Cardiovascular Safety of Tocilizumab Versus Tumor Necrosis Factor Inhibitors in Patients With Rheumatoid Arthritis: A Multi-Database Cohort Study

Seoyoung C Kim, Daniel H Solomon, James R Rogers, Sara Gale, Micki Klearman, Khaled Sarsour, Sebastian Schneeweiss, Seoyoung C Kim, Daniel H Solomon, James R Rogers, Sara Gale, Micki Klearman, Khaled Sarsour, Sebastian Schneeweiss

Abstract

Objective: While tocilizumab (TCZ) is known to increase low-density lipoprotein (LDL) cholesterol levels, it is unclear whether TCZ increases cardiovascular risk in patients with rheumatoid arthritis (RA). This study was undertaken to compare the cardiovascular risk associated with receiving TCZ versus tumor necrosis factor inhibitors (TNFi).

Methods: To examine comparative cardiovascular safety, we conducted a cohort study of RA patients who newly started TCZ or TNFi using claims data from Medicare, IMS PharMetrics, and MarketScan. All patients were required to have previously used a different TNFi, abatacept, or tofacitinib. The primary outcome measure was a composite cardiovascular end point of hospitalization for myocardial infarction or stroke. TCZ initiators were propensity score matched to TNFi initiators with a variable ratio of 1:3 within each database, controlling for >65 baseline characteristics. A fixed-effects model combined database-specific hazard ratios (HRs).

Results: We included 9,218 TCZ initiators propensity score matched to 18,810 TNFi initiators across all 3 databases. The mean age was 72 years in Medicare, 51 in PharMetrics, and 53 in MarketScan. Cardiovascular disease was present at baseline in 14.3% of TCZ initiators and 13.5% of TNFi initiators. During the study period (mean ± SD 0.9 ± 0.7 years; maximum 4.5 years), 125 composite cardiovascular events occurred, resulting in an incidence rate of 0.52 per 100 person-years for TCZ initiators and 0.59 per 100 person-years for TNFi initiators. The risk of cardiovascular events associated with TCZ use versus TNFi use was similar across all 3 databases, with a combined HR of 0.84 (95% confidence interval 0.56-1.26).

Conclusion: This multi-database population-based cohort study showed no evidence of an increased cardiovascular risk among RA patients who switched from a different biologic drug or tofacitinib to TCZ versus to a TNFi.

© 2017, The Authors. Arthritis & Rheumatology published by Wiley Periodicals, Inc. on behalf of American College of Rheumatology.

Figures

Figure 1
Figure 1
Study design overview. Patients with rheumatoid arthritis (RA) who had not received tocilizumab (TCZ) or tumor necrosis factor inhibitors (TNFi) in the previous 365 days were enrolled in the TCZ cohort or TNFi cohort, respectively. The index date was defined as the date of first TCZ dispensing or the date of switching to a new TNFi after being treated with at least 1 other biologic drug (i.e., abatacept, TNFi, or tofacitinib). Dx = diagnosis; Rx = prescription.
Figure 2
Figure 2
Selection of the study cohort. After the inclusion and exclusion criteria were applied, the study cohort included a total of 40,119 patients with rheumatoid arthritis (RA) who started treatment with tocilizumab (TCZ) or a tumor necrosis factor inhibitor (TNFi), including 9,146 from the Medicare database, 12,826 from the PharMetrics database, and 18,147 from the MarketScan database. After propensity score (PS) matching with a variable ratio of up to 1:3, the final cohort consisted of 28,028 patients, including 9,218 TCZ initiators and 18,810 TNFi initiators. Dx = diagnosis.
Figure 3
Figure 3
Secondary end points in a 1:3 variable ratio propensity score–matched as‐treated analysis of rheumatoid arthritis patients starting treatment with tocilizumab (TCZ) or a tumor necrosis factor inhibitor (TNFi). Hazard ratios (HRs) were combined using an inverse variance‐weighted, fixed‐effects model. A “secondary definition of cardiovascular (CV) event” refers to a discharge diagnosis of myocardial infarction in the principal position for any length of hospitalization or a hospital discharge diagnosis of ischemic or hemorrhagic stroke in the principal position. “Any CV event” includes myocardial infarction, stroke, coronary revascularization, or acute coronary syndrome. 95% CI = 95% confidence interval.
Figure 4
Figure 4
Subgroup analysis in a 1:3 variable ratio propensity score–matched as‐treated analysis of rheumatoid arthritis patients starting treatment with tocilizumab (TCZ) or a tumor necrosis factor inhibitor (TNFi). Hazard ratios (HRs) were combined by an inverse variance‐weighted, fixed‐effects model. For the age

References

    1. Solomon DH, Karlson EW, Rimm EB, Cannuscio CC, Mandl LA, Manson JE, et al. Cardiovascular morbidity and mortality in women diagnosed with rheumatoid arthritis. Circulation 2003;107:1303–7.
    1. Avina‐Zubieta JA, Choi HK, Sadatsafavi M, Etminan M, Esdaile JM, Lacaille D. Risk of cardiovascular mortality in patients with rheumatoid arthritis: a meta‐analysis of observational studies. Arthritis Rheum 2008;59:1690–7.
    1. Meissner Y, Zink A, Kekow J, Rockwitz K, Liebhaber A, Zinke S, et al. Impact of disease activity and treatment of comorbidities on the risk of myocardial infarction in rheumatoid arthritis. Arthritis Res Ther 2016;18:183.
    1. Solomon DH, Kremer J, Curtis JR, Hochberg MC, Reed G, Tsao P, et al. Explaining the cardiovascular risk associated with rheumatoid arthritis: traditional risk factors versus markers of rheumatoid arthritis severity. Ann Rheum Dis 2010;69:1920–5.
    1. Singh JA, Saag KG, Bridges SL Jr, Akl EA, Bannuru RR, Sullivan MC, et al. 2015 American College of Rheumatology guideline for the treatment of rheumatoid arthritis. Arthritis Rheumatol 2016;68:1–26.
    1. Roubille C, Richer V, Starnino T, McCourt C, McFarlane A, Fleming P, et al. The effects of tumour necrosis factor inhibitors, methotrexate, non‐steroidal anti‐inflammatory drugs and corticosteroids on cardiovascular events in rheumatoid arthritis, psoriasis and psoriatic arthritis: a systematic review and meta‐analysis. Ann Rheum Dis 2015;74:480–9.
    1. Barnabe C, Martin BJ, Ghali WA. Systematic review and meta‐analysis: anti–tumor necrosis factor α therapy and cardiovascular events in rheumatoid arthritis. Arthritis Care Res (Hoboken) 2011;63:522–9.
    1. Greenberg JD, Kremer JM, Curtis JR, Hochberg MC, Reed G, Tsao P, et al. Tumour necrosis factor antagonist use and associated risk reduction of cardiovascular events among patients with rheumatoid arthritis. Ann Rheum Dis 2011;70:576–82.
    1. Solomon DH, Curtis JR, Saag KG, Lii J, Chen L, Harrold LR, et al. Cardiovascular risk in rheumatoid arthritis: comparing TNF‐α blockade with nonbiologic DMARDs. Am J Med 2013;126:730.e9–17.
    1. Desai RJ, Rao JK, Hansen RA, Fang G, Maciejewski M, Farley J. Tumor necrosis factor‐α inhibitor treatment and the risk of incident cardiovascular events in patients with early rheumatoid arthritis: a nested case‐control study. J Rheumatol 2014;41:2129–36.
    1. Dixon WG, Watson KD, Lunt M, Hyrich KL, British Society for Rheumatology Biologics Register Control Centre Consortium , Silman AJ, et al, on behalf of the British Society for Rheumatology Biologics Register. Reduction in the incidence of myocardial infarction in patients with rheumatoid arthritis who respond to anti–tumor necrosis factor α therapy: results from the British Society for Rheumatology Biologics Register. Arthritis Rheum 2007;56:2905–12.
    1. Jones G, Sebba A, Gu J, Lowenstein MB, Calvo A, Gomez‐Reino JJ, et al. Comparison of tocilizumab monotherapy versus methotrexate monotherapy in patients with moderate to severe rheumatoid arthritis: the AMBITION study. Ann Rheum Dis 2010;69:88–96.
    1. Nishimoto N, Yoshizaki K, Miyasaka N, Yamamoto K, Kawai S, Takeuchi T, et al. Treatment of rheumatoid arthritis with humanized anti–interleukin‐6 receptor antibody: a multicenter, double‐blind, placebo‐controlled trial. Arthritis Rheum 2004;50:1761–9.
    1. Smolen JS, Beaulieu A, Rubbert‐Roth A, Ramos‐Remus C, Rovensky J, Alecock E, et al. Effect of interleukin‐6 receptor inhibition with tocilizumab in patients with rheumatoid arthritis (OPTION study): a double‐blind, placebo‐controlled, randomised trial. Lancet 2008;371:987–97.
    1. Gabay C, McInnes IB, Kavanaugh A, Tuckwell K, Klearman M, Pulley J, et al. Comparison of lipid and lipid‐associated cardiovascular risk marker changes after treatment with tocilizumab or adalimumab in patients with rheumatoid arthritis. Ann Rheum Dis 2016;75:1806–12.
    1. Gabay C, Emery P, van Vollenhoven R, Dikranian A, Alten R, Pavelka K, et al. Tocilizumab monotherapy versus adalimumab monotherapy for treatment of rheumatoid arthritis (ADACTA): a randomised, double‐blind, controlled phase 4 trial. Lancet 2013;381:1541–50.
    1. Rao VU, Pavlov A, Klearman M, Musselman D, Giles JT, Bathon JM, et al. An evaluation of risk factors for major adverse cardiovascular events during tocilizumab therapy. Arthritis Rheumatol 2015;67:372–80.
    1. Hennessy S, Freeman C, Cunningham F. US government claims databases In: Strom B, Kimmel S, Hennessy S, editors. Pharmacoepidemiology. 5th ed Philadelphia: Wiley‐Blackwell; 2012. p. 209–23.
    1. Kappelman MD, Rifas‐Shiman SL, Kleinman K, Ollendorf D, Bousvaros A, Grand RJ, et al. The prevalence and geographic distribution of Crohn's disease and ulcerative colitis in the United States. Clin Gastroenterol Hepatol 2007;5:1424–9.
    1. Quek RG, Fox KM, Wang L, Li L, Gandra SR, Wong ND. A US claims‐based analysis of real‐world lipid‐lowering treatment patterns in patients with high cardiovascular disease risk or a previous coronary event. Am J Cardiol 2016;117:495–500.
    1. Kim SY, Servi A, Polinski JM, Mogun H, Weinblatt ME, Katz JN, et al. Validation of rheumatoid arthritis diagnoses in health care utilization data. Arthritis Res Ther 2011;13:R32.
    1. Kiyota Y, Schneeweiss S, Glynn RJ, Cannuscio CC, Avorn J, Solomon DH. Accuracy of Medicare claims‐based diagnosis of acute myocardial infarction: estimating positive predictive value on the basis of review of hospital records. Am Heart J 2004;148:99–104.
    1. Andrade SE, Harrold LR, Tjia J, Cutrona SL, Saczynski JS, Dodd KS, et al. A systematic review of validated methods for identifying cerebrovascular accident or transient ischemic attack using administrative data. Pharmacoepidemiol Drug Saf 2012;21 Suppl 1:100–28.
    1. Kumamaru H, Judd SE, Curtis JR, Ramachandran R, Hardy NC, Rhodes JD, et al. Validity of claims‐based stroke algorithms in contemporary Medicare data: reasons for geographic and racial differences in stroke (REGARDS) study linked with Medicare claims. Circ Cardiovasc Qual Outcomes 2014;7:611–9.
    1. Ting G, Schneeweiss S, Scranton R, Katz JN, Weinblatt ME, Young M, et al. Development of a health care utilisation data‐based index for rheumatoid arthritis severity: a preliminary study. Arthritis Res Ther 2008;10:R95.
    1. Gagne JJ, Glynn RJ, Avorn J, Levin R, Schneeweiss S. A combined comorbidity score predicted mortality in elderly patients better than existing scores. J Clin Epidemiol 2011;64:749–59.
    1. Schneeweiss S, Rassen JA, Glynn RJ, Myers J, Daniel GW, Singer J, et al. Supplementing claims data with outpatient laboratory test results to improve confounding adjustment in effectiveness studies of lipid‐lowering treatments. BMC Med Res Methodol 2012;12:180.
    1. Rubin DB. Estimating causal effects from large data sets using propensity scores. Ann Intern Med 1997;127:757–63.
    1. Rassen JA, Shelat AA, Myers J, Glynn RJ, Rothman KJ, Schneeweiss S. One‐to‐many propensity score matching in cohort studies. Pharmacoepidemiol Drug Saf 2012;21 Suppl 2:69–80.
    1. Austin PC. Optimal caliper widths for propensity‐score matching when estimating differences in means and differences in proportions in observational studies. Pharm Stat 2011;10:150–61.
    1. Franklin JM, Rassen JA, Ackermann D, Bartels DB, Schneeweiss S. Metrics for covariate balance in cohort studies of causal effects. Stat Med 2014;33:1685–99.
    1. Cummings P, McKnight B, Weiss NS. Matched‐pair cohort methods in traffic crash research. Accid Anal Prev 2003;35:131–41.
    1. Walker AM, Jick H, Hunter JR, Danford A, Watkins RN, Alhadeff L, et al. Vasectomy and non‐fatal myocardial infarction. Lancet 1981;1:13–5.
    1. Munder T, Brutsch O, Leonhart R, Gerger H, Barth J. Researcher allegiance in psychotherapy outcome research: an overview of reviews. Clin Psychol Rev 2013;33:501–11.
    1. Liao KP, Liu J, Lu B, Solomon DH, Kim SC. Association between lipid levels and major adverse cardiovascular events in rheumatoid arthritis compared to non–rheumatoid arthritis patients. Arthritis Rheumatol 2015;67:2004–10.
    1. Myasoedova E, Crowson CS, Kremers HM, Roger VL, Fitz‐Gibbon PD, Therneau TM, et al. Lipid paradox in rheumatoid arthritis: the impact of serum lipid measures and systemic inflammation on the risk of cardiovascular disease. Ann Rheum Dis 2011;70:482–7.
    1. Robertson J, Peters MJ, McInnes IB, Sattar N. Changes in lipid levels with inflammation and therapy in RA: a maturing paradigm. Nat Rev Rheumatol 2013;9:513–23.
    1. Navarro‐Millán I, Yang S, DuVall SL, Chen L, Baddley J, Cannon GW, et al. Association of hyperlipidaemia, inflammation and serological status and coronary heart disease among patients with rheumatoid arthritis: data from the National Veterans Health Administration. Ann Rheum Dis 2016;75:341–7.
    1. Seriolo B, Paolino S, Sulli A, Fasciolo D, Cutolo M. Effects of anti‐TNF‐α treatment on lipid profile in patients with active rheumatoid arthritis. Ann N Y Acad Sci 2006;1069:414–9.
    1. Zhang J, Xie F, Yun H, Chen L, Muntner P, Levitan EB, et al. Comparative effects of biologics on cardiovascular risk among older patients with rheumatoid arthritis. Ann Rheum Dis 2016;75:1813–8.
    1. Ray WA. Evaluating medication effects outside of clinical trials: new‐user designs. Am J Epidemiol 2003;158:915–20.
    1. Yoshida K, Solomon DH, Kim SC. Active‐comparator design and new‐user design in observational studies. Nat Rev Rheumatol 2015;11:437–41.

Source: PubMed

3
Suscribir