Rituximab in patients with acute ST-elevation myocardial infarction: an experimental medicine safety study

Tian X Zhao, Muhammad Aetesam-Ur-Rahman, Andrew P Sage, Saji Victor, Rincy Kurian, Sarah Fielding, Hafid Ait-Oufella, Yi-Da Chiu, Christoph J Binder, Mikel Mckie, Stephen P Hoole, Ziad Mallat, Tian X Zhao, Muhammad Aetesam-Ur-Rahman, Andrew P Sage, Saji Victor, Rincy Kurian, Sarah Fielding, Hafid Ait-Oufella, Yi-Da Chiu, Christoph J Binder, Mikel Mckie, Stephen P Hoole, Ziad Mallat

Abstract

Aims: In pre-clinical models of acute myocardial infarction (MI), mature B cells mobilize inflammatory monocytes into the heart, leading to increased infarct size and deterioration of cardiac function, whilst anti-CD20 antibody-mediated depletion of B cells limits myocardial injury and improves cardiac function. Rituximab is a monoclonal anti-CD20 antibody targeted against human B cells. However, its use in cardiovascular disease is untested and is currently contraindicated. Therefore, we assessed the safety, feasibility, and pharmacodynamic effect of rituximab given to patients with acute ST-elevation MI (STEMI).

Methods and results: Rituximab in patients with acute ST-elevation myocardial infarction (RITA-MI) was a prospective, open-label, dose-escalation, single-arm, phase 1/2a clinical trial, which tested rituximab administered as a single intravenous dose in patients with STEMI within 48 h of symptom onset. Four escalating doses (200, 500, 700, and 1000 mg) were used. The primary endpoint was safety, whilst secondary endpoints were changes in circulating immune cell subsets including B cells, and cardiac and inflammatory biomarkers. A total of 24 patients were dosed. Rituximab appeared well tolerated. Seven serious adverse events were reported, none of which were assessed as being related to the rituximab infusion. Rituximab caused a mean 96.3% (95% confidence interval 93.8-98.8%) depletion of circulating B cells within 30 min of starting the infusion. Maximal B-cell depletion was seen at Day 6, which was significantly lower than baseline for all doses (P < 0.001). B-cell repopulation at 6 months was dose-dependent, with modulation of returning B-cell subsets. Immunoglobulin (IgG, IgM, and IgA) levels were not affected during the 6 months of follow-up.

Conclusions: A single infusion of rituximab appears safe when given in the acute STEMI setting and substantially alters circulating B-cell subsets. We provide important new insight into the feasibility and pharmacodynamics of rituximab in acute STEMI, which will inform further clinical translation of this potential therapy.

Clinical trial registration: NCT03072199 at https://www.clinicaltrials.gov/.

Keywords: B lymphocytes; Immune system; Rituximab; Myocardial infarction.

© The Author(s) 2021. Published by Oxford University Press on behalf of the European Society of Cardiology.

Figures

Graphical abstract
Graphical abstract
Figure 1
Figure 1
Consort diagram for RITA-MI. All patients completed dosing and follow-up. aLogistically not possible = dosing would have been on weekend which was not permitted, doctor/nurse not available to dose patient, mandated follow-up would have been on weekend or doctor not available. Top three reasons for ineligibility were timing of chest pain, previous medical history, clinical instability post-MI.
Figure 2
Figure 2
CD19+ B-cell depletion. Panel (A and B) shows the effect of four different doses of rituximab (200, 500, 700, and 1000 mg) on B-cell count and percentage (of lymphocytes) respectively. Each point and line represent a single patient in each group. Panel (C and D) shows the same data on a logarithmic scale. In panel C, blue and red dashed lines represent pre-specified levels of partial and complete B-cell depletion, respectively. Parametric ANOVA was performed comparing different timepoints against baseline. Significance was corrected for multiple testing. n = 24 patients (6/dose). **P < 0.01, ***P < 0.001, ****P < 0.0001.
Figure 3
Figure 3
Parts of whole stacked bar graph of B-cell subsets. The four columns of graphs show the data from the four rituximab doses (200, 500, 700, and 1000 mg). Panel A shows the absolute counts of B-cell subsets as a ‘part of whole’ stacked bar graph. Each colour represents a B-cell subset. Plasmablasts numbers are low and therefore poorly visible. The total of all the subsets in each bar equals total B-cell count. Panel B shows the same data but normalized so each subset is represented as a percentage of the whole. N = 24 patients (6/dose).
Figure 4
Figure 4
Grouped data showing cardiac biomarkers. Each panel shows the mean and 95% CI for each of the four rituximab doses (200, 500, 700, and 1000 mg) at different timepoints. Panels AD show troponin, NT pro-BNP, hsCRP, and interleukin-6, respectively. N = 24 patients (6/dose). BNP, NT-pro-B-type Natriuretic Peptide; hsCRP, high-sensitivity C-reactive protein.
Figure 5
Figure 5
Grouped data showing immunoglobulin counts. Each panel shows the mean and 95% CI for each of the four rituximab doses (200, 500, 700, and 1000 mg) at different timepoints. Panels AC show IgG, IgM, and IgA, respectively. N = 24 patients (6/dose). Ig, immunoglobulins.

References

    1. Bulluck H, Yellon DM, Hausenloy DJ.. Reducing myocardial infarct size: challenges and future opportunities. Heart 2016;102:341–348.
    1. Zhao TX, Mallat Z.. Targeting the immune system in atherosclerosis: JACC State-of-the-Art Review. J Am Coll Cardiol 2019;73:1691–1706.
    1. Conrad N, Judge A, Tran J, Mohseni H, Hedgecott D, Crespillo AP, Allison M, Hemingway H, Cleland JG, McMurray JJV, Rahimi K.. Temporal trends and patterns in heart failure incidence: a population-based study of 4 million individuals. Lancet 2018;391:572–580.
    1. Savarese G, Lund LH.. Global public health burden of heart failure. Card Fail Rev 2017;03:7.
    1. Ali M, Pulli B, Courties G, Tricot B, Sebas M, Iwamoto Y, Hilgendorf I, Schob S, Dong A, Zheng W, Skoura A, Kalgukar A, Cortes C, Ruggeri R, Swirski FK, Nahrendorf M, Buckbinder L, Chen JW.. Myeloperoxidase inhibition improves ventricular function and remodeling after experimental myocardial infarction. JACC Basic Transl Sci 2016;1:633–643.
    1. Horckmans M, Ring L, Duchene J, Santovito D, Schloss MJ, Drechsler M, Weber C, Soehnlein O, Steffens S.. Neutrophils orchestrate post-myocardial infarction healing by polarizing macrophages towards a reparative phenotype. Eur Heart J 2017;38:187–197.
    1. Nahrendorf M, Swirski FK, Aikawa E, Stangenberg L, Wurdinger T, Figueiredo J-L, Libby P, Weissleder R, Pittet MJ.. The healing myocardium sequentially mobilizes two monocyte subsets with divergent and complementary functions. J Exp Med 2007;204:3037–3047.
    1. Zouggari Y, Ait-Oufella H, Bonnin P, Simon T, Sage AP, Guérin C, Vilar J, Caligiuri G, Tsiantoulas D, Laurans L, Dumeau E, Kotti S, Bruneval P, Charo IF, Binder CJ, Danchin N, Tedgui A, Tedder TF, Silvestre J-S, Mallat Z.. B lymphocytes trigger monocyte mobilization and impair heart function after acute myocardial infarction. Nat Med 2013;19:1273–1280.
    1. Adamo L, Staloch LJ, Rocha-Resende C, Matkovich SJ, Jiang W, Bajpai G, Weinheimer CJ, Kovacs A, Schilling JD, Barger PM, Bhattacharya D, Mann DL.. Modulation of subsets of cardiac B lymphocytes improves cardiac function after acute injury. JCI Insight 2018;3:1–18.
    1. Dutta P, Courties G, Wei Y, Leuschner F, Gorbatov R, Robbins CS, Iwamoto Y, Thompson B, Carlson AL, Heidt T, Majmudar MD, Lasitschka F, Etzrodt M, Waterman P, Waring MT, Chicoine AT, van der Laan AM, Niessen HWM, Piek JJ, Rubin BB, Butany J, Stone JR, Katus HA, Murphy SA, Morrow DA, Sabatine MS, Vinegoni C, Moskowitz MA, Pittet MJ, Libby P, Lin CP, Swirski FK, Weissleder R, Nahrendorf M.. Myocardial infarction accelerates atherosclerosis. Nature 2012;487:325–329.
    1. Sage AP, Tsiantoulas D, Binder CJ, Mallat Z.. The role of B cells in atherosclerosis. Nat Rev Cardiol 2018;388:1459.
    1. Nus M, Sage AP, Lu Y, Masters L, Lam BYH, Newland S, Weller S, Tsiantoulas D, Raffort J, Marcus D, Finigan A, Kitt L, Figg N, Schirmbeck R, Kneilling M, Yeo GSH, Binder CJ, De La Pompa JL, Mallat Z.. Marginal zone B cells control the response of follicular helper T cells to a high-cholesterol diet. Nat Med 2017;23:601–610.
    1. Ait-Oufella H, Herbin O, Bouaziz J-DD, Binder CJ, Uyttenhove C, Laurans L, Taleb S, Van Vré E, Esposito B, Vilar J, Sirvent J, Van Snick J, Tedgui A, Tedder TF, Mallat Z.. B cell depletion reduces the development of atherosclerosis in mice. J Exp Med 2010;207:1579–1587.
    1. Kyaw T, Tay C, Khan A, Dumouchel V, Cao A, To K, Kehry M, Dunn R, Agrotis A, Tipping P, Bobik A, Toh B-H.. Conventional B2 B cell depletion ameliorates whereas its adoptive transfer aggravates atherosclerosis. J Immunol 2010;185:4410–4419.
    1. Salles G, Barrett M, Foà R, Maurer J, O'Brien S, Valente N, Wenger M, Maloney DG.. Rituximab in B-cell hematologic malignancies: a review of 20 years of clinical experience. Adv Ther 2017;34:2232–2273.
    1. EMC. MabThera 500 mg concentrate for solution for infusion—Summary of Product Characteristics (SmPC)—(eMC). 2008;1–21. .
    1. Poterucha JT, Westberg M, Nerheim P, Lovell JP.. Rituximab-induced polymorphic ventricular tachycardia. Texas Hear Inst J 2010;37:218–220.
    1. Arai Y, Tadokoro J, Mitani K.. Ventricular tachycardia associated with infusion of rituximab in mantle cell lymphoma. Am J Hematol 2005;78:317–318.
    1. Coiffier B, Lepage E, Brière J, Herbrecht R, Tilly H, Bouabdallah R, Morel P, Van Den Neste E, Salles G, Gaulard P, Reyes F, Lederlin P, Gisselbrecht C.. CHOP chemotherapy plus rituximab compared with CHOP alone in elderly patients with diffuse large-B-cell lymphoma. N Engl J Med 2002; 346:235–242.
    1. Armitage JD, Montero C, Benner A, Armitage JO, Bociek G.. Acute coronary syndromes complicating the first infusion of rituximab. Clin Lymphoma Myeloma 2008;8:253–255.
    1. Truffa AAM, Granger CB, White KR, Newby LK, Mehta RH, Hochman JS, Patel MR, Pieper KS, Al-Khalidi HR, Armstrong PW, Lopes RD.. Serious infection after acute myocardial infarction: incidence, clinical features, and outcomes. JACC Cardiovasc Interv 2012;5:769–776.
    1. Jose J. Statins and its hepatic effects: newer data, implications, and changing recommendations. J Pharm Bioallied Sci 2016;8:23–28.
    1. Tschöpe C, Van Linthout S, Spillmann F, Posch MG, Reinke P, Volk H-D, Elsanhoury A, Kühl U.. Targeting CD20+ B-lymphocytes in inflammatory dilated cardiomyopathy with rituximab improves clinical course: a case series. Eur Heart J Case Rep 2019;3:ytz131.
    1. Gonzalez-Juanatey C, Llorca J, Vazquez-Rodriguez TR, Diaz-Varela N, Garcia-Quiroga H, Gonzalez-Gay MA.. Short-term improvement of endothelial function in rituximab-treated rheumatoid arthritis patients refractory to tumor necrosis factor α blocker therapy. Arthritis Rheum 2008;59:1821–1824.
    1. Benucci M, Saviola G, Manfredi M, Sarzi-Puttini P, Atzeni F.. Factors correlated with improvement of endothelial dysfunction during rituximab therapy in patients with rheumatoid arthritis. Biol Targets Ther 2013;7:69–75.
    1. Novikova DS, Popkova TV, Lukina GV, Luchikhina EL, Karateev DE, Volkov AV, Novikov AA, Aleksandrova EN, Nasonov EL.. The effects of rituximab on lipids, arterial stiffness, and carotid intima-Media thickness in rheumatoid arthritis. J Korean Med Sci 2016;31:202–207.
    1. Simon Q, Pers JO, Cornec D, Le Pottier L, Mageed RA, Hillion S.. In-depth characterization of CD24highCD38high transitional human B cells reveals different regulatory profiles. J Allergy Clin Immunol 2016;137:1577–1584.e10.
    1. Biburger M, Aschermann S, Schwab I, Lux A, Albert H, Danzer H, Woigk M, Dudziak D, Nimmerjahn F.. Monocyte subsets responsible for immunoglobulin G-dependent effector functions in vivo. Immunity 2011;35:932–944.
    1. Uchida J, Hamaguchi Y, Oliver JA, Ravetch JV, Poe JC, Haas KM, Tedder TF.. The innate mononuclear phagocyte network depletes B lymphocytes through Fc receptor-dependent mechanisms during anti-CD20 antibody immunotherapy. J Exp Med 2004;199:1659–1669.
    1. Gong Q, Ou Q, Ye S, Lee WP, Cornelius J, Diehl L, Lin WY, Hu Z, Lu Y, Chen Y, Wu Y, Meng YG, Gribling P, Lin Z, Nguyen K, Tran T, Zhang Y, Rosen H, Martin F, Chan AC.. Importance of cellular microenvironment and circulatory dynamics in B cell immunotherapy. J Immunol 2005;174:817–826.
    1. Van Vollenhoven RF, Emery P, Bingham CO, Keystone EC, Fleischmann R, Furst DE, Macey K, Sweetser M, Kelman A, Ravi R.. Longterm safety of patients receiving rituximab in rheumatoid arthritis clinical trials. J Rheumatol 2010;37:558–567.
    1. Binder CJ, Papac-Milicevic N, Witztum JL.. Innate sensing of oxidation-specific epitopes in health and disease. Nat Rev Immunol 2016;16:485–497.
    1. Upadhye A, Srikakulapu P, Gonen A, Hendrikx S, Perry HM, Nguyen A, McSkimming C, Marshall MA, Garmey JC, Taylor AM, Bender TP, Tsimikas S, Holodick NE, Rothstein TL, Witztum JL, McNamara CA.. Diversification and CXCR4-dependent establishment of the bone marrow B-1a cell pool governs atheroprotective IgM production linked to human coronary atherosclerosis. Circ Res 2019;125:e55–e70.
    1. Hamaguchi Y, Uchida J, Cain DW, Venturi GM, Poe JC, Haas KM, Tedder TF.. The peritoneal cavity provides a protective niche for B1 and conventional B lymphocytes during anti-CD20 immunotherapy in mice. J Immunol 2005;174:4389–4399.
    1. Giugliano GR, Giugliano RP, Gibson CM, Kuntz RE.. Meta-analysis of corticosteroid treatment in acute myocardial infarction. Am J Cardiol 2003;91:1055–1059.
    1. Gilboa Y, Brauman A.. The effect of glucocorticosteroid administration on the survival of patients with acute myocardial infarction. Acta Cardiol 1977;32:55–63.

Source: PubMed

3
Sottoscrivi