MedPath

Leflunomide Advanced Drug Monograph

Published:Aug 4, 2025

Generic Name

Leflunomide

Brand Names

Arava, Leflunomide medac, Leflunomide Zentiva (previously Leflunomide Winthrop), Leflunomide ratiopharm

Drug Type

Small Molecule

Chemical Formula

C12H9F3N2O2

CAS Number

75706-12-6

Associated Conditions

Juvenile Idiopathic Arthritis (JIA), Rheumatoid Arthritis

A Comprehensive Monograph on Leflunomide (DB01097)

1.0 Executive Summary

Leflunomide is a pivotal isoxazole-derivative, disease-modifying antirheumatic drug (DMARD) that represents a significant therapeutic option in the management of autoimmune inflammatory conditions.[1] It functions as a prodrug, undergoing rapid and extensive conversion following oral administration to its pharmacologically active metabolite, A77 1726, also known as teriflunomide.[3] Teriflunomide is responsible for virtually all of the drug's in-vivo activity and exerts its primary therapeutic effect through the selective and reversible inhibition of the mitochondrial enzyme dihydroorotate dehydrogenase (DHODH).[3] This targeted mechanism interrupts the

de novo pyrimidine synthesis pathway, a metabolic route that is critically essential for the proliferation of activated T and B lymphocytes that drive autoimmune pathology. By depriving these specific cells of the nucleotides required for DNA and RNA synthesis, leflunomide induces a G1 phase cell cycle arrest, resulting in a targeted, cytostatic (proliferation-inhibiting) immunomodulatory effect rather than a broadly cytotoxic one.[3]

Leflunomide is approved by major regulatory bodies, including the U.S. Food and Drug Administration (FDA) and the European Medicines Agency (EMA), for the treatment of active rheumatoid arthritis (RA) and active psoriatic arthritis (PsA).[3] In these conditions, it has demonstrated efficacy in reducing signs and symptoms, improving physical function, and, crucially, inhibiting the progression of structural joint damage.[3] Beyond its approved indications, leflunomide has found notable off-label application in the management of solid organ transplant rejection, for which it holds an FDA orphan drug designation, and as an investigational therapy for lupus nephritis.[3]

The clinical use of leflunomide is profoundly influenced by its unique pharmacokinetic profile, which is characterized by high bioavailability and an exceptionally long elimination half-life of approximately two weeks.[1] This long half-life, a consequence of extensive enterohepatic circulation, necessitates the use of a loading dose to achieve therapeutic concentrations rapidly and, more critically, requires a specific accelerated elimination or "washout" procedure in instances of severe toxicity or planned pregnancy.[1] The drug's risk-benefit profile is defined by two FDA Boxed Warnings that mandate stringent clinical management: one for the risk of severe and potentially fatal hepatotoxicity, and another for the risk of embryo-fetal toxicity.[1] These warnings necessitate rigorous patient selection, counseling, and ongoing monitoring of liver function and hematologic parameters, positioning leflunomide as a potent but complex therapeutic agent in the rheumatologic armamentarium.

2.0 Identification and Physicochemical Properties

Leflunomide is a synthetic, small molecule drug belonging to the isoxazole derivative class.[1] Its precise chemical and physical properties are essential for its identification, formulation, and handling in both research and clinical settings.

2.1 Chemical Identity

The formal chemical identity of leflunomide is established by its systematic nomenclature and universally recognized identifiers. It is a monocarboxylic acid amide resulting from the formal condensation of the carboxy group of 5-methyl-1,2-oxazole-4-carboxylic acid with the anilino group of 4-(trifluoromethyl)aniline.[14]

  • International Union of Pure and Applied Chemistry (IUPAC) Name: 5-methyl-N-[4-(trifluoromethyl)phenyl]-1,2-oxazole-4-carboxamide.[7]
  • Synonyms: The compound is referred to by several synonyms in scientific literature and commercial contexts, including N-(4-Trifluoromethyphenyl)-5-methylisoxazole-4-carboxamide, Leflunomidum, and the internal research code HWA-486.[7] Its most widely known brand name is Arava.[7]
  • Chemical Formula: The empirical formula for leflunomide is C12​H9​F3​N2​O2​.[7]
  • Molecular Weight: The molecular weight is consistently reported as 270.21 g/mol.[7]
  • Chemical Abstracts Service (CAS) Number: The unique registry number for leflunomide is 75706-12-6.[7]
  • DrugBank Accession Number: The compound is cataloged in the DrugBank database under the identifier DB01097.[3]

2.2 Physicochemical Properties

Leflunomide's physical characteristics determine its behavior in solution and its requirements for storage and administration.

  • Appearance: At room temperature, leflunomide is a white to almost white or off-white crystalline solid or powder.[15]
  • Solubility: It is characterized by poor solubility in aqueous solutions, with a reported water solubility of 27 mg/L at 25 °C, rendering it effectively insoluble.[17] However, it is soluble in various organic solvents, including Dimethyl Sulfoxide (DMSO) at concentrations up to 30 mg/ml and Ethanol at 27 mg/ml, as well as in acetone and methanol.[16]
  • Melting Point: The melting point of the crystalline solid ranges from approximately 166.0 °C to 170.0 °C.[17]
  • Stability and Storage: The compound is stable enough for shipment under ambient temperatures. For optimal long-term preservation in a research setting, it is recommended to store it in a dry, dark environment at -20 °C. Short-term storage (days to weeks) is acceptable at refrigerated temperatures of 0 °C to 4 °C.[7]

A summary of these key identifying and physical properties is provided in Table 1.

Table 1: Identification and Physicochemical Properties of Leflunomide

PropertyValueSource(s)
DrugBank IDDB010973
CAS Number75706-12-67
IUPAC Name5-methyl-N-[4-(trifluoromethyl)phenyl]-1,2-oxazole-4-carboxamide7
Chemical FormulaC12​H9​F3​N2​O2​7
Molecular Weight270.21 g/mol7
AppearanceWhite to off-white crystalline solid/powder15
Melting Point166.0 - 170.0 °C17
Water SolubilityInsoluble (27 mg/L at 25 °C)17
Solubility (Organic)Soluble in DMSO, Ethanol, Acetone, Methanol16
Storage (Long-Term)Dry, dark, -20 °C7

3.0 Pharmacology and Mechanism of Action

The pharmacological activity of leflunomide is complex, involving its conversion to an active metabolite that targets a specific and critical metabolic pathway in the immune system. While it has a primary, well-defined mechanism, secondary effects contribute to its overall immunomodulatory profile.

3.1 Prodrug Metabolism and the Active Metabolite, Teriflunomide (A77 1726)

Leflunomide itself is a pharmacologically inert prodrug.[3] Its therapeutic activity is entirely dependent on its conversion to its active metabolite. Following oral administration, leflunomide undergoes rapid and nearly complete first-pass metabolism, which occurs in the cells of the gastrointestinal wall and the liver.[1] This metabolic activation involves the opening of the isoxazole ring structure to form the metabolite A77 1726, which is also known by the non-proprietary name teriflunomide.[1]

This conversion is so efficient that the parent leflunomide compound is rarely detectable in plasma, and teriflunomide is considered responsible for essentially all of the drug's in-vivo activity.[1] The process does not appear to be mediated by a single, specific enzyme, which may contribute to a relatively consistent activation efficiency across patients.[18] Teriflunomide is itself marketed as a separate drug for multiple sclerosis under the brand name Aubagio®, highlighting its central role as the active pharmacological entity.[18]

3.2 Primary Mechanism: Inhibition of Dihydroorotate Dehydrogenase (DHODH)

The principal and most well-characterized mechanism of action for leflunomide's therapeutic effect is the reversible inhibition of the mitochondrial enzyme dihydroorotate dehydrogenase (DHODH) by its active metabolite, teriflunomide.[3]

DHODH is a pivotal, rate-limiting enzyme in the de novo synthesis pathway of pyrimidines. This pathway is responsible for the creation of new pyrimidine nucleotides, which are fundamental building blocks of DNA and RNA. Specifically, DHODH catalyzes the fourth step in this process: the conversion of dihydroorotate to orotate, a precursor to uridine monophosphate (rUMP).[2] By inhibiting DHODH, teriflunomide effectively blocks this pathway, leading to a depletion of the intracellular pyrimidine pool.[3] The resulting shortage of rUMP halts DNA and RNA synthesis, which in turn causes proliferating cells to arrest in the G1 phase of the cell cycle, preventing them from proceeding to the S phase where DNA replication occurs.[3]

The therapeutic utility of this mechanism stems from its relative specificity for certain cell types. Autoimmune diseases such as rheumatoid arthritis are characterized by the massive clonal expansion of self-reactive T and B lymphocytes.[3] During this rapid proliferation, activated lymphocytes must dramatically increase their pyrimidine pool—by an estimated eightfold—and are uniquely dependent on the

de novo synthesis pathway to meet this high metabolic demand.[2] In contrast, most other cell types in the body are either quiescent or can sufficiently meet their needs through an alternative "salvage pathway," which recycles existing pyrimidines.[2] This differential dependency is the cornerstone of leflunomide's targeted effect. It allows the drug to selectively inhibit the proliferation of the key pathogenic lymphocytes driving the autoimmune process while having a much smaller impact on other cells. This explains why the drug's action is primarily

cytostatic (inhibiting cell division) rather than broadly cytotoxic (cell-killing), a feature that helps define its therapeutic window.[3]

3.3 Secondary and Ancillary Mechanisms

While DHODH inhibition is the primary driver of its efficacy at therapeutic concentrations, leflunomide and teriflunomide exhibit other biological activities that may contribute to their overall effect, particularly at higher concentrations or in specific contexts.

  • Tyrosine Kinase Inhibition: Teriflunomide has been shown to inhibit the activity of several protein tyrosine kinases.[3] These enzymes are critical components of intracellular signaling cascades that regulate immune cell activation, proliferation, and survival. However, this inhibitory effect is generally observed at concentrations higher than those achieved during standard clinical treatment for RA.[6] Therefore, while this activity exists and may be relevant in situations of overdose or in investigational uses like oncology, the immunomodulatory effects in RA are primarily attributed to DHODH inhibition.[6]
  • Direct Anti-inflammatory Effects: Beyond its antiproliferative action, teriflunomide exerts more direct anti-inflammatory effects. It can suppress the activation of Nuclear Factor-kappa B (NF-κB), a central transcription factor that orchestrates the expression of numerous pro-inflammatory genes.[6] Furthermore, it has been shown to selectively suppress the production of key inflammatory cytokines, such as tumor necrosis factor-alpha (TNF-α) and interleukin-1 (IL-1), which are major drivers of synovial inflammation in RA.[2]
  • Other Immunomodulatory Effects: The depletion of pyrimidines may have further downstream consequences. Evidence suggests that it can interfere with the glycosylation of cell adhesion molecules on the surface of lymphocytes. This could reduce the ability of these cells to adhere to and interact with synovial cells in the joint, thereby disrupting a key process in the perpetuation of inflammation.[2] Additionally, some studies suggest it may promote the synthesis of the immunosuppressive cytokine Transforming Growth Factor-beta (TGF-β).[3]
  • Antiviral Activity: An increasingly recognized ancillary property of teriflunomide is its broad-spectrum antiviral activity. It has demonstrated effects against several viruses, including Cytomegalovirus (CMV), Herpes Simplex Virus 1 (HSV1), and BK polyomavirus.[4] This action is believed to stem from the same core mechanism: by inhibiting pyrimidine synthesis, it interferes with the ability of the virus to replicate its own genetic material and assemble new virions.[4] This property is of particular clinical interest for leflunomide's off-label use in transplant recipients, who are highly susceptible to such viral infections.[20]

4.0 Pharmacokinetics: A Profile Defined by a Long Half-Life

The clinical application, dosing strategy, and safety management of leflunomide are inextricably linked to its distinctive pharmacokinetic properties. The drug's behavior in the body—its absorption, distribution, metabolism, and particularly its excretion—is dominated by the characteristics of its active metabolite, teriflunomide (M1), and its exceptionally long elimination half-life.

4.1 Absorption, Distribution, Metabolism, and Excretion (ADME)

  • Absorption: Following oral administration, leflunomide is well-absorbed, with an estimated bioavailability of 80% to 100%.[4] As a prodrug, it is rapidly and almost completely converted to its active metabolite, M1, during first-pass metabolism in the gut wall and liver.[1] Peak plasma concentrations ( Tmax​) of M1 are typically reached between 6 and 12 hours after dosing.[1] The absorption of leflunomide is not significantly affected by the presence of food, which provides flexibility in its administration schedule.[18]
  • Distribution: The active metabolite M1 is highly bound to plasma proteins, specifically albumin, with a binding percentage greater than 99.3%.[1] This extensive protein binding is a key factor contributing to its low volume of distribution ( Vd​) of approximately 0.13 L/kg (or 11 L) and its long retention time in the body.[1] This indicates that the drug primarily remains within the vascular and tissue compartments rather than distributing extensively into adipose tissue.
  • Metabolism: As previously detailed, leflunomide's primary metabolic fate is its conversion to teriflunomide (M1).[1] M1 itself is further metabolized to several minor metabolites. The only one that is typically quantifiable in plasma is 4-trifluoromethylaniline (TFMA), though it is present at low levels.[1] While the initial conversion is not attributed to a single enzyme, further biotransformation of M1 involves the cytochrome P450 system, with enzymes such as CYP1A2, CYP2C19, and CYP3A4 having been implicated.[18]
  • Excretion: The elimination of M1 from the body is a slow and balanced process. Over a given period, approximately 43% of a dose is eliminated via the kidneys in the urine (primarily as leflunomide glucuronides and an oxanilic acid derivative of M1), and about 48% is eliminated in the feces (primarily as unchanged M1).[1] The fecal excretion route is of paramount importance because M1 undergoes extensive enterohepatic circulation. In this process, M1 excreted into the bile is reabsorbed from the intestine back into the bloodstream. This continuous recycling is the primary mechanism responsible for the drug's exceptionally long elimination half-life.[18]

4.2 The Clinical Significance of a Prolonged Half-Life and Enterohepatic Circulation

The pharmacokinetic profile of leflunomide is a double-edged sword, offering convenience but demanding careful management of its associated risks.

  • Elimination Half-Life: The elimination half-life (t1/2​) of the active metabolite M1 is remarkably long, typically averaging 14 to 18 days, or approximately two weeks.[1] This prolonged presence ensures stable, sustained therapeutic concentrations with convenient once-daily dosing, which is advantageous for patient adherence.[18]
  • Loading Dose Requirement: A direct consequence of this long half-life is the significant time required to reach steady-state plasma concentrations—estimated to be nearly two months with standard maintenance dosing alone.[1] To overcome this therapeutic lag, clinical practice guidelines recommend initiating therapy with a loading dose of 100 mg once daily for three days, followed by the daily maintenance dose. This strategy allows for the rapid attainment of therapeutic drug levels.[1]
  • Accelerated Elimination ("Washout") Procedure: The drug's persistence in the body becomes a critical liability in situations requiring its rapid removal. These include cases of severe adverse drug reactions (e.g., hepatotoxicity, interstitial lung disease), overdose, or in a woman who wishes to become pregnant.[12] In these scenarios, simply discontinuing the drug is insufficient, as toxic or teratogenic concentrations would persist for weeks or months. Without intervention, it can take up to two years for plasma M1 concentrations to fall below the threshold considered safe for pregnancy (0.02 mg/L).[25] To address this, a mandatory accelerated drug elimination procedure must be employed. This procedure directly targets the enterohepatic circulation. By administering a binding agent—either cholestyramine (8 g taken three times daily) or activated charcoal—for 11 days, the M1 metabolite is trapped within the gastrointestinal tract, preventing its reabsorption.[11] This interruption of the recycling pathway dramatically enhances elimination, reducing the effective half-life of M1 to approximately 24 hours.[19] This "washout" procedure is not merely an option but an indispensable safety tool that is integral to the overall therapeutic and risk management strategy for leflunomide.

A summary of these clinically crucial pharmacokinetic parameters is provided in Table 2.

Table 2: Key Pharmacokinetic Parameters of Leflunomide's Active Metabolite (A77 1726)

ParameterValueClinical ImplicationSource(s)
Bioavailability80% - 100%High and consistent absorption after oral administration.4
Time to Peak (Tmax​)6 - 12 hoursOnset of action is not immediate; peak levels reached several hours post-dose.1
Protein Binding>99.3% (to albumin)Contributes to long half-life and low volume of distribution; potential for displacement interactions.1
Volume of Distribution (Vd​)~0.13 L/kg (~11 L)Drug is primarily confined to plasma and tissues, not widely distributed into fat.1
Elimination Half-Life (t1/2​)~14 - 18 daysAllows for convenient once-daily dosing but creates risk of drug accumulation and persistent toxicity.1
MetabolismProdrug converted to active metabolite (teriflunomide); further metabolism involves CYPs.Activation occurs in GI wall/liver; potential for CYP-mediated drug interactions.1
Excretion43% renal, 48% fecal; extensive enterohepatic circulation.Enterohepatic circulation is the primary cause of the long half-life and the target of the washout procedure.1

5.0 Clinical Efficacy in Approved and Investigational Indications

Leflunomide has a well-established evidence base for its approved indications in rheumatology and a growing body of research supporting its use in several off-label and investigational settings. Its efficacy is rooted in its ability to modulate the underlying autoimmune processes of these diseases.

5.1 Rheumatoid Arthritis (RA)

Leflunomide is a cornerstone therapy for RA, with a formal indication for the management of active disease in adults. The goals of therapy are to reduce the signs and symptoms of inflammation, inhibit the progression of structural joint damage as seen on radiographs, and improve overall physical function.[1]

The efficacy of leflunomide in RA is supported by a robust portfolio of Phase II, III, and IV clinical trials.[1] These studies have consistently demonstrated its superiority over placebo across all major outcome measures. When compared to placebo, leflunomide therapy leads to statistically significant and clinically meaningful improvements in the American College of Rheumatology (ACR) response criteria (ACR20, ACR50, and ACR70). This includes marked reductions in the number of tender and swollen joints, patient-reported pain, and disability as measured by the Health Assessment Questionnaire (HAQ).[8] It also normalizes inflammatory biomarkers such as the Erythrocyte Sedimentation Rate (ESR) and C-Reactive Protein (CRP).[8]

A key feature of a true DMARD is its ability to alter the natural course of the disease. In this regard, leflunomide has been shown to be significantly more effective than placebo in retarding the radiographic progression of RA, meaning it helps to slow or prevent the erosions and joint space narrowing that lead to permanent disability.[3] The clinical response to leflunomide is typically observed within the first month of treatment, with maximal therapeutic benefit achieved within three to six months.[8]

Numerous post-marketing (Phase IV) trials have further defined its role in contemporary RA management. These studies have investigated leflunomide as part of various combination strategies, including with other conventional DMARDs like methotrexate, prednisone, and sulfasalazine, as well as with biologic agents such as etanercept, sarilumab, and tocilizumab, reflecting its integration into modern, aggressive treatment paradigms.[30]

5.2 Psoriatic Arthritis (PsA)

Leflunomide is also approved for the treatment of active psoriatic arthritis in adults, a condition characterized by both joint inflammation and skin manifestations.[4] Its efficacy in this dual-pathology disease was established in key clinical trials, most notably the TOPAS (Treatment of Psoriatic Arthritis) study.[33]

The TOPAS study was a 24-week, randomized, double-blind, placebo-controlled trial that demonstrated significant benefits of leflunomide. The primary endpoint, the Psoriatic Arthritis Response Criteria (PsARC) response rate, was achieved by significantly more patients in the leflunomide group than in the placebo group (58.9% vs. 29.7%, respectively).[33]

Importantly, leflunomide showed efficacy against both the articular and dermatological components of PsA. Beyond joint improvement, patients treated with leflunomide experienced significant improvements in their skin disease, as measured by the Psoriasis Area and Severity Index (PASI) and target lesion response.[33] This translated into better quality of life, evidenced by significant improvements in the Dermatology Life Quality Index (DLQI).[33] Patient-reported outcomes from clinical use confirm its effectiveness for PsA, although common side effects such as gastrointestinal issues and hair loss are frequently noted.[34]

5.3 Off-Label and Investigational Uses

The unique immunomodulatory and ancillary properties of leflunomide have prompted its investigation in a range of other diseases.

5.3.1 Solid Organ Transplantation

Leflunomide holds an FDA orphan drug designation for the prevention of acute and chronic rejection in solid organ transplant recipients.[3] Its appeal in this setting is twofold. First, it provides potent immunosuppression, which is necessary to prevent the recipient's immune system from attacking the transplanted organ. Retrospective clinical studies have shown its substantial immunosuppressive capacity in both liver and kidney transplant recipients, often allowing for the dose reduction or even complete cessation of other mainline immunosuppressants like calcineurin inhibitors (e.g., cyclosporine, tacrolimus) and prednisone, thereby sparing patients from the significant long-term toxicities of those agents.[36]

Second, and of unique value in this population, are its antiviral properties. Transplant patients are at high risk for opportunistic viral infections, particularly with BK polyomavirus (which can cause transplant nephropathy) and Cytomegalovirus (CMV).[20] Leflunomide's ability to inhibit the replication of these viruses provides a "dual benefit" that is highly attractive in this complex clinical scenario.[4] Despite these advantages, concerns about its own toxicity profile (hepatotoxicity, anemia) mean it is not recommended as a first-line agent but rather as a valuable tool for salvage therapy or in specific high-risk situations, ideally with therapeutic drug monitoring.[20]

5.3.2 Systemic Lupus Erythematosus (SLE) and Lupus Nephritis (LN)

Leflunomide has been investigated as a therapeutic option for SLE, with a particular focus on lupus nephritis, one of the most severe manifestations of the disease.[9] It is explored as an alternative to standard-of-care immunosuppressants like cyclophosphamide and azathioprine, especially in patients who are refractory to or intolerant of those therapies.

The evidence for its use in LN is encouraging. A landmark prospective, randomized, multicenter trial demonstrated that leflunomide was non-inferior to azathioprine for the maintenance therapy of LN over a 36-month period.[10] The study found no statistically significant difference in the time to kidney flare or in the rates of adverse events between the two groups.[10] Furthermore, a meta-analysis of available studies suggested that the efficacy and safety of leflunomide for induction therapy in LN were comparable to that of cyclophosphamide.[39] Open-label studies have also reported high response rates (up to 76%) in patients with difficult-to-treat LN.[40] These findings support leflunomide as a potential new candidate for LN treatment, and further clinical trials are ongoing.[38]

5.3.3 Other Investigational Areas

  • Oncology: The fundamental antiproliferative mechanism of leflunomide—inducing pyrimidine starvation—has made it a candidate for cancer therapy. By targeting the metabolic needs of rapidly dividing cancer cells, it has shown promise in preclinical models of various malignancies, including prostate cancer, breast cancer, multiple myeloma, and neuroblastoma.[7] A completed Phase 2 clinical trial has also investigated its use in patients with relapsed anaplastic astrocytoma, a type of brain tumor.[42]
  • Uveitis: A completed Phase 2 trial has evaluated the use of leflunomide for uveitis, an inflammatory condition of the eye, suggesting interest in its application for localized autoimmune processes.[43]
  • Viral Infections: Reflecting the interest in its antiviral properties, a Phase 2 trial was conducted to evaluate its role in preventing BK viruria in renal transplant patients. Although this specific trial was terminated, it underscores the ongoing investigation into this aspect of the drug's activity.[44]

A summary of key efficacy trials is presented in Table 3.

Table 3: Summary of Key Efficacy Trials for Leflunomide in Rheumatic Diseases

Trial Identifier/NameIndicationPhaseComparator(s)DurationPrimary Endpoint(s)Key ResultsSource(s)
US301 / MN301 / MN302Rheumatoid ArthritisIIIPlacebo, Methotrexate, Sulfasalazine6-24 monthsACR20 Response, Radiographic ProgressionSuperior to placebo in ACR response and slowing joint damage. Efficacy comparable to methotrexate and sulfasalazine.1
TOPAS StudyPsoriatic ArthritisIIIPlacebo24 weeksPsARC ResponseSignificantly higher PsARC response vs. placebo (58.9% vs 29.7%). Significant improvement in skin (PASI) and quality of life (DLQI).33
NCT00563849Rheumatoid ArthritisIVMethotrexate (in combination)N/AN/AInvestigated combination therapy of Leflunomide + Methotrexate.30
NCT01172002Lupus NephritisIIIAzathioprine36 monthsTime to kidney flareNon-inferior to azathioprine for maintenance therapy; similar flare rates and safety profile.10

6.0 Safety, Tolerability, and Risk Management

The therapeutic benefits of leflunomide are balanced by a significant and complex safety profile that requires rigorous risk management. Its use is governed by two major FDA Boxed Warnings and a range of potential adverse events that necessitate careful patient selection, education, and ongoing monitoring.

6.1 FDA Boxed Warning: Hepatotoxicity

One of the most serious risks associated with leflunomide is severe liver injury. The FDA has issued a Boxed Warning to highlight this risk, which was strengthened in 2010 following post-marketing reports of severe hepatotoxicity, including 49 cases of severe liver injury and 14 instances of fatal liver failure between 2002 and 2009.[12]

The mechanism for this toxicity is linked to the drug's metabolism. Leflunomide is activated in the liver, and its active metabolite undergoes extensive enterohepatic circulation, leading to high and prolonged exposure of the hepatobiliary system to the drug and its byproducts.[1] This inherent metabolic pathway logically increases the potential for liver strain and injury.

  • Contraindications and High-Risk Patients: To mitigate this risk, leflunomide is strictly contraindicated in patients with pre-existing liver disease (including acute or chronic hepatitis B or C) and in those with baseline serum alanine aminotransferase (ALT) levels greater than two times the upper limit of normal (>2x ULN).[1] The risk is significantly potentiated in patients who are concurrently taking other hepatotoxic drugs, most notably methotrexate, or who consume excessive alcohol.[12]
  • Mandatory Monitoring: Stringent liver function monitoring is required for all patients. ALT levels must be checked at baseline, then at least monthly for the first six months of therapy. After this initial period, monitoring can be extended to every 6-8 weeks for the duration of treatment.[1]
  • Management of Elevated Enzymes: If a patient's ALT level rises to greater than three times the ULN, leflunomide therapy must be discontinued immediately. Concurrently, the accelerated drug elimination (washout) procedure with cholestyramine or activated charcoal must be initiated to rapidly clear the drug from the system. Liver tests must then be monitored weekly until they have normalized.[12]

6.2 FDA Boxed Warning: Embryo-Fetal Toxicity

Leflunomide is a potent teratogen and is absolutely contraindicated for use during pregnancy. This risk is codified in a second Boxed Warning for embryo-fetal toxicity.[1]

The teratogenic risk is a direct and predictable consequence of the drug's primary mechanism of action. By inhibiting pyrimidine synthesis, leflunomide blocks the production of DNA and RNA, processes that are fundamental to the rapid cell division and differentiation that characterize fetal development.[3] Exposing a developing embryo or fetus to such an agent can lead to severe and catastrophic birth defects. Animal reproduction studies confirmed that leflunomide is both teratogenic and embryolethal at exposures lower than those seen in humans receiving therapeutic doses.[1]

  • Clinical Management and Contraception:
  • Pregnancy must be definitively excluded via testing before initiating leflunomide therapy.[13]
  • Females of reproductive potential must be counseled on the severe risk to a fetus and must use a reliable form of contraception throughout treatment.[13]
  • This requirement extends beyond the cessation of the drug due to its long half-life. After stopping leflunomide, a woman must undergo the full accelerated elimination procedure and have her plasma teriflunomide levels verified to be below the safe threshold of 0.02 mg/L on two separate occasions at least 14 days apart before attempting to conceive.[25]
  • Men who are taking leflunomide and wish to father a child should also discuss discontinuing the drug and undergoing a washout procedure with their physician, as the potential risk to a fetus via semen exposure cannot be definitively ruled out.[49]
  • If a patient becomes pregnant while taking leflunomide, the drug must be stopped immediately, and the washout procedure must be administered to minimize fetal exposure as quickly as possible.[13]

6.3 Common and Serious Adverse Events

Beyond the risks highlighted in the Boxed Warnings, leflunomide is associated with a spectrum of other adverse effects.

  • Common Adverse Events (reported in ≥10% of patients): The most frequently encountered side effects are gastrointestinal and dermatological. These include diarrhea (which can be significant, occurring in up to 27% of patients), nausea, headache, abnormal liver function tests, hair loss (alopecia), and skin rash.[28] Hypertension is also a common adverse event that requires monitoring.[29]
  • Serious Adverse Events:
  • Immunosuppression and Infection: As an immunomodulatory agent, leflunomide increases the risk of serious infections, including sepsis and opportunistic infections such as Pneumocystis jiroveci pneumonia and tuberculosis, which can be fatal.[1] It is not recommended for patients with severe immunodeficiency or active, uncontrolled infections.[1]
  • Hematologic Toxicity: Although rare, serious bone marrow suppression—including pancytopenia, agranulocytosis, and thrombocytopenia—has been reported. The risk is highest in patients receiving concomitant myelosuppressive therapy, such as methotrexate.[1]
  • Respiratory Toxicity: Interstitial lung disease (ILD), including interstitial pneumonitis and pulmonary fibrosis, is a rare but potentially fatal adverse reaction. It can occur acutely at any time during therapy. Any new or worsening pulmonary symptoms, such as a dry cough or shortness of breath, warrant immediate investigation and discontinuation of the drug.[1]
  • Dermatologic Reactions: In rare instances, leflunomide has been associated with severe, life-threatening skin reactions, including Stevens-Johnson Syndrome (SJS), Toxic Epidermal Necrolysis (TEN), and Drug Reaction with Eosinophilia and Systemic Symptoms (DRESS). If such a reaction is suspected, the drug must be stopped immediately and a washout procedure initiated.[22]
  • Peripheral Neuropathy: The development of peripheral neuropathy, with symptoms of numbness, tingling, or burning pain in the hands or feet, is a known adverse effect.[29]

6.4 Patient Monitoring and Risk Mitigation Strategies

The safe use of leflunomide hinges on a proactive risk mitigation strategy.

  • Baseline Evaluation: Before starting therapy, a comprehensive evaluation is mandatory. This includes:
  • A negative pregnancy test for females of reproductive potential.[25]
  • A baseline liver function panel, including ALT, and screening for hepatitis B and C.[1]
  • A complete blood count (CBC) with differential and platelet count.[1]
  • Measurement of baseline blood pressure.[50]
  • Screening for latent tuberculosis (TB).[55]
  • Ongoing Monitoring during Treatment:
  • ALT: Monthly for the first 6 months, then every 6-8 weeks thereafter.[12]
  • CBC: Monthly for the first 6 months, then every 8 weeks thereafter.[1]
  • Blood Pressure: Checked regularly at clinic visits.[49]
  • Patient Counseling: Thorough patient education is critical. Patients must be counseled on the absolute need for reliable contraception, the signs and symptoms of liver injury (jaundice, dark urine, abdominal pain), infection (fever, chills, sore throat), and other serious reactions, and instructed to report them immediately.[12] They should also be advised to avoid excessive alcohol consumption and to not receive live vaccines while on therapy.[28]

A detailed summary of adverse effects is provided in Table 4.

Table 4: Comprehensive Adverse Effects Profile of Leflunomide

System Organ ClassAdverse EventFrequencyClinical Considerations/ManagementSource(s)
HepatobiliarySevere Liver Injury / Fatal Liver FailureRareFDA Boxed Warning. Contraindicated in liver disease. Monitor ALT monthly for 6 months, then every 6-8 weeks. Discontinue and perform washout if ALT >3x ULN.12
Abnormal Liver EnzymesCommon (≥10%)Monitor ALT as per guidelines. Dose reduction may be considered for mild elevations.29
ReproductiveEmbryo-Fetal Toxicity / TeratogenicityN/A (Risk)FDA Boxed Warning. Contraindicated in pregnancy. Mandates effective contraception and washout procedure before conception.13
GastrointestinalDiarrheaVery Common (>10%)Often dose-limiting but may improve over time. Antidiarrheal medication or dose reduction may help.28
Nausea, Abdominal PainCommon (1-10%)Can be managed symptomatically.29
Immune SystemSerious Infections (incl. Sepsis, Opportunistic)UncommonIncreased susceptibility to infection. Avoid in severe immunodeficiency. Patient must report signs of infection immediately.22
HematologicPancytopenia, AgranulocytosisRareSerious. Risk increased with concomitant myelosuppressive drugs. Monitor CBC monthly for 6 months, then every 8 weeks.25
RespiratoryInterstitial Lung Disease (ILD)RarePotentially fatal. Discontinue immediately and investigate if new/worsening cough or dyspnea occurs. Consider washout.28
DermatologicHair Loss (Alopecia)Common (≥10%)Generally mild to moderate, may be distressing to patient.28
RashCommon (≥10%)Usually mild.29
SJS / TEN / DRESSVery RareLife-threatening. Discontinue immediately and perform washout if suspected.22
Nervous SystemHeadacheCommon (≥10%)Generally manageable.29
Peripheral NeuropathyUncommonMay manifest as numbness, tingling, or pain. Requires evaluation and possible discontinuation.29
CardiovascularHypertensionCommon (1-10%)Monitor blood pressure regularly during treatment.29

7.0 Clinically Significant Drug Interactions

The clinical use of leflunomide requires careful consideration of potential drug-drug interactions. These interactions can be pharmacodynamic, leading to additive toxicity, or pharmacokinetic, altering the concentration of leflunomide or co-administered drugs.

7.1 Pharmacodynamic Interactions (Additive Toxicity)

These interactions occur when two drugs with similar toxic effects are used together, resulting in an amplified risk.

  • Hepatotoxic Agents: This is one of the most critical areas of concern. Co-administration of leflunomide with other drugs known to cause liver injury significantly potentiates the risk of severe hepatotoxicity, as highlighted in the FDA's Boxed Warning.[1]
  • Methotrexate: The most common and clinically significant interaction in this class. While the combination is used therapeutically, it mandates heightened vigilance and more frequent liver function monitoring due to the increased risk of hepatotoxicity.[1] This creates a therapeutic paradox where a logical combination for efficacy is also a high-risk scenario for safety.
  • Alcohol: Patients must be counseled to avoid or strictly limit alcohol consumption to minimize the cumulative strain on the liver.[28]
  • Other Hepatotoxins: A wide range of other drugs, including the retinoid acitretin, the anti-tuberculosis agent isoniazid, and certain antifungal agents, can increase the risk of liver damage when combined with leflunomide.[3]
  • Immunosuppressive and Myelosuppressive Agents: Combining leflunomide with other drugs that suppress the immune system or bone marrow function increases the risk of serious and opportunistic infections, as well as hematologic toxicities like pancytopenia.[1] This is relevant for combinations with:
  • Biologic DMARDs: Agents like adalimumab, etanercept, and abatacept.[3]
  • Other Conventional DMARDs: Azathioprine, cyclosporine.[3]
  • Antineoplastic Agents: Many chemotherapy drugs are myelosuppressive.[58]

7.2 Pharmacokinetic Interactions

These interactions involve one drug affecting the absorption, distribution, metabolism, or excretion of another.

  • Interactions Involving the Cytochrome P450 (CYP) Enzyme System:
  • Inhibition of CYP2C9 by Leflunomide: The active metabolite of leflunomide, teriflunomide, is a known inhibitor of the enzyme CYP2C9.[1] This is a critical interaction because CYP2C9 is responsible for the metabolism of several commonly used drugs with narrow therapeutic indices. This inhibition can lead to increased plasma concentrations and an exaggerated effect or toxicity of these co-administered drugs. A prime example is warfarin, where CYP2C9 inhibition by leflunomide can lead to a dangerously elevated International Normalized Ratio (INR) and a high risk of bleeding.[1] Other CYP2C9 substrates like phenytoin and certain NSAIDs (e.g., diclofenac, ibuprofen) may also be affected.[1] This interaction underscores the necessity of a thorough medication review before initiating leflunomide.
  • Induction of Leflunomide Metabolism: Strong inducers of CYP enzymes, such as rifampin, can accelerate the metabolism of leflunomide. This can lead to higher peak concentrations of the active teriflunomide metabolite, potentially increasing the risk of toxicity. Caution is advised when these drugs are used together.[1]
  • Inhibition of Leflunomide Metabolism: Conversely, drugs that inhibit the CYP enzymes involved in leflunomide's biotransformation (implicated enzymes include CYP1A2, CYP2C19, and CYP3A4) could theoretically increase leflunomide concentrations, though the clinical significance of this is less well-defined.[3] For instance, abametapir, a CYP inhibitor, may increase leflunomide levels.[3]
  • Interactions with Drug Transporters: Leflunomide may interact with drug transporters that are responsible for moving drugs into and out of cells. Co-administration with drugs that inhibit these transporters, such as afatinib or abemaciclib, could potentially decrease leflunomide's excretion and lead to higher serum levels.[3]
  • Therapeutic Interaction with Binding Agents:
  • Cholestyramine and Activated Charcoal: These agents are not considered toxic interactions but are used therapeutically to enhance leflunomide's elimination. They work by binding teriflunomide in the gut, thereby interrupting its enterohepatic circulation and preventing its reabsorption. This is the mechanistic basis for the accelerated elimination or "washout" procedure.[3]

A summary of major drug interactions is presented in Table 5.

Table 5: Major Drug-Drug Interactions with Leflunomide

Interacting Drug/ClassMechanism of InteractionPotential Clinical EffectManagement Recommendation/MonitoringSource(s)
MethotrexatePharmacodynamic (Additive Toxicity)Potentiated risk of severe hepatotoxicity and myelosuppression.Use with caution. Requires more frequent monitoring of LFTs and CBCs than with either agent alone.22
Other Hepatotoxic Agents (e.g., Alcohol, Acitretin, Isoniazid)Pharmacodynamic (Additive Toxicity)Increased risk of severe liver injury.Avoid or use with extreme caution. Counsel patients to avoid alcohol. Monitor LFTs closely.3
Other Immunosuppressants (e.g., Biologics, Azathioprine)Pharmacodynamic (Additive Toxicity)Increased risk of serious and opportunistic infections, bone marrow suppression.Use with caution. Monitor closely for signs of infection and hematologic toxicity (CBC).3
WarfarinPharmacokinetic (Inhibition of CYP2C9 by teriflunomide)Increased warfarin levels, leading to elevated INR and significant risk of bleeding.Monitor INR very closely upon initiation, dose adjustment, or discontinuation of leflunomide.1
RifampinPharmacokinetic (Induction of leflunomide metabolism)Increased peak levels of the active metabolite teriflunomide, potentially increasing toxicity.Use with caution and monitor for adverse effects.1
CYP2C9 Substrates (e.g., Phenytoin, some NSAIDs)Pharmacokinetic (Inhibition of CYP2C9 by teriflunomide)Increased levels and potential toxicity of the co-administered drug.Monitor for signs of toxicity of the co-administered drug. Dose adjustments may be needed.1
Cholestyramine, Activated CharcoalPharmacokinetic (Interruption of enterohepatic circulation)Dramatically increased clearance of teriflunomide.This is a therapeutic interaction used for the accelerated elimination ("washout") procedure.3

8.0 Dosing, Administration, and Special Populations

The clinical application of leflunomide is guided by specific dosing regimens designed to account for its unique pharmacokinetics, and its use is restricted or requires caution in certain patient populations.

8.1 Dosing for Rheumatoid Arthritis

The dosing strategy for leflunomide in RA is designed to rapidly achieve and then maintain therapeutic plasma concentrations of its active metabolite, teriflunomide.

  • Loading Dose: Standard therapy is initiated with a loading dose of 100 mg once daily for three consecutive days.[1] This initial high-dose regimen is necessary to quickly saturate the body and achieve steady-state plasma concentrations, which would otherwise take nearly two months due to the drug's long half-life.[1] In patients considered to be at a higher risk for adverse events, such as hepatotoxicity or myelosuppression (e.g., those on concomitant methotrexate), clinicians may elect to omit the loading dose and start directly with the maintenance dose. This approach reduces the initial drug exposure and may decrease the risk of acute side effects, though it will delay the onset of full therapeutic benefit.[22]
  • Maintenance Dose: Following the loading dose, the recommended maintenance dose is 20 mg once daily.[25] This is the maximum recommended daily dose.[25]
  • Dose Adjustment: For patients who experience tolerability issues, such as persistent diarrhea or elevated liver enzymes (below the threshold for discontinuation), the maintenance dose can be reduced to 10 mg once daily.[25] Patients should be monitored carefully after any dose reduction.

8.2 Administration

Leflunomide is formulated for oral administration.

  • It is available as tablets in strengths of 10 mg, 20 mg, and 100 mg.[27] The 100 mg tablets are intended for use only during the 3-day loading dose period.
  • The tablets should be swallowed whole with a sufficient amount of liquid and can be taken with or without food, as food does not significantly impact its absorption.[22]

8.3 Special Populations

The use of leflunomide must be carefully considered in specific patient groups due to altered risk-benefit profiles.

  • Hepatic Impairment: Leflunomide is contraindicated in patients with pre-existing severe liver disease or those with baseline ALT levels greater than two times the upper limit of normal.[1] This is due to the high risk of potentiating liver injury and the liver's central role in the drug's metabolic activation and elimination.
  • Renal Impairment: Caution is advised when administering leflunomide to patients with renal insufficiency. While the kidneys are a route of elimination, the primary concern is based on pharmacokinetic studies in dialysis patients which showed that the unbound (free) fraction of the active metabolite was approximately doubled.[1] The clinical significance of this finding is unknown as there is limited clinical experience in this population. Manufacturers do not provide specific dose adjustments for renal impairment.[22]
  • Pediatric Use: The pharmacokinetics of leflunomide have been studied in a limited number of pediatric patients (ages 3 to 17) with polyarticular course Juvenile Rheumatoid Arthritis (JRA).[1] However, it is not a standard or first-line therapy in this population, and its use and dosage must be determined by a specialist physician.[50]
  • Pregnancy and Lactation: Leflunomide is strictly contraindicated in pregnancy due to its demonstrated teratogenicity.[1] It is also contraindicated in women who are breastfeeding, as there is a potential for serious adverse reactions in nursing infants from drug exposure through breast milk.[3]

9.0 Comparative Analysis and Place in Therapy

To understand the clinical role of leflunomide, it is essential to compare it with other cornerstone conventional synthetic DMARDs (csDMARDs), primarily methotrexate and sulfasalazine. This comparative analysis helps to define its specific place within the therapeutic algorithm for rheumatoid arthritis.

9.1 Leflunomide vs. Methotrexate

Methotrexate is widely considered the "anchor" drug in RA therapy, making its comparison with leflunomide the most clinically relevant for positioning.

  • Efficacy: A substantial body of evidence from head-to-head clinical trials and systematic reviews concludes that, overall, leflunomide and methotrexate have comparable efficacy for the treatment of RA.[45] Over treatment periods of one to two years, both drugs demonstrate similar improvements in clinical outcomes (ACR20, ACR50, ACR70 scores) and are equally effective at slowing the rate of radiographic joint damage.[45] While the overall efficacy is similar, some individual studies and meta-analyses have noted a slight, though not always statistically significant, trend favoring methotrexate in specific measures, such as the reduction in swollen joint count.[60]
  • Safety and Tolerability: While the overall rates of adverse events and treatment withdrawals are similar between the two drugs, their specific side effect profiles differ, which often guides therapeutic choice.[45]
  • Leflunomide is more commonly associated with diarrhea and hypertension.[45]
  • Methotrexate is more frequently associated with nausea, vomiting, and stomatitis (mouth sores). However, these gastrointestinal side effects can often be effectively mitigated by co-administration of folic acid.[52]
  • The risk of hepatotoxicity (elevated liver enzymes) is a concern with both medications. The combination of the two agents significantly increases this risk.[57] Some analyses suggest that liver enzyme elevations are more frequent with leflunomide [61], while other trial data (conducted before routine folate supplementation with methotrexate) showed higher withdrawal rates due to liver issues with methotrexate.[60]
  • Hair loss occurs at similar rates with both drugs, with some evidence suggesting a slightly higher incidence with leflunomide.[52]
  • Dosing, Cost, and Patient Preference: Methotrexate offers greater dosing flexibility (oral or subcutaneous administration, with a weekly schedule) and is significantly less expensive than leflunomide.[45] Leflunomide provides the convenience of a simple once-daily oral regimen.[53] In terms of patient-reported outcomes on platforms like Drugs.com, methotrexate tends to receive a slightly higher average satisfaction rating (6.5 out of 10) compared to leflunomide (5.1 out of 10), with a lower percentage of users reporting a negative experience.[62]

This comparative data leads to a clear positioning in clinical practice. Methotrexate, with its long history, dosing flexibility, established role as the anchor for combination therapies, and lower cost, is generally the preferred first-line DMARD for most patients.[57] The American College of Rheumatology conditionally recommends methotrexate over leflunomide for DMARD-naive patients with moderate-to-high disease activity.[57] Leflunomide, with its comparable efficacy but different side effect profile, serves as the primary

first-line alternative. Its therapeutic niche is for patients who cannot tolerate methotrexate (due to severe nausea, for example) or for whom it is contraindicated. It fills a critical therapeutic gap, allowing for continued effective csDMARD therapy in a substantial portion of the RA population.

9.2 Leflunomide vs. Sulfasalazine

  • Efficacy: Leflunomide is generally considered to be more effective than sulfasalazine, particularly with long-term use. While their efficacy may be comparable in the first 6 to 12 months of treatment, by 24 months, studies show that leflunomide leads to significantly greater improvements across most clinical outcome measures, including physician and patient global assessments and functional ability (HAQ score).[45]
  • Safety and Tolerability: The overall safety profiles and rates of discontinuation due to adverse events are comparable between the two drugs.[45] Patient review data shows similar average satisfaction ratings for both medications.[64]
  • Cost: Leflunomide is substantially more expensive than sulfasalazine.[45]

9.3 Role in Combination Therapy

Reflecting the modern paradigm of aggressive RA management, leflunomide is often used as part of a combination regimen for patients who do not achieve adequate control with monotherapy.[65] It can be combined with other csDMARDs, such as methotrexate (with the aforementioned need for heightened safety monitoring), sulfasalazine, and hydroxychloroquine.[30] It is also used in conjunction with biologic agents.[30] The choice of combination often depends on prior treatments, patient tolerance, and specific treatment guidelines. For instance, in patients intolerant to methotrexate, a combination involving leflunomide may be considered as an alternative to standard "triple therapy" (methotrexate + sulfasalazine + hydroxychloroquine).[65]

A summary of the key comparative points between leflunomide and methotrexate is provided in Table 6.

Table 6: Comparative Profile: Leflunomide vs. Methotrexate for Rheumatoid Arthritis

FeatureLeflunomideMethotrexate
Mechanism of ActionInhibition of dihydroorotate dehydrogenase (DHODH), blocking de novo pyrimidine synthesis.Inhibition of dihydrofolate reductase (DHFR), blocking purine synthesis and other folate-dependent pathways.
Dosing RegimenOral, once daily (with initial 3-day loading dose).Oral or subcutaneous, once weekly.
Overall EfficacyComparable to methotrexate in improving symptoms and slowing joint damage.Gold standard; comparable to leflunomide. Often considered the "anchor" DMARD.
Key Side EffectsHigher incidence of diarrhea and hypertension.Higher incidence of nausea/vomiting and stomatitis (mitigated by folic acid).
HepatotoxicityBoxed Warning. Risk present, potentially more frequent enzyme elevations.Risk present, especially without folate. Potentiated when combined with leflunomide.
TeratogenicityBoxed Warning. Absolutely contraindicated. Requires long washout period (up to 2 years naturally).Absolutely contraindicated. Shorter washout period required.
CostSignificantly more expensive.Lower cost generic widely available.
Place in TherapyPrimary alternative to methotrexate; for patients with intolerance or contraindication to methotrexate.First-line anchor drug for most DMARD-naive patients.
Source(s)4545

10.0 Regulatory and Development History

The journey of leflunomide from a novel chemical entity to an established DMARD reflects the evolving landscape of autoimmune disease treatment in the late 20th century.

  • Initial Development: Leflunomide was originally synthesized and investigated as a potential anti-inflammatory agent. However, during early development, its potent immunomodulatory activities became apparent, which redirected its clinical development path toward the treatment of autoimmune diseases like rheumatoid arthritis.[19]
  • U.S. Food and Drug Administration (FDA) Approval:
  • The original formulation, under the brand name Arava and sponsored by Sanofi-Aventis, received its initial approval from the FDA on September 10, 1998.[67] The indication was for the treatment of active rheumatoid arthritis.
  • The approval of leflunomide occurred around the same time as the first biologic TNF inhibitors, marking a period of significant advancement in rheumatology therapeutics.[71]
  • The first generic versions of leflunomide tablets were approved by the FDA starting in September 2005, and numerous manufacturers have since entered the market, increasing accessibility.[69]
  • European Medicines Agency (EMA) Approval:
  • Leflunomide was first approved for use in Europe in 1999, shortly after its US approval.[3]
  • The indication for Arava was expanded by the European Commission in 2004 to include the treatment of active psoriatic arthritis.[72]
  • Generic versions of leflunomide from various manufacturers, such as Leflunomide medac and Leflunomide ratiopharm, began receiving marketing authorization across the European Union around 2010.[73]
  • Key Post-Marketing Label Changes and Safety Updates:
  • The risk of liver injury was recognized early in its post-marketing surveillance. In 2003, a bolded warning regarding the risk of severe liver injury was added to the US drug label.[76]
  • Following a review of accumulating adverse event reports, the FDA took further action in July 2010. The agency mandated that the warning for severe liver injury be elevated to a Boxed Warning, placing it alongside the pre-existing Boxed Warning for embryo-fetal toxicity. This update was intended to highlight the critical importance of appropriate patient selection and stringent liver function monitoring during treatment.[12]

11.0 Conclusion and Future Perspectives

Leflunomide has secured a durable and important role in the management of autoimmune arthritis. It stands as a potent and effective disease-modifying antirheumatic drug for both rheumatoid arthritis and psoriatic arthritis, offering efficacy comparable to the cornerstone therapy, methotrexate. Its distinct mechanism of action—the targeted, cytostatic inhibition of lymphocyte proliferation via the blockade of dihydroorotate dehydrogenase—and the convenience of its once-daily oral dosing regimen make it a valuable therapeutic tool.

However, the clinical utility of leflunomide is fundamentally defined and constrained by its challenging safety profile. Its benefits must be constantly weighed against the significant risks codified in two FDA Boxed Warnings: one for severe, potentially fatal hepatotoxicity, and the other for profound embryo-fetal toxicity. These risks are not arbitrary but are direct consequences of the drug's core pharmacology and unique pharmacokinetics, particularly its extensive hepatic metabolism and exceptionally long half-life. This necessitates a rigorous and non-negotiable risk management strategy, including careful patient selection, comprehensive counseling on contraception, and diligent monitoring of liver and hematologic parameters. The mandatory accelerated elimination procedure is a unique clinical requirement that underscores the complexities of managing this long-acting agent.

In the current therapeutic landscape, leflunomide's primary place in therapy is as a first-line alternative for RA patients who are intolerant of, have contraindications to, or have an inadequate response to methotrexate. It provides a critical option that allows a significant subset of patients to remain on effective conventional DMARD therapy.

Looking forward, the future of leflunomide may lie in the expansion of its off-label and investigational uses. The promising non-inferiority data in maintenance therapy for lupus nephritis suggests it could evolve into a standard-of-care option for this severe condition, pending confirmation from larger, long-term clinical trials. Furthermore, its unique dual profile of providing both immunosuppression and antiviral activity ensures its continued interest as a niche agent in the complex field of solid organ transplantation, especially for managing patients with concomitant viral reactivation. While its potential in oncology remains highly investigational, its fundamental antiproliferative mechanism warrants continued exploration. Ultimately, the story of leflunomide is a powerful clinical lesson in balancing targeted efficacy against the profound management challenges posed by a drug's unique pharmacokinetic properties and its on-target toxicities. Broadening its future application will depend on developing more refined strategies to predict and mitigate its risks, allowing clinicians to better harness its clear benefits for a wider range of patients.

Works cited

  1. Mechanism of Action Pharmacokinetics - accessdata.fda.gov, accessed August 4, 2025, https://www.accessdata.fda.gov/drugsatfda_docs/label/2007/020905s016lbl.pdf
  2. Mechanism of Action for Leflunomide in Rheumatoid Arthritis | Request PDF - ResearchGate, accessed August 4, 2025, https://www.researchgate.net/publication/222903210_Mechanism_of_Action_for_Leflunomide_in_Rheumatoid_Arthritis
  3. Leflunomide: Uses, Interactions, Mechanism of Action | DrugBank ..., accessed August 4, 2025, https://go.drugbank.com/drugs/DB01097
  4. Leflunomide - Wikipedia, accessed August 4, 2025, https://en.wikipedia.org/wiki/Leflunomide
  5. What is the mechanism of Leflunomide? - Patsnap Synapse, accessed August 4, 2025, https://synapse.patsnap.com/article/what-is-the-mechanism-of-leflunomide
  6. Leflunomide: mode of action in the treatment of rheumatoid arthritis, accessed August 4, 2025, https://ard.bmj.com/content/59/11/841.short
  7. Leflunomide | CAS#75706-12-6 - MedKoo Biosciences, accessed August 4, 2025, https://www.medkoo.com/products/8290
  8. Leflunomide: uses, dosing, warnings, adverse events, interactions - MedCentral, accessed August 4, 2025, https://www.medcentral.com/drugs/monograph/16551-300032/leflunomide-oral
  9. Leflunomide an immunomodulator with antineoplastic and antiviral ..., accessed August 4, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC7869628/
  10. Leflunomide versus azathioprine for maintenance therapy of lupus nephritis: a prospective, multicentre, randomised trial and long-term follow-up | Annals of the Rheumatic Diseases, accessed August 4, 2025, https://ard.bmj.com/content/81/11/1549
  11. Clinical pharmacokinetics of leflunomide - PubMed, accessed August 4, 2025, https://pubmed.ncbi.nlm.nih.gov/12074690/
  12. FDA Drug Safety Communication: New boxed warning for severe ..., accessed August 4, 2025, https://www.fda.gov/drugs/postmarket-drug-safety-information-patients-and-providers/fda-drug-safety-communication-new-boxed-warning-severe-liver-injury-arthritis-drug-arava-leflunomide
  13. ARAVA (leflunomide) tablets, for oral use - accessdata.fda.gov, accessed August 4, 2025, https://www.accessdata.fda.gov/drugsatfda_docs/label/2024/020905s033lbl.pdf
  14. Leflunomide | C12H9F3N2O2 | CID 3899 - PubChem, accessed August 4, 2025, https://pubchem.ncbi.nlm.nih.gov/compound/Leflunomide
  15. Leflunomide | CAS 75706-12-6 | SCBT - Santa Cruz Biotechnology, accessed August 4, 2025, https://www.scbt.com/p/leflunomide-75706-12-6
  16. Leflunomide | CAS 75706-12-6 | Immunosupressant - StressMarq Biosciences Inc., accessed August 4, 2025, https://www.stressmarq.com/products/small-molecules/inhibitor/leflunomide-sih-368/
  17. Leflunomide 75706-12-6 | TCI AMERICA - Tokyo Chemical Industry, accessed August 4, 2025, https://www.tcichemicals.com/CA/en/p/L0250
  18. Leflunomide Applicability in Rheumatoid Arthritis: Drug Delivery Challenges and Emerging Formulation Strategies - MDPI, accessed August 4, 2025, https://www.mdpi.com/2813-2998/4/3/36
  19. 20905 Arava Tablets 10mg, 20mg and 100mg Clinical Pharmacology Biopharmaceutics Review - accessdata.fda.gov, accessed August 4, 2025, https://www.accessdata.fda.gov/drugsatfda_docs/nda/98/20905_arava_biopharmr.pdf
  20. Leflunomide in renal transplantation - PubMed, accessed August 4, 2025, https://pubmed.ncbi.nlm.nih.gov/21595593/
  21. Leflunomide: A Comprehensive Guide - Number Analytics, accessed August 4, 2025, https://www.numberanalytics.com/blog/leflunomide-orthopedic-pharmacology-guide
  22. Arava, INN-leflunomide, accessed August 4, 2025, https://ec.europa.eu/health/documents/community-register/2018/20180730141969/anx_141969_en.pdf
  23. Population pharmacokinetics and association between A77 1726 plasma concentrations and disease activity measures following administration of leflunomide to people with rheumatoid arthritis - PMC, accessed August 4, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC1884779/
  24. Inhibition of hepatic cytochrome P450 enzymes and sodium/bile acid cotransporter exacerbates leflunomide-induced hepatotoxicity - PMC, accessed August 4, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC4775851/
  25. highlights of prescribing information - Sanofi U.S., accessed August 4, 2025, https://products.sanofi.us/Leflunomide/Leflunomide.pdf
  26. Leflunomide (Arava): Uses & Side Effects - Cleveland Clinic, accessed August 4, 2025, https://my.clevelandclinic.org/health/drugs/20778-leflunomide-tablets
  27. Arava (leflunomide) dosing, indications, interactions, adverse effects, and more, accessed August 4, 2025, https://reference.medscape.com/drug/arava-leflunomide-343203
  28. Leflunomide (Arava) - American College of Rheumatology, accessed August 4, 2025, https://rheumatology.org/patients/leflunomide-arava
  29. Leflunomide Uses, Side Effects & Warnings - Drugs.com, accessed August 4, 2025, https://www.drugs.com/mtm/leflunomide.html
  30. Leflunomide Completed Phase 4 Trials for Rheumatoid Arthritis Treatment - DrugBank, accessed August 4, 2025, https://go.drugbank.com/drugs/DB01097/clinical_trials?conditions=DBCOND0027961&phase=4&purpose=treatment&status=completed
  31. Leflunomide Completed Phase 3 Trials for Rheumatoid Arthritis Treatment - DrugBank, accessed August 4, 2025, https://go.drugbank.com/drugs/DB01097/clinical_trials?conditions=DBCOND0027961&phase=3&purpose=treatment&status=completed
  32. Leflunomide | Side-effects, uses, time to work - Versus Arthritis, accessed August 4, 2025, https://versusarthritis.org/about-arthritis/treatments/drugs/leflunomide/
  33. Leflunomide improves psoriasis in patients with psoriatic arthritis: an in-depth analysis of data from the TOPAS study - PubMed, accessed August 4, 2025, https://pubmed.ncbi.nlm.nih.gov/16549920/
  34. Leflunomide Reviews & Ratings - Drugs.com, accessed August 4, 2025, https://www.drugs.com/comments/leflunomide/
  35. Leflunomide for Psoriatic Arthritis Reviews - Drugs.com, accessed August 4, 2025, https://www.drugs.com/comments/leflunomide/for-psoriatic-arthritis.html
  36. Experiences with leflunomide in solid organ transplantation - PubMed, accessed August 4, 2025, https://pubmed.ncbi.nlm.nih.gov/11884931/
  37. Leflunomide: Friend or foe for systemic lupus erythematosus? | Request PDF, accessed August 4, 2025, https://www.researchgate.net/publication/230825844_Leflunomide_Friend_or_foe_for_systemic_lupus_erythematosus
  38. Rheumatoid Arthritis Drug Leflunomide Offers Encouraging Results in People with Lupus Nephritis, accessed August 4, 2025, https://www.lupus.org/news/rheumatoid-arthritis-drug-leflunomide-offers-encouraging-results-in-people-with-lupus
  39. SAT0253 Efficacy and safety of leflunomide therapy in lupus nephritis:a meta-analysis, accessed August 4, 2025, https://ard.bmj.com/content/76/Suppl_2/870.1
  40. Safety and efficacy of leflunomide in the treatment of lupus nephritis refractory or intolerant to traditional immunosuppressive therapy: an open label trial, accessed August 4, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC1798050/
  41. Study Details | Leflunomide in Systemic Lupus Erythematosus | ClinicalTrials.gov, accessed August 4, 2025, https://www.clinicaltrials.gov/study/NCT00637819
  42. Leflunomide Completed Phase 2 Trials for Brain and Central Nervous System Tumors Treatment | DrugBank Online, accessed August 4, 2025, https://go.drugbank.com/drugs/DB01097/clinical_trials?conditions=DBCOND0028448&phase=2&purpose=treatment&status=completed
  43. Uveitis Completed Phase 2 Trials for Leflunomide (DB01097) | DrugBank Online, accessed August 4, 2025, https://go.drugbank.com/indications/DBCOND0009937/clinical_trials/DB01097?phase=2&status=completed
  44. Viral sepsis Terminated Phase 2 Trials for Leflunomide (DB01097) | DrugBank Online, accessed August 4, 2025, https://go.drugbank.com/indications/DBCOND0013691/clinical_trials/DB01097?phase=2&status=terminated
  45. Is Leflunomide as Safe and Effective in the Treatment of Rheumatoid ..., accessed August 4, 2025, https://www.aafp.org/pubs/afp/issues/2003/0901/p849.html
  46. Arava (leflunomide): Boxed Warning - Risk of Severe Liver Injury - Moda Health, accessed August 4, 2025, https://www.modahealth.com/pdfs/rx_updates/1007_Arava.pdf
  47. FDA Adds Liver Warning to Leflunomide Label - NEJM Journal Watch, accessed August 4, 2025, https://www.jwatch.org/fw201007140000004/2010/07/14/fda-adds-liver-warning-leflunomide-label
  48. Drug Interaction Report: leflunomide, Norco - Drugs.com, accessed August 4, 2025, https://www.drugs.com/interactions-check.php?drug_list=1440-0,71-8487&professional=1
  49. Leflunomide in rheumatoid arthritis (RA) - NRAS, accessed August 4, 2025, https://nras.org.uk/resource/leflunomide/
  50. Leflunomide (oral route) - Side effects & dosage - Mayo Clinic, accessed August 4, 2025, https://www.mayoclinic.org/drugs-supplements/leflunomide-oral-route/description/drg-20067158
  51. Leflunomide Side Effects: Common, Severe, Long Term - Drugs.com, accessed August 4, 2025, https://www.drugs.com/sfx/leflunomide-side-effects.html
  52. Leflunomide vs. Methotrexate: What to Know About Each - Healthline, accessed August 4, 2025, https://www.healthline.com/health/drugs/leflunomide-vs-methotrexate
  53. Methotrexate vs. Arava for Rheumatoid Arthritis: Important Differences and Potential Risks., accessed August 4, 2025, https://www.goodrx.com/compare/methotrexate-vs-arava
  54. The safety of leflunomide - Australian Prescriber - Therapeutic Guidelines, accessed August 4, 2025, https://australianprescriber.tg.org.au/articles/the-safety-of-leflunomide.html
  55. Leflunomide: MedlinePlus Drug Information, accessed August 4, 2025, https://medlineplus.gov/druginfo/meds/a600032.html
  56. Leflunomide (Arava®) - Information - Johns Hopkins Arthritis Center, accessed August 4, 2025, https://www.hopkinsarthritis.org/patient-corner/drug-information/leflunomide-arava/
  57. What is safer, Arava (Leflunomide) versus Methotrexate (MTX)? - Dr.Oracle AI, accessed August 4, 2025, https://www.droracle.ai/articles/104120/arava-versus-methetextrate-what-is-safer-
  58. Drug Interaction Report: leflunomide, methylprednisolone - Drugs.com, accessed August 4, 2025, https://www.drugs.com/interactions-check.php?drug_list=1440-0,1607-0&professional=1
  59. Leflunomide (Arava): Side Effects, Uses, Dosage, Interactions, Warnings - RxList, accessed August 4, 2025, https://www.rxlist.com/leflunomide_arava/generic-drug.htm
  60. A comparison of the efficacy and safety of leflunomide and methotrexate for the treatment of rheumatoid arthritis - PubMed, accessed August 4, 2025, https://pubmed.ncbi.nlm.nih.gov/10888712/
  61. Systematic review and meta-analysis of the efficacy and safety of leflunomide and methotrexate in the treatment of rheumatoid arthritis - PubMed, accessed August 4, 2025, https://pubmed.ncbi.nlm.nih.gov/28867467/
  62. Leflunomide vs Methotrexate Comparison - Drugs.com, accessed August 4, 2025, https://www.drugs.com/compare/leflunomide-vs-methotrexate
  63. Treatment of active rheumatoid arthritis with leflunomide: two year follow up of a double blind, placebo controlled trial versus sulfasalazine - PubMed, accessed August 4, 2025, https://pubmed.ncbi.nlm.nih.gov/11557646/
  64. Leflunomide vs Sulfasalazine Comparison - Drugs.com, accessed August 4, 2025, https://www.drugs.com/compare/leflunomide-vs-sulfasalazine
  65. Study Details | Clinical Trial Evaluating Methotrexate or Leflunomide + Targeted Therapy Versus Methotrexate or Leflunomide + Sulfasalazine + Hydroxychloroquine in Patients With Rheumatoid Arthritis and Insufficient Response to Methotrexate or Leflunomide | ClinicalTrials.gov, accessed August 4, 2025, https://clinicaltrials.gov/study/NCT02714634?term=NCT00259610%20NCT00422227%20NCT00521924%20NCT00579878%20NCT00764725%20NCT00908089%20NCT01172639%20NCT01659242%20NCT01709578%20NCT01768572%20NCT01941095%20NCT02057250%20NCT02373202%20NCT02374021%20NCT02433184%20NCT02451748%20NCT02466581%20NCT02714634%20NCT02930343%20NCT03414502%20NCT03449758%20NCT03813771&rank=5
  66. Rheumatoid arthritis - Treatment - NHS, accessed August 4, 2025, https://www.nhs.uk/conditions/rheumatoid-arthritis/treatment/
  67. www.accessdata.fda.gov, accessed August 4, 2025, https://www.accessdata.fda.gov/drugsatfda_docs/nda/98/20905_arava.cfm#:~:text=Approval%20Date%3A%209%2F10%2F1998&text=Note%3A%20Documents%20in%20PDF%20format%20require%20the%20Adobe%20Acrobat%20Reader%C2%AE.
  68. Drug Approval Package: Arava (leflunomide tablet) NDA# 20905, accessed August 4, 2025, https://www.accessdata.fda.gov/drugsatfda_docs/nda/98/20905_arava.cfm
  69. Generic Arava Availability - Drugs.com, accessed August 4, 2025, https://www.drugs.com/availability/generic-arava.html
  70. leflunomide - Drug Central, accessed August 4, 2025, https://drugcentral.org/drugcard/1552
  71. 9 Facts about Leflunomide | RheumNow, accessed August 4, 2025, https://rheumnow.com/news/9-facts-about-leflunomide
  72. Aventis Release: European Commission Approval For Arava® (Leflunomide) For Use In Adults With Psoriatic Arthritis - BioSpace, accessed August 4, 2025, https://www.biospace.com/aventis-release-b-european-commission-b-approval-for-arava-leflunomide-for-use-in-adults-with-psoriatic-arthritis
  73. Leflunomide ratiopharm | European Medicines Agency (EMA), accessed August 4, 2025, https://www.ema.europa.eu/en/medicines/human/EPAR/leflunomide-ratiopharm
  74. Human medicines European public assessment report (EPAR): Leflunomide medac, leflunomide, Date of authorisation: 27/07/2010, Revision: 24, Status, accessed August 4, 2025, https://efim.org/node/140199
  75. Leflunomide medac | European Medicines Agency (EMA), accessed August 4, 2025, https://www.ema.europa.eu/en/medicines/human/EPAR/leflunomide-medac
  76. FDA updates Arava label to include sterner liver safety warning - Citeline News & Insights, accessed August 4, 2025, https://insights.citeline.com/SC009525/FDA-updates-Arava-label-to-include-sterner-liver-safety-warning/

Published at: August 4, 2025

This report is continuously updated as new research emerges.

MedPath

Empowering clinical research with data-driven insights and AI-powered tools.

© 2025 MedPath, Inc. All rights reserved.