Allergische reacties als complicatie bij tsDMARD’s
Uitgangsvraag
Welke allergische reacties kunnen als complicatie optreden op als gevolg van een behandeling met tsDMARD’s? Hoe kunnen deze voorkomen worden?
Aanbeveling
Er worden in de beschikbare literatuur geen allergische reacties beschreven bij het gebruik van JAK-remmers.
Overwegingen
In de inleiding van de huidige module is beschreven dat de richtlijn zich focust op JAK-remmers aangezien de overige tsDMARD’s (anno 2024 apremilast) in beperkte mate wordt gebruikt binnen de Nederlandse praktijk.
In de EULAR 2019 review naar de veiligheid van DMARD’s (Sepriano, 2020) en in de bijbehorende aanbevelingen voor de behandeling van RA (Smolen, 2020) werden voor het eerst resultaten meegenomen uit studies naar de veiligheid van JAK-remmers in vergelijking met bDMARD’s en csDMARD’s. Er zijn echter geen resultaten beschreven voor allergische reacties.
Praktijkervaring
Voorschrijfgedrag wordt naast richtlijnen veelal bepaald door het lokale beleid en eigen ervaringen met het voorgeschreven middel. Qua richtlijnen volgen we de (inter)nationale richtlijnen. In de EULAR 2022 update voor de behandeling van RA (Smolen, 2023) is een waarschuwing toegevoegd aan alle aanbevelingen m.b.t. het gebruik van JAK-remmers,: “De volgende risicofactoren voor cardiovasculaire events en maligniteiten moeten in acht genomen worden wanneer overwogen wordt om een JAK-remmer voor te schrijven: leeftijd > 65 jaar, roken (actief of in het verleden), andere cardiovasculaire risicofactoren (zoals diabetes, zwaarlijvigheid en hypertensie), andere risicofactoren voor kanker (maligniteiten, nog aanwezig of in de voorgeschiedenis, met uitzondering van succesvol behandelde niet-melanoom huidkanker), risicofactoren voor veneuze trombo-embolie (voorgeschiedenis van hartinfarct of hartfalen, kanker, erfelijke bloedstollingsstoornis, voorgeschiedenis van bloedstolsels, hormonale anticonceptie en hormoonsubstitutietherapie, het moeten ondergaan van een grote operatieve ingreep, of immobiliteit).” Dit komt overeen met de PRAC-veiligheidswaarschuwing, die begin 2023, is verschenen, maar die in dit geval wel leidend is.
In de dagelijks praktijk start men over het algemeen met een csDMARD en bij een inadequate response en/of bijwerkingen wordt een bDMARD of JAK-remmer (of andere tsDMARD) geïnitieerd (bij ~30% van onze patiënten). De eerste keus is tot op heden veelal een TNF-remmer en pas bij het falen op (meerdere) TNF-remmers wordt een andere mode-of-action gestart, wat dus een non-TNF bDMARD of JAK-remmer kan zijn. De volgorde is veelal afhankelijk van het lokale beleid, en indirect dus de kosten, en ervaringen van de voorschrijver waarbij natuurlijk ook de wensen van de patiënten in de overwegingen worden meegenomen.
Waarden en voorkeuren van patiënten (en evt. hun verzorgers)
Bij de keuze van een behandeling dient er natuurlijk rekening te worden gehouden met de waarden en voorkeuren van patiënt. De voor- en nadelen (o.a. risico op complicaties en bijwerkingen) van de behandeling dienen uitgebreid besproken te worden, waarbij de behandelaar natuurlijk ook rekening houdt met het lokale beleid en de huidige aanbevelingen, o.a. de PRAC-veiligheidswaarschuwing m.b.t. het veilig gebruik van JAK-remmers. Redenen voor het kiezen voor een bepaald medicijn zijn o.a. effectiviteit, bijwerkingenprofiel, toedieningsvorm (bv. oraal vs. subcutaan), comorbiditeit (bv. aanwezigheid van een inflammatoire darmziekte of psoriasis of uveitis of bekend met een myocardinfarct) en de waarden en voorkeuren van patiënt.
Kosten (middelenbeslag)
Bij de keuze van een JAK-remmer (of andere tsDMARD) dient de voorschrijver naast het risico op het optreden van complicaties, ook de mogelijk kosten die gepaard gaan met een complicatie (en de behandeling hiervan) in acht te nemen.
Aanvaardbaarheid, haalbaarheid en implementatie
Na het falen op ≥1 csDMARD worden JAK-remmers (alsmede bDMARD’s) vergoed door de zorgverzekeraar bij RA. In veel ziekenhuizen geldt echter een lokaal beleid, wat tot op zekere hoogte de volgorde van de voor te schrijven bDMARD’s en JAK-remmer bepaald. Daarnaast zijn er in 2023 de PRAC-veiligheidswaarschuwingen verschenen, wat ook zijn weerslag heeft gehad op het voorschrijfgedrag. De adviezen die PRAC geeft, staan hierboven beschreven.
Rationale van de aanbeveling
In de beschikbare literatuur wordt niets beschreven over allergische reacties in vergelijk met andere DMARD’s. Mochten deze onverhoopt toch optreden dan is het advies het middel (tijdelijk) te staken.
Onderbouwing
Achtergrond
De tsDMARD’s, onderverdeeld in JAK-remmers en apremilast, verschillen op het gebied van veiligheid, dan wel het ontstaan van complicaties door gebruik van het middel. Aangezien de tsDMARD apremilast anno 2024 beperkt gebruikt wordt binnen de reumatologie en de recente ontwikkelingen ten aanzien van veiligheid bij JAK-remmers, focust de werkgroep zich in deze module op de JAK-remmers.
Op basis van de literatuur blijkt dat JAK-remmers een bewezen effect hebben op de ziekteactiviteit en patiënt-gerapporteerde uitkomstmaten bij volwassen patiënten met een inflammatoire reumatische aandoening. Lange termijn studies laten tevens een blijvend effect zien. Gezien het zeer uitgebreide therapeutisch arsenaal wordt echter naast de effectiviteit de nadruk ook steeds meer gelegd op de mogelijke (ernstige) bijwerkingen. Zo heeft de Pharmacovigilance Risk Assessment Committee (PRAC) van de Europese Geneesmiddelenbureau (EMA) recentelijk extra aanbevelingen gedaan omtrent het veilig gebruik van JAK-remmers. Om een goede indicatie te krijgen van de huidige plaatsbepaling van JAK-remmers in de dagelijkse praktijk is het van belang dat we risicogroepen herkennen, inzicht hebben in de frequentie waarmee bijwerkingen voorkomen en weten wat de verschillen in (ernstige) bijwerkingen zijn met de bestaande csDMARD’s en bDMARD’s en eventueel tussen de JAK-remmers.
Conclusies / Summary of Findings
Serious infections (other than herpes zoster)
tsDMARDs versus bDMARDs - Evidence from RCTs
Low GRADE |
Evidence from one RCT suggests that there may be an increased risk of serious infections (other than HZ) with tsDMARDs compared to bDMARDs in patients with RA, PsA or SpA.
Source: Sepriano (2023) |
tsDMARDs versus bDMARDs - Evidence from observational studies
Very low GRADE |
Evidence from observational studies suggests no difference in risk of serious infections (other than HZ) with tsDMARDs compared to bDMARDs in patients with RA, PsA or SpA but the evidence is very uncertain.
Sources: Sepriano (2023), Frisell (2023), Hirose (2022), Mok (2023), Uchida, (2023) |
tsDMARDS versus csDMARDs – Evidence from RCTs and observational studies
No GRADE |
No evidence was found regarding the risk of serious infections with tsDMARDs compared to csDMARDs in patients with RA, PsA or SpA.
Sources: None |
Herpes Zoster (HZ)
tsDMARDs versus bDMARDs - Evidence from RCTs
Low GRADE |
Evidence from RCTs suggests that there may be an increased risk of herpes zoster with tsDMARDs compared to bDMARDs in patients with RA, PsA or SpA.
Source: Sepriano (2023) |
tsDMARDs versus bDMARDs - Evidence from observational studies
Very low GRADE |
Evidence from observational studies showed an increased risk of herpes zoster with tsDMARDs compared to bDMARDs in patients with RA, PsA or SpA, but the evidence is very uncertain.
Sources: Sepriano (2023), Frisell (2023), Hirose (2022), Jeong (2022), Mok (2023), Song (2023b), Uchida (2023) |
tsDMARDS versus csDMARDs - Evidence from RCTs
Low GRADE |
Evidence from RCTs suggests that there may be an increased risk of herpes zoster with tsDMARDs compared to csDMARDs in patients with RA, PsA or SpA.
Source: Sepriano (2023) |
tsDMARDS versus csDMARDs - Evidence from observational studies
Very low GRADE |
Evidence from observational studies suggests an increased risk of herpes zoster with tsDMARDs compared to csDMARDs in patients with RA, PsA or SpA, but the evidence is very uncertain.
Source: Sepriano (2023) |
Major cardiovascular events (MACEs), including venous thromboembolism (VTE) and pulmonary embolism (PE)
tsDMARDs versus bDMARDs - Evidence from RCTs
Low GRADE |
Evidence from RCTs suggests that there may be an increased risk of MACE, including VTE and PE, with tsDMARDs compared to bDMARDs in patients with RA, PsA or SpA.
Source: Sepriano (2023) |
tsDMARDs versus bDMARDs - Evidence from observational studies
Very low GRADE |
The evidence from observational studies is very uncertain about the risk of MACE, including VTE and PE, with tsDMARDs compared to bDMARDs in patients with RA, PsA or SpA.
Sources: Sepriano (2023), Fang (2022), Frisell (2023), Hirose (2022), Hoisnard (2022), Khosrow-Khavar (2022), Min (2023), Mok (2023), Molander (2022), Pina Vegas (2022), Song (2023a) |
tsDMARDS versus csDMARDs - Evidence from RCTs
No GRADE |
No evidence from RCTs was found regarding the risk of MACE, including VTE and PE, with tsDMARDs compared to csDMARDs in patients with RA, PsA or SpA.
Sources: None |
tsDMARDS versus csDMARDs - Evidence from observational studies
Very low GRADE |
Evidence from one observational study suggests a lower risk of MACE, including VTE and PE, with tsDMARDs compared to csDMARDs in patients with RA, PsA or SpA, but the evidence is very uncertain.
Source: Sepriano (2023) |
Malignancies
tsDMARDs versus bDMARDs - Evidence from RCTs
Low GRADE |
Evidence from RCTs suggests that there may be an increased risk of malignancies with tsDMARDs compared to bDMARDs in patients with RA, PsA or SpA.
Source: Sepriano (2023) |
tsDMARDs versus bDMARDs - Evidence from observational studies
Very low GRADE |
The evidence from observational studies is very uncertain about the risk of malignancies with tsDMARDs compared to bDMARDs in patients with RA, PsA or SpA.
Sources: Sepriano (2023), Fang (2022), Hirose (2022), Huss (2023), Min (2023), Mok (2023), Song (2022), Uchida (2023), Westermann (2023) |
tsDMARDS versus csDMARDs - Evidence from RCTs and observational studies
No GRADE |
No evidence was found regarding the risk of malignancies with tsDMARDs compared to csDMARDs in patients with RA, PsA or SpA.
Sources: None |
Samenvatting literatuur
Description of EULAR study
Sepriano (2023) was a systematic literature review informing the 2022 update of the EULAR recommendations for the management of rheumatoid arthritis (RA). Databases were searched from 1 January 2019 to 14 January 2022 for publications addressing the safety of DMARD use (csDMARD, bDMARD—including biosimilars—or tsDMARD) in adult (≥18 years old) patients with RA, updating a previous SR. Studies were only eligible if they included a comparator group (either another DMARD, combination therapy, or the general population). Studies on glucocorticoids were excluded, as they were dealt with in a separate SR. The following safety outcomes were considered: infections, including serious infections, opportunistic infections such as tuberculosis (TB) and herpes zoster (HZ), malignancies, mortality, major adverse cardiovascular events (MACEs), venous thromboembolism (VTE), including pulmonary embolism (PE) and deep venous thrombosis (DVT), changes in lipid levels, elevations of creatine phosphokinase, impairments in renal function, elevations of liver enzymes, haematological abnormalities, gastrointestinal side effects, demyelinating disease, induction of autoimmune disease, teratogenicity, fertility and pregnancy outcomes. From a total of 2961 references, authors included 59 observational studies. From an accompanying SR addressing efficacy, two RCTs with a primary safety outcome, and 28 RCTs/long-term extensions (LTEs) were included. An important included RCT (i.e., also referenced and duplicated by later observational cohort studies) was the ORAL-Surveillance trial (In Sepriano, 2023: Ytterberg, 2022), a non-inferiority trial in which patients ≥50 years old who failed methotrexate and had ≥1 cardiovascular risk factor were randomised to tofacitinib 5 mg two times per day, tofacitinib 10 mg two times per day, or TNFi (adalimumab or etanercept). Safety outcomes in the ORAL-Surveillance trial were serious infections, HZ, malignancies, MACE, VTE, gastrointestinal perforations, and mortality. The trial was designed to test whether the upper limit of the 95% CI around the risk ratio of MACE or malignancies for tofacitinib (5 mg and 10 mg two times per day combined) compared with TNFi, was below 1.8 (the non-inferiority question). Studies in the EULAR SR were heterogeneous, precluding data pooling. Therefore, results were presented descriptively.
Description of additional studies
Fang (2022) was a retrospective cohort study in Taiwan examining the safety of tsDMARDs (Janus kinase inhibitors (JAKi)) compared with bDMARDs (tumor necrosis factor inhibitors (TNFi)) in patients with RA. Safety outcomes of interest were: coronary heart disease, stroke, overall thromboembolism, deep vein thrombosis, total hip replacement, total knee replacement, tuberculosis, malignancy, and all-cause mortality. Except for total hip/knee replacement and all-cause-mortality, safety outcomes were in line with our PICO. Data sources were the Taiwan National Health Insurance Research Database and the Taiwan Death Registry. Data from patients, aged >18 and <80, newly diagnosed with RA between 1995 and 2019, using JAKi or TNFi between 2015 and 2017 were selected. Study follow-up ended December 31, 2018. To reduce misclassification of RA, only RA patients with a catastrophic illness certificate, issued by an expert panel, were enrolled. They included 822 patients treated with JAKi (i.e., tofacitinib) and 2357 patients treated with TNFi (i.e., etanercept, adalimumab, and golimumab). All available relevant covariates were taken into account (i.e., age, sex, comorbidities, concomitant medication). The two groups were well-balanced in all these covariates after propensity score stabilized weighting. However, some variables associated with study outcomes, such as cigarette smoking, alcohol consumption, and exercise, were unavailable in the database that they used. Per safety outcome, group incidence rates (IR) per 100 person-years (PY), hazard ratios (HR) and 95% confidence intervals (CI) were provided. Authors also performed sensitivity analyses restricted to patients aged ≥ 50.
Frisell (2023) performed an observational cohort study in Sweden assessing the safety of all b/tsDMARDs used in Sweden. Outcomes of interest were discontinuation due to adverse events, MACE, serious infection (requiring inpatient care), hospital-treated herpes zoster, tuberculosis, non-steatosis liver disease, diagnosed depression, attempted or completed suicide, any hospitalization, and all-cause mortality. Authors used the longitudinal clinical registry infrastructure ‘Anti-Rheumatic Therapies in Sweden’ (ARTIS), which allows for linking of individual-level longitudinal data on treatments, disease activity and other clinical measurements from the Swedish Rheumatology Quality Register (SRQ), to prospectively collected data in Swedish national healthcare registers (i.e., National Patient Register, Prescribed Drug Register, census/taxation registers). They included 20117 patients with RA that started a b/tsDMARD between 1 January 2010 and 31 December 2020, who were followed until 30 June 2021, contributing a total of 34 279 treatment episodes. Authors used inverse probability of treatment weighting (IPTW) modelling to balance treatment groups on all relevant covariates (i.e. age, sex, immigration status, education, smoking, RF/anti-citrullinated peptide antibodies (ACPA), RA duration, previous b/tsDMARD use, comedication with csDMARDs and glucocorticoids, the DAS28-CRP, HAQ, comorbidity, medical history). Crude and adjusted incidence rates per 1000 patient-years, and adjusted hazard ratios comparing JAKi (baricitinib and tofacitinib) and bDMARDs (all but etanercept) to etanercept. Additional analyses included comparisons of b/tsDMARD cohorts to matched general population and b/tsDMARD-naïve cohorts. Sensitivity analyses tested the impact of the study period by restriction to (1) the time after JAKi market entry (excluding all b/tsDMARD starts before 1 January 2017) and (2) the time before the COVID-19 pandemic (follow-up
terminated on 28 February 2020).
Hirose (2022) was a multicenter, longitudinal observational study conducted at 12 hospitals and clinics for rheumatology in Japan. Authors compared tofacitinib (TOFA) and abatacept (ABT) on clinical outcomes, assessing effectiveness and safety in patients with RA. Patients fulfilled ACR/EULAR classification criteria for RA, started one of the two treatments between January 2015 and January 2021, had disease activity that was not controlled by MTX or csDMARDs, or were unable to be treated with csDMARDs, including MTX. Prior use of bDMARDs or JAK inhibitors was not a criterium for exclusion. Data collection was partly retrospective, partly prospective: Data from the patients who started treatments with TOFA or ABT or between January 2015 and December 2017 were obtained retrospectively from the patients’ medical records, data from patients who started the treatment between January 2018 and January 2021 were obtained prospectively. Their primary outcome of interest was the remission rate at week 52, measured by DAS28-ESR. Secondary outcomes were disease activity and safety at week 52, and authors investigated the effects of shared epitope positivity on clinical outcomes in and between treatment groups. RA patients starting TOFA (n=187) and ABT (n=183) were enrolled. Effectiveness outcomes were compared after applying IPTW based on a propensity score that reduces the selection bias to a minimum and adjusts for confounding factors between binary treatment groups. Regarding safety outcomes, authors reported on adverse events observed during the 52 weeks before adjustments with IPTW. The incidence and severity of all adverse events were recorded until week 52. Common terminology criteria for adverse events of the National Cancer Institute (v5.0) were used to describe and grade adverse events and laboratory abnormalities. Regarding the safety outcomes (most in line with our PICO), incidence in n (%) was reported for any adverse event, serious adverse event, death, serious infection, herpes zoster, cancer, major adverse cardiovascular events (MACE), and venous thromboembolism (VTE).
Hoisnard (2022) conducted a nationwide population-based cohort study in France using the French national health data system. Authors assessed the risk of MACEs and VTEs among patients with RA initiating a JAKi (tofacitinib and baricitinib) versus patients with RA initiating adalimumab. Between 1 July 2017 and 31 May 2021 (follow-up until 31 December 2021), authors identified 8481 patients with RA (age ≥ 18 yrs.) who initiated a JAKi and 7354 who initiated adalimumab. Authors balanced groups using IPTW; age, sex, use of csDMARD before and at index date, previous use of bDMARDs, comorbidities, use of systemic corticosteroids and NSAIDs before and at index date, use of antiplatelet agents and anticoagulants were included in the propensity score. Analyses were performed with concomitant administration of MTX as a time-varying variable. Incidence rates per 1000 patient-years, crude and weighted hazard ratios were provided. Sensitivity analyses were performed, adjusted for the same variables, but (1) with death as a competing risk, (2) using a conventional multivariate Cox model, and (3) with a broader definition of MACEs. Subgroup analyses included analyses of patients (1) on tofacitinib or baricitinib, (2) aged ≥ 50 years with ≥ 1 cardiovascular (CV) risk factor, (3) aged ≥ 65 years with ≥ CV risk factor, (4) having ≥ 1 CV risk factor, (5) divided by gender, and (6) divided by MACE subtype (myocardial infarction and stroke).
Huss (2023) was an observational cohort study in Sweden using prospectively collected individual-level clinical data enriched through linkage to other national Swedish registers, running from 1 January 2016 to 31 December 2020. Authors assessed cancer risks of (1) JAKi and (2) non-TNFi bDMARDs versus bDMARDs in clinical practice in patients with RA or PsA. All patients were older than 18 years and without a history of cancer. Authors included 10447 patients with RA and 4443 patients with PsA who initiated treatment with JAKi, a non-TNFi bDMARD or a TNFi. In RA patients, they observed 2143 initiations with a JAKi, 4128 initiations with a non-TNFi bDMARD, and 8580 initiations with a TNFi. In PsA patients, they observed 379 initiations with a JAKi, 185 initiations with a non-TNFi bDMARD, and 4186 initiations with a TNFi. Median follow-up time for JAKi was shorter than for bDMARDs due to their relatively recent introduction on the Swedish market. Authors estimated incidence rates, and hazard ratios via Cox regression, for all cancers excluding non-melanoma skin cancer (NMSC), and for individual cancer types including NMSC. In addition, for cancer incidence, results were not only described for treatment populations, but also for a matched general population. In analyses, all available relevant covariates were taken into account (i.e., age, sex, lifestyle, socioeconomics, comorbidities, disease activity, previous treatments, concomitant medication). Authors performed additional (sensitivity) analyses (1) fitting models by time since treatment initiation (≤1, 1–2, ≥2 years), (2) separating analyses by previous use of b/tsDMARD’s, (3) introducing a latency period of 90 days, so that only cancers diagnoses 90 days or later after treatment start would contribute to analyses, (4) restricting to a CV-enriched subset of treatment cohorts, (5) using an on-drug approach, (6) restricting the follow-up to February 2020, pre-COVID pandemic.
Jeong (2022) was an observational cohort study in South Korea using the Korean Health Insurance Review & Assessment Service database. Authors examined the risk of incident and recurrent herpes zoster (HZ) in seropositive RA patients, comparing the use of tsDMARD tofacitinib and the use of bDMARDs infliximab, adalimumab, golimumab, tocilizumab, rituximab to the use of bDMARDs abatacept and etanercept. Between January 2011 and January 2019, they included a total of 11720 RA patients (tofacitinib n=701; abatacept n=1153; etanercept n=2680), new users of a b/tsDMARD, age 20 years or older, without HZ within 1 month of b/tsDMARD use, and without HZ within 6 months of previous HZ. Authors calculated incidence rates per 1000 patient-years and hazard ratios. Hazard ratio analyses were adjusted for age, sex, number of csDMARD, the Charlson comorbidity index, enrollment year, steroids use, and history of zoster. Authors performed subgroup analyses stratified by sex (men and women), age (≥65 years and <65 years), number of conventional synthetic DMARD types (1–2 and >2), CCI (≤2 and >2), and glucocorticoid use within 6 months after the date of initial bDMARD or tsDMARD use (≥5 mg/day and <5 mg/day). In addition, to normalize the clinical application period of tofacitinib in South Korea, analyses were restricted to patients who started b/tsDMARDs from 2017 onward.
Khosrow-Khavar (2022) was a new user, active comparator observational cohort study in the United States of America using de-identified data from Optum Clinformatics (2012–2020), IBM MarketScan (2012–2018), and Medicare claims databases. Authors created a 1) a “real-world evidence (RWE) cohort” consisting of routine care patients; and 2) a “RCT-duplicate cohort” mimicking inclusion and exclusion criteria of the ORAL surveillance trial to calibrate results against the trial findings. They compared RA patients initiating treatment with tofacitinib with patients initiating treatment with tumor necrosis factor inhibitors (TNFi) on the risk of cardiovascular outcomes (a composite outcome of myocardial infarction and stroke). They included only new users (i.e., no prescription prior to entry date) of at least 18 years of age, without missing data on age or gender, and not admitted to a nursing facility or hospice prior to cohort entry date. In the RWE cohort, 28568, 34083, and 39612 patients who met the inclusion and exclusion criteria were identified from Optum, MarketScan, and Medicare respectively of whom 13.2%, 15.6%, and 9.5% initiated treatment on tofacitinib (the rest of the patients was on TNFi). In the ORAL RCT-duplicate cohort, 6878, 8071, and 20121 patients were identified from Optum, MarketScan, and Medicare, respectively, of whom 11.6%, 14.3%, and 7.7% initiated treatment with tofacitinib (the rest of the patients was on TNFi). Cox proportional hazards models with propensity score fine stratification weighting were used to estimate hazard ratios, accounting for 76 potential confounders. Pre-specified subgroup analyses were conducted based on age (≤65 and > 65), sex, and baseline cardiovascular disease (RWE cohorts only). In addition, analyses were stratified by unique number of previous agents of bDMARDs (i.e., 0 vs ≥1). Secondary analysis was also conducted by using an intention-to-treat exposure definition whereby patients were censored 365 days after initiation of treatment with tofacitinib or TNFI. Also, a 1:1 propensity score matching was conducted where each patient initiating tofacitinib was matched with a patient initiating TNFi using nearest neighbor greedy matching without replacement using a caliper of 0.025 on the natural scale of the propensity score. Finally, sensitivity analyses were conducted by restricting the TNFi comparator group in RWE and RCT-duplicate cohorts to only adalimumab and etanercept users, the specific TNFi which were the comparator in the ORAL Surveillance trial.
Min (2023) was a retrospective observational cohort study in Korea using Korean health insurance data. Authors assessed the risk of cancers, cardiovascular (CV) diseases (MACE, acute myocardial infarction (AMI), stroke), thromboembolisms (VTE, arterial thromboembolism (ATE)), and (CV-related or all-cause-related) mortality for treatment with JAKi compared to treatment with TNFi in either TNFi/JAKi naïve RA patients (set 1) or all RA patients (set 2). All patients were older than 18 years, had used MTX for at least six months before start with JAKi/TNFi, and did not have ankylosing spondylitis (AS), systemic lupus erythematosus (SLE), psoriasis (PS), psoriatic arthritis (PsA), Behçet’s disease, Crohn’s disease, or ulcerative colitis. In set 1, 645 RA patients on JAKi and 951 RA patients on TNFi were included. In set 2, 2498 RA patients on JAKi and 9267 RA patients on TNFi were included. Incidence rates ratios (IRRs) and adjusted HRs (adjusted for medication use, age, sex, and presence of comorbidities) for JAKi users compared to TNFi users were reported. Total possible study/treatment period in set 1 was from start with first JAKi/TNFi on July 1, 2017 to study end in December 31, 2021. Total possible study/treatment period in set 2 was from start with JAKi/ (a new) TNFi in March 1, 2015 to study end in December 31, 2021.
Mok (2023) was a retrospective observational cohort study in Hong Kong using data from the Hong Kong Biologics Registry between 2008 and 2021. Authors assessed the incidence of MACEs, cancers, (serious) infections (including TB, HZ, and COVID-19), (-related) mortality), and withdrawal rates due to a serious adverse event (SAE) or inefficacy for treatment with JAKi compared to treatment with TNFi in patients with RA. Patients with RA who were ever treated with JAKi/TNFi were included (n=1732) with a total of 2471 courses, 551 JAKi courses and 1920 TNFi courses. Incidence per 100 patient-years, adjusted incidence ratios and hazard ratios were provided. Analyses were adjusted for relevant covariates (i.e., age, sex, follow-up duration, and/or RA duration, and/or smoking, and/or BMI, and/or past history of MACE, vascular risk factors that required therapies, and/or history of cancer, and/or use of concomitant csDMARDs, diabetes mellitus). Additional subgroup analyses were performed on patients ≥ 65 years and with ≥ 1 atherosclerotic risk factor and on patients that started treatment since 2014, the year that JAKis were first used in Hong Kong.
Molander (2022) was a nationwide register-based, active comparator, new user design cohort study in Sweden from 2010 to 2021. The Swedish Rheumatology Quality Register was linked to national health registers to identify treatment cohorts (exposure) of RA patients (≥18 yrs of age) who were initiators of a JAKi, a TNFi, or a non-TNFi bDMARD (n=32737 treatment initiations, of which 2354 JAKi and 19950 TNFi)). Also, a general population cohort (matched 1:5; n=92.108) and an ‘overall RA’ comparator cohort (n=85 722) were identified. Cohorts were compared on the risk of venous thromboembolism (VTE; including separate comparisons for pulmonary embolism (PE) and deep vein thrombosis (DVT); TNFi was the reference cohort). Incidence and hazard ratios were provided. Cox proportional hazards regression analyses were adjusted for age, sex and number of previous b/tsDMARDs (ever), relevant comorbidities and treatments, healthcare consumption and socioeconomic variables, and for RA disease-related variables and smoking. Separated analyses by sex, RA serostatus, number of previous b/tsDMARDs and time since start of follow-up (0 to ≤1 year, 1 to ≤5 years and ≥5 years) were performed. To test the proportional hazards assumption, an interaction term between follow-up time (0 to ≤1 year, 1 to ≤5 years and≥5 years) and exposure was used. To test the robustness of the results, sensitivity analyses were performed. To maximize statistical precision, the main analysis used data from 2010. Since JAKis were introduced to the Swedish market in 2016, a separate analysis restricted to data from 2016 to 2021 was performed. To investigate the role of missingness, a complete case analysis (i.e., excluding patients with missing values for the disease-related variables and smoking) was performed, as well as an analysis using multiple imputation for variables with missing data. Finally, a separate analysis of patients fulfilling relevant inclusion and exclusion criteria for the ORAL surveillance study was performed, to assess how such enrichment for cardiovascular risk factors affected the VTE incidence and HRs.
Pina Vegas (2022) was a nationwide register-based, new user observational cohort study in France from 2015 to 2019 including all adult PsA patients starting a bDMARD (n=9510; including etanercept, infliximab, adalimumab, certolizumab, golimumab as TNFi, ustekinumab, secukinumab, ixekizumab) or apremilast (n=1885) and comparing the risk of cardiovascular events (i.e. MACE, a composite outcome combining acute myocardial infarction and ischaemic stroke) with the TNFi group (n=7289) as reference group. Incidence rates per 1000 patient-years and hazard ratios were reported. To control for confounding, inverse probability of treatment weighting (IPTW) was used. The following covariates were taken into account: age, sex, complementary universal health coverage and French deprivation index, inflammatory diseases associated with PsA, cardiovascular risk biomarkers and other comorbidities. Subgroup analyses were performed in patients without skin psoriasis requiring topical therapies and in patients without comorbidities related to CVD. Sensitivity analyses included: (i) a per-protocol analysis to try to avoid bias due to potential differential adherences to the drugs compared: follow-up was additionally censored at the time of treatment discontinuation; (ii) Fine–Gray competing risks analysis, computing IPTW subhazard ratios to account for the competing risk between all-cause out of- hospital death and hospitalization for MACEs; (iii) conventional multivariate Cox model computing adjusted HRs: the co-reimbursement of NSAIDs or prednisone with bDMARD or apremilast considered as time-varying variables; (iv) using a larger definition of MACE including unstable angina and transient ischaemic attack in addition to myocardial infarction and ischaemic stroke (v) modifying the new-user definition as those who had not filled a prescription for a bDMARDs or apremilast for 5 years before the index date (rather than 1 year as in the main analysis); and (vi) defining treatment discontinuation as >60 or >120 days without filling a prescription for the same treatment after the period covered by the previous prescription.
Song (2022) was a retrospective observational cohort study in the Republic of Korea using data from the Korean National Health Insurance database between 2015 and 2019. Authors compared the risk of malignancy (overall, solid, hematological) between JAKi and TNFi, and between different TNFi in patients with RA. They also sought to identify risk factors associated with serious infection and herpes zoster. RA patients were older than 18 years, JAKi/TNFi naïve, had no other systemic inflammatory disease (i.e., AS, PsA, inflammatory bowel disease (IBD) or juvenile idiopathic arthritis (JIA)), had an observation period of ≥6 months, were in remission (i.e., no recurrence of malignancy for 5 years prior to start JAKi/TNFi).
To control potential confounding factors, IPTW was performed to balance characteristics between the JAKi and TNFi groups. To calculate the probability of being prescribed
JAKi, a multivariable logistic regression model was used taking into account numerous demographic and clinical characteristics for the propensity score (i.e., age, sex, geographical region, level of household income, type of insurance, type of institution, year of initiating JAKi or TNFi, seropositivity of RA, comorbidities, medication use and healthcare usage). The study included a total of 4929 patients (1064 JAKi and 3865 TNFi); the observation periods were 1288.6 person-years for JAKi users and 6823.8 Pys for TNFi users. Authors provided incidence rates per 100 patient-years and hazard ratios, crude/before IPTW and weighted/after IPTW. Additionally, subgroup analyses (according to sex, age and concomitant use of MTX) and sensitivity analyses (including a latent period of 6 months or 1 year, and with/without a 24-week permissible gap) were performed.
Song (2023a) was a nationwide observational cohort study in the Republic of Korea using the National Health Insurance Service database between 2015 and 2019 to examine the risk of venous thromboembolism (VTE, including pulmonary embolism (PE) and deep vein thrombosis (DVT)) in RA patients, comparing first JAKi users and first TNFi users. Authors included 4178 new users of JAKi (n=871; tofacitinib or baricitinib) or TNFi (n=3307; adalimumab, etanercept, golimumab, or infliximab), all adult (≥18) RA patients without other autoimmune diseases and without a prior history of VTE and/or anticoagulant therapy within 30 days of start JAKi/TNFi. Groups were balanced using inverse probability of treatment weighting (IPTW). Covariates taken into account calculating the propensity score were age, sex, geographical region, level of household income, type of insurance, type of institution, year of initiating JAKi or TNF inhibitor, seropositivity of RA, comorbidities, medication use, and healthcare utilization. Incidence rates per 100 patient-years and adjusted hazard ratios for VTE and PE and DVT separately, were calculated. Additional analyses included a subgroup study according to sex, age, and comorbidities, and an analysis comparing tofacitinib users with TNF inhibitor users.
Song (2023b) was a single-center prospective study in the Republic of Korea aimed to determine the risk of herpes zoster (HZ) in Korean rheumatoid arthritis (RA) patients on tofacitinib compared with TNFi treatment (i.e., etanercept, infliximab, adalimumab, golimumab). RA patients in an academic referral hospital in Korea (n=912) who started tofacitinib (n=200) between March 2017 and May 2021 and those who started TNFi (n=712) between July 2011 and May 2021 were included. Baseline characteristics of tofacitinib and TNFi users were balanced through inverse probability of treatment weighting (IPTW) using the propensity score including age, sex, duration of RA, comorbidities, disease activity of RA, and medication use. The incidence rates per 100 patient-years and incidence rate ratios (IRR) were calculated for outcomes HZ and serious HZ. In sensitivity analyses, the IRR of HZ development within 12 months of tofacitinib or TNFi use was calculated. Also, the IRR of HZ in the study population after matching the inclusion periods (March 2017 to May 2021) of the two groups was calculated.
Uchida (2023) was a retrospective observational cohort study in Japan. Authors compared the risk of serious infections, herpes zoster, MACE, and malignancies (non-hematopoietic) in RA patients, comparing JAKi users to TNFi users and Authors included 499 patients who were diagnosed with RA and treated with JAKi (tofacitinib (n=192) or baricitinib (n=104)) or TNFi (adalimumab or etanercept) (n=203) at Nagasaki University Hospital, Sasebo Chuo Hospital, or Ureshino Medical Center during the period from March 2013 through December 2020, and consented to the use of their data. To balance groups, authors performed IPTW for which age, sex, RF positivity, ACPA positivity, the coexistence of diabetes mellitus, the coexistence of lung disease, the dose of MTX, the dose of GC, the previous use of b/ts DMARDs, and the disease duration were taken into account. For serious infections, herpes zoster, MACE and malignancies, hazard ratios comparing TNFi to JAKi were provided. For infections, also incidence rates per 100 patient-years were provided per treatment group, and hazard ratios of risk factors. For malignancies, treatment groups were also compared with the general Japanese population in the same region as the hospital population, using the standardized incidence ratio.
Westermann (2023) performed an observational cohort study in Denmark exploring the risk of first primary cancer (excl. NMSC) in patients with RA treated with JAKi (tofacitinib and baricitinib) compared with RA patients treated with bDMARDs in the period 1 January 2017 to 31 December 2020. They used (and linked) data from the Danish Rheumatology Quality Register, the Danish Cancer Registry, the Danish Civil Registration System, the Danish Population Education Register, and the Danish National Patient Register. Authors used IPTW modelling to balance treatment groups on all covariates either associated with cancer or with both cancer and treatment choice (i.e., age, sex, education level, relevant indicators of existing comorbidities, smoking status, covariates reflective of RA characteristics, concomitant medication use, number of previous bDMARDs). Death was considered a competing risk due to its preclusion of cancer occurrence. Therefore, authors calculated 95% bootstrap Cis (95% CI) with 500 iterations for all weighted models. The study included 875 RA patients using JAKi (1315 patient-years) and 4247 RA patients using bDMARDs (8597 patient-years). Authors provided crude incidence rates of cancer per 1000 patient-years for both JAKi and bDMARD groups, and weighted hazard ratios comparing JAKi to bDMARD treatment. For comparison with the ORAL Surveillance study with only patients with RA aged 50+, authors performed subgroup analyses stratifying the main analysis by age groups 50+ and 65+, and also by length of follow-up (<1 year and 1+ years). Also sensitivity analyses were performed: (1) two on-drug analyses taking into account initiation and discontinuation dates, allowing for switching between groups, enabling patients to contribute to both groups; (2) two analyses lagging discontinuation dates with 3 months and 6 months, respectively, to consider ‘carry-over effects’; (3) two analyses testing the impact of various inclusion criteria (i.e. at least one prior bDMARD treatment, only patients who were ‘ever’ smokers); (4) two separate analyses with the bDMARD group consisting only of patients initiating TNFi and non-TNFi bDMARDs, respectively, while follow-up was stopped upon switching to the opposite bDMARD class, establishing less heterogeneous comparator groups and mimicking the pure TNFi exposure comparison of the ORAL Surveillance study.
Results
1. Infections
In addition to the SR (Sepriano, 2023), our search resulted in the selection of six observational cohort studies (Frisell, 2023; Hirose, 2022; Jeong, 2022; Mok, 2023; Song, 2023b; Uchida, 2023) that examined the risk of infections of treatment with tsDMARDs as compared to treatment with TNFi (Frisell, 2023; Jeong, 2022; Mok, 2023; Song, 2023b; Uchida, 2023) or T-cell inhibitor, abatacept (Hirose, 2022; Jeong, 2022), all in patients with RA. Results for serious infections and herpes zoster are reported separately.
1.1. Serious infections (other than herpes zoster)
Results from RCTs
In the ORAL-Surveillance RCT (in Sepriano, 2023: Ytterberg, 2022) dose dependent differences were found in the risk of serious infection for tofacitinib versus TNFi in a RA population of patients ≥50 years old who failed methotrexate and had ≥1 cardiovascular risk factor. The risk of serious infections was increased with tofacitinib 10 mg 2/daily compared with TNFi: HR 1.48 (95% CI 1.17, 1.87), number needed to harm (NNH) = 17. In the same study, the risk of serious infections with tofacitinib 5 mg 2/daily was not increased compared with TNFi: HR 1.17 (95% CI 0.92, 1.50), NNH 48.
Results from observational studies
Three observational studies examined the risk of serious infections of tofacitinib versus bDMARDs in RA patients (in Sepriano, 2023: Chen, 2021; Kremer, 2021; Pawar, 2020), with consistent results. In these studies, serious infections were not more common with tofacitinib than with bDMARDs. The adjusted hazard ratio (aHR) was 0.99 (95% CI 0.75 to 1.30) in the only low risk of bias (RoB) study, the CorEvitas (formerly CORRONA) registry study (in Sepriano, 2023: Kremer, 2021).
In addition to the EULAR SR (Sepriano, 2023), we included four observational studies (Frisell, 2023; Hirose, 2022; Mok, 2023; Uchida, 2023). None of these studies found an increased risk for serious infections comparing tsDMARDs (tofacitinib, baricitinib) versus bDMARDs (adalimumab, etanercept, abatacept) in RA patient populations:
- Frisell (2023): baricitinib (aHR 0.98 (95% CI 0.75, 1.29)) or tofacitinib (aHR 0.89 (95% CI 0.47, 1.69) versus TNFi etanercept.
- Hirose (2022): tofacitinib n=4 (2.1%) versus abatacept n=4 (2.2%).
- Mok (2023): JAKi versus TNFi; IR 0.97 (95% CI 0.76, 1.23).
- Uchida (2023): TNFi (adalimumab and etanercept) vs. JAKi (tofacitinib and baricitinib); aHR 0.792 (95% CI 0.417, 1.502).
1.2 Herpes Zoster (HZ)
Results from RCTs
The included RCTs showed an increased risk of HZ for tofacitinib compared to bDMARDs, but for upadacitinib and filgotinib, compared to csDMARDs and bDMARDs, results were mixed. All studies were in RA patient populations. In the ORAL-Surveillance RCT (in Sepriano, 2023: Ytterberg, 2022), the risk of infections by HZ was increased both with tofacitinib 10 mg and 5 mg compared to TNFi (HR 3.28 (95% CI 2.44, 4.41), NNH = 8 and HR 3.39 (95% CI 2.52, 4.55), NNH = 7 resp.) in RA patients of ≥50 years old who failed methotrexate and had ≥1 cardiovascular risk factor. Three RCTs (in Sepriano, 2023: Fleischmann, 2019; Smolen, 2019; van Vollenhoven, 2020) found that the number of infections caused by HZ was higher with upadacitinib (range: 0.8%–3%), with or without MTX, than with adalimumab plus MTX or MTX alone (0.3%–0.5%), while one RCT (Rubbert-Roth, 2020) found a similar HZ infection risk for upadacitinib plus MTX (1.3%) versus abatacept plus MTX (1.3%). Another RCT (in Sepriano, 2023: Kameda, 2020), found more HZ infections with upadacitinib 30 mg (6.0%), but not upadacitinib 7.5 mg or 15 mg (2.0% and 0.0.% resp.), as compared to a placebo plus csDMARD (2.0%). Two RCTs (in Sepriano, 2023: Combe, 2021; Westhovens, 2021) found a similar number of HZ infections for filgotinib plus MTX (0.4%–1.0%) versus adalimumab plus MTX or versus MTX (0.6%–1%) while one RCT (in Sepriano, 2023: Genovese, 2019) found the number of HZ infections to be elevated in filgotinib plus csDMARD groups (1.3%-1.4%), as compared to a placebo plus csDMARD group (0.0%).
Results from observational studies
In observational studies in RA patient populations, the EULAR review (Sepriano, 2023) found a higher risk of HZ with tsDMARDs (tofacitinib, baricitinib and upadacitinib) compared to csDMARDs and bDMARDs. In one study at low RoB from the German RABBIT registry (in Sepriano, 2023: Redeker, 2022), the risk of HZ was higher with JAKi (tofacitinib, baricitinib and upadacitinib) than with csDMARDs (aHR 3.66 (95% CI 2.38, 5.63)). Moreover, in three studies (in Sepriano, 2023: Khosrow-Khavar, 2022; Kremer, 2021; Pawar, 2020) infections by HZ were more frequent with tofacitinib than with bDMARDs; aHR 2.32 (95% CI 1.43, 3.75) in the low RoB study CorEvitas (formerly CORRONA) registry study (in Sepriano, 2023: Kremer, 2021).
In addition to the results of the EULAR SR (Sepriano, 2023), we included results from six observational studies (Frisell, 2023; Hirose, 2022; Jeong, 2022; Mok, 2023; Song, 2023b; Uchida, 2023). In all these studies, the risk of HZ was increased with tsDMARDs (tofacitinib, baricitinib) versus bDMARDs (etanercept, infliximab, adalimumab, golimumab, abatacept) in RA patient populations:
- Frisell (2023): baricitinib (aHR 3.82 (95% CI 2.05, 7.09)) or tofacitinib (aHR 4.00 (95% CI 1.59, 10.06) versus TNFi etanercept.
- Hirose (2022): tofacitinib n=17 (9.1%) versus abatacept n=5 (2.7%).
- Jeong (2022): tofacitinib versus abatacept (aHR 2.46 (95% CI 1.61, 3.76)); tofacitinib versus etanercept (aHR 2.06 (95% CI 1.38, 3.08).
- Mok (2023): JAKi Incidence/100 patient-years (3.49) versus TNFi (0.94).
- Song (2023b): tofacitinib versus TNFi (etanercept, infliximab, adalimumab, golimumab): incidence risk ratio (IRR) 8.33 (95%CI 3.05–22.76).
- Uchida (2023): TNFi (adalimumab and etanercept) vs. JAKi (tofacitinib and baricitinib); aHR 0.200 (95% CI 0.077, 0.524).
2. Major cardiovascular events (MACEs), including venous thromboembolism (VTE) and pulmonary embolism (PE)
In addition to the EULAR SR (Sepriano, 2023), we selected 10 observational cohort studies that examined the risk of MACEs, VTEs and PE associated with treatment with JAKi as compared to treatment with TNFi (Fang, 2022; Frisell, 2023; Hoisnard, 2022; Khosrow-Khavar, 2022; Min, 2023; Mok, 2023; Molander, 2022; Song, 2023a) or abatacept (Hirose, 2022). Pina Vegas (2022) examined the risk of MACE in a patient population with PsA, comparing apremilast to TNFi.
Results from RCTs
In Sepriano (2023), RCTs comparing JAKi to TNFi, found higher, dose-dependent risks of MACE and VTE, only in patients with cardiovascular risk factors. Sepriano (2023) reported that in the ORAL surveillance RCT (in Sepriano, 2023: Ytterberg, 2022), which examined patients ≥50 years old who failed methotrexate and had ≥1 cardiovascular risk factor, compared with TNFi, tofacitinib was associated with an increased risk of MACE (HR 1.33 (95% CI 0.91, 1.94), NNH 82) over 5.5 years (not statistically significant). Non-inferiority of tofacitinib could not be claimed for MACE. In the same study, also the risk of VTE was increased with tofacitinib compared with TNFi (tofacitinib 5 mg HR 1.66 (95% CI 0.76, 3.63), NNH 153; tofacitinib 10 mg HR 3.52 (95% CI 1.74, 7.12), NNH 40). In two LTEs up to 52 weeks, the incidence of MACE was reported to be similar with JAKi (filgotinib and upadacitinib) and adalimumab in patients who, by design, did not had to have cardiovascular risk factors (in Sepriano, 2023: Combe, 2021 and Fleischmann, 2019).
Results from observational studies
Sepriano (2023) reported a lower to similar risk of MACE with tofacitinib as compared to csDMARDs and bDMARDs. In one observational study, at unclear RoB, patients on tofacitinib had a (non-statistically significant) lower risk of MACE compared with patients on csDMARDs (aHR 0.23 (95% CI 0.03, 1.62); in Sepriano, 2023: Ozen, 2021). In two other observational studies, at low RoB, the risk of MACE and VTE was reported to be similar with tofacitinib and bDMARDs (in Sepriano 2023: Kremer, 2021, MACE aHR: 0.61 (95% CI 0.34 to 1.06); Desai, 2021, VTE aHR: 1.13 (95% CI 0.77, 1.65)).
Comparing tsDMARDs with bDMARDs on cardiovascular risks, results were mixed in the 10 observational studies we added to Sepriano (2023). One observational study (Molander, 2022) reported an increased risk of VTE (PE and deep vein thrombosis (DVT); aHR 1.73 (95% CI 1.24, 2.42)), when comparing JAKi (tofacitinib, baricitinib, upadacitinib) versus TNFi (etanercept, infliximab, adalimumab, golimumab, certolizumab pegol). The increased risk was confined to PE aHR 3.21 (95% CI 2.11, 4.88) and associated with baricitinib use (aHR 1.79 (95% CI 1.25, 2.55). Two other studies, one in RA patients and another in PsA patients, also found an increased risk of MACE, albeit with wide confidence intervals indicating no statistical significance, for JAKi (Mok, 2023: aHR 1.36 (95% CI 0.62, 2.96)) or apremilast (Pina Vegas, 2022: wHR 1.4 (95%CI 0.8-2.4)) compared to TNFi. In contrary, one study (Min, 2023) found a lower risk of MACE, and with less certainty (no statistical significance), also VTE, for JAKi (tofacitinib, baricitinib, upacitinib) compared to TNFi (etanercept, adalimumab, infliximab, golimumab) in both newly diagnosed, b/tsDMARD naive RA patients (MACE aHR 0.59 (95% CI 0.35, 0.99); VTE aHR 0.33 (95% CI 0.07, 1.54)) and a set including all RA patients (MACE aHR 0.80 (95% CI 0.67, 0.97), 0.02; VTE aHR 1.34 (0.90, 1.99)). A study by Song (2023a), in an overlapping but larger cohort than the study by Min (2023) found a similar lower risk of VTE, albeit again with very wide confidence intervals indicating no statistical significance (aHR 0.18 (95% CI 0.01, 3.47)) and found that this was limited to cases of DVT (aHR 0.22 (95% CI 0.01, 4.32)), not PE (no cases in JAKi group)for JAKi tofacitinib and baricitinib compared to TNFi (adalimumab, etanercept, golimumab, infliximab). Four other studies indicated similar risks of MACE, including stroke, VTE, and DVT, when comparing tofacitinib and/or baricitinib to TNFi ([1] Fang, 2022: tofacitinib Coronary heart disease HR 1.03 (95% CI 0.45, 2.36); stroke HR 0.75 (95% CI 0.29, 1.94); VTE HR 0.65 (95% CI 0.25-1.70); DVT HR 0.57 (95% CI 0.20, 1.64); [2] Frisell, 2023: tofacitinib MACE aHR 0.78 (95% CI 0.31-1.99); baricitinib MACE aHR 0.83 (95% CI 0.49-1.42); [3] Hoisnard, 2022: JAKi tofacitinib and baricitinib MACE HRw 1.0 (95% CI 0.7, 1.5); VTE HRw 1.1 (95% CI 0.7, 1.6)) or to abatacept (Hirose, 2022: tofacitinib MACE 1.1%; VTE 0% versus abatacept MACE 0.5%; VTE 0.5%). A study by Khosrow-Khavar (2022), comparing tofacitinib to TNFi, found a similar risk of MACE in a real-world-evidence cohort (wHR 1.01 (95% CI 0.83, 1.23)) and, similar to the results of the ORAL Surveillance RCT, an increased risk of MACE in a ORAL Surveillance-duplicate cohort (wHR 1.24 (95% CI 0.90, 1.69)).
3. Malignancies
In addition to the EULAR SR (Sepriano, 2023), we selected eight observational cohort studies that examined the risk of malignancies associated with treatment with JAKi as compared to treatment with bDMARDs (Westermann, 2023), TNFi only (Fang, 2022; Huss, 2023; Min, 2023; Mok, 2023; Song, 2022; Uchida, 2023) or abatacept (Hirose, 2022) in RA patients. Besides RA patients, Huss (2023) also included a patient cohort with PsA.
Results from RCTs
In Sepriano (2023), RCTs comparing JAKi to TNFi, showed higher risks of malignancies for tofacitinib, but not filgotinib and upadacitinib. Sepriano (2023) reported that, compared with TNFi, tofacitinib (both 5 mg and 10 mg) was associated with an increased risk of malignancies (HR: 1.48 (95% CI 1.04 to 2.09), NNH 55) over 5.5 years in the ORAL-Surveillance RCT (in Sepriano, 2023: Ytterberg, 2022). Non-inferiority of tofacitinib could not be claimed for malignancies. In two LTEs up to 52 weeks, the incidence of malignancies was similar with JAKi (filgotinib and upadacitinib) and adalimumab in patients who, by design, did not had to have malignancy risk factors (in Sepriano, 2023: Combe, 2021 and Fleischmann, 2019).
Results from observational studies
One observational study (in Sepriano, 2023: Kremer, 2021), at low RoB, found a similar risk of malignancies with tofacitinib and bDMARDs (all malignancies except non-melanoma skin cancer (NMSC) aHR 1.04 (95% CI 0.68, 1.61); NMSC aHR 1.02 (0.69; 1.50)).
Comparing JAKi with bDMARDs on malignancy risks, results were mixed in RA patient populations in the eight observational studies we added to Sepriano (2023). Four studies showed a lower risk of any malignancy with JAKi as compared to TNFi, but with wide confidence intervals indicating statistical non-significance (Mok, 2023: aHR 0.87 (95% CI 0.39, 1.95); Song, 2022: wHR 0.83 (95% CI 0.55, 1.27); Uchida, 2023: aHR 0.385 (95% CI 0.095, 1.552); Hirose, 2022: tofacitinib 0% versus abatacept 0.5%). Results by Fang (2022) showed a similar to higher risk of malignancies with JAKi as compared to TNFi, but again with a wide confidence interval indicating statistical non-significance (Fang, 2022: any malignancy HR 1.10 (95% CI 0.44, 2.78)). Results by Min (2023) in a patient cohorts that overlapped the cohort of Song (2022), but were smaller and with less history of comorbidity, varied: after one year of follow-up, Min (2023) found a lower risk of cancer (excluding NMSC) in an all RA (b/tsDMARD naïve and non-naïve) JAKi group as compared to an all RA TNFi group (aHR 0.59 (0.39, 0.89), but when taking the full duration of follow-up into account (mean in all groups > 2 years), Min (2023) found evidence of similar to higher risks of malignancies with JAKi as compared to TNFi (in newly diagnosed, b/tsDMARD naïve patients, all cancers but NMSC aHR 1.29 (95% CI 0.68, 2.45); no NMSC cases; in all RA patients, all cancers but NMSC aHR 0.94 (95% CI 0.74, 1.19); NMSC aHR 1.38 (95% CI 0.39, 4.88)). Huss (2023) found a similar risk for all cancers but NMSC (aHR 0.94 (95% CI 0.65 to 1.38)), but a higher risk of NMSC with JAKi as compared to TNFi (RA NMSC aHR 1.39 (95% CI 1.01, 1.91)). A study by Westermann (2023) compared JAKi with bDMARDs (TNFi and non-TNFi) and found a higher risk of any malignancy but NMSC (wHR 1.41 (95% CI 0.76, 2.37)). One study (Huss, 2023) also included a PsA patient cohort and found a higher risk of any malignancy, but again with a wide confidence interval indicating statistical non-significance (PsA all cancers but NMSC aHR 1.88 (95% CI 0.68 to 5.16); PsA NMSC aHR 2.05 (95% CI 0.79 to 5.31)).
Level of evidence of the literature
Evidence included both results from RCTs and observational cohort studies as reported in Sepriano (2023) and results from observational cohort studies published after Sepriano (2023), 13 nationwide registry studies (Fang, 2022; Frisell, 2023; Hoisnard, 2022; Huss, 2023; Jeong, 2022; Khosrow-Khavar, 2022; Min, 2023; Mok, 2023; Molander, 2022; Pina Vegas, 2022; Song, 2022; Song, 2023a; Westermann, 2023), one single-center prospective study (Song, 2023b), and two multicenter studies (Hirose, 2022; Uchida, 2023). All observational cohort studies had only minor concerns regarding risk of bias; they applied the best statistical methods (IPTW) to control for confounding. With respect to RCTs that evaluated tsDMARDs, Sepriano (2023) reported that all but one of the included RCTs were not designed, and therefore, not powered, to evaluate safety outcomes. The incidence of major adverse events was low and mostly comparable between active treatment, placebo or active comparator. The exception was the ORAL-Surveillance study (in Sepriano, 2023: Ytterberg, 2022). However, this study did not include all RA patients, but a specific subpopulation of patients ≥50 years old who failed methotrexate and had ≥1 cardiovascular risk factor. Therefore, results cannot with certainty be generalized to the RA patient population at large, nor the general PsA and SpA patient populations.
In the following section, level of evidence is reported per safety outcome and separately for RCTs and observational cohort studies, using the GRADE system.
1. Infections
1.1. Serious infections (other than herpes zoster)
tsDMARDs versus bDMARDs - Evidence from RCTs
Considering the results of one RCT, the level of evidence regarding the risk of serious infections with tsDMARDs as compared to bDMARDs started high but was downgraded by one level for indirectness and another level for imprecision resulting in a low level of evidence.
tsDMARDs versus bDMARDs - Evidence from observational studies
Considering the results from observational studies, the level of evidence regarding the risk of serious infections with tsDMARDs as compared to bDMARDs started low and was further downgraded by one level for imprecision resulting in a very low level of evidence.
tsDMARDS versus csDMARDs – Evidence from RCTs and observational studies
No evidence was found regarding the risk of serious infections with tsDMARDs as compared to csDMARDs. The level of evidence for this comparison was not assessed due to lack of data.
Herpes Zoster (HZ)
tsDMARDs compared to bDMARDs - Evidence from RCTs
Considering the results from RCTs, the level of evidence regarding the risk of HZ with tsDMARDs as compared to bDMARDs, started high but was downgraded by one level for inconsistency and another level for imprecision resulting in a low level of evidence.
tsDMARDs compared to bDMARDs - Evidence from observational studies
Considering the results from observational studies, the level of evidence regarding the risk of HZ with tsDMARDs as compared to bDMARDs started low and was further downgraded by one level for imprecision resulting in a very low level of evidence.
tsDMARDS compared to csDMARDs - Evidence from RCTs
Considering the results from RCTs, the level of evidence regarding the risk of HZ with tsDMARDs as compared to csDMARDs started high but was downgraded by one level for inconsistency and another level for imprecision resulting in a low level of evidence.
tsDMARDS compared to csDMARDs - Evidence from observational studies
Considering the results from one observational study, the level of evidence regarding the risk of HZ with tsDMARDs as compared to csDMARDs started low and was further downgraded by one level for imprecision resulting in a very low level of evidence.
2. Major cardiovascular events (MACEs), including venous thromboembolism (VTE) and pulmonary embolism (PE)
tsDMARDs compared to bDMARDs - Evidence from RCTs
Considering the results from RCTs, the level of evidence regarding the risk of MACE, including VTE and PE, with tsDMARDs as compared to bDMARDs, started high but was downgraded by one level for inconsistency and another level for imprecision resulting in a low level of evidence.
tsDMARDs compared to bDMARDs - Evidence from observational studies
Considering the results from observational studies, the level of evidence regarding the risk of MACE, including VTE and PE, with tsDMARDs as compared to bDMARDs started low and was further downgraded by one level for inconsistency and another for imprecision resulting in a very low level of evidence.
tsDMARDS compared to csDMARDs – Evidence from RCTs
No evidence from RCTs was found regarding the risk of MACE, including VTE and PE, with tsDMARDs as compared to csDMARDs. The level of evidence with regard to this comparison was not assessed due to lack of data.
tsDMARDS compared to csDMARDs - Evidence from observational studies
Considering the results from one observational study, the level of evidence regarding the the risk of MACE, including VTE and PE, with tsDMARDs as compared to csDMARDs started low and was further downgraded by one level for imprecision resulting in a very low level of evidence.
3. Malignancies
tsDMARDs compared to bDMARDs - Evidence from RCTs
Considering the results from RCTs, the level of evidence regarding the risk of malignancies with tsDMARDs as compared to bDMARDs, started high but was downgraded by one level for inconsistency and another level for imprecision resulting in a low level of evidence.
tsDMARDs compared to bDMARDs - Evidence from observational studies
Considering the results from observational studies, the level of evidence regarding the risk of malignancies with tsDMARDs as compared to bDMARDs started low and was further downgraded by one level for inconsistency and another for imprecision resulting in a very low level of evidence.
tsDMARDS compared to csDMARDs – Evidence from RCTs and observational studies
No evidence was found regarding the risk of malignancies with tsDMARDs as compared to csDMARDs. The level of evidence with regard to this comparison was not assessed due to lack of data.
Zoeken en selecteren
A systematic review of the literature was performed to answer the following question:
What are the risks of treatment with targeted synthetic (ts)DMARDs compared to treatment with biological (b)DMARDs or conventional synthetic (cs)DMARDs in patients with rheumatoid arthritis (RA), psoriatic arthritis (PsA) or spondylarthritis (SpA)?
The systematic review of literature aimed to answer the following research sub- questions, each directed at specific outcomes only. Not all types of infections were included in the research question / PICO performed for module (ernstige) infecties als complicatie bij tsDMARD’s. No research question / PICO was formulated for module Allergische reacties als complicatie bij tsDMARD’s.
- Sub-question 1: What is the risk of (serious) infections: With what frequency do (serious) infections occur in patients with rheumatoid arthritis (RA), psoriatic arthritis (PsA) or spondylarthritis (SpA) who are treated with tsDMARDs compared to those treated with bDMARDs or csDMARDs?
- Sub-question 2: What is the risk of cardiovascular complications: With what frequency do cardiovascular complications occur in patients with rheumatoid arthritis (RA), psoriatic arthritis (PsA) or spondylarthritis (SpA) who are treated with tsDMARDs compared to those treated with bDMARDs or csDMARDs?
- Sub-question 3: What is the risk of malignancies: With what frequency do malignancies occur in patients with rheumatoid arthritis (RA), psoriatic arthritis (PsA) or spondylarthritis (SpA) who are treated with tsDMARDs compared to those treated with bDMARDs or csDMARDs?
P: | patients with RA, PsA, or SpA |
I: | treatment with tsDMARD |
C1: | treatment with bDMARD |
C2: | treatment with csDMARD |
O: |
sub-question 1: infections, 1.1 serious infections and 1.2 herpes zoster sub-question 2: major cardiovascular events (MACEs), including venous thromboembolism (VTE) and pulmonary embolism (PE) sub-question 3: malignancies |
Relevant outcome measures
- A priori, the working group did not define the outcome measures listed above but used the definitions used in the studies.
- The working group predefined all outcomes as a critical outcome measure for decision making.
- The working group did not define values for minimal clinically (patient) important differences per outcome measure due to the suspected low number of events. In addition, the working group discussed that these values were also based on individual patient characteristics.
Search and select (Methods)
Recently, the EULAR performed a comprehensive systematic literature review (SR) on the safety of synthetic and biological DMARDs (Sepriano, 2023). They performed a systematic search in line with our research question until 14 January 2022. We used the results of this SR to answer our research question and performed a complementary systematic search on the 2nd of June 2023. Databases Embase.com and Ovid/Medline were searched for SRs, randomized controlled trials (RCTs), and observational studies on complications of using tsDMARDs in patients with rheumatoid arthritis, psoriatic arthritis or spondylarthritis. The search resulted in 933 unique hits.
Studies were selected based on the following criteria:
- They should be a SR, RCT, or observational study;
- published since 14 January 2022;
- be in line with our research (sub)question(s).
Results
Initially, in addition to the EULAR SR (Sepriano, 2023), 31 studies were selected based on title and abstract screening. After reading the full text, 16 studies were selected, all observational studies (Fang, 2022; Hirose, 2022; Hoisnard, 2022; Huss, 2023; Jeong, 2022; Khosrow-Khavar, 2022; Min, 2023; Mok, 2023; Molander, 2022; Pina Vegas, 2022; Song 2022; Song, 2023a; Song, 2023b; Uchida, 2023; Westermann, 2023), and 15 papers were excluded (see the table with reasons for exclusion under the tab ’Bijlagen’). Important study characteristics and results are summarized in the evidence tables. The quality assessment is summarized in the table of Quality assessment for systematic reviews of RCTs and observational studies and the Risk of bias table for interventions studies (cohort studies based on risk of bias tool by the CLARITY Group at McMaster University).
Referenties
- Asenjo-Lobos C, González L, Bulnes JF, Roque M, Muñoz Venturelli P, Rodríguez GM. Cardiovascular events risk in patients with systemic autoimmune diseases: a prognostic systematic review and meta-analysis. Clin Res Cardiol. 2024 Feb;113(2):246-259. doi: 10.1007/s00392-023-02291-4. Epub 2023 Aug 31. PMID: 37650912.
- Duru N, van der Goes MC, Jacobs JW, Andrews T, Boers M, Buttgereit F, Caeyers N, Cutolo M, Halliday S, Da Silva JA, Kirwan JR, Ray D, Rovensky J, Severijns G, Westhovens R, Bijlsma JW. EULAR evidence-based and consensus-based recommendations on the management of medium to high-dose glucocorticoid therapy in rheumatic diseases. Ann Rheum Dis. 2013 Dec;72(12):1905-13. doi: 10.1136/annrheumdis-2013-203249. Epub 2013 Jul 19. PMID: 23873876.
- Kerschbaumer A, Sepriano A, Bergstra SA, Smolen JS, van der Heijde D, Caporali R, Edwards CJ, Verschueren P, de Souza S, Pope JE, Takeuchi T, Hyrich KL, Winthrop KL, Aletaha D, Stamm TA, Schoones JW, Landewé RBM. Efficacy of synthetic and biological DMARDs: a systematic literature review informing the 2022 update of the EULAR recommendations for the management of rheumatoid arthritis. Ann Rheum Dis. 2023 Jan;82(1):95-106. doi: 10.1136/ard-2022-223365. Epub 2022 Nov 11. PMID: 36368906.
- Ramiro S, Sepriano A, Chatzidionysiou K, Nam JL, Smolen JS, van der Heijde D, Dougados M, van Vollenhoven R, Bijlsma JW, Burmester GR, Scholte-Voshaar M, Falzon L, Landewé RBM. 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. 2017 Jun;76(6):1101-1136. doi: 10.1136/annrheumdis-2016-210708. Epub 2017 Mar 15. PMID: 28298374.
- Riley TR, George MD. Risk for infections with glucocorticoids and DMARDs in patients with rheumatoid arthritis. RMD Open. 2021 Feb;7(1):e001235. doi: 10.1136/rmdopen-2020-001235. PMID: 33597206; PMCID: PMC7893655.
- Sepriano A, Kerschbaumer A, Bergstra SA, Smolen JS, van der Heijde D, Caporali R, Edwards CJ, Verschueren P, de Souza S, Pope J, Takeuchi T, Hyrich K, Winthrop KL, Aletaha D, Stamm T, Schoones JW, Landewé RBM. Safety of synthetic and biological DMARDs: a systematic literature review informing the 2022 update of the EULAR recommendations for the management of rheumatoid arthritis. Ann Rheum Dis. 2023 Jan;82(1):107-118.
- Sepriano A, Kerschbaumer A, Smolen JS, van der Heijde D, Dougados M, van Vollenhoven R, McInnes IB, Bijlsma JW, Burmester GR, de Wit M, Falzon L, Landewé R. Safety of synthetic and biological DMARDs: a systematic literature review informing the 2019 update of the EULAR recommendations for the management of rheumatoid arthritis. Ann Rheum Dis. 2020 Jun;79(6):760-770. doi: 10.1136/annrheumdis-2019-216653. Epub 2020 Feb 7. PMID: 32033941.
- Seror R, Lafourcade A, De Rycke Y, Pinto S, Castaneda J, Fautrel B, Mariette X, Tubach F. Risk of malignancy in rheumatoid arthritis patients initiating biologics: an historical propensity score matched cohort study within the French nationwide healthcare database. RMD Open. 2022 Jun;8(2):e002139. doi: 10.1136/rmdopen-2021-002139. PMID: 35738803; PMCID: PMC9226991.
- Singh JA, Cameron C, Noorbaloochi S, Cullis T, Tucker M, Christensen R, Ghogomu ET, Coyle D, Clifford T, Tugwell P, Wells GA. Risk of serious infection in biological treatment of patients with rheumatoid arthritis: a systematic review and meta-analysis. Lancet. 2015 Jul 18;386(9990):258-65. doi: 10.1016/S0140-6736(14)61704-9. Epub 2015 May 11. PMID: 25975452; PMCID: PMC4580232
Evidence tabellen
Evidence table for systematic review of RCTs and observational studies (intervention studies)
Research question: What are the benefits/harms of treatment with targeted synthetic (ts)DMARD’s compared to treatment with biological (b)DMARD’s or conventional synthetic (cs)DMARD’s in patients with rheumatoid arthritis (RA), psoriatic arthritis (PsA) or spondylarthritis (SpA)
Study reference |
Study characteristics |
Patient characteristics |
Intervention (I) |
Comparison / control (C)
|
Follow-up |
Outcome measures and effect size |
Comments |
EULAR Sepriano, 2023
|
Type of study: SR without meta-analysis
Databases searched: MEDLINE, Embase, Web of Science and The Cochrane CENTRAL Register of Controlled Trials (Central)
Setting and Country: Worldwide. For details, see article and supplementary files.
Included studies: Studies were heterogeneous, precluding data pooling, and results are presented descriptively. -59 observational studies - 30 RCTs
Observational studies - 51 assessed only 1 outcome, 8 addressed ≥2 outcomes - 27 studies evaluating the risk of infections; 23 included patients on bDMARD’s; of which also patients on JAKi; 3 only patients on csDMARD’s; and 1 patients either on tofacitinib or on csDMARD’s. - 9 studies evaluated the risk of malignancies with bDMARD’s; and one of these also with tofacitinib. - 13 studies assessed the risk of MACE; with 11 including patients on bDMARD’s; 6 of which included patients on JAKi; and 2 had patients only on csDMARD’s. - Intestinal perforations and neuroinflammatory events were assessed in two studies, each in patients on bDMARD’s. - All-cause mortality with bDMARD’s was assessed in seven studies. - Seven studies addressed withdrawals due to adverse events with bDMARD’s and one with JAKi. - One study assessed any serious adverse events, another any adverse event, both in patients with bDMARD’s, - one evaluated pregnancy outcomes in patients on bDMARD’s.
RCTs - 11 RCTs evaluated bDMARD’s and 19 RCTs evaluated tsDMARD’s. - Most RCTs were not designed, and therefore, not powered, to evaluate safety outcomes. - The incidence of major adverse events was low and, mostly comparable between active treatment, placebo or active comparator. - The exception was the ORAL-Surveillance study, a non-inferiority trial in which patients ≥50 years old who failed methotrexate and had ≥1 cardiovascular risk factor were randomised to tofacitinib 5 mg two times per day, tofacitinib 10 mg two times per day, or TNFi (adalimumab or etanercept). The trial was designed to test whether the upper limit of the 95% CI around the risk ratio of MACE or malignancies for tofacitinib (5 mg and 10 mg two times per day combined) compared with TNFi, was below 1.8 (the non-inferiority question). - In addition, the ENTRACTE trial, which had a similar design and non-inferiority margin, compared the risk of MACE (primary endpoint) between tocilizumab and etanercept.
Source of funding and conflicts of interest: Funding European Alliance of Associations for Rheumatology. Competing interests: An extensive list of competing interests is reported in the article. |
Inclusion criteria SR: - No language restrictions - Publications from 1 January 2019 to 14 January 2022, as an update of the previous SLR. - The literature search addressed the safety of DMARD’s. - Participants were adults (≥18 years old) with a clinical diagnosis of RA. - Studies including patients with other diagnoses were eligible only if results from patients with RA were presented separately. - Observational studies, namely cohort studies/registries with >50 cases were the main study type. - RCTs with a primary safety outcome were included. In addition, RCTs and long-term extensions (LTEs), selected in the accompanying SLR addressing efficacy,9 were also included to assess the safety of drugs without, or with limited real-world data available. - Included safety outcomes are mentioned in the outcome measures and effect size column. - For the risk of infection by SARS-CoV-2, only studies published after 31 May 2021 (the limit date of an SLR informing EULAR recommendations focusing on the topic) were considered.
Exclusion criteria SR: Studies on glucocorticoids were excluded, as they were dealt with in a separate SLR.
Important patient characteristics at baseline: For details, see article and supplementary files.
Groups comparable at baseline? For details, see article and supplementary files. |
Describe intervention: The intervention was any DMARD (csDMARD, bDMARD—including biosimilars—or tsDMARD), including all drugs (chloroquine, hydroxychloroquine, leflunomide, methotrexate, sulfasalazine, abatacept, anakinra, adalimumab, brodalumab, certolizumab pegol, etanercept, golimumab, guselkumab, infliximab, ixekizumab, mavrilimumab, ocrelizumab, ofatumumab, olokizumab, otilimab, rituximab, sarilumab, sirukumab, tabalumab, tocilizumab, ustekinumab, apremilast, baricitinib, decernotinib, evobrutinib, fenebrutinib, filgotinib, fostamatinib, peficitinib, ruxolitinib, tofacitinib, upadacitinib), formulations and duration. |
Describe control: Studies were only eligible if they included a comparator group (either another DMARD, combination therapy, or the general population). Studies on glucocorticoids were excluded, as they were dealt with in a separate SLR. |
End-point of follow-up: Follow-up time per included study: For details, see article and supplementary files.
For how many participants were no complete outcome data available? For details, see article and supplementary files.
|
The following safety outcomes were considered:
infections (including serious infections, opportunistic infections such as tuberculosis (TB) and herpes zoster (HZ), risk of infection by SARS-CoV-2)
malignancies
major adverse cardiovascular events (MACEs)
venous thromboembolism (VTE) including pulmonary embolism/deep venous thrombosis
Furthermore: Mortality, changes in lipid levels, elevations of creatine phosphokinase, impairments in renal function, elevations of liver enzymes, haematological abnormalities, gastrointestinal side effects, demyelinating disease, induction of autoimmune disease, teratogenicity, fertility and pregnancy outcomes.
For detailed results per outcome, see article and supplementary files.
|
Risk of bias (high, some concerns or low): For details, see article and supplementary files.
Brief description of author’s conclusion: The safety profile of bDMARD’s was further demonstrated. Whether the difference in incidence of malignancies, MACE and VTE between tofacitinib and TNFi applies to other JAKi needs further evaluation. WHAT THIS STUDY ADDS ⇒ The risk of malignancies and major adverse cardiovascular events (MACEs) is similar, or even decreased, with bDMARD’s compared with conventional synthetic (cs)DMARD’s. ⇒ Malignancies and MACE occurred more with tofacitinib than with TNFi in patients who had certain cardiovascular risk factors, especially in patients older than 65 years of age. ⇒ Herpes zoster is more common with JAKi than with csDMARD’s or bDMARD’s. ⇒ Lower intestinal perforations are rare but occur more often with tocilizumab than with other bDMARD’s.
GRADE (per comparison and outcome measure) including reasons for down/upgrading: Not specified. |
Evidence table for intervention studies randomized controlled trials and non-randomized observational studies
Research question: What are the benefits/harms of treatment with targeted synthetic (ts)DMARD’s compared to treatment with biological (b)DMARD’s or conventional synthetic (cs)DMARD’s in patients with rheumatoid arthritis (RA), psoriatic arthritis (PsA) or spondylarthritis (SpA)
Study reference |
Study characteristics |
Patient characteristics 2 |
Intervention (I) |
Comparison / control (C) 3
|
Follow-up |
Outcome measures and effect size 4 |
Comments |
Fang, 2022 Various relevant outcomes
|
Type of study: retrospective cohort study
Setting/Source and country: NHI Research Database (NHIRD) and Taiwan Death Registry (TDR). Taiwan.
Funding and conflicts of interest: Funding from Chang Gung Memorial Hospital (CMRPG3K0041, CMRPG3K0042). No competing interests. |
Inclusion criteria: - Newly diagnosed RA (ICD-9- CM: 714.0 and ICD-10-CM: M05-06) patients identified from 1995 to 2017. Only RA patients with a catastrophic illness certificate, issued after a formal review by an expert panel commissioned by the NHI Administration, were enrolled. - Patients that received JAKi or TNFi 2015-2017
Exclusion criteria: - Never received cDMARD’s - Never received JAKi and TNFi - Aged<18 or aged>80 - Received TNFi before 2015 or after 2017
N total at baseline: 3179 Intervention: 822 Control: 2357
Important prognostic factors2: age ± SD: I: 56.02 (12.11) C: 55.81 (12.68)
Sex: I: % M 20.45 C: % M 21.91
Groups comparable at baseline? Yes |
Describe intervention (treatment/procedure/test): JAKi (tofacitinib)
|
Describe control (treatment/procedure/test): TNFi (etanercept, adalimumab, or golimumab)
|
Length of follow-up: Follow-up was performed from the first use of JAKis or TNFis until the first occurrence of the individual study outcomes, switch to other agents, or December 31, 2018, whichever came first.
|
Relevant measures and effect size: Coronary heart disease I Incidence (100 PY) 0.48 (0.20-0.92) C Incidence (100 PY) 0.45 (0.26-0.64) HR (95% CI) 1.03 (0.45-2.36) P value .9463 Stroke I Incidence (100 PY) 0.33 (0.11-0.72) C Incidence (100 PY) 0.46 (0.27-0.65) HR (95% CI) 0.75 (0.29-1.94) P value .5519 Overall venous thromboembolism I Incidence (100 PY) 0.32 (0.11-0.70) C Incidence (100 PY) 0.48 (0.29-0.68) HR (95% CI) 0.65 (0.25-1.70) P value .3810 Deep vein thrombosis I Incidence (100 PY) 0.26 (0.07-0.62) C Incidence (100 PY) 0.44 (0.26-0.63) HR (95% CI) 0.57 (0.20-1.64) P value .3010 Tuberculosis I Incidence (100 PY) 0.29 (0.09-0.66) C Incidence (100 PY) 0.49 (0.29-0.68) HR (95% CI) 0.60 (0.22-1.62) P value .3122 Malignancy I Incidence (100 PY) 0.39 (0.14-0.80) C Incidence (100 PY) 0.35 (0.18-0.51) HR (95% CI) 1.10 (0.44-2.78) P value .8353 All-cause mortality I Incidence (100 PY) 1.17 (0.64-1.70) C Incidence (100 PY) 1.29 (0.97-1.61) HR (95% CI) 0.91 (0.54-1.52) P value .7099 |
Article conclusion: Comparable safety issues and mortality rates were observed for JAKis and TNFis in RA patients treated in real-world settings.
Sensitivity analyses: The incidence rates of safety outcomes (CHD, stroke, OVT, DVT, TB, THR, TKR, malignancy, and all-cause mortality) were higher in those aged ≧50 than in the entire sample. After PSSW, an insignificant difference in these incidence rates between the 2 medication groups was seen in those aged ≧50. |
Frisell, 2023 Various relevant outcomes
|
Type of study: retrospective cohort study Setting/Source and country: ARTIS safety monitoring programme linking individual-level longitudinal data on treatments, disease activity and other clinical measurements from the Swedish Rheumatology Quality Register (SRQ) to prospectively collected data in Swedish national healthcare registers ( National Patient Register, Prescribed Drug Register, census /taxation registers). Sweden
Funding and conflicts of interest: TF was supported by the Swedish Research Council (2016-00398, 2021-01418). JA was supported by the Swedish Research Council, Region Stockholm/Karolinska Institutet (ALF), the Swedish HeartLung-Foundation, the Swedish Cancer Society, Nordforsk and Vinnova. |
Inclusion criteria: all patients with RA in Sweden who were recorded as starting any b/ tsDMARD between 1 January 2010 and 31 December 2020. All approved b/tsDMARD’s used for RA in Sweden during the study period were included.
Exclusion criteria: Drugs with fewer than 200 treatment episodes (here: anakinra (n=84) and upadacitinib (n=105)) were excluded from further analysis.
N total at baseline: 20 117 unique patients with RA (contributing a total of 34 279 treatment episodes) Intervention: JAKi: BAR 1837 TOFA 426
bDMARD’s: ADA 5526 IFX 2971 CZP 2179 GOL 1889 ABA 3434 RTX 4220 TCZ 2757 SAR 292
Control: ETA 8748
Important prognostic factors2: age ± SD: I: JAKi: BAR 61 (14); TOFA 59 (13) bDMARD’s: 58 (14); 58 (14); 56 (15;) 57 (14); 61 (13); 64 (13;) 59 (14); 59 (14) C: 58 (14)
Sex: I: % M BAR 18; TOFA 18; bDMARD’s between 26 and 20% C: % M 23
Groups comparable at baseline? No, but weighting successfully balanced mean patient characteristics across the major treatment groups, it was not possible to reach acceptable balance for all variables in the two smallest groups (sarilumab, tofacitinib). Adjustment directly in multivariable Cox regressions gave very similar estimates throughout. |
Describe intervention (treatment/procedure/test): JAKi: baricitinib, tofacitinib and upadacitinib.
TNFi: adalimumab, certolizumab pegol, golimumab and infliximab.
Other bDMARD’s: abatacept, anakinra, rituximab, sarilumab, tocilizumab
|
Describe control (treatment/procedure/test): TNFi: etanercept, |
Length of follow-up: - Max.l study duration 1 January 2010 - 30 June 2021. - Patients were considered exposed to a treatment from their first ever start of that specific b/tsDMARD, as recorded in the SRQ, until treatment switch or discontinuation. - When a patient switched or discontinued treatment, a lag time of 90 days was added after the treatment was stopped (183 days for rituximab) to capture adverse events linked to treatment discontinuation but registered with some delay. If restarted within 90 days (183 days for rituximab), the two treatment episodes were merged. -Swithching between biosimilars was not considered as discontinuation. - Patients could contribute with multiple treatment episodes on different drugs, but only the first ever start for each molecule. - Follow-up was censored at death or first emigration from Sweden after treatment start.
Loss-to-follow-up and incomplete outcome data: Data were complete on treatments, outcomes and most covariates derived from national registers, but about 30% lacked data on baseline DAS28 and HAQ. Missing covariate data were accounted for by multiple imputation. Due to differences in market entry, the average FU pp was +/- 3 yrs for most bDMARD’s, below 2 yrs for JAKi & lowest for SAR, 1.3 yrs. |
Relevant measures and effect size: Only results for JAKi BAR and TOFA vs ETA are presented here. The article also includes results for other TNFi/bDMARD’s vs ETA. Article also mentions results for other outcomes (i.e., treatment discontinuation due to adverse events, liver disease, depression, suicide, hospitalisation)
Crude IR/1000 PY; weighted IR/1000 PY; adjusted HRs (95%CI) vs etanercept
MACE BAR 10.7; 9.9; 0.83 (0.49-1.42) TOFA 12.9; 9.2; 0.78 (0.31-1.99) ETA 10.1; 12.3; 1.0 (ref)
Serious infection BAR 39.5; 33.6; 0.98 (0.75-1.29) TOFA 47.3; 30.5; 0.89 (0.47-1.69) ETA 24.8; 30.8; 1.0 (ref)
Diagnosed herpes zoster BAR 10.0; 9.8; 3.82 (2.05-7.09) TOFA 14.1; 10.2; 4.00 (1.59-10.06) ETA2.0; 2.5; 1.0 (ref)
All-cause mortality BAR 18.8; 22.0; 2.27 (1.51-3.40) TOFA 8.7; 10.5; 1.09 90.40-2.92) ETA 8.8; 12.1; 1.0 (ref)
|
Article conclusion: - Data from ARTIS supports that the b/tsDMARD’s currently used to treat RA have acceptable and largely similar safety profiles, but differences exist in particular concerning tolerability and specific infection risks. - few significant differences were observed for the serious adverse events under study. Neither CV events nor general serious infections were more frequent on BAR or TOFA versus bDMARD’s, but JAKi were associated with almost four times higher rates of hospital-treated HZ (vs ETA). - Mortality rate was similar across TNFi, but about 30%–40% higher on the non-TNFi bDMARD’s, with the numerically highest HR seen for baricitinib.
NB: Low number of events limited some comparisons, in particular for sarilumab and tofacitinib.
Sensitivity analyses: - Restricting the study period to the time after JAKi market entry reduced sample sizes drastically, and several differences between non-TNFi bDMARD’s and etanercept were no longer significant, but it did not materially alter the comparison between the JAKi and etanercept. - Ending follow-up at the onset of the COVID-19 pandemic had little impact on incidence rates or contrasts between bDMARD’s, but the reduced sample size for sarilumab and JAKi excluded them from most comparative analyses for these groups. |
Hirose, 2022 Various relevant outcomes
|
Type of study: multicentre, longitudinal observational study with both retrospective and prospective data
Setting and country: 12 hospitals and clinics for rheumatology in Japan
Funding and conflicts of interest: No specific funding was received from any bodies in the public, commercial or not-for-profit sectors to carry out the work described in this article. Five out of the ten authors have an extensive list of conflicts of interest / liaisons with pharmaceutical companies. |
Inclusion criteria: - patients aged ≥20 years -who fulfilled the 2010 ACR/EULAR classification criteria for RA -who started treatment with TOF or ABT between January 2015 and January 2021. - who had disease activity that was not controlled by MTX or csDMARD’s, or were unable to be treated with csDMARD’s, including MTX
- 2015-2017 retrospective data from patients’ medical records - 2018-jan. 2021 prospective data
Exclusion criteria: non stated. Only that prior use of bDMARD’s or JAKi was not an exclusion criterium.
N total at baseline: 370 Intervention: 187 Control: 183
Important prognostic factors2: age ± SD: I: 66.2 (11.5) C: 70.7 (11.3)
Sex: I: % M 15.5 C: % M 15.8
Groups comparable at baseline? After IPTW, yes |
Describe intervention (treatment/procedure/test): Tofacitinib (TOFA) Patients with an estimated glomerular filtration rate >60 ml/ min/1.73m2 received 5mg of TOF orally twice daily, whereas those with estimated glomerular filtration rate <60 ml/min/ 1.73m2 received 5mg of TOF orally once daily.
|
Describe control (treatment/procedure/test): Abatacept (ABT) ABT was administered as an i.v. infusion (500 mg for patients weighing <60 kg, 750mg for 60–100 kg, and 1000mg for >100 kg) at weeks 0, 2 and 4, then every 4 weeks thereafter. Alternatively, patients received 125mg by s.c. injection once weekly.
|
Length of follow-up: 52 weeks
Discontinuation reasons at week 4, 12, 26 and 52 Loss-to-follow-up: Intervention: N (%) 32 (17) Reasons (describe): 2 consent withdrawal, 2 hospital transfer, 12 lack of efficacy, 1 concern for drug interaction, 1 clinical remission, 1 abdominal pain, 1 headache, 1 infectious arthritis of elbow, 1 non-tuberculous mycobacteria disease, 1 fever, 1 diverticulitis of the colon, 1 infectious enteritis, 3 herpes zoster, angina pectoris, 1 bacterial pneumonia, 1 herpes simplex, 1 generalized eczema, 1 elevation of liver enzymes.
Control: N (%) 37 (20) Reasons (describe): 5 consent withdrawal, 2 hospital transfer, 10 lack of efficacy, 3 clinical remission, 1 nausea, 1 skin rash, 1 cough, 1 fatigue, 2 gastrointestinal bleeding, 1 skin ulcer, 1 bacterial pneumonia, 1 COVID-19 pneumonia, 1 vertebral fracture, 1 bronchial asthma, 1 infectious enteritis, 1 pulmonary non-tuberculous mycobacterial infection, 1 eczema, 1 malignant lymphoma, 1 stomatitis, 1 gynecomastia
Incomplete outcome data: The last observation carried forward method was used for patients who discontinued treatment before week 52 to include all patients in the analysis. |
Relevant measures:
Serious infection, n (%) I: 4 (2.1) C: 4 (2.2) P-value: 1.00 Herpes zoster, n (%) I: 17 (9.1) C: 5 (2.7) P-value: 0.014 Cancer, n (%) I: 0 (0) C: 1 (0.5) P-value: 1.00 MACE, n (%) I: 2 (1.1) C: 1 (0.5) P-value: 0.49 VTE, n (%) I: 0 (0) C: 1 (0.5) P-value: 0.49 |
Conclusion with regard to the relevant safety comparisons as stated in the article: “The incidence of herpes zoster was significantly higher in the TOF group than in the ABT group (9.1 vs 2.7%, P=0.014). Three patients, including one case of grade 3, discontinued TOF prematurely owing to herpes zoster. No significant differences were noted in the incidence of serious infections, cancer or major adverse cardiovascular and venous thromboembolic events between the two treatment groups.”
Note: The primary outcome was the remission rate at week 52 in each group. Analyses and results regarding the comparative effectiveness are described in more detail/length than safety.
Possible bias: between group differences in retrospective and prospective collected data? No information on numbers. Also, in general, but especially for the prospective data, it is unclear how (on what grounds, taking into account data already collected retrospectively, matching on covariates?) patients were assigned to a treatment group. |
Hoisnard, 2022 Cardiovascular outcomes (MACE, VTE)
|
Type of study: nationwide population-based cohort study
Setting/Source and country: French national health data system France
Funding and conflicts of interest: Funding statement: The authors have not declared a specific grant for this research from any funding agency in the public, commercial or not-for- profit sectors. Competing interests: None declared.
|
Inclusion criteria: - All adults (≥ 18 years old) with at least one dispensation of a JAKi between 1 July 2017 and 31 May 2021 were eligible for inclusion in the exposed group. - The unexposed population had to present RA severity comparable to that of the exposed cohort and use adalimumab. - Patients with RA had recorded within the 5 years before the index date: (1) at least one hospitalisation with ICD-10 code as a principal, related or associated diagnosis specific for RA, and (2) long-term disease status with an ICD-10 code specific for RA. - Patients who were JAKi-naive and adalimumab-naïve (new users), defined as those who had not filled a prescription for one of these drugs for 1 year.
Exclusion criteria: None specified in article.
N total at baseline: Intervention: JAKi N=8481, Tofacitinib, N=3416, Baricitinib, N=5065 Control: Adalimumab) N=7354
Important prognostic factors2: age ± SD: I: 59.3 (13.3) C: 55.3 (13.4)
Sex: I: % M 21.7 C: % M 28.8
Groups comparable at baseline? No, but after IPTW yes. |
Describe intervention (treatment/procedure/test): JAKi, baricitinib 2 or 4 mg daily, tofacitinib 5 mg or 10 mg two times per day |
Describe control (treatment/procedure/test): adalimumab
|
Length of follow-up: Patients were followed up to the occurrence of each event (MACE and VTE), death from any cause, exposure discontinuation or 31 December 2021, whichever came first.
Discontinuation date was the date corresponding to 120 days after the last reimbursement.
Only the first therapeutic sequence of a JAKi or adalimumab was considered in this analysis.
Follow-up duration (days) Median (IQR) JAKi 440 (203–846) ADA 344 (185–686)
Loss-to-follow-up: - Incomplete outcome data: -
|
Relevant measures and effect size:
MACE N events; IR (95%CI)/1000PY; N acute myocardial infarctions; N ischaemic strokes JAKi 54; 4.3 (3.1 to 5.4); 29; 28 TOFA 14; 2.8 (1.4 to 4.3); 8; 6 BAR 40; 5.2 (3.6 to 6.8); 21; 22 ADA 35; 3.6 (2.4 to 4.9); 24; 11
Risk of MACEs JAKi vs ADA HRw 1.0 (0.7 to 1.5) (p=0.99)
VTE N events; IR (95%CI)/1000PY; N pulmonary embolisms; N venous embolism and thrombosis JAKi 75; 6.0 (4.6 to 7.3); 41 34 TOFA 29; 5.9 (3.7 to 8.0); 12 17 BAR 46; 6.0 (4.3 to 7.7); 29 17 ADA 32; 3.3 (2.2 to 4.5); 14 18
Risk of VTEs JAKi vs ADA: HRw 1.1 (0.7 to 1.6) (p=0.63) |
Article conclusion: This study provides reassuring data regarding the risks of MACEs and VTEs in patients initiating a JAKi versus adalimumab, including patients at high risk of cardiovascular diseases.
Subgroup analyses: After weighting with the IPTW method, risk of MACEs and VTEs was not significant whatever the subgroup (JAKi subgroups, age subgroups with CV risk factor, gender subgroups).
Sensitivity analyses were also not significant (death as competing risk analysis, a conventional multivariate Cox model with covariates and time-varying MTX [as in main analyses], and analysis with a broader definition of MACEs). |
Huss, 2023 Outcome malignancies
|
Type of study: prospective observational cohort study
Source and country: The Swedish Rheumatology Quality register (SRQ), the National Patient Register, the Longitudinal Database for Insurance and Labor Market Studies (LISA), the Prescribed Drug Register, the Population and Cause of death register and the National Cancer register.
Funding and conflicts of interest: - This work was supported by funding from the Karolinska Institute Region Stockholm funds (ALF), the Swedish Research Council, the Swedish Cancer Society and the Swedish HeartLung Foundation. TF was supported by the Swedish Research Council (2021-01418). Funders had no impact on the design or interpretation of the study or its results. - Competing interests: VH and KH have no competing interests to declare. JA has had or have research agreements with Abbvie, Astra-Zeneca, BMS, Eli Lilly, MSD, Pfizer, Roche, Samsung Bioepis, Sanofi, and UCB, mainly in the context of safety monitoring of biologics via ARTIS/Swedish Biologics Register. TF and HB are partly employed by the ARTIS project. |
Inclusion criteria: -individuals above 18 years of age registered with RA or PsA in SRQ - treatment initiations among patients with RA or PsA with: (1) TNFi, (2) non-TNFi bDMARD or (3) JAKi - included treatment initiations (TNFi and non-TNFi) from 2016. - For JAKi, treatment initiations from their market entry in 2017
Exclusion criteria: - Because of the new-user design, treatment episodes initiated before or ongoing at the start of the study period were not included (but contributed to the number of previously used b/tsDMARD’s). - participants who had a history of any previous cancer other than NMSC were excluded from all analyses.
RA - N total at baseline: 10447 Intervention: (1) 1967 JAKi (2) 3520 non-TNFi Control: (3) 7343 TNFi
PsA - N total at baseline: 4443 Intervention: (1) 379 (2) 185 Control: (3) 4186
Important prognostic factors2: RA age (IQR): I: (1) 59 (50–69) (2) 60 (51–70) C: (3) 56 (46–67) (4) 57 (47–68)
Sex: I: % M (1) 18 (2) 21 C: % M (3) 22 (4) 22
PsA age (IQR): I: (1) 52 (45–61) (2) 54 (45–62) C: (3) 50 (41–59) (4) 49 (41–59)
Sex: I: % M (1) 29 (2) 29 C: % M (3) 45 (4) 46
Groups comparable at baseline? Not on all possible confounders, but analyses were corrected for all possible confounders. |
Describe intervention (treatment/procedure/test):
(1) JAKi (baricitinib, tofacitinib, upadacitinib).
(2) non-TNFi bDMARD (rituximab, abatacept, tocilizumab, sarilumab)
|
Describe control (treatment/procedure/test):
(3) TNFi (adalimumab, certolizumab pegol, etanercept, golimumab, infliximab)
(4) General population (For each patient, five general population comparator subjects were identified, free from RA/PsA at their index case’s first registration of RA/PsA and matched on age, sex and region of domicile, from the Population register) |
Length of follow-up: - Start of follow-up was defined as the date of treatment start with each b/tsDMARD’s during the study period. - Authors used an ever-treated approach, in which each patient in each treatment cohort was followed from every treatment initiation until the occurrence of the outcome, death, emigration from Sweden or end of the study period on 31 December 2020. - The total person time at risk in the JAKi, non-TNFi bDMARD and TNFi cohorts was 585, 418 and 12 623 patient years, respectively. The median follow-up times were 1.52, 2.25 and 2.44 years.
Loss-to-follow-up: - Incomplete outcome data: -
Hazard Ratio analyses were adjusted for (1) age, sex, line of therapy; (2) comorbidities and SES => for these variables, there were no missing values (3) for disease-related factors, => missing values => complete case approach.
Imputation models were adjusted for all covariates included in the analysis model plus the event indicator and the Nelson-Aalen estimate of the cumulative hazard.
|
Outcome measures and effect size: - Main outcome: All cancers other than NMSC. And authors defined 19 sites of cancer as 19 individual outcomes. - Authors only performed comparative analyses (i.e., estimated HRs) where the number events in each cohort were ≥5. - In the JAKi cohort, there were 7 breast cancers (HR=0.73, 95% CI 0.29 to 1.86, vs TNFi), 6 haematopoietic (HR=1.90, 95% CI 0.70 to 5.16) and 7 lung cancers (HR=1.15, 95% CI 0.57 to 2.32). - For all other sites other than NMSC, there were less than five events observed among the JAKi treated and thus are not presented.
Beneath, per outcome, per cohort: Events; PY, no (only for RA patients); Crude IR/1000 PY; Standardised IR/1000 PY; Fully adjusted HR (95%CI)
RA patients Outcome: All cancers other than NMSC Cohort: All JAKi 38; 3996; 9.5; 8.3; 0.94 (0.65 to 1.38) Tofacitinib 8; 793; 10.1; 11.2; 1.08 (0.52 to 2.24) Baricitinib 30; 3187; 9.4; 8.0 ; 0.92 (0.61 to 1.38) Non-TNFi bDMARD 141; 11 051; 12.8; 10.5; 1.12 (0.88 to 1.43) TNFi 213; 21 122; 10.1; 10.1; 1.0 (Reference) General population 1245; 128 224; 9.7; 9.2; n/a
Outcome: NMSC Cohort: All JAKi 59; 3954; 14.9; 12.9; 1.39 (1.01 to 1.91) Tofacitinib 11; 781; 14.1; 14.9; 1.56 (0.83 to 2.92) Baricitinib 48; 3157; 15.2; 12.5; 1.37 (0.97 to 1.92) Non-TNFi bDMARD 126; 11 027; 11.4; 9.0; 1.00 (0.78 to 1.28) TNFi 189; 21 083; 9.0; 9.0; 1.0 (Reference) General population 852; 128 630; 6.6; 6.2; n/a
PsA patients All cancers other than NMSC JAKi 5; 8.6; 7.3; 1.88 (0.68 to 5.16) Non-TNFi 2; 4.8; –; – TNFi 73; 5.8; 5.8; 1.0 (Reference) Gen population 317; 5.9; 6.0; n/a NMSC JAKi 8; 13.9; 11.7; 2.05 (0.79 to 5.31) Non-TNFi 2; 4.8; –; – TNFi 73; 5.8; 5.8; 1.0 (Reference) Gen population 209; 3.9; 3.9; n/a |
Article conclusion: among individuals with RA or PsA, we found no evidence of an increased short-term risk of all cancers other than NMSC for patients initiating JAKi compared with TNFi, but the risk of NMSC may be increased, at least in patients with RA.
Note: Patients that switched to a second drug of the same exposure category within the study period contributed twice to that cohort.
Study included additional analyses: (1) with fitted models by time since treatment initiation (≤1, 1–2, ≥2 years) by inclusion of an interaction term and thereby relaxed the proportional hazards assumption. (2) analyses by previous use of b/tsDMARD’s (0, 1–2, ≥3). (3) with latency period of 90 days, so that only cancers diagnosed 90 days or later after treatment start would contribute to analyses. (4) restricted the main analysis to a CV-enriched subset of the JAKi, non-TNFi and TNFi cohort (5) a sensitivity analysis using an on-drug approach for all cancers other than NMSC and NMSC, respectively, for patients with RA. (6) a sensitivity analysis restricting the follow-up to Feb 2020 (for patients with RA) because the study period ended in December 2020 and the COVID pandemic could theoretically affect the results.
With regard to these additional analyses: They did not appreciably vary from or alter the (fully adjusted HR) results of the main analysis |
Jeong, 2022
Infections (serious, herpes zoster)
|
Type of study:
Setting/Source and country: Korean Health Insurance Review & Assessment Service database. South Korea
Funding and conflicts of interest: This study was supported by the Korea Health Technology R&D Project through the Korea Health Industry Development Institute funded by the Ministry of Health Welfare, Republic of Korea (grant number HI14C1277). The authors declare that they have no competing interests. |
Inclusion criteria: patients with seropositive RA who were prescribed bDMARD’s or tofacitinib between January 2010 and January 2019
Exclusion criteria: - those who were prescribed bDMARD’s and tsDMARD in 2010 to limit the study population to new users - patients with non-seropositive RA - juvenile RA - age <20 years - HZ within 1 month of bDMARD or tsDMARD use - HZ within 6 months of previous HZ
N total at baseline: 11720 Intervention: TOFA 701 Control: ABA 1153; ETA 2680
Important prognostic factors2: age IQR: I: TOFA 56 (47–63) C: ABA 60 (52–68) C: ETA 55 (45–64)
Sex: I: % M TOFA 18.3 C: % M ABA 19.8 C: % M ETA 19.3
Groups comparable at baseline? No, probably not but analyses were adjusted for potential confounders. |
Describe intervention (treatment/procedure/test): tofacitinib
infliximab, adalimumab, golimumab, tocilizumab, rituximab, |
Describe control (treatment/procedure/test): abatacept or etanercept
|
Length of follow-up: Follow-up investigations were performed for all patients until drug failure, development of HZ, or January 31, 2019, whichever occurred earlier.
Patients with longer than 1 year of refill period from the date of the last drug prescription were censored at the date of the last drug prescription. The drug failure-free survival was calculated for each individual to define person-days of bDMARD’s or tsDMARD treatment for the follow-up investigation.
Loss-to-follow-up: -
Incomplete outcome data: -
Patients with missing data, such as variables for adjustment, were excluded from the study.
|
Relevant measures and effect size:
Risk of HZ on RA patients during first bDMARD or tsDMARD use IR/1000PY TOFA 100.4 ABA 59.7 ETA 43.4
aHR (95% CI); p-value – ABA as reference TOFA 2.46 (1.61–3.76); <0.001 ETA 1.19 (0.94–1.51); 0.146
aHR (95% CI); p-value - ETA as reference TOFA 2.06 (1.38–3.08); <0.001 ABA 0.84 (0.66–1.06); 0.146
Risk of recurrent HZ on RA patients during first bDMARD or tsDMARD use IR/1000PY TOFA 185.6 ABA 83.8 ETA 70.7
aHR (95% CI); p-value – ABA as reference TOFA 3.69 (1.77–7.69) <0.001 ETA 1.22 (0.75–2.00); 0.421
aHR (95% CI); p-value - ETA as reference TOFA 3.01 (1.49–6.11); 0.002 ABA 0.82 (0.50–1.34); 0.421
After excluding patients who started bDMARD’s or tsDMARD before 2017 to normalize the clinical application period of TOFA in South Korea, TOFA (aHR, 3.26; 95% CI, 1.63–6.51; P < 0.001) showed an increased HZ risk compared to that of ABA. |
Article conclusion: Patients with seropositive RA treated with bDMARD’s or tsDMARD have a high HZ risk. In addition, HZ risk is significantly increased in RA patients with a history of HZ after the initiation of bDMARD’s or tsDMARD, unlike that in the general population where prior HZ may reduce the risk of recurrence. RA patients who received tofacitinib showed higher overall, incident, and recurrent HZ risks than those who received bDMARD’s.
Subgroup analyses: subgroups of tofacitinib, infliximab, and adalimumab showed increased HZ risk compared to that of the corresponding abatacept subgroups. Higher HZ risk associated with Charlson comorbidity index > 2 (in ETA, IFX, ADA, TOFA) and with steroid use ≥ 5mg/day (in IFX, ADA, RTX, TOFA), for TOFA also with female gender, age above or below 65, more or less than 2 csDMARD’s. |
Khosrow-Khavar, 2022
Cardiovascular outcomes (MACE, VTE)
|
Type of study: observational cohort study
Setting/Source and country: claims data from the Optum Clinformatics, IBM MarketScan, and Medicare databases. USA Funding and conflicts of interest: This study was funded by internal sources of the Division of Pharma-coepidemiology and Pharmacoeconomics, Brigham and Women’s Hospital & Harvard Medical School, Boston, MA, USA. Two authors received research grants for unrelated studies. All other authors have no conflict of interests to disclose. |
Inclusion criteria: - patients initiating treatment on tofacitinib or a TNFI (infliximab, adalimumab, certolizumab pegol, etanercept, and golimumab) - Cohort entry date corresponded to first TNFI or tofacitinib dispensation (“index drug”) with a minimum of 365 days of continuous enrollment in health plan prior to and including the cohort entry date. - Patients required at least two diagnosis codes for RA in any setting during the 365 days baseline period (between 7 and 365 days apart)
Exclusion criteria: - TNFI users with a prescription of index TNFI and tofacitinib users with prescription of tofacitinib in the 365 days prior to cohort entry date; - patients with a prescription of tofacitinib and TNFI on cohort entry date; - patients missing data on age or gender, and those with admission to nursing facility or hospice on or prior to cohort entry date; - TNFI users with history of use of any JAK inhibitor or with prescriptions for multiple agents from the TNFI class on cohort entry date; - tofacitinib users with prescriptions of other approved JAK inhibitors (i.e. baricitinib or upadacitinib) on or at any point prior to cohort entry date.
From this source population of RA patients initiating treatment with tofacitinib or TNFI two cohorts were created: (1) “real-world evidence (RWE)”, included all RA patients from routine care of ≥ 18 years of age in MarketScan and Optum (≥ 65 in Medicare) at cohort entry date. (2) “RCT-duplicate cohort”, mimicked the inclusion and exclusion criteria of the ORAL surveillance trial. This was restricted to - patients ≥ 50 years of age (65 in Medicare) with ≥ 1 methotrexate dispensation in six months prior to cohort entry date. - and patients with at least one CV risk factor including history of smoking, hypertension, dyslipidemia, diabetes mellitus, ischemic heart disease, or family history of ischemic heart disease. - Patients hospitalized with infections in the 30-days prior to cohort entry date and pregnant patients were excluded.
N total at baseline: RWE cohort, 28568, 34083, and 39612 patients who met the inclusion and exclusion criteria were identified from Optum, MarketScan, and Medicare respectively of whom 13.2%, 15.6%, and 9.5% initiated treatment on tofacitinib RCT-duplicate cohort, 6878, 8071, and 20121 patients were identified from Optum, MarketScan, and Medicare, respectively, of whom 11.6%, 14.3%, and 7.7% initiated treatment with tofacitinib.
Important prognostic factors2: Data on covariates are provided per database per group for the RWE cohort. Age; mean (std) TOFA 56.8 (12.5) TNFi 57.1 (13.2) TOFA 54.7 (11.5) TNFi 55.0 (12.0) TOFA 72.1 (5.6) TNFi 72.2 (5.6)
Female Gender; n (%) TOFA 3043 (80.9) TNFi 20046 (81.2) TOFA 4333 (81.8) TNFi 23503 (81.8) TOFA 3134 (82.9) TNFi 29819 (83.3)
Groups comparable at baseline? Authors assessed 76 potential confounders and IPTW achieved excellent covariate balance in study populations with standardized differences close to zero for all covariates. |
Describe intervention (treatment/procedure/test): tofacitinib |
Describe control (treatment/procedure/test): TNFi (infliximab, adalimumab, certolizumab pegol, etanercept, and golimumab).
|
Length of follow-up: Authors used an as-treated exposure definition whereby patients were followed from treatment initiation for study outcomes until treatment discontinuation or switch, insurance disenrollment, death, or end of the study period, whichever occurred first.
Total person years of follow-up: RWE cohort TOFA 11157 TNFi 80992
RCT-duplicate cohort TOFA 3159 TNFi 29030
Loss-to-follow-up: - Incomplete outcome data: -
|
Relevant measures and effect size: Effect estimates were pooled across three databases using fixed effects model with inverse variance weighting.
Results for primary outcome: composite CV outcome consisting of hospitalizations for myocardial infarction or stroke.
RWE Cohort Crude Incidence Rate (95% CI)/100PY TOFA 1.31 (1.10 to 1.56) TNFi 1.24 (1.16 to 1.33)
Crude Incidence Rate Difference (95% CI)/100PY (TNFi=ref): 0.20 (0.01 to 0.39)
Weighted HR (95% CI)(TNFi=ref): 1.01 (0.83 to 1.23)
RCT-Duplicate Cohort Crude Incidence Rate (95% CI)/ 100PY TOFA 1.83 (1.41 to 2.39) TNFi 1.46 (1.32 to 1.61)
Crude Incidence Rate Difference (95% CI)/100PY (TNFi=ref): 0.46 (0.01 to 0.92)
Weighted HR (95% CI)(TNFi=ref): 1.24 (0.90 to 1.69)* *which aligned closely with ORAL-surveillance results (HR: 1.33, 95% CI: 0.91 to 1.94).
Results for secondary outcomes in the RWE cohort: For individual CV outcomes, the pooled weighted HR (95% CI) was 1.04 (0.82 to 1.33) for MI, 0.93 (0.66 to 1.31) for stroke, 1.07 (0.79 to 1.46) for heart failure hospitalization, and 1.04 (0.78 to 1.40) for coronary revascularization (Supplemental Table 10) when comparing tofacitinib users with TNFI users. The pooled weighted HR (95% CI) was 1.20 (0.98 to 1.46) for all-cause mortality. For the positive control outcome, we successfully replicated the known association between tofacitinib and risk of herpes zoster (pooled weighted HR 1.98, 95% CI: 1.78 to 2.19).
|
Article conclusion: Authors did not find evidence for an increased risk of cardiovascular outcomes with tofacitinib in RA patients treated in the real-world setting; however, tofacitinib was associated with an increased risk of cardiovascular outcomes, albeit statistically non-significant, in RA patients with cardiovascular risk factors.
Subgroup analyses in RWE cohort: Some indication of possible increased risk of CV outcomes for CV history y/n and age subgroups: the pooled weighted HR (95% CI) was 1.27 (0.95 to 1.70) and 0.81 (0.61 to 1.07) among patients with and without history of cardiovascular disease respectively. The pooled weighted HR (95% CI) among patients ≤ 65 years of age was 1.00 (0.66 to 1.50) and 1.05 (0.84 to 1.33) for patient aged more than 65 years. No TOFA-risk association was observed across other subgroups (gender, previous bDMARD). Consistent results were observed across other sensitivity and secondary analyses.
Sensitivity analysis in RCT-duplicate cohort: In sensitivity analysis restricting comparator to adalimumab and etanercept (Supplemental Table 13), the pooled weighted HR (95%) CI for primary CV outcome was 1.32 (0.94 to 1.86). |
Min, 2023
Various relevant outcomes
|
Type of study: retrospective observational study
Source and country: Korean National Health Insurance Service (NHIS) from the National Health Information Database (NHID) were used.
Funding and conflicts of interest: This work was supported by the Konkuk University Medical Center Research Grant 2022 (grant No. K220112). The authors have no conflicts of interest to declare for this study. |
Set 1 newly diagnosed RA patients who were not exposed to either bDMARD’s or JAKis. Set 2 included all RA patients, both bDMARD-naive and bDMARD-exposed.
Inclusion criteria set 1: (1) patients who had an ICD-10 code for seropositive or seronegative RA as their first or second diagnosis between January 1, 2014, and December 31, 2020; (2) used MTX for at least 6 months before initiation of JAKi or TNFi treatment.
Exclusion criteria set 1: (1) RA patients were excluded who had an ICD-10 code for seropositive RA or seronegative RA between January 1, 2014, and December 31, 2016 (2) subjects were excluded who had a diagnosis code for AS, SLE, PS, PsA, Behçet’s disease, Crohn’s disease, or ulcerative colitis as their first or second diagnosis. (3) patients with a main ICD-10 diagnosis code for AMI, stroke, VTE, ATE, or cancer 12 months prior to the initiation of TNFi or JAKi were excluded. (4) Patients below 18 years (5) RA patients who did not use JAKis or TNFis, or who started JAKis/TNFis before July 1, 2017, were excluded.
The inclusion and exclusion criteria for set 2 were identical to those of set 1; however, for set 2, the first exclusion step was not performed. In addition, in the last exclusion step for set 2, RA patients who started JAKis/TNFis before March 1, 2015, were excluded.
Set 1 & Set 2 N total at baseline: Intervention: 645 & 2498 Control: 951 & 9267
Important prognostic factors2: age ± SD: I: 52.30±12.80 & 51.50±12.25 C: 50.16±14.44 & 52.00±13.27
Sex: I: % M 24.5 & 16.8 C: % M 27.7 & 18.1
Groups comparable at baseline? Not on all possible covariates, but analyses were corrected for all covariates. |
Describe intervention (treatment/procedure/test): JAKi (i.e., tofacitinib, baricitinib, or upadacitinib)
|
Describe control (treatment/procedure/test): TNFi (i.e., etanercept, adalimumab, infliximab, or golimumab)
|
Length of follow-up: All subjects were followed until December 31, 2021, or until each primary adverse event occurred, or if switching from JAKi to TNFi or TNFi to JAKi.
Loss-to-follow-up: not described
Incomplete outcome data: not described
The mean total follow-up duration was 2.1 years for the JAKi group and 2.5 years for the TNFi group in set 1, and 2.6 years for the JAKi group and 5.8 years for the TNFi group in set 2.
|
Outcome measures and effect size - incidence rate ratio (95%CI): 1yr FU; total FU - hazard ratio (95%CI), p-value: 1yr FU; total FU
Set 1 AMI, acute myocardial infarction 0.95 (0.41, 2.19); 0.77 (0.40, 1.48) 0.79 (0.33, 1.87), 0.59; 0.65 (0.34, 1.27), 0.21 Stroke 0.27 (0.06, 1.20); 0.44 (0.15, 1.33) 0.31 (0.07, 1.43), 0.13; 0.43 (0.14, 1.33), 0.14 cardiovascular related mortality 1.47 (0.43, 5.09); 1.43 (0.57, 3.63) 0.21 (0.04, 1.21), 0.08; 1.26 (0.47, 3.40), 0.64 MACE 0.71 (0.37, 1.34); 0.69 (0.42, 1.14) 0.64 (0.33, 1.22), 0.18; 0.59 (0.35, 0.99), 0.04 All-cause mortality 1.23 (0.38, 4.03); 1.46 (0.70, 3.03) 0.97 (0.29, 3.28), 0.97; 1.41 (0.65, 3.04), 0.39 VTE, venous thromboembolism NA; 0.36 (0.08, 1.62) NA; 0.33 (0.07, 1.54), 0.16 ATE, arterial thromboembolism NA; 1.79 (0.11, 28.62) NA; 1.58 (0.10, 25.47), 0.75 Cancer (excluding non-melanoma skin cancer) 0.80 (0.30, 2.17); 1.53 (0.81, 2.87) 0.68 (0.25, 1.88), 0.46; 1.29 (0.68, 2.45), 0.44 Non-melanoma skin cancer NA; NA NA; NA
Set 2 AMI, acute myocardial infarction 0.62 (0.41, 0.93); 0.69 (0.53, 0.90) 0.65 (0.43, 0.99), 0.04; 0.67 (0.51, 0.87), <0.01 Stroke 0.55 (0.31, 0.96); 0.74 (0.52, 1.05) 0.60 (0.34, 1.06), 0.08; 0.74 (0.52, 1.06), 0.10 cardiovascular related mortality 1.44 (0.92, 2.27); 1.14 (0.85, 1.54) 1.66 (1.05, 2.63), 0.03; 1.34 (0.98, 1.83), 0.07 MACE 0.74 (0.56, 0.98); 0.84 (0.70, 1.00) 0.81 (0.61, 1.06), 0.13; 0.80 (0.67, 0.97), 0.02 All-cause mortality 1.32 (0.86, 2.03); 1.19 (0.94, 1.52) 1.43 (0.93, 2.22), 0.10; 1.71 (1.32, 2.22), <0.01 VTE, venous thromboembolism 1.52 (0.88, 2.63); 1.35 (0.92, 1.97) 1.55 (0.90, 2.70), 0.12; 1.34 (0.90, 1.99), 0.15 ATE, arterial thromboembolism; 1.86 (0.34, 10.13); 0.87 (0.27, 2.86) 1.83 (0.33, 10.05), 0.49; 0.93 (0.27, 3.20), 0.91 Cancer (excluding non-melanoma skin cancer) 0.57 (0.38, 0.87); 1.02 (0.81, 1.29) 0.59 (0.39, 0.89), 0.01; 0.94 (0.74, 1.19); 0.61 Non-melanoma skin cancer NA; 1.10 (0.33, 3.66) NA; 1.38 (0.39, 4.88), 0.61 |
Article conclusion: JAKis did not increase the risk of AMI, stroke, CV-related mortality, MACE, VTE, ATE, or cancer in Korean RA patients relative to TNFis.
In Set 1: Only the total follow-up HR for MACE was significantly lower in the JAKi group compared to the TNFi group.
In Set 2: 1-yr follow-up HRs for AMI, Cancer (excl. NMSC), and total follow-up HRs for AMI and MACE were significantly lower in the JAKi group compared to the TNFi group while 1-yr follow-up HR for CV related mortality, and the total follow-up HR for all cause mortality were significantly higher in the JAKi group compared to the TNFi group. |
Mok, 2023
Various relevant outcomes
|
Type of study: retrospective observational cohort study
Setting and country: the Hong Kong Biologics Registry
Funding and conflicts of interest: This study was funded by the Hong Kong Society of Rheumatology, which has obtained a mini-grant from Pfizer. Disclosure statement: C.C.M. received a one-off speaker’s honorarium from Pfizer at the APLAR 2022 meeting. H.S. received speaker’s honorarium from Pfizer in the past 2 years. |
Inclusion criteria: patients who fulfilled the ACR/ EULAR criteria for RA and were ever treated with JAK and TNF inhibitors according to data from the Hong Kong Biologics registry between 2008 and December 2021.
Exclusion criteria: none reported.
N total at baseline: 2471 courses in 1732 RA patients Intervention: 551 Control: 1920
Important prognostic factors2: age ± SD: I: 57.9 (11.4) C: 52.6 (12.6
Sex: I: % M 18.1 C: % M 15.7
Groups comparable at baseline? No, many significant differences, but analyses are corrected for covariates.
|
Describe intervention (treatment/procedure/test): JAKi
|
Describe control (treatment/procedure/test): TNFi
|
Length of follow-up: Length of treatment/course of JAKi or TNFi (multiple courses per patient possible). Reasons for withdrawal, clinical inefficacy or SAE and mortality were outcome measures.
Loss-to-follow-up: The cumulative withdrawal rates for JAKis and TNFis at year 1, 3 and 5 due to clinical inefficacy were 22.4%, 36.9%, 43.5%; and 28.9%, 43.7%, 52.8%, respectively (log rank test; P¼0.001). The corresponding withdrawal rates of the JAKis and TNFis due to SAEs were 8.6%, 14.5%, 18.5%; and 12.9%, 20.2%, 24.5%, respectively (log rank test; P¼0.004). The hazard ratios (HRs) of withdrawal of the JAKis relative to the TNFis for clinical inefficacy and SAEs were 0.77 (95% CI: 0.63, 0.89) and 0.57 (95% CI: 0.42, 0.79), respectively, after adjustment for age, sex, duration of RA and the use of concomitant csDMARD’s. |
Outcome measures and effect size: Incidence/100PY JAKis; Incidence/100PY TNFi; Incidence ratio (95%CI); P Cardiovascular events 0.63; 0.40; 1.67 (0.65, 4.32); 0.29 Cerebrovascular events 0.54; 0.36; 0.93 (0.36, 2.41); 0.89 Peripheral vascular events 0.18; 0.00; –; – Total MACEs 1.34; 0.75; 1.49 (0.79, 2.84); 0.22 Venous thromboembolism 0.09; 0.02; 3.90 (0.20, 78.1); 0.37 Total cancer 0.81; 0.85; 0.65 (0.32, 1.35); 0.25 Total infections 16.3; 9.90; 1.24 (1.04, 1.48); 0.02 Total serious infections 8.20; 5.90; 0.97 (0.76, 1.23); 0.77 Death due to vascular events 0.09; 0.00; –; – Death due to infections 0.18; 0.17; 0.70 (0.19, 2.62); 0.59 Death due to cancer 0.18; 0.15; 0.70 (0.15, 3.38); 0.66 All-cause death 0.72; 0.47; 0.78 (0.35, 1.74); 0.54
JAKi vs TNFi Univariate HR (95%CI), P JAKi vs TNFi Multivariate HR (95%CI), P MACEs 2.09 (1.10, 3.95), 0.02 1.36 (0.62, 2.96), 0.44 Cancers 0.84 (0.39, 1.83), 0.66 0.87 (0.39, 1.95), 0.74 Infections 1.43 (1.16, 1.77), 0.001 1.08 (0.84, 1.39), 0.55 |
Article conclusion: In a real-life setting, there is no increase in MACEs or cancers in users of JAKis compared with TNFis. However, the incidence of non-serious infections, including herpes zoster, was increased in users of JAKis.
No significant difference in: - incidence of MACE after adjustment for age, sex and follow-up duration. - incidence of new cancers. However, Users of the JAKis had numerically higher incidence of breast, upper gastrointestinal and haematological cancers whereas users of the TNFis had numerically higher incidence of lung, head and neck, genitourinary, and non-melanoma skin cancers compared with their counterparts. - incidence of all-cause mortality. - incidence of serious infections. - incidence of TB, atypical mycobacterium, COVID-19
Significant difference in - incidence of all infections => higher in the JAKi than in the TNFi group after adjustment for age, sex and RA duration, which was contributed by more genitourinary, lower respiratory tract, skin and soft tissue infections. - HZ infection was significantly more common with the JAKis than TNFis (3.49 vs 0.94 per 100 patient-years; P<0.001). |
Molander, 2022
Cardiovascular outcomes (MACE, VTE)
|
Type of study: nationwide register-based, active comparator, new user design cohort study
Setting/Source and country: The Swedish Rheumatology Quality Register was linked to national health registers; Sweden
Funding and conflicts of interest: This study has received funding from Swedish Research Council, the Swedish Heart Lung Foundation, Nordforsk, Vinnova, and the Karolinska Institutet Region Stockholm funds (ALF). Karolinska Institutet, with JA as principal investigator, has or has had research agreements with Abbvie, Astra-Zeneca, BMS, Eli Lilly, Galapagos, MSD, Pfizer, Roche, Samsung Bioepis, Sanofi, and UCB, mainly in the context of safety monitoring of biologics via ARTIS/Swedish Biologics Register. |
Inclusion criteria: Not explicitly mentioned. Authors included b/tsDMARD treatment initiations between 1 January 2010 and 31 December 2020 among all in the Swedish Rheumatology Quality Register (SRQ) registered patients with RA above 18 years of age.
Using validated algorithms applied to National Patient Register (NPR), authors further identified the entire RA population in Sweden (the ‘overall RA cohort’) defined by at least two separate visits listing RA at a rheumatology or internal medicine clinic before or during the study period.
For each patient in b/ts cohorts five individuals from the general population were matched on age, sex and residential area.
Exclusion criteria: Subjects with a VTE registered during the year prior to start of follow-up were excluded.
N total at baseline: 27 610 unique patients with RA contributed with 32 737 b/tsDMARD’s treatment exposures Intervention: 2150 JAKi Control: 15090 TNFi
Important prognostic factors2: age Median (IQR): I: 60 (51–70) C: 57 (47–67)
Sex: I: % M 18 C: % M 23
Groups comparable at baseline? No, but comparative analyses were adjusted. |
Describe intervention (treatment/procedure/test):
JAKi (tofacitinib, baricitinib, upadacitinib)
Also: rituximab, interleukin 6 inhibitors (IL6i) (tocilizumab, sarilumab), abatacept |
Describe control (treatment/procedure/test):
TNFi (etanercept, infliximab, adalimumab, golimumab, certolizumab pegol),
|
Length of follow-up: Treatment initiation was defined as the registered date of treatment start in the SRQ. For each of these treatment cohorts, we used a first initiation per molecule approach, meaning that one individual could contribute to each treatment cohort more than once, but only once with each individual drug. Switch from an originator to biosimilar was considered the same treatment episode, as was restarting the same treatment within 90 days after discontinuation (180 days for rituximab) if no other bDMARD was initiated in between. An on-drug approach was used. Treatment discontinuation was defined as either of (1) registered date for discontinuation in the SRQ, (2) start of alternative b/tsDMARD in SRQ or (3) date of filled prescription for alternative b/tsDMARD from the PDR, whichever came first. For the overall RA cohort, follow-up started at first day of study period (or at the time point of the second ICD10 code registration for RA, if later). For the matched general population cohort, follow-up started at the time point of the first recorded treatment initiation in the corresponding index individual with RA. For all cohorts, follow-up ended at 60 days after discontinuation of the DMARD treatment in question, first VTE event, death, emigration or end of study period (31 December 2021), whichever came first.
Loss-to-follow-up: - Incomplete outcome data: - For covariates with missing data they used the missing indicator method. |
Relevant measures and effect size:
venous thromboembolism (VTE)
JAKi vs. TNFi
Baricitinib vs. TNFi
Tofacitinib vs. TNFi
TNFi
Model 1 adjusted for age, sex and line of therapy. Model 2 additionally adjusted for comorbidities and socioeconomic variables. Model 3 additionally adjusted for RA disease variables, civil status and smoking, using an indicator for missing variables.
pulmonary embolism (PE) JAKi vs. TNF fully adjusted HR 3.21 (95% CI 2.11 to 4.88)
deep vein thrombosis (DVT) JAKi vs. TNF fully adjusted HR 0.83 (95% CI 0.47 to 1.45) |
Article conclusion: Patients with RA treated with JAKi in clinical practice are at increased risk of VTE compared with those treated with bDMARD’s, an increase numerically confined to PE.
Stratified analyses: - analysed separately by sex, the incidence rates of VTE were approximately 50% higher in males than in females, and the HRs were numerically higher for males compared with females for those treated with IL6i versus TNFi as well as JAKi versus TNFi (2.66 (95% CI 1.44 to 4.92) for males and 1.53 (1.02–2.30) for females). - stratified by history of VTE, the incidence rate of VTE was almost nine times higher for individuals with a previous VTE versus those without, but all HRs for b/tsDMARD’s versus TNFi were close to 1. By contrast, among those without a previous VTE, the HRs were increased with IL6i versus TNFi and with JAKi versus TNFi (1.95 (1.33 to 2.87).
Sensitivity analyses -altering the definition of VTE as well as the definition of follow-up showed similar results as the main analyses, although the HRs were generally lower when extending follow-up from 60 days after exposure-drug discontinuation until start of (any) next treatment - limiting the study period to 2016–2021, HRs were very similar to those in the main analysis.
Additional analyses - analysing the subgroup of those fulfilling an emulation of the inclusion and exclusion criteria of the ORAL surveillance study, the incidence rates of VTE were around 50% higher, although the HRs for JAKi versus TNFi were similar or lower compared with the main analysis |
Pina Vegas, 2022
Cardiovascular outcomes (MACE, VTE)
|
Type of study: Observational cohort study Setting/Source and country: the French National Health Insurance database linked with the national hospital discharge database. France
Funding and conflicts of interest: There are no funders to report for this submission. Disclosure statement: L.P.V., P.L.C., L.P., M.P. and E.S. have no conflict of interest to declare. P.C. has received consulting fees from Abbvie, Pfizer, Roche-Chugai, Bristol-Myers Squibb, MSD, UCB, Novartis, Janssen, Lilly and Celgene (<$10 000 each), and has been an investigator for Roche Chugai, Sanofi Aventis, Celgene, Pfizer, MSD, Novartis and BMS. |
Inclusion criteria: - Adults (≥18 years old) with PsA, ICD-10 code M07] registered in the SNDS between 2015 and 2019 - patients with at least one prescription of bDMARD or apremilast for PsA - bDMARD- and apremilast-naive patients, defined as those who had not filled a prescription for one of these drugs for 1 year.
Exclusion criteria: patients with a history of acute myocardial infarction, unstable angina, chronic ischaemic heart disease, ischaemic stoke or transient ischaemic attack identified within 5 years before the index date
N total at baseline: Intervention: 1885 apremilast Control: 7289 TNFi (total bDMARD’s included, n=9510)
Important prognostic factors2: age ± SD: I: 54.0 (12.5) C: 48.2 (12.8)
Sex: I: % M 44.3 C: % M 41.2
Groups comparable at baseline? No, but IPTW was applied. |
Describe intervention (treatment/procedure/test): tsDMARD: Apremilast
IL-12/23i IL-17i: secukinumab and ixekizumab |
Describe control (treatment/procedure/test): TNFi: etanercept, infliximab, adalimumab, certolizumab and golimumab |
Length of follow-up: Exposure to a molecule was defined as the time from initiation to discontinuation. We defined the discontinuation of treatment as (i) a period of >90 days without a dispensation of the same treatment after the period covered by the previous reimbursement [28], or (ii) a switch of systemic treatment (other bDMARD or apremilast). The period covered by a prescription was 30 days for all molecules except infliximab (56 days) and ustekinumab (82 days). The discontinuation date was defined as the end of the 90-day period, and the switch date was defined as the date on which another systemic treatment was first reimbursed. Only the first therapeutic sequence of bDMARD or apremilast was considered in this analysis. In the intention-to-treat analysis, patients were followed up to the MACE event, death from any-cause, systemic treatment switch, lost to follow-up (defined by the absence of any reimbursement for 12 consecutive months) or 31 December 2019, whichever came first.
Loss-to-follow-up: - Incomplete outcome data: -
|
Relevant measures and effect size:
N(%) MACE, IR/1000PY(95%CI) TNF inhibitors (n¼7289) 30 (0.4) 2.8 (1.8, 3.9) IL-12/23i (n=1058): 5 (0.5), 3.1 (0.4, 5.8) IL-17i (n=1163): 8 (0.7), 5.9 (1.8, 9.9) Apremilast (n=1885): 8 (0.4), 5.2 (1.6, 8.9)
IPTW Cox HRw (95%CI), p-value IL-1223i: 2.0 (1.3-3.0), <10-4 IL-17i: 1.9 (1.2-3.0), <10-3 Apremilast: 1.3 (0.8-2.2), 0.31 (ref: TNF inhibitors)
IPTW Fine-Gray sHRw (95%CI), p-value IL-1223i: 2.1 (1.5-2.9), <10-3 IL-17i: 2.3 (1.5-3.0), <10-3 Apremilast: 1.4 (0.8-2.4), 0.12 (ref: TNF inhibitors)
|
Article conclusion Analysis of a large database revealed a small overall number of MACEs, and the risk of MACEs was greater for PsA new users of IL-12/23 and IL-17, not apremilast vs TNF inhibitors.
Subroup and sensitivity analyses - The results did not differ for patients without skin psoriasis requiring local treatment. - Among patients without comorbidities related to CVD, the overall crude incidence was 2.0 (0.0) per 1000 PY. Risk of MACEs was significantly higher (overall P<0.01) for IL- 17 inhibitors (HRw 1.6, 95% CI 1.1, 2.8), but not apremilast (HRw 0.7, 95% CI 0.4, 1.5), than TNF inhibitors. - The per-protocol and additional sensitivity analyses results were consistent with those of the main analysis. |
Song, 2022
Outcome malignancies
|
Type of study: retrospective cohort study
Setting and country: Korean National Health Insurance database
Funding and conflicts of interest: - Supported by a grant from the PACEN funded by the Ministry of Health and Welfare, Republic of Korea (grant number: HI19C0481, HC19C0052), and by the Basic Science Research Programme through the NRF funded by the Ministry of Education (NRF-2021 R1A6A1A03038899). - Y-KS has received research grants from Bristol-Myers Squibb, Eisai, Pfizer and JW Pharmaceutical. Other authors declare no conflict of interest. |
Inclusion criteria: Patients with RA who received their first JAKi or TNFi between 2015-2019.
Exclusion criteria: - all patients with prescriptions of JAKi or TNFi before the index date to ensure patients were TNFi/JAKi naïve. - under 18 years of age - diagnosed with AS, PsA, IBD, JIA patients - with prior malignancy in the past 5 years from the index date (hence not in remission) - with observation periods of less than 6 months
N total at baseline: 4929 patients Intervention: 1064 Control: 3865
Important prognostic factors2: age ± SD: I: 55.7±12.5 C: 54.2±13.3
Sex: I: % M 17.2 C: % M 20.9
Groups comparable at baseline? No, even after IPTW, some differences remained. |
Describe intervention (treatment/procedure/test): First JAKi
|
Describe control (treatment/procedure/test): First TNFi
|
Length of follow-up: Patients were followed up from the index date to the occurrence of malignancy, drug discontinuation, death or the end of the study in December 2019.
Loss-to-follow-up: Patients who were lost to follow-up were not considered separately because they were censored due to drug discontinuation.
Incomplete outcome data: n/a
|
Outcome measures and effect size:
Before IPTW: IR (95%CI); HR (95%CI) Overall malignancy JAKi 0.54 (0.26 to 1.14); 0.69 (0.30 to 1.56) TNFi 0.85 (0.66 to 1.10) Solid malignancy JAKi 0.47 (0.21 to 1.04); 0.61 (0.26 to 1.47) TNFi 0.82 (0.63 to 1.07) Haematological malignancy JAKi 0.08 (0.01 to 0.55); 2.41 (0.15 to 37.99) TNFi 0.03 (0.01 to 0.12)
After IPTW: IR (95%CI); HR (95%CI) Overall malignancy JAKi 0.67 (0.48 to 0.94); 0.83 (0.55 to 1.27) TNFi 0.85 (0.67 to 1.07) Solid malignancy JAKi 0.61 (0.43 to 0.87); 0.77 (0.50 to 1.19) TNFi 0.82 (0.65 to 1.04) Haematological malignancy JAKi 0.06 (0.02 to 0.19); 2.86 (0.41 to 20.00) TNFi 0.02 (0.01 to 0.10)
In the sensitivity and subgroup analyses, there was also no significant difference in the risk of malignancy between the JAKi and TNFi groups. |
Article conclusion: Malignancy risk in Korean patients with RA was not increased with JAKi use compared with TNFi use.
NB: since the observation period was less than 2 years for both groups, it may have been insufficient for malignancies to develop.
NB2: 6% of the total population had used non-TNFis before TNFi/JAKi treatment, but included as covariate. |
Song, 2023a
Cardiovascular outcomes (MACE, VTE)
|
Type of study: Observational cohort study
Setting/Source and country: National Health Insurance Service database Republic of Korea
Funding and conflicts of interest: This research was supported by a grant of Patient-Centered Clinical Research Coordinating center (PACEN) funded by the Ministry of Health & Welfare, Republic of Korea. This research was also supported by Basic Science Research Program through the National Research Foundation of Korea (NRF), funded by the Ministry of Education. YKS has received research grants from Bristol-Myers Squibb, Eisai, Pfizer, and JW Pharmaceutical. Other authors declare that there are no conflicts of interest. |
Inclusion criteria: - Prevalent RA = RA diagnostic code + prescription of DMARD - New user of JAKi or TNFi - Between 2015 and 2019
Exclusion criteria: - age <18 - diagnosed with other autoimmune diseases, such as ankylosing spondylitis, psoriatic arthritis, inflammatory bowel disease and juvenile idiopathic arthritis - prior history of VTE any time before the index date or a prior history of anticoagulant therapy within 30 days of the index date - Patients with an observation period of less than six months
N total at baseline: 4178 patients Intervention: 871 JAKi users Control: 3307 TNF inhibitor users
Important prognostic factors2: age ± SD: I: 54.6 ± 12.3 C: 54.2 ± 13.2
Sex: I: % M 16.9 C: % M 19.5
Groups comparable at baseline? No, but IPTW was applied. After performing sIPTW, 577 JAKi users and 3160 TNF inhibitor users were included. Almost all variables were balanced, including age, sex, comorbidities and medication use. However, several variables were still unbalanced after sIPTW: seropositivity, Charlson Comorbidity Index score group, varicose vein and obesity. |
Describe intervention (treatment/procedure/test): JAKi: tofacitinib or baricitinib |
Describe control (treatment/procedure/test): TNFi: adalimumab, etanercept, golimumab, or infliximab
|
Length of follow-up: - The observation started from the index date and terminated at the incidence of VTE, discontinuation of JAKi or TNF inhibitor, death, or December 2019. - 12 weeks of permissible gap to define discontinuation of JAKi or TNF inhibitor, so a gap of less than 12 weeks in addition to the usual drug interval was not considered drug discontinuation. As-treated analysis was mainly performed for VTE incidence, and intention-to-treat (ITT) analysis was conducted as a sensitivity analysis.
Loss-to-follow-up: - Incomplete outcome data: -
|
Relevant measures and effect size:
The risk of VTE in RA patients treated with JAKi versus TNFi (after sIPTW) N patients, n events, person-years, IR(95%CI), HR (95%CI), aHR (95%CI)
VTE JAKi: 577, 0.4, 688.0, 0.06 (0.00–1.23), 0.18 (0.01–3.47), 0.18 (0.01–3.47) TNFi: 3160, 20.4, 5384.9, 0.38 (0.25–0.58), ref., ref.
PE JAKi: 577, 0, 688.1, N/A, N/A, N/A. TNFi: 3160, 4.1, 5395.2, 0.08 (0.03–0.20)
DVT JAKi: 577, 0.4, 688.0, 0.06 (0.00–1.23), 0.21 (0.01–4.24), 0.22 (0.01–4.32) TNFi: 3160, 17.1, 5388.2, 0.32 (0.20–0.51) |
Article conclusion: There is no increased risk of VTE in RA patients treated with JAKis compared with TNF inhibitors in Korea.
In sensitivity analyses, ITT analysis was conducted in two ways: for the total observation period and 1 year. The adjusted HR of VTE was 0.33 (95% CI 0.06–1.90) in the ITT analysis for the total observation period and 0.40 (95% CI 0.04–3.71) in the ITT analysis for 1 year. A subgroup analysis according to sex, age, hypertension and dyslipidaemia was conducted. The HR could not be calculated in several subgroups due to the absence of VTE incidence amongst JAKi users, but there were no statistically significant differences in those subgroups where the HR could be calculated. When tofacitinib users and TNF inhibitor users were compared, tofacitinib did not increase the risk of VTE compared with TNF inhibitor. |
Song, 2023b
Infections (serious, herpes zoster)
|
Type of study: Prospective observational cohort study
Setting/Source and country: academic referral hospital in Korea
Funding and conflicts of interest: This research was supported by a grant of the Korea Health Technology R&D Project through the Korea Health Industry Development Institute (KHIDI), funded by the Ministry of Health and Welfare, Republic of Korea. Tofacitinib registry and this research was sponsored by Pfizer. YKS has received research grants from Bristol-Myers Squibb, Eisai, Pfizer, and JW Pharmaceutical. JYJ and HJY are employees and shareholders of Pfizer Inc. No potential conflicts of interest relevant to this article are reported. The other authors declare that they have no competing interests. |
Inclusion criteria: All patients with RA who started targeted therapy in our institution were candidates for the cohorts, but those who refused to write an informed consent were excluded. In this study, patients with RA starting tofacitinib between March 2017 and May 2021 and those starting TNFi between July 2011 and May 2021 were included.
Exclusion criteria: No informed consent.
N total at baseline: 912 Intervention: 200 Control: 712
Important prognostic factors2: age ± SD: I: 53.4 ± 12.2 C: 50.8 ± 13.5
Sex: I: % M 9.5 C: % M
Groups comparable at baseline? No, but Differences in the baseline variables were balanced after IPTW between 200 tofacitinib users and 200 TNFi users. |
Describe intervention (treatment/procedure/test):
Tofacitinib |
Describe control (treatment/procedure/test):
TNFi: etanercept, infliximab, adalimumab, golimumab |
Length of follow-up: Demographic and clinical information of RA patients were collected at enrolment, and the disease activity was assessed every 6 months according to DAS28 and PROMs. The observation period commenced from the initiation of tofacitinib or TNFi and continued until the onset of HZ, discontinuation of each agent, or May 2021.
Loss-to-follow-up: - Incomplete outcome data: The medical records of patients were reviewed together to minimize the possibility of missing information on the study-related data points.
|
Relevant measures and effect size:
Incidence and risk of Herpes Zoster and serious HZ, tofacitinib vs. TNFi N cases, IR/100PY, IRR (95%CI), p-value
Incidence and risk of HZ Before IPTW TOFA: 20, 6.03, 3.27 (1.89–5.65), <0.001 TNFi: 36, 1.85, ref.
Incidence and risk of serious HZ Before IPTW TOFA: 1, 0.30, 0.99 (0.12–8.20), 0.987 TNFi: 6, 0.31, ref.
Incidence and risk of HZ after IPTW TOFA: 20, 6.03, 8.33 (3.05–22.76), <0.001 TNFi: 5, 0.77, ref.
Incidence and risk of serious HZ after IPTW TOFA: 1, 0.30, 3.52 (0.14–93.51), 0.452 TNFi: 1, 0.15, ref.
Incidence and risk of HZ within 12 months of use before IPTW TOFA: 12, 7.39, 4.59 (1.93–10.88), <0.001 TNFi: 9, 1.61, ref.
Incidence and risk of HZ within 12 months of use after IPTW TOFA: 12, 7.39, 10.83 (1.58–74.51), 0.015 TNFi: 1, 0.60, ref.
|
Article conclusion: Tofacitinib use increased the risk of HZ compared with TNFi in Korean patients with RA, but the rate of serious HZ or permanent discontinuation of tofacitinib due to HZ event was low.
In the sensitivity analysis, the IRR of HZ development within 12 months increased significantly after IPTW to 10.83 (95% CI 1.58–74.51, p = 0.015) |
Uchida, 2023
Various relevant outcomes
|
Type of study: retrospective cohort study
Setting and country: Nagasaki University Hospital, Sasebo Chuo Hospital and Ureshino Medical Center, Japan
Funding and conflicts of interest: No specific funding was received. No conflicts of interest. |
Inclusion criteria: RA diagnosis treated with tofacitinib, baricitinib, or TNF inhibitors informed consent
Exclusion criteria: None reported.
N total at baseline: 499 Intervention: 296 (TAFA 192, BAR 104) Control: 203
Important prognostic factors2: age (IQR): I: TOFA 67 (58–73) I: BAR 68 (58–75) C: TNF 51 (37–61)
Sex: I: % M TOFA 19.3 I: % M BAR 15.4 C: % M TNF 18.2
Groups comparable at baseline? No, but IPTW is performed. |
Describe intervention (treatment/procedure/test): The JAK inhibitor–treated patients received either tofacitinib 5mg twice or once daily (in patients with renal impairment) or baricitinib 2mg (in patients with renal impairment) or 4mg once daily. |
Describe control (treatment/procedure/test): TNF inhibitors (adalimumab or etanercept)
|
Length of follow-up: Max. March 2013 through December 2020; the period from the initiation of tofacitinib or baricitinib treatment until the treatment’s discontinuation, the patient’s transfer to another hospital or death, or the end of the study period, whichever occurred first.
Loss-to-follow-up: Is when outcomes occur.
Incomplete outcome data: None reported.
Follow-up time, yrs: TOFA 2.0 (0.5–3.8) BAR 0.8 (0.33–1.5) TNF 1.4 (0.4–3.1)
|
Outcome measures and effect size:
n(%); IR per 100 PY (95% CI)
Serious infectious diseases other than HZ JAKi 40 (13.5%); 8.36 (7.79, 8.93) TOFA 33 (17.2%); 8.68 (8.10, 9.26) BAR 7 (6.7%); 7.09 (6.57, 7.61) TNFi 16 (7.9%); 4.09 (3.70–4.49)
Herpes Zoster JAKi 57 (19.3%); 13.00 (12.29, 13.71) TOFA 46 (24.0%); 13.37 (12.65, 14.09) BAR 11 (10.6%); 11.63 (10.96, 12.30) TNFi 6 (3.0%); 1.48 (1.24–1.72)
MACE n = 2 (1.0%) in TOFA 0 and 0 in BAR and TNF
Malignancies n = 11 (3.7%) in JAKi n = 3 (1.4%) in TNFi
The weighted Cox proportional hazard model revealed the following adjusted hazard ratios TNFi vs JAKi
Serious infection 0.792 (95% CI: 0.417, 1.502), p=0.475 HZ 0.200 (95% CI: 0.077, 0.524), p=0.001 Malignancy 0.385 (95% CI: 0.095, 1.552), p=0.179 |
Article conclusions: The infectious disease IR in RA was comparable between tofacitinib and baricitinib, but the IR for HZ in these treatment groups was high compared with that in the TNF inhibitor treatment group. The malignancy rate in the JAK-inhibitor-treated group was high but not significantly different from that of the general population or that of the TNF-inhibitor–treated group.
Note: baseline data were limited and did not include parameters of a smoking history, rate of vaccination other than the HZ vaccine, or disease activity.
|
Westermann, 2023
Outcome malignancies
|
Type of study: observational cohort study
Source and country: the Danish Rheumatology Quality Register, the Danish Cancer Registry, the Danish Civil Registration System, the Danish Population Education Register, and the Danish National Patient Register, Denmark
Funding and conflicts of interest: This work was financially supported by the Danish Rheumatism Association and the Danish Cancer Society. The funders were involved neither in the planning, performance, nor the submission of the manuscript. Two of the seven authors have no conflicts of interests. For the other five, disclosures are provided in an extensive statement |
Inclusion criteria: All patients with RA in DANBIO aged 18 or more and who initiated treatment with either JAKi or bDMARD’s (index date) from 1 January 2017 to 31 December 2020.
Exclusion criteria: Patients with a registered cancer prior to the index date were excluded, except for those with prior non-melanoma skin cancer (NMSC).
For both groups, patients with previous bDMARD treatments were included.
N total at baseline: 4601 Intervention: 875 Control: 4247
Important prognostic factors2: age ± SD: I: 57.8 (13.05) C: 57.6 (13.83)
Sex: I: % M 20.2 C: % M 24.9
Groups comparable at baseline? Not before IPTW, but after all covariates and covariate levels had post-IPTW weighting SMDs <10%, and the majority were <5%. |
Describe intervention (treatment/procedure/test): any available JAKi in Denmark, i.e. tofacitinib or baricitinib
|
Describe control (treatment/procedure/test): interleukin-6 inhibitors (tocilizumab/ sarilumab); B-cell inhibitors via anti-CD20 (rituximab); T-cell co-stimulation inhibitors via CTLA-4 (abatacept); all types of TNFi, including both originators and biosimilars
|
Length of follow-up: Follow-up started on the date upon first registered treatment with JAKi or bDMARD’s after 1 January 2017. Follow-up was performed in a hierarchical ever-treated design, i.e. ‘once exposed always exposed’. However, patients were able to switch from the bDMARD group to the JAKi group, mimicking the usual hierarchical treatment pattern for JAKi-treated patients according to Danish guidelines. Person years (PYRS) of follow-up and number of cancers were allocated to each JAKi and bDMARD group, respectively, by following patients from the index date and until date of cancer, death, emigration, initiation of JAKi (bDMARD group censoring only), or 31 December 2020, whichever occurred first.
During follow-up, the JAKi group contributed 1315 PYRS (median 1.48 years; interquartile range 0.98– 1.93 years) and 19 cancers, while the bDMARD group contributed 8597 PYRS (1.98; 1.11–3.09) and 111 cancers. |
Outcome measures and effect size:
Malignancy (excl. NMSC) JAKi IR/1000PY 14.4 bDMARD IR/1000PY 12.9 weighted CSC model: HR JAKi vs bDMARD: 1.41 (95%CI 0.76, 2.37) |
Article conclusion: JAKi treatment in real-world patients with RA was not associated with a statistically significant increased risk of first primary cancer compared with those who received bDMARD’s.
Note: Analyses stratified by age and by length of follow-up also displayed numerically increased yet statistically non-significant HRs. None of the sensitivity analyses showed any statistically significant HRs |
Notes:
Various relevant outcomes
Infections (serious, herpes zoster)
Cardiovascular outcomes (MACE, VTE)
Outcome malignancies
N = number of patients; NNH = number needed to harm; I = intervention; C = Comparator; IR = incidence rate; PY = patient-years; HR = hazard ratio; RoB = risk of bias; RCT = randomized controlled trial; LTE = long-term extension; NMCS = Non-melanoma skin cancer; BAR = baricitinib; FIL = filgotinib; PEF = peficitinib; TOFA = tofacitinib; UPA = upadacitinib; JAKi = Janus kinase inhibitor; TNFi = tumor necrosis factor inhibitor; bDMARD = biological DMARD; tsDMARD = targeted synthetic DMARD; c(s)DMARD = conventional synthetic DMARD; ABA = abatacept; ADA = adalimumab; CZP = certolizumab pegol; ETA = etanercept; Golimumab = GOL; IFX = infliximab; RTX = rituximab; SAR = sarilumab; TCZ = tocilizumab; MTX = methotrexate; NR = not reported; NA = not applicable
Table of quality assessment for systematic reviews of RCTs and observational studies
Based on AMSTAR checklist (Shea et al.; 2007, BMC Methodol 7: 10; doi:10.1186/1471-2288-7-10) and PRISMA checklist (Moher et al 2009, PLoS Med 6: e1000097; doi:10.1371/journal.pmed1000097)
Study
First author, year |
Appropriate and clearly focused question?1
Yes/no/unclear |
Comprehensive and systematic literature search?2
Yes/no/unclear |
Description of included and excluded studies?3
Yes/no/unclear |
Description of relevant characteristics of included studies?4
Yes/no/unclear |
Appropriate adjustment for potential confounders in observational studies?5
Yes/no/unclear/not applicable |
Assessment of scientific quality of included studies?6
Yes/no/unclear |
Enough similarities between studies to make combining them reasonable?7
Yes/no/unclear |
Potential risk of publication bias taken into account?8
Yes/no/unclear |
Potential conflicts of interest reported?9
Yes/no/unclear |
EULAR Sepriano, 2023 |
Yes |
Yes |
Yes |
Yes |
Unclear The risk of bias varies per included study. |
Yes |
No Studies were heterogeneous, precluding data pooling, and results are presented descriptively. |
Yes |
Yes |
- Research question (PICO) and inclusion criteria should be appropriate and predefined
- Search period and strategy should be described; at least Medline searched; for pharmacological questions at least Medline + EMBASE searched
- Potentially relevant studies that are excluded at final selection (after reading the full text) should be referenced with reasons
- Characteristics of individual studies relevant to research question (PICO), including potential confounders, should be reported
- Results should be adequately controlled for potential confounders by multivariate analysis (not applicable for RCTs)
- Quality of individual studies should be assessed using a quality scoring tool or checklist (Jadad score, Newcastle-Ottawa scale, risk of bias table etc.)
- Clinical and statistical heterogeneity should be assessed; clinical: enough similarities in patient characteristics, intervention and definition of outcome measure to allow pooling? For pooled data: assessment of statistical heterogeneity using appropriate statistical tests (e.g. Chi-square, I2)?
- An assessment of publication bias should include a combination of graphical aids (e.g., funnel plot, other available tests) and/or statistical tests (e.g., Egger regression test, Hedges-Olken). Note: If no test values or funnel plot included, score “no”. Score “yes” if mentions that publication bias could not be assessed because there were fewer than 10 included studies.
- Sources of support (including commercial co-authorship) should be reported in both the systematic review and the included studies. Note: To get a “yes,” source of funding or support must be indicated for the systematic review AND for each of the included studies.
Risk of bias table for interventions studies (cohort studies based on risk of bias tool by the CLARITY Group at McMaster University)
Research question: What are the benefits/harms of treatment with targeted synthetic (ts)DMARD’s compared to treatment with biological (b)DMARD’s or conventional synthetic (cs)DMARD’s in patients with rheumatoid arthritis (RA), psoriatic arthritis (PsA) or spondylarthritis (SpA)
Author, year |
Selection of participants
Was selection of exposed and non-exposed cohorts drawn from the same population?
|
Exposure
Can we be confident in the assessment of exposure?
|
Outcome of interest
Can we be confident that the outcome of interest was not present at start of study?
|
Confounding-assessment
Can we be confident in the assessment of confounding factors?
|
Confounding-analysis
Did the study match exposed and unexposed for all variables that are associated with the outcome of interest or did the statistical analysis adjust for these confounding variables? |
Assessment of outcome
Can we be confident in the assessment of outcome?
|
Follow up
Was the follow up of cohorts adequate? In particular, was outcome data complete or imputed?
|
Co-interventions
Were co-interventions similar between groups?
|
Overall Risk of bias
|
Definitely yes, probably yes, probably no, definitely no |
Definitely yes, probably yes, probably no, definitely no |
Definitely yes, probably yes, probably no, definitely no |
Definitely yes, probably yes, probably no, definitely no |
Definitely yes, probably yes, probably no, definitely no |
Definitely yes, probably yes, probably no, definitely no |
Definitely yes, probably yes, probably no, definitely no |
Definitely yes, probably yes, probably no, definitely no |
Low, Some concerns, High |
|
Fang, 2022 Various relevant outcomes
|
Definitely yes
Reason: Participants were selected from a registry |
Definitely yes
Reason: Source National Health Insurance research database. |
Definitely yes for all outcomes, except malignancy Definitely no for outcome malignancy. NB: groups were matched on comorbidities.
Reason: Outcome malignancy could already be present at baseline, registered as comorbidity. |
Probably yes
Reason: From high quality database, all available confounding factors were assessed. Some variables associated with the study outcomes were not in the database. |
Definitely yes
Reason: Comprehensive matching with propensity score
|
Probably yes
Reason: Outcomes came from a national research database. |
Probably yes
Reason: FU was performed from the first use of agents of interest until the first occurrence of the individual study outcomes, switch to other agents, or end date study, whichever came first. |
Definitely yes
Reason: Additional used medication was balanced between groups |
Some concerns |
Frisell, 2023 Various relevant outcomes |
Definitely yes
Reason: Participants were selected from a registry |
Definitely yes
Reason: Source is a longitudinal clinical registry-infrastructure with consistent definitions of treatment cohorts, follow-up and outcomes (ARTIS) |
Definitely yes
Reason: Patients with a recent history of an outcome (prior 5 yrs, except for infection where only last year was considered) were excluded from analyses of the same outcome. |
Definitely yes
Reason: Source is a longitudinal clinical registry-infrastructure with consistent definitions of treatment cohorts, follow-up and outcomes (ARTIS) |
Definitely yes
Reason: Groups were balanced by IPTW taking into account all relevant confounders |
Definitely yes
ReasonSource is a longitudinal clinical registry-infrastructure with consistent definitions of treatment cohorts, follow-up and outcomes (ARTIS) |
Definitely yes
Reason: Data were complete on treatments, outcomes and most covariates. Missing covariate data were accounted for by multiple imputation. |
Probably no
Reason: Treatment groups seem to differ in concomitant medications, but groups were balanced on these variables. |
Some concerns |
Hirose, 2022 Various relevant outcomes
|
Probably no
Reason: Data were derived retrospectively for patients starting treatment before start of study and prospectively for patients starting within the duration of the study. No information how many patients/data were obtained retrospectively and how many prospectively; not clear if different per treatment group |
Definitily yes
Reason: Secure record |
Probably yes
Reason: incidence and severity of all adverse events were recorded during the study period. Co-morbidities (which could be the same as safety outcomes) were not taken into account in the article, but extensive lab measurements were taken into account. |
Probably yes
Reason: Adjustment for most plausible prognostic variables obtained from medical records (retrospectively or prospectively). |
Probably yes
Reason: inverse probability of treatment weighting (IPTW) based on a propensity score that reduces the selection bias to a minimum and adjusts for confounding factors between binary treatment groups was used |
Probably yes
Reason: Partly from medical records, partly from prospective data collection. The common terminology criteria for adverse events of the National Cancer Institute (v.5.0) were used to describe and grade adverse events and laboratory abnormalities. |
Probably yes
Reason: The last observation carried forward method was used for patients who discontinued treatment before week 52 to include all patients in the analysis. No information: Unclear what they did with respect to data from patients who withdrew their consent for data-usage. |
Definitely yes
Reason: Additional used medication was balanced between groups |
Some concerns |
Hoisnard, 2022 Cardiovascular outcomes (MACE, VTE) |
Definitely yes
Reason: Participants were selected from the same registry over the same time period. |
Definitely yes
Reason: secure record, identification in registry with ATC classification codes. |
Probably yes
Reason: Presence of outcome at baseline was not an exclusion criterium, but also not likely to coincide with index date and risks associated with outcome were identified and taken into account in analyses. |
Probably yes
Reason: All relevant available covariates collected from the French national health data system.Some variables included in the propensity score relied on proxies. No information on missing covariate data. |
Probably yes
Reason: Groups were balanced by IPTW taking into account all available relevant confounders. However, confounders such as disease activity data were not available. |
Definitely yes
Reason: Identification of outcomes by ICD-10 codes in national database. |
Probably yes
Reason: Patients were followed up to the occurrence of each event (MACE and VTE), death from any cause, exposure discontinuation or study end, whichever came first. |
Definitely no
Reason: Groups differed in concomitant csDMARD use, but analyses were corrected / groups balanced on these variables. |
Some concerns |
Huss, 2023 OutcomeMalignancies
|
Probably no
Reason: Same database sources, same locations, but shorter time frame for intervention due to 1 year later introduction of JAKi |
Definitely yes
Reason: Secure records |
Definitely yes
Reason: Patients with previous cancer diagnosis (outcome) were excluded |
Probalby yes
Reason: All plausible confounding variables were recorded, came from various national registries |
Probably yes
Reason: adjustment for most plausible confounding variables |
Definitely yes
Reason: medical record and record linkage |
Probably yes
Reason: FU from every treatment initiation until the occurrence of the outcome, death, emigration from Sweden or end of the study period |
Definitivily no
Reason: % use of MTX and oral steroids seems to vary per group, but no comparative analyses are presented. However, analyses were corrected for confounding of co-interventions. |
Some concerns |
Jeong, 2022 Infections (serious, herpes zoster) |
Definitely yes
Reason: Participants were selected from the same registry over the same time period. |
Definitily yes
Reason: Secure record; high quality national registry |
Definitely yes
Reason: Patients with (recent or rapidly recurring) herpes zoster (outcome) were excluded |
Probalby yes
Reason: All plausible confounding variables were recorded, coming from a good quality national database. No information: article mentions that patients with missing data, such as covariates, were excluded, but they don’t mention anything about subsequently excluded patients; all originally included patients are in analyses. |
Probably yes
Reason: adjustment for most plausible confounding variables |
Probably yes
Reason: Outcomes came from a national research database. |
Probably yes
Reason: FU until until drug failure, development of herpes zoster (outcome), or study end. |
Probably no
Reason: Groups differ, but analyses were corrected for these variables. |
Some concerns |
Khosrow-Khavar, 2022 Cardiovascular outcomes (MACE, VTE) |
Definititely yes
Reason: Participants were selected from the same registry. |
Definitily yes
Reason: Secure record; high quality national registry |
Probably yes
Reason: assessed 76 potential confounders from various national databases, including medical history. |
Definitely yes
Reason: Propensity score fine stratification achieved excellent covariate balance with standardized differences close to zero across all covariates |
Definitely yes
Reason: assessed 76 potential confounders from various national databases. |
Probably yes
Reason: Outcomes came from various national databases. |
Probably yes
Reason: patients were followed from treatment initiation for study outcomes until treatment discontinuation or switch, insurance disenrollment, death, or end of the study period, whichever occurred first. |
Probably no
Reason: Groups differ, but analyses were corrected for this. |
Some concerns |
Min, 2023 Various relevant outcomes
|
Definititely yes
Reason: Participants were selected from the same registry. |
Definititely yes
Reason: Secure records |
Definitely yes
Reason: patients with a main ICD-10 diagnosis code for AMI, stroke, VTE, ATE, or cancer 12 months prior to the initiation of TNFi or JAKi were excluded. |
Probably yes
Reason: Most important covariates as recorded in health insurance database |
Probably yes
Reason: Authors did adjust analyses for all the covariates that they did collect. No information: Not clear what was done with missing covariate information. |
Definitely yes
Reason: National database ICD-10 codes were used |
Probably yes
Reason: FU until outcome occurred, switch in medication type, or end of study. |
Definitely no
Reason: Glucocorticoid and NSAID us differed between groups. However, MTX was used in both groups (inclusion criteria) and analyses were corrected for confounding variables. |
Some concerns |
Mok, 2023 Various relevant outcomes
|
Definitely yes
Reason: Participants were selected from the same registry |
Definitely yes
Reason: Secure records |
Probably yes
Reason: Not entirely clear, but history of outcome diseases was recorded and taken into account in analyses. |
Probably yes
Reason: Analyses were adjusted for important covariates as recorded in a registry; these differed per outcome. |
Probably yes
Reason: Analyses were adjusted for relevant covariates in which groups differed. However, analyses were not always adjusted for all covariates; no information why. |
Definitely yes
Reason: Regional registry used and verification by review of individual medical records by two independent research assistants. |
Probably yes
Reason: FU until outcome occurred, start of a new cours, or end of study. |
Definitely no
Reason: Between group differences in MTX, LEF, HCQ, Glucocorticoids, sulphasalizine. At least the analyses for outcome Infections were adjusted for this. |
Some concerns |
Molander, 2022 Cardiovascular outcomes (MACE, VTE) |
Definitely yes
Reason: Participants were selected from the same registry over the same time period. |
Definitily yes
Reason: Secure record; high quality national registry |
Definitely yes
Reason: Subjects with a VTE (outcome) registered during the year prior to start of follow-up were excluded.
|
Probably yes
Reason: Analyses were adjusted for important covariates as recorded in a registry |
Probably yes
Reason: adjustment for most plausible confounding variables |
Definitely yes
Reason: information from various registries was combined to define an outcome. |
Probably yes
Reason: follow-up ended at 60 days after discontinuation of the DMARD treatment in question, first VTE event, death, emigration or end of study period, whichever came first.
|
Definitely no
Reason: Compared with TNFi, patients starting other b/tsDMARD’s were generally slightly older, had longer-lasting RA, more comorbidities and a higher level of healthcare consumption. However, analyses were adjusted for this. |
Some concerns |
Pina Vegas, 2022 Cardiovascular outcomes (MACE, VTE) |
Definitely yes
Reason: Participants were selected from the same registry over the same time period. |
Definitily yes
Reason: Secure record; high quality national registry |
Definitely yes
Reason: patients with a history of acute myocardial infarction, unstable angina, chronic ischaemic heart disease, ischaemic stoke or transient ischaemic attack were excluded. identified within 5 years before the index date |
Definitely yes
Reason: Analyses were adjusted for important covariates as recorded in a registry. IPTW was performed. |
Definitely yes
Reason: Comprehensive matching with propensity score. |
Probably yes
Reason: Events were identified by a hospital discharge diagnosis - ICD-10 codes - with a previously validated algorithm. |
Probably yes
Reason: Patients were followed up to the MACE event, death from any-cause, systemic treatment switch, lost to follow-up (defined by the absence of any reimbursement for 12 consecutive months) or study end, whichever came first. |
Definitely no
Reason: groups differed on various covariates. However, after IPTW, a pseudo-cohort with a similar distribution of between the different treatment classes was obtained. |
Some concerns |
Song, 2022 OutcomeMalignancies
|
Definitely yes
Reason: Participants were selected from the same registry |
Definitely yes
Reason: Secure records |
Definitely yes
Reason: Outcome was cancer and cancer had to be in remission at start of treatment (cancer in last 5 years meant exclusion) |
Probably yes
Reason: Most relevant covariates, but maybe not all/those available were taken into account, as recorded in health insurance database |
Probably yes
Reason: Comprehensive matching with propensity score. After IPTW, authors report still unbalanced covariates including the year of initiating JAKi or TNFi treatment, seropositivity, cerebrovascular disease, previous csDMARD use, and concomitant MTX and oral corticosteroid use. |
Definitely yes
Reason: linkage of two records |
Definitely yes
Reason: Patients were followed up from the index date to the occurrence of malignancy, drug discontinuation, death or the end of the study in December 2019. Patients who were lost to follow-up were not considered separately because they were censored due to drug discontinuation. |
Definitely no
Reason: Even after IPTW, differences remained on concomitant MTX and oral corticosteroid use. |
Some concerns |
Song, 2023a Cardiovascular outcomes (MACE, VTE) |
Definitely yes
Reason: Participants were selected from the same registry |
Definitely yes
Reason: Secure records |
Definitely yes
Reason: those with a prior history of VTE or a prior history of anticoagulant therapy within 30 days of the index date were excluded. |
Probably yes
Reason: Most relevant covariates, but maybe not all/those available were taken into account, as recorded in health insurance database |
Probably yes
Reason: Comprehensive matching with propensity score. However, several variables were still unbalanced after sIPTW. |
Probably yes
Reason: defined by ICD10 code and use of anticoagulant therapy within 30 days from VTE diagnosis |
Probably yes
Reason: The observation started from the index date and terminated at the incidence of VTE, discontinuation of JAKi or TNF inhibitor, death, or study end. |
Definitely no
Reason: Differences before IPTW, but not after on medication use. |
Some concerns |
Song, 2023b Infections (serious, herpes zoster) |
Probably yes
Reason: Same hospital, all at start target medication |
Definitely yes
Reason: Secure records, medical record |
Probably no:
Reason: no exclusion criteria for (recent or rapidly recurring) herpes zoster (outcome) |
Probabably yes
Reason: Most relevant covariates were recorded. |
Definitely yes
Reason: Comprehensive matching with propensity score. Groups balanced after IPTW. |
Probably yes
Reason: based on HZ diagnose in medical record and reporting as AE or SAE. Medical records reviewed by two people together. |
Probably yes:
Reason: The observation period commenced from the initiation of tofacitinib or TNFi and continued until the onset of HZ, discontinuation of each agent, or study end. |
Definitely no
Reason: Differences before IPTW, but not after on medication use. |
Some concerns |
Uchida, 2023 Various relevant outcomes
|
Probably yes
Reason: participants from same points of care over the same time frame |
Definitely yes
Reason: Medical record |
Probably yes
Reason: For the outcome of malignancies they excluded recurrent ones or occurrence within one month after treatment start. |
Probably no
Reason: Most relevant covariates recorded (medical record) at initiation of treatment, but as the authors note, not all possible important confounders were taken into account |
Definitely yes
Reason: Comprehensive matching with propensity score |
Definitely yes
Reason: Outcomes were determined by treating physician(s) according to described requirements. |
Definitely yes
Reason: Patients were followed up from the initiation of tofacitinib or baricitinib treatment until the treatment’s discontinuation, the patient’s transfer to another hospital or death, or the end of the study period, whichever occurred first. |
No information
Reason: Groups seem to differ on concomitant treatments before IPTW. Not clear if they were balanced after IPTW. |
Some concerns |
Westermann, 2023 OutcomeMalignancies
|
Definitely yes
Reason: Participants were selected from the same registry |
Definitely yes
Reason: high quality databases |
Definitely yes
Reason: patients with a registered cancer (outcome) prior to index date were excluded. |
Probably yes
Reason: Indicators of all possible relevant confounders were collected from high quality national databases. NB: Strangely enough, concomitant and/or previous MTX use is nowhere mentioned in te article. |
Definitely yes
Reason: Comprehensive matching with propensity score |
Definitely yes
Reason: All cancer diagnoses were collected from the DCR. Cancer diagnoses registered in DCR have been estimated to be highly valid and complete. |
Definitely yes
Reason: FU until date of: cancer, death, emigration, initiation of JAKi (bDMARD group censoring only), or 31 December 2020, whichever occurred first. Death was considered a competing risk due to its preclusion of cancer occurrence. We calculated 95% bootstrap CIs (95% CI) with 500 iterations for all weighted models |
Probably no
Reason: Not all possible concomitant medication per group provided (e.g., MTX use is missing) |
Some concerns |
Table of excluded studies
Reference |
Reason for exclusion |
Ahn SS, Han M, Jung I, Kim HW. Cancers and cardiovascular diseases in patients with seropositive rheumatoid arthritis treated with JAK inhibitors, biologics and conventional synthetic DMARD’s. Clin Exp Rheumatol. 2023 Sep. |
Full text could not be found. |
Ytterberg SR, Bhatt DL, Mikuls TR, Koch GG, Fleischmann R, Rivas JL, Germino R, Menon S, Sun Y, Wang C, Shapiro AB. Cardiovascular and cancer risk with tofacitinib in rheumatoid arthritis. New England Journal of Medicine. 2022 Jan 27;386(4):316-26. |
Results are in the EULAR review. |
Takabayashi K, Ando F, Ikeda K, Nakajima H, Hanaoka H, Suzuki T. Incidence of opportunistic infections in patients with rheumatoid arthritis treated with different molecular-targeted drugs: A population-based retrospective cohort study. Modern Rheumatology. 2022 Oct 29:roac133. |
Incomplete results. No comparative analysis. |
Fleischmann R, Mysler E, Bessette L, Peterfy CG, Durez P, Tanaka Y, Swierkot J, Khan N, Bu X, Li Y, Song IH. Long-term safety and efficacy of upadacitinib or adalimumab in patients with rheumatoid arthritis: results through 3 years from the SELECT-COMPARE study. RMD open. 2022 Feb 1;8(1):e002012. |
No comparative analysis. |
Winthrop KL, Tanaka Y, Takeuchi T, Kivitz A, Matzkies F, Genovese MC, Jiang D, Chen K, Bartok B, Jahreis A, Besuyen R. Integrated safety analysis of filgotinib in patients with moderately to severely active rheumatoid arthritis receiving treatment over a median of 1.6 years. Annals of the rheumatic diseases. 2022 Feb 1;81(2):184-92. |
Safety of filgotinib relative to active comparators adalimumab and methotrexate was not presented as part of this analysis. |
Song YJ, Cho SK, Kim H, Kim HW, Nam E, Choi CB, Kim TH, Jun JB, Bae SC, Yoo DH, Sung YK. Risk factors for herpes zoster in Korean patients with rheumatoid arthritis treated with JAK inhibitor: a nested case–control study. RMD open. 2022 Jan 1;8(1):e001892. |
Wrong comparison: herpes zoster positive group vs herpes zoster negative group |
Huss V, Bower H, Wadström H, Frisell T, Askling J, ARTIS group Ahlenius Gerd-Marie Baecklund Eva Chatzidionysiou Katerina Feltelius Nils Forsblad-d’Elia Helena Kastbom Alf Klareskog Lars Lindqvist Elisabet Lindström Ulf Turesson Carl Sjöwall Christopher Askling Johan. Short-and longer-term cancer risks with biologic and targeted synthetic disease-modifying antirheumatic drugs as used against rheumatoid arthritis in clinical practice. Rheumatology. 2022 May 1;61(5):1810-8. |
Wrong comparison: only data on bDMARD’s vs b/tsDMARD naive patients are presented. |
Burmester GR, Cohen SB, Winthrop KL, Nash P, Irvine AD, Deodhar A, Mysler E, Tanaka Y, Liu J, Lacerda AP, Palac H. Safety profile of upadacitinib over 15 000 patient-years across rheumatoid arthritis, psoriatic arthritis, ankylosing spondylitis and atopic dermatitis. RMD open. 2023 Feb 1;9(1):e002735. |
No comparative analysis and is based on the SELECT-COMPARE trial data which are part of the EULAR review. |
Redeker I, Albrecht K, Kekow J, Burmester GR, Braun J, Schäfer M, Zink A, Strangfeld A. Risk of herpes zoster (shingles) in patients with rheumatoid arthritis under biologic, targeted synthetic and conventional synthetic DMARD treatment: data from the German RABBIT register. Annals of the Rheumatic Diseases. 2022 Jan 1;81(1):41-7. |
Is included in the EULAR review. |
Conaghan P, Cohen S, Burmester G, Mysler E, Nash P, Tanaka Y, Rigby W, Patel J, Shaw T, Betts KA, Patel P. Benefit–Risk Analysis of Upadacitinib Compared with Adalimumab in the Treatment of Patients with Moderate-to-Severe Rheumatoid Arthritis. Rheumatology and Therapy. 2021 Nov 23:1-6. |
Is a post hoc analysis of the SELECT-COMPARE trial, which is part of the EULAR review. |
Atsumi T, Tanaka Y, Matsubara T, Amano K, Ishiguro N, Sugiyama E, Yamaoka K, Westhovens R, Ching DW, Messina OD, Burmester GR. Long-term safety and efficacy of filgotinib treatment for rheumatoid arthritis in Japanese patients naïve to MTX treatment (FINCH 3). Modern Rheumatology. 2023 Jul 1;33(4):657-67. |
Is the Japanese part of the FINCH fase 3 trial. This trial is described in the EULAR review. |
Curtis JR, Yamaoka K, Chen YH, Bhatt DL, Gunay LM, Sugiyama N, Connell CA, Wang C, Wu J, Menon S, Vranic I. Malignancy risk with tofacitinib versus TNF inhibitors in rheumatoid arthritis: results from the open-label, randomised controlled ORAL Surveillance trial. Annals of the Rheumatic Diseases. 2023 Mar 1;82(3):331-43. |
Is a ORAL trial subgroup analysis. Relevant ORAL results are reported in the EULAR review. |
Kristensen LE, Danese S, Yndestad A, Wang C, Nagy E, Modesto I, Rivas J, Benda B. Identification of two tofacitinib subpopulations with different relative risk versus TNF inhibitors: an analysis of the open label, randomised controlled study ORAL Surveillance. Annals of the Rheumatic Diseases. 2023 Jul 1;82(7):901-10. |
Is a ORAL trial subgroup analysis. Relevant ORAL results are reported in the EULAR review. |
Balanescu AR, Citera G, Pascual-Ramos V, Bhatt DL, Connell CA, Gold D, Chen AS, Sawyerr G, Shapiro AB, Pope JE, Schulze-Koops H. Infections in patients with rheumatoid arthritis receiving tofacitinib versus tumour necrosis factor inhibitors: results from the open-label, randomised controlled ORAL Surveillance trial. Annals of the Rheumatic Diseases. 2022 Nov 1;81(11):1491-503. |
Is a ORAL trial subgroup analysis. Relevant ORAL results are reported in the EULAR review. |
Charles-Schoeman C, Buch MH, Dougados M, Bhatt DL, Giles JT, Ytterberg SR, Koch GG, Vranic I, Wu J, Wang C, Kwok K. Risk of major adverse cardiovascular events with tofacitinib versus tumour necrosis factor inhibitors in patients with rheumatoid arthritis with or without a history of atherosclerotic cardiovascular disease: a post hoc analysis from ORAL Surveillance. Annals of the rheumatic diseases. 2023 Jan 1;82(1):119-29. |
Is a ORAL trial post hoc analysis. Relevant ORAL results are reported in the EULAR review. |
Verantwoording
Autorisatiedatum en geldigheid
Laatst beoordeeld : 28-02-2025
Laatst geautoriseerd : 28-02-2025
Geplande herbeoordeling : 28-02-2026
Algemene gegevens
De ontwikkeling/herziening van deze richtlijnmodule werd ondersteund door het Kennisinstituut van de Federatie Medisch Specialisten (www.demedischspecialist.nl/kennisinstituut) en werd gefinancierd uit de Kwaliteitsgelden Medisch Specialisten (SKMS). De financier heeft geen enkele invloed gehad op de inhoud van de richtlijnmodule.
Samenstelling werkgroep
Voor het ontwikkelen van de richtlijnmodule is in 2022 een multidisciplinaire werkgroep ingesteld, bestaande uit vertegenwoordigers van alle relevante specialismen (zie hiervoor de Samenstelling van de werkgroep) die betrokken zijn bij de behandeling van patiënten met biological DMARD’s of targeted synthetic DMARD’s die worden ingezet bij inflammatoire reumatische aandoeningen (o.a. reumatoïde artritis (RA), spondylitis ankylopoietica (SpA), artritis psoriatica (PsA)). De werkgroep realiseert zich dat de middelen ook voor andere indicaties worden ingezet, zoals bij overige auto-immuunziekten, en gaat ervan uit dat in essentie hiervoor de regels van verantwoord gebruik niet anders zullen zijn.
Werkgroep
- Dr. D. (David) ten Cate (voorzitter), reumatoloog, werkzaam in Sint Maartenskliniek, NVR.
- Dr. M. (Marlies) van der Goes, reumatoloog, werkzaam in Meander Medisch Centrum, NVR.
- Dr. P. (Pascal) de Jong, reumatoloog, werkzaam in Erasmus Medisch Centrum, NVR.
- Dr. G.J. (Gerrit Jan) Wolbink, reumatoloog, werkzaam in Reade, NVR.
- Drs. S. (Sadaf) Atiqi, AIOS reumatologie, werkzaam in Reade, NVR.
- R. (René) van der Knaap, patiëntvertegenwoordiger, ReumaZorg Nederland.
- MSc. M.J. (Marieke) van Leijden, patiëntvertegenwoordiger, ReumaNederland.
- S.P. (Silvia) van der Windt, reumaverpleegkundige, werkzaam in Reinier de Graaf ziekenhuis, V&VN.
- Dr. J.C.E.M. (Josianne) ten Berge, oogarts, werkzaam in Erasmus Medisch Centrum, NOG.
- Dr. T. (Thijs) Giezen, ziekenhuisapotheker, werkzaam in Apotheek Spaarne Gasthuis, NVZA.
- Dr. T (Teun) van Gelder, internist – klinisch farmacoloog, werkzaam in Leids Universitair Medisch Centrum, NIV/NVKFB.
Klankbordgroep
- Dr. A.E. (Andrea) van der Meulen – de Jong, maag-darm-leverarts, werkzaam in Leids Universitair Medisch Centrum, NVMDL.
- Drs. B.J.M. (Barbara) Bergmans, arts-microbioloog, werkzaam in Labmicta, NVMM.
Met ondersteuning van
- Dr. T. Hoekstra, senior adviseur (vanaf juni 2024), Kennisinstituut van Federatie Medisch Specialisten.
- Dr. C.L. Overman, adviseur (tot december 2023), Kennisinstituut van de Federatie Medisch Specialisten.
- Dr. B.H. Stegeman, senior adviseur, Kennisinstituut van Federatie Medisch Specialisten.
- Dr. M.M.A. Verhoeven, adviseur, Kennisinstituut van de Federatie Medisch Specialisten.
- Mw. A. van der Wal, medisch informatiespecialist, Kennisinstituut van de Federatie Medisch Specialisten.
Belangenverklaringen
De Code ter voorkoming van oneigenlijke beïnvloeding door belangenverstrengeling is gevolgd. Alle werkgroepleden hebben schriftelijk verklaard of zij in de laatste drie jaar directe financiële belangen (betrekking bij een commercieel bedrijf, persoonlijke financiële belangen, onderzoek financiering) of indirecte belangen (persoonlijke relaties, reputatiemanagement) hebben gehad. Gedurende de ontwikkeling of herziening van een module worden wijzigingen in belangen aan de voorzitter doorgegeven. De belangenverklaring wordt opnieuw bevestigd tijdens de commentaarfase.
Een overzicht van de belangen van werkgroepleden en het oordeel over het omgaan met eventuele belangen vindt u in onderstaande tabel. De ondertekende belangenverklaringen zijn op te vragen bij het secretariaat van het Kennisinstituut van de Federatie Medisch Specialisten.
Werkgroeplid |
Functie |
Nevenfuncties |
Gemelde belangen |
Ondernomen actie |
Dr. D. (David) ten Cate (voorzitter) |
reumatoloog, werkzaam in Sint Maartenskliniek |
Geen |
Geen |
Geen |
Dr. M. (Marlies) van der Goes |
reumatoloog, werkzaam in Meander Medisch Centrum |
Geen |
Geen |
Geen |
Dr. P. (Pascal) de Jong |
reumatoloog, werkzaam in Erasmus Medisch Centrum |
Geen |
Deelgenomen aan diverse adviesraden voor: Abbvie, Lilly; Bristol-Myers Squibb; Pfizer; Sanofi Genzyme; Galapagos; AstraZeneca & UCB. Verzorgen van onderwijs uit naam van de industrie: Galapagos; Pfizer; Novartis; Lilly & Abbvie |
Deelname aan adviesraad neerleggen gedurende richtlijnontwikkeling. Restrictie ten aanzien van besluitvorming van een module follow-up (Deze module gaat over de effectiviteit van de middelen, de andere modules meer over het 'veilig gebruik') |
Dr. G.J. (Gerrit Jan) Wolbink |
reumatoloog, werkzaam in Reade |
Werkzaam bij Sanquin. lid Covid werkgroep NVR Werkgroep Immuungecompromiteerden Covid vaccinatie RIVM. |
Extern gefinancierd onderzoek: ADORA Todora COVIDARC ZonMW |
Restrictie m.b.t. besluitvorming module 'follow up' |
Drs. S. (Sadaf) Atiqi |
AIOS reumatologie, werkzaam in Reade |
Geen |
Extern gefinancierd onderzoek: Therapeutic drug monitoring to optimise treatment with adalimumab in rheumatoid arthritis patients. |
Geen |
R. (René) van der Knaap |
patiëntvertegenwoordiger, ReumaZorg Nederland |
Geen |
Geen |
Geen |
MSc. M.J. (Marieke) van Leijden |
patiëntvertegenwoordiger, ReumaNederland |
Geen |
Geen |
Geen |
S.P. (Silvia) van der Windt |
reumaverpleegkundige, werkzaam in Reinier de Graaf ziekenhuis |
Geen |
Geen |
Geen |
Dr. J.C.E.M. (Josianne) ten Berge |
oogarts, werkzaam in Erasmus Medisch Centrum |
Geen |
Geen |
Geen |
Dr. T. (Thijs) Giezen |
ziekenhuisapotheker, werkzaam in Apotheek Spaarne Gasthuis |
Lid, Biosimilar Working Party, EMA, Amsterdam (onbetaald) t/m 2023 Extern expert CBG, Utrecht (onbetaald) t/m 2023 0-uren aanstelling, Division of Pharmacoepidemiology and Clinical Pharmacology, UIPS, Utrecht (onbetaald)
Vanaf 2024: Lid van de Medische AdviesRaad van Sanquin. De werkzaamheden voor Sanquin betreffen het non-profit gedeelte (de Bloedbank). |
Als co-promotor betrokken geweest bij 2 promotietrajecten welke gefinancierd zijn door het CBG en de Saudi FDA. |
Geen |
Dr. T (Teun) van Gelder |
internist – klinisch farmacoloog, werkzaam in Leids Universitair Medisch Centrum |
Klinisch Farmacoloog bij CCMO. Spreker en consulting vergoedingen ontvangen van Roche Diagnostics, Thermo Fisher, Vitaeris, CSL Behring, Astellas and Aurinia Pharma. In alle gevallen is de donatie aan de ziekenhuis rekening gedaan, dus geen persoonlijke betalingen. Geen aandelen of werkzaamheden voor deze bedrijven. |
Extern gefinancierd onderzoek: Ministerie van VWS, geneesmiddel-ontwikkeling ZonMW, IMProving symptomatic treatment with Amifampridine; a randomized double-blinded, placebo controlled AntiCancer Fund, Adaptive therapy in metastatic castration resistant prostate cancer Medical Delta Talent Acceleration Call 2021 A Biofilm-dissolving Gel to Eradicate Cardiac Bacterial Infections during Surgery KNMP, Klinisch redeneren door de apotheker |
Deelname aan adviesraad neerleggen gedurende richtlijnontwikkeling. Geen aanvullende restricties. |
Inbreng patiëntenperspectief
Er werd aandacht besteed aan het patiëntenperspectief door afgevaardigden van patiëntenorganisatiesin de werkgroep. De opzet van de module Organisatie van Zorg is in samenspraak met de patiëntenorganisaties opgezet. De conceptrichtlijn is tevens voor commentaar voorgelegd aan verschillende patiëntenverenigingen en de eventueel aangeleverde commentaren zijn bekeken en verwerkt.
Wkkgz & Kwalitatieve raming van mogelijke substantiële financiële gevolgen
Kwalitatieve raming van mogelijke financiële gevolgen in het kader van de Wet kwaliteit, klachten en geschillen zorg (Wkkgz).
Bij de richtlijn is conform de Wkkgz een kwalitatieve raming uitgevoerd of de aanbevelingen mogelijk leiden tot substantiële financiële gevolgen. Bij het uitvoeren van deze beoordeling zijn richtlijnmodules op verschillende domeinen getoetst (zie het stroomschema op de Richtlijnendatabase).
Uit de kwalitatieve raming blijkt dat er waarschijnlijk geen substantiële negatieve financiële gevolgen zijn, zie onderstaande tabel.
Module |
Uitkomst raming |
Toelichting |
Complicaties tsDMARD’s – allergische reacties |
Geen substantiële financiële gevolgen. |
Hoewel uit de toetsing volgt dat de aanbeveling(en) breed toepasbaar zijn (tussen 5.000-40.000), volgt ook uit de toetsing dat het geen nieuwe manier van zorgverlening of andere organisatie van zorgverlening betreft. Er worden daarom geen substantiële financiële gevolgen verwacht. |
Werkwijze
AGREE
Deze richtlijnmodule is opgesteld conform de eisen vermeld in het rapport Medisch Specialistische Richtlijnen 2.0 van de adviescommissie Richtlijnen van de Raad Kwaliteit. Dit rapport is gebaseerd op het AGREE II instrument (Appraisal of Guidelines for Research & Evaluation II; Brouwers, 2010).
Knelpuntenanalyse en uitgangsvragen
Tijdens de voorbereidende fase inventariseerde de werkgroep de knelpunten in de huidige zorg middels een schriftelijke knelpuntenanalyse. Tijdens deze knelpuntenanalyse werd aan vertegenwoordigers vanuit verschillende organisaties gevraagd hun input te leveren met als doel te inventariseren welke knelpunten men ervaarde rondom de te ontwikkelen richtlijn. De resultaten hiervan zijn opgenomen onder aanverwante producten (zie Bijlage Reacties schriftelijke knelpunteninventarisatie). Op basis van de uitkomsten van de knelpuntenanalyse zijn door de werkgroep uitgangsvragen opgesteld en definitief vastgesteld.
Uitkomstmaten
Na het opstellen van de zoekvraag behorende bij de uitgangsvraag inventariseerde de werkgroep welke uitkomstmaten voor de patiënt relevant zijn, waarbij zowel naar gewenste als ongewenste effecten werd gekeken. De werkgroep waardeerde deze uitkomstmaten volgens hun relatieve belang bij de besluitvorming rondom aanbevelingen, als cruciaal (kritiek voor de besluitvorming), belangrijk (maar niet cruciaal) en onbelangrijk. Tevens definieerde de werkgroep tenminste voor de cruciale uitkomstmaten welke verschillen zij klinisch en/of patiëntrelevant vonden.
Methode literatuursamenvatting
Een uitgebreide beschrijving van de strategie voor zoeken en selecteren van literatuur en de beoordeling van de risk-of-bias van de individuele studies is te vinden onder ‘Search and select’. De beoordeling van de kracht van het wetenschappelijke bewijs wordt hieronder toegelicht.
Beoordelen van de kracht van het wetenschappelijke bewijs
De kracht van het wetenschappelijke bewijs werd bepaald volgens de GRADE-methode. GRADE staat voor ‘Grading Recommendations Assessment, Development and Evaluation’ (zie http://www.gradeworkinggroup.org/). De basisprincipes van de GRADE-methodiek zijn: het benoemen en prioriteren van de klinisch (patiënt) relevante uitkomstmaten, een systematische review per uitkomstmaat, en een beoordeling van de bewijskracht per uitkomstmaat op basis van de acht GRADE-domeinen (domeinen voor downgraden: risk of bias, inconsistentie, indirectheid, imprecisie, en publicatiebias; domeinen voor upgraden: dosis-effect relatie, groot effect, en residuele plausibele confounding).
GRADE onderscheidt vier gradaties voor de kwaliteit van het wetenschappelijk bewijs: hoog, redelijk, laag en zeer laag. Deze gradaties verwijzen naar de mate van zekerheid die er bestaat over de literatuurconclusie, in het bijzonder de mate van zekerheid dat de literatuurconclusie de aanbeveling adequaat ondersteunt (Schünemann, 2013; Hultcrantz, 2017).
GRADE |
Definitie |
Hoog |
|
Redelijk |
|
Laag |
|
Zeer laag |
|
Bij het beoordelen (graderen) van de kracht van het wetenschappelijk bewijs in richtlijnen volgens de GRADE-methodiek spelen grenzen voor klinische besluitvorming een belangrijke rol (Hultcrantz, 2017). Dit zijn de grenzen die bij overschrijding aanleiding zouden geven tot een aanpassing van de aanbeveling. Om de grenzen voor klinische besluitvorming te bepalen moeten alle relevante uitkomstmaten en overwegingen worden meegewogen. De grenzen voor klinische besluitvorming zijn daarmee niet een-op-een vergelijkbaar met het minimaal klinisch relevant verschil (Minimal Clinically Important Difference, MCID). Met name in situaties waarin een interventie geen belangrijke nadelen heeft en de kosten relatief laag zijn, kan de grens voor klinische besluitvorming met betrekking tot de effectiviteit van de interventie bij een lagere waarde (dichter bij het nuleffect) liggen dan de MCID (Hultcrantz, 2017).
Overwegingen (van bewijs naar aanbeveling)
Om te komen tot een aanbeveling zijn naast (de kwaliteit van) het wetenschappelijke bewijs ook andere aspecten belangrijk en worden meegewogen, zoals aanvullende argumenten uit bijvoorbeeld de biomechanica of fysiologie, waarden en voorkeuren van patiënten, kosten (middelenbeslag), aanvaardbaarheid, haalbaarheid en implementatie. Deze aspecten zijn systematisch vermeld en beoordeeld (gewogen) onder het kopje ‘Overwegingen’ en kunnen (mede) gebaseerd zijn op expert-opinie. Hierbij is gebruik gemaakt van een gestructureerd format gebaseerd op het evidence-to-decision framework van de internationale GRADE Working Group (Alonso-Coello, 2016a; Alonso-Coello 2016b). Dit evidence-to-decision framework is een integraal onderdeel van de GRADE methodiek.
Formuleren van aanbevelingen
De aanbevelingen geven antwoord op de uitgangsvraag en zijn gebaseerd op het beschikbare wetenschappelijke bewijs en de belangrijkste overwegingen, en een weging van de gunstige en ongunstige effecten van de relevante interventies. De kracht van het wetenschappelijk bewijs en het gewicht dat door de werkgroep wordt toegekend aan de overwegingen, bepalen samen de sterkte van de aanbeveling. Conform de GRADE-methodiek sluit een lage bewijskracht van conclusies in de systematische literatuuranalyse een sterke aanbeveling niet a priori uit, en zijn bij een hoge bewijskracht ook zwakke aanbevelingen mogelijk (Agoritsas, 2017; Neumann, 2016). De sterkte van de aanbeveling wordt altijd bepaald door weging van alle relevante argumenten tezamen. De werkgroep heeft bij elke aanbeveling opgenomen hoe zij tot de richting en sterkte van de aanbeveling zijn gekomen.
In de GRADE-methodiek wordt onderscheid gemaakt tussen sterke en zwakke (of conditionele) aanbevelingen. De sterkte van een aanbeveling verwijst naar de mate van zekerheid dat de voordelen van de interventie opwegen tegen de nadelen (of vice versa), gezien over het hele spectrum van patiënten waarvoor de aanbeveling is bedoeld. De sterkte van een aanbeveling heeft duidelijke implicaties voor patiënten, behandelaars en beleidsmakers (zie onderstaande tabel). Een aanbeveling is geen dictaat, zelfs een sterke aanbeveling gebaseerd op bewijs van hoge kwaliteit (GRADE gradering HOOG) zal niet altijd van toepassing zijn, onder alle mogelijke omstandigheden en voor elke individuele patiënt.
Implicaties van sterke en zwakke aanbevelingen voor verschillende richtlijngebruikers |
||
|
Sterke aanbeveling |
Zwakke (conditionele) aanbeveling |
Voor patiënten |
De meeste patiënten zouden de aanbevolen interventie of aanpak kiezen en slechts een klein aantal niet. |
Een aanzienlijk deel van de patiënten zouden de aanbevolen interventie of aanpak kiezen, maar veel patiënten ook niet. |
Voor behandelaars |
De meeste patiënten zouden de aanbevolen interventie of aanpak moeten ontvangen. |
Er zijn meerdere geschikte interventies of aanpakken. De patiënt moet worden ondersteund bij de keuze voor de interventie of aanpak die het beste aansluit bij zijn of haar waarden en voorkeuren. |
Voor beleidsmakers |
De aanbevolen interventie of aanpak kan worden gezien als standaardbeleid. |
Beleidsbepaling vereist uitvoerige discussie met betrokkenheid van veel stakeholders. Er is een grotere kans op lokale beleidsverschillen. |
Organisatie van zorg
Meer algemene, overkoepelende, of bijkomende aspecten van de organisatie van zorg worden behandeld in de module organisatie van zorg.
Commentaar- en autorisatiefase
De conceptrichtlijnmodule werd aan de betrokken (wetenschappelijke) verenigingen en (patiënt) organisaties voorgelegd ter commentaar. De commentaren werden verzameld en besproken met de werkgroep. Naar aanleiding van de commentaren werd de conceptrichtlijnmodule aangepast en definitief vastgesteld door de werkgroep. De definitieve richtlijnmodule werd aan de deelnemende (wetenschappelijke) verenigingen en (patiënt)organisaties voorgelegd voor autorisatie en door hen geautoriseerd dan wel geaccordeerd.
Literatuur
Agoritsas T, Merglen A, Heen AF, Kristiansen A, Neumann I, Brito JP, Brignardello-Petersen R, Alexander PE, Rind DM, Vandvik PO, Guyatt GH. UpToDate adherence to GRADE criteria for strong recommendations: an analytical survey. BMJ Open. 2017 Nov 16;7(11):e018593. doi: 10.1136/bmjopen-2017-018593. PubMed PMID: 29150475; PubMed Central PMCID: PMC5701989.
Alonso-Coello P, Schünemann HJ, Moberg J, Brignardello-Petersen R, Akl EA, Davoli M, Treweek S, Mustafa RA, Rada G, Rosenbaum S, Morelli A, Guyatt GH, Oxman AD; GRADE Working Group. GRADE Evidence to Decision (EtD) frameworks: a systematic and transparent approach to making well informed healthcare choices. 1: Introduction. BMJ. 2016 Jun 28;353:i2016. doi: 10.1136/bmj.i2016. PubMed PMID: 27353417.
Alonso-Coello P, Oxman AD, Moberg J, Brignardello-Petersen R, Akl EA, Davoli M, Treweek S, Mustafa RA, Vandvik PO, Meerpohl J, Guyatt GH, Schünemann HJ; GRADE Working Group. GRADE Evidence to Decision (EtD) frameworks: a systematic and transparent approach to making well informed healthcare choices. 2: Clinical practice guidelines. BMJ. 2016 Jun 30;353:i2089. doi: 10.1136/bmj.i2089. PubMed PMID: 27365494.
Brouwers MC, Kho ME, Browman GP, Burgers JS, Cluzeau F, Feder G, Fervers B, Graham ID, Grimshaw J, Hanna SE, Littlejohns P, Makarski J, Zitzelsberger L; AGREE Next Steps Consortium. AGREE II: advancing guideline development, reporting and evaluation in health care. CMAJ. 2010 Dec 14;182(18):E839-42. doi: 10.1503/cmaj.090449. Epub 2010 Jul 5. Review. PubMed PMID: 20603348; PubMed Central PMCID: PMC3001530.
Hultcrantz M, Rind D, Akl EA, Treweek S, Mustafa RA, Iorio A, Alper BS, Meerpohl JJ, Murad MH, Ansari MT, Katikireddi SV, Östlund P, Tranæus S, Christensen R, Gartlehner G, Brozek J, Izcovich A, Schünemann H, Guyatt G. The GRADE Working Group clarifies the construct of certainty of evidence. J Clin Epidemiol. 2017 Jul;87:4-13. doi: 10.1016/j.jclinepi.2017.05.006. Epub 2017 May 18. PubMed PMID: 28529184; PubMed Central PMCID: PMC6542664.
Medisch Specialistische Richtlijnen 2.0 (2012). Adviescommissie Richtlijnen van de Raad Kwalitieit. http://richtlijnendatabase.nl/over_deze_site/over_richtlijnontwikkeling.html
Neumann I, Santesso N, Akl EA, Rind DM, Vandvik PO, Alonso-Coello P, Agoritsas T, Mustafa RA, Alexander PE, Schünemann H, Guyatt GH. A guide for health professionals to interpret and use recommendations in guidelines developed with the GRADE approach. J Clin Epidemiol. 2016 Apr;72:45-55. doi: 10.1016/j.jclinepi.2015.11.017. Epub 2016 Jan 6. Review. PubMed PMID: 26772609.
Schünemann H, Brożek J, Guyatt G, et al. GRADE handbook for grading quality of evidence and strength of recommendations. Updated October 2013. The GRADE Working Group, 2013. Available from http://gdt.guidelinedevelopment.org/central_prod/_design/client/handbook/handbook.html.
Zoekverantwoording
Zoekacties zijn opvraagbaar. Neem hiervoor contact op met de Richtlijnendatabase.