neurological consequences become irreversible 225, 226, 227, 228, 229, 230, 172Vitamin B12 deficiency is a condition caused by low levels of vitamin B12 (cobalamin) in the body, leading to symptoms like megaloblastic anemia, fatigue, weakness, pale skin, nerve problems (numbness, tingling), mood changes, and in severe cases, neurological damage. Vitamin B12 deficiency may affect a significant number of people, especially older adults. One symptom of vitamin B12 deficiency is megaloblastic anemia, which is indistinguishable from that associated with folate deficiency. There is concern that large doses of folic acid given to an individual with an undiagnosed vitamin B12 deficiency could correct the megaloblastic anemia without correcting the underlying vitamin B12 deficiency, leaving the individual at risk of developing irreversible neurologic damage. Such cases of neurologic progression in vitamin B12 deficiency have been mostly seen in case studies at folic acid doses of 5,000 mcg (5 mg) and above. In order to be very sure of preventing irreversible neurologic damage in vitamin B12-deficient individuals, the Food and Nutrition Board of the US National Academy of Medicine advises that all adults limit their intake of folic acid (supplements and fortification) to 1,000 mcg (1 mg) daily (Table 1). The Food and Nutrition Board also noted that vitamin B12 deficiency is very rare in women in their childbearing years, making the consumption of folic acid at or above 1,000 mcg/day unlikely to cause problems 9. Furthermore, there are limited saefty data on the effects of large folic acid doses 231.

However, there are concerns that high folic acid intakes might accelerate the progression of preneoplastic lesions, increasing the risk of colorectal and possibly other cancers in certain individuals 10, 12, 148, 168, 20. In addition, intakes of 1,000 mcg (1 mg) per day or more of folic acid from supplements during the periconception period have been associated with lower scores on several tests of cognitive development in children at age 4–5 years than in children of mothers who took 400 mcg to 999 mcg 232.

Intakes of folic acid that exceed the body’s ability to reduce it to tetrahydrofolate (THF, the biologically active form of folic acid) lead to unmetabolized folic acid in the body, which has been linked to reduced numbers and activity of natural killer cells, suggesting that it could affect the immune system 233, 234. A small study conducted in postmenopausal women also raised concerns about the effect of exposure to unmetabolized folic acid on immune function 233. Recent observational studies have found no link between circulating maternal unmetabolized folate during pregnancy and allergic disease in infancy 235 or autistic traits or language impairment in early childhood 236. In a small, randomized, open-label trial in 38 women of reproductive age receiving 30 weeks of daily multivitamin supplements, daily supplementation with either 1.1 mg or 5 mg of folic acid resulted in the transient appearance of unmetabolized folic acid in blood over the first 12 weeks of supplementation 237. However, unmetabolized folic acid concentrations returned to baseline levels at the end of the study, suggesting that adaptive mechanisms eventually converted folic acid to reduced forms of folate.

Some scientists have hypothesized that unmetabolized folic acid might be related to cognitive impairment among older adults (≥60 years) 226, 238. These potential negative health consequences are not well understood and warrant further research 10, 141. The use of supplemental Levomefolic acid (5-methyltetrahydrofolate [5-MTHF] the most active form of folate) may provide an alternative to prevent any potential negative effects of unconverted folic acid in older adults.

Studies have found unmetabolized folic acid in blood from children, adolescents, and adults 10, 234, 239, 240; breastmilk 241; and cord blood from newborns 242, 243. Limited research suggests that single doses of 300 mcg or 400 mcg folic acid (a common amount in folic acid-containing supplements or servings of fortified foods, such as breakfast cereals) result in detectable serum levels of unmetabolized folic acid, whereas doses of 100 mcg or 200 mcg do not 244, 245. In addition, a dose-frequency interaction appears to occur in which smaller amounts of folic acid consumed more frequently produce higher unmetabolized folic acid concentrations than the same total dose consumed in larger, less frequent amounts 246.

Based on the metabolic interactions between folate and vitamin B12, the Food and Nutrition Board established a Tolerable Upper Intake Level (maximum daily intake unlikely to cause adverse health effects) for the synthetic forms of folate available in dietary supplements and fortified foods (Table 1) 9. The Food and Nutrition Board did not establish a Tolerable Upper Intake Level (maximum daily intake unlikely to cause adverse health effects) for folate from food because high intakes of folate from food sources have not been reported to cause adverse effects 9. Unlike the Recommended Dietary Allowance (RDA), the Tolerable Upper Intake Level (maximum daily intake unlikely to cause adverse health effects) are in mcg, not mcg DFE. For folic acid, 1,000 mcg is equivalent to 1,667 mcg DFE because 0.6 mcg folic acid = 1 mcg DFE 7. The ULs do not apply to individuals taking high doses of supplemental folate under medical supervision 7.

Table 1. Tolerable Upper Intake Levels (ULs) for Folate from Supplements or Fortified Foods

AgeMaleFemalePregnancyLactation
Birth to 6 monthsNot possible to establish*Not possible to establish*
7–12 monthsNot possible to establish*Not possible to establish*
1–3 years300 mcg300 mcg
4–8 years400 mcg400 mcg
9–13 years600 mcg600 mcg
14–18 years800 mcg800 mcg800 mcg800 mcg
19+ years1,000 mcg1,000 mcg1,000 mcg1,000 mcg
[Source 7 ]
  1. Budak M.Ş., Oğlak S.C., Akgöl S., Can B., Arkan K., Erkmen A.D., Halisçelik M.A., Budak A., Tunç Ş., Bolluk G., et al. Folic acid versus folinic acid during methotrexate treatment for low-risk gestational trophoblastic neoplasia. Ginekol. Pol. 2025;96:200–205. doi: 10.5603/gpl.101142[]
  2. Gristan YD, Patel P, Moosavi L. Folinic Acid. [Updated 2024 Feb 28]. In: StatPearls [Internet]. Treasure Island (FL): StatPearls Publishing; 2025 Jan-. Available from: https://www.ncbi.nlm.nih.gov/books/NBK545232[][][][][][]
  3. Askari BS, Krajinovic M. Dihydrofolate reductase gene variations in susceptibility to disease and treatment outcomes. Curr Genomics. 2010 Dec;11(8):578-83. doi: 10.2174/138920210793360925[]
  4. Office on Women’s Health, U.S. Department of Health and Human Services – Folic acid – https://www.womenshealth.gov/a-z-topics/folic-acid[][]
  5. Scaglione F, Panzavolta G. Folate, folic acid and 5-methyltetrahydrofolate are not the same thing. Xenobiotica. 2014 May;44(5):480-8. doi: 10.3109/00498254.2013.845705[][][][][][][][][]
  6. Shulpekova Y, Nechaev V, Kardasheva S, Sedova A, Kurbatova A, Bueverova E, Kopylov A, Malsagova K, Dlamini JC, Ivashkin V. The Concept of Folic Acid in Health and Disease. Molecules. 2021 Jun 18;26(12):3731. doi: 10.3390/molecules26123731[][]
  7. Folate. https://ods.od.nih.gov/factsheets/Folate-HealthProfessional[][][][][][][][][][][][][]
  8. Duthie SJ, Narayanan S, Brand GM, Pirie L, Grant G. Impact of folate deficiency on DNA stability. J Nutr. 2002 Aug;132(8 Suppl):2444S-2449S. doi: 10.1093/jn/132.8.2444S[][]
  9. Food and Nutrition Board, Institute of Medicine. Folate. Dietary Reference Intakes: Thiamin, Riboflavin, Niacin, Vitamin B-6, Folate, Vitamin B-12, Pantothenic Acid, Biotin, and Choline. Washington D.C.: National Academy Press; 1998:196-305. https://nap.nationalacademies.org/read/6015/chapter/10[][][][][][][][][][]
  10. Bailey LB, Caudill MA. Folate. In: Erdman JW, Macdonald IA, Zeisel SH, eds. Present Knowledge in Nutrition. 10th ed. Washington, DC: Wiley-Blackwell; 2012:321-42.[][][][][][][][][][][][][][][][][]
  11. Institute of Medicine. Food and Nutrition Board. Dietary Reference Intakes: Thiamin, Riboflavin, Niacin, Vitamin B6, Folate, Vitamin B12, Pantothenic Acid, Biotin, and Choline. Washington, DC: National Academy Press; 1998.[][]
  12. Stover PJ. Folic acid. In: Ross AC, Caballero B, Cousins RJ, Tucker KL, Ziegler TR, eds. Modern Nutrition in Health and Disease. 11th ed. Baltimore, MD: Lippincott Williams & Wilkins; 2012:358-68.[][][]
  13. Bailey LB, Gregory JFr (2006). Folate. Present Knowledge in Nutrition. B. Bowman and R. Russell. Washington, DC, International Life Sciences Institute. I: 278-301.[]
  14. Carmel R. Folic acid. In: Shils M, Shike M, Ross A, Caballero B, Cousins RJ, eds. Modern Nutrition in Health and Disease. 11th ed. Baltimore, MD: Lippincott Williams & Wilkins; 2005:470-81.[][][][][][]
  15. Ho RC, Cheung MW, Fu E, Win HH, Zaw MH, Ng A, Mak A. Is high homocysteine level a risk factor for cognitive decline in elderly? A systematic review, meta-analysis, and meta-regression. Am J Geriatr Psychiatry. 2011 Jul;19(7):607-17. doi: 10.1097/JGP.0b013e3181f17eed[][]
  16. Williams J, Mai CT, Mulinare J, Isenburg J, Flood TJ, Ethen M, Frohnert B, Kirby RS; Centers for Disease Control and Prevention. Updated estimates of neural tube defects prevented by mandatory folic Acid fortification – United States, 1995-2011. MMWR Morb Mortal Wkly Rep. 2015 Jan 16;64(1):1-5. https://pmc.ncbi.nlm.nih.gov/articles/PMC4584791[][][]
  17. Scholl TO, Johnson WG. Folic acid: influence on the outcome of pregnancy. Am J Clin Nutr. 2000 May;71(5 Suppl):1295S-303S. doi: 10.1093/ajcn/71.5.1295s[][][]
  18. Lamers Y. Folate recommendations for pregnancy, lactation, and infancy. Ann Nutr Metab. 2011;59(1):32-7. doi: 10.1159/000332073[]
  19. Wilson RD; GENETICS COMMITTEE; MOTHERISK. RETIRED: Pre-conceptional vitamin/folic acid supplementation 2007: the use of folic acid in combination with a multivitamin supplement for the prevention of neural tube defects and other congenital anomalies. J Obstet Gynaecol Can. 2007 Dec;29(12):1003-1013. English, French. doi: 10.1016/S1701-2163(16)32685-8. Erratum in: J Obstet Gynaecol Can. 2008 Mar;30(3):193. Goh, Ingrid [corrected to Goh, Y Ingrid]. https://doi.org/10.1016/S1701-2163(16)32685-8[]
  20. Ulrich CM, Potter JD. Folate supplementation: too much of a good thing? Cancer Epidemiol Biomarkers Prev. 2006 Feb;15(2):189-93. doi: 10.1158/1055-9965.EPI-152CO[][][]
  21. Pitkin RM. Folate and neural tube defects. Am J Clin Nutr. 2007 Jan;85(1):285S-288S. doi: 10.1093/ajcn/85.1.285S[]
  22. Scott JM. Evidence of folic acid and folate in the prevention of neural tube defects. Bibl Nutr Dieta. 2001;(55):192-5. doi: 10.1159/000059465[]
  23. Molloy AM, Kirke PN, Brody LC, Scott JM, Mills JL. Effects of folate and vitamin B12 deficiencies during pregnancy on fetal, infant, and child development. Food Nutr Bull. 2008 Jun;29(2 Suppl):S101-11; discussion S112-5. doi: 10.1177/15648265080292S114[][]
  24. Williams LJ, Rasmussen SA, Flores A, Kirby RS, Edmonds LD. Decline in the prevalence of spina bifida and anencephaly by race/ethnicity: 1995-2002. Pediatrics. 2005 Sep;116(3):580-6. doi: 10.1542/peds.2005-0592[]
  25. Rader JI, Schneeman BO. Prevalence of neural tube defects, folate status, and folate fortification of enriched cereal-grain products in the United States. Pediatrics. 2006 Apr;117(4):1394-9. doi: 10.1542/peds.2005-2745[]
  26. Dary O. Nutritional interpretation of folic acid interventions. Nutr Rev. 2009 Apr;67(4):235-44. doi: 10.1111/j.1753-4887.2009.00193.x[]
  27. Shane B. Folate-responsive birth defects: of mice and women. Am J Clin Nutr. 2012 Jan;95(1):1-2. doi: 10.3945/ajcn.111.029595[]
  28. Pietrzik K, Bailey L, Shane B. Folic acid and L-5-methyltetrahydrofolate: comparison of clinical pharmacokinetics and pharmacodynamics. Clin Pharmacokinet. 2010 Aug;49(8):535-48. doi: 10.2165/11532990-000000000-00000[][]
  29. Molloy AM, Pangilinan F, Brody LC. Genetic Risk Factors for Folate-Responsive Neural Tube Defects. Annu Rev Nutr. 2017 Aug 21;37:269-291. doi: 10.1146/annurev-nutr-071714-034235[]
  30. Centers for Disease Control and Prevention (CDC). CDC Grand Rounds: additional opportunities to prevent neural tube defects with folic acid fortification. MMWR Morb Mortal Wkly Rep. 2010 Aug 13;59(31):980-4. https://www.cdc.gov/mmwr/preview/mmwrhtml/mm5931a2.htm[]
  31. Nutrition During Pregnancy. https://www.acog.org/womens-health/faqs/nutrition-during-pregnancy[][][]
  32. Prepregnancy Counseling. https://www.acog.org/clinical/clinical-guidance/committee-opinion/articles/2019/01/prepregnancy-counseling[]
  33. Practice Bulletin No. 187: Neural Tube Defects. Obstet Gynecol. 2017 Dec;130(6):e279-e290. doi: 10.1097/AOG.0000000000002412[][]
  34. Frayne DJ, Verbiest S, Chelmow D, Clarke H, Dunlop A, Hosmer J, Menard MK, Moos MK, Ramos D, Stuebe A, Zephyrin L. Health Care System Measures to Advance Preconception Wellness: Consensus Recommendations of the Clinical Workgroup of the National Preconception Health and Health Care Initiative. Obstet Gynecol. 2016 May;127(5):863-872. doi: 10.1097/AOG.0000000000001379[][]
  35. Buck Louis GM, Sapra KJ, Schisterman EF, Lynch CD, Maisog JM, Grantz KL, Sundaram R. Lifestyle and pregnancy loss in a contemporary cohort of women recruited before conception: The LIFE Study. Fertil Steril. 2016 Jul;106(1):180-188. doi: 10.1016/j.fertnstert.2016.03.009[][]
  36. Khan KM, Jialal I. Folic Acid Deficiency. [Updated 2022 Jun 27]. In: StatPearls [Internet]. Treasure Island (FL): StatPearls Publishing; 2023 Jan-. Available from: https://www.ncbi.nlm.nih.gov/books/NBK535377[][][][][]
  37. Green R, Datta Mitra A. Megaloblastic Anemias: Nutritional and Other Causes. Med Clin North Am. 2017 Mar;101(2):297-317. doi: 10.1016/j.mcna.2016.09.013[]
  38. Carmel R (2005). Folic Acid. Modern Nutrition in Health and Disease. M. Shils, M. Shike, A. Ross, B. Caballero and R. Cousins. Baltimore, MD, Lippincott Williams & Wilkins: 470-481.[]
  39. Institute of Medicine. Food and Nutrition Board (1998). Dietary Reference Intakes: Thiamin, Riboflavin, Niacin, Vitamin B6, Folate, Vitamin B12, Pantothenic Acid, Biotin, and Choline. Washington, DC, National Academy Press. https://www.nap.edu/catalog/6015/dietary-reference-intakes-for-thiamin-riboflavin-niacin-vitamin-b6-folate-vitamin-b12-pantothenic-acid-biotin-and-choline[][][][][]
  40. Colapinto CK, O’Connor DL, Dubois L, Tremblay MS (2012). Folic acid supplement use is the most significant predictor of folate concentrations in Canadian women of childbearing age. Appl Physiol Nutr Metab 37(2): 284-292. http://www.nrcresearchpress.com/doi/full/10.1139/h11-161[]
  41. Green R (2011). Indicators for assessing folate and vitamin B-12 status and for monitoring the efficacy of intervention strategies. Am J Clin Nutr 94(2): 666S-672S. https://www.ncbi.nlm.nih.gov/pmc/articles/PMC3142735/[]
  42. Yeung LF, Cogswell ME, Carriquiry AL, Bailey LB, Pfeiffer CM, Berry RJ. Contributions of enriched cereal-grain products, ready-to-eat cereals, and supplements to folic acid and vitamin B-12 usual intake and folate and vitamin B-12 status in US children: National Health and Nutrition Examination Survey (NHANES), 2003-2006. Am J Clin Nutr. 2011 Jan;93(1):172-85. doi: 10.3945/ajcn.2010.30127[]
  43. Lis K. Hypersensitivity to Folic Acid and/or Folinic Acid-A Review of Clinical Cases, Potential Mechanism, Possible Cross-Allergies and Current Diagnostic Options. Curr Issues Mol Biol. 2025 Aug 14;47(8):654. doi: 10.3390/cimb4708065 []
  44. Folic Acid. https://pubchem.ncbi.nlm.nih.gov/compound/Folic-Acid[]
  45. Lis K. Hypersensitivity to Folic Acid and/or Folinic Acid-A Review of Clinical Cases, Potential Mechanism, Possible Cross-Allergies and Current Diagnostic Options. Curr Issues Mol Biol. 2025 Aug 14;47(8):654. doi: 10.3390/cimb47080654[][]
  46. Frosst P, Blom HJ, Milos R, Goyette P, Sheppard CA, Matthews RG, Boers GJ, den Heijer M, Kluijtmans LA, van den Heuvel LP, et al. A candidate genetic risk factor for vascular disease: a common mutation in methylenetetrahydrofolate reductase. Nat Genet. 1995 May;10(1):111-3. doi: 10.1038/ng0595-111[]
  47. Green R, Miller JW. Folate deficiency beyond megaloblastic anemia: hyperhomocysteinemia and other manifestations of dysfunctional folate status. Semin Hematol. 1999 Jan;36(1):47-64.[]
  48. Hoffbrand AV, Jackson BF. Correction of the DNA synthesis defect in vitamin B12 deficiency by tetrahydrofolate: evidence in favour of the methyl-folate trap hypothesis as the cause of megaloblastic anaemia in vitamin B12 deficiency. Br J Haematol. 1993 Apr;83(4):643-7. doi: 10.1111/j.1365-2141.1993.tb04704.x[]
  49. Quinlivan EP, Davis SR, Shelnutt KP, Henderson GN, Ghandour H, Shane B, Selhub J, Bailey LB, Stacpoole PW, Gregory JF 3rd. Methylenetetrahydrofolate reductase 677C->T polymorphism and folate status affect one-carbon incorporation into human DNA deoxynucleosides. J Nutr. 2005 Mar;135(3):389-96. doi: 10.1093/jn/135.3.389[]
  50. Crider KS, Yang TP, Berry RJ, Bailey LB. Folate and DNA methylation: a review of molecular mechanisms and the evidence for folate’s role. Adv Nutr. 2012 Jan;3(1):21-38. doi: 10.3945/an.111.000992[]
  51. Choi SW, Mason JB. Folate and carcinogenesis: an integrated scheme. J Nutr. 2000 Feb;130(2):129-32. doi: 10.1093/jn/130.2.129[]
  52. Bailey LB, Gregory JF 3rd. Folate metabolism and requirements. J Nutr. 1999 Apr;129(4):779-82. doi: 10.1093/jn/129.4.779[]
  53. Folate. https://lpi.oregonstate.edu/mic/vitamins/folate[]
  54. Shuvalov O, Petukhov A, Daks A, Fedorova O, Vasileva E, Barlev NA. One-carbon metabolism and nucleotide biosynthesis as attractive targets for anticancer therapy. Oncotarget. 2017 Apr 4;8(14):23955-23977. doi: 10.18632/oncotarget.15053[]
  55. Gerhard GT, Duell PB. Homocysteine and atherosclerosis. Curr Opin Lipidol. 1999 Oct;10(5):417-28. doi: 10.1097/00041433-199910000-00006[]
  56. Azzini E, Ruggeri S, Polito A. Homocysteine: Its Possible Emerging Role in At-Risk Population Groups. Int J Mol Sci. 2020 Feb 20;21(4):1421. doi: 10.3390/ijms21041421[]
  57. Wang S, Wang L, Zhou Z, Deng Q, Li L, Zhang M, Liu L, Li Y. Leucovorin Enhances the Anti-cancer Effect of Bortezomib in Colorectal Cancer Cells. Sci Rep. 2017 Apr 6;7(1):682. doi: 10.1038/s41598-017-00839-9[][][]
  58. Yetley EA, Pfeiffer CM, Phinney KW, Fazili Z, Lacher DA, Bailey RL, et al. (2011). Biomarkers of folate status in NHANES: a roundtable summary. Am J Clin Nutr 94(1): 303S-312S. https://www.ncbi.nlm.nih.gov/pmc/articles/PMC3127517/[]
  59. Greenberg JA, Bell SJ, Guan Y, Yu YH. Folic Acid supplementation and pregnancy: more than just neural tube defect prevention. Rev Obstet Gynecol. 2011 Summer;4(2):52-9. https://www.ncbi.nlm.nih.gov/pmc/articles/PMC3218540[]
  60. Henderson AM, Aleliunas RE, Loh SP, Khor GL, Harvey-Leeson S, Glier MB, Kitts DD, Green TJ, Devlin AM. l-5-Methyltetrahydrofolate Supplementation Increases Blood Folate Concentrations to a Greater Extent than Folic Acid Supplementation in Malaysian Women. J Nutr. 2018 Jun 1;148(6):885-890. doi: 10.1093/jn/nxy057[]
  61. Green TJ, Liu Y, Dadgar S, Li W, Böhni R, Kitts DD. 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. 2013 Jun;143(6):867-71. doi: 10.3945/jn.113.174268[]
  62. Lamers Y, Prinz-Langenohl R, Brämswig S, Pietrzik K. 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. 2006 Jul;84(1):156-61. doi: 10.1093/ajcn/84.1.156[]
  63. Desmoulin SK, Hou Z, Gangjee A, Matherly LH. The human proton-coupled folate transporter: Biology and therapeutic applications to cancer. Cancer Biol Ther. 2012 Dec;13(14):1355-73. doi: 10.4161/cbt.22020[]
  64. Solanky N, Requena Jimenez A, D’Souza SW, Sibley CP, Glazier JD. Expression of folate transporters in human placenta and implications for homocysteine metabolism. Placenta. 2010 Feb;31(2):134-43. doi: 10.1016/j.placenta.2009.11.017[]
  65. Greiner PO, Zittoun J, Marquet J, Cheron JM. Pharmacokinetics of (-)-folinic acid after oral and intravenous administration of the racemate. Br J Clin Pharmacol. 1989 Sep;28(3):289-95. doi: 10.1111/j.1365-2125.1989.tb05429.x[][][][][]
  66. Priest DG, Schmitz JC, Bunni MA, Stuart RK. Pharmacokinetics of leucovorin metabolites in human plasma as a function of dose administered orally and intravenously. J Natl Cancer Inst. 1991 Dec 18;83(24):1806-12. doi: 10.1093/jnci/83.24.1806[][][][]
  67. Howard SC, McCormick J, Pui CH, Buddington RK, Harvey RD. Preventing and Managing Toxicities of High-Dose Methotrexate. Oncologist. 2016 Dec;21(12):1471-1482. doi: 10.1634/theoncologist.2015-0164[][][][][][][]
  68. de Gramont A, Figer A, Seymour M, Homerin M, Hmissi A, Cassidy J, Boni C, Cortes-Funes H, Cervantes A, Freyer G, Papamichael D, Le Bail N, Louvet C, Hendler D, de Braud F, Wilson C, Morvan F, Bonetti A. Leucovorin and fluorouracil with or without oxaliplatin as first-line treatment in advanced colorectal cancer. J Clin Oncol. 2000 Aug;18(16):2938-47. doi: 10.1200/JCO.2000.18.16.2938[][]
  69. Gustavsson B, Carlsson G, Machover D, Petrelli N, Roth A, Schmoll HJ, Tveit KM, Gibson F. A review of the evolution of systemic chemotherapy in the management of colorectal cancer. Clin Colorectal Cancer. 2015 Mar;14(1):1-10. doi: 10.1016/j.clcc.2014.11.002[]
  70. Zaniboni A. Adjuvant chemotherapy in colorectal cancer with high-dose leucovorin and fluorouracil: impact on disease-free survival and overall survival. J Clin Oncol. 1997 Jun;15(6):2432-41. doi: 10.1200/JCO.1997.15.6.2432[]
  71. Antifolate. https://www.sciencedirect.com/topics/medicine-and-dentistry/antifolate[]
  72. Shiroky JB, Neville C, Esdaile JM, Choquette D, Zummer M, Hazeltine M, Bykerk V, Kanji M, St-Pierre A, Robidoux L, et al. Low-dose methotrexate with leucovorin (folinic acid) in the management of rheumatoid arthritis. Results of a multicenter randomized, double-blind, placebo-controlled trial. Arthritis Rheum. 1993 Jun;36(6):795-803. doi: 10.1002/art.1780360609[]
  73. Lambie DG, Johnson RH. Drugs and folate metabolism. Drugs. 1985 Aug;30(2):145-55. doi: 10.2165/00003495-198530020-00003[]
  74. Sanz-Altamira PM, Ferrante K, Jenkins RL, Lewis WD, Huberman MS, Stuart KE. A phase II trial of 5-fluorouracil, leucovorin, and carboplatin in patients with unresectable biliary tree carcinoma. Cancer. 1998 Jun 15;82(12):2321-5.[]
  75. Hsu CH, Yeh KH, Chen LT, Liu JM, Jan CM, Lin JT, Chen YC, Cheng AL. Weekly 24-hour infusion of high-dose 5-fluorouracil and leucovorin in the treatment of advanced gastric cancers. An effective and low-toxic regimen for patients with poor general condition. Oncology. 1997 Jul-Aug;54(4):275-80. doi: 10.1159/000227702[]
  76. Vokes EE, Schilsky RL, Weichselbaum RR, Kozloff MF, Panje WR. Induction chemotherapy with cisplatin, fluorouracil, and high-dose leucovorin for locally advanced head and neck cancer: a clinical and pharmacologic analysis. J Clin Oncol. 1990 Feb;8(2):241-7. doi: 10.1200/JCO.1990.8.2.241[]
  77. Neoptolemos JP, Dunn JA, Stocken DD, Almond J, Link K, Beger H, Bassi C, Falconi M, Pederzoli P, Dervenis C, Fernandez-Cruz L, Lacaine F, Pap A, Spooner D, Kerr DJ, Friess H, Büchler MW; European Study Group for Pancreatic Cancer. Adjuvant chemoradiotherapy and chemotherapy in resectable pancreatic cancer: a randomised controlled trial. Lancet. 2001 Nov 10;358(9293):1576-85. doi: 10.1016/s0140-6736(01)06651-x[]
  78. Klimo P, Connors JM. MACOP-B chemotherapy for the treatment of diffuse large-cell lymphoma. Ann Intern Med. 1985 May;102(5):596-602. doi: 10.7326/0003-4819-102-5-596[]
  79. Tomblyn M, Chiller T, Einsele H, Gress R, Sepkowitz K, Storek J, Wingard JR, Young JA, Boeckh MJ; Center for International Blood and Marrow Research; National Marrow Donor program; European Blood and MarrowTransplant Group; American Society of Blood and Marrow Transplantation; Canadian Blood and Marrow Transplant Group; Infectious Diseases Society of America; Society for Healthcare Epidemiology of America; Association of Medical Microbiology and Infectious Disease Canada; Centers for Disease Control and Prevention. Guidelines for preventing infectious complications among hematopoietic cell transplantation recipients: a global perspective. Biol Blood Marrow Transplant. 2009 Oct;15(10):1143-238. doi: 10.1016/j.bbmt.2009.06.019. Erratum in: Biol Blood Marrow Transplant. 2010 Feb;16(2):294. Boeckh, Michael A [corrected to Boeckh, Michael J].[]
  80. Masur H, Brooks JT, Benson CA, Holmes KK, Pau AK, Kaplan JE; National Institutes of Health; Centers for Disease Control and Prevention; HIV Medicine Association of the Infectious Diseases Society of America. Prevention and treatment of opportunistic infections in HIV-infected adults and adolescents: Updated Guidelines from the Centers for Disease Control and Prevention, National Institutes of Health, and HIV Medicine Association of the Infectious Diseases Society of America. Clin Infect Dis. 2014 May;58(9):1308-11. doi: 10.1093/cid/ciu094[]
  81. Touam M, Zingraff J, Jungers P, Chadefaux-Vekemans B, Drüeke T, Massy ZA. Effective correction of hyperhomocysteinemia in hemodialysis patients by intravenous folinic acid and pyridoxine therapy. Kidney Int. 1999 Dec;56(6):2292-6. doi: 10.1046/j.1523-1755.1999.00792.x[]
  82. Amos RJ, Amess JA, Nancekievill DG, Rees GM. Prevention of nitrous oxide-induced megaloblastic changes in bone marrow using folinic acid. Br J Anaesth. 1984 Feb;56(2):103-7. doi: 10.1093/bja/56.2.103[]
  83. Finer LB, Zolna MR. Unintended pregnancy in the United States: incidence and disparities, 2006. Contraception. 2011 Nov;84(5):478-85. doi: 10.1016/j.contraception.2011.07.013[]
  84. Czeizel AE, Puhó EH, Langmar Z, Acs N, Bánhidy F. Possible association of folic acid supplementation during pregnancy with reduction of preterm birth: a population-based study. Eur J Obstet Gynecol Reprod Biol. 2010 Feb;148(2):135-40. doi: 10.1016/j.ejogrb.2009.10.016[]
  85. Wilson RD; GENETICS COMMITTEE; MOTHERISK. RETIRED: Pre-conceptional vitamin/folic acid supplementation 2007: the use of folic acid in combination with a multivitamin supplement for the prevention of neural tube defects and other congenital anomalies. J Obstet Gynaecol Can. 2007 Dec;29(12):1003-1013. English, French. doi: 10.1016/S1701-2163(16)32685-8. Erratum in: J Obstet Gynaecol Can. 2008 Mar;30(3):193. Goh, Ingrid [corrected to Goh, Y Ingrid].[][]
  86. Liu S, Joseph KS, Luo W, León JA, Lisonkova S, Van den Hof M, Evans J, Lim K, Little J, Sauve R, Kramer MS; Canadian Perinatal Surveillance System (Public Health Agency of Canada). Effect of Folic Acid Food Fortification in Canada on Congenital Heart Disease Subtypes. Circulation. 2016 Aug 30;134(9):647-55. doi: 10.1161/CIRCULATIONAHA.116.022126[]
  87. Botto LD, Mulinare J, Erickson JD. Occurrence of congenital heart defects in relation to maternal mulitivitamin use. Am J Epidemiol. 2000 May 1;151(9):878-84. doi: 10.1093/oxfordjournals.aje.a010291[]
  88. Shaw GM, O’Malley CD, Wasserman CR, Tolarova MM, Lammer EJ. Maternal periconceptional use of multivitamins and reduced risk for conotruncal heart defects and limb deficiencies among offspring. Am J Med Genet. 1995 Dec 4;59(4):536-45. doi: 10.1002/ajmg.1320590428[]
  89. Watkins D, Rosenblatt DS. Update and new concepts in vitamin responsive disorders of folate transport and metabolism. J Inherit Metab Dis. 2012 Jul;35(4):665-70. doi: 10.1007/s10545-011-9418-1[]
  90. Goldman ID. Hereditary Folate Malabsorption. 2008 Jun 17 [Updated 2024 Feb 15]. In: Adam MP, Feldman J, Mirzaa GM, et al., editors. GeneReviews® [Internet]. Seattle (WA): University of Washington, Seattle; 1993-2025. Available from: https://www.ncbi.nlm.nih.gov/books/NBK1673[][][][][][][][][]
  91. SLC46A1 gene. https://medlineplus.gov/genetics/gene/slc46a1[]
  92. Zhao R, Min SH, Qiu A, Sakaris A, Goldberg GL, Sandoval C, Malatack JJ, Rosenblatt DS, Goldman ID. The spectrum of mutations in the PCFT gene, coding for an intestinal folate transporter, that are the basis for hereditary folate malabsorption. Blood. 2007 Aug 15;110(4):1147-52. doi: 10.1182/blood-2007-02-077099[][]
  93. Borzutzky A, Crompton B, Bergmann AK, Giliani S, Baxi S, Martin M, Neufeld EJ, Notarangelo LD. Reversible severe combined immunodeficiency phenotype secondary to a mutation of the proton-coupled folate transporter. Clin Immunol. 2009 Dec;133(3):287-94. doi: 10.1016/j.clim.2009.08.006[]
  94. Sofer Y, Harel L, Sharkia M, Amir J, Schoenfeld T, Straussberg R. Neurological manifestations of folate transport defect: case report and review of the literature. J Child Neurol. 2007 Jun;22(6):783-6. doi: 10.1177/0883073807304004[]
  95. Lubout CMA, Goorden SMI, van den Hurk K, Jaeger B, Jager NGL, van Koningsbruggen S, Chegary M, van Karnebeek CDM. Successful Treatment of Hereditary Folate Malabsorption With Intramuscular Folinic Acid. Pediatr Neurol. 2020 Jan;102:62-66. doi: 10.1016/j.pediatrneurol.2019.06.009[][]
  96. FOLATE MALABSORPTION, HEREDITARY. https://www.omim.org/entry/229050[]
  97. Zhao R, Aluri S, Goldman ID. The proton-coupled folate transporter (PCFT-SLC46A1) and the syndrome of systemic and cerebral folate deficiency of infancy: Hereditary folate malabsorption. Mol Aspects Med. 2017 Feb;53:57-72. doi: 10.1016/j.mam.2016.09.002[][]
  98. Geller J, Kronn D, Jayabose S, Sandoval C. Hereditary folate malabsorption: family report and review of the literature. Medicine (Baltimore). 2002 Jan;81(1):51-68. doi: 10.1097/00005792-200201000-00004[]
  99. Verbeek MM, Blom AM, Wevers RA, Lagerwerf AJ, van de Geer J, Willemsen MA. Technical and biochemical factors affecting cerebrospinal fluid 5-MTHF, biopterin and neopterin concentrations. Mol Genet Metab. 2008 Nov;95(3):127-32. doi: 10.1016/j.ymgme.2008.07.004[]
  100. Torres A, Newton SA, Crompton B, Borzutzky A, Neufeld EJ, Notarangelo L, Berry GT. CSF 5-Methyltetrahydrofolate Serial Monitoring to Guide Treatment of Congenital Folate Malabsorption Due to Proton-Coupled Folate Transporter (PCFT) Deficiency. JIMD Rep. 2015;24:91-6. doi: 10.1007/8904_2015_445[]
  101. Aluri S, Zhao R, Lubout C, Goorden SMI, Fiser A, Goldman ID. Hereditary folate malabsorption due to a mutation in the external gate of the proton-coupled folate transporter SLC46A1. Blood Adv. 2018 Jan 5;2(1):61-68. doi: 10.1182/bloodadvances.2017012690[][]
  102. Manea E, Gissen P, Pope S, Heales SJ, Batzios S. Role of Intramuscular Levofolinate Administration in the Treatment of Hereditary Folate Malabsorption: Report of Three Cases. JIMD Rep. 2018;39:7-12. doi: 10.1007/8904_2017_39[]
  103. Frye RE, Sequeira JM, Quadros EV, James SJ, Rossignol DA. Cerebral folate receptor autoantibodies in autism spectrum disorder. Mol Psychiatry. 2013 Mar;18(3):369-81. doi: 10.1038/mp.2011.175[][]
  104. Grapp M, Just IA, Linnankivi T, Wolf P, Lücke T, Häusler M, Gärtner J, Steinfeld R. Molecular characterization of folate receptor 1 mutations delineates cerebral folate transport deficiency. Brain. 2012 Jul;135(Pt 7):2022-31. doi: 10.1093/brain/aws122[]
  105. Kanmaz S, Simsek E, Yilmaz S, Durmaz A, Serin HM, Gokben S. Cerebral folate transporter deficiency: a potentially treatable neurometabolic disorder. Acta Neurol Belg. 2023 Feb;123(1):121-127. doi: 10.1007/s13760-021-01700-7[]
  106. Ramaekers VT, Quadros EV. Cerebral Folate Deficiency Syndrome: Early Diagnosis, Intervention and Treatment Strategies. Nutrients. 2022 Jul 28;14(15):3096. doi: 10.3390/nu14153096[]
  107. Cerebral Folate Deficiency. https://rarediseases.org/rare-diseases/cerebral-folate-deficiency[][][][][][][]
  108. Cerebral folate transport deficiency. https://medlineplus.gov/genetics/condition/cerebral-folate-transport-deficiency[]
  109. Masingue M, Benoist JF, Roze E, Moussa F, Sedel F, Lubetzki C, Nadjar Y. Cerebral folate deficiency in adults: A heterogeneous potentially treatable condition. J Neurol Sci. 2019 Jan 15;396:112-118. doi: 10.1016/j.jns.2018.11.014[]
  110. Ramaekers VT, Blau N, Sequeira JM, Nassogne MC, Quadros EV. Folate receptor autoimmunity and cerebral folate deficiency in low-functioning autism with neurological deficits. Neuropediatrics. 2007 Dec;38(6):276-81. doi: 10.1055/s-2008-1065354[]
  111. Ramaekers VT, Häusler M, Opladen T, Heimann G, Blau N. Psychomotor retardation, spastic paraplegia, cerebellar ataxia and dyskinesia associated with low 5-methyltetrahydrofolate in cerebrospinal fluid: a novel neurometabolic condition responding to folinic acid substitution. Neuropediatrics. 2002 Dec;33(6):301-8. doi: 10.1055/s-2002-37082[]
  112. Trimble J.J., Murthy S.C., Bakker A., Grassmann R., Desrosiers R.C. A gene for dihydrofolate reductase in a herpesvirus. Science. 1988;239:1145–1147. doi: 10.1126/science.2830673[]
  113. Dihydrofolate Reductase. https://www.sciencedirect.com/topics/neuroscience/dihydrofolate-reductase[]
  114. Banka S, Blom HJ, Walter J, Aziz M, Urquhart J, Clouthier CM, Rice GI, de Brouwer AP, Hilton E, Vassallo G, Will A, Smith DE, Smulders YM, Wevers RA, Steinfeld R, Heales S, Crow YJ, Pelletier JN, Jones S, Newman WG. Identification and characterization of an inborn error of metabolism caused by dihydrofolate reductase deficiency. Am J Hum Genet. 2011 Feb 11;88(2):216-25. doi: 10.1016/j.ajhg.2011.01.004[][]
  115. Cario H, Smith DE, Blom H, Blau N, Bode H, Holzmann K, Pannicke U, Hopfner KP, Rump EM, Ayric Z, Kohne E, Debatin KM, Smulders Y, Schwarz K. Dihydrofolate reductase deficiency due to a homozygous DHFR mutation causes megaloblastic anemia and cerebral folate deficiency leading to severe neurologic disease. Am J Hum Genet. 2011 Feb 11;88(2):226-31. doi: 10.1016/j.ajhg.2011.01.007[][]
  116. Tauro G.P., Danks D.M., Rowe P.B., Van der Weyden M.B., Schwarz M.A., Collins V.L., Neal B.W. Dihydrofolate reductase deficiency causing megaloblastic anemia in two families. N. Engl. J. Med. 1976;294:466–470. doi: 10.1056/NEJM197602262940903[][]
  117. Erbe R.W. Inborn errors of folate metabolism (second of two parts) N. Engl. J. Med. 1975;293:807–812. doi: 10.1056/NEJM197510162931606[]
  118. Hoffbrand A.V., Tripp E., Jackson B.F., Luck W.E., Frater-Schroder M. Hereditary abnormal transcobalamin II previously diagnosed as congenital dihydrofolate reductase deficiency. N. Engl. J. Med. 1984;310:789–790. doi: 10.1056/nejm198403223101217[]
  119. Hodis B, Mughal S, Saadabadi A. Autism Spectrum Disorder. [Updated 2025 Jan 17]. In: StatPearls [Internet]. Treasure Island (FL): StatPearls Publishing; 2025 Jan-. Available from: https://www.ncbi.nlm.nih.gov/books/NBK525976[]
  120. Hosseini SA, Molla M. Asperger Syndrome. [Updated 2024 Feb 12]. In: StatPearls [Internet]. Treasure Island (FL): StatPearls Publishing; 2025 Jan-. Available from: https://www.ncbi.nlm.nih.gov/books/NBK557548[]
  121. Smith O, Jones SC. ‘Coming Out’ with Autism: Identity in People with an Asperger’s Diagnosis After DSM-5. J Autism Dev Disord. 2020 Feb;50(2):592-602. doi: 10.1007/s10803-019-04294-5[]
  122. Tietze FA, Hundertmark L, Roy M, Zerr M, Sinke C, Wiswede D, Walter M, Münte TF, Szycik GR. Auditory Deficits in Audiovisual Speech Perception in Adult Asperger’s Syndrome: fMRI Study. Front Psychol. 2019 Oct 10;10:2286. doi: 10.3389/fpsyg.2019.02286[]
  123. Autism Spectrum Disorder. https://www.nimh.nih.gov/health/topics/autism-spectrum-disorders-asd[]
  124. Love C, Sominsky L, O’Hely M, Berk M, Vuillermin P, Dawson SL. Prenatal environmental risk factors for autism spectrum disorder and their potential mechanisms. BMC Med. 2024 Sep 16;22(1):393. doi: 10.1186/s12916-024-03617-3[]
  125. Conklin L, Hviid A, Orenstein WA, Pollard AJ, Wharton M, Zuber P. Vaccine safety issues at the turn of the 21st century. BMJ Glob Health. 2021 May;6(Suppl 2):e004898. doi: 10.1136/bmjgh-2020-004898[]
  126. Gabis LV, Attia OL, Goldman M, Barak N, Tefera P, Shefer S, Shaham M, Lerman-Sagie T. The myth of vaccination and autism spectrum. Eur J Paediatr Neurol. 2022 Jan;36:151-158. doi: 10.1016/j.ejpn.2021.12.011[]
  127. Rylaarsdam L, Guemez-Gamboa A. Genetic Causes and Modifiers of Autism Spectrum Disorder. Front Cell Neurosci. 2019 Aug 20;13:385. doi: 10.3389/fncel.2019.00385[]
  128. Emberti Gialloreti L, Mazzone L, Benvenuto A, Fasano A, Alcon AG, Kraneveld A, Moavero R, Raz R, Riccio MP, Siracusano M, Zachor DA, Marini M, Curatolo P. Risk and Protective Environmental Factors Associated with Autism Spectrum Disorder: Evidence-Based Principles and Recommendations. J Clin Med. 2019 Feb 8;8(2):217. doi: 10.3390/jcm8020217[]
  129. Taylor LE, Swerdfeger AL, Eslick GD. Vaccines are not associated with autism: an evidence-based meta-analysis of case-control and cohort studies. Vaccine. 2014 Jun 17;32(29):3623-9. doi: 10.1016/j.vaccine.2014.04.085[]
  130. Berry RJ, Crider KS, Yeargin-Allsopp M. Periconceptional folic acid and risk of autism spectrum disorders. JAMA. 2013 Feb 13;309(6):611-3. doi: 10.1001/jama.2013.198[]
  131. Bjørk M, Riedel B, Spigset O, Veiby G, Kolstad E, Daltveit AK, Gilhus NE. Association of Folic Acid Supplementation During Pregnancy With the Risk of Autistic Traits in Children Exposed to Antiepileptic Drugs In Utero. JAMA Neurol. 2018 Feb 1;75(2):160-168. doi: 10.1001/jamaneurol.2017.3897. Erratum in: JAMA Neurol. 2018 Apr 1;75(4):518. doi: 10.1001/jamaneurol.2018.0085[][][]
  132. Schmidt RJ, Kogan V, Shelton JF, Delwiche L, Hansen RL, Ozonoff S, Ma CC, McCanlies EC, Bennett DH, Hertz-Picciotto I, Tancredi DJ, Volk HE. Combined Prenatal Pesticide Exposure and Folic Acid Intake in Relation to Autism Spectrum Disorder. Environ Health Perspect. 2017 Sep 8;125(9):097007. doi: 10.1289/EHP604[][][]
  133. Goodrich AJ, Volk HE, Tancredi DJ, McConnell R, Lurmann FW, Hansen RL, Schmidt RJ. Joint effects of prenatal air pollutant exposure and maternal folic acid supplementation on risk of autism spectrum disorder. Autism Res. 2018 Jan;11(1):69-80. doi: 10.1002/aur.1885[][]
  134. Roffman JL. Neuroprotective Effects of Prenatal Folic Acid Supplementation: Why Timing Matters. JAMA Psychiatry. 2018 Jul 1;75(7):747-748. doi: 10.1001/jamapsychiatry.2018.0378[]
  135. Caffrey A, Irwin RE, McNulty H, Strain JJ, Lees-Murdock DJ, McNulty BA, Ward M, Walsh CP, Pentieva K. Gene-specific DNA methylation in newborns in response to folic acid supplementation during the second and third trimesters of pregnancy: epigenetic analysis from a randomized controlled trial. Am J Clin Nutr. 2018 Apr 1;107(4):566-575. doi: 10.1093/ajcn/nqx069[]
  136. DeVilbiss EA, Gardner RM, Newschaffer CJ, Lee BK. Maternal folate status as a risk factor for autism spectrum disorders: a review of existing evidence. Br J Nutr. 2015 Sep 14;114(5):663-72. doi: 10.1017/S0007114515002470[]
  137. Surén P, Roth C, Bresnahan M, Haugen M, Hornig M, Hirtz D, Lie KK, Lipkin WI, Magnus P, Reichborn-Kjennerud T, Schjølberg S, Davey Smith G, Øyen AS, Susser E, Stoltenberg C. Association between maternal use of folic acid supplements and risk of autism spectrum disorders in children. JAMA. 2013 Feb 13;309(6):570-7. doi: 10.1001/jama.2012.155925[]
  138. Schmidt RJ, Tancredi DJ, Ozonoff S, Hansen RL, Hartiala J, Allayee H, Schmidt LC, Tassone F, Hertz-Picciotto I. 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. 2012 Jul;96(1):80-9. doi: 10.3945/ajcn.110.004416[]
  139. Levine SZ, Kodesh A, Viktorin A, Smith L, Uher R, Reichenberg A, Sandin S. 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. 2018 Feb 1;75(2):176-184. doi: 10.1001/jamapsychiatry.2017.4050[]
  140. Virk J, Liew Z, Olsen J, Nohr EA, Catov JM, Ritz B. Preconceptional and prenatal supplementary folic acid and multivitamin intake and autism spectrum disorders. Autism. 2016 Aug;20(6):710-8. doi: 10.1177/1362361315604076[]
  141. Bailey LB, Stover PJ, McNulty H, Fenech MF, Gregory JF 3rd, Mills JL, Pfeiffer CM, Fazili Z, Zhang M, Ueland PM, Molloy AM, Caudill MA, Shane B, Berry RJ, Bailey RL, Hausman DB, Raghavan R, Raiten DJ. Biomarkers of Nutrition for Development-Folate Review. J Nutr. 2015 Jul;145(7):1636S-1680S. doi: 10.3945/jn.114.206599[][]
  142. He H, Shui B. Folate intake and risk of bladder cancer: a meta-analysis of epidemiological studies. Int J Food Sci Nutr. 2014 May;65(3):286-92. doi: 10.3109/09637486.2013.866641[]
  143. Kim YI. Will mandatory folic acid fortification prevent or promote cancer? Am J Clin Nutr. 2004 Nov;80(5):1123-8. doi: 10.1093/ajcn/80.5.1123[][][]
  144. Kim YI. Folate and carcinogenesis: evidence, mechanisms, and implications. J Nutr Biochem. 1999 Feb;10(2):66-88. doi: 10.1016/s0955-2863(98)00074-6[]
  145. Kim YI. Folate and cancer: a tale of Dr. Jekyll and Mr. Hyde? Am J Clin Nutr. 2018 Feb 1;107(2):139-142. doi: 10.1093/ajcn/nqx076[][]
  146. Andreeva VA, Touvier M, Kesse-Guyot E, Julia C, Galan P, Hercberg S. B vitamin and/or ω-3 fatty acid supplementation and cancer: ancillary findings from the supplementation with folate, vitamins B6 and B12, and/or omega-3 fatty acids (SU.FOL.OM3) randomized trial. Arch Intern Med. 2012 Apr 9;172(7):540-7. doi: 10.1001/archinternmed.2011.1450[]
  147. Ebbing M, Bønaa KH, Nygård O, Arnesen E, Ueland PM, Nordrehaug JE, Rasmussen K, Njølstad I, Refsum H, Nilsen DW, Tverdal A, Meyer K, Vollset SE. Cancer incidence and mortality after treatment with folic acid and vitamin B12. JAMA. 2009 Nov 18;302(19):2119-26. doi: 10.1001/jama.2009.1622[]
  148. Mason JB. Unraveling the complex relationship between folate and cancer risk. Biofactors. 2011 Jul-Aug;37(4):253-60. doi: 10.1002/biof.174[][][]
  149. Giovannucci E, Stampfer MJ, Colditz GA, Rimm EB, Trichopoulos D, Rosner BA, Speizer FE, Willett WC. Folate, methionine, and alcohol intake and risk of colorectal adenoma. J Natl Cancer Inst. 1993 Jun 2;85(11):875-84. doi: 10.1093/jnci/85.11.875[]
  150. Gibson TM, Weinstein SJ, Pfeiffer RM, Hollenbeck AR, Subar AF, Schatzkin A, Mayne ST, Stolzenberg-Solomon R. Pre- and postfortification intake of folate and risk of colorectal cancer in a large prospective cohort study in the United States. Am J Clin Nutr. 2011 Oct;94(4):1053-62. doi: 10.3945/ajcn.110.002659[][]
  151. Kennedy DA, Stern SJ, Moretti M, Matok I, Sarkar M, Nickel C, Koren G. Folate intake and the risk of colorectal cancer: a systematic review and meta-analysis. Cancer Epidemiol. 2011 Feb;35(1):2-10. doi: 10.1016/j.canep.2010.11.004[]
  152. Sanjoaquin MA, Allen N, Couto E, Roddam AW, Key TJ. Folate intake and colorectal cancer risk: a meta-analytical approach. Int J Cancer. 2005 Feb 20;113(5):825-8. doi: 10.1002/ijc.20648[]
  153. Bassett JK, Severi G, Hodge AM, Baglietto L, Hopper JL, English DR, Giles GG. Dietary intake of B vitamins and methionine and colorectal cancer risk. Nutr Cancer. 2013;65(5):659-67. doi: 10.1080/01635581.2013.789114[]
  154. de Vogel S, Dindore V, van Engeland M, Goldbohm RA, van den Brandt PA, Weijenberg MP. Dietary folate, methionine, riboflavin, and vitamin B-6 and risk of sporadic colorectal cancer. J Nutr. 2008 Dec;138(12):2372-8. doi: 10.3945/jn.108.091157[]
  155. Neuhouser ML, Cheng TY, Beresford SA, Brown E, Song X, Miller JW, Zheng Y, Thomson CA, Shikany JM, Vitolins MZ, Rohan T, Green R, Ulrich CM. Red blood cell folate and plasma folate are not associated with risk of incident colorectal cancer in the Women’s Health Initiative observational study. Int J Cancer. 2015 Aug 15;137(4):930-9. doi: 10.1002/ijc.29453[]
  156. Chuang SC, Rota M, Gunter MJ, Zeleniuch-Jacquotte A, Eussen SJ, Vollset SE, Ueland PM, Norat T, Ziegler RG, Vineis P. Quantifying the dose-response relationship between circulating folate concentrations and colorectal cancer in cohort studies: a meta-analysis based on a flexible meta-regression model. Am J Epidemiol. 2013 Oct 1;178(7):1028-37. doi: 10.1093/aje/kwt083[]
  157. Song Y, Manson JE, Lee IM, Cook NR, Paul L, Selhub J, Giovannucci E, Zhang SM. Effect of combined folic acid, vitamin B(6), and vitamin B(12) on colorectal adenoma. J Natl Cancer Inst. 2012 Oct 17;104(20):1562-75. doi: 10.1093/jnci/djs370[]
  158. Figueiredo JC, Mott LA, Giovannucci E, Wu K, Cole B, Grainge MJ, Logan RF, Baron JA. Folic acid and prevention of colorectal adenomas: a combined analysis of randomized clinical trials. Int J Cancer. 2011 Jul 1;129(1):192-203. doi: 10.1002/ijc.25872[][]
  159. Cole BF, Baron JA, Sandler RS, Haile RW, et al. Polyp Prevention Study Group. Folic acid for the prevention of colorectal adenomas: a randomized clinical trial. JAMA. 2007 Jun 6;297(21):2351-9. doi: 10.1001/jama.297.21.2351[][]
  160. Vollset SE, Clarke R, Lewington S, et al. B-Vitamin Treatment Trialists’ Collaboration. Effects of folic acid supplementation on overall and site-specific cancer incidence during the randomised trials: meta-analyses of data on 50,000 individuals. Lancet. 2013 Mar 23;381(9871):1029-36. doi: 10.1016/S0140-6736(12)62001-7[]
  161. van Wijngaarden JP, Swart KM, Enneman AW, Dhonukshe-Rutten RA, van Dijk SC, Ham AC, Brouwer-Brolsma EM, van der Zwaluw NL, Sohl E, van Meurs JB, Zillikens MC, van Schoor NM, van der Velde N, Brug J, Uitterlinden AG, Lips P, de Groot LC. Effect of daily vitamin B-12 and folic acid supplementation on fracture incidence in elderly individuals with an elevated plasma homocysteine concentration: B-PROOF, a randomized controlled trial. Am J Clin Nutr. 2014 Dec;100(6):1578-86. doi: 10.3945/ajcn.114.090043. Erratum in: Am J Clin Nutr. 2020 Jul 1;112(1):239. doi: 10.1093/ajcn/nqaa129[]
  162. Tu H, Dinney CP, Ye Y, Grossman HB, Lerner SP, Wu X. Is folic acid safe for non-muscle-invasive bladder cancer patients? An evidence-based cohort study. Am J Clin Nutr. 2018 Feb 1;107(2):208-216. doi: 10.1093/ajcn/nqx019[]
  163. Kim SJ, Zuchniak A, Sohn KJ, Lubinski J, Demsky R, Eisen A, Akbari MR, Kim YI, Narod SA, Kotsopoulos J. Plasma folate, vitamin B-6, and vitamin B-12 and breast cancer risk in BRCA1- and BRCA2-mutation carriers: a prospective study. Am J Clin Nutr. 2016 Sep;104(3):671-7. doi: 10.3945/ajcn.116.133470[]
  164. Figueiredo JC, Grau MV, Haile RW, Sandler RS, Summers RW, Bresalier RS, Burke CA, McKeown-Eyssen GE, Baron JA. Folic acid and risk of prostate cancer: results from a randomized clinical trial. J Natl Cancer Inst. 2009 Mar 18;101(6):432-5. doi: 10.1093/jnci/djp019[]
  165. Tomaszewski JJ, Cummings JL, Parwani AV, Dhir R, Mason JB, Nelson JB, Bacich DJ, O’Keefe DS. Increased cancer cell proliferation in prostate cancer patients with high levels of serum folate. Prostate. 2011 Sep;71(12):1287-93. doi: 10.1002/pros.21346[]
  166. Wien TN, Pike E, Wisløff T, Staff A, Smeland S, Klemp M. Cancer risk with folic acid supplements: a systematic review and meta-analysis. BMJ Open. 2012 Jan 12;2(1):e000653. doi: 10.1136/bmjopen-2011-000653[]
  167. Kim S, Choi BY, Nam JH, Kim MK, Oh DH, Yang YJ. Cognitive impairment is associated with elevated serum homocysteine levels among older adults. Eur J Nutr. 2019 Feb;58(1):399-408. doi: 10.1007/s00394-017-1604-y[][][]
  168. Kim YI. Folate: a magic bullet or a double edged sword for colorectal cancer prevention? Gut. 2006 Oct;55(10):1387-9. doi: 10.1136/gut.2006.095463[][]
  169. Mason JB, Tang SY. Folate status and colorectal cancer risk: A 2016 update. Mol Aspects Med. 2017 Feb;53:73-79. doi: 10.1016/j.mam.2016.11.010[]
  170. Lee JE, Willett WC, Fuchs CS, Smith-Warner SA, Wu K, Ma J, Giovannucci E. Folate intake and risk of colorectal cancer and adenoma: modification by time. Am J Clin Nutr. 2011 Apr;93(4):817-25. doi: 10.3945/ajcn.110.007781[]
  171. National Toxicology Program. NTP monograph: identifying research needs for assessing safe use of high intakes of folic acid. National Toxicology Program, 2015. https://ntp.niehs.nih.gov/sites/default/files/ntp/ohat/folicacid/final_monograph_508.pdf[][]
  172. Clarke R, Halsey J, Lewington S, Lonn E, Armitage J, Manson JE, Bønaa KH, Spence JD, Nygård O, Jamison R, Gaziano JM, Guarino P, Bennett D, Mir F, Peto R, Collins R; B-Vitamin Treatment Trialists’ Collaboration. Effects of lowering homocysteine levels with B vitamins on cardiovascular disease, cancer, and cause-specific mortality: Meta-analysis of 8 randomized trials involving 37 485 individuals. Arch Intern Med. 2010 Oct 11;170(18):1622-31. doi: 10.1001/archinternmed.2010.348[][][][]
  173. Huang T, Chen Y, Yang B, Yang J, Wahlqvist ML, Li D. Meta-analysis of B vitamin supplementation on plasma homocysteine, cardiovascular and all-cause mortality. Clin Nutr. 2012 Aug;31(4):448-54. doi: 10.1016/j.clnu.2011.01.003[]
  174. Toole JF, Malinow MR, Chambless LE, Spence JD, Pettigrew LC, Howard VJ, Sides EG, Wang CH, Stampfer M. Lowering homocysteine in patients with ischemic stroke to prevent recurrent stroke, myocardial infarction, and death: the Vitamin Intervention for Stroke Prevention (VISP) randomized controlled trial. JAMA. 2004 Feb 4;291(5):565-75. doi: 10.1001/jama.291.5.565[]
  175. Lonn E, Yusuf S, Arnold MJ, Sheridan P, Pogue J, Micks M, McQueen MJ, Probstfield J, Fodor G, Held C, Genest J Jr; Heart Outcomes Prevention Evaluation (HOPE) 2 Investigators. Homocysteine lowering with folic acid and B vitamins in vascular disease. N Engl J Med. 2006 Apr 13;354(15):1567-77. doi: 10.1056/NEJMoa060900. Epub 2006 Mar 12. Erratum in: N Engl J Med. 2006 Aug 17;355(7):746.[][]
  176. Albert CM, Cook NR, Gaziano JM, Zaharris E, MacFadyen J, Danielson E, Buring JE, Manson JE. Effect of folic acid and B vitamins on risk of cardiovascular events and total mortality among women at high risk for cardiovascular disease: a randomized trial. JAMA. 2008 May 7;299(17):2027-36. doi: 10.1001/jama.299.17.2027[][][]
  177. Ebbing M, Bleie Ø, Ueland PM, Nordrehaug JE, Nilsen DW, Vollset SE, Refsum H, Pedersen EK, Nygård O. Mortality and cardiovascular events in patients treated with homocysteine-lowering B vitamins after coronary angiography: a randomized controlled trial. JAMA. 2008 Aug 20;300(7):795-804. doi: 10.1001/jama.300.7.795[]
  178. Christen WG, Cook NR, Van Denburgh M, Zaharris E, Albert CM, Manson JE. Effect of Combined Treatment With Folic Acid, Vitamin B6, and Vitamin B12 on Plasma Biomarkers of Inflammation and Endothelial Dysfunction in Women. J Am Heart Assoc. 2018 May 18;7(11):e008517. doi: 10.1161/JAHA.117.008517[]
  179. Huo Y, Li J, Qin X, Huang Y, et al. CSPPT Investigators. Efficacy of folic acid therapy in primary prevention of stroke among adults with hypertension in China: the CSPPT randomized clinical trial. JAMA. 2015 Apr 7;313(13):1325-35. doi: 10.1001/jama.2015.2274[][]
  180. Study of the Effectiveness of Additional Reductions in Cholesterol and Homocysteine (SEARCH) Collaborative Group; Armitage JM, Bowman L, Clarke RJ, Wallendszus K, Bulbulia R, Rahimi K, Haynes R, Parish S, Sleight P, Peto R, Collins R. Effects of homocysteine-lowering with folic acid plus vitamin B12 vs placebo on mortality and major morbidity in myocardial infarction survivors: a randomized trial. JAMA. 2010 Jun 23;303(24):2486-94. doi: 10.1001/jama.2010.840[]
  181. Huo Y, Qin X, Wang J, Sun N, Zeng Q, Xu X, Liu L, Xu X, Wang X. Efficacy of folic acid supplementation in stroke prevention: new insight from a meta-analysis. Int J Clin Pract. 2012 Jun;66(6):544-51. doi: 10.1111/j.1742-1241.2012.02929.x[][]
  182. Christen WG, Cook NR, Van Denburgh M, Zaharris E, Albert CM, Manson JE. Effect of Combined Treatment With Folic Acid, Vitamin B6, and Vitamin B12 on Plasma Biomarkers of Inflammation and Endothelial Dysfunction in Women. J Am Heart Assoc. 2018 May 18;7(11):e008517. doi: 10.1161/JAHA.117.008517.[]
  183. Kong X, Huang X, Zhao M, Xu B, Xu R, Song Y, Yu Y, Yang W, Zhang J, Liu L, Zhang Y, Tang G, Wang B, Hou FF, Li P, Cheng X, Zhao S, Wang X, Qin X, Li J, Huo Y. Platelet Count Affects Efficacy of Folic Acid in Preventing First Stroke. J Am Coll Cardiol. 2018 May 15;71(19):2136-2146. doi: 10.1016/j.jacc.2018.02.072[]
  184. Stampfer M, Willett W. Folate supplements for stroke prevention: targeted trial trumps the rest. JAMA. 2015 Apr 7;313(13):1321-2. doi: 10.1001/jama.2015.1961[]
  185. Martí-Carvajal AJ, Solà I, Lathyris D. Homocysteine-lowering interventions for preventing cardiovascular events. Cochrane Database Syst Rev. 2015 Jan 15;1:CD006612. doi: 10.1002/14651858.CD006612.pub4. Update in: Cochrane Database Syst Rev. 2017 Aug 17;8:CD006612. doi: 10.1002/14651858.CD006612.pub5[]
  186. Jenkins DJA, Spence JD, Giovannucci EL, et al. Supplemental Vitamins and Minerals for CVD Prevention and Treatment. J Am Coll Cardiol. 2018 Jun 5;71(22):2570-2584. doi: 10.1016/j.jacc.2018.04.020[]
  187. Tian T, Yang KQ, Cui JG, Zhou LL, Zhou XL. Folic Acid Supplementation for Stroke Prevention in Patients With Cardiovascular Disease. Am J Med Sci. 2017 Oct;354(4):379-387. doi: 10.1016/j.amjms.2017.05.020[]
  188. Seshadri S, Beiser A, Selhub J, Jacques PF, Rosenberg IH, D’Agostino RB, Wilson PW, Wolf PA. Plasma homocysteine as a risk factor for dementia and Alzheimer’s disease. N Engl J Med. 2002 Feb 14;346(7):476-83. doi: 10.1056/NEJMoa011613[][]
  189. Ravaglia G, Forti P, Maioli F, Martelli M, Servadei L, Brunetti N, Porcellini E, Licastro F. Homocysteine and folate as risk factors for dementia and Alzheimer disease. Am J Clin Nutr. 2005 Sep;82(3):636-43. doi: 10.1093/ajcn.82.3.636[][]
  190. Clarke R. B-vitamins and prevention of dementia. Proc Nutr Soc. 2008 Feb;67(1):75-81. doi: 10.1017/S0029665108006046[]
  191. Smith AD, Refsum H. Homocysteine, B Vitamins, and Cognitive Impairment. Annu Rev Nutr. 2016 Jul 17;36:211-39. doi: 10.1146/annurev-nutr-071715-050947[][][][]
  192. Smith AD, Refsum H, Bottiglieri T, Fenech M, Hooshmand B, McCaddon A, Miller JW, Rosenberg IH, Obeid R. Homocysteine and Dementia: An International Consensus Statement. J Alzheimers Dis. 2018;62(2):561-570. doi: 10.3233/JAD-171042[][]
  193. Hooshmand B, Solomon A, Kåreholt I, Rusanen M, Hänninen T, Leiviskä J, Winblad B, Laatikainen T, Soininen H, Kivipelto M. Associations between serum homocysteine, holotranscobalamin, folate and cognition in the elderly: a longitudinal study. J Intern Med. 2012 Feb;271(2):204-12. doi: 10.1111/j.1365-2796.2011.02484.x[]
  194. Eussen SJ, de Groot LC, Joosten LW, Bloo RJ, Clarke R, Ueland PM, Schneede J, Blom HJ, Hoefnagels WH, van Staveren WA. Effect of oral vitamin B-12 with or without folic acid on cognitive function in older people with mild vitamin B-12 deficiency: a randomized, placebo-controlled trial. Am J Clin Nutr. 2006 Aug;84(2):361-70. doi: 10.1093/ajcn/84.1.361[]
  195. van der Zwaluw NL, Dhonukshe-Rutten RA, van Wijngaarden JP, Brouwer-Brolsma EM, van de Rest O, In ‘t Veld PH, Enneman AW, van Dijk SC, Ham AC, Swart KM, van der Velde N, van Schoor NM, van der Cammen TJ, Uitterlinden AG, Lips P, Kessels RP, de Groot LC. Results of 2-year vitamin B treatment on cognitive performance: secondary data from an RCT. Neurology. 2014 Dec 2;83(23):2158-66. doi: 10.1212/WNL.0000000000001050[]
  196. Kang JH, Cook N, Manson J, Buring JE, Albert CM, Grodstein F. A trial of B vitamins and cognitive function among women at high risk of cardiovascular disease. Am J Clin Nutr. 2008 Dec;88(6):1602-10. doi: 10.3945/ajcn.2008.26404[]
  197. Aisen PS, Schneider LS, Sano M, Diaz-Arrastia R, van Dyck CH, Weiner MF, Bottiglieri T, Jin S, Stokes KT, Thomas RG, Thal LJ; Alzheimer Disease Cooperative Study. High-dose B vitamin supplementation and cognitive decline in Alzheimer disease: a randomized controlled trial. JAMA. 2008 Oct 15;300(15):1774-83. doi: 10.1001/jama.300.15.1774[]
  198. Walker JG, Batterham PJ, Mackinnon AJ, Jorm AF, Hickie I, Fenech M, Kljakovic M, Crisp D, Christensen H. Oral folic acid and vitamin B-12 supplementation to prevent cognitive decline in community-dwelling older adults with depressive symptoms–the Beyond Ageing Project: a randomized controlled trial. Am J Clin Nutr. 2012 Jan;95(1):194-203. doi: 10.3945/ajcn.110.007799. Epub 2011 Dec 14. Erratum in: Am J Clin Nutr. 2012 Aug;96(2):448. Dosage error in article text.[]
  199. Clarke R, Bennett D, Parish S, Lewington S, Skeaff M, Eussen SJ, Lewerin C, Stott DJ, Armitage J, Hankey GJ, Lonn E, Spence JD, Galan P, de Groot LC, Halsey J, Dangour AD, Collins R, Grodstein F; B-Vitamin Treatment Trialists’ Collaboration. Effects of homocysteine lowering with B vitamins on cognitive aging: meta-analysis of 11 trials with cognitive data on 22,000 individuals. Am J Clin Nutr. 2014 Aug;100(2):657-66. doi: 10.3945/ajcn.113.076349[]
  200. Balk EM, Raman G, Tatsioni A, Chung M, Lau J, Rosenberg IH. Vitamin B6, B12, and folic acid supplementation and cognitive function: a systematic review of randomized trials. Arch Intern Med. 2007 Jan 8;167(1):21-30. doi: 10.1001/archinte.167.1.21[]
  201. Malouf R, Grimley Evans J. Folic acid with or without vitamin B12 for the prevention and treatment of healthy elderly and demented people. Cochrane Database Syst Rev. 2008 Oct 8;(4):CD004514. doi: 10.1002/14651858.CD004514.pub2[]
  202. Dangour AD, Whitehouse PJ, Rafferty K, Mitchell SA, Smith L, Hawkesworth S, Vellas B. B-vitamins and fatty acids in the prevention and treatment of Alzheimer’s disease and dementia: a systematic review. J Alzheimers Dis. 2010;22(1):205-24. doi: 10.3233/JAD-2010-090940[]
  203. Ford AH, Almeida OP. Effect of homocysteine lowering treatment on cognitive function: a systematic review and meta-analysis of randomized controlled trials. J Alzheimers Dis. 2012;29(1):133-49. doi: 10.3233/JAD-2012-111739[]
  204. Durga J, van Boxtel MP, Schouten EG, Kok FJ, Jolles J, Katan MB, Verhoef P. Effect of 3-year folic acid supplementation on cognitive function in older adults in the FACIT trial: a randomised, double blind, controlled trial. Lancet. 2007 Jan 20;369(9557):208-16. doi: 10.1016/S0140-6736(07)60109-3[]
  205. Huang X, Fan Y, Han X, Huang Z, Yu M, Zhang Y, Xu Q, Li X, Wang X, Lu C, Xia Y. Association between Serum Vitamin Levels and Depression in U.S. Adults 20 Years or Older Based on National Health and Nutrition Examination Survey 2005⁻2006. Int J Environ Res Public Health. 2018 Jun 9;15(6):1215. doi: 10.3390/ijerph15061215[][]
  206. Gougeon L, Payette H, Morais JA, Gaudreau P, Shatenstein B, Gray-Donald K. Intakes of folate, vitamin B6 and B12 and risk of depression in community-dwelling older adults: the Quebec Longitudinal Study on Nutrition and Aging. Eur J Clin Nutr. 2016 Mar;70(3):380-5. doi: 10.1038/ejcn.2015.202[][]
  207. Morris MS, Fava M, Jacques PF, Selhub J, Rosenberg IH. Depression and folate status in the US Population. Psychother Psychosom. 2003 Mar-Apr;72(2):80-7. doi: 10.1159/000068692[][]
  208. Papakostas GI, Petersen T, Mischoulon D, Ryan JL, Nierenberg AA, Bottiglieri T, Rosenbaum JF, Alpert JE, Fava M. Serum folate, vitamin B12, and homocysteine in major depressive disorder, Part 1: predictors of clinical response in fluoxetine-resistant depression. J Clin Psychiatry. 2004 Aug;65(8):1090-5. doi: 10.4088/jcp.v65n0810[]
  209. Trujillo J, Vieira MC, Lepsch J, Rebelo F, Poston L, Pasupathy D, Kac G. A systematic review of the associations between maternal nutritional biomarkers and depression and/or anxiety during pregnancy and postpartum. J Affect Disord. 2018 May;232:185-203. doi: 10.1016/j.jad.2018.02.004[]
  210. Chong MF, Wong JX, Colega M, Chen LW, van Dam RM, Tan CS, Lim AL, Cai S, Broekman BF, Lee YS, Saw SM, Kwek K, Godfrey KM, Chong YS, Gluckman P, Meaney MJ, Chen H; GUSTO study group. Relationships of maternal folate and vitamin B12 status during pregnancy with perinatal depression: The GUSTO study. J Psychiatr Res. 2014 Aug;55:110-6. doi: 10.1016/j.jpsychires.2014.04.006[]
  211. Blunden CH, Inskip HM, Robinson SM, Cooper C, Godfrey KM, Kendrick TR. Postpartum depressive symptoms: the B-vitamin link. Ment Health Fam Med. 2012 Jan;9(1):5-13. https://pmc.ncbi.nlm.nih.gov/articles/PMC3487611[]
  212. Yan J, Liu Y, Cao L, Zheng Y, Li W, Huang G. Association between Duration of Folic Acid Supplementation during Pregnancy and Risk of Postpartum Depression. Nutrients. 2017 Nov 2;9(11):1206. doi: 10.3390/nu9111206[]
  213. Coppen A, Bailey J. Enhancement of the antidepressant action of fluoxetine by folic acid: a randomised, placebo controlled trial. J Affect Disord. 2000 Nov;60(2):121-30. doi: 10.1016/s0165-0327(00)00153-1[]
  214. Bedson E, Bell D, Carr D, Carter B, Hughes D, Jorgensen A, Lewis H, Lloyd K, McCaddon A, Moat S, Pink J, Pirmohamed M, Roberts S, Russell I, Sylvestre Y, Tranter R, Whitaker R, Wilkinson C, Williams N. Folate Augmentation of Treatment–Evaluation for Depression (FolATED): randomised trial and economic evaluation. Health Technol Assess. 2014 Jul;18(48):vii-viii, 1-159. doi: 10.3310/hta18480[]
  215. Roberts E, Carter B, Young AH. Caveat emptor: Folate in unipolar depressive illness, a systematic review and meta-analysis. J Psychopharmacol. 2018 Apr;32(4):377-384. doi: 10.1177/0269881118756060[][][]
  216. Sarris J, Murphy J, Mischoulon D, Papakostas GI, Fava M, Berk M, Ng CH. Adjunctive Nutraceuticals for Depression: A Systematic Review and Meta-Analyses. Am J Psychiatry. 2016 Jun 1;173(6):575-87. doi: 10.1176/appi.ajp.2016.15091228[][]
  217. Cleare A, Pariante CM, Young AH, Anderson IM, Christmas D, Cowen PJ, Dickens C, Ferrier IN, Geddes J, Gilbody S, Haddad PM, Katona C, Lewis G, Malizia A, McAllister-Williams RH, Ramchandani P, Scott J, Taylor D, Uher R; Members of the Consensus Meeting. Evidence-based guidelines for treating depressive disorders with antidepressants: A revision of the 2008 British Association for Psychopharmacology guidelines. J Psychopharmacol. 2015 May;29(5):459-525. doi: 10.1177/0269881115581093[][]
  218. Ravindran AV, Balneaves LG, Faulkner G, Ortiz A, McIntosh D, Morehouse RL, Ravindran L, Yatham LN, Kennedy SH, Lam RW, MacQueen GM, Milev RV, Parikh SV; CANMAT Depression Work Group. Canadian Network for Mood and Anxiety Treatments (CANMAT) 2016 Clinical Guidelines for the Management of Adults with Major Depressive Disorder: Section 5. Complementary and Alternative Medicine Treatments. Can J Psychiatry. 2016 Sep;61(9):576-87. doi: 10.1177/0706743716660290[][]
  219. Papakostas GI, Shelton RC, Zajecka JM, Etemad B, Rickels K, Clain A, Baer L, Dalton ED, Sacco GR, Schoenfeld D, Pencina M, Meisner A, Bottiglieri T, Nelson E, Mischoulon D, Alpert JE, Barbee JG, Zisook S, Fava M. L-methylfolate as adjunctive therapy for SSRI-resistant major depression: results of two randomized, double-blind, parallel-sequential trials. Am J Psychiatry. 2012 Dec;169(12):1267-74. doi: 10.1176/appi.ajp.2012.11071114[][]
  220. Jardine LF, Ingram LC, Bleyer WA. Intrathecal leucovorin after intrathecal methotrexate overdose. J Pediatr Hematol Oncol. 1996 Aug;18(3):302-4. doi: 10.1097/00043426-199608000-00014[]
  221. Apeland T, Mansoor MA, Strandjord RE. Antiepileptic drugs as independent predictors of plasma total homocysteine levels. Epilepsy Res. 2001 Nov;47(1-2):27-35. doi: 10.1016/s0920-1211(01)00288-1[]
  222. Folate. In: Hendler SS, Rorvik, D.R., ed. PDR for Nutritional Supplements. 2nd ed. Montvale: Physicians’ Desk Reference Inc.; 2008.[]
  223. Desmoulin SK, Wang L, Polin L, White K, Kushner J, Stout M, Hou Z, Cherian C, Gangjee A, Matherly LH. Functional loss of the reduced folate carrier enhances the antitumor activities of novel antifolates with selective uptake by the proton-coupled folate transporter. Mol Pharmacol. 2012 Oct;82(4):591-600. doi: 10.1124/mol.112.079004[]
  224. Wilson SM, Bivins BN, Russell KA, Bailey LB. Oral contraceptive use: impact on folate, vitamin B₆, and vitamin B₁₂ status. Nutr Rev. 2011 Oct;69(10):572-83. doi: 10.1111/j.1753-4887.2011.00419.x[]
  225. Johnson MA. If high folic acid aggravates vitamin B12 deficiency what should be done about it? Nutr Rev. 2007 Oct;65(10):451-8. doi: 10.1111/j.1753-4887.2007.tb00270.x[]
  226. Morris MS, Jacques PF, Rosenberg IH, Selhub J. Folate and vitamin B-12 status in relation to anemia, macrocytosis, and cognitive impairment in older Americans in the age of folic acid fortification. Am J Clin Nutr. 2007 Jan;85(1):193-200. doi: 10.1093/ajcn/85.1.193[][]
  227. Selhub J, Morris MS, Jacques PF. In vitamin B12 deficiency, higher serum folate is associated with increased total homocysteine and methylmalonic acid concentrations. Proc Natl Acad Sci U S A. 2007 Dec 11;104(50):19995-20000. doi: 10.1073/pnas.0709487104[]
  228. Selhub J, Morris MS, Jacques PF, Rosenberg IH. Folate-vitamin B-12 interaction in relation to cognitive impairment, anemia, and biochemical indicators of vitamin B-12 deficiency. Am J Clin Nutr. 2009 Feb;89(2):702S-6S. doi: 10.3945/ajcn.2008.26947C. Epub 2009 Jan 13. Erratum in: Am J Clin Nutr. 2009 Jun;89(6):1951.[]
  229. Berry RJ, Carter HK, Yang Q. Cognitive impairment in older Americans in the age of folic acid fortification. Am J Clin Nutr. 2007 Jul;86(1):265-7; author reply 267-9. doi: 10.1093/ajcn/86.1.265[]
  230. Carmel R. Does high folic acid intake affect unrecognized cobalamin deficiency, and how will we know it if we see it? Am J Clin Nutr. 2009 Dec;90(6):1449-50. doi: 10.3945/ajcn.2009.28835[]
  231. Field MS, Stover PJ. Safety of folic acid. Ann N Y Acad Sci. 2018 Feb;1414(1):59-71. doi: 10.1111/nyas.13499[]
  232. Valera-Gran D, Navarrete-Muñoz EM, Garcia de la Hera M, Fernández-Somoano A, Tardón A, Ibarluzea J, Balluerka N, Murcia M, González-Safont L, Romaguera D, Julvez J, Vioque J; INMA Project. Effect of maternal high dosages of folic acid supplements on neurocognitive development in children at 4-5 y of age: the prospective birth cohort Infancia y Medio Ambiente (INMA) study. Am J Clin Nutr. 2017 Sep;106(3):878-887. doi: 10.3945/ajcn.117.152769[]
  233. Troen AM, Mitchell B, Sorensen B, Wener MH, Johnston A, Wood B, Selhub J, McTiernan A, Yasui Y, Oral E, Potter JD, Ulrich CM. Unmetabolized folic acid in plasma is associated with reduced natural killer cell cytotoxicity among postmenopausal women. J Nutr. 2006 Jan;136(1):189-94. doi: 10.1093/jn/136.1.189[][]
  234. Paniz C, Bertinato JF, Lucena MR, De Carli E, Amorim PMDS, Gomes GW, Palchetti CZ, Figueiredo MS, Pfeiffer CM, Fazili Z, Green R, Guerra-Shinohara EM. A Daily Dose of 5 mg Folic Acid for 90 Days Is Associated with Increased Serum Unmetabolized Folic Acid and Reduced Natural Killer Cell Cytotoxicity in Healthy Brazilian Adults. J Nutr. 2017 Sep;147(9):1677-1685. doi: 10.3945/jn.117.247445[][]
  235. Best KP, Green TJ, Sulistyoningrum DC, Sullivan TR, Aufreiter S, Prescott SL, Makrides M, Skubisz M, O’Connor DL, Palmer DJ. Maternal Late-Pregnancy Serum Unmetabolized Folic Acid Concentrations Are Not Associated with Infant Allergic Disease: A Prospective Cohort Study. J Nutr. 2021 Jun 1;151(6):1553-1560. doi: 10.1093/jn/nxab040[]
  236. Husebye ESN, Wendel AWK, Gilhus NE, Riedel B, Bjørk MH. 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. 2022 May 1;115(5):1432-1440. doi: 10.1093/ajcn/nqab436[]
  237. Tam C, O’Connor D, Koren G. Circulating unmetabolized folic Acid: relationship to folate status and effect of supplementation. Obstet Gynecol Int. 2012;2012:485179. doi: 10.1155/2012/485179[]
  238. Morris MS, Jacques PF, Rosenberg IH, Selhub J. Circulating unmetabolized folic acid and 5-methyltetrahydrofolate in relation to anemia, macrocytosis, and cognitive test performance in American seniors. Am J Clin Nutr. 2010 Jun;91(6):1733-44. doi: 10.3945/ajcn.2009.28671. Epub 2010 Mar 31. Erratum in: Am J Clin Nutr. 2010 Oct;92(4):1002.[]
  239. Pfeiffer CM, Sternberg MR, Fazili Z, Yetley EA, Lacher DA, Bailey RL, Johnson CL. Unmetabolized folic acid is detected in nearly all serum samples from US children, adolescents, and adults. J Nutr. 2015 Mar;145(3):520-31. doi: 10.3945/jn.114.201210[]
  240. Stamm RA, March KM, Karakochuk CD, Gray AR, Brown RC, Green TJ, Houghton LA. Lactating Canadian Women Consuming 1000 µg Folic Acid Daily Have High Circulating Serum Folic Acid Above a Threshold Concentration of Serum Total Folate. J Nutr. 2018 Jul 1;148(7):1103-1108. doi: 10.1093/jn/nxy070. Erratum in: J Nutr. 2019 Apr 1;149(4):698. doi: 10.1093/jn/nxy214[]
  241. Page R, Robichaud A, Arbuckle TE, Fraser WD, MacFarlane AJ. Total folate and unmetabolized folic acid in the breast milk of a cross-section of Canadian women. Am J Clin Nutr. 2017 May;105(5):1101-1109. doi: 10.3945/ajcn.116.137968[]
  242. Obeid R, Kasoha M, Kirsch SH, Munz W, Herrmann W. Concentrations of unmetabolized folic acid and primary folate forms in pregnant women at delivery and in umbilical cord blood. Am J Clin Nutr. 2010 Dec;92(6):1416-22. doi: 10.3945/ajcn.2010.29361[]
  243. Plumptre L, Masih SP, Ly A, Aufreiter S, Sohn KJ, Croxford R, Lausman AY, Berger H, O’Connor DL, Kim YI. High concentrations of folate and unmetabolized folic acid in a cohort of pregnant Canadian women and umbilical cord blood. Am J Clin Nutr. 2015 Oct;102(4):848-57. doi: 10.3945/ajcn.115.110783[]
  244. Sweeney MR, McPartlin J, Scott J. Folic acid fortification and public health: report on threshold doses above which unmetabolised folic acid appear in serum. BMC Public Health. 2007 Mar 22;7:41. doi: 10.1186/1471-2458-7-41[]
  245. Kelly P, McPartlin J, Goggins M, Weir DG, Scott JM. Unmetabolized folic acid in serum: acute studies in subjects consuming fortified food and supplements. Am J Clin Nutr. 1997 Jun;65(6):1790-5. doi: 10.1093/ajcn/65.6.1790[]
  246. Sweeney MR, McPartlin J, Weir DG, Daly L, Scott JM. Postprandial serum folic acid response to multiple doses of folic acid in fortified bread. Br J Nutr. 2006 Jan;95(1):145-51. doi: 10.1079/bjn20051618[]
Health Jade