Safety, pharmacokinetics, and pharmacodynamic activity of obinutuzumab, a type 2 anti-CD20 monoclonal antibody for the desensitization of candidates for renal transplant

Robert R Redfield, Stanley C Jordan, Stephan Busque, Flavio Vincenti, E Steve Woodle, Niraj Desai, Elaine F Reed, Simon Tremblay, Andrea A Zachary, Ashley A Vo, Richard Formica, Thomas Schindler, Ha Tran, Caroline Looney, Candice Jamois, Cherie Green, Alyssa Morimoto, Richa Rajwanshi, Aaron Schroeder, Matthew D Cascino, Paul Brunetta, Dominic Borie, Robert R Redfield, Stanley C Jordan, Stephan Busque, Flavio Vincenti, E Steve Woodle, Niraj Desai, Elaine F Reed, Simon Tremblay, Andrea A Zachary, Ashley A Vo, Richard Formica, Thomas Schindler, Ha Tran, Caroline Looney, Candice Jamois, Cherie Green, Alyssa Morimoto, Richa Rajwanshi, Aaron Schroeder, Matthew D Cascino, Paul Brunetta, Dominic Borie

Abstract

The limited effectiveness of rituximab plus intravenous immunoglobulin (IVIG) in desensitization may be due to incomplete B cell depletion. Obinutuzumab is a type 2 anti-CD20 antibody that induces increased B cell depletion relative to rituximab and may therefore be more effective for desensitization. This open-label phase 1b study assessed the safety, pharmacokinetics, and pharmacodynamics of obinutuzumab in highly sensitized patients with end-stage renal disease. Patients received 1 (day 1, n = 5) or 2 (days 1 and 15; n = 20) infusions of 1000-mg obinutuzumab followed by 2 doses of IVIG on days 22 and 43. Eleven patients received additional obinutuzumab doses at the time of transplant and/or at week 24. The median follow-up duration was 9.4 months. Obinutuzumab was well tolerated, and most adverse events were grade 1-2 in severity. There were 11 serious adverse events (SAEs) in 9 patients (36%); 10 of these SAEs were infections and 4 occurred after kidney transplant. Obinutuzumab plus IVIG resulted in profound peripheral B cell depletion and appeared to reduce B cells in retroperitoneal lymph nodes. Reductions in anti-HLA antibodies, number of unacceptable antigens, and the calculated panel reactive antibody score as centrally assessed using single-antigen bead assay were limited and not clinically meaningful for most patients (NCT02586051).

Keywords: B cell biology; alloantibody; clinical research/practice; clinical trial; immunosuppressant - fusion proteins and monoclonal antibodies: B cell specific; immunosuppression/immune modulation; kidney transplantation/nephrology; pharmacology.

© 2019 The Authors. American Journal of Transplantation published by Wiley Periodicals, Inc. on behalf of The American Society of Transplantation and the American Society of Transplant Surgeons.

Figures

Figure 1
Figure 1
Patient disposition and number of obinutuzumab (Obi) doses. aThis patient received 1 obinutuzumab dose before transplant and 1 dose after. bThis patient received a transplant at week 1 and was withdrawn from study treatment. cThese 5 patients received 2 obinutuzumab doses before transplant and 1 dose after. dThis patient received 2 obinutuzumab doses before transplant and 2 doses after
Figure 2
Figure 2
CD19+ peripheral B cell counts in patients from cohort 1 and cohort 2 during the desensitization phase. Cell counts are given in number of cells per μL as a function of study days. The lower limit of quantification of the high‐sensitivity flow cytometry assay was 0.441 cells/μL and is indicated with a dashed line. Traces for the 2 patients with detectable antidrug antibodies are marked with an asterisk
Figure 3
Figure 3
CD19+ B cell frequency in peritoneal lymph nodes of patients who received a transplant (7 patients, 13 nodes)
Figure 4
Figure 4
Mean percent reduction in MFI and 95% CIs from baseline to week 24 (filled circles) or last valid MFI measurement (open circles); limited to alleles with MFI > 3000 at any time point and baseline or posttreatment MFI > 500. Marker size is proportional to the number of alleles per patient. Mean and 95% CIs were calculated using bootstrapping with bias‐corrected CI. Patients who have received a transplant are encircled in green; patients identified as biological responders by the DASP are boxed. Abbreviations: CI, confidence interval; DASP, desensitization assessment scientific panel; MFI, mean fluorescence intensity
Figure 5
Figure 5
Change in the number of UAs (A, B) and CPRA score (C, D) from baseline to week 24 or last valid MFI measurement. For UAs and CPRA presented in panels B and D, change in acceptability of a given antigen needed not only to surpass the MFI threshold of 3000 but also display a robust change in MFI of 50%. Abbreviations: CFB, change from baseline; CPRA, calculated panel reactive antibody; MFI, mean fluorescence intensity; UA, unacceptable antigen

References

    1. Terasaki PI, Ozawa M. Predicting kidney graft failure by HLA antibodies: a prospective trial. Am J Transplant. 2004;4(3):438‐443.
    1. Jordan SC, Pescovitz MD. Presensitization: the problem and its management. Clin J Am Soc Nephrol. 2006;1(3):421‐432.
    1. Hart A, Smith JM, Skeans MA, et al. OPTN/SRTR 2016 annual data Report: kidney. Am J Transplant. 2018;18(suppl 1):18‐113.
    1. Meier‐Kriesche HU, Port FK, Ojo AO, et al. Effect of waiting time on renal transplant outcome. Kidney Int. 2000;58(3):1311‐1317.
    1. Nankivell BJ, Alexander SI. Rejection of the kidney allograft. N Engl J Med. 2010;363(15):1451‐1462.
    1. Matas AJ, Smith JM, Skeans MA, et al. OPTN/SRTR 2013 annual data report: kidney. Am J Transplant. 2015;15(suppl 2):1‐34.
    1. Vo AA, Lukovsky M, Toyoda M, et al. Rituximab and intravenous immune globulin for desensitization during renal transplantation. N Engl J Med. 2008;359(3):242‐251.
    1. Ramos EJ, Pollinger HS, Stegall MD, Gloor JM, Dogan A, Grande JP. The effect of desensitization protocols on human splenic B‐cell populations in vivo. Am J Transplant. 2007;7(2):402‐407.
    1. Marfo K, Ling M, Bao Y, et al. Lack of effect in desensitization with intravenous immunoglobulin and rituximab in highly sensitized patients. Transplantation. 2012;94(4):345‐351.
    1. Genberg H, Hansson A, Wernerson A, Wennberg L, Tyden G. Pharmacodynamics of rituximab in kidney allotransplantation. Am J Transplant. 2006;6(10):2418‐2428.
    1. Thaunat O, Patey N, Gautreau C, et al. B cell survival in intragraft tertiary lymphoid organs after rituximab therapy. Transplantation. 2008;85(11):1648‐1653.
    1. Kamburova EG, Koenen HJ, Borgman KJ, ten Berge IJ, Joosten I, Hilbrands LB. A single dose of rituximab does not deplete B cells in secondary lymphoid organs but alters phenotype and function. Am J Transplant. 2013;13(6):1503‐1511.
    1. Mossner E, Brunker P, Moser S, et al. Increasing the efficacy of CD20 antibody therapy through the engineering of a new type II anti‐CD20 antibody with enhanced direct and immune effector cell‐mediated B‐cell cytotoxicity. Blood. 2010;115(22):4393‐4402.
    1. Goede V, Fischer K, Busch R, et al. Obinutuzumab plus chlorambucil in patients with CLL and coexisting conditions. N Engl J Med. 2014;370(12):1101‐1110.
    1. Levey AS, Stevens LA, Schmid CH, et al. A new equation to estimate glomerular filtration rate. Ann Intern Med. 2009;150(9):604‐612.
    1. Reed EF, Rao P, Zhang Z, et al. Comprehensive assessment and standardization of solid phase multiplex‐bead arrays for the detection of antibodies to HLA. Am J Transplant. 2013;13(7):1859‐1870.
    1. Beal S, Sheiner L, Boeckmann A, Bauer RJ. NONMEM Users Guides (1989–2011). Ellicott City, MD: Icon Development Solutions; 2011.
    1. Gibiansky E, Gibiansky L, Carlile DJ, Jamois C, Buchheit V, Frey N. Population pharmacokinetics of obinutuzumab (GA101) in chronic lymphocytic leukemia (CLL) and non‐Hodgkin's lymphoma and exposure‐response in CLL. CPT Pharmacometrics Syst Pharmacol. 2014;3:e144.
    1. Gosset C, Lefaucheur C, Glotz D. New insights in antibody‐mediated rejection. Curr Opin Nephrol Hypertens. 2014;23(6):597‐604.
    1. Dirks NL, Meibohm B. Population pharmacokinetics of therapeutic monoclonal antibodies. Clin Pharmacokinet. 2010;49(10):633‐659.
    1. Alexander T, Sarfert R, Klotsche J, et al. The proteasome inhibitior bortezomib depletes plasma cells and ameliorates clinical manifestations of refractory systemic lupus erythematosus. Ann Rheum Dis. 2015;74(7):1474‐1478.
    1. Diwan TS, Raghavaiah S, Burns JM, Kremers WK, Gloor JM, Stegall MD. The impact of proteasome inhibition on alloantibody‐producing plasma cells in vivo. Transplantation. 2011;91(5):536‐541.
    1. Moreno Gonzales MA, Gandhi MJ, Schinstock CA, et al. 32 doses of bortezomib for desensitization is not well tolerated and is associated with only modest reductions in anti‐HLA antibody. Transplantation. 2017;101(6):1222‐1227.
    1. Driscoll J, Tremblay S, Dasgupta N, Aronow B, Singh H, Woodle ES. Genomic and biochemical proteasomal adaptations in carfilzomib‐resistant plasma cells that limit HLA‐desensitization in transplantation candidates. Blood. 2017;130(suppl 1):5448.
    1. Woodle ES, Shields AR, Ejaz NS, et al. Prospective iterative trial of proteasome inhibitor‐based desensitization. Am J Transplant. 2015;15(1):101‐118.
    1. Kwun J, Burghuber C, Manook M, et al. Humoral compensation after bortezomib treatment of allosensitized recipients. J Am Soc Nephrol. 2017;28(7):1991‐1996.
    1. van de Donk NW. Carfilzomib versus bortezomib: no longer an ENDEAVOR. Lancet Oncol. 2017;18(10):1288‐1290.
    1. Tremblay S, Shields AR, Alloway R, et al. A prospective iterative trial of carfizomib‐based desensitization trial: initial comparative observations. Am J Transplant. 2017;17(suppl 3).
    1. Chu VT, Beller A, Nguyen TT, Steinhauser G, Berek C. The long‐term survival of plasma cells. Scand J Immunol. 2011;73(6):508‐511.
    1. Jordan SC, Tyan D, Stablein D, et al. Evaluation of intravenous immunoglobulin as an agent to lower allosensitization and improve transplantation in highly sensitized adult patients with end‐stage renal disease: report of the NIH IG02 trial. J Am Soc Nephrol. 2004;15(12):3256‐3262.
    1. Tambur AR, Campbell P, Claas FH, et al. Sensitization in Transplantation: assessment of Risk (STAR) 2017 Working Group meeting report. Am J Transplant. 2018;18(7):1604‐1614.
    1. Vo AA, Choi J, Cisneros K, et al. Benefits of rituximab combined with intravenous immunoglobulin for desensitization in kidney transplant recipients. Transplantation. 2014;98(3):312‐319.
    1. Zachary AA, Lucas DP, Montgomery RA, Leffell MS. Rituximab prevents an anamnestic response in patients with cryptic sensitization to HLA. Transplantation. 2013;95(5):701‐704.

Source: PubMed

3
Se inscrever