Content » Vol 97, Issue 4

Special Report

Methotrexate Use and Monitoring in Patients with Psoriasis: A Consensus Report Based on a Danish Expert Meeting

Line Raaby1, Claus Zachariae2, Monika Østensen3, Lene Heickendorff4, Peter Thielsen5, Henning Grønbæk6, Lone Skov2, Nini Kyvsgaard7, Jakob T. Madsen8, Michael Heidenheim9, Anne T. Funding10, Gitte Strauss11, Rune Lindberg12 and Lars Iversen1

Departments of 1Dermatology, 4Clinical Biochemistry, 6Hepatology and Gastroenterology and 7Paediatrics, Aarhus University Hospital, Aarhus, Departments of 2Dermatology and Allergy and 5Hepatology and Gastroenterology, Herlev and Gentofte Hospital, University of Copenhagen, Hellerup, Denmark, 3National Advisory Unit on Pregnancy and Rheumatic Diseases, Department of Rheumatology, Trondheim University Hospital, Trondheim, Norway, 8Department of Dermatology and Allergy Centre, Odense University Hospital, Institute of Clinical Research, University of Southern Denmark, Odense, 9Department of Dermatology, Copenhagen University Hospital, Roskilde, 10Dermatology Clinic, Hudlaegecenter Nord, Aalborg, 11Dermatology Clinic, Copenhagen, and 12Dermatology Clinic, Odense, Denmark

ABSTRACT

Methotrexate (MTX) has been used in the treatment of psoriasis and other dermatological diseases for more than 50 years. However, there is limited evidence regarding its effect, dose and monitoring, and a lack of consensus regarding how the drug should be used in daily practice. Although the use of MTX is governed by guidelines, such as the European S3-Guidelines and the National Institute for Health and Care Excellence (NICE) guideline, it is important to discuss and adjust these guidelines to national standards. An expert meeting was held in Denmark at the end of 2014, in order to reach consensus regarding the use of MTX in dermatological practice in Denmark. Participants included dermatologists, hepatologists, paediatricians, clinical biochemists and a rheumatologist. Topics discussed were: liver disease monitoring, teratogenic effects of MTX, risk of cancer, and use of MTX in children. We report here the conclusions of this expert meeting regarding use of MTX in dermatological practice.

Key words: psoriasis; skin diseases; methotrexate.

Accepted Dec 12, 2016; Epub ahead of print Dec 13, 2016

Acta Derm Venereol 2017; 97: XX–XX.

Corr: Claus Zachariae, Department of Dermatology and Allergy, Herlev and Gentofte Hospital, University of Copenhagen, DK-2900 Hellerup, Denmark. E-mail: claus.zachariae@regionh.dk; and Lars Iversen, Department of Dermatology, Aarhus University Hospital, DK-8000 Aarhus, Denmark. E-mail: lars.iversen@clin.au.dk

INTRODUCTION

Methotrexate (MTX) is a folic acid antagonist that was first used in the treatment of acute leukaemia in the early 1950s and subsequently for the treatment of solid tumours. Low-dose MTX has also been used successfully for the treatment of rheumatoid arthritis and psoriasis; and over the past 25 years, MTX has become the standard of care in the treatment of these 2 diseases (1–3). The effect of MTX was originally described as anti-proliferative, as the drug induces inhibition of purine, methionine and thymidylate synthesis, and thereby inhibits DNA synthesis. MTX is transported into the cells by either a folate carrier or by passive diffusion, and is polyglutamated once inside the cells (4). Whereas MTX has a half-life of 5–8 h, the MTX polyglutamates are retained in cells and tissues for several weeks or months (5). It was suggested that low-dose MTX treat-ment, e.g. the doses used in psoriasis, may also have anti-inflammatory effects, including increased adenosine levels, and MTX has been shown to modulate immune cells and to decrease the level of tumour necrosis factor alpha (TNFα), among other effects (3, 6).

DERMATOLOGICAL USE OF METHOTREXATE

MTX has been used in the treatment of moderate-to-severe psoriasis for many years; yet, the first randomized controlled trials were not performed until 2003. These studies compared MTX with cyclosporine (7, 8). MTX was later compared with different biologics in other randomized controlled trials (9–11). Even so, evidence for its effect remains limited. Yet, MTX is used for the treatment of a wide range of dermatological disorders, including pityriasis rubra pilaris, atopic dermatitis, chronic urticaria, pityriasis lichenoides, blistering disorders, cutaneous lupus erythematosus, localized scleroderma, vasculitis, cutaneous sarcoidosis, dermatomyositis and granuloma annulare (6). This report focuses on experience with MTX in the treatment of psoriasis.

For dermatological indications, MTX is used in low doses compared with those used in oncology. No studies have established the best starting or maintaining dose for dermatological indications (12), although a few studies have compared different doses of MTX in the treatment of psoriasis (13, 14). The general recommendation in psoriasis is to start at 5–15 mg once weekly, with dose escalation up to 25–30 mg weekly, depending on the clinical response (15, 16). The doses of MTX used for other dermatological diseases are similar to those used in psoriasis (6). Caution is recommended when treating elderly patients and patients with impaired kidney function (15).

According to the National Institute for Health and Care Excellence (NICE) guideline, the maximum treatment response is usually seen 16–24 weeks after treatment initiation, although maximal effect will occasionally be reached within 8–12 weeks of treatment with 15 mg weekly doses (16). Both the PASI75, which is equal to a 75% reduction in the skin manifestation of psoriasis, and patients’ quality of life improve significantly after treatment with MTX (17). In a randomized controlled trial, 62% of patients reached a PASI50 after 16 weeks of treatment, whereas 36% reached a PASI75. However, due to a low MTX starting dose (7.5 mg), these numbers are likely to be underestimated (9). This is supported by retrospective studies in which 40% and 62% of the patients achieved a PASI75 after 12 and 24 weeks of MTX treatment, respectively (18). In Denmark, it is usually recommended to evaluate the effect after 12 weeks of psoriasis treatment with the maximum tolerated dose of MTX (17).

In dermatology, MTX is administered orally or as a subcutaneous injection. In general, the oral administration route is the first choice owing to its simplicity and lower price. However, if there is lack of efficacy or unacceptable side-effects, it is recommended to change the administration route to subcutaneous injection, which can increase the efficacy and tolerability (19, 20).

Some of the most common side-effects of low-dose MTX, such as nausea, anorexia, fatigue and malaise, are very inconvenient for the patient, although not dangerous. These side-effects are dose-dependent and usually occur at initiation of therapy (6), and they may be reduced by folic acid supplementation (21). MTX can cause haematopoietic suppression, and haematopoietic biochemical monitoring is therefore recommended during treatment. MTX can also lead to pancytopaenia; however, this is a rare side-effect at the low-level doses used in dermatological diseases (22). The ability of MTX to induce liver toxicity is an important side-effect discussed in detail below.

Even though the efficacy of MTX is lower than the biological therapies used in psoriasis, MTX is the first-line systemic treatment for psoriasis in Denmark owing to its cost-effectiveness and the extensive experience in the use of this drug accrued over many years (23, 24). MTX is therefore used very frequently in dermatological settings; however, national guidelines on how to monitor MTX treatment are lacking. The aim of this paper is to provide recommendations regarding the use of MTX in a dermatological setting. Recommendations regarding the indication, dose and laboratory tests are summarized in Table I (15).


Table I. Recommendations regarding use of methotrexate (MTX)

METHOTREXATE AND LIVER MONITORING

MTX has been recognized as a hepatotoxic drug, which may limit its therapeutic use (25). Based on the literature and the discussion at our meeting, we suggest the approach summarized in Fig. 1 before and during treatment with MTX. Briefly, before treatment, liver enzymes and procollagen III aminopeptide (PIIINP) are tested; and if baseline values are elevated, fibroscan or change to another treatment is recommended. After treatment initiation, alanine aminotransferase (ALT) is measured after one week and then every second week for the first 2 months and then every 3 months. PIIINP is measured every 6 months and measurement is repeated within weeks if levels are elevated compared with baseline values. Further control is needed (Fig. 1) if ALT is increased by more than 1.5-fold or if PIIINP is elevated in 3 successive samples and cannot be explained by, for example, psoriatic arthritis.


Fig. 1. Algorithm for methotrexate (MTX) treatment and monitoring of liver toxicity and fibrosis. #Reference Limit (RL), method-specific. *Primary use of alanine aminotransferase (ALT) to monitor liver toxicity if procollagen III aminopeptide (PIIINP) is elevated due to, for example, psoriatic arthritis.

The pathogenesis of MTX hepatotoxicity includes hepatic folate depletion by MTX. Folate supplementation therefore results in a marked reduction in hepatotoxicity (26). In addition, polymorphisms in the gene encoding the MTHFR enzyme have been related to low-dose MTX-induced hepatotoxicity (27). The pathological changes in the liver following MTX are characterized by fat infiltration with subsequent inflammation and fibrosis and potentially development of cirrhosis. A number of risk factors associated with liver steatosis, inflammation and fibrosis may increase the risk of MTX-induced liver toxicity (Table II). These risk factors include obesity, dyslipidaemia and diabetes as part of the metabolic syndrome and are strongly associated with non-alcoholic fatty liver disease (NAFLD) including steatohepatitis (NASH) (28). Furthermore, excessive alcohol intake and other hepatotoxic drugs, e.g. non-steroidal anti-inflammatory drugs (NSAIDs), are important risk factors.


Table II. Risk factors for methotrexate (MTX)-induced liver toxicity

The incidence and prevalence of MTX toxicity is debated. However, there is little doubt that cumulative doses are associated with hepatotoxicity. In older studies, a worsening on Roenigk’s 5-grade histological classification scale was observed in 28% of patients; patients had a 7% risk of progressing at least one histological grade for each gram of MTX taken. The overall incidence of advanced pathological changes ascertained in liver biopsies (grades IIIB or IV) was 5% (29). In a recent meta-analysis of observational studies, a 22% increased risk of “any fibrosis” on biopsy was reported following MTX therapy. In this meta-analysis, the cumulative dose and duration of MTX therapy was not associated with biopsy-verified fibrosis or cirrhosis. Recent data also show that the risk of cirrhosis development with end-stage liver disease is quite limited, as determined by the very low number of patients listed for liver transplantation (30).

Liver biopsy is the gold standard for assessment of the severity of any liver disease, e.g. NASH or alcoholic liver disease with fibrosis, before MTX treatment is initiated, and may also be used during MTX treatment. However, performing a liver biopsy is associated with increased morbidity and mortality due to the risk of bleeding (31), and surrogate markers for liver disease are therefore often used. Most often, liver injury is assessed via the liver enzyme ALT, which is elevated in 7.5–26% of all patients treated with MTX depending on the cut-off level used (32). However, more specific methods are needed, in order to limit the number of liver biopsies and to evaluate the risk of liver fibrosis.

The PIIINP has been used in the assessment of liver fibrosis, as fibrogenesis results in the release of certain extracellular matrix proteins, including PIIINP, into the bloodstream. PIIINP is a molecule that derives from type III procollagen biosynthesis (33). Changes in PIIINP
levels are not specific to a particular disease, but reflect the involvement and altered metabolism of type III collagen depending on the activity and the extent of the tissue involved. Levels are high in children and adolescents as a result of physiological growth (34). In conditions where accumulation and/or degradation of connective tissue takes place, PIIINP levels are increased; and interpretation of the individual PIIINP values can be challenging because active joint involvement, smoking and other factors may lead to an increase in PIIINP levels that is unrelated to hepatic fibrosis (15). PIIINP is therefore less useful for monitoring of patients with psoriatic arthritis (35, 36), and therefore there is a need for further development of biomarkers.

A meta-analysis on assessments of liver toxicity in MTX-treated patients with psoriasis calculated that
PIIINP had a sensitivity of 77% and a specificity of 92%. Thus, PIIINP can be expected to have a good negative predictive value when the prevalence of severe fibrosis is expected to be low (37). In general, the risk of development of hepatic fibrosis is minimal if serial measurements of PIIINP are normal (35, 36).

The Enhanced Liver Fibrosis test (ELF™, Siemens Healthcare Diagnostic Inc., Tarrytown, NY, USA) was introduced recently as a non-invasive test of liver fibrosis. An ELF score is calculated using an algorithm based on analysis of PIIINP, hyaluronic acid and tissue inhibitor of matrix metalloproteinase-1 (TIMP-1). Studies indicate that the ELF test can be used to detect fibrosis in chronic liver diseases, such as primary biliary cirrhosis, NAFLD and chronic hepatitis C infection, and that the score has a higher diagnostic value than the individual tests alone (38–41). At present, only a single study has studied ELF as a non-invasive marker of MTX-induced hepatotoxicity (42). From this study it was concluded that ELF may be at least equivalent or possibly superior to PIIINP. The PIIINP assay that is part of the ELF test is different from the Orion Ercopharm PIIINP radioimmunoassay, which has been studied extensively. The Siemens PIIINP from the ELF test gives higher values due to a different standardization, but the values compare relatively well (43).

During the past decade other non-invasive tools, such as transient elastography and fibro test to monitor hepatotoxicity in MTX-treated patients with psoriasis, have been introduced, but large prospective studies are needed (44). Also fibroscan for liver fibrosis have become available and have proven effective in other fibrotic liver diseases (45). This has been evaluated in a few trials studying MTX-induced liver toxicity. In a study of patients with psoriasis only, the median fibroscan value was 6.4 kPa (range 3.3–18.4 kPa) and the fibroscan correctly identified 88% of the patients without significant liver fibrosis based on a liver biopsy (METAVIR score < F2, fibroscan ≤ 7.1 kPa) (46). However, fibroscan has yet to be validated for use in MTX-treated patients. Other scanning modalities are required and better biomarkers for liver disease severity and progression are needed.

Conclusion: Liver monitoring

MTX hepatotoxicity remains a clinical challenge in patients with psoriasis, and monitoring is important. The algorithm shown in Fig. 1 is recommended, and the importance of recognizing the increased risk of liver fibrosis in at-risk patient groups (Table II) is stressed. New imaging modalities and biomarkers for liver fibrosis provide the basis for non-invasive assessment of MTX liver toxicity.These tools will reduce the need for liver biopsies during follow-up of patients who receive MTX treatment.

METHOTREXATE AND PREGNANCY

MTX is embryotoxic in humans and animals (47). Different forms of embryopathies have been described after exposure during gestational weeks 5–8. The most severe malformation is “aminopterin syndrome” consisting of multiple central nervous system (CNS), skeletal and cardiac abnormalities. New prospective studies in patients with rheumatoid arthritis have shown no increase in spontaneous abortion or in congenital malformations when MTX was given 6–0 months before conception. However, when given at doses of 10–20 mg/week during the first trimester, a high rate of spontaneous abortion (20–40%) and a slight increase in congenital malformations with a variety of single and combined defects, as well as cardiac defects at high doses have been observed (48). Therefore, MTX must be discontinued in women 1–3 months before a planned pregnancy. Folic acid supplementation should be given to all patients of fertile age who are receiving MTX treatment, and supplementation should be continued during the first trimester even after withdrawal of MTX before a planned pregnancy.

In general, MTX at doses between 5 and 25 mg/week do not impair spermatogenesis. Several recent cohort studies investigating the outcome of pregnancies fathered by men who had taken MTX within 3 months before conception have failed to show any increase in congenital malformations in their children (49). A 3-month paternal MTX-free interval prior to conception does not appear to be necessary (50, 51). However, the drug information leaflet and the European S3-Guidelines still recommend a 3-month MTX-free interval before conception (15). The need for this interval should be considered on an individual basis.

Conclusion: Pregnancy and methotrexate

Low-dose MTX given for treatment of skin diseases can induce congenital malformations if administered during gestational weeks 5–8. MTX treatment during the first trimester may increase the rate of spontaneous abortion. MTX treatment must therefore be discontinued 3 months prior to planned pregnancy in females. There is no evidence to indicate that male fertility is impaired by standard doses of MTX; nor is there any evidence to support an increased rate of birth defects after paternal low-dose MTX intake around conception. However, the European S3-Guidelines still recommend a 3-month paternal MTX-free interval prior to conception, and it should therefore be considered on an individual basis.

METHOTREXATE AND CANCER

The risk of cancer in patients with immune-mediated inflammatory diseases has been debated in recent years. Chronic inflammation may have a pro-tumourigenic effect and immunosuppressive treatment may increase the risk, but, at the same time, inflammatory cells and cytokines may have an anti-tumour effect.

Studies of patients with psoriasis have shown an increased risk of some solid cancers, especially cancers related to the respiratory tract, urinary tract and liver cancer (52). These are cancers that have been related to alcohol consumption and smoking habits, which are more common in patients with psoriasis (53). The risk of lymphomas has been shown to be slightly increased in patients with psoriasis (54). Patients with psoriasis also have an increased risk of non-melanoma skin cancer, especially squamous cell carcinomas (SCC) (55). The increase in non-melanoma skin cancer has been associated with previous use of ultraviolet (UV) treatment, especially psoralen plus ultraviolet A (PUVA), but immunosuppressive treatment, such as cyclosporine, may also increase the risk (56–58). Furthermore, MTX has been associated with an increased cancer risk in 1 study (56). Whether MTX treatment alone increases the risk of cancer in patients with psoriasis is debated. There have been some cases of lymphoproliferative diseases in patients treated with MTX; however, most of these patients had rheumatoid arthritis, and only a few cases have been reported in patients with psoriasis (59).

MTX is also used for treatment of many types of cancers and, in line with acitretin, MTX is recommended as treatment for moderate-to-severe psoriasis in patients with a history of previous cancer.

One study in patients with psoriasis who had previously been treated with PUVA and subsequently with MTX showed an increased risk of SCC (58). No studies have found an increased risk of non-melanoma skin cancer following a combination of UVB treatment and MTX. However, it is generally recommended to routinely screen the skin for skin cancer during treatment with immunosuppressive drugs, including MTX. This should be done at least once a year.

Conclusion: Cancer and methotrexate

MTX clearly exhibits anti-tumourigenic effects, as demonstrated by its common use in treating different malignancies. However, based on the literature, it is not possible to conclude whether treatment with low-dose MTX in patients with psoriasis exhibits pro-tumourigenic effects. Owing to many years of experience with MTX treatment (compared with other treatments), we do not list previous cancer as a relative contraindication for MTX treatment in patients with psoriasis. However, patients with psoriasis have an increased risk of non-melanoma skin cancer, and annual screening of the skin for skin cancer is recommended when patients are being treated with all immunosuppressive drugs, including MTX.

METHOTREXATE USE IN CHILDREN

The evidence for use of MTX in paediatric patients with psoriasis is limited to case reports and observational studies. Furthermore, MTX is not registered for paediatric psoriasis, and thus has to be used off-label. In general, a good clinical effect is demonstrated. One randomized controlled study comparing adalimumab with MTX has been conducted; however, the data have yet to be published (60). The MTX doses used in psoriasis are 0.2–0.4 mg/kg/week (~6–12 mg/m2/week), although higher doses can be used (maximum 20 mg/week) (61, 62).

Most evidence regarding the use of low-dose MTX in paediatric patients is found for rheumatic indications, especially juvenile idiopathic arthritis (JIA), juvenile dermato-myositis (JDM) and localized scleroderma (LS). The recommendations regarding the administration, monitoring, adverse events and long-term effect of low-dose MTX treatment in paediatric patients are based primarily on experiences from MTX treatment in JIA as no recommendations have yet been published in children with psoriasis. In JIA, the dosage of MTX is usually 10–15 mg/m2/week (60, 63–67).

MTX in children may be administered orally or subcutaneously. Subcutaneous administration of MTX is more often preferred in children because it has fewer side-effects and allows administration of higher doses. Furthermore, the bioavailability is higher and more stable (68). In dermatology, it is common practice to monitor children via blood samples prior to initiation, weekly for 4 weeks after treatment start and then every other week up to week 8. When the paediatric patient is on a stable dose, laboratory tests should be similar to those recommended for adult patients (Table I) (69). However, use of PIIINP is difficult in paediatric patients because it increases during growth (70). Similar to adults, the adverse events most commonly associated with low-dose MTX treatment are nausea, vomiting and elevated liver enzymes (60). As in the treatment of adult patients, it is recommended to prescribe 5 mg folic acid (71).

In general, low-dose MTX is safe both in the short and the long term. It has not been found to affect growth nor fertility, nor has it been proven to be associated with malignancies (60, 72).

Conclusion: Methotrexate use in children

The evidence regarding the use and safety of low-dose MTX in paediatric patients is limited and based mostly on JIA studies. In general, doses of 0.2–0.4 mg/kg/week for the treatment of psoriasis and close monitoring are recommended in paediatric patients with psoriasis. Treat-ment with low-dose MTX is effective and seems not to affect growth or fertility or to be associated with an increased risk of malignancies.

CONCLUSION

This report was based on an expert meeting. It is hoped that it will provide further consensus regarding the use and monitoring of MTX treatment in patients with psoriasis in Denmark and other countries. In general, the recommendations given above are in line with the European S3-Guidelines.

The authors declare no conflicts of interest.

REFERENCES
  1. Meyer LM, Miller FR, Rowen MJ, Bock G, Rutzky J. Treatment of acute leukemia with amethopterin (4-amino, 10-methyl pteroyl glutamic acid). Acta Haematol 1950; 4: 157–167.
    View article    Google Scholar
  2. Gubner R, August S, Ginsberg V. Therapeutic suppression of tissue reactivity. II. Effect of aminopterin in rheumatoid arthritis and psoriasis. Am J Med Sci 1951; 221: 176–182.
    View article    Google Scholar
  3. Cipriani P, Ruscitti P, Carubbi F, Liakouli V, Giacomelli R. Methotrexate: an old new drug in autoimmune disease. Expert Rev Clin Immunol 2014; 10: 1519–1530.
    View article    Google Scholar
  4. Grim J, Chladek J, Martinkova J. Pharmacokinetics and pharmacodynamics of methotrexate in non-neoplastic diseases. Clin Pharmacokinet 2003; 42: 139–151.
    View article    Google Scholar
  5. Bannwarth B, Pehourcq F, Schaeverbeke T, Dehais J. Clinical pharmacokinetics of low-dose pulse methotrexate in rheumatoid arthritis. Clin Pharmacokinet 1996; 30: 194–210.
    View article    Google Scholar
  6. Shen S, O’Brien T, Yap LM, Prince HM, McCormack CJ. The use of methotrexate in dermatology: a review. Australas J Dermatol 2012; 53: 1–18.
    View article    Google Scholar
  7. Heydendael VMSP, Opmeer BC, de Borgie CA, Reitsma JB, Goldschmidt WF, Bossuyt PM, et al. Methotrexate versus cyclosporine in moderate-to-severe chronic plaque psoriasis. N Engl J Med 2003; 349: 658–665.
    View article    Google Scholar
  8. Sandhu K, Kaur I, Kumar B, Saraswat A. Efficacy and safety of cyclosporine versus methotrexate in severe psoriasis: a study from north India. J Dermatol 2003; 30: 458–463.
    View article    Google Scholar
  9. Saurat JH, Stingl G, Dubertret L, Papp K, Langley RG, Ortonne JP, et al. Efficacy and safety results from the randomized controlled comparative study of adalimumab vs. methotrexate vs. placebo in patients with psoriasis (CHAMPION). Br J Dermatol 2008; 158: 558–566.
    View article    Google Scholar
  10. Barker J, Hoffmann M, Wozel G, Ortonne JP, Zheng H, van Hoogstraten H, et al. Efficacy and safety of infliximab vs. methotrexate in patients with moderate-to-severe plaque psoriasis: results of an open-label, active-controlled, randomized trial (RESTORE1). Br J Dermatol 2011; 165: 1109–1117.
    View article    Google Scholar
  11. Reich K, Langley RG, Papp KA, Ortonne JP, Unnebrink K, Kaul M, et al. A 52-week trial comparing briakinumab with methotrexate in patients with psoriasis. N Engl J Med 2011; 365: 1586–1596.
    View article    Google Scholar
  12. Menting SP, Dekker PM, Limpens J, Hooft L, Spuls PI. Methotrexate dosing regimen for plaque-type psoriasis: a systematic review of the use of test-dose, start-dose, dosing scheme, dose adjustments, maximum dose and folic acid supplementation. Acta Derm Venereol 2016; 96: 23–28.
    View article    Google Scholar
  13. Chladek J, Grim J, Martinkova J, Simkova M, Vaniekova J, Koudelkova V, et al. Pharmacokinetics and pharmacodynamics of low-dose methotrexate in the treatment of psoriasis. Br J Clin Pharmacol 2002; 54: 147–156.
    View article    Google Scholar
  14. Dogra S, Krishna V, Kanwar AJ. Efficacy and safety of systemic methotrexate in two fixed doses of 10 mg or 25 mg orally once weekly in adult patients with severe plaque-type psoriasis: a prospective, randomized, double-blind, dose-ranging study. Clin Exp Dermatol 2012; 37: 729–734.
    View article    Google Scholar
  15. Nast A, Gisondi P, Ormerod AD, Saiag P, Smith C, Spuls PI, et al. European S3-Guidelines on the systemic treatment of psoriasis vulgaris – Update 2015 – Short version – EDF in cooperation with EADV and IPC. J Eur Acad Dermatol Venereol 2015; 29: 2277–2294.
    View article    Google Scholar
  16. NICE. Psoriasis: assessment and management of psoriasis. NICE guidelines [CG153]. London: Royal College of Physicians (UK) CTI – National Institute for Health and Clinical Excellence: Guidance; 2012 [updated 20141024]. Available from: http://www.nice.org.uk/guidance/cg153/evidence/full-guideline-188351533.
    View article    Google Scholar
  17. SST. National klinisk retningslinje for psoriasis. Copenhagen: The Danish Health Authority; 2016 [updated 2016-03-16; cited 2016]. Available from: https://sundhedsstyrelsen.dk/da/udgivelser/2016/nkr-psoriasis.
    View article    Google Scholar
  18. Inzinger M, Weger W, Heschl B, Salmhofer W, Quehenberger F, Wolf P. Methotrexate vs. fumaric acid esters in moderate-to-severe chronic plaque psoriasis: data registry report on the efficacy under daily life conditions. J Eur Acad Dermatol Venereol 2013; 27: 861–866.
    View article    Google Scholar
  19. Hazlewood GS, Thorne JC, Pope JE, Lin D, Tin D, Boire G, et al. The comparative effectiveness of oral versus subcutaneous methotrexate for the treatment of early rheumatoid arthritis. Ann Rheum Dis 2015; 75: 1003–1008.
    View article    Google Scholar
  20. Wegrzyn J, Adeleine P, Miossec P. Better efficacy of methotrexate given by intramuscular injection than orally in patients with rheumatoid arthritis. Ann Rheum Dis 2004; 63: 1232–1234.
    View article    Google Scholar
  21. Shea B, Swinden MV, Tanjong Ghogomu E, Ortiz Z, Katchamart W, Rader T, et al. Folic acid and folinic acid for reducing side effects in patients receiving methotrexate for rheumatoid arthritis. Cochrane Database Syst Rev 2013; 5: Cd000951.
    View article    Google Scholar
  22. Van Dooren-Greebe RJ, Kuijpers AL, Mulder J, De Boo T, Van de Kerkhof PC. Methotrexate revisited: effects of long-term treatment in psoriasis. Br J Dermatol 1994; 130: 204–210.
    View article    Google Scholar
  23. Kuster D, Nast A, Gerdes S, Weberschock T, Wozel G, Gutknecht M, et al. Cost-effectiveness of systemic treatments for moderate-to-severe psoriasis in the German health care setting. Arch Dermatol Res 2016; 308: 249–261.
    View article    Google Scholar
  24. DDS. Retningslinjer for behandling af psoriasis med 2. generations immunomodulatorisk behandling 2014. Available from: http://www.dds.nu/wp-content/uploads/2012/08/Dansk-Dermatologisk-Selskabs-retningslinjer-for-behandling-af-psoriasis-med-2.-generations-immunomodulatorisk-behandling.pdf.
    View article    Google Scholar
  25. Maybury CM, Jabbar-Lopez ZK, Wong T, Dhillon AP, Barker JN, Smith CH. Methotrexate and liver fibrosis in people with psoriasis: a systematic review of observational studies. Br J Dermatol 2014; 171: 17–29.
    View article    Google Scholar
  26. Bath RK, Brar NK, Forouhar FA, Wu GY. A review of methotrexate-associated hepatotoxicity. J Dig Dis 2014; 15: 517–524.
    View article    Google Scholar
  27. Davila-Fajardo CL, Swen JJ, Cabeza Barrera J, Guchelaar HJ. Genetic risk factors for drug-induced liver injury in rheumatoid arthritis patients using low-dose methotrexate. Pharmacogenomics 2013; 14: 63–73.
    View article    Google Scholar
  28. Rosenberg P, Urwitz H, Johannesson A, Ros AM, Lindholm J, Kinnman N, et al. Psoriasis patients with diabetes type 2 are at high risk of developing liver fibrosis during methotrexate treatment. J Hepatol 2007; 46: 1111–1118.
    View article    Google Scholar
  29. Whiting-O’Keefe QE, Fye KH, Sack KD. Methotrexate and histologic hepatic abnormalities: a meta-analysis. Am J Med 1991; 90: 711–716.
    View article    Google Scholar
  30. Dawwas MF, Aithal GP. End-stage methotrexate-related liver disease is rare and associated with features of the metabolic syndrome. Aliment Pharmacol Ther 2014; 40: 938–948.
    View article    Google Scholar
  31. Ott P. [Percutaneous liver biopsy]. Ugeskr Laeger 2003; 165: 1571–1573 (in Danish).
    View article    Google Scholar
  32. Sotoudehmanesh R, Anvari B, Akhlaghi M, Shahraeeni S, Kolahdoozan S. Methotrexate hepatotoxicity in patients with rheumatoid arthritis. Middle East J Dig Dis 2010; 2: 104–109.
    View article    Google Scholar
  33. Risteli J, Sogaard H, Oikarinen A, Risteli L, Karvonen J, Zachariae H. Aminoterminal propeptide of type III procollagen in methotrexate-induced liver fibrosis and cirrhosis. Br J Dermatol 1988; 119: 321–325.
    View article    Google Scholar
  34. Trivedi P, Hindmarsh P, Risteli J, Risteli L, Mowat AP, Brook CG. Growth velocity, growth hormone therapy, and serum concentrations of the amino-terminal propeptide of type III procollagen. J Pediatr 1989; 114: 225–230.
    View article    Google Scholar
  35. Lindsay K, Fraser AD, Layton A, Goodfield M, Gruss H, Gough A. Liver fibrosis in patients with psoriasis and psoriatic arthritis on long-term, high cumulative dose methotrexate therapy. Rheumatology (Oxford) 2009; 48: 569–572.
    View article    Google Scholar
  36. Zachariae H, Heickendorff L, Sogaard H. The value of amino-terminal propeptide of type III procollagen in routine screening for methotrexate-induced liver fibrosis: a 10-year follow-up. Br J Dermatol 2001; 144: 100–103.
    View article    Google Scholar
  37. Montaudie H, Sbidian E, Paul C, Maza A, Gallini A, Aractingi S, et al. Methotrexate in psoriasis: a systematic review of treatment modalities, incidence, risk factors and monitoring of liver toxicity. J Eur Acad Dermatol Venereol 2011; 25 Suppl 2: 12–18.
    View article    Google Scholar
  38. Guha IN, Parkes J, Roderick P, Chattopadhyay D, Cross R, Harris S, et al. Noninvasive markers of fibrosis in nonalcoholic fatty liver disease: validating the European Liver Fibrosis Panel and exploring simple markers. Hepatology 2008; 47: 455–460.
    View article    Google Scholar
  39. Lichtinghagen R, Pietsch D, Bantel H, Manns MP, Brand K, Bahr MJ. The Enhanced Liver Fibrosis (ELF) score: normal values, influence factors and proposed cut-off values. J Hepatol 2013; 59: 236–242.
    View article    Google Scholar
  40. Parkes J, Guha IN, Roderick P, Harris S, Cross R, Manos MM, et al. Enhanced Liver Fibrosis (ELF) test accurately identifies liver fibrosis in patients with chronic hepatitis C. J Viral Hepat 2011; 18: 23–31.
    View article    Google Scholar
  41. Rosenberg WM, Voelker M, Thiel R, Becka M, Burt A, Schuppan D, et al. Serum markers detect the presence of liver fibrosis: a cohort study. Gastroenterology 2004; 127: 1704–1713.
    View article    Google Scholar
  42. Martyn-Simmons CL, Rosenberg WM, Cross R, Wong T, Smith CH, Barker JN. Validity of noninvasive markers of methotrexate-induced hepatotoxicity: a retrospective cohort study. Br J Dermatol 2014; 171: 267–273.
    View article    Google Scholar
  43. Knudsen CS, Heickendorff L, Nexo E. Measurement of amino terminal propeptide of type III procollagen (PIIINP) employing the ADVIA Centaur platform. Validation, reference interval and comparison to UniQ RIA. Clin Chem Lab Med 2014; 52: 237–241.
    View article    Google Scholar
  44. Lynch M, Higgins E, McCormick PA, Kirby B, Nolan N, Rogers S, et al. The use of transient elastography and FibroTest for monitoring hepatotoxicity in patients receiving methotrexate for psoriasis. JAMA Dermatol 2014; 150: 856–862.
    View article    Google Scholar
  45. Wilder J, Patel K. The clinical utility of FibroScan((R)) as a noninvasive diagnostic test for liver disease. Med Devices (Auckl) 2014; 7: 107–114.
    View article    Google Scholar
  46. Berends MA, Snoek J, de Jong EM, Van Krieken JH, de Knegt RJ, van Oijen MG, et al. Biochemical and biophysical assessment of MTX-induced liver fibrosis in psoriasis patients: Fibrotest predicts the presence and Fibroscan predicts the absence of significant liver fibrosis. Liver Int 2007; 27: 639–645.
    View article    Google Scholar
  47. Ostensen M, Hartmann H, Salvesen K. Low dose weekly methotrexate in early pregnancy. A case series and review of the literature. J Rheumatol 2000; 27: 194–210.
    View article    Google Scholar
  48. Weber-Schoendorfer C, Chambers C, Wacker E, Beghin D, Bernard N, Network of French Pharmacovigilance C, et al. Pregnancy outcome after methotrexate treatment for rheumatic disease prior to or during early pregnancy: a prospective multicenter cohort study. Arthritis Rheumatol 2014; 66: 1101–1110.
    View article    Google Scholar
  49. Weber-Schoendorfer C, Hoeltzenbein M, Wacker E, Meister R, Schaefer C. No evidence for an increased risk of adverse pregnancy outcome after paternal low-dose methotrexate: an observational cohort study. Rheumatology (Oxford) 2014; 53: 757–763.
    View article    Google Scholar
  50. Wallenius M, Lie E, Daltveit AK, Salvesen KA, Skomsvoll JF, Kalstad S, et al. No excess risks in offspring with paternal preconception exposure to disease-modifying antirheumatic drugs. Arthritis Rheumatol 2015; 67: 296–301.
    View article    Google Scholar
  51. Viktil KK, Engeland A, Furu K. Outcomes after anti-rheumatic drug use before and during pregnancy: a cohort study among 150,000 pregnant women and expectant fathers. Scand J Rheumatol 2012; 41: 196–201.
    View article    Google Scholar
  52. Pouplard C, Brenaut E, Horreau C, Barnetche T, Misery L, Richard MA, et al. Risk of cancer in psoriasis: a systematic review and meta-analysis of epidemiological studies. J Eur Acad Dermatol Venereol 2013; 27 Suppl 3: 36–46.
    View article    Google Scholar
  53. Hayes J, Koo J. Psoriasis: depression, anxiety, smoking, and drinking habits. Dermatol Ther 2010; 23: 174–180.
    View article    Google Scholar
  54. Gelfand JM, Shin DB, Neimann AL, Wang X, Margolis DJ, Troxel AB. The risk of lymphoma in patients with psoriasis. J Invest Dermatol 2006; 126: 2194–2201.
    View article    Google Scholar
  55. Hannuksela-Svahn A, Pukkala E, Laara E, Poikolainen K, Karvonen J. Psoriasis, its treatment, and cancer in a cohort of Finnish patients. J Invest Dermatol 2000; 114: 587–590.
    View article    Google Scholar
  56. Patel RV, Clark LN, Lebwohl M, Weinberg JM. Treatments for psoriasis and the risk of malignancy. J Am Acad Dermatol 2009; 60: 1001–1017.
    View article    Google Scholar
  57. Marcil I, Stern RS. Squamous-cell cancer of the skin in patients given PUVA and ciclosporin: nested cohort crossover study. The Lancet 2001; 358: 1042–1045.
    View article    Google Scholar
  58. Stern RS, Study PF-U. The risk of squamous cell and basal cell cancer associated with psoralen and ultraviolet A
    View article    Google Scholar
  59. therapy: a 30-year prospective study. J Am Acad Dermatol 2012; 66: 553–562.
    View article    Google Scholar
  60. Paul C LTA, Cayuela JM, Devidas A, Robert C, Molinié V, Dubertret L. Epstein-Barr virus-associated lymphoproliferative disease during methotrexate therapy for psoriasis. Arch Dermatol 1997; 133: 867–871.
    View article    Google Scholar
  61. Hashkes PJ, Becker ML, Cabral DA, Laxer RM, Paller AS, Rabinovich CE, et al. Methotrexate: new uses for an old drug. J Pediatr 2014; 164: 231–236.
    View article    Google Scholar
  62. Bertelsen T, Iversen L. Systemic treatment of psoriasis in children. J Clin Exp Dermatol Res 2015; 6: 313.
    View article    Google Scholar
  63. van Geel MJ, Mul K, de Jager ME, van de Kerkhof PC, de Jong EM, Seyger MM. Systemic treatments in paediatric psoriasis: a systematic evidence-based update. J Eur Acad Dermatol Venereol 2015; 29: 425–437.
    View article    Google Scholar
  64. Giannini EH BE, Kuzmina N, Shaikov A, Maximov A, Vorontsov I, Fink CW, Newman AJ, Cassidy JT, Zemel LS. Methotrexate in resistant juvenile rheumatoid arthritis. Results of the U.S.A.-U.S.S.R. double-blind, placebo-controlled trial. The Pediatric Rheumatology Collaborative Study Group and The Cooperative Children’s Study. N Engl J Med 1992; 326: 1043–1049.
    View article    Google Scholar
  65. Kremer JM. Still trying to understand methotrexate. J Rheumatol 2014; 41: 2009–2101.
    View article    Google Scholar
  66. Petty RE ST, Manners P, Baum J, Glass DN, Goldenberg J, He X, Maldonado-Cocco J, et al. International League of Associations for Rheumatology classification of juvenile idiopathic arthritis: second revision, Edmonton, 2001. J Rheumatol 2004; 31: 390–392.
    View article    Google Scholar
  67. Ruperto N, Murray KJ, Gerloni V, Wulffraat N, de Oliveira SK, Falcini F, et al. A randomized trial of parenteral methotrexate comparing an intermediate dose with a higher dose in children with juvenile idiopathic arthritis who failed to respond to standard doses of methotrexate. Arthritis Rheum 2004; 50: 2191–2201.
    View article    Google Scholar
  68. Vilca I, Munitis PG, Pistorio A, Ravelli A, Buoncompagni A, Bica B, et al. Predictors of poor response to methotrexate in polyarticular-course juvenile idiopathic arthritis: analysis of the PRINTO methotrexate trial. Ann Rheum Dis 2010; 69: 1479–1483.
    View article    Google Scholar
  69. Wallace CA, Giannini EH, Spalding SJ, Hashkes PJ, O’Neil KM, Zeft AS, et al. Trial of early aggressive therapy in polyarticular juvenile idiopathic arthritis. Arthritis Rheum 2012; 64: 2012–2021.
    View article    Google Scholar
  70. Beukelman T, Patkar NM, Saag KG, Tolleson-Rinehart S, Cron RQ, DeWitt EM, et al. 2011 American College of Rheumatology recommendations for the treatment of juvenile idiopathic arthritis: initiation and safety monitoring of therapeutic agents for the treatment of arthritis and systemic features. Arthritis Care Res (Hoboken) 2011; 63: 465–482.
    View article    Google Scholar
  71. Trivedi P, Cheeseman P, Portmann B, Hegarty J, Mowat AP. Variation in serum type III procollagen peptide with age in healthy subjects and its comparative value in the assessment of disease activity in children and adults with chronic active hepatitis. Eur J Clin Invest 1985; 15: 69–74.
    View article    Google Scholar
  72. Ravelli A, Migliavacca D, Viola S, Ruperto N, Pistorio A, Martini A. Efficacy of folinic acid in reducing methotrexate toxicity in juvenile idiopathic arthritis. Clin Exp Rheumatol 1999; 17: 625–627.
    View article    Google Scholar
  73. Beukelman T, Haynes K, Curtis JR, Xie F, Chen L, Bemrich-Stolz CJ, et al. Rates of malignancy associated with juvenile idiopathic arthritis and its treatment. Arthritis Rheum 2012; 64: 1263–1271.
    View article    Google Scholar