Skip to main content

Safety outcomes in patients with rheumatoid arthritis treated with abatacept: results from a multinational surveillance study across seven European registries

Abstract

Background

Patients with rheumatoid arthritis (RA) have an increased risk of infection and malignancy compared with the general population. Infection risk is increased further with the use of disease-modifying antirheumatic drugs (DMARDs), whereas evidence on whether the use of biologic DMARDs increases cancer risk remains equivocal. This single-arm, post-marketing study estimated the incidence of prespecified infection and malignancy outcomes in patients with RA treated with intravenous or subcutaneous abatacept.

Methods

Data were included from seven European RA quality registries: ATTRA (Anti-TNF Therapy in Rheumatoid Arthritis [Czech Republic]), DANBIO (Danish Rheumatologic Database), ROB-FIN (National Registry of Antirheumatic and Biological Treatment in Finland), ORA (Orencia and Rheumatoid Arthritis [France]), GISEA (Italian Group for the Study of Early Arthritis), BIOBADASER (Spanish Register of Adverse Events of Biological Therapies in Rheumatic Diseases), and the SCQM (Swiss Clinical Quality Management) system. Each registry is unique with respect to design, data collection, definition of the study cohort, reporting, and validation of outcomes. In general, registries defined the index date as the first day of abatacept treatment and reported data for infections requiring hospitalization and overall malignancies; data for other infection and malignancy outcomes were not available for every cohort. Abatacept exposure was measured in patient-years (p-y). Incidence rates (IRs) were calculated as the number of events per 1000 p-y of follow-up with 95% confidence intervals.

Results

Over 5000 patients with RA treated with abatacept were included. Most patients (78–85%) were female, and the mean age range was 52–58 years. Baseline characteristics were largely consistent across registries. Among patients treated with abatacept, IRs for infections requiring hospitalization across the registries ranged from 4 to 100 events per 1000 p-y, while IRs for overall malignancy ranged from 3 to 19 per 1000 p-y.

Conclusions

Despite heterogeneity between registries in terms of design, data collection, and ascertainment of safety outcomes, as well as the possibility of under-reporting of adverse events in observational studies, the safety profile of abatacept reported here was largely consistent with previous findings in patients with RA treated with abatacept, with no new or increased risks of infection or malignancy.

Introduction

Compared with the general population, patients with rheumatoid arthritis (RA) are at an increased risk of certain events, specifically infections and malignancies [1,2,3,4]. Patients with RA are at a modestly increased risk of malignancy overall—for lung cancer and lymphoma, in particular—but at a decreased risk of colorectal and breast cancers [3, 4]. Data suggest that the risk of infections in patients with RA is further increased with the use of conventional synthetic (cs) disease-modifying antirheumatic drugs (DMARDs) and biologic (b)/targeted synthetic (ts) DMARDs [1, 2]. Although there has been no finding that the risk of malignancies with the use of b/tsDMARDs is increased in the RA population, evidence regarding long-term safety continues to emerge [2, 5,6,7,8,9,10,11,12,13,14].

The bDMARD abatacept, a selective T cell co-stimulation modulator that blocks the interaction between CD80/CD86 on antigen-presenting cells and CD28 on T cells [15], is approved for the treatment of RA [16, 17]. Abatacept is effective in reducing disease activity, preventing or slowing radiologic disease progression, and improving physical function and health-related quality of life in adult patients with early [18, 19] and established [15, 20,21,22,23,24,25] RA. Data from randomized, controlled clinical trials have shown abatacept to be well tolerated, with adverse event rates similar to those of placebo, and to have a consistent and favourable safety profile over the longer term [15, 26,27,28]. A recently published, integrated safety analysis for abatacept included data from nine clinical trials in adult patients with RA treated with abatacept (n = 2653) or placebo (n = 1485) for a mean (standard deviation) duration of exposure of 10.8 (3.3) and 10.3 (3.5) months, respectively [29]. The study showed that the incidence rates (IRs) for overall infections, serious infections/infestations, opportunistic infections, and malignancies were similar between the abatacept and placebo groups.

An abatacept global post-marketing epidemiology programme was initiated in 2006 in North America and 2008 in Europe to monitor the safety, specifically infections and malignancies, of abatacept using real-world data sources. The programme included observational data from a geographically diverse group of patients and clinical practices gathered in bDMARD or disease registries in Europe and North America, as well as administrative healthcare claims databases from North America. A retrospective, observational study using administrative data from three large US healthcare databases showed a slightly but statistically significantly increased risk in total malignancies but no increased risk for specific cancers (lung, breast, lymphoma), infections requiring hospitalization, or opportunistic infections with abatacept versus b/tsDMARDs [30]. In a cohort study, the use of abatacept was associated with an increased incidence of cancer overall and a significantly increased incidence of non-melanoma skin cancer compared with other bDMARDs [11]. Additionally, a recent large pooled analysis of randomized trials demonstrated no increased risk of infections with abatacept compared with non-bDMARDs [31]. An observational study using VigiBase, the World Health Organization’s global database of individual case safety reports, showed that exposure to abatacept (compared with other bDMARDs) in patients with RA was significantly associated with an increased risk of reporting melanoma [32]. In a register-based prospective cohort study of patients with RA, abatacept was associated with an increased risk of developing squamous cell skin cancer compared with TNFi [7].

The present study was designed to complement the abatacept global post-marketing epidemiology programme, comply with the European regulatory authority requirements, and contribute to the body of evidence on the safety of abatacept. The study used data from seven European-based RA registries to estimate the incidence of prespecified outcomes, namely infections and malignancies, in patients with RA treated with abatacept.

Methods

Study design and data sources

This single-arm study included data from patients treated with intravenous or subcutaneous abatacept taken from seven established longitudinal observational RA registries based in Europe: ATTRA (Anti-TNF Therapy in Rheumatoid Arthritis [Czech Republic]), DANBIO (Danish Rheumatologic Database), ROB-FIN (National Registry of Antirheumatic and Biological Treatment in Finland), ORA (Orencia and Rheumatoid Arthritis [France]), GISEA (Italian Group for the Study of Early Arthritis), BIOBADASER (Spanish Register of Adverse Events of Biological Therapies in Rheumatic Diseases), and the SCQM (Swiss Clinical Quality Management) system.

The study was conducted initially using data from ATTRA, DANBIO, ROB-FIN, and SCQM, with the addition of three registries—ORA, GISEA, and BIOBADASER—in 2014. Each data source is unique with respect to the design, data collection, reporting, and validation of outcomes. The characteristics of each registry are shown in Table 1.

Table 1 Characteristics of registries used for identifying patients with RA in the post-marketing epidemiology abatacept study

This study was conducted in accordance with the International Society for Pharmacoepidemiology Guidelines for Good Pharmacoepidemiology Practices, applicable regulatory requirements, and the principles of the Declaration of Helsinki.

Patient population

Patients diagnosed with RA who had a record of treatment with abatacept and follow-up information available were eligible for inclusion in the study. The definition of the study cohort for each registry is included in Table 1. Patient identification, eligibility criteria, and definition of study cohorts that qualified a patient for inclusion varied between the individual registries.

Outcomes

Each registry was unique with respect to the outcomes reported; thus, not all outcomes are available for every cohort (Table 1). All seven registries reported data for infections requiring hospitalization and overall malignancies. For the GISEA registry, infections requiring hospitalization and malignancies were reported as adverse event counts. For specific infection outcomes, opportunistic infections were reported only for the ATTRA, DANBIO, and BIOBADASER registries; tuberculosis was reported by all registries apart from ORA and GISEA. For specific malignancy outcomes, lymphoma, breast, and lung cancers were reported by all registries apart from GISEA. For the ORA registry, the occurrence of breast cancer, lung cancer, and lymphoma was reported in the patient adverse event summary narratives. The exclusion criteria for patients with a history of cancer prior to abatacept exposure were not standardized in this study; however, in the DANBIO registry, patients who had previous diagnoses of cancer (except non-melanoma skin cancer) were excluded from the IR calculations.

Exposure

The calculation of abatacept exposure was computed by the individual registries. In general, the index date was the first day of treatment with abatacept and exposure was measured in patient-years (p-y).

For infections, patient time was calculated from the index date until the event of interest, the end of the newly initiated treatment plus 90-day lag time, death, emigration (DANBIO), the end of the study, or the last follow-up date (if earlier than the 90-day lag time in the ORA registry), whichever occurred first.

As malignancies often have latency periods, the risk associated with a specific drug is expected to continue beyond the immediate period of exposure to any treatment. Thus, patient time of exposure to abatacept for malignancies was extended to the end of the follow-up period regardless of whether the patient discontinued the drug. In addition, different registries calculated exposure time for malignancy slightly differently (Additional file 1).

Statistical analyses

Within each registry, baseline demographics and clinical characteristics were computed with descriptive statistics for categorical and continuous variables.

There was some variation in the methodology for the calculation of IRs for each registry (Additional file 1). IRs for each prespecified outcome were provided by the individual registries as the number of events per 1000 total p-y of follow-up with 95% confidence intervals (CIs), except for all IRs reported for GISEA and for some malignancy IRs reported for ORA. For GISEA, IRs were calculated using the mean duration of therapy (in months) and adverse event counts for infections requiring hospitalization and overall malignancies. For ORA, IRs for breast cancer, lung cancer, and lymphoma were based on patient narrative data and p-y provided; follow-up for incidence of cancers is the overall follow-up period. CIs were not provided by the ORA or GISEA registries for any of the IRs.

Results

Patient demographics and clinical characteristics

Over 5000 patients with RA treated with abatacept were included in this study (Table 2). Most patients (78–85%) were female, and the mean age ranged from 52.1 to 58.0 years across all registries. Baseline characteristics were largely consistent across the seven different registries, apart from a longer disease duration in the ORA registry. The mean duration of RA ranged from 8.1 to 14.0 years across the seven registries.

Table 2 Baseline demographics and clinical characteristics of patients with RA in the post-marketing epidemiology abatacept study

Infections

Among patients treated with abatacept, the IRs for infections requiring hospitalization ranged from 4 to 100 per 1000 p-y (Table 3, Fig. 1A). The highest rates were observed in the DANBIO registry, with an IR of 100 per 1000 p-y (95% CI: 87, 115), corresponding to 196 events in 1213 patients with 1955 p-y of exposure. The lowest IRs were observed in the SCQM (4 per 1000 p-y [95% CI: 2, 8]) and ATTRA (6 per 1000 p-y [95% CI: 0, 12]) registries.

Table 3 Incidence rates/1000 patient-years (95% CI), number of events, and patient-years of exposure for infections
Fig. 1
figure 1

Incidence rates/1000 patient-years (95% CI) for A infections requiring hospitalization and B overall malignancy. For GISEA, IRs were calculated using the mean duration of therapy (in months) and adverse event counts for infections requiring hospitalization and overall malignancies; for overall malignancy, the calculated IR used first- and second-line abatacept adverse event counts from November 2015. No CIs were provided by the ORA or GISEA registries for any of the IRs. For SCQM, IRs are by exposure group with mid-imputed dates of occurrence. ATTRA, Anti-TNF Therapy in Rheumatoid Arthritis; BIOBADASER, Spanish Register of Adverse Events of Biological Therapies in Rheumatic Diseases; DANBIO, Danish Rheumatologic Database; CI, confidence interval; GISEA, Italian Group for the Study of Early Arthritis; IR, incidence rate; ORA, Orencia and Rheumatoid Arthritis; p-y, patient-years; ROB-FIN, National Registry of Antirheumatic and Biological Treatment in Finland; SCQM, Swiss Clinical Quality Management

The IRs for opportunistic infections ranged from 0 to 14 per 1000 p-y (Table 3). In BIOBADASER, the IR was 14 per 1000 p-y (95% CI: 7, 27), corresponding to 8 events in 350 patients with 578 p-y of exposure. In DANBIO, there were two cases of opportunistic infections occurring in 1213 patients with 2233 p-y of exposure. There were no occurrences of opportunistic infections reported in the ATTRA registry.

The IRs for tuberculosis ranged from 0 to 6 per 1000 p-y with the number of events ranging from 0 in the ROB-FIN and BIOBADASER registries to 5 in the ATTRA registry (Table 3).

Malignancies

The IRs for overall malignancies in patients treated with abatacept were low and ranged from 3 to 19 per 1000 p-y (Table 4, Fig. 1B). The highest rates were observed in the DANBIO registry with an IR of 19 per 1000 p-y (95% CI: 15, 24), corresponding to 70 events in 1213 patients with 3642 p-y of exposure.

Table 4 Incidence rates/1000 patient-years (95% CI), number of events, and patient-years of exposure for malignancies

The IRs for breast cancer ranged from 1.7 to 2 per 1000 p-y (Table 4). The IRs for lung cancer and lymphoma were low and ranged from 1 to 2 and 0 to 1 per 1000 p-y, respectively (Table 4).

Discussion

This real-world observational study of over 5000 patients with RA treated with abatacept showed a range in rates of infection and malignancy events across the included registries. No new or increased risks of infection or malignancy with abatacept were identified. These findings are consistent with the safety profile of abatacept reported in adult patients with active RA in multiple randomized controlled clinical trials, large international observational trials, and several extensive real-world data sources from the literature [29,30,31, 35].

The IRs of infections requiring hospitalization observed with abatacept in the present study ranged from 4 to 100 per 1000 p-y. A recent study from the abatacept global post-marketing epidemiology programme included observational data from five large international data sources including a total of 6450 patients treated with abatacept, reported the following IRs of infections requiring hospitalization for patients with RA treated with abatacept, csDMARDs, or b/tsDMARDs: 16–56/1000 p-y, 19–46/1000 p-y, and 18–40/1000 p-y, respectively [31]. Compared with these data, the IRs for infections requiring hospitalization in the present study were lower in the ATTRA (6/1000 p-y) and SCQM (4/1000 p-y) registries but higher in the DANBIO (100/1000 p-y) and ROB-FIN (84/1000 p-y) registries. Methodologic differences across the seven registries in this study included the determination of index date, data collection methods, definition of outcomes, and reporting of adverse events. The differences in methodology may have contributed to the wide range of IRs observed. Overall, with the exception of DANBIO, the IRs for infections requiring hospitalization reported in the present study were consistent with reference ranges published previously from the abatacept global post-marketing epidemiology programme [31]. In DANBIO, the higher rates of infections requiring hospitalization may be explained by linkage to the Danish National Patient Registry, which represents a nearly complete Danish population and, therefore, ensured that a high rate of adverse events was captured. Another observational cohort study (2010–2015), which included data from DANBIO and the Anti-Rheumatic Treatment in Sweden Register/Swedish Rheumatology Quality Register, investigated the risk of serious (hospitalized) infections in patients with RA who started treatment with non-tumour necrosis factor (TNF) inhibitor bDMARDs [36]. Over the first 12 months, crude IRs for serious infections ranged from 61 to 70/1000 p-y for abatacept, 71 to 88/1000 p-y for rituximab, and 46 to 61/1000 p-y for tocilizumab, with the Danish rates being consistently higher than the Swedish rates [36].

For other infection outcomes reported in the present study, the IRs for opportunistic infections were low in ATTRA (0/1000 p-y) and DANBIO (1/1000 p-y) and consistent with recently published data for abatacept (0.4–7.8/1000 p-y), csDMARDs (0.3–4.3/1000 p-y), and b/tsDMARDs (0.5–3.8/1000 p-y) [31] but were higher in BIOBADASER (14/1000 p-y). Additionally, IRs of tuberculosis in this study were low (0–1.5/1000 p-y) across four of the five registries that reported data but were higher (IR: 6/1000 p-y) in the ATTRA database. These findings may, in part, be reflective of the variability in the incidence of tuberculosis across Europe [37]. Data from the abatacept global post-marketing epidemiology programme reported IRs of tuberculosis per 1000 p-y of 0.0–8.4, 0.0–6.0, and 0.0–6.3 in patients receiving treatment with abatacept, csDMARDs, and b/tsDMARDs, respectively (data on file). Data from the British Society for Rheumatology Biologics Register (BSRBR), a national prospective observational study comparing rates of tuberculosis in more than 10,000 patients treated with a TNF inhibitor, reported crude IRs of ~ 1.4/1000 p-y [38], while a retrospective cohort study from Taiwan (n = 951) reported IRs of tuberculosis of 0.9 and 1.1 per 1000 p-y in patients treated with etanercept and adalimumab, respectively [33]. Another larger retrospective study using data from Taiwan’s National Health Insurance Database reported rates of tuberculosis of 0.55/1000 p-y in 7888 patients with RA treated with a tsDMARD, 1.5/1000 p-y in 3459 patients treated with a bDMARD, and rates of 1.2/1000 p-y for etanercept and 2.9/1000 p-y for adalimumab specifically [34]. Overall, the data reported here are consistent with previously reported data for abatacept, csDMARDs, and other b/tsDMARDs [31, 33, 34, 38].

Similar to the results observed for infections, there was a wide variability in the incidence of overall malignancies across the seven registries included in the present study. Again, this variability may be explained by methodologic differences between the different data sources. Furthermore, although the IR of overall malignancy reported in the DANBIO registry was higher than expected, despite the vast methodologic differences, the IRs for the remaining six registries were similar to those from a recent observational study from the abatacept global post-marketing epidemiology programme that reported IRs/1000 p-y for overall malignancy of 8–11 for abatacept, 9–13 for csDMARDs, and 5–12 for b/tsDMARDs [35]. Data from the present study are also consistent with data reported in an integrated analysis of data from nine randomized, controlled clinical trials, where IRs of malignancy were 13/1000 p-y for patients treated with abatacept versus 14/1000 p-y for patients receiving placebo [29]. Other studies have reported variability in the incidence of overall malignancy with different therapeutic agents used in the treatment of patients with RA. A 2016 study from the BSRBR reported rates of incident malignancy of 33.3 events/1000 p-y for patients treated with a TNF inhibitor, 24.7/1000 p-y for rituximab, and 53.8/1000 p-y for those treated with a non-bDMARD [39]. Other observational studies also reported an increased risk of total malignancies [30] and skin cancers [7, 32] in patients with RA exposed to abatacept. Recently, another Swedish observational study noted a potential increased overall cancer risk in patients with RA treated for 2–5 years with abatacept (IR of 13.6 1000 p-y compared to the general population) [12], but the models presented in the study did not account for the number of prior biologics. There is evidence showing that patients with RA who received abatacept have been exposed to, on average, more than 2 biologic treatments [unpublished data]; this cumulative exposure to biologics may result in an increase in cancer risk [unpublished data].

In contrast, other studies have reported much lower rates of overall malignancy, including IRs of 5.4/1000 p-y observed with bDMARDs versus 7.4/1000 p-y with non-bDMARD treatment in the Taiwan National Health Insurance Research Database [40] and 8.1/1000 p-y with TNF inhibitor therapy and 11.7/1000 p-y with non-bDMARDs (including methotrexate) in another study from the BSRBR [8].

In terms of specific types of cancer, the IRs reported here for breast cancer (1.7–2/1000 p-y), lung cancer (1–2/1000 p-y), and lymphoma (0–1/1000 p-y) are within the expected ranges for abatacept based on previously reported data from the abatacept global post-marketing epidemiology programme (IRs/1000 p-y were 0–4.4 for breast cancer, 0.1–2.8 for lung cancer, and 0–1.1 for lymphoma) [35]. This previous study, which included data from over 110,000 patients across geographically diverse data sources, demonstrated similar rates of breast cancer, lung cancer, and lymphoma among patients treated with abatacept, csDMARDs, and other b/tsDMARDs [35]. It is important to note, however, that issues of channelling bias, differences in methodology between studies, and regional differences in healthcare systems, such as cancer screening methods and surveillance programmes, make direct comparisons of malignancy rates between DMARD cohorts challenging.

The data reported here contribute to the existing body of evidence on the safety of abatacept from randomized controlled trials and the post-marketing epidemiology abatacept programme. The strength of the present study lies in its provision of additional information on the safety of abatacept in over 5000 patients with RA from multiple databases across Europe. A geographically diverse group of patients and clinical practices are represented, with data reported for about 10 years for most of the registries included in the analysis. Baseline characteristics were similar across databases, confirming the validity of combining data. Although these analyses are not adjusted for age or other factors, a patient population of this size and breadth suggests the generalizability of the result.

The lack of a control group was a limitation of this study. Furthermore, many of the included registries were in early development at study initiation (in 2008) with less comprehensive data collection or fewer analysis options (e.g., comparator analyses) than are currently available. In addition, there was considerable variability among the registries in terms of design, determination of index date, completeness and manner of recording of concomitant treatments and known risk factors for serious infections or malignancies, data collection methods, computation of p-y exposed, definition/reporting of outcomes, validation of events, and calculation of IRs—all of which may have resulted in the wide range of IRs observed. This heterogeneity, as well as the possibility of under-reporting of adverse events in observational studies, in general, should be considered when assessing the potential clinical impact of these data.

Conclusions

In this collaborative, international, real-world observational study using data from existing RA registries, no new or increased risks of infection or malignancy were identified for abatacept in the treatment of patients with RA. Despite the heterogeneity between registries, the data reported are consistent with the overall known safety profile for abatacept. Overall, most IRs were consistent with previously reported reference ranges.

Availability of data and materials

Bristol Myers Squibb policy on data sharing may be found at https://www.bms.com/researchers-and-partners/clinical-trials-and-research/disclosure-commitment.html.

Abbreviations

ATTRA:

Anti-TNF Therapy in Rheumatoid Arthritis

bDMARD:

Biologic disease-modifying antirheumatic drug

BIOBADASER:

Spanish Register of Adverse Events of Biological Therapies in Rheumatic Diseases

BSRBR:

British Society for Rheumatology Biologics Register

CI:

Confidence interval

csDMARD:

Conventional synthetic disease-modifying antirheumatic drug

DANBIO:

Danish Rheumatologic Database

DMARD:

Disease-modifying antirheumatic drug

GISEA:

Italian Group for the Study of Early Arthritis

HILMO:

Finnish Nationwide Social and Healthcare Data Collection and Reporting System

ICD(-10):

International Classification of Diseases (Tenth Edition)

IR:

Incidence rate

MAH:

Marketing authorization holder

MedDRA:

Medical Dictionary for Drug Regulatory Activities

NR:

Not reported

ORA:

Orencia and Rheumatoid Arthritis

p-y:

Patient-years

RA:

Rheumatoid arthritis

ROB-FIN:

National Registry of Antirheumatic and Biological Treatment in Finland

SCQM:

Swiss Clinical Quality Management

SD:

Standard deviation

TNF:

Tumour necrosis factor

tsDMARD:

Targeted synthetic disease-modifying antirheumatic drug

References

  1. Winthrop KL. Infections and biologic therapy in rheumatoid arthritis: our changing understanding of risk and prevention. Rheum Dis Clin North Am. 2012;38:727–45.

    Article  PubMed  Google Scholar 

  2. Bongartz T, Sutton AJ, Sweeting MJ, Buchan I, Matteson EL, Montori V. Anti-TNF antibody therapy in rheumatoid arthritis and the risk of serious infections and malignancies: systematic review and meta-analysis of rare harmful effects in randomized controlled trials. JAMA. 2006;295:2275–85.

    Article  CAS  PubMed  Google Scholar 

  3. Simon TA, Thompson A, Gandhi KK, Hochberg MC, Suissa S. Incidence of malignancy in adult patients with rheumatoid arthritis: a meta-analysis. Arthritis Res Ther. 2015;17:212.

    Article  PubMed  PubMed Central  Google Scholar 

  4. Smitten AL, Simon TA, Hochberg MC, Suissa S. A meta-analysis of the incidence of malignancy in adult patients with rheumatoid arthritis. Arthritis Res Ther. 2008;10:R45.

    Article  PubMed  PubMed Central  Google Scholar 

  5. Lopez-Olivo MA, Tayar JH, Martinez-Lopez JA, Pollono EN, Cueto JP, Gonzales-Crespo MR, et al. Risk of malignancies in patients with rheumatoid arthritis treated with biologic therapy: a meta-analysis. JAMA. 2012;308:898–908.

    Article  CAS  PubMed  Google Scholar 

  6. Askling J, Fahrbach K, Nordstrom B, Ross S, Schmid CH, Symmons D. Cancer risk with tumor necrosis factor alpha (TNF) inhibitors: meta-analysis of randomized controlled trials of adalimumab, etanercept, and infliximab using patient level data. Pharmacoepidermiol Drug Saf. 2011;20:119–30.

    Article  CAS  Google Scholar 

  7. Wadstrom H, Frisell T, Askling J, Anti-Rheumatic Therapy in Sweden Study Group. Malignant neoplasms in patients with rheumatoid arthritis treated with tumor necrosis factor inhibitors, tocilizumab, abatacept, or rituximab in clinical practice: a nationwide cohort study from Sweden. JAMA Intern Med. 2017;177:1605–12.

    Article  PubMed  PubMed Central  Google Scholar 

  8. Mercer LK, Lunt M, Low AL, Dixon WG, Watson KD, Symmons DP, et al. Risk of solid cancer in patients exposed to anti-tumour necrosis factor therapy: results from the British Society for Rheumatology Biologics Register for Rheumatoid Arthritis. Ann Rheum Dis. 2015;74:1087–93.

    Article  CAS  PubMed  Google Scholar 

  9. Dreyer L, Mellemkjaer L, Andersen AR, Bennett P, Poulsen UE, Juulsgaard Ellingsen T, et al. Incidences of overall and site specific cancers in TNF alpha inhibitor treated patients with rheumatoid arthritis and other arthritides - a follow-up study from the DANBIO Registry. Ann Rheum Dis. 2013;72:79–82.

    Article  PubMed  Google Scholar 

  10. Ozen G, Pedro S, Schumacher R, Simon TA, Michaud K. Safety of abatacept compared with other biologic and conventional synthetic disease-modifying antirheumatic drugs in patients with rheumatoid arthritis: data from an observational study. Arthritis Res Ther. 2019;21:141.

    Article  PubMed  PubMed Central  Google Scholar 

  11. Montastruc F, Renoux C, Dell’Aniello S, Simon TA, Azoulay L, Hudson M, et al. Abatacept initiation in rheumatoid arthritis and the risk of cancer: a population-based comparative cohort study. Rheumatology (Oxford). 2019;58:683–91.

    Article  CAS  PubMed  Google Scholar 

  12. Huss V, Bower H, Wadström H, Frisell T, Askling J. Short- and longer-term cancer risks with biologic and targeted synthetic disease-modifying antirheumatic drugs as used against rheumatoid arthritis in clinical practice. Rheumatology (Oxford). 2022;61:1810–8.

    Article  CAS  PubMed  Google Scholar 

  13. Grøn KL, Glintborg B, Nørgaard M, Mehnert F, Østergaard M, Dreyer L, Krogh NS, Hetland ML. Overall infection risk in rheumatoid arthritis during treatment with abatacept, rituximab and tocilizumab; an observational cohort study. Rheumatology (Oxford). 2020;59:1949–56.

    Article  PubMed  Google Scholar 

  14. Mercer LK, Askling J, Raaschou P, Dixon WG, Dreyer L, Hetland ML, et al. Risk of invasive melanoma in patients with rheumatoid arthritis treated with biologics: results from a collaborative project of 11 European biologic registers. Ann Rheum Dis. 2017;76:386–91.

    Article  PubMed  Google Scholar 

  15. Weinblatt M, Schiff M, Goldman A, Kremer J, Luggen M, Li T, et al. Selective costimulation modulation using abatacept in patients with active rheumatoid arthritis while receiving etanercept: a randomised clinical trial. Ann Rheum Dis. 2007;66:228–34.

    Article  CAS  PubMed  Google Scholar 

  16. Toupin-April K, Barton J, Fraenkel L, Li LC, Brooks P, De Wit M, et al. Toward the development of a core set of outcome domains to assess shared decision-making interventions in rheumatology: results from an OMERACT Delphi Survey and Consensus Meeting. J Rheumatol. 2017;44:1544–50.

    Article  PubMed  Google Scholar 

  17. Orencia (250 mg powder for concentrate for solution for infusion) Summary of Product Characteristics. 2013. http://www.medicines.org.uk/emc/medicine/19714/SPC/. Accessed 2/2/2018.

  18. Emery P, Durez P, Dougados M, Legerton CW, Becker JC, Vratsanos G, et al. Impact of T-cell costimulation modulation in patients with undifferentiated inflammatory arthritis or very early rheumatoid arthritis: a clinical and imaging study of abatacept (the ADJUST trial). Ann Rheum Dis. 2010;69:510–6.

    Article  CAS  PubMed  Google Scholar 

  19. Emery P, Burmester GR, Bykerk VP, Combe BG, Furst DE, Barre E, et al. Evaluating drug-free remission with abatacept in early rheumatoid arthritis: results from the phase 3b, multicentre, randomised, active-controlled AVERT study of 24 months, with a 12-month, double-blind treatment period. Ann Rheum Dis. 2015;74:19–26.

    Article  CAS  PubMed  Google Scholar 

  20. Genovese MC, Becker JC, Schiff M, Luggen M, Sherrer Y, Kremer J, et al. Abatacept for rheumatoid arthritis refractory to tumor necrosis factor alpha inhibition. N Engl J Med. 2005;353:1114–23.

    Article  CAS  PubMed  Google Scholar 

  21. Kremer JM, Westhovens R, Leon M, Di Giorgio E, Alten R, Steinfeld S, et al. Treatment of rheumatoid arthritis by selective inhibition of T-cell activation with fusion protein CTLA4Ig. N Engl J Med. 2003;349:1907–15.

    Article  CAS  PubMed  Google Scholar 

  22. Kremer JM, Dougados M, Emery P, Durez P, Sibilia J, Shergy W, et al. Treatment of rheumatoid arthritis with the selective costimulation modulator abatacept: twelve-month results of a phase IIb, double-blind, randomized, placebo-controlled trial. Arthritis Rheum. 2005;52:2263–71.

    Article  CAS  PubMed  Google Scholar 

  23. Kremer JM, Genant HK, Moreland LW, Russell AS, Emery P, Abud-Mendoza C, et al. Effects of abatacept in patients with methotrexate-resistant active rheumatoid arthritis: a randomized trial. Ann Intern Med. 2006;144:865–76.

    Article  CAS  PubMed  Google Scholar 

  24. Schiff M, Keiserman M, Codding C, Songcharoen S, Berman A, Nayiager S, et al. Efficacy and safety of abatacept or infliximab vs placebo in ATTEST: a phase III, multi-centre, randomised, double-blind, placebo-controlled study in patients with rheumatoid arthritis and an inadequate response to methotrexate. Ann Rheum Dis. 2008;67:1096–103.

    Article  CAS  PubMed  Google Scholar 

  25. Schiff M, Pritchard C, Huffstutter JE, Rodriguez-Valverde V, Durez P, Zhou X, et al. The 6-month safety and efficacy of abatacept in patients with rheumatoid arthritis who underwent a washout after anti-tumour necrosis factor therapy or were directly switched to abatacept: the ARRIVE trial. Ann Rheum Dis. 2009;68:1708–14.

    Article  CAS  PubMed  Google Scholar 

  26. Westhovens R, Kremer JM, Moreland LW, Emery P, Russell AS, Li T, et al. Safety and efficacy of the selective costimulation modulator abatacept in patients with rheumatoid arthritis receiving background methotrexate: a 5-year extended phase IIB study. J Rheumatol. 2009;36:736–42.

    Article  CAS  PubMed  Google Scholar 

  27. Genovese MC, Schiff M, Luggen M, Le Bars M, Aranda R, Elegbe A, et al. Longterm safety and efficacy of abatacept through 5 years of treatment in patients with rheumatoid arthritis and an inadequate response to tumor necrosis factor inhibitor therapy. J Rheumatol. 2012;39:1546–54.

    Article  CAS  PubMed  Google Scholar 

  28. Kremer JM, Russell AS, Emery P, Abud-Mendoza C, Szechinski J, Westhovens R, et al. Long-term safety, efficacy and inhibition of radiographic progression with abatacept treatment in patients with rheumatoid arthritis and an inadequate response to methotrexate: 3-year results from the AIM trial. Ann Rheum Dis. 2011;70:1826–30.

    Article  CAS  PubMed  Google Scholar 

  29. Simon T, Soule B, Hochberg M, Fleming D, Torbeyns A, Banerjee S, et al. Safety of abatacept versus placebo in rheumatoid arthritis: integrated data analysis of nine clinical trials. ACR Open Rheumatol. 2019;1:251–7.

    Article  PubMed  PubMed Central  Google Scholar 

  30. Simon TA, Boers M, Hochberg M, Baker N, Skovron ML, Ray N, et al. Comparative risk of malignancies and infections in patients with rheumatoid arthritis initiating abatacept versus other biologics: a multi-database real-world study. Arthritis Res Ther. 2019;21:228.

    Article  PubMed  PubMed Central  Google Scholar 

  31. Simon TA, Suissa S, Skovron ML, Frisell T, Askling J, Michaud K, et al. FRI0118 Infection outcomes in patients with RA treated with abatacept and other DMARDs: results from a 10-year international post-approval study [abstract]. Ann Rheum Dis. 2019;78(Suppl 2):726–7.

  32. de Germay S, Bagheri H, Despas F, Rousseau V, Montastruc F. Abatacept in rheumatoid arthritis and the risk of cancer: a world observational post-marketing study. Rheumatology (Oxford). 2020;59:2360–7.

    Article  PubMed  Google Scholar 

  33. Lim CH, Chen HH, Chen YH, Chen DY, Huang WN, Tsai JJ, et al. The risk of tuberculosis disease in rheumatoid arthritis patients on biologics and targeted therapy: a 15-year real world experience in Taiwan. PLoS ONE. 2017;12:e0178035.

    Article  PubMed  PubMed Central  Google Scholar 

  34. Chiu YM, Lang HC, Lin HY, Yang MT, Fang CH, Yang YW, et al. Risk of tuberculosis, serious infection and lymphoma with disease-modifying biologic drugs in rheumatoid arthritis patients in Taiwan. Int J Rheum Dis. 2014;17(Suppl 3):9–19.

    Article  CAS  PubMed  Google Scholar 

  35. Simon TA, Suissa S, Skovron ML, Frisell T, Askling J, Michaud K, et al. OP0226 Solid tumour outcomes in patients with RA treated with abatacept and other DMARDs: results from a 10-year international post-approval study [abstract]. Ann Rheum Dis. 2019;78(Suppl 2):190.

  36. Gron KL, Arkema EV, Glintborg B, Mehnert F, Ostergaard M, Dreyer L, et al. Risk of serious infections in patients with rheumatoid arthritis treated in routine care with abatacept, rituximab and tocilizumab in Denmark and Sweden. Ann Rheum Dis. 2019;78:320–7.

    Article  CAS  PubMed  Google Scholar 

  37. European Centre for Disease Prevention and Control and World Health Organization ROfE. Tuberculosis surveillance and monitoring in Europe, 2017 data. 2019. https://www.ecdc.europa.eu/sites/default/files/documents/tuberculosis-surveillance-monitoring-Europe-2019-20_Mar_2019.pdf. Accessed 4/2/2021.

  38. Dixon WG, Hyrich KL, Watson KD, Lunt M, Galloway J, Ustianowski A, et al. Drug-specific risk of tuberculosis in patients with rheumatoid arthritis treated with anti-TNF therapy: results from the British Society for Rheumatology Biologics Register (BSRBR). Ann Rheum Dis. 2010;69:522–8.

    Article  CAS  PubMed  Google Scholar 

  39. Silva-Fernandez L, Lunt M, Kearsley-Fleet L, Watson KD, Dixon WG, Symmons DP, et al. The incidence of cancer in patients with rheumatoid arthritis and a prior malignancy who receive TNF inhibitors or rituximab: results from the British Society for Rheumatology Biologics Register-Rheumatoid Arthritis. Rheumatology (Oxford). 2016;55:2033–9.

    Article  CAS  PubMed  Google Scholar 

  40. Wu CY, Chen DY, Shen JL, Ho HJ, Chen CC, Kuo KN, et al. The risk of cancer in patients with rheumatoid arthritis taking tumor necrosis factor antagonists: a nationwide cohort study. Arthritis Res Ther. 2014;16:449.

    Article  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

The authors are grateful to all investigators who contributed to the registries. DANBIO: Mette Nørgaard, MD, PhD (consultant); Nickolaj Risbo Kristensen, MSc; Frank Mehnert, MSc; Professor Henrik Toft Sørensen, MD, PhD, DMSc; Bente Glintborg, MD, PhD (consultant); and Kathrine Lederballe Grøn, MD. Myriam Riek (SCQM Foundation, Zürich, Switzerland) performed the statistical analysis of the SCQM data. ROB-FIN: Nina Trokovic, PhD (data manager), and Arja Kaarto (study nurse). ORA: Xavier Mariette, MD, PhD (co-coordinated the study). Professional medical writing and editorial assistance was provided by Catriona McKay, PhD, and Candice Dcosta, MSc, at Caudex and was funded by Bristol Myers Squibb.

Funding

This study was sponsored by Bristol Myers Squibb.

Author information

Authors and Affiliations

Authors

Contributions

AD, MLH, AF, J-EG, FI, RC, TDK, DN, MVH, CS-P, FS-A, KP, TCB, and TAS made substantial contributions to the conception OR design of the work; OR the acquisition, analysis, OR interpretation of data; OR the creation of new software used in the work; OR have drafted the work or substantively revised it. AD, MLH, AF, J-EG, FI, RC, TDK, DN, MVH, CS-P, FS-A, KP, TCB, and TAS approved the submitted version (and any substantially modified version that involves the author's contribution to the study). AD, MLH, AF, J-EG, FI, RC, TDK, DN, MVH, CS-P, FS-A, KP, TCB, and TAS agreed both to be personally accountable for the author's own contributions and to ensure that questions related to the accuracy or integrity of any part of the work, even ones in which the author was not personally involved, are appropriately investigated, resolved, and the resolution documented in the literature.

Corresponding author

Correspondence to T. Christopher Bond.

Ethics declarations

Ethics approval and consent to participate

This study was conducted in accordance with the International Society for Pharmacoepidemiology Guidelines for Good Pharmacoepidemiology Practices, applicable regulatory requirements in each country, and the principles of the Declaration of Helsinki.

Consent for publication

Not applicable.

Competing interests

AD: employee and shareholder: Bristol Myers Squibb (at the time of analysis); employee: Pfizer (current); MLH: grant/research support: AbbVie, Biogen, Bristol Myers Squibb, Merck, Novartis, Pfizer, Roche, Sandoz; consulting fees: Biogen, Celltrion, Eli Lilly, Merck, Orion Pharma, Pfizer, Samsung Bioepis; AF: grant/research support: AbbVie, Bristol Myers Squibb, Eli Lilly, Galapagos, Pfizer; speaker fees/honoraria: AB2Bio, AbbVie, Bristol Myers Squibb, Eli Lilly, Pfizer, Sandoz, Sanofi; J-EG: grant/research support, consulting fees: Bristol Myers Squibb, Eli Lilly, Pfizer; consulting fees: AbbVie, Bristol Myers Squibb, CSL Behring, Galapagos, Gilead, Merck Sharp & Dohme, Pfizer, Sanofi-Regeneron; FI: speaker fees/honoraria: AbbVie, Bristol Myers Squibb, Eli Lilly, Galapagos, Janssen, Merck Sharp & Dohme, Pfizer, Roche, Sanofi, UCB; RC: speaker fees/honoraria: AbbVie, Bristol Myers Squibb, Eli Lilly, Galapagos, Gilead, Merck Sharp & Dohme, Pfizer, Roche, Sanofi, UCB; TDK: employee: Bristol Myers Squibb (at the time of analysis), BeiGene USA, Inc. (current); DN: consulting fees, speaker fees/honoraria: AbbVie, Bristol Myers Squibb, Eli Lilly, Merck Sharp & Dohme, Novartis, Pfizer, Roche, UCB; MVH, CS-P, FS-A: nothing to disclose; KP: speaker fees/honoraria: AbbVie, Amgen, Biogen, Bristol Myers Squibb, Egis, Merck Sharp & Dohme, Novartis, Pfizer, Roche, UCB; TCB: employee and shareholder: Bristol Myers Squibb; TAS: employee and shareholder: Bristol Myers Squibb (at the time of analysis); employee: Physicians Research Center (current); consulting fees: Bristol Myers Squibb.

Additional information

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Affiliations for Alyssa Dominique, Tzuyung Douglas Kou, Carlos Sánchez-Piedra, and Teresa A. Simon were those at the time of analysis.

Supplementary Information

Additional file 1.

 Methods used to calculate patient-time of exposure and incidence rates for each individual registry included in the post-marketing epidemiology abatacept study.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Dominique, A., Hetland, M.L., Finckh, A. et al. Safety outcomes in patients with rheumatoid arthritis treated with abatacept: results from a multinational surveillance study across seven European registries. Arthritis Res Ther 25, 101 (2023). https://0-doi-org.brum.beds.ac.uk/10.1186/s13075-023-03067-x

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://0-doi-org.brum.beds.ac.uk/10.1186/s13075-023-03067-x

Keywords