Cytotoxic Drugs

Comprehensive Guide to Methotrexate Toxicity: Symptoms, Prevention, and Management

Methotrexate (MTX) is vital in treating various cancers and autoimmune diseases, such as rheumatoid arthritis and severe psoriasis. Its unique ability to inhibit dihydrofolate reductase, essential for DNA synthesis, makes it effective. However, it also poses significant toxicity risks, particularly at high doses (HDMTX). Understanding and managing these risks is crucial for optimizing patient outcomes. MTX's chemical structure includes a pteridine ring, a benzene ring, and a glutamic acid derivative, enabling its therapeutic actions. High-dose MTX (exceeding 500 mg/m²) is effective in treating malignancies like acute lymphoblastic leukemia and osteosarcoma but requires meticulous monitoring to avoid severe toxicities. Common drug interactions with MTX include NSAIDs, penicillins, PPIs, sulfonamides, and aspirin, which can elevate MTX levels and increase toxicity. Symptoms of MTX toxicity range from mild drowsiness and gastrointestinal discomfort to severe myelosuppression, hepatotoxicity, renal impairment, and pulmonary issues. Monitoring MTX plasma levels at 24, 48, and 72 hours post-administration is essential for detecting toxic levels and guiding interventions. Management strategies for MTX toxicity involve supportive care, hydration, leucovorin rescue therapy, and, in severe cases, treatments like intravenous fluids, alkaline diuresis, and glucarpidase administration. Preventive measures include folic acid supplementation, careful dosing, and patient education on safe MTX use. Effective MTX therapy requires balancing its powerful therapeutic benefits against potential toxicities through vigilant monitoring, appropriate dosing, and prompt intervention. This comprehensive approach ensures the safer use of this critical medication, enhancing patient care and treatment success.

Chemical Structure of Methotrexate

Chemical structure of methotrexate, a widely used chemotherapy drug and immune system suppressant. Methotrexate features a pteridine ring with amino groups, a benzene ring, and a glutamic acid derivative with carboxyl groups. It is commonly used for treating cancers, rheumatoid arthritis, and other autoimmune diseases
Methotrexate chemical structure

Methotrexate (MTX) has a unique chemical structure that significantly contributes to its therapeutic effects and potential toxicity. Its molecular structure allows it to inhibit dihydrofolate reductase, thereby impeding DNA synthesis and targeting rapidly dividing cells. The therapeutic range for MTX varies depending on the indication. Generally, it falls between 0.01 to 0.1 µM for low-dose therapy (used in rheumatoid arthritis) and 1 to 10 µM for high-dose therapy (used in certain cancers).

Therapeutic Effects of Methotrexate

Methotrexate's efficacy stems from its ability to inhibit dihydrofolate reductase, which is crucial for treating various conditions.

  • Methotrexate in Oncology: High-dose methotrexate (HDMTX) regimens, defined as doses exceeding 500 mg/m², are pivotal in treating malignancies such as acute lymphoblastic leukemia (ALL), osteosarcoma, and certain types of lymphoma. HDMTX significantly improves outcomes but brings about severe toxicities that necessitate meticulous monitoring and intervention (Howard et al., 2016).

  • Methotrexate for Rheumatology: It is widely used in rheumatology because it inhibits dihydrofolate reductase, thereby impeding DNA synthesis and targeting rapidly dividing cells.

  • Methotrexate for Dermatology: Effective in treating severe psoriasis and other skin conditions.

Drug Interactions with Methotrexate

 Various factors, including interactions with other drugs and substances such as NSAIDs and antibiotics, can influence methotrexate's metabolism. These interactions can lead to significant variations in methotrexate serum levels, potentially resulting in either subtherapeutic effects or increased toxicity (Chan & Rajakumar, 2014).

Common Drugs That Interact with Methotrexate: Risks and Management

  • NSAIDs: Increase methotrexate levels, leading to toxicity.

  • Penicillins: Decrease renal clearance of methotrexate, increasing toxicity.

  • PPIs: Delay the renal clearance of methotrexate.

  • Sulfonamides: Increase the risk of MTX toxicity.

  • Trimethoprim/Sulfamethoxazole: Increase the risk of bone marrow suppression.

  • Aspirin: Increase methotrexate levels, leading to toxicity.

  • Cyclosporine: Increase the toxicity of methotrexate.

  • Phenytoin: Increase methotrexate levels, leading to toxicity.

Epidemiology of Methotrexate Toxicity

Prevalence and health risks in different populations, especially those undergoing high-dose methotrexate therapy, show significant health risks associated with its use, particularly in oncology settings where HDMTX is common (Howard et al., 2016).

Case Studies of Methotrexate Toxicity

  • Pediatric Exposures to Methotrexate: Clinical effects such as mucocutaneous toxicity, bone marrow suppression, and acute kidney injury (Esteve et al., 2006).

  • Methotrexate in Adolescents and Adults: Symptoms include mucositis, bone marrow suppression, and neurotoxicity (Rask et al., 1998).

  • Methotrexate Toxicity in General Population: Adverse effects include nephrotoxicity, mucocutaneous toxicity, and bone marrow suppression (Abelson et al., 1983).

Pharmacokinetics of Methotrexate

  • Absorption of Methotrexate: Absorption is inversely related to dose. Oral doses below 30 mg/m² are completely absorbed, but absorption decreases with higher doses.

  • Peak Concentration of Methotrexate: Methotrexate reaches peak plasma concentrations post-ingestion.

  • Methotrexate Metabolism and Half-Life: Undergoes hepatic metabolism primarily via cytochrome P450 enzymes, leading to variability in plasma levels.

  • Methotrexate Excretion: Primarily excreted in the urine and feces.

  • Methotrexate Bioavailability: Methotrexate has moderate protein binding (50-70%) and a low volume of distribution (0.6-0.9 L/kg), which influences its serum levels and toxicity profile (Abelson et al., 1983).

Mechanisms of Methotrexate Toxicity

Mechanisms of Methotrexate Toxicity diagram illustrating the pathways of pulmonary toxicity, gastrointestinal toxicity, folate depletion, oxidative stress and apoptosis, hypersensitivity reactions in the lungs, genetic variability, and renal toxicity caused by methotrexate.
Mechanisms of Methotrexate Toxicity

Understanding the molecular mechanisms underlying MTX toxicity is crucial for optimizing its therapeutic use while minimizing adverse effects.

Key Mechanisms of Methotrexate Toxicity:

1.     Folate Depletion Due to Methotrexate:

o   MTX inhibits dihydrofolate reductase, reducing folate levels and subsequent impairment in DNA synthesis. Accumulation of methotrexate polyglutamate in tissues further exacerbates folate depletion, contributing to toxicity, particularly in rapidly dividing cells such as bone marrow and gastrointestinal mucosa (Kamen et al., 1981).

2.     Methotrexate -Induced Oxidative Stress and Apoptosis:

o   MTX-induced oxidative stress results from producing reactive oxygen species (ROS), leading to cellular damage and apoptosis. This mechanism is particularly significant in hepatotoxicity and nephrotoxicity (Almalki et al., 2021).

3.     Immune-Mediated Mechanisms of Methotrexate toxicity:

o   MTX can trigger immune-mediated toxicities, including hypersensitivity reactions in the lungs, resulting in pulmonary toxicity. These reactions can manifest as allergic or cytotoxic responses involving immune cell activation and inflammation (Kim et al., 2009).

4.     Genetic Variability and Methotrexate toxicity:

o   Genetic polymorphisms such as the C677T mutation in the methylenetetrahydrofolate reductase (MTHFR) gene affect MTX metabolism and toxicity. Individuals with the TT genotype exhibit higher toxicity due to reduced enzyme activity, leading to increased MTX plasma levels and prolonged exposure (Ulrich et al., 2001).

5.     Renal Toxicity of Methotrexate:

o   MTX-induced renal impairment involves both functional and morphological changes in the kidneys, often due to the direct toxic effects of the drug. These effects include necrosis of the renal tubular epithelium and decreased clearance of methotrexate, exacerbating systemic toxicity( Garcia et al., 1969).

6.     Gastrointestinal Toxicity of Methotrexate:

o   MTX can cause significant gastrointestinal toxicity, including mucositis and intestinal damage. These adverse effects are partly due to their impact on rapidly dividing cells in the gastrointestinal tract and alterations in gut microbiota. Leucovorin rescue therapy can mitigate these effects by restoring folate levels and modulating gut microbiota composition. (Huang et al., 2020).

7.     Pulmonary Toxicity of Methotrexate:

o   Pulmonary toxicity associated with MTX can present as interstitial pneumonitis, pulmonary fibrosis, or hypersensitivity pneumonitis. These conditions result from direct cytotoxic effects, immune reactions, and inflammation in the lung tissue (Lateef et al., 2005).

The toxicity of methotrexate is multifaceted, involving folate depletion, oxidative stress, immune responses, genetic factors, renal impairment, and gastrointestinal and pulmonary damage. Understanding these mechanisms is essential for developing strategies to prevent and manage MTX-induced toxicities, ensuring this critical therapeutic agent's safer and more effective use.

Toxic Doses of Methotrexate

  • Toxic Plasma Levels: Therapeutic plasma levels range significantly, and toxic effects are more likely when plasma levels exceed certain thresholds. Methotrexate toxicity is indicated by serum levels exceeding specific thresholds post-administration: 10 µM at 24 hours, 1 µM at 48 hours, and 0.1 µM at 72 hours. Exceeding these levels necessitates immediate medical intervention to prevent severe adverse effects such as myelosuppression and organ toxicity. Clinical studies support these thresholds, highlighting the importance of timely monitoring and intervention to mitigate toxicity and ensure patient safety.

  • Toxicity is indicated by methotrexate levels exceeding 10 µM at 24 hours, 1 µM at 48 hours, and 0.1 µM at 72 hours post-administration. These thresholds necessitate immediate medical intervention to prevent severe adverse effects.

  • Pediatrics: Significant toxicity symptoms at doses as low as 50 mg.

  • Adolescents and Adults: Significant toxicity at doses above 100 mg, with severe toxicity often occurring at doses above 1000 mg (Rask et al., 1998).

Determining Methotrexate Toxicity: Steps, Formulas, and Thresholds

Step-by-Step Guide for Calculating Methotrexate Toxicity

Step 1: Calculate the Body Surface Area (BSA) for Methotrexate

BSA (m^2)=√[(height (cm) x weight (kg)) / 3600]

Step 2: Calculate the Dose per BSA

                                              Total Methotrexate Dose (mg) / BSA (m^2)

Step 3: Identify Methotrexate Toxicity Thresholds

High-Dose Methotrexate (HDMTX) Protocols:

  • Doses higher than 500 mg/m² are associated with significant toxicity risks.

  • Plasma methotrexate concentrations above 10^-7 M at 48 hours post-administration are linked to high toxicity.

Step 4: Monitor Methotrexate Plasma Levels

Key Time Points:

  • Measure methotrexate levels at 24, 48, and 72 hours post-administration.

  • Levels above 10^-7 M (0.1 μmol/L) at 48 hours are considered toxic.

Step 5: Evaluate Clinical Symptoms

Symptoms of Methotrexate Toxicity

  • Myelosuppression: Pancytopenia, leukopenia, thrombocytopenia

  • Gastrointestinal: Mucositis, diarrhea, stomatitis

  • Hepatotoxicity: Elevated liver enzymes

  • Renal Toxicity: Increased serum creatinine, decreased urine output

  • Pulmonary Toxicity: Pneumonitis

  • Additional Symptoms:

    • Renal failure

    • Pancytopenia

    • Nausea

    • Vomiting

    • Diarrhea

    • Stomatitis

    • Acute lung injury

    • Respiratory depression

Step 6: Assess Risk Factors

Factors Increasing Risk of Methotrexate Toxicity:

  • Renal Impairment: Reduced clearance of methotrexate

  • Drug Interactions: NSAIDs, penicillins, and other nephrotoxic agents can increase methotrexate levels

  • Genetic Polymorphisms: MTHFR C677T polymorphism can predict an increased risk of toxicity.

Using the Dose of Methotrexate per BSA in Practice

1.     Compare with Methotrexate Toxicity Thresholds: Ensure the dose per BSA does not exceed 500 mg/m² for high-dose protocols.

2.     Monitor Methotrexate Plasma Levels: Ensure methotrexate plasma concentrations do not exceed 0.1 μmol/L 48 hours after administration.

3.     Evaluate Clinical Symptoms of Methotrexate toxicity: Regularly assess the patient for symptoms of myelosuppression, gastrointestinal distress, hepatotoxicity, renal impairment, and pulmonary issues.

4.     Adjust for Risk Factors: Adjust dosing or provide additional monitoring for patients with renal impairment, potential drug interactions, or genetic predispositions.

Example Calculation and Application

Example 1:

  • Height: 144.78 cm

  • Weight: 52.6 kg

  • Total Methotrexate Dose: 140 mg

Step-by-Step Example Calculation

Step 1: Calculate BSA

BSA = √[(144.78 × 52.6) / 3600]

BSA = √[(7611.708) / 3600]

BSA = √[2.114]

BSA ≈ 1.453 m²

Step 2: Calculate the Dose per BSA

Dose per BSA = 140 mg / 1.453 m² ≈ 96.3 mg/m²

Step 3: Identify Toxicity Thresholds

The calculated dose per BSA (96.3 mg/m²) is well below the 500 mg/m² threshold for high-dose toxicity.

Step 4: Monitor Methotrexate Plasma Levels

Check methotrexate plasma levels at 24, 48, and 72 hours. Ensure levels do not exceed 0.1 μmol/L at 48 hours.

Step 5: Evaluate Clinical Symptoms

Monitor the patient for symptoms such as myelosuppression, gastrointestinal issues, hepatotoxicity, renal impairment, and pulmonary toxicity.

Step 6: Assess Risk Factors

Adjust dosing or increase monitoring for patients with renal impairment, potential drug interactions, or genetic predispositions.

Example 2:

  • Height: 165 cm

  • Weight: 60 kg

  • Total Methotrexate Dose: 1500 mg

Step-by-Step Example Calculation

Step 1: Calculate BSA

BSA = √[(165 × 60) / 3600]

BSA = √[(9900) / 3600]

BSA = √[2.75]

BSA ≈ 1.66 m²

Step 2: Calculate the Dose per BSA

Dose per BSA = 1500 mg / 1.66 m² ≈ 903.6 mg/m²

Step 3: Identify Toxicity Thresholds

The calculated dose per BSA (903.6 mg/m²) exceeds the 500 mg/m² threshold for high-dose toxicity, indicating a high risk for toxicity.

Step 4: Monitor Methotrexate Plasma Levels

Check methotrexate plasma levels at 24, 48, and 72 hours. Ensure levels do not exceed 0.1 μmol/L at 48 hours.

Step 5: Evaluate Clinical Symptoms

Monitor the patient for symptoms such as myelosuppression, gastrointestinal issues, hepatotoxicity, renal impairment, and pulmonary toxicity.

Step 6: Assess Risk Factors

Adjust dosing or increase monitoring for patients with renal impairment, potential drug interactions, or genetic predispositions.

By following these steps and using the calculations, healthcare providers can determine if a methotrexate dose is toxic and take appropriate measures to manage and mitigate the risk.

Clinical Presentation of Methotrexate Toxicity Based on Severity

  • Mild Methotrexate Toxicity: Symptoms include drowsiness and lethargy.

  • Moderate Methotrexate Toxicity: Symptoms include pronounced sedation, hypotension, tachycardia, nausea, vomiting, and stomatitis.

  • Severe Methotrexate Toxicity: Symptoms include significant CNS depression, seizures, serious cardiac effects, pancytopenia, acute lung injury, pneumonitis, renal failure, respiratory depression, elevated liver enzymes, increased serum creatinine, and decreased urine output

Close-up images showing severe oral mucositis, with inflammation, ulceration, and redness of the oral mucosa, typically seen as a side effect of chemotherapy treatments such as methotrexate. The images highlight the painful sores and lesions affecting the inner lips, tongue, and gums.
Severe oral mucositis from methotrexate (https://doi.org/10.14476/jomp.2015.40.2.82)

Management of Methotrexate Toxicity Based on Severity

  • Mild to Moderate Methotrexate Toxicity Management: The primary treatments include supportive care, monitoring, and stomatitis management.

  • Severe Methotrexate Toxicity Management: Emergency interventions and advanced supportive care, including airway management and continuous cardiac monitoring (Howard et al., 2016).

Leucovorin Dosage and Method of Administration for methotrexate toxicity

  • Early administration is crucial; within 1 hour

  • An intravenous dose equal to or greater than the methotrexate dose is needed. If the dose is unknown, administer 75 mg (children, 10 mg/m²/dose) initially, followed by 12 mg every 6 hours for four doses.

  • Use serum methotrexate levels to guide subsequent therapy.

  • Avoid oral therapy.

Leucovorin Dosing Based on Methotrexate Levels

Flowchart diagram illustrating the management of Methotrexate toxicity based on serum levels. It shows different treatment protocols for Methotrexate levels of 0.1–1 µmol/L, 1–5 µmol/L, and 5–10 µmol/L, and also outlines steps to take if there is a 50% increase in serum creatinine within 24 hours post-exposure.
Leucovorin Dosing Based on Methotrexate Levels
  • Methotrexate serum levels between 0.1–1 mcmol/L: Administer 10–15 mg/m² of leucovorin every 6 hours for 12 doses.

  • Levels between 1–5 mcmol/L: Increase the leucovorin dose to 50 mg/m² every 6 hours until the serum level is less than 0.1 mcmol/L.

  • Levels between 5–10 mcmol/L: Administer 100 mg/m² of leucovorin every 6 hours until the serum level drops below 0.1 mcmol/L.

  • If serum creatinine increases by 50% in the first 24 hours after methotrexate exposure, Increase the dose frequency of leucovorin to every 3 hours until the methotrexate level is less than 5 mcmol/L (Howard et al., 2016).

Methotrexate Levels and Toxicity Management

  • Importance of Methotrexate Levels: Monitoring methotrexate levels is essential in HDMTX therapy. Unlike repeated supratherapeutic exposures, which may not necessitate serum level checks, HDMTX requires precise monitoring. Serum methotrexate levels provide critical information for guiding rescue interventions and managing potential toxicities (Stoller et al., 1977).

Additional Treatment Strategies for Methotrexate Toxicity:

  • Intravenous leucovorin: Administer as soon as possible.

  • Intravenous fluids: Begin to support renal function.

  • Alkaline diuresis: Implement with a bicarbonate infusion to prevent renal precipitation of methotrexate.

  • Colony-stimulating factors: Administer filgrastim or sargramostim to counteract severe neutropenia.

  • Transfusion: Provide platelets and/or packed red cells for severe thrombocytopenia, anemia, or hemorrhage.

  • Severe nausea and vomiting: May respond to a combination of agents from different drug classes.

  • Glucarpidase: Rapidly reduces methotrexate levels, administered as formerly known as carboxypeptidase CPDG2 (Esteve et al., 2006).

Hemodialysis Considerations for Methotrexate Toxicity

The pharmacokinetics of methotrexate also influence its elimination during hemodialysis. Although methotrexate has a low volume of distribution and moderate protein binding, it can rebound after dialysis sessions. Therefore, prolonged dialysis sessions may be necessary to effectively reduce methotrexate levels in cases of severe toxicity (Abelson et al., 1983).

Intrathecal Methotrexate Overdose Management

An accidental intrathecal overdose of methotrexate is a medical emergency. Immediate measures include CSF exchange to reduce the concentration of the drug. This procedure, often called a "CSF oil change," involves removing and replacing CSF to lower drug levels. Intrathecal glucarpidase administration can also be highly effective, reducing methotrexate levels by 98% (Howard et al., 2016).

Evaluation of Methotrexate-Induced Stomatitis Management Strategies

Preventive Measures for Methotrexate-Induced Stomatitis

1.     Folic Acid Supplementation:

o   Several studies support the use of folic acid supplementation to reduce the toxicity associated with methotrexate therapy, including stomatitis (Bauer et al., 1999).

2.     Leucovorin (Folinic Acid) Rescue:

o   Leucovorin is widely recognized for its role in mitigating methotrexate toxicity, especially at higher doses, and for its effectiveness in preventing severe mucositis (Batchelor et al., 2003).

Methotrexate-Induced Stomatitis Symptomatic Treatment

1.     Oral Hygiene:

o   Maintaining good oral hygiene is a general recommendation to prevent secondary infections and promote the healing of oral ulcers (Seino et al., 2018).

2.     Topical Treatments:

o   Magic Mouthwash: This compounded mixture has components that help relieve pain and reduce inflammation. Its effectiveness is supported by clinical use in various cases (Troeltzsch et al., 2013).

o   Chlorhexidine Mouthwash: Clinical practice guidelines support the benefits of this antiseptic, which reduces bacterial load and aids in healing.

o   Benzydamine Hydrochloride Rinse: This topical NSAID helps reduce pain and inflammation, making it a recommended treatment for oral mucositis (Akagi et al., 2020).

3.     Pain Management:

o   Topical Anesthetics: Lidocaine gel provides localized pain relief, crucial for managing discomfort in patients with stomatitis (Seino et al., 2018).

o   Systemic Analgesics: NSAIDs or acetaminophen are recommended if topical treatments are insufficient.

Hydration and Nutrition

1.     Hydration:

o   Adequate fluid intake is essential to prevent dehydration and support overall health, especially in patients with oral pain, preventing normal intake.

2.     Soft, Bland Diet:

o   A soft diet minimizes irritation and allows for easier consumption, which is important for maintaining nutrition during episodes of stomatitis (Yamada et al., 2019).

Barrier Protection

1.     Oral Balance Gel or Biotene:

o   These products help protect the mucosa and provide moisture, which can be beneficial in managing dry and painful oral conditions.

Treating Secondary Infections

1.     Antifungal Treatment:

o   Secondary fungal infections, such as oral thrush, require antifungal medications like nystatin or fluconazole (Akagi et al., 2020).

2.     Antibiotic Treatment:

o   Appropriate antibiotics are necessary if secondary bacterial infections are present (Seino et al., 2018).

Modifying Methotrexate Therapy

1.     Dose Adjustment:

o   Temporarily reducing the dose of methotrexate or adjusting the dosing schedule can help manage severe stomatitis (Seino et al., 2018).

2.     Alternative Therapies:

o   In cases of severe and recurrent stomatitis, switching to an alternative medication may be necessary (Troeltzsch et al., 2013).

Monitoring and Follow-up

1.     Regular follow-up with a healthcare provider ensures that complications are managed promptly and treatment is adjusted as needed. This continuous monitoring is vital for effective management and early intervention. Regular monitoring of plasma levels is essential, especially at 24, 48, and 72 hours after administration, to ensure levels are within safe limits. Close monitoring helps in the timely intervention and management of potential toxicities.

The management strategies for methotrexate-induced stomatitis outlined above are scientifically valid and supported by clinical evidence from the literature. These strategies encompass preventive measures, symptomatic treatments, nutritional support, and adjustments in methotrexate therapy to mitigate the side effects and promote healing.

Observation Criteria for Methotrexate Toxicity

Home Observation

  • Threshold: Patients who have ingested doses below the toxic threshold exhibit mild or no symptoms.

  • Monitoring: Patients must be asymptomatic for at least 6 hours before considering home observation. If they remain asymptomatic for 6 hours, they may be sent home with instructions to seek medical attention if symptoms develop later due to potential delayed toxic effects.

  • Instructions: Return to a healthcare provider if any symptoms develop.

Emergency Department Observation

  • Symptoms: Patients with moderate to severe symptoms or ingested doses above the toxic threshold.

  • Action: Immediate medical evaluation and treatment to manage symptoms and prevent complications.

Extended Observation for Severe Cases

  • Severity: Patients with severe toxicity require intensive care and continuous monitoring.

  • Conditions: Includes patients presenting with:

    • Renal failure

    • Pancytopenia

    • Nausea

    • Vomiting

    • Diarrhea

    • Stomatitis

    • Acute lung injury

    • Respiratory depression

  • Monitoring: Patients with deliberate overdose and symptomatic patients need monitoring until clear improvement and clinical stability are evident.

Special Considerations

  • Psychiatric Evaluation: Patients with deliberate overdose require a psychiatric evaluation before discharge to ensure appropriate mental health support (Rask et al., 1998).

Prevention and Public Health Measures for Methotrexate Toxicity

  • Safe Prescribing Practices of Methotrexate: Follow dosage and monitoring guidelines to prevent toxicity.

  • Safe Storage and Handling of Methotrexate: Preventing accidental ingestion, particularly in households with children.

  • Public Education and Awareness about Methotrexate Toxicity: Raising awareness and educating healthcare providers and the public about the risks associated with methotrexate.

  • Poison Control and Emergency Preparedness: Ensuring accessibility and clear protocols for managing methotrexate toxicity (Esteve et al., 2006).

Conclusion: Managing and Preventing Methotrexate Toxicity

High-dose methotrexate is a powerful chemotherapeutic agent with a complex toxicity profile. Understanding the pharmacokinetics, monitoring serum levels, and implementing appropriate management strategies are crucial for optimizing patient outcomes. Continued education and awareness, implementing the latest guidelines, and balancing the therapeutic benefits with the potential risks of high-dose methotrexate are essential for future research and practice.

 

References:

  • Abelson, H., et al. "Methotrexate-Induced Renal Impairment: Clinical Studies and Rescue from Systemic Toxicity with High-Dose Leucovorin and Thymidine." Journal of Clinical Oncology, vol. 1, no. 3, 1983, pp. 208–216.

  • Akagi, Takahiko, et al. "Severe Oral Stomatitis Due to Reactivation of Herpes Simplex Virus Type 1 in a Methotrexate-Treated Patient with Dermatomyositis." Immunological Medicine, vol. 44, 2020, pp. 56-60.

  • Almalki, R. S., et al. "Methotrexate Toxicity: Molecular Mechanisms and Management." Journal of Pharmaceutical Research International, vol. 33, no. 49B, 2021, pp. 204–217, doi:10.9734/jpri/2021/v33i49B33357.

  • Batchelor, T., et al. "Treatment of Primary CNS Lymphoma with Methotrexate and Deferred Radiotherapy: A Report of NABTT 96-07." Journal of Clinical Oncology, vol. 21, no. 6, 2003, pp. 1044-1049.

  • Bauer, J., et al. "Ulcerative Stomatitis as Clinical Clue to Inadvertent Methotrexate Overdose." Der Hautarzt; Zeitschrift Fur Dermatologie, Venerologie, und Verwandte Gebiete, 1999.

  • Chan, E. S. L., and Rajakumar, D. V. "High-Dose Methotrexate in Adult Oncology Patients: Pharmacokinetic Principles and Practical Application." Therapeutic Drug Monitoring, vol. 36, no. 5, 2014, pp. 547-556.

  • Esteve, M., et al. "Severe Toxicity Associated with Methotrexate Therapy: Use of Therapeutic Drug Monitoring and Test-Dose to Guide Carboxypeptidase G2 Rescue and Methotrexate Continuation." European Journal of Clinical Pharmacology, vol. 63, 2006, pp. 39-42.

  • Garcia, H., et al. "Toxicité Rénale du Méthotrexate à Haute Dose." Nephrol Ther, vol. 14, suppl. 1, 2018, pp. S103-S113.

  • Howard, S. C., et al. "Preventing and Managing Toxicities of High-Dose Methotrexate." The Oncologist, vol. 21, 2016, pp. 1471–1482.

  • Huang, X., et al. "Leucovorin Ameliorated Methotrexate Induced Intestinal Toxicity via Modulation of the Gut Microbiota." Toxicology and Applied Pharmacology, vol. 391, 2020, p. 114900.

  • Kamen, B., et al. "Methotrexate Accumulation and Folate Depletion in Cells as a Possible Mechanism of Chronic Toxicity to the Drug." British Journal of Haematology, 1981.

  • Kim, Y. J., Song, M., and Ryu, J. "Mechanisms Underlying Methotrexate-Induced Pulmonary Toxicity." Expert Opinion on Drug Safety, vol. 8, no. 4, 2009, pp. 451-458.

  • Kivity, S., et al. "Clinical Characteristics and Risk Factors for Low-Dose Methotrexate Toxicity: A Cohort of 28 Patients." Autoimmunity Reviews, vol. 13, no. 11, 2014, pp. 1109-1113.

  • Lateef, O., Shakoor, N., and Balk, R. "Methotrexate Pulmonary Toxicity." Expert Opinion on Drug Safety, vol. 4, no. 4, 2005, pp. 723-730.

  • Lee, H.-J., et al. "Methotrexate-Induced Oral Mucositis." Journal of Oral Medicine and Pain, 2015, doi:10.14476/jomp.2015.40.2.82.

  • Mahmoud, L., et al. "Use of MTHFR C677T Polymorphism and Plasma Pharmacokinetics to Predict Methotrexate Toxicity in Patients with Acute Lymphoblastic Leukemia." Advances in Clinical and Experimental Medicine, vol. 27, no. 8, 2018, pp. 1061-1068.

  • Rask, C., et al. "Clinical and Pharmacokinetic Risk Factors for High-Dose Methotrexate-Induced Toxicity in Children with Acute Lymphoblastic Leukemia: A Logistic Regression Analysis." Acta Oncologica, vol. 37, no. 3, 1998, pp. 277-284.

  • Seino, Sayaka, et al. "A Case of Severe Stomatitis Associated with Pancytopenia that Developed During Methotrexate Therapy for Rheumatoid Arthritis." Japanese Journal of Oral and Maxillofacial Surgery, 2018.

  • Stoller, R. G., et al. "Use of Plasma Pharmacokinetics to Predict and Prevent Methotrexate Toxicity." The New England Journal of Medicine, vol. 297, no. 12, 1977, pp. 630-634.

  • Troeltzsch, Matthias, et al. "Oral Mucositis in Patients Receiving Low-Dose Methotrexate Therapy for Rheumatoid Arthritis: Report of 2 Cases and Literature Review." Oral Surgery, Oral Medicine, Oral Pathology, and Oral Radiology, vol. 115, no. 5, 2013, pp. e28-33.

  • Ulrich, C., et al. "Pharmacogenetics of Methotrexate: Toxicity Among Marrow Transplantation Patients Varies with the Methylenetetrahydrofolate Reductase C677T Polymorphism." Blood, vol. 98, no. 1, 2001, pp. 231-234.

  • Yamada, Kohta, et al. "Severe Stomatitis Caused by Misuse of Methotrexate in an Elderly Patient with Chronic Rheumatoid Arthritis." Journal of Oral and Maxillofacial Surgery, Medicine, and Pathology, 2019.