Biomarkers to predict risk of venous thromboembolism in patients with rheumatoid arthritis receiving tofacitinib or tumour necrosis factor inhibitors

Jeffrey I Weitz, Zoltán Szekanecz, Christina Charles-Schoeman, Ivana Vranic, Burak Sahin, Sara A Paciga, Zhenyu Wang, Craig Hyde, David A Martin, Jeffrey I Weitz, Zoltán Szekanecz, Christina Charles-Schoeman, Ivana Vranic, Burak Sahin, Sara A Paciga, Zhenyu Wang, Craig Hyde, David A Martin

Abstract

Objective: In the ORAL (Oral Rheumatoid Arthritis triaL) Surveillance study of patients with rheumatoid arthritis aged ≥50 years with ≥1 additional cardiovascular risk factor, incidence of pulmonary embolism was higher with tofacitinib 10 mg two times per day than with tumour necrosis factor inhibitors (TNFi). This exploratory post hoc analysis examined whether biomarkers explained the associations of tofacitinib versus TNFi with venous thromboembolism (VTE).

Methods: ORAL Surveillance was a prospective, open-label, event-driven, non-inferiority, postauthorisation safety study. Patients were randomised 1:1:1 to receive tofacitinib 5 mg or 10 mg two times per day or a TNFi. For this analysis, 294 soluble, proteomic, genetic and antibody biomarkers (of which 79 had a known role in inflammation, coagulation, vascular biology or Janus kinase signalling) were quantified in serum collected at baseline, month 12 and study end.

Results: Overall, 4362 patients were randomised and treated. The exploratory biomarker data set included 285 patients (57 VTE cases; 228 matched controls). D-dimer was quantified in 3732 patients (54 VTE cases; 3678 controls). No biomarker demonstrated a clear mechanistic association with the increased risk of VTE for tofacitinib versus TNFi. Month 12 D-dimer levels were positively associated with risk of a subsequent VTE within the tofacitinib 5 mg and 10 mg two times per day arms.

Conclusions: Overall, this post hoc analysis did not identify biomarkers that explained the increased VTE risk for tofacitinib versus TNFi. Individual VTE risk should be considered when making decisions about initiation or maintenance of tofacitinib treatment.

Trial registration number: NCT02092467; ClinicalTrials.gov.

Keywords: antirheumatic agents; arthritis, rheumatoid; cardiovascular diseases.

Conflict of interest statement

Competing interests: JIW received honoraria from Anthos, Bayer AG, Boehringer Ingelheim, Bristol-Myers Squibb, Daiichi Sankyo, Ionis, Janssen, Novartis, Pfizer, PhaseBio, Portola and Servier Pharmaceuticals, and institutional grants from Bayer AG and Boehringer Ingelheim. ZS has received consulting fees and honoraria from AbbVie, Amgen, BMS, Gedeon Richter, Lilly, MSD, Pfizer, Roche, Sanofi and UCB. CC-S has received consulting fees, research grants and/or honoraria from AbbVie, Amgen, BMS, Gilead, Octapharma, Pfizer and Sanofi/Regeneron. IV, BS, SAP, ZW, CH and DAM are employees and/or shareholders of Pfizer Inc.

© Author(s) (or their employer(s)) 2022. Re-use permitted under CC BY-NC. No commercial re-use. See rights and permissions. Published by BMJ.

Figures

Figure 1
Figure 1
(A) CRP levels at baseline and (B) changes from baseline in CRP levels at month 12 by treatment arm in controls and VTE cases (full clinical data set). Normal assay range:

Figure 2

D-dimer levels at baseline, month…

Figure 2

D-dimer levels at baseline, month 12 and end of study by treatment arm…

Figure 2
D-dimer levels at baseline, month 12 and end of study by treatment arm in controls and VTE cases (D-dimer data set). Normal assay range:

Figure 3

(A) TPO levels at baseline…

Figure 3

(A) TPO levels at baseline and changes from baseline in TPO levels at…

Figure 3
(A) TPO levels at baseline and changes from baseline in TPO levels at month 12; (B) exploratory biomarker data set; (C) changes from baseline in platelet counts at month 12 (full clinical data set) in controls and VTE cases. For patients assigned to receive tofacitinib at a dose of 10 mg two times per day who had their dose reduced to 5 mg two times per day, the data collected after patients had been switched to 5 mg two times per day were counted in the arm receiving 10 mg two times per day. BID, twice daily; CI, confidence interval; ln, natural logarithm; LS, least squares; TNFi, tumour necrosis factor inhibitor; TPO, thrombopoietin; VTE, venous thromboembolism.
Similar articles
References
    1. Khan F, Tritschler T, Kahn SR, et al. . Venous thromboembolism. Lancet 2021;398:64–77. 10.1016/S0140-6736(20)32658-1 - DOI - PubMed
    1. Heit JA, Spencer FA, White RH. The epidemiology of venous thromboembolism. J Thromb Thrombolysis 2016;41:3–14. 10.1007/s11239-015-1311-6 - DOI - PMC - PubMed
    1. Huang W, Goldberg RJ, Anderson FA, et al. . Secular trends in occurrence of acute venous thromboembolism: the Worcester VTE study (1985-2009). Am J Med 2014;127:829–39. 10.1016/j.amjmed.2014.03.041 - DOI - PMC - PubMed
    1. Kim SC, Schneeweiss S, Liu J. Risk of venous thromboembolism in patients with rheumatoid arthritis. Arthritis Care Res (Hoboken) 2013;65:1600–7. - PMC - PubMed
    1. Molander V, Bower H, Frisell T, et al. . Risk of venous thromboembolism in rheumatoid arthritis, and its association with disease activity: a nationwide cohort study from Sweden. Ann Rheum Dis 2021;80:169–75. 10.1136/annrheumdis-2020-218419 - DOI - PubMed
Show all 29 references
Publication types
MeSH terms
Associated data
Full text links [x]
[x]
Cite
Copy Download .nbib
Format: AMA APA MLA NLM
Figure 2
Figure 2
D-dimer levels at baseline, month 12 and end of study by treatment arm in controls and VTE cases (D-dimer data set). Normal assay range:

Figure 3

(A) TPO levels at baseline…

Figure 3

(A) TPO levels at baseline and changes from baseline in TPO levels at…

Figure 3
(A) TPO levels at baseline and changes from baseline in TPO levels at month 12; (B) exploratory biomarker data set; (C) changes from baseline in platelet counts at month 12 (full clinical data set) in controls and VTE cases. For patients assigned to receive tofacitinib at a dose of 10 mg two times per day who had their dose reduced to 5 mg two times per day, the data collected after patients had been switched to 5 mg two times per day were counted in the arm receiving 10 mg two times per day. BID, twice daily; CI, confidence interval; ln, natural logarithm; LS, least squares; TNFi, tumour necrosis factor inhibitor; TPO, thrombopoietin; VTE, venous thromboembolism.
Figure 3
Figure 3
(A) TPO levels at baseline and changes from baseline in TPO levels at month 12; (B) exploratory biomarker data set; (C) changes from baseline in platelet counts at month 12 (full clinical data set) in controls and VTE cases. For patients assigned to receive tofacitinib at a dose of 10 mg two times per day who had their dose reduced to 5 mg two times per day, the data collected after patients had been switched to 5 mg two times per day were counted in the arm receiving 10 mg two times per day. BID, twice daily; CI, confidence interval; ln, natural logarithm; LS, least squares; TNFi, tumour necrosis factor inhibitor; TPO, thrombopoietin; VTE, venous thromboembolism.

References

    1. Khan F, Tritschler T, Kahn SR, et al. . Venous thromboembolism. Lancet 2021;398:64–77. 10.1016/S0140-6736(20)32658-1
    1. Heit JA, Spencer FA, White RH. The epidemiology of venous thromboembolism. J Thromb Thrombolysis 2016;41:3–14. 10.1007/s11239-015-1311-6
    1. Huang W, Goldberg RJ, Anderson FA, et al. . Secular trends in occurrence of acute venous thromboembolism: the Worcester VTE study (1985-2009). Am J Med 2014;127:829–39. 10.1016/j.amjmed.2014.03.041
    1. Kim SC, Schneeweiss S, Liu J. Risk of venous thromboembolism in patients with rheumatoid arthritis. Arthritis Care Res (Hoboken) 2013;65:1600–7.
    1. Molander V, Bower H, Frisell T, et al. . Risk of venous thromboembolism in rheumatoid arthritis, and its association with disease activity: a nationwide cohort study from Sweden. Ann Rheum Dis 2021;80:169–75. 10.1136/annrheumdis-2020-218419
    1. Ramagopalan SV, Wotton CJ, Handel AE, et al. . Risk of venous thromboembolism in people admitted to hospital with selected immune-mediated diseases: record-linkage study. BMC Med 2011;9:1. 10.1186/1741-7015-9-1
    1. Yusuf HR, Hooper WC, Grosse SD, et al. . Risk of venous thromboembolism occurrence among adults with selected autoimmune diseases: a study among a U.S. cohort of commercial insurance enrollees. Thromb Res 2015;135:50–7. 10.1016/j.thromres.2014.10.012
    1. van den Oever IAM, Sattar N, Nurmohamed MT. Thromboembolic and cardiovascular risk in rheumatoid arthritis: role of the haemostatic system. Ann Rheum Dis 2014;73:954–7. 10.1136/annrheumdis-2013-204767
    1. Xue L, Tao L, Li X, et al. . Plasma fibrinogen, D-dimer, and fibrin degradation product as biomarkers of rheumatoid arthritis. Sci Rep 2021;11. 10.1038/s41598-021-96349-w
    1. Ytterberg SR, Bhatt DL, Mikuls TR, et al. . Cardiovascular and cancer risk with tofacitinib in rheumatoid arthritis. N Engl J Med 2022;386:316–26. 10.1056/NEJMoa2109927
    1. Rosendaal FR, Koster T, Vandenbroucke JP, et al. . High risk of thrombosis in patients homozygous for factor V Leiden (activated protein C resistance) [see comments]. Blood 1995;85:1504–8. 10.1182/blood.V85.6.1504.bloodjournal8561504
    1. Poort SR, Rosendaal FR, Reitsma PH, et al. . A common genetic variation in the 3'-untranslated region of the prothrombin gene is associated with elevated plasma prothrombin levels and an increase in venous thrombosis. Blood 1996;88:3698–703. 10.1182/blood.V88.10.3698.bloodjournal88103698
    1. Edelmann B, Gupta N, Schnoeder TM, et al. . JAK2-V617F promotes venous thrombosis through β1/β2 integrin activation. J Clin Invest 2018;128:4359–71. 10.1172/JCI90312
    1. Galli M, Borrelli G, Jacobsen EM, et al. . Clinical significance of different antiphospholipid antibodies in the WAPS (Warfarin in the Antiphospholipid Syndrome) study. Blood 2007;110:1178–83. 10.1182/blood-2007-01-066043
    1. Devreese KMJ, Ortel TL, Pengo V, et al. . Laboratory criteria for antiphospholipid syndrome: communication from the SSC of the ISTH. J Thromb Haemost 2018;16:809–13. 10.1111/jth.13976
    1. Cohen AT, Spiro TE, Spyropoulos AC, et al. . D-dimer as a predictor of venous thromboembolism in acutely ill, hospitalized patients: a subanalysis of the randomized controlled MAGELLAN trial. J Thromb Haemost 2014;12:479–87. 10.1111/jth.12515
    1. Spyropoulos AC, Ageno W, Albers GW, et al. . Rivaroxaban for thromboprophylaxis after hospitalization for medical illness. N Engl J Med 2018;379:1118–27. 10.1056/NEJMoa1805090
    1. Cohen AT, Harrington RA, Goldhaber SZ, et al. . Extended thromboprophylaxis with betrixaban in acutely ill medical patients. N Engl J Med 2016;375:534–44. 10.1056/NEJMoa1601747
    1. Weitz JI, Fredenburgh JC, Eikelboom JW. A test in context: D-dimer. J Am Coll Cardiol 2017;70:2411–20. 10.1016/j.jacc.2017.09.024
    1. Poudel A, Poudel Y, Adhikari A, et al. . D-dimer as a biomarker for assessment of COVID-19 prognosis: D-dimer levels on admission and its role in predicting disease outcome in hospitalized patients with COVID-19. PLoS One 2021;16:e0256744. 10.1371/journal.pone.0256744
    1. Yao Y, Cao J, Wang Q, et al. . D-dimer as a biomarker for disease severity and mortality in COVID-19 patients: a case control study. J Intensive Care 2020;8:49. 10.1186/s40560-020-00466-z
    1. Wei F, Chang Y, Wei W. The role of BAFF in the progression of rheumatoid arthritis. Cytokine 2015;76:537–44. 10.1016/j.cyto.2015.07.014
    1. Weinhold KJ, Bukowski JF, Brennan TV, et al. . Reversibility of peripheral blood leukocyte phenotypic and functional changes after exposure to and withdrawal from tofacitinib, a Janus kinase inhibitor, in healthy volunteers. Clin Immunol 2018;191:10–20. 10.1016/j.clim.2018.03.002
    1. Lee J, Lee J, Kwok S-K, et al. . JAK-1 inhibition suppresses interferon-induced BAFF production in human salivary gland: potential therapeutic strategy for primary Sjögren's syndrome. Arthritis Rheumatol 2018;70:2057–66. 10.1002/art.40589
    1. Yoshimoto K, Tanaka M, Kojima M. JAK-STAT pathways are involved in the BAFF signaling of peripheral monocytes of patients with primary Sjögren’s syndrome.. J Immunol 2012;188 (Suppl):174.9.
    1. Hodge JA, Kawabata TT, Krishnaswami S, et al. . The mechanism of action of tofacitinib - an oral Janus kinase inhibitor for the treatment of rheumatoid arthritis. Clin Exp Rheumatol 2016;34:318–28.
    1. Meyer SC, Keller MD, Woods BA, et al. . Genetic studies reveal an unexpected negative regulatory role for Jak2 in thrombopoiesis. Blood 2014;124:2280–4. 10.1182/blood-2014-03-560441
    1. Mori S, Ogata F, Tsunoda R. Risk of venous thromboembolism associated with Janus kinase inhibitors for rheumatoid arthritis: case presentation and literature review. Clin Rheumatol 2021;40:4457–71. 10.1007/s10067-021-05911-4
    1. Ceresa IF, Noris P, Ambaglio C, et al. . Thrombopoietin is not uniquely responsible for thrombocytosis in inflammatory disorders. Platelets 2007;18:579–82. 10.1080/09537100701593601

Source: PubMed

3
Se inscrever