Journal Information
Vol. 11. Issue 5.
Pages 279-294 (September - October 2015)
Visits
6558
Vol. 11. Issue 5.
Pages 279-294 (September - October 2015)
Special Article
Full text access
2014 Update of the Consensus Statement of the Spanish Society of Rheumatology on the Use of Biological Therapies in Rheumatoid Arthritis
Actualización 2014 del Documento de Consenso de la Sociedad Española de Reumatología sobre el uso de terapias biológicas en la artritis reumatoide
Visits
6558
Raimon Sanmartía,
Corresponding author
sanmarti@clinic.ub.es

Corresponding author.
, Susana García-Rodríguezb, José María Álvaro-Graciac, José Luis Andreud, Alejandro Balsae, Rafael Cálizf, Antonio Fernández-Nebrog, Iván Ferraz-Amaroh, Juan Jesús Gómez-Reinoi, Isidoro González-Álvaroc, Emilio Martín-Molae, Víctor Manuel Martínez-Taboadaj, Ana M. Ortizc, Jesús Tornerok, Sara Marsall, José Vicente Moreno-Muelasl,m
a Servicio de Reumatología, Hospital Clínic de Barcelona, Barcelona, Spain
b Unidad de Investigación, Sociedad Española de Reumatología, Madrid, Spain
c Servicio de Reumatología, Hospital Universitario de la Princesa, Madrid, Spain
d Servicio de Reumatología, Hospital Universitario Puerta de Hierro, Madrid, Spain
e Servicio de Reumatología, Hospital Universitario La Paz, Madrid, Spain
f Servicio de Reumatología, Hospital Universitario Virgen de las Nieves, Granada, Spain
g Unidad de Gestión Clínica de Reumatología, Instituto de Investigación Biomédica de Málaga, Hospital Regional Universitario de Málaga, Universidad de Málaga, Málaga, Spain
h Servicio de Reumatología, Hospital Universitario de Canarias, Tenerife, Spain
i Servicio de Reumatología, Hospital Clínico Universitario de Santiago, Santiago de Compostela, A Coruña, Spain
j Servicio de Reumatología, Hospital Universitario Marqués de Valdecilla, Santander, Cantabria, Spain
k Servicio de Reumatología, Hospital Universitario de Guadalajara, Guadalajara, Spain
l Servicio de Reumatología, Hospital Universitario Vall d'Hebron, Barcelona, Spain
m Sociedad Española de Reumatología, Madrid, Spain
Ver más
This item has received
Article information
Abstract
Full Text
Bibliography
Download PDF
Statistics
Figures (1)
Tables (2)
Table 1. SER 2014 Consensus Recommendations.
Table 2. Biological Therapy Risk Management.
Show moreShow less
Abstract
Objective

To establish recommendations for the management of patients with rheumatoid arthritis (RA) to serve as a reference for all health professionals involved in the care of these patients, and focusing on the role of available synthetic and biologic disease-modifying antirheumatic drugs (DMARDs).

Methods

Consensual recommendations were agreed on by a panel of 14 experts selected by the Spanish Society of Rheumatology (SER). The available scientific evidence was collected by updating three systematic reviews (SR) used for the EULAR 2013 recommendations. A new SR was added to answer an additional question. The literature review of the scientific evidence was made by the SER reviewer's group. The level of evidence and the degree of recommendation was classified according to the Oxford Centre for Evidence-Based Medicine system. A Delphi panel was used to evaluate the level of agreement between panellists (strength of recommendation).

Results

Thirteen recommendations for the management of adult RA were emitted. The therapeutic objective should be to treat patients in the early phases of the disease with the aim of achieving clinical remission, with methotrexate playing a central role in the therapeutic strategy of RA as the reference synthetic DMARD. Indications for biologic DMARDs were updated and the concept of the optimisation of biologicals was introduced.

Conclusions

We present the fifth update of the SER recommendations for the management of RA with synthetic and biologic DMARDs.

Keywords:
Rheumatoid arthritis
Disease management
Recommendations
Spanish Society of Rheumatology
Biologic DMARD
Synthetic DMARD
Resumen
Objetivo

Establecer recomendaciones para el manejo de pacientes con artritis reumatoide (AR) centrado en el papel de los fármacos antirreumáticos modificadores de enfermedad (FAME) sintéticos y biológicos disponibles, que sirvan de referencia para todos los profesionales implicados en la atención de estos pacientes.

Métodos

Las recomendaciones se consensuaron a través de un panel de 14 expertos previamente seleccionados por la Sociedad Española de Reumatología (SER). Se recogió la evidencia disponible mediante la actualización de las 3 revisiones sistemáticas (RS) que se utilizaron para las recomendaciones EULAR 2013, a las que se añadió una nueva RS para dar respuesta a una pregunta adicional. Todas fueron realizadas por miembros del grupo de revisores de la SER. La clasificación del nivel de la evidencia y del grado de la recomendación se realizó utilizando el sistema del Centre for Evidence-Based Medicine de Oxford. Se utilizó la metodología Delphi para evaluar el grado de acuerdo entre los panelistas para cada recomendación.

Resultados

Se emiten un total de 13 recomendaciones sobre el manejo terapéutico de pacientes con AR del adulto. El objetivo terapéutico debe ser tratar al paciente en fases precoces de la enfermedad, con el objetivo de la remisión clínica, teniendo un papel central el metotrexato como FAME sintético de referencia. Se actualizan las indicaciones de los FAME biológicos disponibles, se enfatiza la importancia de los factores pronósticos y se incide en el concepto de optimización de biológicos.

Conclusiones

Se presenta la quinta actualización de las recomendaciones SER para el manejo de la AR con FAME sintéticos y biológicos.

Palabras clave:
Artritis reumatoide
Manejo de la enfermedad
Recomendaciones
Sociedad Española de Reumatología
FAME biológico
FAME sintético
Full Text
Introduction

Rheumatoid arthritis (RA) is the most prevalent form of chronic polyarthritis and has the greatest socio-sanitary impact in society today. RA can cause different degrees of disability, loss of quality of life and even increased rates of mortality. In recent years, important advances have been made in the management and treatment of this disease that have resulted in better patient prognosis, although we are still far from a definitive cure.1

In 2010, we witnessed publication of the fourth and latest update of the consensus document by the Spanish Society of Rheumatology (SER – Sociedad Española de Reumatología) regarding the use of biological therapies in RA.2 In recent years, a vast amount of scientific evidence has been generated about the effectiveness of diverse therapeutic strategies. Meanwhile, new concepts have been developed, such as the optimisation of biological therapies, and new disease-modifying anti-rheumatic drugs (DMARDs) have surpassed their original clinical targets with new indications. This is the reasoning behind this fifth update, which incorporates discussion of these advances and latest insights into a new consensus document.

The present document not only compiles the main aspects of control and treatment with biologic drugs, but it also deals with important aspects in the management of RA, such as early diagnosis, therapeutic objectives, use of chemical drugs (synthetic DMARDs) and comorbidities. The recommendations, however, mainly focus on therapeutic strategies with synthetic and biologic DMARDs.

Similar to previous versions, this document is aimed at healthcare professionals treating patients with RA, especially rheumatologists, as they are usually involved in the management of this disease. These recommendations are not intended to embody a strict protocol for management and treatment of the disease. Instead, they have been created as a foundation to increase the quality of RA patient care and to aid the therapeutic decision-making process.

Methodology

Development of this document began with the empanelling of experts, drawn from an open call to all members of SER. The Committee for Clinical Practice Guidelines and Consensus of SER assessed the candidates’ curricula vitae by evaluating contributions made towards the better understanding of RA. Their objective criteria mainly focused on candidates participation in articles published in high-impact journals during the last 5 years. In the end, the panel of experts was comprised of 14 rheumatologists who are members of SER.

At the first meeting, three different strategies were considered: (a) update the systematic reviews (SR) on which the recommendations of the previous SER Consensus was based2; (b) adapt the recommendations made by the European League Against Rheumatism (EULAR) 20133 for the management of RA with synthetic and biologic DMARDs by updating the 3 EULAR SR4–6 that were the basis for said recommendations; and (c) restructure the recommendations in order to include new clinical questions. After a vote, the expert panel opted to update the EULAR 2013 SR and adapt its recommendations to our environment.4–6 However, some aspects of the EULAR 2013 SR, such as those referring to the effectiveness and safety of biosimilars and kinase inhibitors, were excluded from the present update by SER.

An additional SR was created to respond to a new clinical question posed by the panel of experts: “In adult patients with AR and osteoporosis, how safe is the combined treatment of denosumab and biologic DMARD?” Reports of the 4 SRs written by members of the Evidence-Based Rheumatology group, as well as all related methodological materials (development protocol and search strategies), are available for consultation on request (proyectos@ser.es).

The levels of evidence and grades of recommendation were classified according to the system developed by the Oxford Centre for Evidence-Based Medicine (http://www.cebm.net/oxford-centre-evidence-based-medicine-levels-evidence-march-2009).7

At a second meeting, the panellists presented written proposals for each of the recommendations in order to establish a group consensus on the final draft proposal. The degree of agreement for each of the recommendations was established by means of a 3-round Delphi method.

This document presents the 13 recommendations (Table 1) made by the panel of experts, accompanied by their corresponding levels of evidence (LE), grades of recommendation (GR), degrees of agreement (DA) and percentages of panellists who agreed with the recommendation. Recommendations 3, 5, 6, 11 and 12 have been subdivided into several sections. In accordance with the recommendations, a treatment algorithm has been created (Fig. 1) to summarise the treatment approach after a diagnosis of RA. This document also includes a table that summarises the risk management of each of the biological therapies that are currently available in our setting (Table 2).

Table 1.

SER 2014 Consensus Recommendations.

  Recommendation  LE  GR  DA% votes in favour (DA ≥4) 
        Mean  SD   
It is recommended to begin treatment with a synthetic DMARD as soon as the diagnosis for RA is made.  1a  5.00  0.00  100% 
The recommended therapeutic goal is to reach a state of clinical remission or, if not, a persistently low degree of inflammatory activity, as evaluated by objective, validated indices.  1a  4.92  0.28  100% 
3a  Disease activity in patients with RA should be frequently monitored.  1b  4.69  0.85  92.31% 
3b  Said frequency should be every 1–3 months if the disease is active, a new treatment has been initiated, or the therapeutic goal has not been reached.  4.77  0.60  92.31% 
3c  Said frequency will be every 3–6 months once the therapeutic goal has been reached.  4.46  0.88  92.31% 
3d  DMARD treatment should be adjusted 3 months after the start of the latest treatment regimen if there is no improvement and after 6 months if the therapeutic goal has not been reached.  4.46  0.78  84.61% 
The inclusion of MTX is recommended in the initial therapeutic strategy of patients with RA.  1a  4.92  0.28  100% 
5a  In patients with a contraindication for MTX, it is recommended to initiate treatment with another synthetic DMARD; in Spain, leflunomide is the most commonly used.  1a  4.69  0.63  92.31% 
5b  Sulfasalazine is also an effective therapeutic alternative.  1a  4.46  0.66  92.31% 
6a  The use of a synthetic DMARD, either combined or as monotherapy, is recommended in patients with RA who have not previously received it.  1a  4.85  0.55  92.31% 
6b  This recommendation is made independently of the concomitant use of glucocorticoids.  1a  4.62  0.77  84.61% 
The use of low doses of glucocorticoids is recommended in the initial treatment of RA (combined with one or more synthetic DMARDs) during the first months, with progressive reductions until definitive withdrawal is achieved.  1a  4.31  1.11  84.61% 
When the therapeutic goal is not reached with an initial strategy involving a synthetic DMARD, another synthetic DMARD can be used in sequential or combined therapy, or a biologic DMARD can be added depending on the characteristics of the patient and the presence of factors for a poor prognosis.  4.85  0.38  100% 
In patients with active RA in whom there is an indication to initiate therapy with a biological DMARD, anti-TNF, abatacept, tocilizumab or, under certain circumstances, rituximab can be used in combination with MTX/other synthetic DMARD.  1b  4.69  0.48  100% 
10  In patients with intolerance or a contraindication for a synthetic DMARD, biological treatment can be used in monotherapy. In this case, tocilizumab can be considered as the preferred option.  1b  4.15  0.99  76.92% 
11a  After a failure to respond to an initial biologic DMARD, it is recommended that the patient be treated with another biologic DMARD.  1b  4.69  0.63  92.31% 
11b  If the first agent used was an anti-TNF, the patient can receive another anti-TNF or another biological DMARD with a different mechanism of action.  1b  4.62  0.65  92.31% 
12a  In patients with established RA in remission or persistently low activity, the biological dose can be progressively reduced, especially when used in combination with a synthetic DMARD.  2b  4.38  0.77  84.61% 
12b  It is not recommended to suspend the biological treatment without previous reduction due to the high risk of relapse.  2b  4.62  0.87  92.31% 
13  When defining the therapeutic goal and treatment strategy, including dose adjustments, in addition to the parameters for disease activity and structural damage progression, the presence of comorbidities and drug safety should be considered.  2b  4.77  0.44  100% 

RA: rheumatoid arthritis; SD: standard deviation; DMARD: disease-modifying anti-rheumatic drugs; DA: degree of agreement (1: absolute disagreement, 2: moderate disagreement, 3: neither agreement nor disagreement, 4: moderate agreement, 5: absolute agreement); GR: grade of recommendation; MTX: methotrexate; LE: level of evidence.

Fig. 1.

Therapeutic algorithm for rheumatoid arthritis.

(0.75MB).
Table 2.

Biological Therapy Risk Management.

  Considerations common to all BT  Specific considerations by biological drug
    Anti-TNF  Anakinra  Abatacept  Rituximab  Tocilizumab 
Prior to treatment  (a) Clinical aspects:- Rule out: active infection (including TB, cancer, CHF, cytopenia, demyelinating disease, relevant comorbidity- Rule out recent contacts with TB(b) Complementary studies:- Blood work, biochemistry- Serology HBV, HCV- Chest Rx- Mantoux and Booster; quantiFERON(c) Other measures:- Preventive measures:• Pneumonia and flu vaccines• Assess HBV, meningitis and Haemophilus vaccines according to baseline disease or comorbidities• Assess antiviral treatment if positive HBV; assessment by specialist in hepatology• Avoid vaccines with attenuated or live germs- Instructions for patient:• Symptoms for concern• Hygienic-dietary measures• Planning for pregnancy(d) Special clinical situations:- Pregnancy and breastfeeding: Advise against pregnancy and breastfeeding- Major elective surgery- Immigrant population: evaluation of infections uncommon to our setting  - Consider administration in patients with a history of CHF, ILD, demyelinating disease and HBV/C+Pregnancy:• FDA category: B• Transference through the placenta or breast milk is very low with certolizumab pegol• Interrupt 3–24 weeks before, depending on the biological agent (see drug data sheet)  Pregnancy:• FDA category: B• Interrupt 10–12 weeks before  Pregnancy:• FDA category: C• Interrupt 14 weeks before  - Consider its administration in patients with a history of CHF or other serious cardiac diseases, and ILD- Consider antiviral treatment if HBV+Before each perfusion, premedication should be given with glucocorticoids, analgesia/antipyretic and an antihistamine.Determine immunoglobulin levelsPregnancy:FDA category: CInterrupt one year before  - Consider its administration in patients with a history of demyelinating disease- Evaluate neutrophils, platelets and hepatic enzymesPregnancy:FDA category: CInterrupt 12 weeks before 
During treatment  (a) Clinical aspects:- Appearance of infections (including TB), severe cytopenia, demyelinating disease or optical neuritis, cancer- Appearance or worsening of CHF and lung disease(b) Complementary studies:- Monthly general blood work and biochemistry for the first 3 months, later every 3–6 months.(c) Other measures:- According to patient progress  Appearance of worsened CHFSkin neoplasmsWatch for appearance of neoplasms in COPD patients or with a smoking history    Appearance of COPD or declining lung function in patients with previous COPD  Reactions during perfusionPotential risk for infections, including PMLWatch for development of late neutropenia   
Suspension of treatment  - Appearance of cancer, demyelinating disease or optical neuritis, severe cytopenia, new interstitial lung disease or decline in previous disease, or other severe events related with the drug- Temporary suspension if infection or major elective surgery in perioperative periodAssess pregnancy or breastfeeding           

ILD: interstitial lung disease; COPD: chronic obstructive pulmonary disease; CHF: congestive heart failure; PML: progressive multifocal leukoencephalopathy; BT: biologic therapy; TNF: tumour necrosis factor; HBV: hepatitis B virus; HCV: hepatitis C virus.

Results and DiscussionRecommendation 1. It Is Recommended That Treatment With Synthetic DMARD Be Initiated as Soon as a Diagnosis of RA Is Made

The EULAR 2013 systematic review on the efficacy of synthetic DMARD4 and the related update by SER for this consensus document both demonstrate that treatment with synthetic DMARDs improves the symptoms and signs of RA (LE 1a). Thus, it is logical that the panellists are in complete agreement for this recommendation. Additionally, the panel emphasised the importance of the diagnosis and early treatment of the disease because a recently published SR supports the concept of a “window of opportunity”. Said review has found a significant correlation between disease duration before the start of treatment with DMARD and radiological progression (LE 1a). Moreover, the same study reported that the duration of symptoms before the start of treatment with DMARD has a significant negative influence on the possibility of later maintaining remission in the absence of treatment with DMARD (LE 1a).8

This first recommendation differs from those of EULAR,3 which established the option of initiating treatment with both synthetic and biologic DMARDs. One reason for starting treatment with a synthetic DMARD stems from the fact that subanalysis of the TEAR study in patients with RA and factors for a poor prognosis showed no relevant mid-term differences when treatment was initiated with methotrexate (MTX) monotherapy versus combined therapy either with or without a biologic agent.9 This study revealed the possibility of changing to a combined therapy regimen (with another synthetic DMARD or a biologic drug) 6 months after the start of treatment, which is feasible in our standard clinical practice. Two years after follow-up, 30% of patients continued treatment with MTX monotherapy, and no relevant differences were observed with those groups that received combined therapy.9

Given that early therapy with DMARDs has a relevant effect on later disease progression, and despite the fact that diagnosis of RA is based on the personal impression of the rheumatologist treating the patient, the 2010 classification criteria can help re-adjust the way in which patients with initial onset arthritis are treated by providing greater sensitivity for the diagnosis of RA.10 Considering that the risk-benefit correlation of early therapy with a synthetic DMARD, especially MTX, is favourable and its management is well known,6 the prescribing of synthetic DMARDs could be extended to patients with undifferentiated arthritis if the rheumatologist suspects that it could progress to RA. Different therapeutic strategies have been tried in these patients in an attempt to halt the progression towards RA and to improve the possibility of achieving treatment-free remission.11–13

Recommendation 2. The Recommended Therapeutic Goal Is to Reach Either a State of Clinical Remission or, Alternatively, a Low Degree of Persistent Inflammatory Activity, as Evaluated by Objective, Validated Indices

This recommendation is based on the fact that it is now possible to achieve clinical remission in a significant percentage of patients. Although there is no unequivocal definition for the concept of “remission”, the current conviction is that clinical remission in RA is an attainable therapeutic goal.

Several observational studies show that 20%–35% of patients with early RA reach clinical remission.14 Furthermore, approximately half or more of patients with early RA who are treated with intensive therapy (i.e., with a combination of biological and synthetic DMARD, begun either at detection or very early on)15,16 or with a strategy of close therapeutic control of the disease,17,18 achieve short-term remission. Nonetheless, long-lasting clinical remission is much less frequent.19,20 The presence of long-term remission is associated with less advanced structural damage and lower disability rates.21,22 It has also been well established that the outcomes are worse if the patient has persistent inflammatory activity, even if low grade.23

The previous SER consensus proposed a low degree of activity as a therapeutic goal, defined quantitatively by the cut-off points of validated activity indices.2 The latest EULAR 2013 recommendations3 insist that remission is an attainable objective in a significant percentage of patients and advocate the use of stricter definitions, such as the ACR/EULAR criteria,24 which correlate better with a state of practical absence of synovitis. The reason is that the definition of remission based on the Disease Activity Index (DAS28), which is widely used in our clinical practice, has important limitations; for example, patients with several inflamed joints and radiological progression can be considered in remission based on this scale.25 The ACR/EULAR remission criteria, initially designed for use in clinical trials24 and validated under daily clinical practice conditions,26 are stricter; only 7%–20% of patients in observational studies meet these criteria.27–29 In addition, one must also consider that factors unrelated to RA, such as subjective patient characteristics30 or the presence of comorbidities,29 can mean that some patients do not meet remission criteria. Thus, a significant percentage of patients either do not reach clinical remission or their remission is not long-lasting. In these cases, especially in patients with established RA, a low degree of inflammatory activity can also be considered a reasonable therapeutic goal (defined by validated indices)3,24 since better outcomes have also been described in the presence of low inflammatory activity compared to intermediate or high inflammatory activity.31

As for the need to include criteria from imaging studies when defining remission, it is well known that a significant percentage of patients in apparent clinical remission have subclinical synovitis that is detectable with MRI or ultrasound.32 Approximately half of such patients have active synovitis with a positive Doppler signal on ultrasound.33,34 The presence of subclinical synovitis is a predictive factor for the loss of remission during follow-up34,35 and could explain the radiological progression observed in patients in an apparent state of clinical remission.33,34 Furthermore, the use of stricter remission criteria using the ACR/EULAR or SDAI criteria is associated with lower rates of subclinical sinovitis36,37 than when DAS28 is used. Nonetheless, the panel holds that there is still no solid evidence to establish a definition of remission that includes an “imaging studies” dimension. However, the panel recognises the importance of imaging techniques for assessing the presence of synovitis in cases of difficult or uncertain physical exploration, which can provide additional information for the management of some patients.

Moreover, as in the previous SER consensus document,2 the therapeutic goal has not been reached if there is persistent inflammation that remains unresolved following local therapeutic measures in important joints or when significant radiological progression is detected.

Recommendation 3. Frequent Monitoring of Disease Activity in Patients With RA Is Recommended. This Frequency Should Be:

  • Every 1–3 months if the disease is active, a new treatment has been initiated, or the therapeutic goal has not been reached.

  • Every 3–6 months once the therapeutic goal has been reached.

Treatment with DMARDs should be adjusted 3 months after the start of a new dosage if there is no improvement, and 6 months thereafter if the therapeutic objective has not been reached.

The “treat to target” strategy has demonstrated clear benefits in efficacy with regards to overall disease evolution over the mid- and long-term, regardless of the pharmacological treatment applied. In the context of this strategy, this recommendation attempts to establish follow-up schedules and times for changes in therapy.

There are no specific data regarding the appropriate periodicity required for monitoring patients with RA. The studies included in both the EULAR 2013 systematic review on the efficacy of DMARDs,4,5 as well as the updates of said revisions done by SER for this consensus, were designed to assess treatment effectiveness, not to define monitoring strategies. Even so, several studies designed to assess therapeutic strategies propose monitoring periods ranging from 4 weeks to 4 months16,38–49 (LE 1b-2b). Only one study assessed disease activity every 24 weeks.50 The period that is considered appropriate before making treatment changes in these studies ranges from 4 to 26 weeks.

In this context, the study by Aletaha et al.51 demonstrates a correlation between disease activity after 3 months and after one year of treatment Therefore, the response reached after 3 months of treatment is highly predictive of the clinical progression during the first year.

In general, the results of the studies mentioned above are applicable to the Spanish healthcare system. Nonetheless, given the organisational differences at various hospitals, the creation of monographic treatment units or, at least, the structure needed, would provide for preferential and frequent care during periods of treatment adjustment or inflammatory activity. In addition, given the variability of resources in our health care system, the panel believes that the collaboration of other medical professionals, such as the nursing staff, would be necessary for successful compliance with this recommendation.

Recommendation 4. It Is Recommended to Include MTX in the Initial Therapeutic Strategy of Patients With RA

This recommendation, included in those of EULAR 2013,3 has been adopted unchanged by the panel of experts responsible for this consensus. Said recommendation sanctions the inclusion of MTX in the therapeutic strategy, without making allusions to administration as monotherapy, therapy in combination with other synthetic DMARDs, or even exceptional cases in which therapy combined with another biologic agent could be initiated.

MTX is not merely very effective for the treatment of RA, but in fact is regarded as the drug of choice. In the most important studies involving MTX as a monotherapy in patients who had not previously received it, 25%–50% reached ACR70, which in many cases meant remission or low activity.52–54 No less relevant is its impact on structural damage. The first study that compared MTX monotherapy with a biological agent (etanercept) in monotherapy or combined with MTX demonstrated that, over the course of 2 years, 60% of patients who received MTX monotherapy did not progress radiologically; in 85%, the change in the Sharp score was less than or equal to the minimal detectable change (5.72 units).55 MTX should be used in rapid escalation such that, in a period of 8–16 weeks, the optimal dose is reached; i.e., at least 15mg (the maximum is generally 25mg).56 Administration is always weekly and preferably in one oral or parenteral dose. Although it is difficult to determine how rapidly dosage is escalated in our country, one must be aware of the fact that, in most cases, the minimum effective dose is approximately 15mg weekly and can be increased to 25–30mg per week. The emAR II study reported that the average maximum dose of MTX in 2008–2009 for patients with RA was 15mg/week (interquartile range from 10 to 20mg/week). Thus, a significant number of patients were probably not receiving the optimal drug dosage.57

Treatment with MTX involves the administration of folic or folinic acid 24h after receiving a dose of MTX; the former does not affect the efficacy of MTX and reduces the toxicity of this drug.58,59 When this occurs, it is not severe and does not require suspension of treatment; rather, a common practice is to increase folate supplementation.58,59 Patients who do not tolerate weekly 15mg doses are usually given smaller weekly doses ranging between 7.5 and 12.5mg.

Generally speaking, MTX is taken orally. However, when a patient receives weekly doses of 20mg or greater, parenteral MTX administration is preferable, since the oral bioavailability of this drug diminishes as the dosage increases. Moreover, the parenteral bioavailability increases in a linear manner.60 There is ample evidence that patients who respond insufficiently to oral doses have a better response to the parenteral route.61–64 Digestive tolerability, as well as that of other organs, seems to be better with parenteral administration.61–64

The efficacy data for MTX remain indisputable, both in monotherapy and when administered in combination with either synthetic or biologic DMARDs. In fact, only 2 agents, tocilizumab and tofacitinib, have been shown to be superior in monotherapy when compared with MTX.65,66

In general, MTX is well tolerated by most patients. Nonetheless, there are certain patients with severe hematologic, hepatic or pulmonary involvement in whom MTX would be contraindicated from the outset. In these cases, it is necessary to start the treatment with other drugs that have also proven effective. See recommendation 5.

Recommendation 5. In Patients With Contraindications to MTX, It Is Advisable to Initiate Treatment With Other Synthetic DMARDs. In Spain, the Most Frequently Used DMARD Is Leflunomide, Although Sulfasalazine Is Also an Effective Therapeutic Alternative

Leflunomide is an effective drug in RA from both a clinical standpoint and in terms of radiological progression. No differences have been found in comparative studies with MTX, whether the doses of MTX used in these studies were optimal for this drug remains debatable.67,68 Leflunomide is also effective when combined with biological agents,69,70 although the available evidence compared to MTX is scant. Indeed, the main clinical assays involving biological agents have used MTX.

The results from studies done with leflunomide are consistent; therefore, this drug can be considered the first alternative to MTX when use of the latter is not possible. In Spain, leflunomide (at a dose of 20mg per day in monotherapy) has become the most widely used drug in patients with intolerance or contraindications to MTX.57

Sulfasalazine with enteric coating at a dose of 3–4g per day has demonstrated effectiveness similar to MTX from both a clinical and radiological standpoint.71–75 Sulfasalazine has the advantage of being safe during pregnancy, which offers an added advantage over the use of MTX or leflunomide. One disadvantage to using such doses is the number of tablets that the patient must take each day (between 6 and 8), which could affect treatment compliance. Nonetheless, a randomised clinical trial (RCT) was designed with 3 therapeutic approaches in patients with RA who received: (a) leflunomide at 20mg per day after a load of 100mg the first 3 days; (b) placebo; and (c) sulfasalazine up to a maximum of 2g/per day for 24 weeks. The results demonstrated that both drugs boasted superior efficacy versus placebo, with no differences between the two.76 The decision to administer 2g of sulfasalazine as a monotherapy was based on previous studies in which this dose had been shown to be effective both at the initiation of treatment as well as during maintenance,74,75,77–79 although occasionally the dose of 2g was administered in combined therapy.

Therefore, in those patients in whom MTX is contraindicated, sulfasalazine could be taken at a dose of 2g in monotherapy if the patient does not tolerate higher doses. However, what is most common is to prescribe these doses in combination therapy with MTX and hydroxychloroquine.80–82 In Spain, sulfasalazine with enteric coating is not available, which explains the poor tolerability of 3–4g doses in a large number of patients. Thus, the use of this drug in RA is very limited in our country.57,83

Another widely used drug in the 1970s and 1980s was gold salts. This drug was considered an alternative to MTX in the first EULAR recommendations.84 However, gold salts disappeared from the list of recommended drugs in the EULAR 2013 recommendations.3 The main reason why gold salts are not used anymore is due to the advent of MTX and from the severe side effects (hematologic and renal toxicity) caused by gold salts in some patients. As a result, in many countries (including Spain), the drug is not widely available.

Last of all, chloroquine and, fundamentally hydroxychloroquine, are drugs that are usually found to be safe, effective agents in cases of very mild RA with little inflammation.85,86 Nonetheless, their use as a monotherapy in RA is practically non-existent and has been almost wholly limited to combined therapy, where they have been widely used with sulfasalazine and MTX.80–82 Like sulfasalazine, hydroxychloroquine is safe for use during pregnancy,87 which offers some advantages in certain clinical situations. Other drugs, such as azathioprine, cyclosporine and cyclophosphamide, are not discussed in this section because their initial use in RA is practically non-existent.

Recommendation 6. The Use of Synthetic DMARDs Is Recommended in Patients With RA Who Have not Previously Taken Them, Either as Monotherapy or in Combined Therapy. This Recommendation Is Made Independently of the Concomitant Use of Glucocorticoids

This recommendation once again underlines the fact that the first treatment strategy in a patient with early stage RA should include synthetic DMARD(s). Given the current controversy over the use of DMARDs in monotherapy or combined therapy, this recommendation was initially much debated, although it ultimately met with a high level of agreement.

The EULAR 2013 SR regarding the efficacy of synthetic DMARDs4 confirmed the effectiveness of these drugs in RA based on several RCTs verifying this fact (LE 1a). Specifically, the efficacy of MTX was demonstrated in both first- and second-line DMARD use (LE 1a). Likewise, the EULAR 2013 recommendations3 identified 5 studies43,88–91 suggesting that combined therapy with synthetic DMARDs was superior to monotherapy with MTX (LE 1a). Among these 5 studies, one even proposed that the efficacy of combined therapy with a synthetic DMARD could be equal to that of biological therapies in combination with MTX. Nonetheless, the EULAR 2013 SR also argued that said studies suffered methodological limitations that hindered their interpretation. It also noted that other studies had demonstrated that sequential monotherapy (by changing a DMARD if there is no response) was as effective as combined therapy in terms of clinical, functional and structural results.92 In spite of the latter, the EULAR 2013 recommendations concluded that combined therapy with or without steroids was an adequate strategy in patients with RA. This combined therapy should generally include MTX, since other combinations that do not include it have not been sufficiently studied.

The previously mentioned SR4 specifically addressed 2 of the 5 studies mentioned above. The tREACH study (LE 1b) is an RCT with three treatment modalities in which patients with recent-onset RA were randomised to receive: (a) a combination of MTX, sulfasalazine and hydroxychloroquine with intramuscular glucocorticoids; (b) the same combination, but with oral glucocorticoids at a decreasing dosage; and (c) only MTX together with the same descending dosage of oral glucocorticoids. The study found that, after 3 months of follow-up, there were no differences in DAS28, ESR and swollen joints among the three treatment groups. However, there were differences in HAQ, painful joints and CRP which were superior in the monotherapy group compared to the combined therapy groups.88 In contrast, the TEAR study (LE 1b) found no differences in clinical results or in radiological damage between the combined therapy used from the start versus sequential therapy.89

The update of this SR has also identified 2 new studies on this subject. The first (LE 1b), a non-inferiority study, compared a COBRA-light prescription (prednisolone in decreasing dosage + MTX) with the original COBRA prescription (decreasing doses of prednisolone+MTX+sulfasalazine). After 6 months, the authors found that both interventions reduced disease activity in early RA and no differences were seen between the two.93 The second study (LE 1b), a post hoc study of the aforementioned TEAR study, analysed the MTX group versus the combined therapy groups (etanercept+MTX, and MTX+sulfasalazine+hydroxychloroquine) and found that patients who received MTX monotherapy (after 102 weeks) maintained benefits similar to groups that received combined treatments.9

As for safety, the different studies analysed revealed that synthetic DMARDs are safe, both in monotherapy and in combined therapy. Only 2 studies (LE 2a) suggest an increase in the rates of cancer94 and infectious lung disease95 when MTX (versus another synthetic DMARD) was involved.

The results of the identified studies can be directly applied to our healthcare system since the therapeutic agents evaluated are commonly used in our setting. Both strategies, monotherapy and combined therapy, are effective in patients with RA, with or without steroids. Preferences, expectations and side effects should be considered when discussing treatment options with patients.

Recommendation 7. Low Doses of Glucocorticoids Are Recommended in the Initial Treatment of RA (in Combination With One or More Synthetic DMARDs) During the First Few Months. Doses Should Be Progressively Reduced Until Their Use Can Be Completely Withdrawn

There is evidence about the usefulness of low doses of glucocorticoids in the initial treatment of RA. What has not been defined, however, is the optimal duration of treatment since only 2-year studies have been published. Based on expert opinions, the EULAR 2013 SR suggests not prolonging administration for more than 6 months.

The EULAR 2010 SR on the management of RA with glucocorticoids96 identified 11 RCT (including 3 Cochrane SR). It concluded that the addition of low glucocorticoid doses (7.5mg/day or less) to the initial synthetic DMARD treatment of recent-onset RA is clinically effective and reduces radiological progression when used continuously for 2 years (LE 1b).97–99 The use of glucocorticoids in RA patients with more than 2 years of disease progression only led to improvements in terms of signs, symptoms and functional state.100 The use of glucocorticoids thus offers a bridge therapy until the effect of a new DMARD provides better clinical results after one month of treatment (LE 1b). However, its long-term clinical and radiological usefulness still has not been determined.101,102

The EULAR 2013 SR4 identified 2 new RCT (SAVE and CAMERA-II [LE 1b]) that evaluated the efficacy of glucocorticoids in recent-onset RA combined with synthetic DMARDs. The CAMERA-II study demonstrated that 10mg per day of prednisone in a control strategy that was closely associated with MTX (up to 30mg/week) reduced radiological damage after 2 years of treatment, and increased the probability of a clinical response (e.g., higher remission rates, lower levels of DAS and better results in HAQ). This treatment strategy also yielded clinical improvement in less time and had less need for other future treatments (e.g., other synthetics or biologic DMARDs).103

The SAVE study demonstrated that administration of a single dose of 120mg of prednisolone to patients with very early stage undifferentiated arthritis was not able to prevent the later progression of RA. Nor did it induce clinical remission or the need to initiate a synthetic DMARD.104 The tREACH trial (LE 1b), mentioned previously, compared clinical effectiveness in patients with early-phase RA (<1 year of symptoms) for one year in 3 treatment modalities: (a) initial triple therapy with a single intramuscular dose of glucocorticoids (120mg 6-methylprednisolone or 80mg triamcinolone); (b) initial triple therapy with oral glucocorticoids withdrawn over 10 weeks; and (c) MTX as a monotherapy with glucocorticoids as in (b). The authors used a “treat to target” strategy for the dose adjustments and intensification to biological therapy. After 3 months, better clinical results were observed (40% less intensifications), but there were no differences in the radiological progression after one year. No clinical or radiological differences were found between the 2 bridge therapies involving glucocorticoids.105

The results of the different studies identified in the 2 EULAR SR are consistent and demonstrate the effectiveness of adding glucocorticoids to the initial treatment (up to 24 months) in patients with recent-onset RA. Some studies also showed that monotherapy with glucocorticoids (e.g., with no associated synthetics or biologic DMARDs) was effective in controlling the symptoms and signs of RA.106,107 Despite the fact that some controlled clinical trials suggest that the side effects associated with the use of low doses of prednisone are modest,108 there is still not sufficient evidence about its long-term safety and efficacy in order to recommend its prolonged use, except in unique situations.

Recommendation 8. When the Therapeutic Goal Has not Been Reached With the Initial Synthetic DMARD Strategy, Either Another Synthetic DMARDs Can Be Used in Sequential or Combined Therapy, or a Biologic Agent Can Be Added, Depending on Patient Characteristics and the Presence of Poor Prognostic Factors

The panel of experts determined that risk stratification is an important aspect in the management of RA. Factors indicating a poor prognosis include a state of high disease activity, positivity for rheumatoid factor and/or anti-citrullinated protein antibodies (ACPA) and the presence of radiographic erosions.109,110

In patients who do not present factors for a poor prognosis and who have failed to respond to an initial strategy with a synthetic DMARD (which almost always includes MTX) the panel recommends using another strategy with a synthetic DMARD as monotherapy (leflunomide or sulfasalazine) or in combination (MTX+hydroxychloroquine+sulfasalazine, or MTX+leflunomide). In patients with some type of factor indicative of a poor prognosis, the recommendations are as follows: add a biologic DMARD or, if the first strategy involved an initial synthetic DMARD in monotherapy, then a combination of a synthetic DMARD could be used (MTX+hydroxychloroquine+sulfasalazine or MTX+leflunomide).

In RA patients who do not adequately respond to MTX, the addition of hydroxychloroquine+sulfasalazine has demonstrated a clinical effectiveness equivalent to the addition of an anti-TNF agent in 3 RCT (IFX in the SWEFOT study,91 etanercept in the TEAR9,89 and RACAT90 study) all of which were of high quality (LE 1b), although the radiological evolution in one of the clinical trials was significantly less in the patient group that received an additional anti-TNF agent.91

There are no data from clinical assays on the use of MTX+leflunomide compared with the addition of a biological agent in patients with RA and insufficient response to MTX. Therefore, based on existing scientific evidence, this panel cannot recommend its use in RA patients with factors for a poor prognosis and insufficient response to monotherapy with MTX. Nonetheless, the panel is conscious of the poor tolerability of Spanish patients to sulfasalazine, as well as the widespread acceptance in our healthcare system regarding the effectiveness and safety of combining MTX and leflunomide. In selected cases, the combination of MTX and leflunomide could be a reasonable alternative to the use of MTX+hydroxychloroquine+sulfasalazine.

In addition, after an insufficient response to MTX in RA patients with no factors for a poor prognosis, the use of a biologic DMARD could be considered in those cases in which (1) the patient characteristics suggest the likelihood of side effects with other synthetic DMARDs (monotherapy or combined) or (2) when there is deficient compliance with the therapeutic prescription.

The panel emphasises that, following a poor response to the initial strategy using a synthetic DMARD, it is of the utmost importance to closely follow the patient. The basic objective is to at least reach a state of low disease activity and, ideally, to enter remission within 6 months of having initiated therapy with DMARD.

Given the importance of the efficient use of healthcare resources, the panel believes that higher mid- and long-term cost-effectiveness is best achieved by inducing a state of disease remission during the first 6 months of treatment. Therefore, the use of a biologic DMARD is justified when the first strategy (synthetic DMARD) has failed in those patients with factors for a poor prognosis.

Recommendation 9. In Patients With Active RA in Whom There Is an Indication to Initiate Therapy With a Biologic DMARD, It Can Be Used in Combination With MTX/Another Synthetic DMARD, Anti-TNF Drugs, Abatacept, Tocilizumab or, in Certain Circumstances, Rituximab

Based on new indications and evidence for certain biological drugs, the number of available therapeutic agents has increased.

In patients with RA and insufficient response to MTX, the 9 biological agents available in Spain (abatacept, adalimumab, anakinra, certolizumab, etanercept, golimumab, infliximab, rituximab and tocilizumab) show promising evidence (LE 1a) not only of the rescue effect, but also of the greater therapeutic effectiveness of the combine use of a biologic DMARD+MTX versus continued MTX monotherapy.111–116 In this scenario, combined biologic DMARD+MTX vs MTX presents a relative risk (RR) of reaching an ACR70 response of 4.07 (95% CI: 3.21–5.17). In this context, there are also data that confirm demonstrating the higher efficacy of combining a biologic DMARD+synthetic DMARD (other than MTX) vs a synthetic DMARD in those patients with an insufficient response to a synthetic DMARD (RR to obtain an ACR70 response: 4.74 [95% CI: 2.63–8.56]).5

Combining a biologic agent with another is not recommended since such a regimen does not provide greater effectiveness in controlling RA and the risk of adverse events, especially infections, increases. Currently there remains a lack of quality scientific data analysing the safety of using denosumab with other biological therapies in patients with RA.

All biologic DMARDs (except anakinra) show a similar therapeutic efficacy in the treatment of those RA patients who respond poorly to MTX (LE 1a).5 Two clinical assays (LE 1b) have directly compared the therapeutic effect of 2 biological agents: the AMPLE study,116 which compared abatacept+MTX versus adalimumab+MTX in cases of RA with less than 2 years of mean disease duration; and the ADACTA study,117 which compared tocilizumab on monotherapy versus adalimumab on monotherapy. In the former, the clinical and radiological responses were similar, while in the latter tocilizumab was more effective than adalimumab.

In active RA patients with an insufficient response to MTX, biological treatment is usually initiated with anti-TNF combined with MTX. This is done for several reasons, including the greater likelihood that the physician has experience with these agents, as well as the greater availability of long-term safety data. Clinical trials and meta-analyses52,118 (LE 1a) have demonstrated that anti-TNF combined with MTX is superior to these biological agents in monotherapy, especially with regards to the slower radiological progression of joint damage (although there is still some doubt about the actual magnitude of this effect).

In RA patients who respond poorly to MTX, the choice of abatacept, rituximab or tocilizumab depends on different factors: patient preference, drug cost, comorbidities, administration pathway, physician experience and indications (rituximab has not been approved when there is failed response to MTX). These biological agents seem very similar to each other, with the following considerations (LE 1b)119–122:

  • The response to rituximab is greater in patients with rheumatoid factor and/or ACPA+.

  • The immunosuppression/immunomodulation exerted by these 3 biologic DMARDs reverts more rapidly with abatacept and tocilizumab than with rituximab, the effects of which on immune function can be longer lasting.

  • Certain comorbidity factors (demyelinating disease, congestive heart failure, lymphoma) can contraindicate the use of an anti-TNF and favour administration of a biological agent.

  • Abatacept may carry less risk of triggering serious infections, which is of particular relevance in elderly patients and those with comorbidities.

Recommendation 10. In Patients With an Intolerance or Contraindication to a Synthetic DMARD, biological treatment can be used in monotherapy. In Such Cases, Tocilizumab Can Be Considered the Preferential Option

This recommendation was the subject of intense discussion among members of the panel of experts, and it was necessary to twice revise it in order to reach agreement consensus of 77% after the third Delphi round. The writing in the present recommendation attempts to explain that while tocilizumab is more likely to be effective in this patient profile, any of the other options approved for RA are also acceptable.

This recommendation is based on 3 lines of evidence. The first stems from the only direct comparative study between 2 biologic DMARDs in monotherapy: ADACTA.117 This was a randomised, double-blind study with a low risk for bias (LE 1b) that demonstrated the superiority of tocilizumab versus anti-TNF adalimumab. This proved true both in monotherapy, when the efficacy was measured by DAS28, and when using other indices with less weight of the acute phase reactants (ACR20, 50, 70, SDAI or CDAI). Moreover, tocilizumab is the only biologic DMARD that has demonstrated its biological superiority over MTX in monotherapy65,123,124 (LE 1b). In fact, none of the other biologic DMARDs have consistently shown an effectiveness superior to MTX in monotherapy, except in terms of its effect on structural damage or its quicker action.5,56,118 Third, monotherapy with tocilizumab has an effectiveness similar to the combined MTX therapy125 (LE 1b), while anti-TNF is less effective in monotherapy than when combined with MTX.5,118,126 Therefore, these 3 lines of evidence support the contention that tocilizumab is more effective than anti-TNF in monotherapy treatment of RA patients.

A possible limitation of this recommendation lies in the fact that there is only direct comparative data for tocilizumab with adalimumab. Nonetheless, considering the existence of data from multiple studies reporting an effectiveness very similar to that of other biologic DMARDs in monotherapy,5,56,118,126 as well as the results mentioned in the previous paragraph, it is reasonable to extrapolate the data on adalimumab to other anti-TNF drugs in this context, although for this reason the grade of recommendation is reduced to B.

One clarification is necessary in the case of etanercept. There was an open 16-week study (ADORE) in which etanercept monotherapy reached an efficacy similar to its combination with MTX.127 However, two other studies, the 52-week double-blind randomised TEMPO study118 (LE 1b) and the 52-week randomised JESMR study126 (LE 1b), showed that the combination of etanercept+MTX was more effective compared to etanercept monotherapy. Furthermore, this greater effectiveness was also noted in a recent Cochrane meta-analysis.128 There are no studies comparing etanercept with other anti-TNF drugs in monotherapy. Therefore, despite certain contradictory evidence surrounding this drug, and in the absence of direct comparative studies with other anti-TNF in monotherapy, the majority of the panel decided that there is no consistent evidence to regard etanercept differently from the use of other anti-TNF drugs in monotherapy.

Lastly, the panel would like to emphasise that the intention of this recommendation is not to support monotherapy with biological agents. Rather, in line with the EULAR recommendations,117 there is agreement that a given biologic DMARD should preferentially be used in combination with a synthetic DMARD, even in the case of tocilizumab.

Recommendation 11. Following a Failed Response to an Initial Biological DMARD, the Panel's Recommendation Is to Treat the Patient With Another Biologic DMARD. If an Anti-TNF Had Been Used, the Patient Can Receive Another Anti-TNF or Another Biologic DMARD That Utilises a Different Mechanism of Action

At least 5 randomised, double-blind clinical trials129–133 have demonstrated the effectiveness of abatacept, rituximab, tocilizumab and golimumab in an analysis of the primary objective. Moreover, a subanalysis studied the efficacy of certolizumab in patients who had not previously responded to another biologic agent (LE 1b). There are no clinical trials comparing the effectiveness of secondary biological agents. However, a recent meta-analysis134 involving indirect comparisons concluded that in patients who have failed to respond to an anti-TNF, the use of abatacept, rituximab or tocilizumab can be superior to the use of a second anti-TNF (LE 1a). This meta-analysis supports observational studies of registries135–139 that compared the effectiveness of a second anti-TNF with rituximab, abatacept or certolizumab (LE 2b).

All of the published evidence shows the effectiveness of using a second biological agent in patients who failed to respond to an anti-TNF. There is a lack of published data on the effectiveness of a second biological agent in patients who did not respond to biologicals other than anti-TNF.

These results are applicable to our health care system, where anti-TNF is generally used as the first biological agent.

In clinical trials, the safety of the second biological drug did not seem very different from the control group129–133 (LE 1b).

Recommendation 12. In Patients With Established RA in Remission or Low Persistent Activity, the Biological Dose Can Be Progressively Reduced, Especially When Used in Combination With a Synthetic DMARD. Due to the High Risk of Relapse, Suspension of Such a Biologic Treatment Is not Recommended Without Previous Reduction

One area of great interest and uncertainty is whether the dose can be reduced, or the biological treatment interrupted, while still retaining the same degree of efficacy.

Several studies46,140–147 indicate that in cases of established RA, withdrawal of anti-TNF is usually not very effective.

In the PRESERVE study, 12 months after the suspension of anti-TNF therapy, 57% of patients with low activity had a new disease episode, compared with 21% of those who reduced the dose of etanercept (25mg etanercept/week+MTX) and 18% who continued with complete doses (50mg etanercept/week+MTX).143 In the CERTAIN study, only 3 out of 17 patients who had reached remission (CDAI ≤2.8) by week 24 of treatment with certolizumab were able to maintain it by week 52.46 Last of all, in the DOSERA study, only 13% of patients who suspended anti-TNF maintained a state of low activity/remission (DAS28 ≤3.2) versus 44% of patients who received half the dose and 52% that continued at full doses (50mg etanercept/week+MTX).147

In established RA, several observational studies about the withdrawal of anti-TNF found 12-month remission rates between 25 and 43%,140,145 and for low activity rates ranging from 16146 to 18.5141 to 55%.145

In cases of recent-onset RA initially treated with anti-TNF, withdrawal of the biological drug is more effective. After suspension of anti-TNF, the TNF 20 study found 12-month remission rates of 70%148 and in the BeST study the percentage was 52% after 5 years. The remaining 48% of the patients required treatment renewal after 17 months (IQR 3–47).149,150 In the IDEA study, 78.6% of the patients in remission for more than 6 months maintained said state when the anti-TNF was suspended.44 In the PRIZE study, 38.5% of patients in remission who continued with MTX alone remained in remission at week 39, versus 63.5% of the patients treated with half of etanercept+MTX and 23% treated with placebo.151 Last of all, in the HIT-HARD study, there were no differences in the DAS 28 between patients randomised to receive MTX+placebo versus MTX+adalimumab during the first 24 weeks versus those in whom the treatment with adalimumab was interrupted and then continued with MTX+placebo, although significant differences were observed in terms of radiographic progression.152 The OPTIMA study reported that most patients who discontinued treatment with adalimumab after reaching the objective of low activity after 6 months of treatment with adalimumab+MTX exhibited either low activity or remained in remission for the following 52 weeks.16

There are studies with agents other than anti-TNF. In the ORION study, which included patients treated with abatacept in remission (DAS28-CRP <2.3), 41.2% whose treatment was suspended maintained remission at week 52 versus 64.7% of those who continued with treatment.153 In the AVERT study, 14.8% of the patients in remission with abatacept+MTX after 12 months of treatment maintained remission 6 months after the withdrawal of all treatment. In those treated only with abatacept, 12.4% were able to maintain said state 6 months after suspension of the biological drug.154

The DREAM study, which evaluated remission rates after the suspension of tocilizumab, found that after 52 weeks 13.4% of the patients continued with low activity and 9.1% reached remission.155 In the ACT-RAY, 50.4% of the patients in remission at week 52 (those treated with either tocilizumab+MTX or tocilizumab+placebo) remained in remission for 52 weeks after tocilizumab was withdrawn.156

Regarding the effectiveness of biological therapy dose reduction in maintaining the state of remission or low activity, the PRESERVE,143 DOSERA147 and PRIZE151 studies-all of which compared the efficacy of the complete withdrawal of a biological agent+MTX (etanercept in 3 cases) versus dose reduction-found that reduced dosing was less effectiveness compared to a full dosage. These differences, however, were not statistically significant.

Additionally, three observational studies showed that dose reductions within a “treat to target” strategy can be effective in clinical practice.146,157,158

A more realistic approach, although one with much less evidence behind it, is dose reduction once the therapeutic objectives are reached. With this strategy the results are slightly lower than with treatment at full doses. Recently, SER has published a consensus document that makes recommendations based mainly on expert opinions about this subject.159

Recommendation 13. Comorbidities and Drug Safety Should Be Considered When Defining Therapeutic Goals and Treatment Strategies (Including Dose Adjustments, Disease Activity Parameters and Progression of Structural Damage)

The purpose of this recommendation is to address the potential existence of comorbidities when treating RA. The presence of comorbidities should be considered in addition to the risks and possible adverse effects of these drugs.

The presence of comorbidities can effect both treatment strategies and efficacy results (LE 2b).160–162 Currently, there are no specific criteria to indicate biological agents for use in patients with significant comorbidities. In such patients, even in those with high disease activity or progressive structural deterioration, the start of a given therapies is often delayed. The assessment of the risk/benefit ratio in these patients would help to select the most appropriate medication and adjust the dose (LE 5). In studies that have examined treatment adjustment strategies, the safety profiles among the different dosages prescribed proved similar.16,143,163

The therapeutic objective should be to reach remission or low disease activity. Nevertheless, due to the aforementioned considerations, attaining either goal in patients with comorbidities should not assume all precedence and, in fact, good results can also be achieved.

In patients with RA, a persistently high degree of inflammation can be associated with the appearance of comorbidities. Therefore, effective RA treatment can also help prevent them (LE 2b).164–168

The results on which this recommendation is based have a low LE given that the presence of comorbidities is usually a cause for exclusion in clinical trials. The conclusions are indirectly extracted from case–control studies or clinical trials in which such patients were not the primary objects of study. Clinical trials are usually short term; due to the heterogeneity of patient groups in observational studies, it is therefore difficult to make definitive conclusions.

Nevertheless, this recommendation was easily agreed upon because it is not uncommon to encounter patients presenting comorbidities, factors for poor prognosis or with secondary effects and in whom complete remission or low disease activity should not be the primary objective. Nonetheless, this does not mean that good results cannot also be achieved.

Ethical disclosuresProtection of human and animal subjects

The authors declare that no experiments were performed on humans or animals for this study.

Confidentiality of data

The authors declare that no patient data appear in this article.

Right to privacy and informed consent

The authors declare that no patient data appear in this article.

Conflict of Interests

Raimon Sanmartí is a member of the Advisory Council for Hospital Outpatient Medication Dispensation for the Catalonian Public Healthcare System (Catsalut). He has received: funding to attend courses and medical congresses from MSD, Abbvie, Roche and Pfizer; professional fees as a lecturer from MSD, Abbvie, Roche, Pfizer, UCB and Bristol; funding for educational programmes or courses from Bristol and MSD; funding for research participation from Roche; and consulting fees from MSD, Abbvie, Roche, Pfizer, UCB and Bristol.

Susana García declares no conflict of interests.

José María Álvaro-Gracia has received funding to attend courses and medical congresses from Pfizer and Roche; professional fees as a lecturer from BMS, Abbvie, MSD, Pfizer, UCB, Roche, Jansen-Cilag, Abbvie and Amgen; funding for research participation from UCB, Roche and Amgen; and consulting fees from BMS, Pfizer, UCB, Roche and Jansen-Cilag.

José Luis Andreu has received funding to attend courses and medical congresses from Pfizer, Abbvie, Gebro and Menarini; professional fees as a lecturer from Abbvie, GSK, Roche and UCB; and consulting fees from UCB and GSK.

Alejandro Balsa has received funding to attend courses and medical congresses from Roche, BMS, Pfizer, Abbvie and MSD; professional fees as a lecturer from Roche, BMS, Pfizer and Abbvie; funding for educational programmes or courses from Roche and Pfizer; funding for research participation from Pfizer; and consulting fees from Roche, BMS, Pfizer, Abbvie and MSD.

Rafael Cáliz has received funding to attend courses/medical congresses from MSD, Roche, Abbvie and Pfizer; and professional fees as a lecturer from BMS, Menarini, Roche, Pfizer and GSK.

Antonio Fernández-Nebro has received funding to attend courses and medical congresses from MSD, Pfizer, BMS, Abbvie, Roche and Angen; professional fees as a lecturer from MSD, Pfizer, BMS, Abbvie and Roche; and consulting fees from MSD, Pfizer, BMS, Abbvie and Roche.

Iván Ferraz-Amaro has received funding to attend courses and medical congresses from Abbvie, MSD and Pfizer; and professional fees as a lecturer from Abbvie.

Juan Jesús Gómez-Reino has received funding to attend courses and medical congresses from Abbott, BMS, MSD, Pfizer, Roche and UCB; professional fees as a lecturer from Abbott, BMS, MSD, Pfizer, Roche and UCB; funding for educational programmes or courses from MSD, Pfizer, Roche and UCB; and consulting fees from Abbott, BMS, MSD, Pfizer, Roche and UCB.

Isidoro González has received funding to attend courses and medical congresses from MSD, Actelion, Abbvie and Pfizer; professional fees as a lecturer from UCB, Roche, Abbott and BMS; funding for research participation from Amgen and Tigenix; and consulting fees from Pfizer. He is also a stockholder in Zeltia.

Sara Marsal has received funding to attend courses and medical congresses from Pfizer and Roche; professional fees as a lecturer from BMS, Roche and UCB; funding for educational programmes or courses from Roche and UCB; funding for research participation from Roche; and consulting fees from Pfizer, Roche, UCB and BMS.

Emilio Martín-Mola has received professional fees as a lecturer from Pfizer, MSD, BMS and Abbott; and consulting fees from MSD, Pfizer, Celgene, Abbott and Roche.

Víctor Manuel Martínez-Taboada has received funding to attend courses and medical congresses from Abbott, Menarini and Pfizer; professional fees as a lecturer from Esteve, Abbott, Roche and UCB; funding for research participation from Roche, MSD, Amgen, GlaxoSmithKline, UCB and Pfizer; and consulting fees from UCB, Roche, Cellerix, Pfizer, Sobi, Servier and Hospira.

José Vicente Moreno Muelas has received funding to attend courses and medical congresses from Gebro, Pfizer and Abbvie.

Ana M. Ortiz has received funding to attend courses and medical congresses from Abbvie, Amgen, GSK, Lilly, Menarini, MSD, Pfizer, Roche and UCB; professional fees as a lecturer from Abbvie, Esteve, MSD, Pfizer, Roche and UCB; funding for research participation from Roche; and consulting fees from Abbvie.

Jesús Tornero has received funding to attend courses and medical congresses from Pfizer; professional fees as a lecturer from Gebro, Pfizer, FAES FARMA, UCB, Menarini and Grunenthal; and funding for research participation from Roche.

Acknowledgements

We would like to acknowledge the authors of the systematic reviews: Ana M. Ortiz, Miguel Ángel Abad, Claudia Alejandra Pereda, María Betina Nishishinya, Jesús Maese and Eugenio Chamizo. We would also like to thank Federico Díaz-González, Director of the SER Research Unit, for his contributions towards safeguarding the independent nature of this document, along with Mercedes Guerra, SER documentalist, and Daniel Seoane, SER methodologist.

References
[1]
D.L. Scott, F. Wolfe, T.W. Huizinga.
Rheumatoid arthritis.
Lancet, 376 (2010), pp. 1094-1108
[2]
J. Tornero Molina, R. Sanmarti Sala, V. Rodriguez Valverde, E. Martin Mola, J.L. Marenco de la Fuente, I. Gonzalez Alvaro, et al.
Update of the Consensus Statement of the Spanish Society of Rheumatology on the management of biologic therapies in rheumatoid arthritis.
Reumatol Clin, 6 (2010), pp. 23-36
[3]
J.S. Smolen, R. Landewe, F.C. Breedveld, M. Buch, G. Burmester, M. Dougados, et al.
EULAR recommendations for the management of rheumatoid arthritis with synthetic and biological disease-modifying antirheumatic drugs: 2013 update.
Ann Rheum Dis, 73 (2014), pp. 492-509
[4]
C. Gaujoux-Viala, J. Nam, S. Ramiro, R. Landewe, M.H. Buch, J.S. Smolen, et al.
Efficacy of conventional synthetic disease-modifying antirheumatic drugs, glucocorticoids and tofacitinib: a systematic literature review informing the 2013 update of the EULAR recommendations for management of rheumatoid arthritis.
Ann Rheum Dis, 73 (2014), pp. 510-515
[5]
J.L. Nam, S. Ramiro, C. Gaujoux-Viala, K. Takase, M. Leon-Garcia, P. Emery, 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, 73 (2014), pp. 516-528
[6]
S. Ramiro, C. Gaujoux-Viala, J.L. Nam, J.S. Smolen, M. Buch, L. Gossec, et al.
Safety of synthetic and biological DMARDs: a systematic literature review informing the 2013 update of the EULAR recommendations for management of rheumatoid arthritis.
Ann Rheum Dis, 73 (2014), pp. 529-535
[7]
Centre for Evidence Based Medicine.
Levels of evidence (March 2009).
CEBM, (2014),
[8]
J.A.B. Van Nies, A. Krabben, J.W. Schoones, T.W.J. Huizinga, M. Kloppenburg, A.H.M. van der Helm-van Mil.
What is the evidence for the presence of a therapeutic window of opportunity in rheumatoid arthritis? A systematic literature review.
Ann Rheum Dis, 73 (2014), pp. 861-870
[9]
J.R. O’Dell, J.R. Curtis, T.R. Mikuls, S.S. Cofield, S.L. Bridges Jr., V.K. Ranganath, et al.
Validation of the methotrexate-first strategy in patients with early, poor-prognosis rheumatoid arthritis: results from a two-year randomized, double-blind trial.
Arthritis Rheum, 65 (2013), pp. 1985-1994
[10]
D. Aletaha, T. Neogi, A.J. Silman, J. Funovits, D.T. Felson, C.O. Bingham 3rd, et al.
Rheumatoid arthritis classification criteria: an American College of Rheumatology/European League Against Rheumatism collaborative initiative.
Ann Rheum Dis, 69 (2010), pp. 1580-1588
[11]
P. Emery, P. Durez, M. Dougados, C.W. Legerton, J.C. Becker, G. Vratsanos, et al.
Impact of T-cell costimulation modulation in patients with undifferentiated inflammatory arthritis or very early rheumatoid arthritis: a clinical and imaging study of abatacept (the ADJUST trial).
Ann Rheum Dis, 69 (2010), pp. 510-516
[12]
B. Saleem, S. Mackie, M. Quinn, S. Nizam, E. Hensor, S. Jarrett, et al.
Does the use of tumour necrosis factor antagonist therapy in poor prognosis, undifferentiated arthritis prevent progression to rheumatoid arthritis?.
Ann Rheum Dis, 67 (2008), pp. 1178-1180
[13]
S.M. Verstappen, M.J. McCoy, C. Roberts, N.E. Dale, A.B. Hassell, D.P. Symmons.
Beneficial effects of a 3-week course of intramuscular glucocorticoid injections in patients with very early inflammatory polyarthritis: results of the STIVEA trial.
Ann Rheum Dis, 69 (2010), pp. 503-509
[14]
M.H. Ma, I.C. Scott, G.H. Kingsley, D.L. Scott.
Remission in early rheumatoid arthritis.
J Rheumatol, 37 (2010), pp. 1444-1453
[15]
P. Emery, F.C. Breedveld, S. Hall, P. Durez, D.J. Chang, D. Robertson, et al.
Comparison of methotrexate monotherapy with a combination of methotrexate and etanercept in active, early, moderate to severe rheumatoid arthritis (COMET): a randomised, double-blind, parallel treatment trial.
[16]
J.S. Smolen, P. Emery, R. Fleischmann, R.F. van Vollenhoven, K. Pavelka, P. Durez, et al.
Adjustment of therapy in rheumatoid arthritis on the basis of achievement of stable low disease activity with adalimumab plus methotrexate or methotrexate alone: the randomised controlled OPTIMA trial.
[17]
C. Grigor, H. Capell, A. Stirling, A.D. McMahon, P. Lock, R. Vallance, et al.
Effect of a treatment strategy of tight control for rheumatoid arthritis (the TICORA study): a single-blind randomised controlled trial.
[18]
L.G. Schipper, M. Vermeer, H.H. Kuper, M.O. Hoekstra, C.J. Haagsma, A.A. Den Broeder, et al.
A tight control treatment strategy aiming for remission in early rheumatoid arthritis is more effective than usual care treatment in daily clinical practice: a study of two cohorts in the Dutch Rheumatoid Arthritis Monitoring registry.
Ann Rheum Dis, 71 (2012), pp. 845-850
[19]
K. Jayakumar, S. Norton, J. Dixey, D. James, A. Gough, P. Williams, et al.
Sustained clinical remission in rheumatoid arthritis: prevalence and prognostic factors in an inception cohort of patients treated with conventional DMARDS.
Rheumatology (Oxford), 51 (2012), pp. 169-175
[20]
F.H. Prince, V.P. Bykerk, N.A. Shadick, B. Lu, J. Cui, M. Frits, et al.
Sustained rheumatoid arthritis remission is uncommon in clinical practice.
Arthritis Res Ther, 14 (2012), pp. R68
[21]
D. Aletaha, M.M. Ward, K.P. Machold, V.P. Nell, T. Stamm, J.S. Smolen.
Remission and active disease in rheumatoid arthritis: defining criteria for disease activity states.
Arthritis Rheum, 52 (2005), pp. 2625-2636
[22]
L.H. Van Tuyl, D.T. Felson, G. Wells, J. Smolen, B. Zhang, M. Boers.
Evidence for predictive validity of remission on long-term outcome in rheumatoid arthritis: a systematic review.
Arthritis Care Res (Hoboken), 62 (2010), pp. 108-117
[23]
J.S. Smolen, C. Han, D.M. van der Heijde, P. Emery, J.M. Bathon, E. Keystone, et al.
Radiographic changes in rheumatoid arthritis patients attaining different disease activity states with methotrexate monotherapy and infliximab plus methotrexate: the impacts of remission and tumour necrosis factor blockade.
Ann Rheum Dis, 68 (2009), pp. 823-827
[24]
D.T. Felson, J.S. Smolen, G. Wells, B. Zhang, L.H. van Tuyl, J. Funovits, et al.
American College of Rheumatology/European League Against Rheumatism provisional definition of remission in rheumatoid arthritis for clinical trials.
Arthritis Rheum, 63 (2011), pp. 573-586
[25]
D. Aletaha, J.S. Smolen.
Joint damage in rheumatoid arthritis progresses in remission according to the Disease Activity Score in 28 joints and is driven by residual swollen joints.
Arthritis Rheum, 63 (2011), pp. 3702-3711
[26]
B. Zhang, B. Combe, N. Rincheval, D.T. Felson.
Validation of ACR/EULAR definition of remission in rheumatoid arthritis from RA practice: the ESPOIR cohort.
Arthritis Res Ther, 14 (2012), pp. R156
[27]
C. Iking-Konert, M. Aringer, J. Wollenhaupt, T. Mosch, S. Tuerk, E. Feist, et al.
Performance of the new 2011 ACR/EULAR remission criteria with tocilizumab using the phase IIIb study TAMARA as an example and their comparison with traditional remission criteria.
Ann Rheum Dis, 70 (2011), pp. 1986-1990
[28]
I. Navarro-Millan, L. Chen, J.D. Greenberg, D.A. Pappas, J.R. Curtis.
Predictors and persistence of new-onset clinical remission in rheumatoid arthritis patients.
Semin Arthritis Rheum, 43 (2013), pp. 137-143
[29]
K. Thiele, D. Huscher, S. Bischoff, S. Spathling-Mestekemper, M. Backhaus, M. Aringer, et al.
Performance of the 2011 ACR/EULAR preliminary remission criteria compared with DAS28 remission in unselected patients with rheumatoid arthritis.
Ann Rheum Dis, 72 (2013), pp. 1194-1199
[30]
K.R. Masri, T.S. Shaver, S.H. Shahouri, S. Wang, J.D. Anderson, R.E. Busch, et al.
Validity and reliability problems with patient global as a component of the ACR/EULAR remission criteria as used in clinical practice.
J Rheumatol, 39 (2012), pp. 1139-1145
[31]
N.B. Klarenbeek, R. Koevoets, D.M. van der Heijde, A.H. Gerards, S. ten Wolde, P.J. Kerstens, et al.
Association with joint damage and physical functioning of nine composite indices and the 2011 ACR/EULAR remission criteria in rheumatoid arthritis.
Ann Rheum Dis, 70 (2011), pp. 1815-1821
[32]
A.K. Brown, M.A. Quinn, Z. Karim, P.G. Conaghan, C.G. Peterfy, E. Hensor, et al.
Presence of significant synovitis in rheumatoid arthritis patients with disease-modifying antirheumatic drug-induced clinical remission: evidence from an imaging study may explain structural progression.
Arthritis Rheum, 54 (2006), pp. 3761-3773
[33]
A.K. Brown, P.G. Conaghan, Z. Karim, M.A. Quinn, K. Ikeda, C.G. Peterfy, et al.
An explanation for the apparent dissociation between clinical remission and continued structural deterioration in rheumatoid arthritis.
Arthritis Rheum, 58 (2008), pp. 2958-2967
[34]
H. Nguyen, A. Ruyssen-Witrand, F. Gandjbakhch, A. Constantin, V. Foltz, A. Cantagrel.
Prevalence of ultrasound-detected residual synovitis and risk of relapse and structural progression in rheumatoid arthritis patients in clinical remission: a systematic review and meta-analysis.
Rheumatology (Oxford), 53 (2014), pp. 2110-2118
[35]
C.A. Scire, C. Montecucco, V. Codullo, O. Epis, M. Todoerti, R. Caporali.
Ultrasonographic evaluation of joint involvement in early rheumatoid arthritis in clinical remission: Power Doppler signal predicts short-term relapse.
Rheumatology (Oxford), 48 (2009), pp. 1092-1097
[36]
A. Balsa, E. de Miguel, C. Castillo, D. Peiteado, E. Martin-Mola.
Superiority of SDAI over DAS-28 in assessment of remission in rheumatoid arthritis patients using power Doppler ultrasonography as a gold standard.
Rheumatology (Oxford), 49 (2010), pp. 683-690
[37]
G. Sakellariou, C.A. Scire, S.M. Verstappen, C. Montecucco, R. Caporali.
In patients with early rheumatoid arthritis, the new ACR/EULAR definition of remission identifies patients with persistent absence of functional disability and suppression of ultrasonographic synovitis.
Ann Rheum Dis, 72 (2013), pp. 245-249
[38]
B. Combe, B. Dasgupta, I. Louw, S. Pal, J. Wollenhaupt, C.A. Zerbini, et al.
Efficacy and safety of golimumab as add-on therapy to disease-modifying antirheumatic drugs: results of the GO-MORE study.
Ann Rheum Dis, 73 (2014), pp. 1477-1486
[39]
Y.P. Goekoop-Ruiterman, J.K. de Vries-Bouwstra, C.F. Allaart, D. van Zeben, P.J. Kerstens, J.M. Hazes, et al.
Clinical and radiographic outcomes of four different treatment strategies in patients with early rheumatoid arthritis (the BeSt study): a randomized, controlled trial.
Arthritis Rheum, 52 (2005), pp. 3381-3390
[40]
L. Heimans, K.V. Boer, K.M. Koudijs, K. Visser, Y.P. Goekoop-Ruiterman, J.B. Harbers, et al.
Health-related quality of life and functional ability in patients with early arthritis during remission steered treatment: results of the IMPROVED study.
Arthritis Res Ther, 15 (2013), pp. R173
[41]
L. Heimans, K.V. Wevers-de Boer, K. Visser, R.J. Goekoop, M. van Oosterhout, J.B. Harbers, et al.
A two-step treatment strategy trial in patients with early arthritis aimed at achieving remission: the IMPROVED study.
Ann Rheum Dis, 73 (2014), pp. 1356-1361
[42]
K. Horslev-Petersen, M.L. Hetland, P. Junker, J. Podenphant, T. Ellingsen, P. Ahlquist, et al.
Adalimumab added to a treat-to-target strategy with methotrexate and intra-articular triamcinolone in early rheumatoid arthritis increased remission rates, function and quality of life. The OPERA Study: an investigator-initiated, randomised, double-blind, parallel-group, placebo-controlled trial.
Ann Rheum Dis, 73 (2014), pp. 654-661
[43]
M. Leirisalo-Repo, H. Kautiainen, L. Laasonen, M. Korpela, M.J. Kauppi, O. Kaipiainen-Seppanen, et al.
Infliximab for 6 months added on combination therapy in early rheumatoid arthritis: 2-year results from an investigator-initiated, randomised, double-blind, placebo-controlled study (the NEO-RACo Study).
Ann Rheum Dis, 72 (2013), pp. 851-857
[44]
J.L. Nam, E. Villeneuve, E.M. Hensor, P.G. Conaghan, H.I. Keen, M.H. Buch, et al.
Remission induction comparing infliximab and high-dose intravenous steroid, followed by treat-to-target: a double-blind, randomised, controlled trial in new-onset, treatment-naive, rheumatoid arthritis (the IDEA study).
Ann Rheum Dis, 73 (2014), pp. 75-85
[45]
J.L. Nam, E. Villeneuve, E.M. Hensor, R.J. Wakefield, P.G. Conaghan, M.J. Green, et al.
A randomised controlled trial of etanercept and methotrexate to induce remission in early inflammatory arthritis: the EMPIRE trial.
Ann Rheum Dis, 73 (2014), pp. 1027-1036
[46]
J.S. Smolen, P. Emery, G.F. Ferraccioli, W. Samborski, F. Berenbaum, O.R. Davies, et al.
Certolizumab pegol in rheumatoid arthritis patients with low to moderate activity: the CERTAIN double-blind, randomised, placebo-controlled trial.
Ann Rheum Dis, 74 (2015), pp. 843-850
[47]
S.M. Van der Kooij, J.K. de Vries-Bouwstra, Y.P. Goekoop-Ruiterman, J.A. Ewals, K.H. Han, J.M. Hazes, et al.
Patient-reported outcomes in a randomized trial comparing four different treatment strategies in recent-onset rheumatoid arthritis.
Arthritis Rheum, 61 (2009), pp. 4-12
[48]
S.M. Van der Kooij, Y.P. Goekoop-Ruiterman, J.K. de Vries-Bouwstra, M. Guler-Yuksel, A.H. Zwinderman, P.J. Kerstens, et al.
Drug-free remission, functioning and radiographic damage after 4 years of response-driven treatment in patients with recent-onset rheumatoid arthritis.
Ann Rheum Dis, 68 (2009), pp. 914-921
[49]
I.C. Van Eijk, M.M. Nielen, I. van der Horst-Bruinsma, G.J. Tijhuis, M. Boers, B.A. Dijkmans, et al.
Aggressive therapy in patients with early arthritis results in similar outcome compared with conventional care: the STREAM randomized trial.
Rheumatology (Oxford), 51 (2012), pp. 686-694
[50]
J.E. Pope, B. Haraoui, J.C. Thorne, A. Vieira, M. Poulin-Costello, E.C. Keystone.
The Canadian Methotrexate and Etanercept Outcome Study: a randomised trial of discontinuing versus continuing methotrexate after 6 months of etanercept and methotrexate therapy in rheumatoid arthritis.
Ann Rheum Dis, 73 (2014), pp. 2144-2151
[51]
D. Aletaha, J. Funovits, E.C. Keystone, J.S. Smolen.
Disease activity early in the course of treatment predicts response to therapy after one year in rheumatoid arthritis patients.
Arthritis Rheum, 56 (2007), pp. 3226-3235
[52]
F.C. Breedveld, M.H. Weisman, A.F. Kavanaugh, S.B. Cohen, K. Pavelka, R. van Vollenhoven, 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, 54 (2006), pp. 26-37
[53]
A. Kavanaugh, R.M. Fleischmann, P. Emery, H. Kupper, L. Redden, B. Guerette, et al.
Clinical, functional and radiographic consequences of achieving stable low disease activity and remission with adalimumab plus methotrexate or methotrexate alone in early rheumatoid arthritis: 26-week results from the randomised, controlled OPTIMA study.
Ann Rheum Dis, 72 (2013), pp. 64-71
[54]
E.W. St Clair, D.M. van der Heijde, J.S. Smolen, R.N. Maini, J.M. Bathon, P. Emery, et al.
Combination of infliximab and methotrexate therapy for early rheumatoid arthritis: a randomized, controlled trial.
Arthritis Rheum, 50 (2004), pp. 3432-3443
[55]
D. Van der Heijde, L. Klareskog, V. Rodriguez-Valverde, C. Codreanu, H. Bolosiu, J. Melo-Gomes, et al.
Comparison of etanercept and methotrexate, alone and combined, in the treatment of rheumatoid arthritis: two-year clinical and radiographic results from the TEMPO study, a double-blind, randomized trial.
Arthritis Rheum, 54 (2006), pp. 1063-1074
[56]
J.M. Bathon, R.W. Martin, R.M. Fleischmann, J.R. Tesser, M.H. Schiff, E.C. Keystone, et al.
A comparison of etanercept and methotrexate in patients with early rheumatoid arthritis.
N Engl J Med, 343 (2000), pp. 1586-1593
[57]
Sociedad Española de Reumatología. Proyecto EMAR II.
Variabilidad en el manejo de la artritis reumatoide y las espondiloartritis en España. Informe de resultados.
SER, (2011),
[58]
S.L. Morgan, J.E. Baggott, W.H. Vaughn, J.S. Austin, T.A. Veitch, J.Y. Lee, et al.
Supplementation with folic acid during methotrexate therapy for rheumatoid arthritis. A double-blind, placebo-controlled trial.
Ann Intern Med, 121 (1994), pp. 833-841
[59]
J.B. Shiroky, C. Neville, J.M. Esdaile, D. Choquette, M. Zummer, M. Hazeltine, et al.
Low-dose methotrexate with leucovorin (folinic acid) in the management of rheumatoid arthritis. Results of a multicenter randomized, double-blind, placebo-controlled trial.
Arthritis Rheum, 36 (1993), pp. 795-803
[60]
M.H. Schiff, J.S. Jaffe, B. Freundlich.
Head-to-head, randomised, crossover study of oral versus subcutaneous methotrexate in patients with rheumatoid arthritis: drug-exposure limitations of oral methotrexate at doses >/=15mg may be overcome with subcutaneous administration.
Ann Rheum Dis, 73 (2014), pp. 1549-1551
[61]
J. Braun, P. Kastner, P. Flaxenberg, J. Wahrisch, P. Hanke, W. Demary, 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, 58 (2008), pp. 73-81
[62]
V.C.S.T. Chalam, T. Price, S. Baskar, D. Mulherin, C. Molloy, F. Keay, et al.
A retrospective study of the effects of switching from oral to subcutaneous methotrexate on disease activity in patients with rheumatoid arthritis.
Rheumatology, 52 (2013), pp. i84
[63]
G. Hazlewood, C. Thorne, J. Pope, G. Boire, B. Haraoui, C. Hitchon, et al.
The comparative effectiveness of oral methotrexate versus subcutaneous methotrexate for the initial treatment of early rheumatoid arthritis.
Ann Rheum Dis, 72 (2013), pp. A246
[64]
J. Wegrzyn, P. Adeleine, P. Miossec.
Better efficacy of methotrexate given by intramuscular injection than orally in patients with rheumatoid arthritis.
Ann Rheum Dis, 63 (2004), pp. 1232-1234
[65]
G. Jones, A. Sebba, J. Gu, M.B. Lowenstein, A. Calvo, J.J. Gomez-Reino, et al.
Comparison of tocilizumab monotherapy versus methotrexate monotherapy in patients with moderate to severe rheumatoid arthritis: the AMBITION study.
Ann Rheum Dis, 69 (2010), pp. 88-96
[66]
E.B. Lee, R. Fleischmann, S. Hall, B. Wilkinson, J.D. Bradley, D. Gruben, et al.
Tofacitinib versus methotrexate in rheumatoid arthritis.
N Engl J Med, 370 (2014), pp. 2377-2386
[67]
J.T. Sharp, V. Strand, H. Leung, F. Hurley, I. Loew-Friedrich, Leflunomide Rheumatoid Arthritis Investigators Group.
Treatment with leflunomide slows radiographic progression of rheumatoid arthritis: results from three randomized controlled trials of leflunomide in patients with active rheumatoid arthritis.
[68]
V. Strand, S. Cohen, M. Schiff, A. Weaver, R. Fleischmann, G. Cannon, Leflunomide Rheumatoid Arthritis Investigators Group, et al.
Treatment of active rheumatoid arthritis with leflunomide compared with placebo and methotrexate.
Arch Intern Med, 159 (1999), pp. 2542-2550
[69]
G.R. Burmester, X. Mariette, C. Montecucco, I. Monteagudo-Saez, M. Malaise, A.G. Tzioufas, et al.
Adalimumab alone and in combination with disease-modifying antirheumatic drugs for the treatment of rheumatoid arthritis in clinical practice: the Research in Active Rheumatoid Arthritis (ReAct) trial.
Ann Rheum Dis, 66 (2007), pp. 732-739
[70]
A. Strangfeld, F. Hierse, J. Kekow, U. von Hinueber, H.P. Tony, R. Dockhorn, et al.
Comparative effectiveness of tumour necrosis factor alpha inhibitors in combination with either methotrexate or leflunomide.
Ann Rheum Dis, 68 (2009), pp. 1856-1862
[71]
H.A. Capell.
Clinical efficacy of sulphasalazine—a review.
Br J Rheumatol, 34 (1995), pp. 35-39
[72]
E.C. Keystone, M.M. Wang, M. Layton, S. Hollis, I.B. McInnes.
Clinical evaluation of the efficacy of the P2X7 purinergic receptor antagonist AZD9056 on the signs and symptoms of rheumatoid arthritis in patients with active disease despite treatment with methotrexate or sulphasalazine.
Ann Rheum Dis, 71 (2012), pp. 1630-1635
[73]
T. Pullar, J.A. Hunter, H.A. Capell.
Sulphasalazine in the treatment of rheumatoid arthritis: relationship of dose and serum levels to efficacy.
Br J Rheumatol, 24 (1985), pp. 269-276
[74]
D.M. Van der Heijde, P.L. van Riel, I.H. Nuver-Zwart, F.W. Gribnau, L.B. vad de Putte.
Effects of hydroxychloroquine and sulphasalazine on progression of joint damage in rheumatoid arthritis.
Lancet, 1 (1989), pp. 1036-1038
[75]
P.L. Van Riel, A.M. van Gestel, L.B. van de Putte.
Long-term usage and side-effect profile of sulphasalazine in rheumatoid arthritis.
Br J Rheumatol, 34 (1995), pp. 40-42
[76]
J.S. Smolen, J.R. Kalden, D.L. Scott, B. Rozman, T.K. Kvien, A. Larsen, European Leflunomide Study Group, et al.
Efficacy and safety of leflunomide compared with placebo and sulphasalazine in active rheumatoid arthritis: a double-blind, randomised, multicentre trial.
Lancet, 353 (1999), pp. 259-266
[77]
M. Boers, A.C. Verhoeven, H.M. Markusse, M.A. van de Laar, R. Westhovens, J.C. van Denderen, et al.
Randomised comparison of combined step-down prednisolone, methotrexate and sulphasalazine with sulphasalazine alone in early rheumatoid arthritis.
[78]
D.T. Felson, J.J. Anderson, R.F. Meenan.
Use of short-term efficacy/toxicity tradeoffs to select second-line drugs in rheumatoid arthritis. A meta-analysis of published clinical trials.
Arthritis Rheum, 35 (1992), pp. 1117-1125
[79]
V.C. Neumann, K.A. Grindulis, S. Hubball, B. McConkey, V. Wright.
Comparison between penicillamine and sulphasalazine in rheumatoid arthritis: Leeds-Birmingham trial.
Br Med J (Clin Res Ed), 287 (1983), pp. 1099-1102
[80]
M. Calguneri, S. Pay, Z. Caliskaner, S. Apras, S. Kiraz, I. Ertenli, et al.
Combination therapy versus monotherapy for the treatment of patients with rheumatoid arthritis.
Clin Exp Rheumatol, 17 (1999), pp. 699-704
[81]
T. Mottonen, P. Hannonen, M. Leirisalo-Repo, M. Nissila, H. Kautiainen, M. Korpela, FIN-RACo trial group, et al.
Comparison of combination therapy with single-drug therapy in early rheumatoid arthritis: a randomised trial.
Lancet, 353 (1999), pp. 1568-1573
[82]
J.R. O’Dell, C.E. Haire, N. Erikson, W. Drymalski, W. Palmer, P.J. Eckhoff, et al.
Treatment of rheumatoid arthritis with methotrexate alone, sulfasalazine and hydroxychloroquine, or a combination of all three medications.
N Engl J Med, 334 (1996), pp. 1287-1291
[83]
I. Gonzalez-Alvaro, M.A. Descalzo, L. Carmona.
Trends towards an improved disease state in rheumatoid arthritis over time: Influence of new therapies and changes in management approach: analysis of the EMECAR cohort.
Arthritis Res Ther, 10 (2008), pp. R138
[84]
J.S. Smolen, R. Landewe, F.C. Breedveld, M. Dougados, P. Emery, C. Gaujoux-Viala, et al.
EULAR recommendations for the management of rheumatoid arthritis with synthetic and biological disease-modifying antirheumatic drugs.
Ann Rheum Dis, 69 (2010), pp. 964-975
[85]
S.J. Katz, A.S. Russell.
Re-evaluation of antimalarials in treating rheumatic diseases: re-appreciation and insights into new mechanisms of action.
Curr Opin Rheumatol, 23 (2011), pp. 278-281
[86]
M.E. Suarez-Almazor, E. Belseck, B. Shea, J. Homik, G. Wells, P. Tugwell.
Antimalarials for treating rheumatoid arthritis.
Cochrane Database Syst Rev, (2000), pp. Cd000959
[87]
R. Partlett, E. Roussou.
The treatment of rheumatoid arthritis during pregnancy.
Rheumatol Int, 31 (2011), pp. 445-449
[88]
P.H. De Jong, J.M. Hazes, P.J. Barendregt, M. Huisman, D. van Zeben, P.A. van der Lubbe, et al.
Induction therapy with a combination of DMARDs is better than methotrexate monotherapy: first results of the tREACH trial.
Ann Rheum Dis, 72 (2013), pp. 72-78
[89]
L.W. Moreland, J.R. O’Dell, H.E. Paulus, J.R. Curtis, J.M. Bathon, E.W. St Clair, et al.
A randomized comparative effectiveness study of oral triple therapy versus etanercept plus methotrexate in early aggressive rheumatoid arthritis: the treatment of Early Aggressive Rheumatoid Arthritis Trial.
Arthritis Rheum, 64 (2012), pp. 2824-2835
[90]
J.R. O’Dell, T.R. Mikuls, T.H. Taylor, V. Ahluwalia, M. Brophy, S.R. Warren, et al.
Therapies for active rheumatoid arthritis after methotrexate failure.
N Engl J Med, 369 (2013), pp. 307-318
[91]
R.F. Van Vollenhoven, P. Geborek, K. Forslind, K. Albertsson, S. Ernestam, I.F. Petersson, et al.
Conventional combination treatment versus biological treatment in methotrexate-refractory early rheumatoid arthritis: 2 year follow-up of the randomised, non-blinded, parallel-group Swefot trial.
Lancet, 379 (2012), pp. 1712-1720
[92]
N.B. Klarenbeek, M. Guler-Yuksel, S.M. van der Kooij, K.H. Han, H.K. Ronday, P.J. Kerstens, et al.
The impact of four dynamic, goal-steered treatment strategies on the 5-year outcomes of rheumatoid arthritis patients in the BeSt study.
Ann Rheum Dis, 70 (2011), pp. 1039-1046
[93]
D. Den Uyl, M. ter Wee, M. Boers, P. Kerstens, A. Voskuyl, M. Nurmohamed, et al.
A non-inferiority trial of an attenuated combination strategy (‘COBRA-light’) compared to the original COBRA strategy: clinical results after 26 weeks.
Ann Rheum Dis, 73 (2014), pp. 1071-1078
[94]
D.H. Solomon, J.M. Kremer, M. Fisher, J.R. Curtis, V. Furer, L.R. Harrold, et al.
Comparative cancer risk associated with methotrexate, other non-biologic and biologic disease-modifying anti-rheumatic drugs.
Semin Arthritis Rheum, 43 (2014), pp. 489-497
[95]
R. Conway, C. Low, R.J. Coughlan, M.J. O’Donnell, J.J. Carey.
Methotrexate and lung disease in rheumatoid arthritis: a meta-analysis of randomized controlled trials.
Arthritis Rheumatol, 66 (2014), pp. 803-812
[96]
S.L. Gorter, J.W. Bijlsma, M. Cutolo, J. Gomez-Reino, M. Kouloumas, J.S. Smolen, et al.
Current evidence for the management of rheumatoid arthritis with glucocorticoids: a systematic literature review informing the EULAR recommendations for the management of rheumatoid arthritis.
Ann Rheum Dis, 69 (2010), pp. 1010-1014
[97]
E.H. Choy, G.H. Kingsley, B. Khoshaba, N. Pipitone, D.L. Scott.
A two year randomised controlled trial of intramuscular depot steroids in patients with established rheumatoid arthritis who have shown an incomplete response to disease modifying antirheumatic drugs.
Ann Rheum Dis, 64 (2005), pp. 1288-1293
[98]
B. Svensson, A. Boonen, K. Albertsson, D. van der Heijde, C. Keller, I. Hafstrom.
Low-dose prednisolone in addition to the initial disease-modifying antirheumatic drug in patients with early active rheumatoid arthritis reduces joint destruction and increases the remission rate: a two-year randomized trial.
Arthritis Rheum, 52 (2005), pp. 3360-3370
[99]
S. Wassenberg, R. Rau, P. Steinfeld, H. Zeidler.
Very low-dose prednisolone in early rheumatoid arthritis retards radiographic progression over two years: a multicenter, double-blind, placebo-controlled trial.
Arthritis Rheum, 52 (2005), pp. 3371-3380
[100]
H.A. Capell, R. Madhok, J.A. Hunter, D. Porter, E. Morrison, J. Larkin, et al.
Lack of radiological and clinical benefit over two years of low dose prednisolone for rheumatoid arthritis: results of a randomised controlled trial.
Ann Rheum Dis, 63 (2004), pp. 797-803
[101]
M.J. Van der Veen, J.W. Bijlsma.
The effect of methylprednisolone pulse therapy on methotrexate treatment of rheumatoid arthritis.
Clin Rheumatol, 12 (1993), pp. 500-505
[102]
A.M. Van Gestel, R.F. Laan, C.J. Haagsma, L.B. van de Putte, P.L. van Riel.
Oral steroids as bridge therapy in rheumatoid arthritis patients starting with parenteral gold. A randomized double-blind placebo-controlled trial.
Br J Rheumatol, 34 (1995), pp. 347-351
[103]
M.F. Bakker, J.W. Jacobs, P.M. Welsing, S.M. Verstappen, J. Tekstra, E. Ton, et al.
Low-dose prednisone inclusion in a methotrexate-based, tight control strategy for early rheumatoid arthritis: a randomized trial.
Ann Intern Med, 156 (2012), pp. 329-339
[104]
K.P. Machold, R. Landewe, J.S. Smolen, T.A. Stamm, D.M. van der Heijde, K.N. Verpoort, et al.
The Stop Arthritis Very Early (SAVE) trial, an international multicentre, randomised, double-blind, placebo-controlled trial on glucocorticoids in very early arthritis.
Ann Rheum Dis, 69 (2010), pp. 495-502
[105]
P.H. De Jong, J.M. Hazes, H.K. Han, M. Huisman, D. van Zeben, P.A. van der Lubbe, et al.
Randomised comparison of initial triple DMARD therapy with methotrexate monotherapy in combination with low-dose glucocorticoid bridging therapy: 1-year data of the tREACH trial.
Ann Rheum Dis, 73 (2014), pp. 1331-1339
[106]
J.W. Bijlsma, J.N. Hoes, A.A. van Everdingen, S.M. Verstappen, J.W. Jacobs.
Are glucocorticoids DMARDs?.
Ann N Y Acad Sci, 1069 (2006), pp. 268-274
[107]
J.R. Kirwan, The Arthritis and Rheumatism Council Low-Dose Glucocorticoid Study Group.
The effect of glucocorticoids on joint destruction in rheumatoid arthritis.
N Engl J Med, 333 (1995), pp. 142-146
[108]
J.A. Da Silva, J.W. Jacobs, J.R. Kirwan, M. Boers, K.G. Saag, L.B. Ines, et al.
Safety of low dose glucocorticoid treatment in rheumatoid arthritis: published evidence and prospective trial data.
Ann Rheum Dis, 65 (2006), pp. 285-293
[109]
N. Vastesaeger, S. Xu, D. Aletaha, E.W. St Clair, J.S. Smolen.
A pilot risk model for the prediction of rapid radiographic progression in rheumatoid arthritis.
Rheumatology (Oxford), 48 (2009), pp. 1114-1121
[110]
K. Visser, Y.P.M. Goekoop-Ruiterman, J.K. de Vries-Bouwstra, H.K. Ronday, P.E.H. Seys, P.J.S.M. Kerstens, et al.
A matrix risk model for the prediction of rapid radiographic progression in patients with rheumatoid arthritis receiving different dynamic treatment strategies: post hoc analyses from the BeSt study.
Ann Rheum Dis, 69 (2010), pp. 1333-1337
[111]
E. Choy, F. McKenna, J. Vencovsky, R. Valente, N. Goel, B. van Lunen, et al.
Certolizumab pegol plus MTX administered every 4 weeks is effective in patients with RA who are partial responders to MTX.
Rheumatology, 51 (2012), pp. 1226-1234
[112]
H. Kameda, Y. Ueki, K. Saito, S. Nagaoka, T. Hidaka, T. Atsumi, et al.
Etanercept (ETN) with methotrexate (MTX) is better than ETN monotherapy in patients with active rheumatoid arthritis despite MTX therapy: a randomized trial.
Mod Rheumatol, 20 (2010), pp. 531-538
[113]
Y.M. Kang, W. Park, Y.E. Park, J.Y. Choe, S.-C. Bae, C.S. Cho, et al.
Efficacy and safety of certolizumab pegol (CZP) with concomitant methotrexate (MTX) in Korean rheumatoid arthritis (RA) patients (PTS) with an inadequate response to MTX.
Ann Rheum Dis, 71 (2013), pp. 666
[114]
R. Knevel, M. Schoels, T.W. Huizinga, D. Aletaha, G.R. Burmester, B. Combe, et al.
Current evidence for a strategic approach to the management of rheumatoid arthritis with disease-modifying antirheumatic drugs: a systematic literature review informing the EULAR recommendations for the management of rheumatoid arthritis.
Ann Rheum Dis, 69 (2010), pp. 987-994
[115]
J.L. Nam, K.L. Winthrop, R.F. van Vollenhoven, K. Pavelka, G. Valesini, E.M. Hensor, et al.
Current evidence for the management of rheumatoid arthritis with biological disease-modifying antirheumatic drugs: a systematic literature review informing the EULAR recommendations for the management of RA.
Ann Rheum Dis, 69 (2010), pp. 976-986
[116]
M.E. Weinblatt, M. Schiff, R. Valente, D. van der Heijde, G. Citera, C. Zhao, et al.
Head-to-head comparison of subcutaneous abatacept versus adalimumab for rheumatoid arthritis: findings of a phase IIIb, multinational, prospective, randomized study.
Arthritis Rheum, 65 (2013), pp. 28-38
[117]
C. Gabay, P. Emery, R. van Vollenhoven, A. Dikranian, R. Alten, K. Pavelka, et al.
Tocilizumab monotherapy versus adalimumab monotherapy for treatment of rheumatoid arthritis (ADACTA): a randomised, double-blind, controlled phase 4 trial.
Lancet, 381 (2013), pp. 1541-1550
[118]
L. Klareskog, D. van der Heijde, J.P. de Jager, A. Gough, J. Kalden, M. Malaise, 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.
[119]
P. Guyot, P. Taylor, R. Christensen, L. Pericleous, C. Poncet, M. Lebmeier, et al.
Abatacept with methotrexate versus other biologic agents in treatment of patients with active rheumatoid arthritis despite methotrexate: a network meta-analysis.
Arthritis Res Ther, 13 (2011), pp. R204
[120]
A. Rubbert-Roth, P.P. Tak, C. Zerbini, J.L. Tremblay, L. Carreno, G. Armstrong, et al.
Efficacy and safety of various repeat treatment dosing regimens of rituximab in patients with active rheumatoid arthritis: results of a phase III randomized study (MIRROR).
Rheumatology (Oxford), 49 (2010), pp. 1683-1693
[121]
P.P. Tak, W.F. Rigby, A. Rubbert-Roth, C.G. Peterfy, R.F. van Vollenhoven, W. Stohl, et al.
Inhibition of joint damage and improved clinical outcomes with rituximab plus methotrexate in early active rheumatoid arthritis: the IMAGE trial.
Ann Rheum Dis, 70 (2011), pp. 39-46
[122]
T. Tanaka, M. Narazaki, A. Ogata, T. Kishimoto.
A new era for the treatment of inflammatory autoimmune diseases by interleukin-6 blockade strategy.
Semin Immunol, 26 (2014), pp. 88-96
[123]
G.R. Burmester, W. Rigby, R.F. van Vollenhoven, J. Kay, A. Rubbert-Roth, A. Kelman, et al.
Tocilizumab in combination therapy and monotherapy versus methotrexate in methotrexate-naïve patients with early rheumatoid arthritis: clinical and radiographic outcomes from a randomized, placebo-controlled trial.
Arthritis Rheumatol, 65 (2013), pp. S1182-S1183
[124]
N. Nishimoto, J. Hashimoto, N. Miyasaka, K. Yamamoto, S. Kawai, T. Takeuchi, et al.
Study of active controlled monotherapy used for rheumatoid arthritis, an IL-6 inhibitor (SAMURAI): evidence of clinical and radiographic benefit from an X-ray reader-blinded randomised controlled trial of tocilizumab.
Ann Rheum Dis, 66 (2007), pp. 1162-1167
[125]
M. Dougados, K. Kissel, T. Sheeran, P.P. Tak, P.G. Conaghan, E.M. Mola, et al.
Adding tocilizumab or switching to tocilizumab monotherapy in methotrexate inadequate responders: 24-week symptomatic and structural results of a 2-year randomised controlled strategy trial in rheumatoid arthritis (ACT-RAY).
Ann Rheum Dis, 72 (2013), pp. 43-50
[126]
H. Kameda, K. Kanbe, E. Sato, Y. Ueki, K. Saito, S. Nagaoka.
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, 38 (2011), pp. 1585-1592
[127]
P.L.C.M. Van Riel, A.J. Taggart, J. Sany, M. Gaubitz, H.W. Nab, R. Pedersen, et al.
Efficacy and safety of combination etanercept and methotrexate versus etanercept alone in patients with rheumatoid arthritis with an inadequate response to methotrexate: the ADORE study.
Ann Rheum Dis, 65 (2006), pp. 1478-1483
[128]
A. Lethaby, M.A. Lopez-Olivo, L. Maxwell, A. Burls, P. Tugwell, G.A. Wells.
Etanercept for the treatment of rheumatoid arthritis.
Cochrane Database Syst Rev, 5 (2013), pp. CD004525
[129]
S.B. Cohen, P. Emery, M.W. Greenwald, M. Dougados, R.A. Furie, M.C. Genovese, et al.
Rituximab for rheumatoid arthritis refractory to anti-tumor necrosis factor therapy: results of a multicenter, randomized, double-blind, placebo-controlled, phase III trial evaluating primary efficacy and safety at twenty-four weeks.
Arthritis Rheum, 54 (2006), pp. 2793-2806
[130]
P. Emery, E. Keystone, H.P. Tony, A. Cantagrel, R. van Vollenhoven, A. Sanchez, et al.
IL-6 receptor inhibition with tocilizumab improves treatment outcomes in patients with rheumatoid arthritis refractory to anti-tumour necrosis factor biologicals: results from a 24-week multicentre randomised placebo-controlled trial.
Ann Rheum Dis, 67 (2008), pp. 1516-1523
[131]
M.C. Genovese, M. Schiff, M. Luggen, J.C. Becker, R. Aranda, J. Teng, et al.
Efficacy and safety of the selective co-stimulation modulator abatacept following 2 years of treatment in patients with rheumatoid arthritis and an inadequate response to anti-tumour necrosis factor therapy.
Ann Rheum Dis, 67 (2008), pp. 547-554
[132]
J.S. Smolen, J. Kay, M.K. Doyle, R. Landewe, E.L. Matteson, J. Wollenhaupt, et al.
Golimumab in patients with active rheumatoid arthritis after treatment with tumour necrosis factor alpha inhibitors (GO-AFTER study): a multicentre, randomised, double-blind, placebo-controlled, phase III trial.
[133]
M.E. Weinblatt, R. Fleischmann, T.W. Huizinga, P. Emery, J. Pope, E.M. Massarotti, et al.
Efficacy and safety of certolizumab pegol in a broad population of patients with active rheumatoid arthritis: Results from the REALISTIC phase IIIb study.
Rheumatology (Oxford), 51 (2012), pp. 2204-2214
[134]
H.L. Kim, M.Y. Lee, S.Y. Park, S.K. Park, J.H. Byun, S. Kwon, et al.
Comparative effectiveness of cycling of tumor necrosis factor-alpha (TNF-alpha) inhibitors versus switching to non-TNF biologics in rheumatoid arthritis patients with inadequate response to TNF-alpha inhibitor using a Bayesian approach.
Arch Pharm Res, 37 (2014), pp. 662-670
[135]
A. Finckh, A. Ciurea, L. Brulhart, D. Kyburz, B. Moller, S. Dehler, et al.
B cell depletion may be more effective than switching to an alternative anti-tumor necrosis factor agent in rheumatoid arthritis patients with inadequate response to anti-tumor necrosis factor agents.
Arthritis Rheum, 56 (2007), pp. 1417-1423
[136]
J.J. Gomez-Reino, J.R. Maneiro, J. Ruiz, R. Rosello, R. Sanmarti, A.B. Romero.
Comparative effectiveness of switching to alternative tumour necrosis factor (TNF) antagonists versus switching to rituximab in patients with rheumatoid arthritis who failed previous TNF antagonists: the MIRAR study.
Ann Rheum Dis, 71 (2012), pp. 1861-1864
[137]
S. Hirabara, N. Takahashi, N. Fukaya, H. Miyake, Y. Yabe, A. Kaneko, et al.
Clinical efficacy of abatacept, tocilizumab, and etanercept in Japanese rheumatoid arthritis patients with inadequate response to anti-TNF monoclonal antibodies.
Clin Rheumatol, 33 (2014), pp. 1247-1254
[138]
H.C. Leffers, M. Ostergaard, B. Glintborg, N.S. Krogh, H. Foged, U. Tarp, et al.
Efficacy of abatacept and tocilizumab in patients with rheumatoid arthritis treated in clinical practice: results from the nationwide Danish DANBIO registry.
Ann Rheum Dis, 70 (2011), pp. 1216-1222
[139]
M.M. Soliman, K.L. Hyrich, M. Lunt, K.D. Watson, D.P. Symmons, D.M. Ashcroft.
Rituximab or a second anti-tumor necrosis factor therapy for rheumatoid arthritis patients who have failed their first anti-tumor necrosis factor therapy? Comparative analysis from the British Society for Rheumatology Biologics Register.
Arthritis Care Res (Hoboken), 64 (2012), pp. 1108-1115
[140]
O. Brocq, E. Millasseau, C. Albert, C. Grisot, P. Flory, C.H. Roux, et al.
Effect of discontinuing TNFalpha antagonist therapy in patients with remission of rheumatoid arthritis.
Joint Bone Spine, 76 (2009), pp. 350-355
[141]
M. Harigai, T. Takeuchi, Y. Tanaka, T. Matsubara, H. Yamanaka, N. Miyasaka.
Discontinuation of adalimumab treatment in rheumatoid arthritis patients after achieving low disease activity.
Mod Rheumatol, 22 (2012), pp. 814-822
[142]
M. Nawata, K. Saito, S. Nakayamada, Y. Tanaka.
Discontinuation of infliximab in rheumatoid arthritis patients in clinical remission.
Mod Rheumatol, 18 (2008), pp. 460-464
[143]
J.S. Smolen, P. Nash, P. Durez, S. Hall, E. Ilivanova, F. Irazoque-Palazuelos, et al.
Maintenance, reduction, or withdrawal of etanercept after treatment with etanercept and methotrexate in patients with moderate rheumatoid arthritis (PRESERVE): a randomised controlled trial.
[144]
Y. Tanaka, S. Hirata, S. Kubo, S. Fukuyo, K. Hanami, N. Sawamukai, et al.
Discontinuation of adalimumab after achieving remission in patients with established rheumatoid arthritis: 1-year outcome of the HONOR study.
Ann Rheum Dis, 74 (2015), pp. 389-395
[145]
Y. Tanaka, T. Takeuchi, T. Mimori, K. Saito, M. Nawata, H. Kameda, et al.
Discontinuation of infliximab after attaining low disease activity in patients with rheumatoid arthritis: RRR (remission induction by Remicade in RA) study.
Ann Rheum Dis, 69 (2010), pp. 1286-1291
[146]
A. Van der Maas, W. Kievit, B.J. van den Bemt, F.H. van den Hoogen, P.L. van Riel, A.A. den Broeder.
Down-titration and discontinuation of infliximab in rheumatoid arthritis patients with stable low disease activity and stable treatment: an observational cohort study.
Ann Rheum Dis, 71 (2012), pp. 1849-1854
[147]
R.F. Van Vollenhoven, M. Ostergaard, M. Leirisalo-Repo, T. Uhlig, M. Jansson, A. Klackenberg, et al.
In rheumatoid arthritis patients with stable low disease activity on methotrexate plus etanercept, continuation of etanercept 50mg weekly or 25mg weekly are both clinically superior to discontinuation: results from a randomized, 3-armed, double-blind clinical trial.
Arthritis Rheum, 64 (2012), pp. 4171
[148]
M.A. Quinn, P.G. Conaghan, P.J. O’Connor, Z. Karim, A. Greenstein, A. Brown, et al.
Very early treatment with infliximab in addition to methotrexate in early, poor-prognosis rheumatoid arthritis reduces magnetic resonance imaging evidence of synovitis and damage, with sustained benefit after infliximab withdrawal: results from a twelve-month randomized, double-blind, placebo-controlled trial.
Arthritis Rheum, 52 (2005), pp. 27-35
[149]
M. Van den Broek, N.B. Klarenbeek, L. Dirven, D. van Schaardenburg, H.M. Hulsmans, P.J. Kerstens, et al.
Discontinuation of infliximab and potential predictors of persistent low disease activity in patients with early rheumatoid arthritis and disease activity score-steered therapy: subanalysis of the BeSt study.
Ann Rheum Dis, 70 (2011), pp. 1389-1394
[150]
S.M. Van der Kooij, S. le Cessie, Y.P. Goekoop-Ruiterman, J.K. de Vries-Bouwstra, D. van Zeben, P.J. Kerstens, et al.
Clinical and radiological efficacy of initial vs delayed treatment with infliximab plus methotrexate in patients with early rheumatoid arthritis.
Ann Rheum Dis, 68 (2009), pp. 1153-1158
[151]
P. Emery, M. Hammoudeh, O. FitzGerald, B. Combe, E. Martin-Mola, M.H. Buch, et al.
Sustained remission with etanercept tapering in early rheumatoid arthritis.
N Engl J Med, 371 (2014), pp. 1781-1792
[152]
J. Detert, H. Bastian, J. Listing, A. Weiss, S. Wassenberg, A. Liebhaber, et al.
Induction therapy with adalimumab plus methotrexate for 24 weeks followed by methotrexate monotherapy up to week 48 versus methotrexate therapy alone for DMARD-naive patients with early rheumatoid arthritis: HIT HARD, an investigator-initiated study.
Ann Rheum Dis, 72 (2013), pp. 844-850
[153]
T. Takeuchi, T. Matsubara, S. Ohta, M. Mukai, K. Amano, S. Tohma, et al.
Abatacept biologic-free remission study in established rheumatoid arthritis patients. Orion Study.
Ann Rheum Dis, 72 (2013), pp. A613
[154]
P. Emery, G. Burmester, V. Bykerk, B. Combe, D.E. Furst, E. Barre, et al.
Induction of clinical remission followed by drug-free withdrawal with abatacept combination and monotherapy in early RA: results from the AVERT study over 18 months.
[155]
N. Nishimoto, K. Amano, Y. Hirabayashi, T. Horiuchi, T. Ishii, M. Iwahashi, et al.
Drug free REmission/low disease activity after cessation of tocilizumab (Actemra) monotherapy (DREAM) study.
Mod Rheumatol, 24 (2014), pp. 17-25
[156]
T.W. Huizinga, P.G. Conaghan, E. Martin-Mola, G. Schett, H. Amital, R.M. Xavier, et al.
Clinical and radiographic outcomes at 2 years and the effect of tocilizumab discontinuation following sustained remission in the second and third year of the ACT-RAY study.
Ann Rheum Dis, 73 (2014), pp. 685
[157]
B.J. Van den Bemt, A.A. den Broeder, G.F. Snijders, Y.A. Hekster, P.L. van Riel, B. Benraad, et al.
Sustained effect after lowering high-dose infliximab in patients with rheumatoid arthritis: a prospective dose titration study.
Ann Rheum Dis, 67 (2008), pp. 1697-1701
[158]
N. Van Herwaarden, S. Herfkens-Hol, A. van der Maas, B.J. van den Bemt, R.F. van Vollenhoven, J.W. Bijlsma, et al.
Dose reduction of tocilizumab in rheumatoid arthritis patients with low disease activity.
Clin Exp Rheumatol, 32 (2014), pp. 390-394
[159]
I. Gonzalez-Alvaro, C. Martinez-Fernandez, B. Dorantes-Calderon, R. Garcia-Vicuna, B. Hernandez-Cruz, A. Herrero-Ambrosio, et al.
Spanish Rheumatology Society and Hospital Pharmacy Society Consensus on recommendations for biologics optimization in patients with rheumatoid arthritis, ankylosing spondylitis and psoriatic arthritis.
Rheumatology (Oxford), (2014),
[160]
M. Dougados, M. Soubrier, A. Antunez, P. Balint, A. Balsa, M.H. Buch, et al.
Prevalence of comorbidities in rheumatoid arthritis and evaluation of their monitoring: results of an international, cross-sectional study (COMORA).
Ann Rheum Dis, 73 (2014), pp. 62-68
[161]
A. Nakajima, E. Inoue, Y. Shimizu, A. Kobayashi, K. Shidara, N. Sugimoto, et al.
Presence of comorbidity affects both treatment strategies and outcomes in disease activity, physical function, and quality of life in patients with rheumatoid arthritis.
Clin Rheumatol, 34 (2015), pp. 441-449
[162]
V.K. Ranganath, P. Maranian, D.A. Elashoff, T. Woodworth, D. Khanna, T. Hahn, et al.
Comorbidities are associated with poorer outcomes in community patients with rheumatoid arthritis.
Rheumatology (Oxford), 52 (2013), pp. 1809-1817
[163]
X. Mariette, S. Rouanet, J. Sibilia, B. Combe, X. le Loet, J. Tebib, et al.
Evaluation of low-dose rituximab for the retreatment of patients with active rheumatoid arthritis: a non-inferiority randomised controlled trial.
Ann Rheum Dis, 73 (2014), pp. 1508-1514
[164]
J. Askling, E. Baecklund, F. Granath, P. Geborek, M. Fored, C. Backlin, et al.
Anti-tumour necrosis factor therapy in rheumatoid arthritis and risk of malignant lymphomas: Relative risks and time trends in the Swedish Biologics Register.
Ann Rheum Dis, 68 (2009), pp. 648-653
[165]
E. Baecklund, A. Iliadou, J. Askling, A. Ekbom, C. Backlin, F. Granath, et al.
Association of chronic inflammation, not its treatment, with increased lymphoma risk in rheumatoid arthritis.
Arthritis Rheum, 54 (2006), pp. 692-701
[166]
L. Carmona, L. Abasolo, M.A. Descalzo, B. Perez-Zafrilla, A. Sellas, F. de Abajo, et al.
Cancer in patients with rheumatic diseases exposed to TNF antagonists.
Semin Arthritis Rheum, 41 (2011), pp. 71-80
[167]
A. Gonzalez, H. Maradit Kremers, C.S. Crowson, K.V. Ballman, V.L. Roger, S.J. Jacobsen, et al.
Do cardiovascular risk factors confer the same risk for cardiovascular outcomes in rheumatoid arthritis patients as in non-rheumatoid arthritis patients?.
Ann Rheum Dis, 67 (2008), pp. 64-69
[168]
A. Low, K. Hyrich, M. Lunt, L. Mercer, C. Gale, K. Watson, et al.
P2. Incidence and severity of myocardial infarction in subjects receiving anti-tumour necrosis factor drugs for rheumatoid arthritis: results from linking the British Society for Rheumatology Biologics Register for Rheumatoid Arthritis and Myocardial Ischaemia National Audit Project.
Rheumatology, 53 (2014), pp. i34-i35

Please cite this article as: Sanmartí R, García-Rodríguez S, Álvaro-Gracia JM, Andreu JL, Balsa A, Cáliz R, et al. Actualización 2014 del Documento de Consenso de la Sociedad Española de Reumatología sobre el uso de terapias biológicas en la artritis reumatoide. Reumatol Clin. 2015;11:279–294.

Copyright © 2015. Elsevier España, S.L.U.. All rights reserved
Idiomas
Reumatología Clínica (English Edition)
Article options
Tools
es en

¿Es usted profesional sanitario apto para prescribir o dispensar medicamentos?

Are you a health professional able to prescribe or dispense drugs?