C20H23N7O7
58-05-9
Advanced Colorectal Cancer, Advanced Esophageal Cancers, Anemia of Pregnancy, Bladder Cancer, Folate and iron deficiency, Folate deficiency, Folic acid antagonist overdose, Iron Deficiency (ID), Macrocytic anemia, Megaloblastic anemia, Pancreatic Metastatic Cancer, Postpartum Anemia, Stage IV Gastric Cancer, Hypochromic anemia, Methotrexate toxicity, Normochromic anemia, Pyrimethamine hematologic toxicity
Leucovorin, known interchangeably as folinic acid, is a chemically reduced derivative of folic acid that occupies a unique and critical position in modern pharmacotherapy.[1] Its clinical utility is defined by two distinct and seemingly contradictory pharmacological roles. Primarily, it functions as a "rescue" agent, or antidote, administered to mitigate the severe, dose-limiting toxicities of folic acid antagonists, most notably the chemotherapeutic agent methotrexate.[1] In this capacity, it selectively protects healthy host cells from cytotoxic damage. Conversely, Leucovorin also serves as a chemopotentiator, purposefully administered to enhance the cytotoxic efficacy of fluoropyrimidine-based chemotherapy, such as 5-fluorouracil (5-FU), particularly in the treatment of colorectal cancer.[1] This monograph is structured around this central duality, exploring the molecular basis and clinical implications of these opposing functions.
The drug's history dates back to its 1948 discovery as an essential growth cofactor for the bacterium Leuconostoc citrovorum, leading to its original name, "citrovorum factor".[1] With a long history of clinical use, it received its initial approval from the U.S. Food and Drug Administration (FDA) in 1952, underscoring its enduring importance in medicine.[6]
Leucovorin is classified as a small molecule folate analog.[2] Its precise chemical identity and properties are fundamental to its biological activity.
For clarity and ease of reference, the key chemical and physical identifiers for Leucovorin are consolidated in Table 1.1. This centralization is crucial for accurate substance identification in research, procurement, and regulatory contexts, particularly given the multiple CAS numbers associated with the drug and its various salt forms.
Table 1.1: Key Chemical and Physical Identifiers for Leucovorin
Identifier | Value | Source(s) |
---|---|---|
Common Names | Leucovorin, Folinic Acid, Citrovorum Factor | 1 |
IUPAC Name | (2S)-2-{[(2-amino-5-formyl-4-oxo-5,6,7,8-tetrahydro-1H-pteridin-6-yl)methylamino] benzoyl]amino}pentanedioic acid | 1 |
DrugBank ID | DB00650 | 1 |
CAS Number (Free Acid) | 58-05-9 | 10 |
CAS Number (Calcium Salt) | 1492-18-8 | 1 |
UNII | RPR1R4C0P4 (Calcium Salt) | 1 |
PubChem CID | 6006 (Free Acid) | 1 |
ChEMBL ID | CHEMBL1679 (Free Acid) | 1 |
ATC Code | V03AF03 | 1 |
Chemical Formula | C20H23N7O7 | 1 |
Molar Mass | 473.446 g·mol⁻¹ | 1 |
Physical State | White to light yellow powder | 13 |
Water Solubility | ~0.3 mg/mL (20 °C) | 1 |
Melting Point | 245 °C (473 °F) with decomposition | 1 |
A point of critical pharmacological importance is the stereochemistry of Leucovorin. The commercially available drug, often referred to simply as Leucovorin, is a racemic mixture containing equal amounts of two diastereoisomers of the 5-formyl derivative of tetrahydrofolic acid (THF): the dextrorotatory (d) and levorotatory (l) forms.[2]
Extensive research has demonstrated that only the levorotatory l-isomer, also known as (-)-folinic acid or levoleucovorin, is biologically active.[2] This is the isomer that can be metabolized and utilized by the body to participate in folate-dependent one-carbon transfer reactions. The d-isomer is pharmacologically inert and does not contribute to the therapeutic effect.[14]
This distinction led to the development and subsequent FDA approval in 2008 of levoleucovorin, a formulation containing only the pure, active l-isomer.[7] Reflecting its composition of 100% active drug, levoleucovorin is dosed at one-half the usual dose of racemic Leucovorin, possessing approximately twice the potency of the racemic mixture.[9]
The presence of the inactive d-isomer in racemic formulations creates a significant "pharmacokinetic burden." Pharmacokinetic studies reveal that the two isomers have vastly different fates in the body. The active l-isomer is rapidly metabolized and cleared, with a plasma half-life of about 32 minutes.[3] In contrast, the inactive d-isomer cannot be metabolized and is cleared much more slowly, exclusively through renal excretion, resulting in a half-life that is over ten times longer, at approximately 451 minutes.[14] This differential clearance leads to the accumulation and prolonged persistence of the inactive d-isomer in the plasma, where its concentrations can greatly exceed those of the active l-isomer and its metabolites.[15] While the d-isomer is considered pharmacologically inert, the sustained presence of a high concentration of any chemical entity is undesirable. It introduces a theoretical risk of competitive inhibition at cellular transport sites, unforeseen off-target effects, or an increased load on renal clearance mechanisms. This provides a compelling scientific and clinical rationale for the development and use of the pure active isomer, levoleucovorin, which delivers the intended therapeutic benefit without this unnecessary and prolonged chemical exposure.
The clinical utility of Leucovorin stems from its role as a prodrug that is readily converted within the body to other biologically active reduced folate derivatives, such as 5,10-methylenetetrahydrofolate and 5-methyltetrahydrofolate (5-MTHF).[1] The defining feature of this process is that it completely bypasses the enzymatic step requiring dihydrofolate reductase (DHFR), the very enzyme targeted by a major class of antifolate chemotherapeutics.[1] This unique metabolic characteristic underpins Leucovorin's two primary, and opposing, pharmacodynamic functions.
Leucovorin's primary role as an antidote is in the context of high-dose methotrexate (MTX) chemotherapy.[1] Methotrexate functions by potently inhibiting the DHFR enzyme. This blockade prevents the reduction of dihydrofolate (DHF) to tetrahydrofolate (THF), a critical step in the folate metabolic pathway. The resulting depletion of the cellular pool of reduced folates halts the
de novo synthesis of purines and thymidylate, which are essential precursors for DNA and RNA synthesis. This leads to cell cycle arrest and apoptosis, affecting not only rapidly dividing cancer cells but also healthy tissues with high turnover rates, such as bone marrow and gastrointestinal mucosa, causing severe toxicity.[1]
Leucovorin "rescues" these healthy cells by providing a direct source of reduced folates that is downstream of the MTX-induced enzymatic block. Once inside the cell, Leucovorin is converted to THF and other active derivatives, replenishing the coenzyme pool and allowing normal cells to resume nucleic acid synthesis and thus survive the otherwise lethal effects of high-dose MTX.[1] This strategy, known as "Leucovorin rescue," is a cornerstone of high-dose MTX therapy. The timing of administration is paramount; Leucovorin is typically given 24 hours
after the MTX infusion to allow the chemotherapeutic to exert its antitumor effect before the rescue of normal tissues begins.[3] The efficacy of the rescue diminishes markedly as the time interval from MTX administration increases.[17]
In stark contrast to its rescue function, Leucovorin acts as a powerful synergistic agent when combined with fluoropyrimidine chemotherapies, principally 5-fluorouracil (5-FU), in the treatment of malignancies like colorectal cancer.[1] The mechanism of this potentiation is well-defined. Inside the cell, 5-FU is converted to its active metabolite, fluorodeoxyuridine monophosphate (FdUMP). FdUMP exerts its cytotoxic effect by inhibiting the enzyme thymidylate synthase (TS), which is responsible for the synthesis of thymidylate, a necessary component of DNA.[3]
Leucovorin's role is to enhance and prolong this inhibition. It is metabolized within the cell to 5,10-methylenetetrahydrofolate (5,10−CH2FH4). This specific folate derivative acts as a molecular "glue," forming a highly stable ternary covalent complex with both FdUMP and the TS enzyme (the TS-FdUMP-5,10−CH2FH4 complex).[2] This stabilization effectively locks the enzyme in an inhibited state, leading to a more profound and sustained depletion of thymidylate and a corresponding enhancement of 5-FU's DNA-damaging and cytotoxic effects.[2]
The primary molecular target that Leucovorin's action is designed to circumvent is dihydrofolate reductase (DHFR).[2] In its potentiating role, the ultimate target of the Leucovorin/5-FU combination is
thymidylate synthase (TS).[2] Furthermore, Leucovorin utilizes and can compete with methotrexate for the same cellular transport systems, such as the reduced folate carrier and the proton-coupled folate transporter, which contributes to its rescue capabilities.[2]
The clinical use of Leucovorin is profoundly influenced by its complex pharmacokinetic profile, which exhibits significant dependence on the route of administration and the stereochemistry of the formulation.
Leucovorin is absorbed rapidly following oral administration, but this process is mediated by a saturable transport mechanism in the gastrointestinal tract.[3] This saturation has direct and critical clinical implications. The apparent bioavailability is highly dose-dependent: at a 25 mg dose, it is 97%, but this value decreases to 75% for a 50 mg dose and plummets to just 37% for a 100 mg dose.[16] This pharmacokinetic limitation is the scientific foundation for the clinical guideline that oral doses greater than 25 mg are not recommended, as they provide unreliable and disproportionately low systemic drug exposure.[3] For indications requiring high, predictable plasma concentrations, such as MTX rescue, parenteral administration is mandatory.
Furthermore, oral absorption is stereoselective, strongly favoring the active l-isomer. At a 25 mg dose, nearly 100% of the active l-isomer is absorbed, whereas only about 20% of the inactive d-isomer enters circulation.[15] The time to reach peak plasma concentration (Tmax) also varies by route: intravenous (IV) administration results in a peak of the parent compound in approximately 10 minutes, intramuscular (IM) administration peaks at around 52 minutes, and the oral route is the slowest, with a peak at approximately 2.3 hours.[16]
Once absorbed, Leucovorin is widely distributed throughout the body tissues, with some concentration in the liver.[3] Its volume of distribution (Vd) is reported to be around 3.2 L/kg.[3] Plasma protein binding is relatively low, with reported values ranging from approximately 15% to 35-45%.[1] Leucovorin readily crosses the blood-brain barrier, primarily in the form of its active metabolite 5-MTHF. However, the resulting concentrations in the cerebrospinal fluid (CSF) are one to three orders of magnitude lower than the typical CSF concentrations of methotrexate following intrathecal administration.[3]
Leucovorin undergoes extensive and rapid metabolism. A significant portion of an oral dose is subject to first-pass metabolism in the intestinal mucosa and the liver, where it is converted to its primary active metabolite, 5-MTHF.[3] Consequently, 5-MTHF is the predominant circulating folate species following oral administration.[16] The metabolic pathway involves the conversion of the parent compound (5-formyl-THF) into other reduced folates, including 5,10-methenyl-THF and 5,10-methylene-THF (the key metabolite for 5-FU potentiation), before its eventual conversion to the terminal metabolite 5-MTHF.[1] This metabolism is specific to the active l-isomer; the inactive d-isomer is not metabolized and is cleared from the body unchanged.[14]
The primary route of elimination for Leucovorin and its metabolites is via the kidneys, with 80-90% of a dose being excreted in the urine.[1] A smaller fraction, 5-8%, is eliminated in the feces.[3] The terminal elimination half-life (
t1/2) for total reduced folates is approximately 6.2 hours.[1] However, this composite value masks the significant differences in the half-lives of the individual components: the active l-leucovorin has a very short half-life of about 32 minutes, its active metabolite 5-MTHF has a half-life of about 227 minutes (3.8 hours), and the inert d-leucovorin persists for a much longer duration with a half-life of about 451 minutes (7.5 hours).[3]
Table 2.1: Comparative Pharmacokinetic Parameters of Leucovorin by Route of Administration
Parameter | Oral Route | Intramuscular (IM) Route | Intravenous (IV) Route |
---|---|---|---|
Bioavailability | 97% (25 mg), 75% (50 mg), 37% (100 mg) 16 | Not applicable (assumed 100%) | Not applicable (100%) |
Tmax (Parent Drug) | ~1.2 hours 16 | ~28 minutes 16 | ~10 minutes 16 |
Cmax (Total Folates) | 393 ng/mL (25 mg dose) 16 | 436 ng/mL (25 mg dose) 16 | 1259 ng/mL (25 mg dose) 16 |
Terminal Half-Life | ~5.7 hours 16 | ~6.2 hours 16 | ~6.2 hours 1 |
The mechanistic duality of Leucovorin creates two entirely different sets of clinical imperatives and safety considerations. When used with methotrexate, its function is to reverse toxicity. The primary clinical risk is therefore under-dosing Leucovorin or administering it too late, which could lead to unchecked, life-threatening MTX toxicity.[17] Conversely, when used with 5-fluorouracil, its function is to enhance toxicity. Here, the primary clinical risk is
over-potentiation, which can lead to severe and potentially fatal 5-FU side effects like diarrhea and myelosuppression.[3] This means that for MTX protocols, clinicians must be vigilant in ensuring adequate Leucovorin levels, often monitoring MTX clearance and increasing the Leucovorin dose if necessary.[13] For 5-FU protocols, the opposite is true: clinicians must often reduce the dose of 5-FU and must monitor closely for signs of enhanced toxicity, being prepared to stop treatment immediately if severe gastrointestinal symptoms appear.[25] Leucovorin cannot be viewed as a simple vitamin supplement; its safe administration requires a sophisticated, context-dependent understanding of its interaction with its partner drug.
Leucovorin has several well-established, FDA-approved indications that span both its antidote and chemopotentiation functions.[5]
This is a cornerstone indication, particularly in the treatment of osteosarcoma and other malignancies where high-dose methotrexate regimens are employed.[2] The explicit purpose is to selectively "rescue" normal, healthy cells from the profound cytotoxic effects of methotrexate, thereby permitting the administration of tumoricidal drug levels that would otherwise be unacceptably toxic to the patient.[3]
Leucovorin is indicated to diminish the toxicity resulting from an inadvertent overdosage of methotrexate or other folic acid antagonists. It is also crucial for patients in whom methotrexate elimination is impaired due to factors such as pre-existing renal insufficiency, drug-induced nephrotoxicity, or the presence of third-space fluid accumulations (e.g., ascites, pleural effusion) that can act as a reservoir for the drug.[1]
In its role as a potentiator, Leucovorin is approved for use in combination with 5-fluorouracil to prolong survival in the palliative treatment of patients with advanced colorectal cancer.[1] This indication is based on the synergistic mechanism that enhances the antitumor activity of 5-FU.
Leucovorin is indicated for the treatment of megaloblastic anemias that arise from a deficiency of folic acid, specifically in situations where oral therapy with standard folic acid is not feasible.[1] This indication carries a critical caveat: pernicious anemia, which is a megaloblastic anemia caused by Vitamin B12 deficiency, must be definitively ruled out before initiating treatment.[1]
The indication for megaloblastic anemia highlights a significant clinical paradox. Using Leucovorin in a patient with an undiagnosed Vitamin B12 deficiency can have catastrophic consequences. Both folate and B12 deficiencies can present with the same hematologic finding: megaloblastic anemia, characterized by abnormally large and immature red blood cells.[1] Administering a folate, such as Leucovorin, can provide the necessary substrate to partially bypass the B12-dependent step in red blood cell maturation, thereby correcting the anemia and making the patient's blood work appear to improve.[1] However, this hematologic improvement masks the underlying B12 deficiency. Vitamin B12 is also independently critical for the maintenance of the myelin sheath that insulates nerve fibers. By "fixing" the anemia, Leucovorin can provide a false sense of security while the severe and often irreversible neurological damage of B12 deficiency—such as subacute combined degeneration of the spinal cord—progresses silently and unchecked.[5] This establishes a non-negotiable diagnostic mandate for clinicians: before treating any megaloblastic anemia with a folate product, Vitamin B12 deficiency must be ruled out as the primary cause.
Beyond its formal approvals, Leucovorin is widely used in other clinical contexts based on established practice and ongoing research.
The safety profile of Leucovorin is highly context-dependent, differing greatly based on whether it is used alone or in combination with a cytotoxic agent.
There are several situations where the use of Leucovorin is absolutely contraindicated due to the risk of severe harm.
This is the most severe warning associated with the drug. Intrathecal administration of Leucovorin is strictly contraindicated. This route of administration has been associated with severe adverse neurological events, including death.[1] This warning is based on case reports, such as that of an 11-year-old boy who suffered severe neurotoxicity after receiving intrathecal Leucovorin.[1]
As detailed previously, Leucovorin is contraindicated for the treatment of pernicious anemia or any other megaloblastic anemia resulting from Vitamin B12 deficiency. Its use can mask the hematologic signs of the disease while allowing irreversible neurological damage to progress unabated.[1]
Leucovorin is contraindicated in any patient with a history of a severe hypersensitivity reaction to leucovorin, folic acid, or folinic acid products.[17]
While a formal "Boxed Warning" from the FDA is not present on the Leucovorin label itself, the gravity of the warnings provided is functionally equivalent.[25] The language used to describe the risks of intrathecal administration ("death has been reported") and 5-FU potentiation ("rapid clinical deterioration leading to death can occur") conveys a level of danger on par with, or exceeding, that of many drugs with boxed warnings.[7] The absence of the specific black box format should not lead to any downplaying of these life-threatening risks. Clinicians and institutions must treat these warnings with the utmost seriousness, recognizing that the danger arises not from the drug in isolation, but from its specific use in a high-risk context or via an improper route of administration.
Leucovorin's interactions with other drugs are clinically significant and central to both its efficacy and its toxicity profile.
Table 4.1: Clinically Significant Drug Interactions with Leucovorin
Interacting Drug/Class | Effect of Interaction | Mechanism | Management Recommendation |
---|---|---|---|
Fluoropyrimidines (5-Fluorouracil, Capecitabine) | Increased therapeutic and toxic effects of the fluoropyrimidine 3 | Leucovorin metabolite (5,10−CH2FH4) stabilizes the inhibitory binding of FdUMP to thymidylate synthase 3 | This is an intended interaction. Monitor closely for enhanced gastrointestinal and hematologic toxicity. The dose of 5-FU is often reduced when given with Leucovorin.19 |
Methotrexate | Decreased therapeutic and toxic effects of methotrexate 3 | Leucovorin bypasses the DHFR enzyme block, replenishing the reduced folate pool. It may also compete for cellular transport.1 | This is the intended "rescue" interaction. Administer Leucovorin after methotrexate. Monitor MTX levels to ensure adequate rescue.5 |
Antiepileptics (Phenobarbital, Phenytoin, Primidone) | Decreased efficacy of the antiepileptic drug, leading to increased seizure frequency 3 | Mechanism is not fully known but may involve interference with folate-dependent microsomal metabolism of the antiepileptics.3 | Primarily a concern with high doses of Leucovorin. Monitor for seizure control and consider checking antiepileptic drug levels.3 |
Trimethoprim/ Sulfamethoxazole | Increased rates of treatment failure and mortality when used for P. jiroveci pneumonia in HIV patients 18 | The mechanism is not fully elucidated but suggests an antagonistic interaction in this specific clinical setting. | This combination should be used with caution or avoided for this indication. |
Glucarpidase | Decreased plasma concentrations of Leucovorin 19 | Glucarpidase is an enzyme that rapidly metabolizes both methotrexate and Leucovorin.20 | Do not administer Leucovorin within two hours before or after a dose of glucarpidase to avoid its inactivation.20 |
Raltitrexed | Decreased efficacy of raltitrexed 3 | Raltitrexed is a folate analogue that inhibits thymidylate synthase; Leucovorin may interfere with this action. | Co-administration is not recommended.3 |
The dosing of Leucovorin is highly specific to the indication and, in the case of methotrexate rescue, is dynamically adjusted based on laboratory monitoring.
Table 5.1: Leucovorin Dosing Guidelines for Methotrexate Rescue
Clinical Situation | Laboratory Findings | Leucovorin Dose and Duration |
---|---|---|
Normal Methotrexate Elimination | Serum MTX level: ~10 µM at 24 hrs, ~1 µM at 48 hrs, and <0.2 µM at 72 hrs 13 | 15 mg (or ~10 mg/m²) IV/IM/PO every 6 hours for 10 doses (60 hours total), starting 24 hours after the start of the MTX infusion.13 |
Delayed Late MTX Elimination | Serum MTX level remains >0.2 µM at 72 hrs, and >0.05 µM at 96 hrs 30 | Continue 15 mg (or 7.5 mg levoleucovorin) IV/IM every 6 hours until MTX level is <0.05 µM.30 |
Delayed Early MTX Elimination or Acute Renal Injury | 24-hr serum creatinine increased ≥50% over baseline, OR 24-hr MTX level >5 µM, OR 48-hr MTX level >0.9 µM 23 | Increase Leucovorin dose to 100-150 mg/m² IV every 3 hours until MTX level is <1 µM, then decrease dose to 15 mg IV every 3 hours until MTX level is <0.05 µM.7 |
Strict adherence to preparation and administration guidelines is essential for the safe and effective use of parenteral Leucovorin.
While Leucovorin is a long-established drug, it remains an active area of clinical investigation, both for refining its role in oncology and for exploring novel applications.
Despite decades of use, several key questions about Leucovorin remain, pointing toward future avenues of research:
Leucovorin is a mature yet indispensable therapeutic agent whose clinical value is defined by a remarkable pharmacodynamic paradox. In one context, it is a life-saving antidote, rescuing healthy tissues from the brink of chemotherapeutic devastation. In another, it is a powerful synergistic partner, intentionally used to amplify the cytotoxic potential of another drug. This dual nature makes it a unique and versatile tool in the pharmacopeia, but also one that demands a high level of clinical sophistication for its safe and effective use. Its long history, from its discovery as a bacterial growth factor to its current role as a cornerstone of complex cancer regimens and a potential therapy for neurodevelopmental disorders, speaks to its enduring relevance.
The optimal and safe application of Leucovorin is entirely contingent on the clinician's deep understanding of its specific role within any given therapeutic regimen. Its use is not a one-size-fits-all proposition. Mastery of its complex pharmacokinetics—particularly the saturable nature of its oral absorption and the implications of its stereoisomerism—is essential for appropriate dose and route selection. Strict adherence to established administration protocols, especially the absolute prohibition of intrathecal use and the careful control of intravenous infusion rates, is non-negotiable for patient safety. Finally, vigilant clinical monitoring for context-specific toxicities, whether it be the risk of under-rescue from methotrexate or the risk of over-potentiation of 5-fluorouracil, is paramount. The future of Leucovorin will likely involve both the refinement of its use in oncology, potentially through the broader adoption of the purer levoleucovorin formulations, and the exciting exploration of its potential in non-oncological fields like neurology. This represents a potential new chapter for a drug that has already been a vital part of medicine for over half a century.
Published at: July 16, 2025
This report is continuously updated as new research emerges.
Empowering clinical research with data-driven insights and AI-powered tools.
© 2025 MedPath, Inc. All rights reserved.