GET THE APP

Methotrexate Side Effects: Review Article
Journal of Drug Metabolism & Toxicology

Journal of Drug Metabolism & Toxicology
Open Access

ISSN: 2157-7609

+44-20-4587-4809

Review Article - (2012) Volume 3, Issue 4

Methotrexate Side Effects: Review Article

Emna Gaies*, Nadia Jebabli, Sameh Trabelsi, Issam Salouage, Rim Charfi, Mohamed Lakhal and Anis Klouz
Service de Pharmacologie Clinique - Centre National de Pharmacovigilance de Tunis, Tunisie
*Corresponding Author: Dr. Emna Gaies, Service de Pharmacologie Clinique - Centre National de Pharmacovigilance, 9 Avenue Dr. Zouheir Essafi, 1006 Tunis, Tunisie Email:

Abstract

Methotrexate (MTX) is an antifolate first developed to treat certain types of cancer. It was used at higher doses as a cancer therapy and since 1990 it is used at much lower doses to treat rheumatic diseases [1]. Side effects of MTX high dose (MTX-HD) may be life threatening, however those of various doses of oral MTX are variable because of the interindividual variability of gastrointestinal absorption of this drug. Bone marrow, gastrointestinal mucosa and hair are particularly vulnerable to the effects of MTX, secondary to their high rate of cellular turnover [2] and because MTX concentration is inversly proportianal to renal clearance [2], renal toxicity is frequent with MTX-HD. This review aimed at exposing MTX toxicity in different organs, at explaining pathogenic mechanisms of these toxicities and their prevention or treatment.

Introduction

Methotrextae (MTX) is an antifolate first developed to treat certain types of cancer. It was used at higher doses as a cancer therapy and since 1990 it is used at much lower doses to treat rheumatic diseases [1].

Side effects of MTX high dose (MTX-HD) may be life threatening, however those of various doses of oral MTX are variable because of the interindividual variability of gastrointestinal absorption of this drug. Bone marrow, gastrointestinal mucosa and hair are particularly vulnerable to the effects of MTX, secondary to their high rate of cellular turnover [2] and because MTX concentration is inversly proportianal to renal clearance [2], renal toxicity is frequent with MTX-HD.

This review is aimed at exposing MTX toxicity in different organs, at explaining pathogenic mechanisms of these toxicities and their prevention or treatment.

Renal Toxicity

Acute renal failure due to acute tubular necrosis induced by MTXHD is rare (2 à 4%) [3] but serious and redoubtable. This toxicity is due to the precipitation of MTX or its metabolites in the renal tubules [4-6] causing obstruction and diminition of renal clearance with consequently prolongation of MTX high levels. High MTX levels may in turn lead to ineffective rescue by leucoverin and an enhancement of MTX’s other toxicities [7-9]. MTX may also acts as a direct toxin on the tubular epithelium [10] and causes vasoconstriction of the afferent arteriole [12].

MTX and its metabolites are relatively insoluble in acid urine [4,11]. An increase in the urine pH results on a greater solubility of the MTX and its metabolites. For that reason, it is recommended to monitor renal function before, during and after MTX infusion in order to control its plsamtic levels. Intravenous hydratation and urine alkalinization were made before, during and after the infusion of MTX-HD [2,3]. Clinically, patients with acute renal failure are usually asymptomatic. It consists frequently on a nonoliguric renal failure which generally disappears in two to three weeks [12-14]. Some patients may develop nausea, vomiting or diarrhea as prodroms [14,15]. Urine alkalinization and leucoverin rescue are the cornerstones of the management of the earlier signs of renal dysfunction. Peritoneal dialysis, hemodialysis and hemofiltration have been tried and may be helpful for the management of the MTX intoxication with acute renal failure. However these methods are invasive and are associated in postdialysis to a marked rebound in plasma MTX concentrations [16-19]. Thymidine, an endogenous nucleoside, has been shown to rescue cells from the MTX effects. It does not compete with MTX for transport into the cell but directed converted to thymidine monophosphate and circumvent de novo pathway blocked by MTX [8,20]. Furthermore carboxypeptidase (CPDG2) an enzyme able to reduce MTX levels by metabolising circulating MTX to the inactive metabolite DAMPA (deglutamated 4-amino-4-deoxy-N10-methyl pteroic acid) providing another route for MTX elimination [8,15,21].

Neurotoxicity

MTX can induce acute, subacute or chronic neurotoxicity. This toxicity is mainly observed after intrathecal or intravenous administration of MTX [22-24]. Mechanisms of toxicity are not yet fully known but many hypotheses may explain this neurotoxicity, such as interference of MTX with transmethylation reactions which are important for the formation of proteins, lipids and myelin [25]. MTX decreases also the rate of methionine and S-adenosyl methionine in cerebrospinal fluid and increases levels of S-adenosyl homocysteine and homocysteine. It was demonstrated that elevated rates of homocysteine may be responsible for the vascular phenomena of MTX neurotoxicity [26,27].

The most frequent acute neurologic manifestation is leucoencephalopathy [11]. This entity can be subclinical diagnosed only by magnetic resonance or manifested by insomnia, confusion, agitation, seizure and coma [28,29]. Headache, nausea, vomiting and aseptic meningitis are also observed after intrathecal administration of MTX [30,31]. These symptoms represent another aspect of acute MTX neurotoxicity. They are generally not severe, last 12 to 72 hours [31] and desappear with the discontinuation of the drug.

Paraplegia, cerebellar dysfunction, seizure are reported as a subacute MTX neurotoxicity [30] and occured a few weeks after MTX initiation [32].

Chronic neurotoxicity is observed several months to years after MTX therapy. It is an irreversible complication and it is observed especially if an encephalic radiotherapy was associated to the therapeutic protocol [2,24]. Necrotic leucoencephalopathy is the most frequent complication characterised by a slow progressive cognitive deterioration, seizures, ataxia, spasticity and/or coma [2,12,28].

MTX neurotoxicity is usually treated with aminophylline or leucoverin administration [33,34].

Hematologic Toxicity

Hematologic toxicity is a serious complication commonly observed with MTX-HD [35]. This complication consists of a thrombocytopenia followed by a rapidly progressive leukoneutropenia [12]. Leukopenia occurs from one to three weeks and marrow recovery is generally observed within approximately 3 weeks [36].

Hematologic toxicity including thrombocytopenia, megaloblastic anemia, leukopenia and pancytopenia with MTX low doses are rare [37,38]. Their prevalence is about 3% in patients with rheumatoid arthritis treated by MTX and the incidence of pancytopenia in these subjects is approximatively 1.4% [39,40]. The frequency of pancytopenia may increase with co-administration of other drugs, folic acid deficiency, hypoalbumenia, concomitant infections, advanced age, dehydratation and renal impairment [41,42]. Pathogenesis of MTX inducing panctopenia is unclear. Pancytopenia may be acute or chronic and thought to be an allergy-like reaction [43,44]. Discontinuation of MTX represents the basis of therapy but the use of G-CSF and methylprednisolone are also beneficial [45].

Cutaneomucous Toxicity

MTX has a variety of cutaneous side effects, particularly when it is administered at high doses. It generally occurs when recommended guidelines are ignored or renal excretion is decreased.

The most frequent mucocutaneous reactions to MTX are ulcerations of the oral mucosa, burning sensation of the skin, photosensitivity, acral erythema, multiform erythema, urticaria and vasculitis [46].

The pathogenesis of skin adverse reactions due to MTX is not well known. Skin reactions to MTX-HD seem to be due to a cytotoxic-T lymphocytes and mononuclear cells that induce apoptosis in keratinocytes expressing drug-derived antigens at their surfaces [47,48]. Hypersensitivity reactions may also explain some cutaneous side effects. It has been also suggested that MTX can induce cutaneous small-vessel vasculitis in patients with collagen vascular disease who were treated with low-dose of MTX [49].

Gastro-intestinal Toxicity

MTX can induce a variety of gastro-intestinal disorder. Patients usually present abdominal pain, vomiting and diarrehea. These side effects may occur either with high or low MTX doses [2].

Hepatotoxicity is a common complication of long term treatment with MTX [2,50]. In rhumatoid arthritis and psoriatic arthritis, an increase in aminotransferases levels was obseserved with a frequency varing from 7, 5 to 26% [50,51]. In another hand, typical histopathologic changes in the liver were observed with MTX and have been divided into four grades according to Roenigk classification [52]; Grade I: mild fatty infiltration, nuclear variability, with or without portal inflammation; Grade II : moderate to severe fatty infiltration, nuclear variability, and portal tract expansion, inflammation and necrosis; Grade IIIA: mild fibrosis; Grade IIIB: moderate to severe fibrosis and Grade IV: cirrhosis.

Many risk factors may increase the occurrence of severe hepatotoxicity (Grade III or IV) such as a long duration of exposure to MTX and its cumulative dose, hepatitis B or C infection, alcoholism, diabetes, obesity and non alcoholic steatohepatitis [53,54].

The mechanism by which MTX affects the liver is unclear. Hepatic folate stores are depleted by MTX in the doses used in rhumatoid arthritis and these stores can be repleted by short-term administration of oral folinic acid [55]. A relationship between folate depletion and hepatic toxicity has not been established. However, supplementation with either folic acid 1 mg per day or folinic acid 2.5 mg per week is associated with a reduced incidence of aminotransferases elevation [56,57].

Pulmonary Toxicity

Pneumonitis is one of the most serious but infrequent side effects of MTX low doses. Its prevalence seems about 0.9 to 1% [58,59]. Mechanism of pneumonitis is an hypersensitivity reaction to MTX mediated by activated T-cells [2,60]. In fact, MTX leads to a cytokines release by type 2 alveolar cells causing an alveolitis by recruitment of inflammatory cells [61]. MTX can also stimulate lung fibroblasts and epithelial cells to induce recruitment of eosinophils [62]. It has been also demonstrated that neutrophiles are implicated in the pathogenesis of lung fibrosis [63,64]. Clinically symptoms may occur from few days to more than a year after the begining of MTX therapy [65,66] and also several weeks after MTX discontinuation [67]. An insidious nonproductive cough is the most common symptom [66]. Fever, malaise, dyspnea may also occur [68]. Chest x-ray may show either localised or diffuse interstitiel changes [69]. Tomography has a higher sensitivity and shows commonly diffuse bilateral and patchy ground glass [70]. Peripheral eosinophilia was seen in one third of patients [69], bronchoalveolar lavage shows lymphocytosis about 33-68% with a disproportionate increase in the CD4+/CD8+ ratio [71]. Lung biopsy is not done all the times because it is an invasive method and because of the availability of tomography.

Treatment of MTX pneumonitis is based on corticotherapy and immediate discontinuation of MTX [59]. In difficult cases cyclophosphamide has been successfully used [72]. The prognosis of MTX-associated lung injury is generally favourable [73].

Prevention and Management of MTX-HD Toxicity

To avoid MTX toxicity, there are some general aspects of MTX-HD administration and post-treatment management that are common to all regimens.

Assessing renal function

Because MTX is eliminated essentially by the kidneys. Therefore it is mandatory to determine renal function for MTX-HD administration. In the presence of an impaired renal fuction, MTX dose adjustement is necessary. So, when creatinine clearance (CrCl) is between 30 and 60 ml/min, dose of MTX is reduced by 50% and when CrCl is between 10 and 30 ml/min, dose of MTX should be reduced by 75% [74].

Maintaining adequate hydratation

Agressive hydration is important to promote diuresis and to prevent intratubular precipitation of MTX. Most protocols recommend at least 2.5 to 3.5 L/m2 of IV fluid hydration per day, starting 4 to 12 hours prior to the initiation of the MTX infusion [2,3].

Maintaining alkaline urine pH

MTX and its metabolite 7-OH-MTX, which is predominent with MTX-HD, show respectively 20 and 12 fold increased solubility when pH increases from 5 to 7 [4]. Renal tubular precipitation of MTX and 7-OH-MTX occurs when pH is lower than 5.7 [75]. In clinical practice, it is essential to begin the MTX infusion only after the urine pH is ≥ 7.0 and to maintain it in this range until plasma MTX levels decline to less than 0.1 μM.

Avoiding drug interactions

Toxicity with MTX-HD may be increased when there is coadministartion of drugs having the potentiel to displace MTX from serum proteins and/or to reduce MTX clearance. The most known are interaction with trimethoprim and sulfamethoxazole (TMP-SMX) and non steroidal anti-inflammatory drugs (NSAID) [44,76,77]. Alteration of the elimination of MTX was also reported with pyrazoles, aminoglycosides, probenecid, some penicillins and macrolides [78], omeprazole [79,80], mezocilline [81], piperacillin [82], amphotericine B [83] and ciprofloxacin [84].

Drainage of third space fluids: The presence of a third space fluid like pleural effusions or ascites is an important contraindication to the administartion of MTX-HD. Third space fluids lead to a prolonged MTX plasma half life and subsequently to a prolonged exposure to MTX and to the risk of toxicity. Drainage of third space fluids before MTX-HD is recommended to prevent toxicity [75].

Leucoverin rescue: Administration of reduced folate such as leucoverin is mandatory in order to circumvent the metabolic blockage imposed by MTX [85]. Leucoverin rescue should be started within 24 to 36 hours of the start of the MTX infusion. The dose and the frequency of leucoverin rescue have been developed empirically and differ according to the regimen of MTX-HD. Rescue doses of leucoverin that are commonly given are between 10 to 15 mg/m2. Leucoverin is given every six hours until plasma MTX levels are less than 0.2 μM [2].

Monitoring plasma MTX concentration

Monitoring plasma MTX is an essentiel part of MTX-HD therapy. It aimed at identifying patients with the highest risk for MTX toxicity. MTX levels should be followed daily. Plasma MTX levels are usually measured at 24, 48, and 72 hours after starting the MTX infusion. For 24-hour infusional regimens, the initial MTX measurement may be at 36 hours. To avoid MTX toxicity, levels should be above 10 μM at 24 hours, 1 μM at 48 hours and 0.15 μM at 72 hours [86,12].

Conclusion

Because MTX can cause many side effects and some of them are life threatening, it is important to recognize them as the drug must be discontinued immediately and rescue measures instituted. Many of these side effects can be avoided by a close monitoring and a good prevention.

References

  1. Benedek TG (2010) Methotrexate: from its introduction to non-oncologic therapeutics to anti-TNF. Clin Exp Rheumatol 28: S3-8.
  2. Olsen EA (1991) The pharmacology of methotrexate. J Am Acad Dermatol 25: 306-318.
  3. Widemann BC, Adamson PC (2006) Understanding and managing methotrexate nephrotoxicity. Oncologist 11: 694-703.
  4. Jacobs SA, Stoller RG, Chabner BA, Johns DG (1976) 7-Hydroxymethotrexate as a urinary metabolite in human subjects and rhesus monkeys receiving high dose methotrexate. J Clin Invest 57: 534-538.
  5. Lankelma J, van der Klein E, Ramaekers F (1980) The role of 7-hydroxymethotrexate during methotrexate anti-cancer therapy. Cancer Lett 9: 133-142.
  6. Smeland E,Fuskevåg OM, Nymann K, Svendesn JS, Olsen R, et al. (1996) High-dose 7-hydromethotrexate: acute toxicity and lethality in a rat model. Cancer Chemother Pharmacol 37: 415-422.
  7. Frei E 3rd, Blum RH, Pitman SW, Kirkwood JM, Henderson IC, et al. (1980) High dose methotrexate with leucovorin rescue. Rationale and spectrum of antitumor activity. Am J Med 68: 370-376.
  8. Abelson HT, Fosburg MT, Beardsley GP, Goorin AM, Gorka C, et al. (1983) Methotrexate-induced renal impairment: clinical studies and rescue from systemic toxicity with high-dose leucovorin and thymidine. J Clin Oncol 1: 208-216.
  9. Stark AN, Jackson G, Carey PJ, Arfeen S, Proctor SJ (1989) Severe renal toxicity due to intermediate-dose methotrexate. Cancer Chemother Pharmacol 24: 243-245.
  10. Messmann R, Allegra C (2001) Antifolates. In: Chabner B, Longo D (Eds) Cancer Chemotherapy and Biotherapy. Philadelphia, Lippincott Williams & Wilkins 139-84.
  11. Donehower RC, Hande KR, Drake JC, Chabner BA (1979) Presence of 2,4-diamino-N10-methylpteroic acid after high-dose methotrexate. Clin Pharmacol Ther 26: 63-72.
  12. Retenauer S, chauveau D, Récher C (2009) Surdsage au méthotrexate:: complications, prise en charge et prevention High-dose methotrexate: toxicity, management and prevention. Reanimation 18: 654-658.
  13. Relling MV, Fairclough D, Ayers D, Crom WR, Rodman JH, et al. (1994) Patient characteristics associated with high-risk methotrexate concentrations and toxicity. J Clin Oncol 12: 1667-1672.
  14. Widemann BC, Balis FM, Murphy RF, Sorensen JM, Montello MJ, et al. (1997) Carboxypeptidase-G2, thymidine, and leucovorin rescue in cancer patients with methotrexate-induced renal dysfunction. J Clin Oncol 15: 2125-2134.
  15. Lawrenz-Wolf B, Wolfrom C, Frickel C, Fengler R, Wehinger H, et al. (1994) [Severe renal impairment of methotrexate elimination after high dose therapy]. Klin Padiatr 206: 319-326.
  16. Hande KR, Balow JE, Drake JC, Rosenberg SA, Chabner BA (1977) Methotrexate and hemodialysis. Ann Intern Med 87: 495-496.
  17. Gibson TP, Reich SD, Krumlovsky FA, Ivanovich P, Gonczy C (1978) Hemoperfusion for methotrexate removal. Clin Pharmacol Ther 23: 351-355.
  18. Greil J, Wyss PA, Ludwig K, Bonakdar S, Scharf J, et al. (1997) Continuous plasma resin perfusion for detoxification of methotrexate. Eur J Pediatr 156: 533-536.
  19. Bouffet E, Frappaz D, Laville M, Finaz J, Pinkerton CR, et al. (1986) Charcoal haemoperfusion and methotrexate toxicity. Lancet 1: 1497.
  20. Grem JL, King SA, Sorensen JM, Christian MC (1991) Clinical use of thymidine as a rescue agent from methotrexate toxicity. Invest New Drugs 9: 281-290.
  21. Widemann BC, Hetherington ML, Murphy RF, Balis FM, Adamson PC (1995) Carboxypeptidase-G2 rescue in a patient with high dose methotrexate-induced nephrotoxicity. Cancer 76: 521-526.
  22. Brugnoletti F, Morris EB, Laningham FH, Patay Z, Pauley JL, et al. (2009) Recurrent intrathecal methotrexate induced neurotoxicity in an adolescent with acute lymphoblastic leukemia: Serial clinical and radiologic findings. Pediatr Blood Cancer 52: 293-295.
  23. Shuper A, Stark B, Kornreich L, Cohen IJ, Avrahami G, et al. (2002) Methotrexate-related neurotoxicity in the treatment of childhood acute lymphoblastic leukemia. Isr Med Assoc J 4: 1050-1053.
  24. Keime-Guibert F, Napolitano M, Delattre JY (1998) Neurological complications of radiotherapy and chemotherapy. J Neurol 245: 695-708.
  25. Harila-Saari AH, Vainionpää LK, Kovala TT, Tolonen EU, Lanning BM (1998) Nerve lesions after therapy for childhood acute lymphoblastic leukemia. Cancer 82: 200-207.
  26. Hankey GJ, Eikelboom JW (2001) Homocysteine and stroke. Curr Opin Neurol 14: 95-102.
  27. Cronstein BN, Naime D, Ostad E (1993) The antiinflammatory mechanism of methotrexate. Increased adenosine release at inflamed sites diminishes leukocyte accumulation in an in vivo model of inflammation. J Clin Invest 92: 2675-2682.
  28. Shuper A, Stark B, Kornreich L, Cohen IJ, Aviner S, et al. (2000) Methotrexate treatment protocols and the central nervous system: significant cure with significant neurotoxicity. J Child Neurol 15: 573-580.
  29. Kishi T, Tanaka Y, Ueda K (2000) Evidence for hypomethylation in two children with acute lymphoblastic leukemia and leukoencephalopathy. Cancer 89: 925-931.
  30. Bleyer WA (1978) The clinical pharmacology of methotrexate: new applications of an old drug. Cancer 41: 36-51.
  31. Weiss HD, Walker MD, Wiernik PH (1974) Neurotoxicity of commonly used antineoplastic agents (first oftwo parts). N Engl J Med 291: 75-81.
  32. Bleyer WA (1977) Current status of intrathecal chemotherapy for human meningeal neoplasms. Natl Cancer Inst Monogr 46: 171-178.
  33. Bernini JC, Fort DW, Griener JC, Kane BJ, Chappell WB, et al. (1995) Aminophylline for methotrexate-induced neurotoxicity. Lancet 345: 544-547.
  34. Winick NJ, Bowman WP, Kamen BA, Roach ES, Rollins N, et al. (1992) Unexpected acute neurologic toxicity in the treatment of children with acute lymphoblastic leukemia. J Natl Cancer Inst 84: 252-256.
  35. Isacoff WH, Townsend CM, Eiber FR, Forster T, Morton DL, et al. (1976) High dose methotrexate therapy of solid tumors: observations relating to clinical toxicity. Med Pediatr Oncol 2: 319-325.
  36. Bertino JR (1982) Clinical pharmacology of methotrexate. Med Pediatr Oncol 10: 401-411.
  37. Agarwal V, Chauhan S, Singh R, Sachdev A, D’Cruz S, et al. (2005) Pancytopenia with the first dose of methotrexate in a patient with psoriatic arthritis. J Indian Rheumatol Assoc 13: 60-1.
  38. Gülerr N, Tander B, Kisacik B, Utku U, Cevlk A, et al. (2008) Low Dose Methotrexate Associated Pancytopenia in a Patient with Rheumatoid Arthritis. Turk J Phys Med Rehab 54: 79-81.
  39. Weinblatt ME (1985) Toxicity of low dose methotrexate in rheumatoid arthritis. J Rheumatol Suppl 12: 35-39.
  40. Gutierrez-Ureña S, Molina JF, García CO, Cuéllar ML, Espinoza LR (1996) Pancytopenia secondary to methotrexate therapy in rheumatoid arthritis. Arthritis Rheum 39: 272-276.
  41. Yang CP, Kuo MC, Guh JY, Chen HC (2006) Pancytopenia after low dose methotrexate therapy in a hemodialysis patient: case report and review of literature. Ren Fail 28: 95-97.
  42. Preet Singh Y, Aggarwal A, Misra R, Agarwal V (2007) Low-dose methotrexate-induced pancytopenia. Clin Rheumatol 26: 84-87.
  43. Kuitunen T, Malmström J, Palva E, Pettersson T (2005) Pancytopenia induced by low-dose methotrexate. A study of the cases reported to the Finnish Adverse Drug Reaction Register from 1991 to 1999. Scand J Rheumatol 34: 238-241.
  44. Doolittle GC, Simpson KM, Lindsley HB (1989) Methotrexate-associated, early-onset pancytopenia in rheumatoid arthritis. Arch Intern Med 149: 1430-1431.
  45. Kondo H, Date Y (1997) Benefit of simultaneous rhG-CSF and methylprednisolone 'pulse' therapy for methotrexate-induced bone marrow failure in rheumatoid arthritis. Int J Hematol 65: 159-163.
  46. Del Pozo J, Martínez W, García-Silva J, Almagro M, Peña-Penabad C, et al. (2001) Cutaneous ulceration as a sign of methotrexate toxicity. Eur J Dermatol 11: 450-452.
  47. Heenen M, Laporte M, Noel JC, de Graef C (1998) Methotrexate induces apoptotic cell death in human keratinocytes. Arch Dermatol Res 290: 240-245.
  48. Bell R, Sullivan JR, Burdon JG, Sinclair R (1979) Toxic rash associated with high dose methotrexate therapy. Clin Exp Pharmacol Physiol Suppl 5: 57-61.
  49. Goerttler E, Kutzner H, Peter HH, Requena L (1999) Methotrexate-induced papular eruption in patients with rheumatic diseases: a distinctive adverse cutaneous reaction produced by methotrexate in patients with collagen vascular diseases. J Am Acad Dermatol 40: 702-707.
  50. Sotoudehmanesh R, Anvari B, Akhlaghi M, Shahraeeni S, Kolahdoozan S (2010) Methotrexate Hepatotoxicity in Patients with Rheumatoid Arthritis. Middle East Journal of Digestive Diseases 2: 104-9.
  51. Tilling L, Townsend S, David J (2006) Methotrexate and hepatic toxicity in rheumatoid arthritis and psoriatic arthritis. Clin Drug Investig 26: 55-62.
  52. Roenigk HH Jr, Auerbach R, Maibach HI, Weinstein GD (1988) Methotrexate in psoriasis: revised guidelines. J Am Acad Dermatol 19: 145-156.
  53. Diouf ML, Diallo S, Mbengue M, Moreira-Diop T (2001) [Methotrexate, liver and rheumatoid arthritis in tropical areas]. Sante 11: 195-200.
  54. Kremer JM, Petrillo GF, Hamilton RA (1995) Pharmacokinetics and renal function in patients with rheumatoid arthritis receiving a standard dose of oral weekly methotrexate: association with significant decrease in creatinin clearance and renal clearance of the drug after 6 month of therapy. J Rheumatol 22: 38-40.
  55. Rodenhuis S, Kremer JM, Bertino JR (1987) Increase of dihydrofolate reductase in peripheral blood lymphocytes of rheumatoid arthritis patients treated with low-dose oral methotrexate. Arthritis Rheum 30: 369-374.
  56. Prey S, Paul C (2009) Effect of folic or folinic acid supplementation on methotrexate-associated safety and efficacy in inflammatory disease: a systematic review. Br J Dermatol 160: 622-628.
  57. van Ede AE, Laan RF, Rood MJ, Huizinga TW, van de Laar MA, et al. (2001) Effect of folic or folinic acid supplementation on the toxicity and efficacy of methotrexate in rheumatoid arthritis: a forty-eight week, multicenter, randomized, double-blind, placebo-controlled study. Arthritis Rheum 44: 1515-1524.
  58. Kinder AJ, Hassell AB, Brand J, Brownfield A, Grove M, et al. (2005) The treatment of inflammatory arthritis with methotrexate in clinical practice: treatment duration and incidence of adverse drug reactions. Rheumatology (Oxford) 44: 61-66.
  59. Chikura B, Sathi N, K. Dawson JK (2009) Methotrexate induced pneumonitis: A Review Article. Curr Respir Med Rev 5: 12-20.
  60. Akoun GM, Gauthier-Rahman S, Mayaud CM, Touboul JL, Denis MF (1987) Leukocyte migration inhibition in methotrexate-induced pneumonitis. Evidence for an immunologic cell-mediated mechanism. Chest 91: 96-99.
  61. Koyama S, Sato E, Takamizawa A, Tsukadaira A, Haniuda M, et al. (2003) Methotrexate stimulates lung epithelial cells to release inflammatory cell chemotactic activities. Exp Lung Res 29: 91-111.
  62. Sato E, Camhi SL, Koyama S, Robbins RA (2001) Methotrexate stimulates lung fibroblasts and epithelial cells to release eosinophil chemotactic activity. J Rheumatol 28: 502-508.
  63. Obayashi Y, Yamadori I, Fujita J, Yoshinouchi T, Ueda N, et al. (1997) The role of neutrophils in the pathogenesis of idiopathic pulmonary fibrosis. Chest 112: 1338-1343.
  64. Rosner V, Chabot F, Moreau L, Grignon Y, Delorme N, et al. (1995) [Contribution of bronchoalveolar lavage and transbronchial biopsy to diagnosis and prognosis of drug-induced pneumopathies]. Rev Pneumol Clin 51: 269-274.
  65. Kremer JM,Alarcón GS, Weinblatt ME, Kaymakcian MV, Macaluso M, et al. (1997) Clinical, laboratory, radiographic, and histopathologic features of methotrexate-associated lung injury in patients with rheumatoid arthritis: a multicenter study with literature review. Arthritis Rheum 40: 1829-1837.
  66. Bartram SA (1998) Experience with methotrexate-associated pneumonitis in northeastern England: comment on the article by Kremer et al. Arthritis Rheum 41: 1327-1328.
  67. Elsasser S, Dalquen P, Soler M, Perruchoud AP (1989) Methotrexate-induced pneumonitis: appearance four weeks after discontinuation of treatment. Am Rev Respir Dis 140: 1089-1092.
  68. Schnabel A, Dalhoff K, Bauerfeind S, Barth J, Gross WL (1996) Sustained cough in methotrexate therapy for rheumatoid arthritis. Clin Rheumatol 15: 277-282.
  69. Zisman DA, McCune WJ, Tino G, Lynch JP 3rd (2001) Drug-induced pneumonitis: the role of methotrexate. Sarcoidosis Vasc Diffuse Lung Dis 18: 243-252.
  70. Arakawa H, Yamasaki M, Kurihara Y, Yamada H, Nakajima Y (2003) Methotrexate-induced pulmonary injury: serial CT findings. J Thorac Imaging 18: 231-236.
  71. Schnabel A, Richter C, Bauerfeind S, Gross WL (1997) Bronchoalveolar lavage cell profile in methotrexate induced pneumonitis. Thorax 52: 377-379.
  72. Suwa A, Hirakata M, Satoh S, Mimori T, Utsumi K, et al. (1999) Rheumatoid arthritis associated with methotrexate-induced pneumonitis: improvement with i.v. cyclophosphamide therapy. Clin Exp Rheumatol 17: 355-358.
  73. Sostman HD, Matthay RA, Putman CE, Smith GJ (1976) Methotrexate-induced pneumonitis. Medicine (Baltimore) 55: 371-388.
  74. Treon SP, Chabner BA (1996) Concepts in use of high-dose methotrexate therapy. Clin Chem 42: 1322-1329.
  75. Fox RM (1979) Methotrexate nephrotoxicity. Clin Exp Pharmacol Physiol 5: 43-44.
  76. Katchamart W, Bourré--Tessier J, Donka T, Drouin J, Rohekar G, et al. (2010) Canadian recommendations for use of methotrexate in patients with rheumatoid arthritis. J Rheumatol 37: 1422-1430.
  77. Franck H, Rau R, Herborn G (1996) Thrombocytopenia in patients with rheumatoid arthritis on long-term treatment with low dose methotrexate. Clin Rheumatol 15: 266-270.
  78. Illiger HJ, Bornmann L, Herdrich K (1995) Methotrexate. In: Drug interactions in the therapy of malignant diseases. (3rdedn), Munich: W. Zuckschwerdt Verlag 114-21.
  79. Suzuki K, Doki K, Homma M, Tamaki H, Hori S, et al. (2009) Co-administration of proton pump inhibitors delays elimination of plasma methotrexate in high-dose methotrexate therapy. Br J Clin Pharmacol 67: 44-49.
  80. Reid T, Yuen A, Catolico M, Carlson RW (1993) Impact of omeprazole on the plasma clearance of methotrexate. Cancer Chemother Pharmacol 33: 82-84.
  81. Dean R, Nachman J, Lorenzana AN (1992) Possible methotrexate-mezlocillin interaction. Am J Pediatr Hematol Oncol 14: 88-89.
  82. Najjar TA, Abou-Auda HS, Ghilzai NM (1998) Influence of piperacillin on the pharmacokinetics of methotrexate and hydroxymethotrexate. Cancer Chemother Pharmacol 42: 423-8.
  83. Gaïes E, Trabelsi S, Sahnoun R, Salouage I, Hamza I, et al. (2010) Modification de la pharmacocinétique du méthotrexate suite à l’administration d’amphotéricine B : à propos d’un cas. J Afr Cancer 2: 264-6.
  84. Dalle JH, Auvrignon A, Vassal G, Leverger G, Kalifa C (2001) [Methotrexate-ciprofloxacin interaction: report of two cases of severe intoxication]. Arch Pediatr 8: 1078-1081.
  85. Ackland SP, Schilsky RL (1987) High-dose methotrexate: a critical reappraisal. J Clin Oncol 5: 2017-2031.
  86. Le Guellec C, Blasco H, Benz I, Hulin A (2010) Niveau de preuve du suivi thérapeutique pharmacologique du méthotrextae au décours de son administration à haute dose. Thérapie 653: 163-9.3-9.
Citation: Gaies E, Jebabli N, Trabelsi S, Salouage I, Charfi R, et al. (2012) Methotrexate Side Effects: Review Article. J Drug Metab Toxicol 3:125.

Copyright: © 2012 Gaies E, et al. This is an open-access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.
Top