8,373
Views
92
CrossRef citations to date
0
Altmetric
Review Article

Japan College of Rheumatology guideline for the use of methotrexate in patients with rheumatoid arthritis

, , , , , , , , & show all
Pages 31-40 | Received 16 Apr 2018, Accepted 25 Apr 2018, Published online: 24 May 2018

Abstract

Methotrexate (MTX), the anchor drug in the current treatment strategy for rheumatoid arthritis (RA), was first approved for treatment of RA in Japan in 1999 at the recommended dose of 6–8 mg/week; it was approved as first-line drug with the maximum dose of 16 mg/week in February 2011. However, more than half of Japanese patients with RA are unable to tolerate a dose of 16 mg/week of MTX. Moreover, some serious adverse events during the treatment with MTX, such as pneumocystis pneumonia (PCP) and lymphoproliferative disorders (LPD) have been observed much more frequently in Japan than in other countries. Therefore, this article, an abridged English translation summarizing the 2016 update of the Japan College of Rheumatology (JCR) guideline for the use of MTX in Japanese patients with RA, is not intended to be valid for global use; however, it is helpful for the Japanese community of rheumatology and its understanding might be useful to the global community of rheumatology.

Introduction

Methotrexate (MTX) is the anchor drug in current treatment strategy for rheumatoid arthritis (RA) [Citation1,Citation2]. In Japan, MTX was first approved for refractory RA in adults in 1999 at the recommended dose of 6–8 mg/week. In February 2011, MTX was approved for adult RA with the maximum dose up to 16 mg/week, and as first-line conventional synthetic disease-modifying anti-rheumatic drug (csDMARD). The Japan College of Rheumatology (JCR) subcommittee on the guideline for the use of MTX in patients with RA published the first Japanese version of the guideline in 2011 and updated it in 2016. Since the adverse events during the treatment with MTX in Japan are rather serious and unique, such as pneumocystis pneumonia (PCP) and lymphoproliferative disorders (LPD), and its maximum dose is limited, it is important to publish the guideline in English for better understanding of the use of MTX for RA in Japan. This article is an abridged English translation summarizing the 2016 update of the JCR guideline for the use of MTX in Japanese patients with RA with a minimal update, and it is not intended to be valid for global use.

Indications, contraindications, and precautions

The paradigm of ‘Treat to Target (T2T)’ [Citation1], in which long-term health-related quality of life is maximized by estimating disease activity and adjusting the therapy accordingly until reaching the target, has been widely accepted, and the first-line use of MTX as the anchor drug for the treatment of RA has been recommended [Citation2]. The evidences from clinical trials on Japanese patients with poorly prognostic RA treated with first-line MTX therapy have been accumulated [Citation3,Citation4]. Therefore, MTX should be considered as the first choice among csDMARDs to maintain a balance between risk factors associated with MTX treatment, such as advanced age and comorbidities and benefits obtained by prompt control of disease activity (). In addition, MTX should be used as far as possible in patients who are likely to develop a functional disorder or its progression despite optimal dose of other csDMARDs for 2–3 months.

Table 1. 2016 Update of the JCR recommendations for the use of MTX in patients with RA.

Contraindications for MTX include severe hematological and lymphatic disorders, namely, myelodysplasia, aplastic anemia, pure red cell aplasia, LPD within recent 5 years, and severe leukopenia or thrombocytopenia (white blood cell [WBC] count <3000/uL, platelet count <50,000/uL) [Citation5]; hepatic disorders, namely, acute or chronic active viral hepatitis B or C, and liver cirrhosis; severe renal disorders, including end-stage kidney disorder requiring dialysis with estimated glomerular filtration rate (eGFR) < 30 mL/min/1.73 m2; and severe respiratory disorders diagnosed with hypoxemia (PaO2 < 70 Torr in room air), % vital capacity <80% as estimated using lung function test, or severe pulmonary fibrosis as seen on the chest radiographs.

MTX should be administered in elderly patients with particular caution. Patients with infectious diseases or those susceptible to them may need appropriate vaccination and/or prophylactic medications. Patients with mild leukopenia (3000/μL ≤ WBC <4000/μL), thrombocytopenia (50,000/μL ≤ platelet <100,000/μL) or with a history of drug-induced bone marrow disorder should receive MTX therapy along with folic acid supplementation. Therapies other than MTX should be fully considered for patients with a history of LPD. A reduced dosing of MTX with folic acid supplementation should be considered for patients with hypoalbuminemia (<3 g/dL), because those are susceptible to dose-dependent adverse events, such as pancytopenia due to delayed clearance of MTX [Citation6]. Patients with habitual drinking should be instructed to avoid alcohol as much as possible. Patients who show a positive test for hepatitis B virus (HBV) surface antigen (HBs-Ag) or HBV-DNA should receive anti-viral therapy prior to starting the MTX treatment after consulting with a hepatology/gastroenterology expert [Citation7]. Patients who show a positive test result for hepatitis C virus (HCV) antibodies should be referred to a hepatology/gastroenterology expert to discuss the risk-benefit from MTX treatment. When the levels of liver transaminases (aspartate aminotransferase [AST] or alanine aminotransferase [ALT]) and alkaline phosphatase (ALP) are greater than 2-fold of the upper limit of normal without any positive tests for hepatitis viruses, intensive evaluation should be carried out before the commencement of MTX therapy, and if needed, MTX should be initiated at a lower dose along with folic acid supplementation. Renal function should be evaluated by estimating eGFR, creatinine clearance, or cystatin C, and in the patients with renal dysfunction (eGFR <60 mL/min/1.73 m2 or equivalent), the therapy should be started with a reduced dose of MTX along with folic acid supplementation. A consultation with a respiratory expert should be considered for patients whose chest X-rays show possible signs of interstitial pneumonia, chronic obstructive pulmonary disease (COPD) or non-tuberculous mycobacterium (NTM) infection.

The dosing of methotrexate

Based on the global evidences on the use of MTX, it has been recommended that the initial optimal dose of MTX should be 10–15 mg/week [Citation8,Citation9], and this has been confirmed by subsequent systematic literature reviews [Citation10,Citation11]. The comparison among 2, 6, and 9 mg/week doses of MTX in terms of efficacy and safety was performed to determine the optimal clinical dose of MTX in Japanese patients with RA [Citation12]. It was observed that the dose of 2 mg/week of MTX was less effective than other doses, whereas, 9 mg/week of MTX showed numerically greater, but statistically insignificant improvement in some clinical parameters compared with 6 mg/week of MTX. However, leukopenia and liver dysfunction were more frequently observed in patients receiving 9 mg/week of MTX than in those receiving 6 mg/week, which led to the maximum approved dose of MTX as 8 mg/week for patients with RA in 1999. After the approval of 16 mg/week as the maximum dose in 2011, the C-OPERA trial was conducted, in which MTX therapy was started with the dose of 8 mg/week and was escalated by 4 mg at every 4th week up to 16 mg/week by week 8 [4]. As a result, the efficacy and safety of protocol starting from 8 mg/week of MTX was confirmed in Japan. Clinical trials for comparing the effectiveness of parenteral MTX up to 10 versus 25 mg/week [Citation13] and oral MTX up to 10 versus 25 mg/week as the initial dose [Citation14] showed that MTX >15 mg/week is not needed as a starting dose of the therapy. In addition, the dose of MTX has been escalated from 7.5 mg/week to the maximum until week 8 in the escalation-protocols of many clinical trials on biological DMARDs (bDMARDs) versus MTX [Citation15,Citation16]. With respect of the T2T strategy aiming at the achievement of therapeutic goal within 6 months [Citation1], 6–8 mg/week of MTX is appropriate for Japanese patients as the initial dose of the MTX therapy. For non-elderly patients with high disease activity and poor-prognostic factors or refractory arthritis, starting MTX therapy with the dose of 8 mg/week is recommended.

A French guideline on the clinical use of MTX has recommended a dose-escalation at the interval of 6 weeks in patients showing an inadequate response to MTX [Citation8]. The clinical response rate in patients receiving MTX is 20–30% at week 4, which corresponds to approximately a half of the total response rate [Citation17]. On the other hand, tight control for rheumatoid arthritis (TICORA) [Citation18] and Computer-Assisted Management in Early Rheumatoid Arthritis (CAMERA) [Citation19] studies examining the T2T strategy demonstrated higher remission rate and less radiographic progression in patients undergoing therapeutic adjustment at every 4th week as compared to those evaluated conventionally at every 12th week. Based on the tolerability of the dose-escalation protocol for MTX from the initial 7.5–8 mg/week up to 16–20 mg/week within 6–8 weeks in Japan and other countries [Citation4,Citation15,Citation16], patients should be assessed every 2–4 weeks after the commencement or dose-escalation of MTX, followed by performing the dose-escalation, if necessary and tolerable, up to the maximum targeted dose within 8–12 weeks. Although the dose of MTX may be escalated up to 16 mg/week, the primarily targeted dose is 10–12 mg/week, because some clinical parameters have been shown to reach the plateau above 15 mg/week [Citation20], and parenteral MTX showed better efficacy and fewer adverse gastrointestinal events than oral MTX at the dose above 15 mg/week [Citation21]. The C-OPERA study in Japan was conducted with the protocol of rapid dose-escalation up to 16 mg/week by 8th week. However, the mean MTX dose during the 1st year was 11.6 mg/week (median 11.9 mg/week) and 10.5 mg/week (median 12.0 mg/week) by 1st year, and only ∼30% of patients were tolerable to 16 mg/week of MTX [Citation4]. On the other hand, a post-marketing surveillance of dose of MTX over 8 mg/week demonstrated that dose-escalation to >8 mg/week resulted in an increase in the remission rate from 10.5 to 32.5% in 24 weeks, and from 16.9 to 47.1% in week 52 [Citation22]. Therefore, the dose-escalation up to 10–12 mg/week of MTX has been recommended in patients showing inadequate response to 6–8 mg/week of MTX. After reaching a dose of 10–12 mg/week of MTX, a few options are available according to the risk-benefit in each patient: further dose-escalation of MTX to 16 mg/week or the addition of other csDMARDs, bDMARDs, or targeted synthetic DMARDs (tsDMARDs) to MTX therapy.

Although single dosing regimen is simple and globally popular for MTX therapy for RA, the bioavailability of oral MTX is not comparable between single and divided dosing [Citation23–26]. Subcutaneous MTX is more effective than oral administration of 15 mg/week of MTX [Citation27]. Furthermore, the bioavailability of subcutaneous MTX is higher than that of oral MTX by 25% even for the dose of 10 mg/week, and much more for the dose greater than 15 mg/week [Citation28]. Thus, the fact that 10–15 mg/week dose of oral MTX is less effective than that of subcutaneous MTX may be explained by the differences in the bioavailability of the respective forms of MTX. In addition, it is reported that bioavailability of MTX is similar between single and divided dosing for the dose of 8 mg/week or less, while the dose of 25–35 mg/week of MTX has better bioavailability in divided dosing than that in single dosing [Citation29]. Subcutaneous MTX, which is recommended for patients who are unable to tolerate oral MTX or in whom oral MTX is less effective [Citation30], has not yet been approved for RA in Japan. Therefore, divided dosing every 12 h for 1–2 d may be beneficial for using more than 8 mg/week of MTX in terms of less GI toxicity and higher bioavailability. In fact, a post-marketing surveillance of the MTX dose over 8 mg/week in Japan revealed that approximately two-thirds of patients received 3-divided dosing for 2 d, and approximately 25% of patients received 2-divided dosing within a day, while single dose administration was observed in 4% of patients. Therefore, a dose ≤8 mg/week of MTX may be administered either as a single dose or as 2–3 divided doses for 1–2 d, while a dose >8 mg/week of MTX should be received 2–4 divided doses into every 12 h for 1–2 d.

Methotrexate combined with other disease-modifying anti-rheumatic drugs

MTX is the anchor drug, which is effective with concomitant csDMARDs in patients showing inadequate response to MTX monotherapy (MTX-IR or step-up), and in those with severe RA (parallel or step-down) [Citation1,Citation31–33]. Although cyclosporin A (CyA) has not been approved for RA in Japan, MTX + CyA has been reported to be effective in early RA and MTX-IR patients [Citation34,Citation35]. MTX + salazosulfapyridine (SASP) has been reported to be effective in early RA and SASP-IR patients [Citation36–38], although the difference in effectiveness between MTX + SASP therapy and MTX monotherapy was modest [Citation39]. The addition of leflunomide (LEF) [Citation40], gold sodium thiomalate (GST) [Citation41], tacrolimus (TAC) [Citation42,Citation43], as well as iguratimode (IGU: a csDMARD suppressing the production of immunoglobulins and inflammatory cytokines through the inhibition of nuclear factor-kappa B [NF-κB] activity) [Citation44,Citation45] to the MTX monotherapy has been effective in MTX-IR patients, although the use of GST is currently very limited in Japan. MTX + LEF therapy should be considered with caution in Japan where interstitial pneumonia associated with LEF is of serious concern [Citation46], in addition to its additive immunosuppressive mode of action and accompanied profiles of adverse events, such as interstitial pneumonia, cytopenia, and liver dysfunction. MTX + bucillamine (BUC: a csDMARD derived from d-penicillamine), has shown a superior American College of Rheumatology 20% (ACR20) response rate and inhibition of radiographic progression as compared to BUC monotherapy or MTX monotherapy in MTX-naïve patients with early RA [Citation47].

A systematic literature review of 51 full papers and 57 abstracts demonstrated that bDMARD and MTX combination therapy with all classes of bDMARD is more efficacious than bDMARD monotherapy, despite some additional evidence on the use of bDMARD monotherapy [Citation48]. Most of the clinical trials involving MTX + anti-tumor necrosis factor (TNF) bDMARDs, such as etanercept (ETN; TEMPO [Citation49], and JESMR [Citation50]), adalimumab (ADA; PREMIER [Citation51]), and golimumab (GLM; GO-BEFORE [Citation52] and GO-FORWARD [Citation53]) have shown superior clinical response rates, remission rates, improvement in health assessment questionnaire-disability index (HAQ-DI), and the inhibition of radiographic progression than that shown by anti-TNF bDMARD monotherapy. Therefore, anti-TNF bDMARDs should be used in combination with MTX therapy, if not indicated otherwise. The difference in the efficacy between anti-interleukin 6 (IL-6) receptor antibody, tocilizumab (TCZ) plus MTX and TCZ monotherapy is modest as compared to that with anti-TNF bDMARDs monotherapy as reported by a recent systematic literature review and network meta-analysis [Citation54,Citation55]. However, a combination therapy with MTX and TCZ has been recommended in MTX-naïve and MTX-IR patients without risk factors for MTX-related adverse events based on the evidences from CHARITHMA [Citation56], ACT-RAY [Citation57], FUNCTION [Citation10], and SURPRISE [Citation58] trials. The 28-joint count disease activity score (DAS28) showed a remission rate of 61.3% in MTX-naïve patients with early RA receiving MTX + abatacept (ABT), which was greater than that in those receiving MTX monotherapy (43.1%) and ABT monotherapy (45.7%) [Citation15], while the results in MTX-IR patients are unavailable till date.

Tofacitinib (TOF), a tsDMARD against Janus kinase (JAK), has shown superiority to placebo (ORAL Scan) [Citation59] and comparable efficacy to ADA (ORAL Standard) [Citation60] in MTX-IR patients. The JCR guideline has recommended using TOF in MTX-IR patients. Therefore, the addition of TOF to MTX monotherapy is an option for patients not achieving the treatment goal despite 10–12 mg/week of MTX. However, a post-marketing surveillance for the safe and effective use of TOF in Japanese patients with RA is still underway; therefore, the addition of other csDMARDs or bDMARDs may be currently preferred to using TOF.

Folic acid supplementation

A meta-analysis examining the effects of folic acid supplementation on the outcome of MTX therapy showed a decrease in the rates of development of gastrointestinal symptoms, liver dysfunction, and MTX discontinuation [Citation61–66]. However, folic acid or folinic acid supplementation may cause exacerbation of RA in some patients [Citation67–69]. Nevertheless, folic acid supplementation has been globally recommended for all patients with RA receiving MTX based on the evidences from the trials involving 15–25 mg/week of MTX along with 1–2 mg/d of folic acid [Citation8,Citation9]. Folic acid supplementation has been especially recommended for patients receiving MTX above 0.2 mg/kg/week or 8 mg/week, and for patients with high risk for MTX-related adverse events, whereas, folic acid is not necessary for the patients receiving MTX <0.15 mg/kg/week.

Folinic acid is an active form of folic acid, not requiring enzymatic activation by dihydrofolate reductase (DHFR). Therefore, the administration of folinic acid, instead of folic acid, has been recommended in severe adverse events of MTX. Generally, 10 mg of folinic acid is administered orally at every 6 h, or 6–12 mg is administered intramuscularly or intravenously at every 6 h (the daily dose of folinic acid is approximately equal to three times the weekly dose of MTX) concomitantly with hydration and urinary alkalinization until improvement in adverse events is observed.

Screening tests before methotrexate therapy

Prior to MTX therapy, RA activity and risk factors for MTX-related adverse events should be assessed by history taking and physical examination. The estimation of CBC, ESR, blood chemistry, C-reactive protein (CRP), and urinalysis are required as general laboratory tests. Since renal disorder is the most important among the risk factors for MTX-related adverse events, renal function should be evaluated with reference to eGFR or cystatin C in elderly patients, in those with elevated serum creatinine levels, and in those with low body weight [Citation70]. It has been reported that many patients manifest a replication of HBV during or after the immunosuppressive therapies or chemotherapies followed by the development of hepatitis, including fatal fulminant hepatitis, and that is also the case with MTX therapy [Citation71,Citation72]. Therefore, the screening and monitoring of HBV, according to the guidelines for preventing HBV reactivation [Citation73], are needed along with a referral to a gastroenterology/hepatology expert. All patients should be screened for HBs-Ag and HCV-Ab before the commencement of MTX therapy. If HBs-Ag is positive, which suggests HBV-carrier, the patient should be referred to a gastroenterology/hepatology expert for prophylactic anti-viral therapy before the initiation of MTX therapy. If HBs-Ag is negative, high sensitivity testing for HBc-Ab and HBs-Ab should be performed for investigating the possibility of resolved HBV infection. If HBc-Ab and/or HBs-Ab is positive, the screening test for HBV-DNA should be performed every 1–3 months except for vaccinated patients, showing negative reports for HBc-Ab. In addition, when ≥20 IU/mL (2.1 log copies/mL) of HBV-DNA are observed in the sample, the patient should be referred to a gastroenterology/hepatology expert for prophylactic anti-viral therapy along with continuing the MTX therapy.

History taking, chest X-ray, interferon gamma releasing assay (IGRA) or tuberculin (TB) skin test, and chest computed tomography (CT) if needed, should be performed in order to investigate the possibility of TB infection. Two types of IGRAs, namely, Quantiferon®TB Gold (Quest Diagnostics, Secaucus, NJ) and T-SPOT®TB (Oxford Diagnostic Laboratories, Memphis, TN) are approved in Japan for this purpose. In principle, patients who meet the below criteria should receive prophylactic chemotherapy for TB before starting MTX:

  1. An abnormal shadow in chest images compatible with old TB infection.

  2. History of receiving TB chemotherapy.

  3. Probable latent TB infection (LTBI) based on IGRA and/or TB skin test.

  4. Close contact with patients with active TB.

  5. Possible active TB as advised by an expert of pulmonary medicine.

Chest X-ray is helpful in the screening of interstitial pneumonia and other lung infections, which is also helpful in comparing with that obtained for the diagnosis of the lung diseases developing during MTX therapy. When interstitial pneumonia or other pulmonary complications are suspected, percutaneous oxygen saturation (SpO2), high-resolution CT of lung, and tests for detecting β-D-glucan and biomarkers of interstitial pneumonia should be considered in addition to physical examination of the chest. Serum anti-MAC-GPL IgA antibody test can be helpful in the patients suspected of NTM infection with Mycobacterium avium/intracellulare (Mycobacterium avium complex; MAC) [Citation74].

Monitoring during methotrexate therapy

Safety monitoring aims at the adequate management of potentially serious or frequently observed adverse events during MTX therapy. In Japan, pancytopenia, lung injury, severe infection, and LPD are the serious MTX-related adverse events, which are of special concern [Citation22]. Laboratory blood tests and urinalysis should be performed every 2–4 weeks within 6 months of starting the MTX therapy or dose-escalation of MTX. The frequency of these tests may be reduced to every 4–12 weeks in patients receiving constant dose of MTX. However, those with risk factors for MTX-related adverse events, such as renal disorder should be monitored more closely [Citation75]. CBC monitoring should include WBC differentiation, to rule out the possibility of granulocytopenia and lymphocytopenia, and estimation of MCV. Blood chemistry includes estimation of AST, ALT, ALP, albumin, glucose, creatinine, and blood urea nitrogen. Transaminase elevation is the most frequently observed abnormal finding among the laboratory investigations, and the sustained elevation suggests possibility of chronic liver diseases [Citation76]. Sustained hypoalbuminemia is a risk factor for the development of cytopenia and various infectious diseases, in addition to the implication of chronic liver diseases [Citation77]. Chest X-ray should be obtained for investigating drug-induced lung injury and opportunistic infections, such as TB. Chest X-ray or chest CT should be performed in patients with lung comorbidities, such as interstitial pneumonia, and tests for the detection of biomarkers for interstitial pneumonia (KL-6 and SP-D) and fungal infection (β-D glucan) may be considered. Serum anti-MAC-GPL IgA antibody test can be helpful in the patients suspected of MAC infection.

The assessment of effectiveness of MTX should be based on a comprehensive assessment of disease activity, including joint examination, and estimation of acute phase reactants. The disease activity should be initially evaluated every 4–8 weeks of MTX dose adjustment, followed by every 12 weeks in patients with sustained remission or low disease activity. In addition, relevant X-ray images of the joints should be obtained at least once a year. Ultrasound examination of the joints has been reported to be useful for the assessment of active synovitis in patients undergoing MTX therapy [Citation78], and it may be performed in addition to the physical examination of joints.

Perioperative management

A majority of the evidences suggest that continuation of 5–12.5 mg/week of MTX in the perioperative period of elective orthopedic surgery does not produce any impact on the postoperative complications and the wound healing, but it reduces the flare rate of RA [Citation79–86]; however, some reports indicated an increase in the rate of postoperative infections [Citation87,Citation88]. Since there is lack of evidences concerning the surgeries other than elective orthopedic surgery, the judgment regarding the continuation, discontinuation, and restart of MTX therapy should be based on the perioperative condition of patients (for example the status of renal function, bleeding volume, and hypoalbuminemia) and the dose of MTX (more than 12 mg/week or not).

Management during pregnancy and lactation

The exposure to MTX during pregnancy may lead to MTX-related embryopathy, which includes CNS deformity, skull bone deformity, growth deficiency of extremities, and cleft palate [Citation89]. A report from the European League Against Rheumatism (EULAR) task force on the use of DMARDs before pregnancy and during pregnancy and lactation recommends the discontinuation of MTX 1–3 months prior to the conception [Citation90]. It is stated in the package insert of MTX in Japan that, female patients of childbearing age should be instructed to avoid pregnancy during MTX therapy and at least one menstrual cycle after the latest dosing of MTX. MTX is contraindicated during lactation, because MTX has been detected in human breast milk [Citation91–93].

The management of methotrexate-related adverse events

When MTX therapy is started, its major adverse effects and their prevention, early recognition, and treatment should be well explained to the patients. Patient education as described above should also be repeated during MTX therapy by doctors and other medical staff. The pamphlet (in Japanese) prepared by JCR (http://www.ryumachi-jp.com/pdf/mtx_2017.pdf) may be helpful for the above purpose.

Blood disorders (myelosuppression with few exceptions) are observed as MTX-related fatal outcomes in 29.5% of cases [Citation22]. The risk factors include renal disorders [Citation22,Citation94–97], advanced age [Citation22,Citation94,Citation96,Citation97], folic acid deficiency [Citation96,Citation97], multiple concomitant drugs [Citation95–97], hypoalbuminemia [Citation95,Citation97], and dehydration.

MTX-related pneumonia develops in 1–7% of RA patients [Citation98,Citation99] and 13% of them are fatal [Citation100]. MTX-related pneumonia tends to develop within 1 year of the commencement of MTX therapy in Japan and other countries, although it may occur abruptly at any time during the therapy. The risk factors for MTX-related pneumonia include pre-existing rheumatoid lung, advanced age, diabetes mellitus, hypoalbuminemia, and previous use of DMARDs [Citation101,Citation102]. When patients develop interstitial pneumonia, MTX should be immediately discontinued, and differential diagnosis to rule out other possibilities should be performed. MTX-related pneumonia is not distinguishable from PCP on the basis of chest HRCT images alone. A publication by the Japanese Respiratory Society Committee on the formulation of Consensus statement is helpful in the diagnosis and treatment of drug-induced lung injuries [Citation103]. Antibiotics, such as TMP/SMX 6–12 g/d are administered along with glucocorticoids. Re-exposure to MTX should be avoided because it has been reported that 25% of such patients developed repeated MTX-related pneumonia [Citation100].

The risk factors for infectious diseases to be evaluated prior to MTX therapy include age, preexisting lung disease, extra-articular manifestations, diabetes mellitus, glucocorticoids use, and serious infection within recent 3 years [Citation104,Citation105], in addition to chronic infectious complications, renal disorder, bone marrow disorder, and the history of opportunistic infections. The adequate prophylaxis includes complete antibiotic therapy for active infection or necessary vaccination, such as that against pneumococcus and influenza viruses, isoniazid for LTBI, TMP/SMX, pentamidine isethionate inhalation, atovaquone for those with elevated risk for PCP, and consultation with an expert of pulmonary medicine for patients with possible NTM infection.

MTX-related liver disorders are classified as dose-dependent, principally hepatotoxic liver dysfunction and viral hepatitis reactivated or aggravated by MTX. When patients showing positive test results for HBV or HCV develop liver dysfunction, they should be referred to a gastroenterology/hepatology expert for the management including discontinuation of MTX administration. When patients without viral hepatitis show elevated AST and/or ALT levels not greater than 3-fold of the upper limit of the normal levels, reduction in the dose of MTX, or starting or increasing the dose of folic acid should be considered; whereas, for those showing elevated AST and/or ALT levels greater than 3-fold of the upper limit of normal, dose reduction or discontinuation of MTX, and starting or increasing the dose of folic acid as daily administration should be encouraged.

A 2–6 fold increased risk of malignant lymphoma in patients with RA as compared to that in the general population have been reported [Citation106–109]. Diffuse large B-cell lymphoma (DLBCL) is the most frequently observed histological type [Citation109], and the presence of extra-nodular lesions, such as in skin, oral cavity, pharynx, and lungs are characteristic of LPD in the patients with receiving MTX therapy [Citation110]. Many cases of iatrogenic immunodeficiency-associated LPD demonstrating complete or partial regression of nodular or extra-nodular lesions shortly after discontinuing the administration of MTX or other immunosuppressive drugs have been reported. A recent report suggested that the elevated mean dose of MTX was a risk factor for LPD; however, the cumulative disease activity was not adjusted in that study [Citation111].

Conflict of interest

HK has received consulting fees and/or speaker fees from AbbVie GK, Bristol-Myers Squibb, Chugai Pharmaceutical Co., Ltd., Eli Lilly and Company, Janssen Pharmaceutical K.K., Mitsubishi Tanabe Pharma, Novartis Pharma K.K., and Sanofi K.K., and has received research grants from AbbVie GK, Asaki Kasei Pharma, Astellas Pharma Inc., Bristol-Myers Squibb, Chugai Pharmaceutical Co., Ltd., Eisai Co., Ltd., Mitsubishi Tanabe Pharma, Novartis Pharma K.K., and Sanofi K.K.

TF has received a research grant and/or speaker fee from Pfizer Japan Inc., Ono Pharmaceutical Co., Ltd., Daiichi Sankyo Co., Ltd., Mitsubishi Tanabe Pharma, Eisai Co., Ltd., AbbVie GK, and Astellas Pharma Inc.

AN has received consulting or speaker fees from Astellas Pharma Inc., Bristol-Myers Squibb, Chugai Pharmaceutical Co., Ltd., Daiichi-Sankyo Healthcare Co., Ltd., Eisai Co., Ltd., Janssen Pharmaceutical K.K., Mitsubishi Tanabe Pharma, Novartis Pharma K.K., Pfizer Japan Inc., Taisho Toyama Pharmaceutical Co., Ltd., and has received research grants from Kissei Pharmaceutical Co., Ltd.

KK has received speaker fee from Mitsubishi Tanabe Pharma.

MH has received speaker fees from Mitsubishi Tanabe Pharma, Chugai Pharmaceutical Co., Ltd., and Eli Lilly and Company, research grants from Abbvie GK, Ayumi Co., Ltd., Eisai Co., Ltd., Nippon-Kayaku Co., Ltd., Taisho-Toyama Pharmaceutical Co., Ltd., Mitsubishi Tanabe Pharma, and Teijin Pharma Co., Ltd.

YS has received research funding from Chugai Pharmaceutical Co., Ltd., and Teijin Pharma Co., Ltd., has received honoraria or fees for promotional materials from Pfizer Japan Inc., Eisai Co., Ltd., Asaki Kasei Pharma, and Ayumi Co., Ltd. All other authors declared no conflicts of interest.

Acknowledgments

The authors thank to Editage Japan for editing the manuscript, which fee was supported by Research on rare and intractable diseases, Health and Labour Sciences Research Grant H29-028.

References

  • Smolen JS, Breedveld FC, Burmester GR, Bykerk V, Dougados M, Emery P, et al. Treating rheumatoid arthritis to target: 2014 update of the recommendations of an international task force. Ann Rheum Dis. 2016;75:3–15.
  • Smolen JS, Landewé R, Breedveld FC, Buch M, Burmester G, Dougados M, et al. EULAR recommendations for the management of rheumatoid arthritis with synthetic and biological disease-modifying antirheumatic drugs: 2013 update. Ann Rheum Dis. 2014;73:492–509.
  • Takeuchi T, Yamanaka H, Ishiguro N, Miyasaka N, Mukai M, Matsubara T, et al. Adalimumab, a human anti-TNF monoclonal antibody, outcome study for the prevention of joint damage in Japanese patients with early rheumatoid arthritis: the HOPEFUL 1 study. Ann Rheum Dis. 2014;73:536–43.
  • Atsumi T, Yamamoto K, Takeuchi T, Yamanaka H, Ishiguro N, Tanaka Y, et al. The first double-blind, randomized, parallel-group certolizumab pegol study in methotrexate-naive early rheumatoid arthritis patients with poor-prognostic factors, C-OPERA, shows inhibition of radiographic progression. Ann Rheum Dis. 2016;75:75–83.
  • Saag KG, Teng GG, Patkar NM, Anuntiyo J, Finney C, Curtis JR, et al. American College of Rheumatology 2008 recommendations for the use of nonbiologic and biologic disease-modifying antirheumatic drugs in rheumatoid arthritis. Arthritis Rheum. 2008;59:762–84.
  • Claudepierre P, Urien S, Chevalier X, Chassany O, Larget-Piet B, Tillement JP. Methotrexate serum binding in rheumatoid arthritis. Int J Clin Pharmacol Ther. 1994;32:113–15.
  • Drafting Committee for Hepatitis Management Guidelines and the Japan Society of Hepatology. JSH guidelines for the management of hepatitis B virus infection. Hepatol Res. 2014;44(S1):1–58.
  • Pavy S, Constantin A, Pham T, Gosseec L, Maillefert JF, Cantagrel A, et al. Methotrexate therapy for rheumatoid arthritis: clinical practice guidelines based on published evidence and expert opinion. Joint Bone Spine. 2006;73:388–95.
  • Visser K, Katchamart W, Loza E, Martinez-Lopez JA, Salliot C, Trudeau J, et al. Multinational evidence-based recommendations for the arthritis: integrating systematic literature research and expert opinion of a broad international panel of rheumatologists in the 3E Initiative. Ann Rheum Dis. 2009;68:1086–93.
  • Kashiwazaki S, Ichikawa Y, Sugawara S, Nagaya I, Kawai S, Hakota M, et al. Determination of the clinical optimal dose of L-377 (methotrexate capsule) for the treatment of rheumatoid arthritis. Jpn J Inflammat. 1996;16:437–58.
  • Visser K, van der Heijde D. Optimal dosage and route of administration of methotrexate in rheumatoid arthritis: a systematic review of the literature. Ann Rheum Dis. 2009;68:1094–9.
  • Mouterde G, Baillet A, Gaujoux-Viala C, Cantagrel A, Wendling D, Le Loët X, et al. Optimizing methotrexate therapy in rheumatoid arthritis: a systematic literature review. Joint Bone Spine. 2011;78:587–92.
  • Thompson RN, Watts C, Edelman J, Esdaile J, Russell AS. A controlled two-centre trial of parenteral methotrexate therapy for refractory rheumatoid arthritis. J Rheumatol. 1984;11:760–3.
  • Hobl EL, Mader RM, Jilma B, Duhm B, Mustak M, Bröll H, et al. A randomized, double-blind, parallel, single-site pilot trial to compare two different starting doses of methotrexate in methotrexate-naïve adult patients with rheumatoid arthritis. Clin Ther. 2012;34:1195–203.
  • Emery P, Burmester GR, Bykerk VP, Combe BG, Furst DE, Barré E, et al. Evaluating drug-free remission with abatacept in early rheumatoid arthritis: results from the phase 3b, multicentre, randomized, active-controlled AVERT study of 24 months, with a 12-month, double-blind treatment period. Ann Rheum Dis. 2015;74:19–26.
  • Burmester GR, Rigby WF, van Vollenhoven RF, Kay J, Rubbert-Roth A, Kelman A, et al. Tocilizumab in early progressive rheumatoid arthritis: FUNCTION, a randomised controlled trial. Ann Rheum Dis. 2016;75:1081–91.
  • Strand V, Cohen S, Schiff M, Weaver A, Fleischmann R, Cannon G, et al. Treatment of active rheumatoid arthritis with leflunomide compared with placebo and methotrexate. Leflunomide Rheumatoid Arthritis Investigators Group. Arch Intern Med. 1999;159:2542–50.
  • Grigor C, Capell H, Stirling A, McMahon AD, Lock P, Vallance R, et al. Effect of a treatment strategy of tight control for rheumatoid arthritis (the TICORA study): a single-blind randomised controlled trial. Lancet. 2004;364:263–9.
  • Verstappen SM, Jacobs JW, van der Veen MJ, Heurkens AH, Schenk Y, ter Borg EJ, et al. Intensive treatment with methotrexate in early rheumatoid arthritis: aiming for remission. Computer Assisted Management in Early Rheumatoid Arthritis (CAMERA, an open-label strategy trial). Ann Rheum Dis. 2007;66:1443–9.
  • Seideman P. Methotrexate-the relationship between dose and clinical effect. Br J Rheumatol. 1993;32:751–3.
  • 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;63:1232–4.
  • RHEUMATREX® CAPSULES 2 mg. Proper use information. Vol. 21. New York, NY: Pfizer Inc.; 2015.
  • Hoekstra M, Haagsma C, Neef C, Proost J, Knuif A, van de Laar M. Bioavailability of higher dose methotrexate comparing oral and subcutaneous administration in patients with rheumatoid arthritis. J Rheumatol. 2004;31:645–8.
  • Korber H, Iven H, Gross WL. Bioavailability and pharmacokinetics of methotrexate (MTX) and its metabolite 7-hydroxy MTX after low dose MTX (25 mg) in patients with chronic rheumatic diseases. Arthritis Rheum. 1992;35(l):S142.
  • Freeman-Narrod M, Gerstley BJ, Engstrom PF, Bomstein RS. Comparison of serum concentrations of methotrexate after various routes of administration. Cancer. 1975;36:1619–24.
  • Teresi ME, Crom WR, Choi KE, Mirro J, Evans WE. Methotrexate bioavailability after oral and intramuscular administration in children. J Pediatr. 1987;110:788–92.
  • Braun J, Kästner P, Flaxenberg P, Währisch J, Hanke P, Demary W, et al. Comparison of the clinical efficacy and safety of subcutaneous versus oral administration of methotrexate in patients with active rheumatoid arthritis: results of a six-month, multicenter, randomized, double-blind, controlled, phase IV trial. Arthritis Rheum. 2008;58:73–81.
  • Schiff MH, Jaffe JS, Freundlich B. Head-to-head, randomised, crossover study of oral versus subcutaneous methotrexate in patients with rheumatoid arthritis: drug-exposure limitations of oral methotrexate at doses ≥15 mg may be overcome with subcutaneous administration. Ann Rheum Dis. 2014;73:1549–51.
  • Hoekstra M, Haagsma C, Neef C, Proost J, Knuif A, van de Laar M. Splitting high-dose oral methotrexate improves bioavailability: a pharmacokinetic study in patients with rheumatoid arthritis. J Rheumatol. 2006;33:481–5.
  • Bianchi G, Caporali R, Todoerti M, Mattana P. Methotrexate and rheumatoid arthritis: current evidence regarding subcutaneous versus oral routes of administration. Adv Ther. 2016;33:369–78.
  • Singh JA, Saag SK, Bridges SL Jr., Akl EA, Bannuru RR, Sullivan MC, et al. 2015 American College of Rheumatology recommendations for the treatment of rheumatoid arthritis. Arthritis Rheumatol. 2016;68:1–26.
  • Rath T, Rubbert A. Drug combinations with methotrexate to treat rheumatoid arthritis. Clin Exp Rheumatol. 2010;28(61):S52–S7.
  • Dale J, Alcorn N, Capell H, Madhok R. Combination therapy for rheumatoid arthritis: methotrexate and sulfasalazine together or with other DMARDs. Nat Clin Pract Rheumatol. 2007;3:450–8.
  • Tugwell P, Pincus T, Yocum D, Stein M, Gluck O, Kraag G, et al. Combination therapy with cyclosporine and methotrexate in severe rheumatoid arthritis. The Methotrexate-Cyclosporine Combination Study Group. N Engl J Med. 1995;333:137–41.
  • Marchesoni A, Battafarano N, Arreghini M, Panni B, Gallazzi M, Tosi S. Radiographic progression in early rheumatoid arthritis: a 12-month randomized controlled study comparing the combination of cyclosporin and methotrexate with methotrexate alone. Rheumatology (Oxford). 2003;42:1545–9.
  • Capell HA, Madhok R, Porter DR, Munro RA, McInnes IB, Hunter JA, et al. Combination therapy with sulfasalazine and methotrexate is more effective than either drug alone in patients with rheumatoid arthritis with a suboptimal response to sulfasalazine: results from the double-blind placebo-controlled MASCOT study. Ann Rheum Dis. 2007;66:235–41.
  • Dougados M, Combe B, Cantagrel A, Goupille P, Olive P, Schattenkirchner M, et al. Combination therapy in early rheumatoid arthritis: a randomised, controlled, double blind 52 week clinical trial of sulphasalazine and methotrexate compared with the single components. Ann Rheum Dis. 1999;58:220–5.
  • Haagsma CJ, van Riel PL, de Rooij DJ, Vree TB, Russel FJ, van’t Hof MA, et al. Combination of methotrexate and sulphasalazine vs methotrexate alone: a randomized open clinical trial in rheumatoid arthritis patients resistant to sulphasalazine therapy. Br J Rheumatol.1994;33:1049–55.
  • Haagsma CJ, van Riel PL, de Jong AJ, van de Putte LB. Combination of sulphasalazine and methotrexate versus the single components in early rheumatoid arthritis: a randomized, controlled, double-blind, 52 week clinical trial. Br J Rheumatol. 1997;36:1082–8.
  • Kremer JM, Genovese MC, Cannon GW, Caldwell JR, Cush JJ, Furst DE, et al. Concomitant leflunomide therapy in patients with active rheumatoid arthritis despite stable doses of methotrexate. A randomized, double-blind, placebo-controlled trial. Ann Intern Med. 2002;137:726–33.
  • Lehman AJ, Esdaile JM, Klinkhoff AV, Grant E, Fitzgerald A, Canvin J, et al. A 48-week, randomized, double-blind, double-observer, placebo-controlled multicenter trial of combination methotrexate and intramuscular gold therapy in rheumatoid arthritis: results of the METGO study. Arthritis Rheum. 2005;52:1360–70.
  • Kremer JM, Habros JS, Kolba KS, Kaine JL, Borton MA, Mengle-Gaw LJ, et al. Tacrolimus in rheumatoid arthritis patients receiving concomitant methotrexate: a six-month, open-label study. Arthritis Rheum. 2003;48:2763–8.
  • Kitahama M, Nakajima A, Inoue E, Taniguchi A, Momohara S, Yamanaka H, et al. Efficacy of adjunct tacrolimus treatment in patients with rheumatoid arthritis with inadequate responses to methotrexate. Mod Rheumatol. 2013;23:788–93.
  • Ishiguro N, Yamamoto K, Katayama K, Kondo M, Sumida T, Mimori T, et al. Concomitant iguratimod therapy in patients with active rheumatoid arthritis despite stable doses of methotrexate: a randomized, double-blind, placebo-controlled trial. Mod Rheumatol. 2013;23:430–9.
  • Hara M, Ishiguro N, Katayama K, Kondo M, Sumida T, Mimori T, et al. Safety and efficacy of combination therapy of iguratimod with methotrexate for patients with active rheumatoid arthritis with an inadequate response to methotrexate: an open-label extension of a randomized, double-blind, placebo-controlled trial. Mod Rheumatol. 2014;24:410–18.
  • Sawada T, Inokuma S, Sato T, Otsuka T, Saeki Y, Takeuchi T, et al. Leflunomide-induced interstitial lung disease: prevalence and risk factors in Japanese patients with rheumatoid arthritis. Rheumatology (Oxford). 2009;48:1069–72.
  • Ichikawa Y, Saito T, Yamanaka H, Akizuki M, Kondo H, Kobayashi S, et al. Therapeutic effects of the combination of methotrexate and bucillamine in early rheumatoid arthritis: a multicenter, double-blind, randomized controlled study. Mod Rheumatol. 2005;15:323–8.
  • Nam JL, Ramiro S, Gaujoux-Viala C, Takase K, Leon-Garcia M, Emery P, et al. Efficacy of biological disease-modifying antirheumatic drugs: a systematic literature review informing the 2013 update of the EULAR recommendations for the management of rheumatoid arthritis. Ann Rheum Dis. 2014;73:516–28.
  • Klareskog L, vander Heijde D, de Jager JP, Gough A, Kalden J, Malaise M, et al. Therapeutic effect of the combination of etanercept and methotrexate compared with each treatment alone in patients with rheumatoid arthritis: double-blind randomised controlled trial. Lancet. 2004;363:675–81.
  • Kameda H, Kanbe K, Sato E, Ueki Y, Saito K, Nagaoka S, et al. Continuation of methotrexate resulted in better clinical and radiographic outcomes than discontinuation upon starting etanercept in patients with rheumatoid arthritis: 52-week results from the JESMR study. J Rheumatol. 2011;38:1585–92.
  • Breedveld FC, Weisman MH, Kavanaugh AF, Cohen SB, Pavelka K, van Vollenhoven R, et al. The PREMIER study: a multicenter, randomized, double-blind clinical trial of combination therapy with adalimumab plus methotrexate versus methotrexate alone or adalimumab alone in patients with early, aggressive rheumatoid arthritis who had not had previous methotrexate treatment. Arthritis Rheum. 2006;54:26–37.
  • Emery P, Fleischmann RM, Moreland LW, Hsia EC, Strusberg I, Durez P, et al. Golimumab, a human anti-tumor necrosis factor alpha monoclonal antibody, injected subcutaneously every four weeks in methotrexate-naive patients with active rheumatoid arthritis: twenty-four-week results of a phase III, multicenter, randomized, double-blind, placebo-controlled study of golimumab before methotrexate as first-line therapy for early-onset rheumatoid arthritis. Arthritis Rheum. 2009;60:2272–83.
  • Keystone EC, Genovese MC, Klareskog L, Hsia EC, Hall ST, Miranda PC, et al. Golimumab, a human antibody to tumour necrosisfactor a given by monthly subcutaneous injections, in active rheumatoid arthritis despite methotrexate therapy: the GO-FORWARD study. Ann Rheum Dis. 2009;68:789–96.
  • Jansen JP, Buckley F, Dejonckheere F, Ogale S. Comparative efficacy of biologics as monotherapy and in combination with methotrexate on patient reported outcomes (PROs) in rheumatoid arthritis patients with an inadequate response to conventional DMARDs—a systematic review and network meta-analysis. Health Qual Life Outcomes. 2014;12:102.
  • Buckley F, Finckh A, Huizinga TW, Dejonckheere F, Jansen JP. Comparative efficacy of novel DMARDs as monotherapy and in combination with methotrexate in rheumatoid arthritis patients with inadequate response to conventional DMARDs: a network meta-analysis. J Manag Care Spec Pharm. 2015;21:409–23.
  • Maini RN, Taylor PC, Szechinski J, Pavelka K, Bröll J, Balint G, et al. Double-blind randomized controlled clinical trial of the interleukin-6 receptor antagonist, tocilizumab, in European patients with rheumatoid arthritis who had an incomplete response to methotrexate. Arthritis Rheum. 2006;54:2817–29.
  • Dougados M, Kissel K, Conaghan PG, Mola EM, Schett G, Gerli R, et al. Clinical, radiographic and immunogenic effects after 1 year of tocilizumab-based treatment strategies in rheumatoid arthritis: the ACT-RAY study. Ann Rheum Dis. 2014;73:803–9.
  • Kaneko Y, Atsumi T, Tanaka Y, Inoo M, Kobayashi-Haraoka H, Amano K, et al. Comparison of adding tocilizumab to methotrexate with switching to tocilizumab in patients with rheumatoid arthritis with inadequate response to methotrexate: 52-week results from a prospective, randomised, controlled study (SURPRISE study). Ann Rheum Dis. 2016;75:1917–23.
  • van Vollenhoven RF, Fleischmann R, Cohen S, Lee EB, García Meijide JA, Wagner S, et al. Tofacitinib or adalimumab versus placebo in rheumatoid arthritis. N Engl J Med. 2012;367:508–19.
  • van der Heijde D, Tanaka Y, Fleischmann R, Keystone E, Kremer J, Zerbini C, et al. Tofacitinib (CP-690,550) in patients with rheumatoid arthritis receiving methotrexate: twelve-month data from a twenty-four-month phase III randomized radiographic study. Arthritis Rheum. 2013;65:559–70.
  • Ortiz Z, Shea B, Suarez-Almazor ME, Moher D, Wells GA, Tugwell P. The efficacy of folic acid and folinic acid in reducing methotrexate gastrointestinal toxicity in rheumatoid arthritis: a metaanalysis of randomized controlled trials. J Rheumatol. 1998;25:36–43.
  • Pincus T. Folic and folinic acid supplementation reduces methotrexate gastrointestinal side effects in rheumatoid arthritis. Clin Exp Rheumatol. 1998;16:667–8.
  • Whittle SL, Hughes RA. Folate supplementation and methotrexate treatment in rheumatoid arthritis: a review. Rheumatology (Oxford). 2004;43:267–71.
  • Morgan SL, Baggott JE. Folate supplementation during methotrexate therapy for rheumatoid arthritis. Clin Exp Rheumatol. 2010;28(61):S102–S9.
  • Strober BE, Menon K. Folate supplementation during methotrexate therapy for patients with psoriasis. J Am Acad Dermatol. 2005;53:652–9.
  • Shea B, Swinden MV, Ghogomu ET, Ortiz Z, Katchamart W, Rader T, et al. Folic acid and folinic acid for reducing side effects in patients receiving methotrexate for rheumatoid arthritis. J Rheumatol. 2014;41:1049–60.
  • Suzuki Y, Uehara R, Tajima C, Noguchi A, Ide M, Ichikawa Y, et al. Elevation of serum hepatic aminotransferases during treatment of rheumatoid arthritis with low-dose methotrexate. Risk factors and response to folic acid. Scand J Rheumatol. 1999;28:273–81.
  • Tishler M, Caspi D, Fishel B, Yaron M. The effects of leucovorin (folinic acid) on methotrexate therapy in rheumatoid arthritis patients. Arthritis Rheum. 1988;31:906–9.
  • Joyce DA, Will RK, Hoffman DM, Laing B, Blackboum SJ. Exacerbation of rheumatoid arthritis in patients treated with methotrexate after administration of folinic acid. Ann Rheum Dis. 1991;50:913–14.
  • Köttgen A, Pattaro C, Böger CA, Fuchsberger C, Olden M, Glazer NL, et al. New loci associated with kidney function and chronic kidney disease. Nat Genet. 2010;42:376–84.
  • Ito S, Nakazono K, Murasawa A, Mita Y, Hata K, Saito N, et al. Development of fulminant hepatitis B (precore variant mutant type) after the discontinuation of low-dose methotrexate therapy in a rheumatoid arthritis patient. Arthritis Rheum. 2001;44:339–42.
  • Hagiyama H, Kubota T, Komano Y, Kurosaki M, Watanabe M, Miyasaka N. Fulminant hepatitis in an asymptomatic chronic carrier of hepatitis B virus mutant after withdrawal of low-dose methotrexate therapy for rheumatoid arthritis. Clin Exp Rheumatol. 2004;22:375–6.
  • Oketani M, Ido A, Uto H, Tsubouchi H. Prevention of hepatitis B virus reactivation in patients receiving immunosuppressive therapy or chemotherapy. Hepatol Res. 2012;42:627–36.
  • Watanabe M, Banno S, Sasaki K, Naniwa T, Hayami Y, Ueda R. Serodiagnosis of Mycobacterium avium-complex pulmonary disease with an enzyme immunoassay kit that detects anti-glycopeptidolipid core antigen IgA antibodies in patients with rheumatoid arthritis. Mod Rheumatol. 2011;21:144–9.
  • The effect of age and renal function on the efficacy and toxicity of methotrexate in rheumatoid arthritis. Rheumatoid Arthritis Clinical Trial Archive Group. J Rheumatol. 1995;22:218–23.
  • Guidelines for monitoring drug therapy in rheumatoid arthritis. American College of Rheumatology Ad Hoc Committee on Clinical Guidelines. Arthritis Rheum. 1996;39:723–31.
  • Kent PD, Luthra HS, Michet C Jr. Risk factors for methotrexate-induced abnormal laboratory monitoring results in patients with rheumatoid arthritis. J Rheumatol. 2004;31:1727–31.
  • Cheung PP, Dougados M, Gossec L. Reliability of ultrasonography to detect synovitis in rheumatoid arthritis: a systematic literature review of 35 studies (1,415 patients). Arthritis Care Res. 2010;62:323–34.
  • del Olmo L, Hemández B, Galindo-Izquierdo M, Tébar D, Balsa A, Carmona L. Perioperative management of disease modifying anti-rheumatic drugs: recommendations based on a meta-analysis. Rev Esp Cir Ortop Traumatol. 2012;56:393–412.
  • Loza E, Martinez-Lopez JA, Carmona L. A systematic review on the optimum management of the use of methotrexate in rheumatoid arthritis patients in the perioperative period to minimize perioperative morbidity and maintain disease control. Clin Exp Rheumatol. 2009;27:856–62.
  • Pieringer H, Stuby U, Biesenbach G. The place of methotrexate perioperatively in elective orthopedic surgeries in patients with rheumatoid arthritis. Clin Rheumatol. 2008;27:1217–20.
  • Pieringer H, Stuby U, Biesenbach G. Patients with rheumatoid arthritis undergoing surgery: how should we deal with antirheumatic treatment? Semin Arthritis Rheum. 2007;36:278–86.
  • Perhala RS, Wilke WS, Clough JD, Segal AM. Local infectious complications following large joint replacement in rheumatoid arthritis patients treated with methotrexate versus those not treated with methotrexate. Arthritis Rheum. 1991;34:146–52.
  • Murata K, Yasuda T, Ito H, Yoshida M, Shimizu M, Nakajima T. Lack of increase in postoperative complications with low-dose methotrexate therapy in patients with rheumatoid arthritis undergoing elective orthopedic surgery. Mod Rheumatol. 2006;16:14–19.
  • Sany J, Anaya JM, Canovas F, Combe B, Jorgensen C, Saker S, et al. Influence of methotrexate on the frequency of postoperative infectious complications in patients with rheumatoid arthritis. J Rheumatol. 1993;20:1129–32.
  • Grennan DM, Gray J, Loudon J, Fear S. Methotrexate and early postoperative complications in patients with rheumatoid arthritis undergoing elective orthopaedic surgery. Ann Rheum Dis. 2001;60:214–17.
  • Bridges SL Jr, López-Méndez A, Han KH, Tracy IC, Alarcón GS. Should methotrexate be discontinued before elective orthopedic surgery in patients with rheumatoid arthritis? J Rheumatol. 1991;18:984–8.
  • Carpenter MT, West SG, Vogelgesang SA, Casey Jones DE. Postoperative joint infections in rheumatoid arthritis patients on methotrexate therapy. Orthopedics. 1996;19:207–10.
  • Hyoun SC, Običan SG, Scialli AR. Teratogen update: methotrexate. Birth Defects Res Part A Clin Mol Teratol. 2012;94:187–207.
  • Götestam Skorpen C, Hoeltzenbein M, Tincani A, Fischer-Betz R, Elefant E, Chambers C, et al. The EULAR points to consider for use of antirheumatic drugs before pregnancy, and during pregnancy and lactation. Ann Rheum Dis. 2016;75:795–810.
  • Johns DG, Rutherford LD, Leighton PC, Vogel CL. Secretion of methotrexate into human milk. Am J Obstet Gynecol. 1972;112:978–80.
  • Thorne JC, Nadarajah T, Moretti M, Ito S. Methotrexate use in a breastfeeding patient with rheumatoid arthritis. J Rheumatol. 2014;41:2332.
  • Sammaritano LR, Bermas BL. Rheumatoid arthritis medications and lactation. Curr Opin Rheumatol. 2014;26:354–60.
  • Buchbinder R, Hall S, Sambrook PN, Champion GD, Harkness A, Lewis D, et al. Methotrexate therapy in rheumatoid arthritis: a life table review of 587 patients treated in community practice. J Rheumatol. 1993;20:639–44.
  • Gutierrez-Urena S, Molina JF, Garcia CO, Cuellar ML, Espinoza LR. Pancytopenia secondary to methotrexate therapy in rheumatoid arthritis. Arthritis Rheum. 1996;39:272–6.
  • Kuitunen T, Maimström J, Palva E, Pettersson T. 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. 2005;34:238–41.
  • Lim AY, Gaffney K, Scott DG. Methotrexate-induced pancytopenia: serious and under-reported? Our experience of 25 cases in 5 years. Rheumatology (Oxford). 2005;44:1051–5.
  • Kinder AJ, Hassell AB, Brand J, Brownfield A, Grove M, Shadforth MF. The treatment of inflammatory arthritis with methotrexate in clinical practice: treatment duration and incidence of adverse drug reactions. Rheumatology (Oxford). 2005;44:61–6.
  • Saravanan V, Kelly CA. Reducing the risk of methotrexate pneumonitis in rheumatoid arthritis. Rheumatology (Oxford). 2004;43:143–7.
  • Imokawa S, Colby TV, Leslie KO, Helmers RA. Methotrexate pneumonitis: review of the literature and histopathological findings in nine patients. Eur Respir J. 2000;15:373–81.
  • Alarcón GS, Kremer JM, Macaluso M, Weinblatt ME, Cannon GW, Palmer WR, et al. Risk factors for methotrexate-induced lung injury in patients with rheumatoid arthritis. A multicenter, case-control study. Methotrexate-Lung Study Group. Ann Intern Med. 1997;127:356–64.
  • Ohosone Y, Okano Y, Kameda H, Fujii T, Hama N, Hirakata M, et al. Clinical characteristics of patients with rheumatoid arthritis and methotrexate induced pneumonitis. J Rheumatol. 1997;24:2299–303.
  • Kubo K, Azuma A, Kanazawa M, Kameda H, Kusumoto M, Genma A, et al. Consensus statement for the diagnosis and treatment of drug-induced lung injuries. Respir Investig. 2013;51:260–77.
  • Doran MF, Crowson CS, Pond GR, O’Fallon WM, Gabriel SE. Predictors of infection in rheumatoid arthritis. Arthritis Rheum. 2002;46:2294–300.
  • Crowson CS, Hoganson DD, Fitz-Gibbon PD, Matteson EL. Development and validation of a risk score for serious infection in patients with rheumatoid arthritis. Arthritis Rheum. 2012;64:2847–55.
  • Kaiser R. Incidence of lymphoma in patients with rheumatoid arthritis: systematic review of the literature. Clin Lymphoma Myeloma. 2008;8:87–93.
  • Hashimoto A, Chiba N, Tsuno H, Komiya A, Furukawa H, Matsui T, et al. Incidence of malignancy and the risk of lymphoma in Japanese patients with rheumatoid arthritis compared to the general population. J Rheumatol. 2015;42:564–71.
  • Yamada T, Nakajima A, Inoue E, Tanaka E, Taniguchi A, Momohara S, et al. Incidence of malignancy in Japanese patients with rheumatoid arthritis. Rheumatol Int. 2011;31:1487–92.
  • Simon TA, Thompson A, Gandhi KK, Hochberg MC, Suissa S. Incidence of malignancy in adult patients with rheumatoid arthritis: a meta-analysis. Arthritis Res Ther. 2015;17:212–21.
  • Baecklund E, Iliadou A, Askling J, Ekbom A, Backlin C, Granath F, et al. Association of chronic inflammation, not its treatment, with increased lymphoma risk in rheumatoid arthritis. Arthritis Rheum. 2006;54:692–701.
  • Kameda T, Dobashi H, Miyatake N, Inoo M, Onishi I, Kurata N, et al. Association of higher methotrexate dose with lymphoproliferative disease onset in rheumatoid arthritis patients. Arthritis Care Res (Hoboken). 2014;66:1302–9.