We use cookies to improve your experience. By continuing to browse this site, you accept our cookie policy.×

In vitro cytokine release assays: reducing the risk of adverse events in man

    ,
    Daniel R Gliddon

    Huntingdon Life Sciences, Woolley Road, Alconbury, Cambridgeshire, PE28 4HS, UK

    ,
    Gregory Bannish

    Huntingdon Life Sciences, 100 Mettlers Road, Somerset, NJ 08873, USA

    ,
    Gary P Bembridge

    Huntingdon Life Sciences, Woolley Road, Alconbury, Cambridgeshire, PE28 4HS, UK

    &
    Lee A Coney

    Huntingdon Life Sciences, Woolley Road, Alconbury, Cambridgeshire, PE28 4HS, UK

    Published Online:https://doi.org/10.4155/bio.11.272

    The induction of cytokine release is a common consequence of the administration of therapeutic antibodies and in most cases is either tolerated by the patient or can be managed clinically by the administration of corticosteroids. However, in 2006, the administration of TGN1412 to six patients in a Phase I trial resulted in a unprecedentedly high level of cytokine release, systemic organ failure and the hospitalization of the subjects. Whilst the path to failure in this incident was multifactorial, at least one contributing factor was the lack of a robust in vitro model that would allow the prediction of the in vivo activity of a therapeutic antibody. In this article we review the current ‘state of the art’ of in vitro cytokine release assays and explore potential future developments.

    References

    • Abramowicz D, Schandene L, Goldman M et al. Release of tumor necrosis factor, interleukin-2, and γ-interferon in serum after injection of OKT3 monoclonal antibody in kidney transplant recipients. Transplantation47(4),606–608 (1989).
    • Winkler U, Jensen M, Manzke O, Schulz H, Diehl V, Engert A. Cytokine-release syndrome in patients with B cell chronic lymphocytic leukemia and high lymphocyte counts after treatment with an anti-CD20 monoclonal antibody (rituximab, IDEC-C2B8). Blood94(7),2217–2224 (1999).
    • Wing MG, Moreau T, Greenwood J et al. Mechanism of first dose cytokine-release syndrome by CAMPATH-1H: involvement of CD16 (FcγRIII) and CD11a/CD18(LFA-1) on NK cells. J. Clin. Invest.98,2819–2826 (1996).
    • Uppenkamp M, Engert A, Diehl V, Bunjes D, Huhn D, Brittinger G. Monoclonal antibody therapy with CAMPATH-1H in patients with relapsed high- and low-grade non-Hodgkin’s lymphomas: a multicenter Phase I/II study. Ann. Hematol.81(1),26–32 (2002).
    • Suntharalingam G, Perry MR, Ward S et al. Cytokine storm in a Phase I trial of the anti-CD28 monoclonal antibody TGN1412. N. Engl. J. Med.355(10),1018–1028 (2006).
    • Rebello PR, Hale G, Friend PJ, Cobbold SP, Waldmann H. Anti-globulin responses to rat and humanized CAMPATH-1 monoclonal antibody used to treat transplant rejection. Transplantation68(9),1417–1420 (1999).
    • Albert D, Dunham J, Khan S et al. Variability in the biological response to anti-CD20 B cell depletion in systemic lupus erythaematosus. Ann. Rheum. Dis.67(12),1724–1731 (2008).
    • Carpenter PA, Pavlovic S, Tso JY et al. Non-Fc receptor-binding humanized anti-CD3 antibodies induce apoptosis of activated human T cells. J. Immunol.165(11),6205–6213 (2000).
    • Xu D, Alegre ML, Varga SS et al.In vitro characterization of five humanized OKT3 effector function variant antibodies. Cell. Immunol.1,16–26 (2000).
    • 10  Armour KL, Parry-Jones DR, Beharry N et al. Intravascular survival of red cells coated with a mutated human anti-D antibody engineered to lack destructive activity. Blood107(7),2619–2626 (2006).
    • 11  Kirton CM, Laukkanen ML, Nieminen A et al. Function-blocking antibodies to human vascular adhesion protein-1: a potential anti-inflammatory therapy. Eur. J. Immunol.35(11),3119–3130 (2005).
    • 12  Desjarlais JR, Lazar GA. Modulation of antibody effector function. Exp. Cell Res.317(9),1278 (2011).
    • 13  Stavenhagen JB, Gorlatov S, Tuaillon N et al. Enhancing the potency of therapeutic monoclonal antibodies via Fc optimization. Adv. Enzyme Regul.48,152–164 (2008).
    • 14  Wilson NS, Yang B, Yang A et al. An Fcγ receptor-dependent mechanism drives antibody-mediated target-receptor signaling in cancer cells. Cancer Cell1(19),101–113 (2011).
    • 15  White AL, Chan HT, Roghanian A et al. Interaction with FcγRIIB is critical for the agonistic activity of anti-CD40 monoclonal antibody. J. Immunol.187,1754–1763 (2011).
    • 16  Li F, Ravetch JV. Inhibitory Fcγ receptor engagement drives adjuvant and anti-tumor activities of agonistic CD40 antibodies. Science333, 6045, 1030–1034 (2011).
    • 17  Chan AC, Carter PJ. Therapeutic antibodies for autoimmunity and inflammation. Nat. Rev. Immunol.10,301–316 (2010).
    • 18  Wing MG. Monoclonal antibody first dose cytokine release syndromes-mechanisms and prediction. J. Immunotoxicol.5(1),11–15 (2008).
    • 19  Bugelski PJ, Achuthanandam R, Capocasale RJ et al. Monoclonal antibody-induced cytokine-release syndrome. Expert Rev. Clin. Immunol.5(5),499–521 (2009).
    • 20  Wing MG, Waldmann H, Isaacs J, Compston DA, Hale G. Ex vivo whole blood cultures for predicting cytokine-release syndrome: dependence on target antigen and antibody isotype. Ther. Immunol.2(4),183–190 (1995).
    • 21  Findlay L, Eastwood F, Stebbings R et al. Improved in vitro methods to predict the in vivo toxicity in man of therapeutic monoclonal antibodies including TGN1412. J. Immunol. Meth.352(1–2),1–12 (2010).
    • 22  Walker M, Makropoulos D, Achuthanandam R, Bugelski PJ. Recent advances in the understanding of drug-mediated infusion reactions and cytokine release syndrome. Curr. Opin Drug Discov. Devel.13(1),124–135 (2010).
    • 23  Yang B, Pham TH, Goldbach-Mansky R, Gadina M. Accurate and simple measurement of the pro-inflammatory cytokine IL-1β using a whole blood stimulation assay. J. Vis. Exp.49,2662 (2011).
    • 24  Liebers V, Stubel H, Düser M, Brüning T, Raulf-Heimsoth M. Standardization of whole blood assay for determination of pyrogenic activity in organic dust samples. Int. J. Hyg. Environ. Health212(5),547–556 (2009).
    • 25  Bruhns P, Iannascoli B, England P et al. Specificity and affinity of human Fcγ receptors and their polymorphic variants for human IgG subclasses. Blood113(16),3716–3725 (2008).
    • 26  Chowdhury F, Williams A, Johnson P. Validation and comparison of two multiplex technologies, Luminex® and Mesoscale Discovery, for human cytokine profiling. J. Immunol. Meth.340(11),55–64 (2009).
    • 27  Arvå E, Andersson B. Kinetics of cytokine release and expression of lymphocyte cell-surface activation markers after in vitro stimulation of human peripheral blood mononuclear cells with Streptococcus pneumoniae.Scand. J. Immunol.49(3),411–416 (1999).
    • 28  Armour KL, van de Winkel JG, Williamson LM, Clark MR. Differential binding to human FcγRIIa and FcγRIIb receptors by human IgG wildtype and mutant antibodies. Mol. Immunol.40(9),585–593 (2003).
    • 29  Jefferis R. Glycosylation as a strategy to improve antibody-based therapeutics. Nat. Rev. Drug Discov.8,226–234 (2009).
    • 30  van der Neut Kolfschoten M, Schuurman J, Losen M et al. Anti-inflammatory activity of human IgG4 antibodies by dynamic Fab arm exchange. Science317,1507–1508 (2007).
    • 101  Common Terminology Criteria for Adverse Events (CTCAE). www.eortc.be/services/doc/ctc/CTCAE_4.03_2010-06-14_QuickReference_5x7.pdf