Journal Information
Vol. 17. Issue 4.
Pages 215-228 (April 2021)
Visits
4616
Vol. 17. Issue 4.
Pages 215-228 (April 2021)
Clinicpathologic Conference
Full text access
Update of the Mexican College of Rheumatology guidelines for the pharmacological treatment of rheumatoid arthritis, 2018
Actualización de las guías del tratamiento farmacológico de la artritis reumatoide del Colegio Mexicano de Reumatología 2018
Visits
4616
Mario H. Cardiela, Sandra Carrillob, Marcela Pérezc, Lilia Andraded, César Pacheco Tenae, Luis H. Silveiraf, Leonardo Limóng, Sergio Cerpah, Sergio Gutiérrez Ureñah,i, Sergio Duráni,j, Fedra Irazoque Palazuelosd, Sandra Muñoz Lópezd, Sandra Araceli Sicsik Ayalak, Leonor Barilel, María Azucena Ramos Sánchezm, Daniel Grajeda Portesm, Margarita Portelan, Alina Hernández Bedollan, José Luis García-Figueroao, Mauricio Monterop..., Carlos Abud-Mendozaq, Marco Ulises Martínez Martínezq, David Herrera van Ostdamq, Virginia Pascual-Ramosr, Javier Merayo-Chalicor, Istar Guzmán-Sánchezs, María Esther Pérez-Bastidast, Jorge Enrique Aguilar Arreolau, Alejandra López Rodríguezv, Greta Reyes-Corderow, Humberto Alfredo Ricardezx, María Fernanda Hernández Cabreray, Guadalupe Olvera-Sotoz, Daniel Xibillé Friedmannaa,
Corresponding author
daniel.xibille@uaem.mx

Corresponding author.
Ver más
a Centro de Investigación Clínica de Morelia, Morelia, Mich, Mexico
b Hospital Regional 1ro de Octubre, Mexico City, Mexico
c Coordinación de Investigación, Instituto Mexicano del Seguro Social (IMSS), Mexico City, Mexico
d Servicio de Reumatología, CMN 20 de Noviembre, ISSSTE, Mexico City, Mexico
e Facultad de Medicina, Universidad Autónoma de Chihuahua, Chihuahua, Mexico
f Departamento de Reumatología, Instituto Nacional de Cardiología Ignacio Chávez, Mexico City, Mexico
g Jefatura de Enseñanza e Investigación Hospital Central Norte PEMEX, Mexico City, Mexico
h Servicio de Reumatología, Hospital Civil de Guadalajara Fray Antonio Alcalde, Guadalajara, Jalisco, Mexico
i Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara, Jalisco, Mexico
j Clínica de Investigación en Reumatología y Obesidad, Guadalajara, Jalisco, Mexico
k Hospital de Especialidades No 71, Unidad Médica de Alta Especialidad (UMAE), Torreón, Coahuila, Mexico
l Hospital Ángeles del Pedregal, Mexico City, Mexico
m Unidad Médica de Alta Especialidad No. 25, Hospital de Especialidades IMSS, Monterrey, Nuevo León, Mexico
n Hospital de Especialidades CMN Siglo XXI, IMSS, Mexico City, Mexico
o Colegio Mexicano de Reumatología AC, Mexico City, Mexico
p Hospital Ángeles, Hospital Regional ISSSTE, Puebla, Puebla, Mexico
q Unidad de Investigaciones Reumatológicas, Hospital Central Dr. Ignacio Morones Prieto, Facultad de Medicina, Universidad Autónoma de San Luis Potosí, San Luis Potosí, S.L.P., Mexico
r Departamento de Inmunología y Reumatología, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
s Centro Médico Instituto de Seguridad Social del Estado de México y Municipios, Toluca, Estado de México, Mexico
t Colegio Jalisciense de Reumatología, Guadalajara, Jalisco, Mexico
u Hospital Civil de Guadalajara Dr. Juan I. Menchaca, Guadalajara, Jalisco, Mexico
v Dermatológico Country, Centro de Investigación de Enfermedades Autoinmunes, Guadalajara, Jalisco, Mexico
w Hospital Ángeles Chihuahua, Chihuahua, Mexico
x Centro de Especialidades Médicas del Instituto de Seguridad Social del Estado de Tabasco, Universidad Juárez Autónoma de Tabasco, División Académica de Ciencias de la Salud, Villahermosa, Tabasco, Mexico
y Hospital General Regional 220 IMSS, Toluca, Estado de México, Mexico
z Comisión Nacional de Protección Social en Salud, Mexico City, Mexico
aa Departamento de Investigación y Capacitación, Servicios de Salud de Morelos, Cuernavaca, Morelos, Mexico
Ver más
This item has received
Article information
Abstract
Full Text
Bibliography
Download PDF
Statistics
Figures (1)
Tables (3)
Table 1. Tuberculosis screening and management.
Table 2. Pregnancy and lactation and fertility.
Table 3. Vaccination in patients with RA.
Show moreShow less
Abstract

Therapeutic advances in rheumatoid arthritis require periodic review of treatment guidelines.

Objective

To update the Mexican College of Rheumatology guidelines on the pharmacological treatment of rheumatoid arthritis.

Method

Board certified rheumatologists from different health institutions and regions of the country participated. Work teams were formed that reviewed the previous guidelines, elaborated new questions, reviewed the literature, and scored the evidence that was presented and discussed in plenary session. The conclusions were presented to infectologists, gynaecologists and patients. Recommendations were based on levels of evidence according to GRADE methodology.

Results

Updated recommendations on the use of available medications for rheumatoid arthritis treatment in Mexico up to 2017 are presented. The importance of adequate and sustained control of the disease is emphasised and relevant safety aspects are described. Bioethical conflicts are included, and government action is invited to strengthen correct treatment of the disease.

Conclusions

The updated recommendations of the Mexican College of Rheumatology on the pharmacological treatment of rheumatoid arthritis incorporate the best available information to be used in the Mexican health care system.

Keywords:
Rheumatoid arthritis
Recommendations
Treatment
Therapeutic strategy and vaccination
Resumen

Los avances terapéuticos en la artritis reumatoide obligan a revisión periódica de las guías de tratamiento.

Objetivo

Actualizar las guías del Colegio Mexicano de Reumatología del tratamiento farmacológico de la artritis reumatoide.

Método

Participaron reumatólogos certificados de diferentes instituciones de salud y regiones del país. Se conformaron equipos de trabajo que revisaron las guías previas, elaboraron nuevas preguntas, revisaron la literatura y calificaron la evidencia, que fue presentada y discutida en sesión plenaria. Las conclusiones se comentaron con infectólogos, ginecobstetras y pacientes. Se emiten recomendaciones basadas en niveles de evidencia de acuerdo con la metodología GRADE.

Resultados

Se presentan recomendaciones actualizadas para el empleo de los medicamentos disponibles en México hasta 2017 para el tratamiento de la artritis reumatoide. Se enfatiza la importancia del control adecuado y sostenido de la enfermedad y se describen aspectos relevantes de seguridad. Se incluyen conflictos bioéticos y se invita a la acción gubernamental para fortalecer el tratamiento adecuado de la enfermedad.

Conclusiones

La actualización de las recomendaciones del Colegio Mexicano de Reumatología del tratamiento farmacológico de la artritis reumatoide integra la mejor información disponible para ser utilizada en el sistema de salud de México.

Palabras clave:
Artritis reumatoide
Recomendaciones
Tratamiento
Estrategia terapéutica y vacunación
Full Text
Methodology

In August 2017, a committee of experts from the Mexican College of Rheumatology (CMR) met to update its guidelines for the pharmacological treatment of rheumatoid arthritis (RA). Rheumatologists and methodological advisors from various health institutions participated. Coordinators were assigned if the subject did not represent a conflict of interest. The analytical framework was defined with a review of the previous version,1 some questions were removed, others were kept or restructured, and others were added due to the availability of new treatments. The words MeSH (Medical Subject Heading) were identified; where these did not exist, the keywords for the search were included under the term “All fields”. The PICO methodology, which identifies population (P), intervention (I), comparator (C) and outcome (O), was used in the search. Randomised clinical trials, systematic reviews and meta-analyses over the period January 2012 to September 2017, published in English or Spanish, on an adult population were considered. In the event of lack of evidence, observational studies or other clinical practice guidelines were included as a source of information. We searched PubMed, the Cochrane Library and websites.

Articles were screened by title, abstract and finally by full text. Participants were trained in the GRADE methodology for standardising the quality rating of evidence and strength of recommendation.2 A face-to-face meeting was held in April 2018 to discuss the recommendations until reaching a consensus. The relevant sections were discussed with infectologists, obstetrician/gynaecologists and patients. The new version was posted electronically on the CMR website for 10 days for comments. The final version was integrated and the treatment algorithm was updated. The quality of evidence by GRADE was classified as very low, low, moderate, or high. Expert opinion was classified as very low quality.3 The strength of the recommendations was rated as strong or weak. Recommendations that were considered important but could not be rated in terms of quality of evidence or strength of recommendation were classified as “good clinical practice”.4

ResultsWhat are the general principles of the treatment of rheumatoid arthritis?

  • Diagnose in the initial stages.

  • Treat appropriately.

  • Management preferably by a rheumatologist.

  • Aim to achieve and maintain remission or a low level of disease activity.

  • Individualise treatment.

  • Monitor periodically.

  • Address comorbidities.

  • Adjust management to clinical practice.

  • Take decisions together with the patient.

What are the principal aims of treatment?

The main objective of the treatment of RA is to achieve and maintain remission of the disease or at least a low level of disease activity taking the perspective of the patient into consideration.5–18

What is the efficacy of glucocorticoids in reducing pain and radiographic progression?

Twenty articles were reviewed and seven were included: (1) glucocorticoids added to conventional synthetic disease modifying anti-rheumatic drug (csDMARD) therapy in early RA19; (2) comparison of triple therapy with csDMARDs vs methotrexate in monotherapy and with low doses of glucocorticoids20; (3) comparison of dexamethasone vs methylprednisolone in pulses for control of relapses21; (4) radiographic follow-up at 2 years of patients with early RA treated with decreasing glucocorticoids vs increasing doses followed by strict control22; (5) response to glucocorticoids at 2 weeks as a predictor of effectiveness of induction therapy with csDMARDs at 3 months23; (6) intra-articular glucocorticoids and csDMARDs in treat-to-target in early RA,24 and (7) usefulness of glucocorticoids to decrease radiographic progression in RA.25

Glucocorticoids can be used with different doses and administration routes in RA.22,23 Use of the lower doses for the shortest time possible is preferred.

Glucocorticoids in combination with csDMARDs can be used as “bridge” therapy to achieve rapid remission.19

In patients who do not achieve complete remission with csDMARDs, the use of intra-articular glucocorticoids may be considered, especially in cases of persistent monoarthritis.24

Potential long-term adverse reactions should be monitored.25

Recommendations

  • The use of prednisone10mg/day or equivalent is recommended as “bridge” therapy with csDMARDs in patients with a diagnosis of RA. High quality of evidence. Strong recommendation.

  • In early RA (less than 6 months’ onset) with poor prognostic factors (seropositivity, high clinical activity, erosions) low doses of glucocorticoids (prednisone10mg/day or equivalent) are recommended to reduce disease activity and radiographic progression. High quality of evidence. Strong recommendation.

  • The use of high doses (methylprednisolone pulses) of intravenous (IV) glucocorticoids is only recommended in patients with severe extra-articular manifestations (mononeuritis multiplex, rheumatoid vasculitis). High quality of evidence. Strong recommendation.

When should treatment with csDMARDs be started to achieve remission, low disease activity and arrest of radiographic damage?

The evidence for this recommendation comes from studies of early RA whose aims were to achieve remission or low activity of RA and to arrest radiographic progression with the use of csDMARDs.

The results of one of the studies show that between 8.7% and 22.5% of the participants achieved remission and between 35.5 and 42.7% low disease activity at 3 and 6 months, respectively, with csDMARDs.26 Remission was observed at week 17 in 77% of the participants.27 Another study showed that the csDMARD group achieved remission in 49%, compared to 21% in the untreated group.28 Another publication29 reported that, in early RA, the use of methotrexate in combination with other csDMARDs was not superior to methotrexate in monotherapy in achieving remission, if glucocorticoids were used at the same time. Early remission predicts sustained clinical remission in patients with recent onset RA (OR=1.95; 95% CI: 1.02–3.74).30,31

Recommendation

  • We recommend the use of csDMARDs as soon as possible. High quality of evidence. Strong recommendation.

Which csDMARD and administration route should be considered first choice?

Methotrexate is still considered the cornerstone of treatment for RA. The recommendations are based on a meta-analysis of clinical trials that included 1432 leflunomide users, 922 methotrexate users, 133 sulfasalazine users, and 312 placebo users.32 The recommendation regarding the route of administration of methotrexate is based on a systematic review that included studies with different designs that compared the oral route with the parenteral route.33 The efficacy and toxicity of methotrexate are mainly related to the absorbed dose and not to the route of administration. Bioavailability is greater with parenteral methotrexate than with the oral route.33

Additional considerations

Oral methotrexate is preferred to parenteral methotrexate due to its ease of administration

Recommendations

  • -

    Methotrexate is recommended as the first line of treatment; when methotrexate is contraindicated we recommend leflunomide. High quality of evidence, strong recommendation.

  • -

    It is suggested that methotrexate should be started orally due to ease of administration. If doses>15mg/week are required, parenteral methotrexate should be considered. High quality of evidence. Strong recommendation.

When should monotherapy be used and when should combined therapy with csDMARDs be used?

A meta-analysis reported a higher probability of achieving ACR 50 with combined therapy (56%–67%) vs methotrexate as monotherapy (41%); additionally, patients who had not received methotrexate and who were treated with combination therapy had lower radiographic progression, and with triple therapy (methotrexate, sulfasalazine and hydroxychloroquine) a 61% probability of achieving an ACR score of 50 vs methotrexate and biologics, which had responses between 24% and 64%.34 There was a higher rate of adverse events (AE) in the combined therapy groups.34

Additional considerations

In patients with RA who do not respond to csDMARDs in monotherapy, similar results are obtained with the use of triple csDMARD therapy (methotrexate, sulfasalazine and hydroxychloroquine) or combined therapy with methotrexate and biological DMARDs (BioDMARDs). In our setting, triple therapy should be the first option of combined therapy.35

Recommendation

  • -

    We recommend the use of monotherapy with csDMARDs in patients with active RA with no previous exposure to DMARDS. Moderate quality of evidence. Strong recommendation.

  • -

    We suggest the use of combined therapy with csDMARDS in patients with active RA with poor prognostic factors or with previous failure with a csDMARD. Low quality of evidence. Weak recommendation.

When is it indicated to start biological DMARDs or JAK inhibitors?

The biological DMARDs (BioDMARDs) and JAK inhibitors available in Mexico are innovative anti-TNF drugs such as etanercept, infliximab, adalimumab, certolizumab pegol and golimumab; anti-IL6 (tocilizumab); abatacept, which is a costimulation blocker; and rituximab (anti-B cells). Biosimilars of etanercept and infliximab are also available. The JAK inhibitors are tofacitinib and baricitinib.

We considered the efficacy and safety data from clinical trials of BioDMARDs or JAK inhibitors in different clinical settings in RA. Early or established RA, without previous treatment or with inadequate response to csDMARDs or biological therapy, their use in monotherapy or in combination with csDMARDS were taken into account. Since the indications and their precautions are similar, they are described in a general manner and special considerations with some of them are mentioned separately. The assumption that the efficacy of different biological therapies and JAK inhibitors is similar comes not only from indirect comparisons of clinical trials conducted,36–39 but also from direct comparison studies: ADACTA, AMPLE, EXXELERATE, ACTION, ORAL STRATEGY, RA-BEAM.40–45

The use of BioDMARDS or JAK inhibitors in patients with established RA, with moderate to intense clinical activity, who have had insufficient response to csDMARDS has proved effective. Similar percentages are reported in ACR responses 20, 50 and 70, with average responses of 70%, 40% and 25%,36 respectively, at 24 weeks of treatment and reduction of disease activity by DAS281.2 up to 70% at 2 years.38,39 The rates of ACR response 20, 50 and 70 are slightly lower: 60%, 30% and 15%, respectively, when the population has a history of a failure to respond to biological therapy. In patients with early RA, the clinical responses are better, with low disease activity achieved in up to 40% of patients and remission in 25%, maintained for up to 5 years.36 Similarly, controlled clinical and extension trials have shown that biological therapy or with JAK inhibitors, combined in both cases with csDMARDs (generally methotrexate), is superior to monotherapy for arresting structural damage.36–39 In general, the best clinical responses are achieved by administering these treatments in combination with methotrexate, especially in the case of anti-TNF.46–49

Some studies with tocilizumab and JAK inhibitors have shown that monotherapy treatment is superior to methotrexate and equivalent to therapy in combination with methotrexate.50,51

There is unequal availability in Mexico's health institutions of these therapeutic options, since availability is limited in the Ministry of Health and the Seguro Popular health initiative. It would be desirable for all institutions to have some of these drugs, with a different mechanism of action and administration route, for patients with moderate to high disease activity, without response to treatment with csDMARDs at appropriate doses over sufficient time.

Additional considerations

No BioDMARD has an obvious therapeutic advantage over another.

Recommendations

  • -

    We recommend the use of BioDMARDs or JAK inhibitors in patients with moderate to high RA activity with inadequate response to treatment with csDMARDs. High quality of evidence. Strong recommendation.

  • -

    In patients who have failed to respond to the first anti-TNF, a second anti-TNF can be used or they can be changed to a BioDMARD with a different mechanism of action. High quality of evidence. Strong recommendation.

  • -

    We recommend a change of mechanism of action in patients with RA with moderate or severe disease activity with inadequate response to BioDMARDS or JAK inhibitors. High quality of evidence. Strong recommendation.

  • -

    We recommend the use of tocilizumab or JAK inhibitors in patients with RA with moderate to severe disease activity in whom monotherapy is decided. High quality of evidence. Strong recommendation.

  • -

    In patients who require biological treatment, the decision about therapeutics with approved biological products (innovative or biocomparable) should be dictated by an individual risk/benefit assessment and not based solely on economic aspects. Good clinical practice.52

What are the safety aspects to be taken into account when using BioDMARDs or JAK inhibitors?Anti-TNF

Infections. In patients with RA who receive ant-TNF, a higher risk of infections has been described, and some severe.53 The adjusted risk of any serious infections, compared with the use of csDMARDs, ranges from 1.1 to 1.8,4,55 which seems to be higher during the first year.56 British and Japanese registers show that etanercept could have a lower risk of serious infections than the monoclonal antibodies.38,57,58 The evidence indicates that patients that use anti-TNF should be closely monitored for the risk of infections, that they should be treated early when they acquire an infection, and that these drugs should be discontinued while the infection is active.

Tuberculosis (TB). The adjusted risk of TB in patients with RA using anti-TNF ranges from 2.7 to 12.5.54 All patients should be screened for TB, and those with a diagnosis of latent or active disease should receive the appropriate treatment. Retrospective studies have shown that it is safe to restart anti-TNF in patients receiving appropriate treatment for latent or active TB59,60 (Table 1).

Table 1.

Tuberculosis screening and management.

Recommendation  Quality of evidence  Strength of recommendation 
Tuberculosis screening (TB)
• TB screening is recommended by taking a clinical history asking about a history of living with people with active TB, physical examination, tuberculin test (or IGRA) and chest x-ray, in all patients with RA to be protocolised for the use of BioDMARDs or JAK inhibitors.  Very low  Good clinical practice 
• It is suggested that screening should be repeated annually in patients who are under treatment with BioDMARDs and JAK inhibitors, given the risk of reactivation of latent TB or de novo infection.  Very low  Good clinical practice 
• It is recommended that prophylactic TB therapy should be started in all patients presenting positive PPD5mm or positive IGRA, regardless of whether they have been BCG-vaccinated or not, and in COMBE positive patients with chest x-ray and signs suggestive of TB before starting therapy with BioDMARDs (especially with anti-TNF type drugs)61,62 or JAK inhibitors  Very low  Good clinical practice 
Start of treatment with BioDMARDs or JAK inhibitors in patients who are receiving prophylaxis or treatment for established TB
• In patients with latent TB it is recommended to start or restart treatment with BioDMARDs or JAK inhibitors after one month of treatment with antituberculous drugs62,63  Very low  Good clinical practice 
• In patients with active TB it is recommended to start or restart BioDMARD/s or JAK inhibitors on completion of antituberculous treatment  Very low  Good clinical practice 
• Patients with latent TB with positive tuberculin or IGRA results require clinical follow-up for appropriate diagnosis in case of reactivation, since repeated tests do not help in diagnosing recurrent TB  Very low  Good clinical practice 

Opportunistic germs. As for other specific infections, recent studies do not show an increased risk of infection by herpes zoster virus,54,64 and a systematic review found a greater risk of opportunistic infections.64

Reactions to application. These occur between 5.8% and 53.8%; most are mild or moderate and in very few cases require the drug to be discontinued.65

Cancer. Several recent meta-analyses have concluded that there is no increased risk of cancer associated with the use of anti-TNF drugs in patients with RA.54–68 A 2012 meta-analysis found a slight increase in non-melanoma skin cancer, which has not been confirmed in more recent meta-analyses and population studies.55,58,66,67,69 There is evidence for a slight increase in the risk of melanoma (aHR 1.5).54

The use of adalimumab or certolizumab during pregnancy appears to be safe; recent studies show that the use of these drugs is not associated with a higher risk of teratogenicity, miscarriage or foetal death compared to that expected for the general population.70–72

Rituximab

The most relevant safety aspects of the use of rituximab are associated with its therapeutic effect and route of administration. Study of the long-term safety of rituximab with follow-up to more than 11 years summarises information from 8 controlled clinical trials, 2 long-term extension studies and one prospective observational study with experience in 14,816 patients/year. Most discontinuations of rituximab occurred in the first two infusion cycles; the main reason for discontinuation was not associated with the safety of the drug (22%) and only 7% discontinued due to adverse events (AE). Most of these AE occurred in the first 6 months and the frequency and nature of the events did not change throughout follow-up. There were 78 deaths (.58 patients/year) in the rituximab group, and 7 in the placebo group (.63 patient/year). The most common infections were respiratory and similar to the rest of the BioDMARDs.58 The frequency of infections did not increase with follow-up time or the number of applications. The most frequent severe infection was pneumonia (2%). The incidence of severe infections by opportunistic microorganisms was infrequent (.05 events/patients/year). Two cases of uncomplicated pulmonary TB were reported, and one new case of hepatitis B. Myocardial infarction had a rate of .39events/100 patients/year, which is similar to that described in patients with RA. No evidence of an increased risk of the development of neoplasms was found. The most frequent solid tumour was breast cancer, and its frequency does not represent an increase with respect to the benchmark population with RA.73

Due to the inhibition of B-lymphocytes (CD20) and the secondary decrease of immunoglobulins, measurement of IgG, IgM and IgA levels prior to the administration of each cycle of rituximab is recommended. European guidelines on the use of rituximab recommend monitoring immunoglobulin levels, with close monitoring for the possibility of infection in patients with low IgG levels.74

Rituximab increases the risk of reactivation of hepatitis B (OR: 7.2; 95%CI: 5.3–9.9), and therefore its administration in these patients should be monitored and its use avoided in cases of active infection.75

Progressive multifocal leukoencephalopathy has been associated with the administration of rituximab in 9 cases out of 351,396 RA patients treated with rituximab until 2015. The incidence rate of leukoencephalopathy in RA patients receiving rituximab is considered to be 2.56 per 100,000 patients.

Special consideration: rituximab can be used as a first choice of treatment in patients with a history of cancer.

Abatacept

Although abatacept has been shown to have a better safety profile than other biologics in the treatment of RA, hepatitis B (HBV) deserves special mention. This has been analysed in several studies; in one study, 72 patients received abatacept; 47 were inactive carriers, 21 were occult, and 4 were chronic carriers of HBV, they all had normal baseline liver function tests or undetectable levels of HBV DNA, except those with chronic active hepatitis; 13 patients received prophylaxis with lamivudine and 4 with adefovir or tenofovir, and after 316 follow-up visit none of the patients had reactivation of hepatitis B, and there were no related AEs of interest.76

One study showed an association of older patients with a higher rate of discontinuation due to AE per 100 patients/year, especially severe infections (1.73 in the very young, 4.65 in the middle-aged, 5.90 in the elderly and 19.38 in the very elderly, P<.001).77 In 219 patients with abatacept combined with methotrexate in a long-term extension study, 114 (52.1%) completed 7 years, and safety was consistent with the initial study periods. Another long-term extension study conducted in patients with RA who were non-responders to methotrexate, received subcutaneous (SC) abatacept, identified 1385 patients who completed the double-blind stage; 1372 entered the long-term extension study and 945 (68.8%) completed ≥5 years of treatment. During the extension study, 97 (7.1%) patients discontinued treatment due to an AE. The incidence rate (event/100 patients/year of exposure) of AE of interest was: serious AE 7.73 (95% CI: 6.96–8.58), infection 38.60 (95% CI: 36.24–41.12), severe infections 1.68 (95% CI: 1.35–2.07), malignancy 1.09 (95%CI: .84–1.42), and autoimmune disease 1.33 (95%CI: 1.05–1.69), remaining stable over time. No association was observed between immunogenicity and worsened safety or loss of efficacy of abatacept. This study at 5 years established the safety of SC abatacept (125mg/week), showing consistent safety and lasting efficacy in the long-term treatment of patients with RA and inadequate response to csDMARDs.78 Although the presence of anti-drug-antibodies associated with abatacept has been demonstrated, these are not neutralisers and are not associated with a loss of efficacy, as seen with other biological drugs.79 Based on the above, we believe that abatacept maintains constant safety, as well as sustained efficacy over 7 years in patients with established RA and inadequate response to methotrexate,80 with fewer discontinuations due to AE (3.8% vs 9.5%), severe AE (1.6% vs 4.9%) and serious infections (0/12 vs 9/19 patients) than the comparison group.42

The use of abatacept has not been associated with interstitial lung disease.81

Tocilizumab

Tocilizumab IV and SC as monotherapy or in combination with other csDMARDs has generally been well tolerated in clinical trials and in clinical practice after 9 years’ experience of the treatment.

The most common AE (>10%) were respiratory infections and hypercholesterolaemia.82 In clinical trials at 6 months the rate of infections with tocilizumab IV (8mg/kg) plus csDMARDs was 127events/100 patients/year, with a rate of severe infections of 5.3events/100 patients/year. In long-term exposure the general infection rate with tocilizumab was 108events/100 patients/year, and that of severe infections, 4.7events/100 patients/year.82

The most common AE were “infections and infestations” with a frequency of 7%–43% in the monotherapy group, and 6%–42% in the combined treatment group, nasopharyngitis being the most common. The drug was discontinued due to an AE in 6.2% of the patients, 9% in monotherapy, and 5.5% in combination with csDMARDs. The reasons for discontinuing treatment were skin and subcutaneous disorders with monotherapy (1.4%), and laboratory abnormalities in combination with csDMARDs (1.1%). The drug was discontinued due to lack of efficacy in 29 patients (1.6%). In addition, headache (7%) and arterial hypertension (6%) have been reported.82,83

Cases of diverticular perforation have been reported. Clinical trials at 6 months show rates of .26/100 people/year, and long-term exposure shows similar rates of .28/100 people/year.82

In studies with post hoc analysis and in long-term studies of treatment with tocilizumab IV (average of 3.7 years of follow-up) was associated with a cardiovascular event rate of 3.4events/100 people/year, with an average time for the first event of 680 days. A multivariate analysis found that the predictors of CVD were: advanced age (HR: 1.07), history of heart disease (HR: 2.32), higher DAS28 (HR: 1.36) and higher total cholesterol to HDL cholesterol ratio (HR: 1.33).82

Severe AE that occurred in between 3% and 5% of the total population studied were pneumonia, herpes zoster, increased aminotransferases, hypersensitivity and erythema at the puncture site.83 In addition, Dyslipidaemia/hypercholesterolaemia and gastrointestinal disorders hove been reported: pain, distension and intestinal haemorrhage, and folliculitis.84

Liver AE had a rate of .78events/100 patients/year. Of the patients, 2.5% had to discontinue the drug due to elevation of aminotransferases.85

Tocilizumab should not be used during pregnancy. There is a risk of miscarriage and foetal death with exposure to high doses in animals; the risk to humans is not known with certainty. Of the prospective and retrospective cases, 21.7% and 28.7% resulted in miscarriage, respectively. Malformations have been reported in 4.5% and premature births in 31.2%.82

JAK inhibitors

Tofacitinib and baricitinib share safety aspects that are important to monitor.

Because these drugs are of recent use in our setting, many recommendations set out here are based on expert advice and not necessarily on information from pivotal studies. Clinicians should become familiar with the following general recommendations on JAK inhibitors:

Infections. These are the most frequently reported AEs. Most are usually mild or moderate, upper respiratory (14%–17%), urinary tract (3.4%–11%) and bronchitis (up to 11%) are the main infections.86–88 Herpes zoster has been reported in 3.9% of patients treated with baricitinib and in 4.39% of those treated with tofacitinib in the global programme, and 3.39% in Latin America.86,89 The frequency of severe infections ranges from 1.1% to 3.1%.40,89 Although opportunistic infections (specifically TB) have been reported in patients treated with JAK inhibitors, the frequency tends to be the same as for non-anti-TNF BioDMARDs. In general, the following risk factors have been described for infections: age>65 years, comorbidities such as diabetes mellitus, obesity and chronic obstructive pulmonary disease, and the use of prednisone>7.5mg/day.88,90 Discontinuing administration of these drugs is suggested if moderate or severe infection is suspected or confirmed, and it should be restarted once it has been resolved.

Lymphopenia. Lymphopenia500cells/ml3 can confer an increased risk of serious infections. It is suggested that treatment with JAK inhibitors should be temporarily or permanently discontinued if this figure is reached.40,90

Alterations in liver function tests. Elevations of ALT and/or AST levels have been observed with JAK inhibitors, but elevations ≥3 times (1.4%) occur infrequently and are generally asymptomatic and transient. They are more frequent in combination therapy with methotrexate. Periodic surveillance is recommended.86,88,91–93

Lipid profile and cardiovascular safety. Elevated total cholesterol, LDL cholesterol and HDL cholesterol has been described, with no changes in the total cholesterol/HDL ratio, in up to 49% of patients treated with JAK inhibitors, and is more frequent during the first 3 months of treatment.94 These alterations are usually reversed with statins.95 The clinical relevance of long-term cardiovascular risk is unknown.

  • Thrombosis. There is an event of special interest in the safety profile of baricitinib, due to an increase in deep vein thrombosis and/or pulmonary thromboembolism. Special caution is advised with the use of baricitinib in patients with risk factors for deep vein thrombosis or pulmonary thromboembolism. If any of these events occur, baricitinib should be discontinued, promptly assessed and appropriate treatment started. The presentations of 2mg and 4mg every 24h have been approved in Mexico.96 However, the USA's Food and Drug Administration (FDA) have only approved the 2mg presentation, taking events of thrombosis into account.97

  • Other laboratory alterations. These have been described with both drugs and very infrequently can cause anaemia, neutropenia, non-significant creatinine elevation86,92,93 and transient elevations of CPK. Up to 2% of patients who receive barcitinib present thrombocytosis.

  • Neoplasias. To date, JAK inhibitors have not shown an increased risk of neoplasia. However, as with all other drugs, long-term follow-up is recommended with international registers and strict monitoring in the event of high suspicion.98,99

Recommendations

  • -

    We recommend strict monitoring of patients with RA receiving BioDMARDs or JAK inhibitors for the appropriate diagnosis and treatment of infections. High quality of evidence. Strong recommendation.

  • -

    The use of BioDMARDs or JAK inhibitors is not recommended in patients with active infection. High quality of evidence. Strong recommendation.

  • -

    We recommend considering other treatment options in patient with a high risk of recurring infections. High quality of evidence. Strong recommendation.

  • -

    We recommend monitoring AE related to the safety profile and mechanism of action of each BioDMARD or JAK inhibitor. High quality of evidence. Strong recommendation.

  • -

    We recommend screening and monitoring for skin cancer in patients with RA under treatment with BioDMARDs. High quality of evidence. Strong recommendation.

When should reduction or discontinuation of csDMARDs and/or BioDMARDs be considered in patients with rheumatoid arthritis (optimisation)?

Consistently, the information shows a higher relapse rate in patients with RA in whom treatment with csDMARDs or BioDMARDs100–105 is reduced or discontinued. The incidence of relapse is lower with a decrease in dose than with discontinuing BioDMARDs. The incidence of relapse in patients following decrease of csDMARDs was 41%, and of BioDMARDs 37%.100

Risk factors for relapse

The following are considered predictors of relapse: positivity of anti-PCC or rheumatoid factor, longer duration of disease, and late start of treatment. Another predictor of relapse is the degree of remission, intense remission with DAS28<1.98 conferring a lower risk; serological markers have also been proposed, such as levels of interleukin-6 and matrix metalloproteinase-3, as well as the detection of synovitis with Power Doppler by musculoskeletal ultrasound.102,104,106–111

Recommendations

  • -

    We suggest optimising treatment in patients in sustained clinical remission (at least 6 months). Optimisation should be individualised and consensual. The logistics of the hospital should be taken into consideration.1,6,106,112,113 Moderate quality of evidence. Weak recommendation.

  • -

    We suggest sequential reduction of the treatment under consideration to start drugs that generate a greater risk of AE in the short or long term, and an increase in costs.100,101,106,113 It is desirable to start glucocorticoid reduction and discontinuation. Moderate quality of evidence. Weak recommendation.

  • -

    We suggest reducing the dose from 20% to 50% of csDMARDs or BioDMARDs, or increasing the period of application, in patients who have remained in remission (DAS28<2.6 or equivalent) for at least 6 months.5,104,112–116 Moderate quality of evidence. Weak recommendation.

  • -

    We suggest discontinuing BioDMARDs in patients with early RA or established RA if the patient is in remission, preferably intensive (DAS28<1.98) for at least 6 months, if they are receiving the minimum dose of BioDMARDs and when there is no evidence of progression on imaging tests.114,116–119 Moderate quality of evidence. Weak recommendation.

  • -

    We recommend strict monitoring to detect relapse, at least every 12.16 weeks, and measuring disease activity by standardised scales.6,102,113,114 Moderate quality of evidence. Weak recommendation.

  • -

    In the event of a relapse, we recommend adjusting the dose or interval of the DMARDs with the aim of achieving remission again.113,114,116 Moderate quality of evidence. Weak recommendation.

What is the role of pharmacological treatment of rheumatoid arthritis in pregnancy, lactation and fertility?

The treatment of RA in pregnancy should be team managed where the patient, the obstetrician/gynaecologist and the rheumatologist maintain adequate communication before, during and after the pregnancy. Disease activity, therapeutic modifications, treatment options in the case of RA activity during gestation, puerperium and lactation should be carefully assessed in the preconception clinic. Disease control should be prioritised and the potential risks to the mother and foetus from both the use and discontinuation of a drug must be considered. Table 2 summarises the recommendations in pregnancy, lactation and fertility.

Table 2.

Pregnancy and lactation and fertility.

Discontinuation of DMARDs in planned pregnancy
Drug and references  Time of discontinuation prior to conception  Quality of evidence  Strength of recommendation 
Methotrexate129–133  At least 3 months  High  Strong 
Leflunomide131,132,134,135  At least 2 years or cholestyramine wash-out  High  Strong 
Rituximab136,137  12 months  High  Strong 
Tocilizumab133,136,138,139  3 months (5 half lives)  Low  Weak 
Abatacept132,140,141  10 weeks  Low  Weak 
Anti-TNF132,142–146  Can continue  Low  Weak 
Tofacitinib132,136,145  2 months  Low  Weak 
Control of clinical activity in pregnancy
Drug and references  Recommended, suggested or contraindicated  Gestational stage  Quality of evidence  Strength of recommendation 
Prednisone132–134,147–149  Recommended  Entire pregnancy  High  Strong 
Antimalarials150  Recommended  Entire pregnancy  High  Strong 
Sulfasalazine2g/day+5mg/d folic acid151  Recommended  Entire pregnancy  High  Strong 
Azathioprine132,134  Recommended  Entire pregnancy  High  Strong 
Certolizumab pegola,152  Suggested  Entire pregnancy  High  Weak 
Infliximab  Suggested  First 20 weeks  Low  Weak 
Adalimumab72  Suggested  Entire pregnancy  Moderate  Weak 
Etanercept  Suggested  Up to week 30  Low  Weak 
Golimumab  Contraindicated  Entire gestation  Very low  Weak 
Anti-TNF biocomparable  Contraindicated  Entire gestation  Very low  Weak 
Treatment during lactation in patients with active RA
Drug and references  Additional considerations  Quality of evidence  Strength of recommendation 
Prednisone130,149  Dose 7.5–10mg  High  Strong 
NSAID133  Preferably use those with a short half-life such as ibuprofen and paracetamol and administer after each breast feeding  Moderate  Weak 
Antimalarials130,133  Hydroxichloroquine and chloroquine  High  Strong 
Sulfasalazine130,132,133  Precaution with premature infants, with G6PD deficiency or hyperbilirubinaemia  Moderate  Weak 
Azathioprine  Precaution in patients with thiopurine methyltransferase deficiency  Moderate  Weak 
Certolizumab pegola,53,154  Low responsibility of the doctor  Moderate  Weak 
Methotrexate and leflunomide130–133  These are contraindicated  Moderate  Strong 
Fertility
Recommendations and references  Quality of evidence  Strength of recommendation 
• Evaluate gestational desire from diagnosis of RA and regularly  Low  Good clinical practice 
• Offer preconception counselling about fertility, attitude to unplanned pregnancy and preconception planning with non-teratogenic drugs155–157  Low  Good clinical practice 
• Recommend control of disease before conception  Low  Good clinical practice 
• Avoid the use of NSAIDs  Low  Good clinical practice 
a

In pregnant patients with moderate to high disease activity and in cases of lactation, the panel discussed and concluded that caution is necessary with the administration of certolizumab pegol, that studies showing little bioavailability in the newborn and in milk do not guarantee the absence of toxicity, that the risk benefit should be assessed and the possibility of infection should be carefully monitored.

What are the vaccination recommendations for patients with rheumatoid arthritis?

Infection constitutes one of the main causes of morbidity and mortality in RA patients, and some can be prevented with vaccination. Vaccination coverage in patients with RA is estimated as suboptimal and the role of the rheumatologist in promoting vaccination is stressed, preferably before starting immunosuppressive therapy.120Table 3 describes the main recommendations on vaccination for patients with RA.

Table 3.

Vaccination in patients with RA.

Vaccine  Recommendation  Quality of evidence  Strength of recommendation 
All  When possible, administer in the stable phase of the disease or prior to drug therapy  Low  Weak 
Influenza24,158 (annual in specified season)  Use is recommended before the season. Tetravalent use is preferred to trivalent. The live virus vaccine (through inhalation) is contraindicated.In the case of the use of methotrexate discontinuation is recommended 2 weeks prior to vaccination.In the case of rituximab an interval between vaccination and the biological drug of at least 6 months is recommended.High dose vaccine for people over 65  Low  Weak 
Pneumococcus158  PCV 13 (few studies as yet). Use single dose preferably prior to starting immunosuppressant treatment, followed by PPV23 one dose 8 weeks after the initial dose of PCV13, and one more dose of PPV23 5 years after the first dose of PPV23161  Low  Weak 
Herpes zoster75,159–163(attenuated live virus)  Apply before synthetic DMARD therapy. It is suggested it should be applied 2 weeks prior to starting biological therapy and 4 weeks before JAK inhibitors  Low  Weak 
HPV  It is suggested that it should be given to women under the age of 26 and men under the age of 21  Low  Weak 
Hepatitis B  It is suggested that it should be applied for patients with RA with negative surface antigen and those with risk factors for acquiring hepatitis B  Low  Weak 
Tdap (tetanus, diphtheria, pertussis)  Single dose for those aged over 19 years who have not received the vaccine  Low  Weak 
What are the recommendations for the screening and treatment of latent tuberculosis in patients with rheumatoid arthritis under management with BioDMARDs and JAK inhibitors?

Good clinical practice recommendations on screening for TB are based on the TBNET study,121 which is limited to intradermal reaction positivity or Interferon Gamma Release Assay (IGRA) in patients with RA; however, TB was not sought in positive cases. The Enzyme-Linked ImmunoSorbent Assay (ELISA) and the Enzyme-Linked Immunosorbent Spot (ELISPOT) are recommended due to supposed greater sensitivity and specificity compared to tuberculin in patients who have previously received BCG.122

IGRA is not readily available in Mexico, and is also expensive as a screening test. Sensitivity for latent TB in general is similar for PPD and IGRA. In the case of active TB the use of IGRA is recommended.123

In case of PPD (≥5mm) or IGRA positivity, the use of prophylactic therapy with isoniazid is recommended for 9 months.62 However, a recent study showed that the use of rifampicin for 4 months was not inferior in efficacy to the use of isoniazid for 9 months.63

Additional considerations

Due to the prevalence of TB in Mexico and the low availability of IGRA as a screening test, starting prophylaxis of latent TB in patients with RA treated with BioDMARDs or JAK inhibitors with positive PPD is recommended.

Bioethical aspects

In the recommendations issued, priority is given to the individual needs and preferences of each patient, prioritising the principle of autonomy.124 In the context of chronic diseases such as RA, the patient not only authorised but, together with the medical team, becomes a fundamental part of the execution of the treatment plan (decision autonomy).125

On the other hand, it is necessary to recognise the impact of health disparities on the outcomes of RA patients. In particular, we refer to the lack of distributive justice,125,126 and one of the dimensions of this problem is non-existent or limited access to high-cost drugs such as the BioDMARDs and JAK inhibitors. Bioethical reflection in this regard must address two aspects. The first is the decision (or otherwise) only to include ideal (albeit unreal) clinical scenarios in the current treatment guidelines in which total health coverage is assumed, with the purpose of flagging up the path to follow to the relevant authorities. However, we should not overlook the fact that bioethics must also give specific answers to real situations and not be limited to ethics of unattainable maximums127; in this regard, we as rheumatologists have learned that the early identification of patients with RA and treatment with csDMARDs favourably changes outcomes of patients; implies greater effort and dedication, but implementation is feasible, necessary and ethically justified, since in many clinical contexts it can even overcome the lack of high-cost drugs. The second aspect, linked to the previous one, concerns the effort we must make to raise the awareness of the relevant authorities and include RA as a disease with catastrophic expenditure.128

Finally, possible conflicts of interest have not gone unnoticed in the planning and development of this document (https://es.unesco.org/). Many of the authors were selected based on their experience in the subject, which possibly implies participation and authorship with some of the assessed therapeutic options or with certain reviewed papers. We consider that this experience is valuable, but there is a potential conflict of interest that we declare in this article.

Conclusions

Although there are different updated therapeutic guidelines for the treatment of RA, it is very important that the recommendations issued consider the health system that they address. The CMR has updated their treatment recommendations for RA, as well as the treatment algorithm (Fig. 1). The main contributions of this update are as follows: the recommendations of BioDMARDs and JAK inhibitors were combined in the same treatment line, information on pregnancy, lactation, fertility and vaccination is included in the text and the management algorithm was updated. This update was prepared for aspirational purposes and highlights the importance of keeping at least some of these high-cost drugs available in the different health subsystems.

Fig. 1.

Rheumatoid arthritis treatment algorithm. NSAID: nonsteroidal anti-inflammatory drug; CDAI: Clinical Disease Activity Index; CQL: Chloroquine; DAS28: Disease Activity Score 28; csDMARD: Conventional synthetic disease-modifying antirheumatic drug; BioDMARD: biological disease-modifying antirheumatic drug; HCQ: hydroxychloroquine; LEF: leflunomide; MTX: methotrexate; PDN: prednisone; SDAI: Simplified Disease Activity Index; hidroxychloroquine; LEF: leflunomide. * Initial clinical, serological and radiological assessment of the disease. ** Factors of poor prognosis: seropositivity, erosive disease, high level of clinical activity and extra-articular manifestations. *** Assessment of clinical response can be from 4 to 12 weeks in the case of active disease, every 12 weeks in the case of low disease activity and up to every 6 months in the case of sustained remission, without neglecting pharmacovigilance. Clinical remission or a low level of activity is considered adequate response.

(0.4MB).

Appropriate treatment of RA should include more efficient systems and processes, with an optimal number of committed rheumatologists, with sufficient time to provide adequate care, as well as pharmacological options that enable good control of the disease. None of the above can be achieved without government support and the commitment of all participants.

Financing

This project was funded by the Mexican College of Rheumatology AC. The Mexican College of Rheumatology thanks Lilly, Roche, Sanofi and UCP for their financial support through an unrestricted educational fund.

Conflict of interests

Mario H. Cardiel declares that he has been a principal investigator, consultant and speaker for Abbvie, Astellas, Eli-Lilly, Janssen, Gilead, Pfizer, Roche.

Sandra M. Carrillo-Vázquez declares that she has been a speaker with Roche, Eli Lilly, Pfizer, Janssen, Novartis, Abbvie, Bristol. She has been sponsored to attend congresses in the past 3 years by Roche, Pfizer, Novartis, Asofarma, UCB, and has collaborated in research protocols with Genentech, GSK, Pfizer, SunPharma, Mallinkrodt ARD, Anthera Pharmaceuticals, Celgene.

Leonardo Limón-Camacho declares having collaborated with Pfizer, Lilly, BMS, Roche, Janssen, UCB, Abbvie, Amgen and Novartis. Sergio R. Gutiérrez Ureña declares having been a consultant, speaker and having received sponsorship for academic events from Pfizer, Lilly, Abbvie, Amgen, Takeda. Sergio Duran-Barragán declares having collaborated with: AMGEN, BMS, Eli Lilly, Glaxo, Merck, Pfizer, Mallinckrodt, Amylin, R-Pharm Speaker: BMS, Roche, Eli Lilly.

Lilia Andrade Ortega declares having received sponsorship from Bristol Myers Squibb, Celgene, Janssen, Novartis, Pfizer, Roche and UCB.

Sandra Araceli Sicsik Ayala declares having been a speaker for Roche, UCB, Pfizer, Bristol, Novartis, Abbvie y Probiomed. She received sponsorship as an investigator from Abbvie, Eli Lily and GlaxoSmithKline.

Leonor Barile Fabris declares being a speaker and consultant for UCB, Pfizer, Abbvie, Roche, Celgene.

María Azucena Ramos Sánchez declares having received sponsorship from Abbvie Farmacéutica, Novartis, Pfizer, Roche and UCB.

Daniel Octavio Grageda Portes declares being a speaker for Roche.

Margarita Portela Hernández declares having received sponsorship from Roche, Pfizer, Bristol, UCB, Abbvie, Janssen and Lily.

José Luis García-Figueroa declares having been sponsored by Expanscience and Bristol Myers Squibb.

Cesar Pacheco-Tena declares having collaborated with Abbvie, Roche, Bristol-Myers-Squibb, UCB, Merck-Serono, Astra-Zeneca, Novartis, Eli-Lilly, R-Pharm, Actelion, Sanofi, Janssen, Pfizer and Sandoz.

Mauricio Montero Luna declares having been sponsored for consultancy, academic events and conferences organised by Novartis, Bristol-Myers-Squibb, Roche, Pfizer, UCB, Abbvie, Janssen, Expanscience and MSD.

Fedra Consuelo Irazoque Palazuelos declares having been sponsored by Roche, Novartis, Celgene, Takeda, Pfizer, Lily, Janssen, Expanscience, Sanofi and Abbvie.

Carlos Abud-Mendoza declares having participated as a consultant and speaker for Pfizer BMS, UCB, Lilly and Roche.

Virginia Pascual Ramos declares having been an investigator in projects of Pfizer and with UCB.

Sergio Cerpa Cruz declares having participated as an investigator for Sanofi, Astra Zeneca and Neovacs.

Jorge Enrique Aguilar Arreola declares having participated as a speaker for Roche and Pfizer and having received support to attend congresses of Pfizer, Roche, Lilly, Abbvie and Asofarma.

Alina Hernández-Bedolla declares having received a grant for congresses for Bristol and ABBVIE, and being a conference speaker for Roche.

Luis H. Silveira declares having received a grant from Novartis to attend the two last Mexican Rheumatology congresses.

Alejandra López Rodríguez declares having participated as a speaker for Roche, Eli Lilly, Pfizer, Celgene, BMS and in clinical research with Pfizer, Novartis, Eli Lily, Glaxo; she is also a consultant with: Roche, Novartis, UCB, Eli Lily.

Greta Reyes-Cordero declares a conflict of interest with Pfizer, UCB, Novartis, Bristol-Myers Squibb, Roche.

Humberto Alfredo Ricardez Puente declares a conflict of interest with Bristol-Myers Squibb as a subinvestigator and for attending congresses and/or updates for Abbive, Pfizer, Janssen, Lilly.

María Fernanda Hernández Cabrera declares a conflict of interests with Roche and Abbvie.

Javier Merayo-Chalico was a speaker for Pfizer and was sponsored to attend two academic events.

Daniel Xibillé Friedmann declares having been a principal investigator, consultant and speaker for Abbvie, BMS, Eli-Lilly, Janssen, Pfizer, Roche.

Sandra Muñoz López, Istar Guzmán Sánchez, María Esther Pérez-Bastidas, Marco Ulises Martínez Martínez, David Herrera van Ostdam, Guadalupe Olvera-Soto and Marcela Pérez have no conflict of interest to declare.

Acknowledgements

The Mexican College of Rheumatology would like to acknowledge the valuable contributions and suggestions of Concepción Cazariego from AMEPAR, Dr. Cecilia Guerrero Almeida, Dr. Polita del Rocío Cruz Cruz, Dr. Sara Morales Hernández and Dr. Alejandro E. Macías, who in their role as patients, infectologists and obstetrician-gynaecologists improved the content of our guidelines.

References
[1]
M.H. Cardiel, A. Díaz-Borjón, M. Vázquez del Mercado Espinosa, J.I. Gámez-Nava, L.A. Barile Fabris, C. Pacheco Tena, et al.
Update of the Mexican College of Rheumatology guidelines for the pharmacologic treatment of rheumatoid arthritis.
Reumatol Clin, 10 (2014), pp. 227-240
[2]
C. Mendoza Pinto, M. García Carrasco.
GRADE system, systematic and transparent evaluation.
Reumatol Clin, 14 (2018), pp. 65-67
[3]
G. Guyatt, G. Vist, Y. Falck-Ytter, R. Kunz, N. Magrini, H. Schunemann.
An emerging consensus on grading recommendations?.
ACP J Club, 144 (2006), pp. A8-A9
[4]
G.H. Guyatt, H.J. Schünemann, B. Djulbegovic, E.A. Akl.
Guideline panels should not GRADE good practice statements.
J Clin Epidemiol, 68 (2015), pp. 597-600
[5]
J.S. Smolen, R. Landewé, 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 anti-rheumatic drugs: 2013 update.
Ann Rheum Dis, 73 (2014), pp. 492-509
[6]
J.A. Singh, K.G. Saag, S.L. Bridges, E.A. Akl, R.R. Bannuru, M.C. Sullivan, et al.
2015 American College of Rheumatology guideline for the treatment of rheumatoid arthritis.
Arthritis Care Res (Hoboken), 68 (2016), pp. 1-25
[7]
C. Abud-Mendoza.
Grupo Mexicano de Estudio de Manejo Integral de la Artritis Reumatoide. Consideraciones a las recomendaciones terapéuticas para la artritis reumatoide.
Reumatol Clin, 11 (2015), pp. 193-195
[8]
S.A. Bergstra, C.F. Allaart.
What is the optimal target for treat-to-target strategies in rheumatoid arthritis?.
Curr Opin Rheumatol, 30 (2018), pp. 282-287
[9]
V. Bykerk, P. Akhavan, G.S. Hazlewood, O. Schieir, A. Dooley, B. Haraoui, et al.
Canadian Rheumatology Association recommendations for the pharmacological management of RA.
J Rheumatol, 39 (2012), pp. 1559-1582
[10]
C. Bombardier, G.S. Hazlewood, P. Akhavan, O. Schieir, A. Dooley, B. Haraoui, et al.
Canadian Rheumatology Association recommendations for the pharmacological management of rheumatoid arthritis with traditional and biologic disease-modifying antirheumatic drugs: Part II. Safety.
J Rheumatol, 39 (2012), pp. 1583-1602
[11]
National Institute for Clinical Excellence (NICE).
Clinical guidelines development methods.
NICE, (2012),
[12]
L. Ventura-Ríos, D. Bañuelos-Ramírez, M.C. Hernández-Quiroz, M. Robles-SanRomán, F. Irazoque-Palazuelos, M.V. Goycochea-Robles.
Patient survival and safety with biologic therapy. Results of the Mexican National Registry Biobadamex 1.0.
Reumatol Clin, 8 (2012), pp. 189-194
[13]
M. Bogas, M.J. Leandro.
Biologic therapy and pregnancy. A systematic literature review.
Acta Reumatol Port, 36 (2011), pp. 219-232
[14]
L. Massardo, B.A. Pons-Estel, D. Wojdyla, M.H. Cardiel, C.M. Galarza Maldonado, M.P. Sacnun, et al.
Early rheumatoid arthritis in Latin America. Low socioeconomic status relates to high disease activity at baseline.
Arthritis Care Res (Hoboken), 64 (2012), pp. 1135-1143
[15]
E. Álvarez-Hernández, I. Peláez-Ballestas, A. Boonen, J. Vázquez-Mellado, A. Hernández-Garduño, F.C. Rivera, et al.
Catastrophic health expenses and impoverishment of households of patients with rheumatoid arthritis.
Reumatol Clin, 8 (2012), pp. 168-173
[16]
R. Burgos-Vargas, L.J. Catoggio, C. Galarza-Maldonado, K. Ostojich, M.H. Cardiel.
Current therapies in rheumatoid arthritis: a Latin American perspective.
Reumatol Clin, 9 (2013), pp. 106-112
[17]
M.H. Cardiel.
Treat to target strategy in rheumatoid arthritis: real benefits.
Reumatol Clin, 9 (2013), pp. 101-105
[18]
M.H. Cardiel, Latin American Rheumatology Associations of the Pan-American League of Associations for Rheumatology (PANLAR); Grupo Latinoamericano de Estudio de Artritis Reumatoide (GLADAR).
First Latin American position paper on the pharmacological treatment of rheumatoid arthritis.
Rheumatology (Oxford), 45 (2006), pp. ii7-ii22
[19]
R.B. Mueller, N. Reshiti, T. Kaegi, A. Finckh, S.R. Haile, H. Schulze-Koops, et al.
Does addition of glucocorticoids to the initial therapy influence the later course of the disease in patients with early RA? Results from the Swiss prospective observational registry (SCQM).
Clin Rheumatol, 36 (2017), pp. 59-66
[20]
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
[21]
V. Sadra, A. Khabbazi, S. Kolahi, M. Hajialiloo, M. Ghojazadeh.
Randomized double-blind study of the effect of dexamethasone and methylprednisolone pulse in the control of rheumatoid arthritis flare-up: a preliminary study.
Int J Rheum Dis, 17 (2014), pp. 389-393
[22]
D. De Cock, G. Vanderschueren, S. Meyfroidt, J. Joly, R. Westhovens, P. Verschueren.
Two-year clinical and radiologic follow-up of early RA patients treated with initial step up monotherapy or initial step down therapy with glucocorticoids, followed by a tight control approach: lessons from a cohort study in daily practice.
Clin Rheumatol, 33 (2014), pp. 125-130
[23]
P.H.P. De Jong, R.A. Quax, M. Huisman, A.H. Gerards, R.A. Feelders, P.B. de Sonnaville, et al.
Response to glucocorticoids at 2 weeks predicts the effectiveness of DMARD induction therapy at 3 months: post hoc analyses from the tREACH study.
Ann Rheum Dis, 72 (2013), pp. 1659-1663
[24]
M.L. Hetland, K. Hørslev-Petersen.
The CIMESTRA study: Intra-articular glucocorticosteroids and synthetic DMARDs in a treat-to-target strategy in early rheumatoid arthritis.
Clin Exp Rheumatol, 30 (2012), pp. S44-S49
[25]
J.R. Kirwan, J.W.J. Bijlsma, M. Boers, B.J. Shea.
Effects of glucocorticoids on radiological progression in rheumatoid arthritis.
Cochrane Database Syst Rev, 1 (2007), pp. CD006356
[26]
T. Uhlig, E. Lie, V. Norvang, Å.S. Lexberg, E. Rødevand, F. Krøll, et al.
Achievement of remission and low disease activity definitions in patients with rheumatoid arthritis in clinical practice: results from the NOR-DMARD study.
J Rheumatol, 43 (2016), pp. 716-723
[27]
L.M. Steunebrink, H.E. Vonkeman, P.M. ten Klooster, M. Hoekstra, P.L. van Riel, M.A. van de Laar.
Recently diagnosed rheumatoid arthritis patients benefit from a treat-to-target strategy: results from the DREAM registry.
Clin Rheumatol, 35 (2016), pp. 609-615
[28]
L. Heimans, G. Akdemir, K.V. Boer, Y.P. Goekoop-Ruiterman, E.T. Molenaar, J.H. van Groenendael, et al.
Two-year results of disease activity score (DAS)-remission-steered treatment strategies aiming at drug-free remission in early arthritis patients (the IMPROVED-study).
Arthritis Res Ther, 18 (2016), pp. 23
[29]
P. Verschueren, D. de Cock, L. Corluy, R. Joos, C. Langenaken, V. Taelman, et al.
Methotrexate in combination with other DMARDs is not superior to methotrexate alone for remission induction with moderate-to-high-dose glucocorticoid bridging in early rheumatoid arthritis after 16 weeks of treatment: The CareRA trial.
Ann Rheum Dis, 74 (2015), pp. 27-34
[30]
B. Kuriya, J. Xiong, G. Boire, B. Haraoui, C. Hitchon, J. Pope, et al.
Earlier time to remission predicts sustained clinical remission in early rheumatoid arthritis—results from the Canadian Early Arthritis Cohort (CATCH).
J Rheumatol, 41 (2014), pp. 2161-2166
[31]
B. Darawankul, S. Chaiamnuay, R. Pakchotanon, P. Asavatanabodee, P. Narongroeknawin.
The good EULAR response at the first year is strongly predictive of clinical remission in rheumatoid arthritis: results from the TARAC cohort.
Clin Rheumatol, 34 (2015), pp. 43-49
[32]
D. Golicki, M. Newada, J. Lis, K. Pol, T. Hermanowski, M. Tłustochowicz.
Leflunomide in monotherapy of rheumatoid arthritis: meta-analysis of randomized trials.
Pol Arch Med Wewn, 122 (2012), pp. 22-32
[33]
S.M. Goodman, M.D. Cronstein, V.P. Bykerk.
Outcomes related to methotrexate dose and route of administration in patients with rheumatoid arthritis: a systematic literature review.
Clin Exp Rheumatol, 33 (2015), pp. 272-278
[34]
G.S. Hazlewood, C. Barnabe, G. Tomlinson, D. Marshall, D.J. Devoe, C. Bombardier.
Methotrexate monotherapy and methotrexate combination therapy with traditional and biologic disease modifying anti-rheumatic drugs for rheumatoid arthritis: a network meta-analysis.
Cochrane Database Syst Rev, 8 (2016), pp. CD010227
[35]
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
[36]
J.A. Singh, A. Hossain, A.S. Mudano, E. Tanjong Ghogomu, M.E. Suarez-Almazor, R. Buchbinder, et al.
Biologics or tofacitinib for people with rheumatoid arthritis naive to methotrexate: a systematic review and network meta-analysis.
Cochrane Database Syst Rev, 5 (2017), pp. CD012657
[37]
J.A. Singh, A. Hossain, E. Tanjong Ghogomu, A.S. Mudano, L.J. Maxwell, R. Buchbinder, et al.
Biologics or tofacitinib for people with rheumatoid arthritis unsuccessfully treated with biologics: a systematic review and network meta-analysis.
Cochrane Database Syst Rev, 3 (2017), pp. CD012591
[38]
K.J. Aaltonen, L.M. Virkki, A. Malmivaara, Y.T. Konttinen, D.C. Nordström, M. Blom.
Systematic review and meta-analysis of the efficacy and safety of existing TNF blocking agents in treatment of rheumatoid arthritis.
[39]
P. Baji, M. Péntek, L. Czirják, Z. Szekanecz, G. Nagy, L. Gulácsi, et al.
Efficacy and safety of infliximab-biosimilar compared to other biological drugs in rheumatoid arthritis: a mixed treatment comparison.
Eur J Health Econ, 15 (2014), pp. S53-S64
[40]
P.C. Taylor, E.C. Keystone, D. van der Heijde, M.E. Weinblatt, L.C. Morales, J. Reyes Gonzaga, et al.
Baricitinib versus placebo or adalimumab in rheumatoid arthritis.
N Engl J Med, 376 (2017), pp. 652-662
[41]
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
[42]
M. Schiff, M.E. Weinblatt, R. Valente, D. van der Heijde, G. Citera, A. Elegbe, et al.
Head-to-head comparison of subcutaneous abatacept versus adalimumab for rheumatoid arthritis: two-year efficacy and safety findings from AMPLE trial.
Ann Rheum Dis, 73 (2014), pp. 86-94
[43]
R. Fleischmann, E. Mysler, S. Hall, A.J. Kivitz, R.J. Moots, Z. Luo, et al.
Efficacy and safety of tofacitinib monotherapy, tofacitinib with methotrexate, and adalimumab with methotrexate in patients with rheumatoid arthritis (ORAL Strategy): a phase 3b/4, double-blind, head-to-head, randomised controlled trial.
[44]
J.S. Smolen, G.R. Burmester, B. Combe, J.R. Curtis, S. Hall, B. Haraoui, et al.
Head-to-head comparison of certolizumab pegol versus adalimumab in rheumatoid arthritis: 2-year efficacy and safety results from the randomised EXXELERATE study.
Lancet, 388 (2016), pp. 2763-2774
[45]
E.H. Choy, C. Bernasconi, M. Aassi, J.F. Molina, O.M. Epis.
Treatment of rheumatoid arthritis with anti-tumor necrosis factor or tocilizumab therapy as first biologic agent in a global comparative observational study.
Arthritis Care Res (Hoboken), 69 (2017), pp. 1484-1494
[46]
P. Hamann, R. Holland, K. Hyrich, J.D. Pauling, G. Shaddick, A. Nightingale, et al.
Factors associated with sustained remission in rheumatoid arthritis in patients treated with anti-tumor necrosis factor.
Arthritis Care Res (Hoboken), 69 (2017), pp. 783-793
[47]
J.O. Costa, L.L. Lemos, M.A. Machado, A.M. Almeida, A.M. Kakehasi, V.E. Araújo, et al.
Infliximab, methotrexate and their combination for the treatment of rheumatoid arthritis: a systematic review and meta-analysis.
Rev Bras Reumatol, 55 (2015), pp. 146-158
[48]
M.A. Machado, A.A. Maciel, L.L. de Lemos, J.O. Costa, A.M. Kakehasi, E.I. Andrade, et al.
Adalimumab in rheumatoid arthritis treatment: a systematic review and meta-analysis of randomized clinical trials.
Rev Bras Reumatol, 53 (2013), pp. 419-430
[49]
M. Chen, D. Peng, Z. Zhang, G. Zuo, G. Zhao.
Efficacy of etanercept for treating the active rheumatoid arthritis: an updated meta-analysis.
Int J Rheum Dis, 19 (2016), pp. 1132-1142
[50]
P. Emery, J.E. Pope, K. Kruger, R. Lippe, R. DeMasi, S. Lula, et al.
Efficacy of monotherapy with biologics and JAK inhibitors for the treatment of rheumatoid arthritis: a systematic review.
Adv Ther, 35 (2018), pp. 1535-1563
[51]
A. Migliore, E. Bizzi, C.G. Egan, M. Bernardi, L. Petrella.
Efficacy of biological agents administered as monotherapy in rheumatoid arthritis: a Bayesian mixed-treatment comparison analysis.
Ther Clin Risk Manag, 11 (2015), pp. 1325-1335
[52]
D. Xibille, S. Carrillo, G. Huerta-Sil, R. Hernández, L. Limón, G. Olvera-Soto, et al.
Current state of biosimilars in Mexico: the position of the Mexican College of Rheumatology, 2016.
Reumatol Clin, 14 (2018), pp. 127-136
[53]
S. Minozzi, S. Bonovas, T. Lytras, V. Pecoraro, M. González-Lorenzo, A.J. Bastiampillai, et al.
Risk of infections using anti-TNF agents in rheumatoid arthritis, psoriatic arthritis, and ankylosing spondylitis: a systematic review and meta-analysis.
Expert Opin Drug Saf, 15 (2016), pp. 11-34
[54]
S. Ramiro, A. Sepriano, K. Chatzidionysiou, J.L. Nam, J.S. Smolen, D. van der Heijde, et al.
Safety of synthetic and biological DMARDs: a systematic literature review informing the 2016 update of the EULAR recommendations for management of rheumatoid arthritis.
Ann Rheum Dis, 76 (2017), pp. 1101-1136
[55]
T.L. Michaud, Y.H. Rho, T. Shamliyan, K.M. Kuntz, H.K. Choi.
The comparative safety of tumor necrosis factor inhibitors in rheumatoid arthritis: a meta-analysis update of 44 trials.
Am J Med, 127 (2014), pp. 1208-1232
[56]
R. Sakai, Y. Komano, M. Tanaka, T. Nanki, R. Koike, H. Nagasawa, et al.
Time-dependent increased risk for serious infection from continuous use of tumor necrosis factor antagonists over three years in patients with rheumatoid arthritis.
Arthritis Care Res (Hoboken), 64 (2012), pp. 1125-1134
[57]
C.L. Morgan, P. Emery, D. Porter, A. Reynolds, A. Young, H. Boyd, et al.
Treatment of rheumatoid arthritis with etanercept with reference to disease-modifying anti-rheumatic drugs: long-term safety and survival using prospective, observational data.
Rheumatology (Oxford), 53 (2014), pp. 186-194
[58]
K.J. Aaltonen, J.T. Joensuu, L. Virkki, T. Sokka, P. Aronen, H. Relas, et al.
Rates of serious infections and malignancies among patients with rheumatoid arthritis receiving either tumor necrosis factor inhibitor or rituximab therapy.
J Rheumatol, 42 (2015), pp. 372-378
[59]
K.W. Jo, Y. Hong, Y.J. Jung, B. Yoo, C.K. Lee, Y.G. Kim, et al.
Incidence of tuberculosis among anti-tumor necrosis factor users in patients with a previous history of tuberculosis.
Respir Med, 107 (2013), pp. 1797-1802
[60]
Y.J. Kim, Y.G. Kim, T.S. Shim, B.S. Koo, S. Hong, C.K. Lee, et al.
Safety of resuming tumour necrosis factor inhibitors in patients who developed tuberculosis as a complication of previous TNF inhibitors.
Rheumatology (Oxford), 53 (2014), pp. 1477-1481
[61]
G.S. Hazlewood, D. Naimark, M. Gardam, V. Bykerk, C. Bombardier.
Prophylaxis for latent tuberculosis infection prior to anti-tumor necrosis factor therapy in low-risk elderly patients with rheumatoid arthritis: a decision analysis.
Arthritis Care Res (Hoboken), 65 (2013), pp. 1722-1731
[62]
L. Carmona, J.J. Gómez-Reino, V. Rodríguez-Valverde, D. Montero, E. Pascual-Gómez, E.M. Mola, et al.
Effectiveness of recommendations to prevent reactivation of latent tuberculosis infection in patients treated with tumor necrosis factor antagonists.
Arthritis Rheum, 52 (2005), pp. 1766-1772
[63]
D. Menzies, M. Adjobimey, R. Ruslami, A. Trajman, O. Sow, H. Kim, et al.
Four months of rifampin or nine months of isoniazid for latent tuberculosis in adults.
N Engl J Med, 379 (2018), pp. 440-453
[64]
R.M. Nanau, M.G. Neuman.
Safety of anti-tumor necrosis factor therapies in arthritis patients.
J Pharm Pharm Sci, 17 (2014), pp. 324-361
[65]
F. Bartoli, C. Bruni, L. Cometi, J. Blagojevic, G. Fiori, L. Tofani, et al.
Premedication prevents infusion reactions and improves retention rate during infliximab treatment.
Clin Rheumatol, 35 (2016), pp. 2841-2845
[66]
S. Bonovas, S. Minozzi, T. Lytras, M. González-Lorenzo, V. Pecoraro, S. Colombo, et al.
Risk of malignancies using anti-TNF agents in rheumatoid arthritis, psoriatic arthritis, and ankylosing spondylitis: a systematic review and meta-analysis.
Expert Opin Drug Saf, 15 (2016), pp. 35-54
[67]
Y. Liu, W. Fan, H. Chen, M. Yu.
Risk of breast cancer and total malignancies in rheumatoid arthritis patients undergoing TNF-α antagonist therapy: a meta-analysis of randomized control trials.
Asian Pac J Cancer Prev, 15 (2014), pp. 3403-3410
[68]
G. Moulis, A. Sommet, J. Béné, F. Montastruc, L. Sailler, J.L. Montastruc, et al.
Cancer risk of anti-TNF-α at recommended doses in adult rheumatoid arthritis: a meta-analysis with intention to treat and per protocol analyses.
[69]
L.K. Mercer, M. Lunt, A.L.S. Low, W.G. Dixon, K.D. Watson, D.P. Symmons, et al.
Risk of solid cancer in patients exposed to anti-tumour necrosis factor therapy: results from the British Society for Rheumatology Biologics Register for Rheumatoid Arthritis.
Ann Rheum Dis, 74 (2015), pp. 1087-1093
[70]
A.W. Friedman, G.R. Burmester, R. Landewé, M.C. Genovese, N.A. Varothai, A.P. Lacerda, et al.
Adalimumab long-term safety: infections, vaccination response and pregnancy outcomes in patients with rheumatoid arthritis.
Ann Rheum Dis, 76 (2016), pp. 414-417
[71]
C. Chambers, A.B. Kimball, A. Afzali, M. Cooney, J.J. Cush, M.E.B. Clowse, et al.
Pregnancy outcomes after exposure to certolizumab pegol.
Arthritis Rheumatol, 70 (2018), pp. 1399-1407
[72]
Y. Kawai, T. Tsuchiya, S. Aoki.
Pregnancy outcomes of patients exposed to adalimumab in Japan.
Dig Dis, 37 (2019), pp. 123-130
[73]
R.F. Van Vollenhoven, R.M. Fleischmann, D.E. Furst, S. Lacey, P.B. Lehane.
Longterm safety of rituximab: final report of the rheumatoid arthritis global clinical trial program over 11 years.
J Rheumatol, 42 (2015), pp. 1761-1766
[74]
M.H. Buch, J.S. Smolen, N. Betteridge, Rituximab Consensus Expert Committee.
Updated consensus statement on the use of rituximab in patients with rheumatoid arthritis.
Ann Rheum Dis, 70 (2011), pp. 909-920
[75]
Y. Oshima, H. Tsukamoto, A. Tojo.
Association of hepatitis B with antirheumatic drugs: a case–control study.
Mod Rheumatol, 23 (2013), pp. 694-704
[76]
M. Padovan, M. Filippini, A. Tincani, E. Lanciano, E. Bruschi, O. Epis, et al.
Safety of abatacept in rheumatoid arthritis with serologic evidence of past or present hepatitis B virus infection.
Arthritis Care Res (Hoboken), 68 (2016), pp. 738-743
[77]
C. Lahaye, M. Soubrier, A. Mulliez, T. Bardin, A. Cantagrel, B. Combe, et al.
Effectiveness and safety of abatacept in elderly patients with rheumatoid arthritis enrolled in the French Society of Rheumatology's ORA registry.
Rheumatology (Oxford), 55 (2016), pp. 874-882
[78]
M.C. Genovese, C. Pacheco-Tena, A. Covarrubias, G. Leon, E. Mysler, M. Keiserman, et al.
Longterm safety and efficacy of subcutaneous abatacept in patients with rheumatoid arthritis: 5-year results from a phase IIIb trial.
J Rheumatol, 45 (2018), pp. 1085-1092
[79]
V. Strand, A. Balsa, J. al-Saleh, L. Barile-Fabris, T. Horiuchi, T. Takeuchi.
Immunogenicity of biologics in chronic inflammatory diseases: a systematic review.
BioDrugs, 31 (2017), pp. 299-316
[80]
R. Westhovens, J.M. Kremer, P. Emery, A.S. Russell, R. Alten, E. Barré, et al.
Long-term safety and efficacy of abatacept in patients with rheumatoid arthritis and an inadequate response to methotrexate: a 7-year extended study.
Clin Exp Rheumatol, 32 (2014), pp. 553-562
[81]
A. Mera-Varela, E. Perez-Pampin.
Abatacept therapy in rheumatoid arthritis with interstitial lung disease.
J Clin Rheumatol, 20 (2014), pp. 445-446
[82]
L.J. Scott.
Tocilizumab: a review in rheumatoid arthritis.
Drugs, 77 (2017), pp. 1865-1879
[83]
E. Choy, R. Caporali, R. Xavier, B. Fautrel, R. Sanmarti, M. Bao, et al.
Subcutaneous tocilizumab in rheumatoid arthritis: findings from the common-framework phase 4 study programme TOZURA conducted in 22 countries.
Rheumatology, 57 (2018), pp. 499-507
[84]
F. Montoya, J.L.C. Moreno, G.C. Casado, G. Citera, B.J. Velasco Zamora, A. Quinteros, et al.
Estudio multicéntrico no intervencional en pacientes con artritis reumatoidea tratados con tocilizumab.
Rev Argent Reumatolg, 28 (2017), pp. 23-28
[85]
M.C. Genovese, J.M. Kremer, R.F. Vollenhoven, R. van Alten, J.J. Scali, A. Kelman, et al.
Transaminase levels and hepatic events during tocilizumab treatment: Pooled analysis of long-term clinical trial safety data in rheumatoid arthritis.
Arthritis Rheumatol, 69 (2017), pp. 1751-1761
[86]
S. Kunwar, C.E. Collins, F. Constantinescu.
Baricitinib, a Janus kinase inhibitor, in the treatment of rheumatoid arthritis: a systematic literature review and meta-analysis of randomized controlled trials.
Clin Rheumatol, 37 (2018), pp. 2611-2620
[87]
A. Berhan.
Efficacy, safety and tolerability of tofacitinib in patients with an inadequate response to disease modifying anti-rheumatic drugs: a meta-analysis of randomized double-blind controlled studies.
BMC Musculoskelet Disord, 14 (2013), pp. 332
[88]
Y. He, A.Y.S. Wong, E.W. Chan, W.C.Y. Lau, K.K.C. Man, C.S.L. Chui, et al.
Efficacy and safety of tofacitinib in the treatment of rheumatoid arthritis: a systematic review and meta-analysis.
BMC Musculoskelet Disord, 14 (2013), pp. 298
[89]
O.M. Castañeda, F.J. Romero, A. Salinas, G. Citera, E. Mysler, O. Rillo, et al.
Safety of tofacitinib in the treatment of rheumatoid arthritis in Latin America compared with the rest of the world population.
J Clin Rheumatol, 23 (2017), pp. 193-199
[90]
S.B. Cohen, Y. Tanaka, X. Mariette, J.R. Curtis, E.B. Lee, P. Nash, et al.
Long-term safety of tofacitinib for the treatment of rheumatoid arthritis up to 8.5years: Integrated analysis of data from the global clinical trials.
Ann Rheum Dis, 76 (2017), pp. 1253-1262
[91]
G.G. Song, S.-C. Bae, Y.H. Lee.
Efficacy and safety of tofacitinib for active rheumatoid arthritis with an inadequate response to methotrexate or disease-modifying antirheumatic drugs: a meta-analysis of randomized controlled trials.
Korean J Intern Med, 29 (2014), pp. 656-663
[92]
J.D. Isaacs, A. Zuckerman, S. Krishnaswami, C. Nduaka, S. Lan, M.M. Hutmacher, et al.
Changes in serum creatinine in patients with active rheumatoid arthritis treated with tofacitinib: results from clinical trials.
Arthritis Res Ther, 16 (2014), pp. R158
[93]
J.M. Kremer, A.J. Kivitz, J.A. Simon-Campos, E.L. Nasonov, H.P. Tony, S.K. Lee, et al.
Evaluation of the effect of tofacitinib on measured glomerular filtration rate in patients with active rheumatoid arthritis: results from a randomised controlled trial.
Arthritis Res Ther, 17 (2015), pp. 95
[94]
A. Souto, E. Salgado, J.R. Maneiro, A. Mera, L. Carmona, J.J. Gómez-Reino.
Lipid profile changes in patients with chronic inflammatory arthritis treated with biologic agents and tofacitinib in randomized clinical trials: a systematic review and meta-analysis.
Arthritis Rheumatol, 67 (2015), pp. 117-127
[95]
C. Charles-Schoeman, P. Wicker, M.A. Gonzalez-Gay, M. Boy, A. Zuckerman, K. Soma, et al.
Cardiovascular safety findings in patients with rheumatoid arthritis treated with tofacitinib, an oral Janus kinase inhibitor.
Semin Arthritis Rheum, 46 (2016), pp. 261-271
[96]
Comisión Federal para la Protección contra Riesgos Sanitarios. Available from: https://www.gob.mx/cms/uploads/attachment/file/326787/Alop_ticos_2017.pdf [accessed 25.03.19].
[97]
Drug Administration. Available from: https://www.accessdata.fda.gov/drugsatfda_docs/nda/2018/207924Orig1s000Approv.pdf [accessed 25.03.19].
[98]
C. Charles-Schoeman, G. Burmester, P. Nash, C.A.F. Zerbini, K. Soma, K. Kwok, et al.
Efficacy and safety of tofacitinib following inadequate response to conventional synthetic or biological disease-modifying antirheumatic drugs.
Ann Rheum Dis, 75 (2016), pp. 1293-1301
[99]
J.R. Curtis, E.B. Lee, G. Martin, X. Mariette, K.K. Terry, Y. Chen, et al.
Analysis of non-melanoma skin cancer across the tofacitinib rheumatoid arthritis clinical programme.
Clin Exp Rheumatol, 35 (2017), pp. 614-622
[100]
T.M. Kuijper, J.J. Luime, P.H. de Jong, A.H. Gerards, D. van Zeben, I. Tchetverikov, et al.
Tapering conventional synthetic DMARDs in patients with early arthritis in sustained remission: 2-year follow-up of the tREACH trial.
Ann Rheum Dis, 75 (2016), pp. 2119-2123
[101]
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
[102]
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
[103]
B. Fautrel, T. Pham, T. Alfaiate, F. Gandjbakhch, V. Foltz, J. Morel, et al.
Step-down strategy of spacing TNF-blocker injections for established rheumatoid arthritis in remission: results of the multicentre non-inferiority randomised open-label controlled trial (STRASS: spacing of TNF-blocker injections in Rheumatoid ArthritiS Study).
Ann Rheum Dis, 75 (2016), pp. 59-67
[104]
J. Haschka, M. Englbrecht, A.J. Hueber, B. Manger, A. Kleyer, M. Reiser, et al.
Relapse rates in patients with rheumatoid arthritis in stable remission tapering or stopping antirheumatic therapy: interim results from the prospective randomised controlled RETRO study.
Ann Rheum Dis, 75 (2016), pp. 45-51
[105]
Y. El Miedany, M. el Gaafary, S. Youssef, I. Ahmed, S. Bahlas, M. Hegazi, et al.
Optimizing therapy in inflammatory arthritis: prediction of relapse after tapering or stopping treatment for rheumatoid arthritis patients achieving clinical and radiological remission.
Clin Rheumatol, 35 (2016), pp. 2915-2923
[106]
G. Schett, P. Emery, Y. Tanaka, G. Burmester, D.S. Pisetsky, E. Naredo, et al.
Tapering biologic and conventional DMARD therapy in rheumatoid arthritis: current evidence and future directions.
Ann Rheum Dis, 75 (2016), pp. 1428-1437
[107]
S. Hirata, K. Saito, S. Kubo, S. Fukuyo, Y. Mizuno, S. Iwata, et al.
Discontinuation of adalimumab after attaining disease activity score 28-erythrocyte sedimentation rate remission in patients with rheumatoid arthritis (HONOR study): an observational study.
Arthritis Res Ther, 15 (2013), pp. R135
[108]
D. Liu, N. Yuan, G. Yu, G. Song, Y. Chen.
Can rheumatoid arthritis ever cease to exist: a review of various therapeutic modalities to maintain drug-free remission?.
Am J Transl Res, 9 (2017), pp. 3758-3775
[109]
G. Bosch, R.N. Garcia Salinas, M.V. Martire, P. Arturi, S.J. Magri, D. Giacomone, et al.
Optimizing biologic therapy in rheumatology: frequency and characteristics from an Argentine referral centre.
Int J Clin Rheumatol, 11 (2016), pp. 108-112
[110]
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
[111]
J.R. Maneiro, E. Perez-Pampin, E. Salgado, L. Carmona, J.J. Gomez-Reino.
Observational study of optimization of biologic therapies in rheumatoid arthritis: a single-centre experience.
Rheumatol Int, 34 (2014), pp. 1059-1063
[112]
J.S. Smolen, R. Landewé, J. Bijlsma, G. Burmester, K. Chatzidionysiou, M. Dougados, et al.
EULAR recommendations for the management of rheumatoid arthritis with synthetic and biological disease-modifying antirheumatic drugs: 2016 update.
Ann Rheum Dis, 76 (2017), pp. 960-977
[113]
B. Fautrel, A.A. den Broeder.
De-intensifying treatment in established rheumatoid arthritis (RA): why, how, when and in whom can DMARDs be tapered?.
Best Pract Res Clin Rheumatol, 29 (2015), pp. 550-565
[114]
I. González-Álvaro, C. Martínez-Fernández, B. Dorantes-Calderón, R. García-Vicuña, B. Hernández-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), 54 (2015), pp. 1200-1209
[115]
R.F. Van Vollenhoven, M. Østergaard, M. Leirisalo-Repo, T. Uhlig, M. Jansson, E. Larsson, et al.
Full dose, reduced dose or discontinuation of etanercept in rheumatoid arthritis.
Ann Rheum Dis, 75 (2016), pp. 52-58
[116]
Y. Tanaka, S. Hirata.
Intensive intervention can lead to a treatment holiday from biological DMARDs in patients with rheumatoid arthritis.
Drugs, 74 (2014), pp. 2129-2139
[117]
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
[118]
S.M. Van der Kooij, Y.P. Goekoop-Ruiterman, J.K. de Vries-Bouwstra, M. Güler-Yüksel, 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
[119]
N.B. Klarenbeek, S.M. van der Kooij, M. Güler-Yüksel, J.H. van Groenendael, K.H. Han, P.J. Kerstens, et al.
Discontinuing treatment in patients with rheumatoid arthritis in sustained clinical remission: exploratory analyses from the BeSt study.
Ann Rheum Dis, 70 (2011), pp. 315-319
[120]
D.W. Baker, T. Brown, J.Y. Lee, A. Ozanich, D.T. Liss, D.S. Sandler, et al.
A multifaceted intervention to improve influenza pneumococcal, and herpes zoster vaccination among patients with rheumatoid arthritis.
J Rheumatol, 43 (2016), pp. 1030-1037
[121]
M. Sester, F. van Leth, J. Bruchfeld, D. Bumbacea, D.M. Cirillo, A.G. Dilektasli, et al.
Risk assessment of tuberculosis in immunocompromised patients. A TBNET study.
Am J Respir Crit Care Med, 190 (2014), pp. 1168-1176
[122]
S.E. McMullen, D.A. Pegues, F.S. Shofer, A.C. Sheller, E.B. Wiener.
Performance of QuantiFERON-TB Gold and tuberculin skin test relative to subjects’ risk of exposure to tuberculosis.
Clin Infect Dis, 58 (2014), pp. 1260-1266
[123]
L.C. Kahwati, C. Feltner, M. Halpern, C.L. Woodell, E. Boland, H.R. Amick.
Primary care screening and treatment for latent tuberculosis infection in adults: evidence report and systematic review for the US Preventive Services Task Force.
JAMA, 316 (2016), pp. 970-983
[124]
T. Beauchamp, R. Childress, F. James.
Principles of biomedical ethics.
6th ed., Oxford University Press, (2009),
[125]
M. Rom, J. Fins, R. Mackenzie.
Articulating a justice ethic for rheumatology: a critical analysis of disparities in rheumatic Diseases.
Arthritis Rheum, 57 (2007), pp. 1343-1345
[126]
J.M. Jordan, R. Lawrence, R. Kington, P. Fraser, E. Karlson, K. Lorig, et al.
Ethnic health disparities in arthritis and musculoskeletal diseases. Report of a scientific conference.
Arthritis Rheum, 46 (2002), pp. 2280-2286
[127]
J.M. Jordan, R. Baker, O. Ferraz, S. Hawkes, C. Krubiner, P. Littlejohns, et al.
Universal health coverage, priority setting, and the human right to health.
Lancet, 390 (2017), pp. 712-714
[128]
K. Xu, D.B. Evans, K. Kawabata, R. Zeramdini, J. Klavus, C.J. Murray.
Household catastrophic health expenditure: a multicountry analysis.
[129]
S.C. Hyoun, S.G. Običan, A.R. Scialli.
Teratogen update: methotrexate.
Birth Defects Res A Clin Mol Teratol, 94 (2012), pp. 187-207
[130]
G.S. Ngian, A.M. Briggs, I.N. Ackerman, S. van Doornum.
Safety of anti-rheumatic drugs for rheumatoid arthritis in pregnancy and lactation.
Int J Rheum Dis, 19 (2016), pp. 834-843
[131]
B.L. Bermas.
Non-steroidal anti-inflammatory drugs, glucocorticoids and disease modifying anti-rheumatic drugs for the management of rheumatoid arthritis before and during pregnancy.
Curr Opin Rheumatol, 26 (2014), pp. 334-340
[132]
C. Gotestam Skorpen, M. Hoeltzenbein, A. Tincani, R. Fischer-Betz, E. Elefant, C. Chambers, et al.
The EULAR points to consider for use of antirheumatic drugs before pregnancy, and during pregnancy and lactation.
Ann Rheum Dis, 75 (2016), pp. 795-810
[133]
H. Ince-Askan, R.J. Dolhain.
Pregnancy and rheumatoid arthritis.
Best Pract Res Clin Rheumatol, 29 (2015), pp. 580-596
[134]
M. Gerosa, T. Schioppo, P.L. Meroni.
Challenges and treatment options for rheumatoid arthritis during pregnancy.
Expert Opin Pharmacother, 17 (2016), pp. 1539-1547
[135]
A. Bérard, J.P. Zhao, I. Shui, S. Colilla.
Leflunomide use during pregnancy and the risk of adverse pregnancy outcomes.
Ann Rheum Dis, 77 (2018), pp. 500-509
[136]
F. Förger, P.M. Villiger.
Treatment of rheumatoid arthritis during pregnancy: present and future.
Expert Rev Clin Immunol, 12 (2016), pp. 937-944
[137]
E.F. Chakravarty, E.R. Murray, A. Kelman, P. Farmer.
Pregnancy outcomes after maternal exposure to rituximab.
Blood, 117 (2011), pp. 1499-1506
[138]
M. Hoeltzenbein, E. Beck, R. Rajwanshi, C. Gøtestam Skorpen, E. Berber, C. Schaefer, et al.
Tocilizumab use in pregnancy: analysis of a global safety database including data from clinical trials and post-marketing data.
Semin Arthritis Rheum, 46 (2016), pp. 238-245
[139]
K. Kaneko, M. Sugitani, M. Goto, A. Murashima.
Tocilizumab and pregnancy: four cases of pregnancy in young women with rheumatoid arthritis refractory to anti-TNF biologics with exposure to tocilizumab.
Mod Rheumatol, 26 (2016), pp. 672-675
[140]
M. Kumar, L. Ray, S. Vemuri, T.A. Simon.
Pregnancy outcomes following exposure to abatacept during pregnancy.
Semin Arthritis Rheum, 45 (2015), pp. 351-356
[141]
M. Ojeda-Uribe, N. Afif, E. Dahan, L. Sparsa, C. Haby, J. Sibilia, et al.
Exposure to abatacept or rituximab in the first trimester of pregnancy in three women with autoimmune diseases.
Clin Rheumatol, 32 (2013), pp. 695-700
[142]
S. Kalari, F. Granath, C.Y. Guo, D. Harrison, G. Broms, A. Geldhof, et al.
FRI0274 pregnancy outcomes in women with rheumatologic conditions exposed to infliximab.
Ann Rheum Dis, 73 (2014), pp. 482-483
[143]
O.H. Nielsen, E.V. Loftus Jr., T. Jess.
Safety of TNF-alpha inhibitors during IBD pregnancy: a systematic review.
[144]
C. Weber-Schoendorfer, M. Oppermann, E. Wacker, N. Bernard, network of French pharmacovigilance centres.
Pregnancy outcome after TNF-α inhibitor therapy during the first trimester: a prospective multicentre cohort study.
Br J Clin Pharmacol, 80 (2015), pp. 727-739
[145]
M.E. Clowse, D.C. Wolf, F. Förger, J. Cush, A. Golembesky, L. Shaughnessy, et al.
Pregnancy outcomes in subjects exposed to certolizumab pegol.
J Rheumatol, 42 (2015), pp. 2270-2278
[146]
F. Förger, A. Zbinden, P.M. Villiger.
Certolizumab treatment during late pregnancy in patients with rheumatic diseases: low drug levels in cord blood but possible risk for maternal infections. A case series of 13 patients.
Joint Bone Spine, 83 (2016), pp. 341-343
[147]
R. Benediktsson, A.A. Calder, C.R. Edwards, J.R. Seckl.
Placental 11 beta-hydroxysteroid dehydrogenase: a key regulator of fetal glucocorticoid exposure.
Clin Endocrinol (Oxf), 46 (1997), pp. 161-166
[148]
M.A. Saavedra Salinas, A. Barrera Cruz, A.R. Cabral Castañeda, L.J. Jara Quezada, C.A. Arce-Salinas, J. Álvarez Nemegyei, et al.
Clinical practice guidelines for the management of pregnancy in women with autoimmune rheumatic diseases of the Mexican College of Rheumatology. Part II.
Reumatol Clin, 11 (2015), pp. 305-315
[149]
S. Levi, M. Liberman, A.J. Levi, I. Bjarnason.
Reversible congenital neutropenia associated with maternal sulphasalazine therapy.
Eur J Pediatr, 148 (1988), pp. 174-175
[150]
N. Costedoat-Chalumeau, Z. Amoura, G. Aymard, T.H. Le, B. Wechsler, D. Vauthier, et al.
Evidence of transplacental passage of hydroxychloroquine in humans.
Arthritis Rheum, 46 (2002), pp. 1123-1124
[151]
A.M. Bay Bjørn, V. Ehrenstein, H.H. Hundborg, E.A. Nohr, H.T. Sørensen, M. Nørgaard.
Use of corticosteroids in early pregnancy is not associated with risk of oral clefts and other congenital malformations in offspring.
Am J Ther, 21 (2014), pp. 73-80
[152]
A. Kubota, T. Nakamura, Y. Miyazaki, M. Sekiguchi, T. Suguro.
Perioperative complications in elective surgery in patients with rheumatoid arthritis treated with biologics.
Mod Rheumatol, 22 (2012), pp. 844-848
[153]
X. Mariette, F. Förger, B. Abraham, A.D. Flynn, A. Moltó, R.M. Flipo, et al.
Lack of placental transfer of certolizumab pegol during pregnancy: results from CRIB, a prospective, postmarketing, pharmacokinetic study.
Ann Rheum Dis, 77 (2018), pp. 228-233
[154]
M.E. Clowse, F. Förger, C. Hwang, J. Thorp, R.J. Dolhain, A. van Tubergen, et al.
Minimal to no transfer of certolizumab pegol into breast milk: results from CRADLE, a prospective, postmarketing, multicentre, pharmacokinetic study.
Ann Rheum Dis, 76 (2017), pp. 1890-1896
[155]
J. Brouwer, J.M. Hazes, J.S. Laven, R.J. Dolhain.
Fertility in women with rheumatoid arthritis: influence of disease activity and medication.
Ann Rheum Dis, 74 (2015), pp. 1836-1841
[156]
J. Brouwer, R. Fleurbaaij, J.M.W. Hazes, R.J.E.M. Dolhain, J.S.E. Laven.
Subfertility in women with rheumatoid arthritis and the outcome of fertility assessments.
Arthritis Care Res (Hoboken), 69 (2017), pp. 1142-1149
[157]
M. Provost, J.L. Eaton, M.E. Clowse.
Fertility and infertility in rheumatoid arthritis.
Curr Opin Rheumatol, 26 (2014), pp. 308-314
[158]
A.J. Kivitz, J. Schechtman, M. Texter, A. Fichtner, M. de Longueville, E.K. Chartash.
Vaccine responses in patients with rheumatoid arthritis treated with certolizumab pegol: results from a single-blind randomized phase IV trial.
J Rheumatol, 41 (2014), pp. 648-657
[159]
K.L. Dooling, A. Guo, M. Patel, G.M. Lee, K. Moore, E.A. Belongia, et al.
Recommendations of the advisory committee on immunization practices for use of herpes zoster vaccines.
MMWR Morb Mortal Wkly Rep, 67 (2018), pp. 103-108
[160]
Centers for Disease Control, Prevention (CDC).
Use of 13-valent pneumococcal conjugate vaccine and 23-valent pneumococcal polysaccharide vaccine for adults with immunocompromising conditions: recommendations of the Advisory Committee on Immunization Practices (ACIP).
MMWR Morb Mortal Wkly Rep, 61 (2012), pp. 816-819
[161]
J. Zhang, E. Delzell, F. Xie, J.W. Baddley, C. Spettell, R.M. McMahan, et al.
The use, safety, and effectiveness of herpes zoster vaccination in individuals with inflammatory and autoimmune diseases: a longitudinal observational study.
Arthritis Res Ther, 13 (2011), pp. R174
[162]
J. Zhang, F. Xie, E. Delzell, L. Chen, K.L. Winthrop, J.D. Lewis, et al.
Association between vaccination for herpes zoster and risk of herpes zoster infection among older patients with selected immune-mediated diseases.
[163]
M.J. Burton, J.R. Curtis, S. Yang, L. Chen, J.A. Singh, T.R. Mikuls, et al.
Safety of biologic and nonbiologic disease-modifying antirheumatic drug therapy in veterans with rheumatoid arthritis and hepatitis B virus infection: a retrospective cohort study.
Arthritis Res Ther, 17 (2015), pp. 136

Please cite this article as: Cardiel MH, Carrillo S, Pérez M, Andrade L, Pacheco Tena C, Silveira LH, et al. Actualización de las guías del tratamiento farmacológico de la artritis reumatoide del Colegio Mexicano de Reumatología 2018. Reumatol Clin. 2021;17:215–228.

The constant therapeutic advances in rheumatoid arthritis (RA) make it necessary to periodically review treatment guidelines in order to offer the treating physician updated, evidence-based recommendations adapted to real clinical practice.

Copyright © 2019. Elsevier España, S.L.U. and Sociedad Española de Reumatología y Colegio Mexicano de Reumatología
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?