Safety and efficacy of eculizumab in the prevention of antibody-mediated rejection in living-donor kidney transplant recipients requiring desensitization therapy: A randomized trial

William H Marks, Nizam Mamode, Robert A Montgomery, Mark D Stegall, Lloyd E Ratner, Lynn D Cornell, Ajda T Rowshani, Robert B Colvin, Bradley Dain, Judith A Boice, Denis Glotz, C10-001 Study Group, John Kanellis, Graeme Russ, Nassim Kamar, Yvon Lebranchu, Christophe Legendre, Lionel Rostaing, Christian Hugo, Giacomo Colussi, Paolo Rigotti, Anna Reisaeter, Frederic Oppenheimer, Lars Mjörnstedt, Gunnar Tufveson, Robert Higgins, Philip Mason, Nicholas Torpey, Anil Chandraker, Matthew Cooper, Jose El-Amm, A Osama Gaber, Roslyn Mannon, Christopher Marsh, Anup Patel, Flavio Vincenti, Patricia West-Thielke, Joshua Wolf, Steve Woodle, William H Marks, Nizam Mamode, Robert A Montgomery, Mark D Stegall, Lloyd E Ratner, Lynn D Cornell, Ajda T Rowshani, Robert B Colvin, Bradley Dain, Judith A Boice, Denis Glotz, C10-001 Study Group, John Kanellis, Graeme Russ, Nassim Kamar, Yvon Lebranchu, Christophe Legendre, Lionel Rostaing, Christian Hugo, Giacomo Colussi, Paolo Rigotti, Anna Reisaeter, Frederic Oppenheimer, Lars Mjörnstedt, Gunnar Tufveson, Robert Higgins, Philip Mason, Nicholas Torpey, Anil Chandraker, Matthew Cooper, Jose El-Amm, A Osama Gaber, Roslyn Mannon, Christopher Marsh, Anup Patel, Flavio Vincenti, Patricia West-Thielke, Joshua Wolf, Steve Woodle

Abstract

We report results of a phase 2, randomized, multicenter, open-label, two-arm study evaluating the safety and efficacy of eculizumab in preventing acute antibody-mediated rejection (AMR) in sensitized recipients of living-donor kidney transplants requiring pretransplant desensitization (NCT01399593). In total, 102 patients underwent desensitization. Posttransplant, 51 patients received standard of care (SOC) and 51 received eculizumab. The primary end point was week 9 posttransplant treatment failure rate, a composite of: biopsy-proven acute AMR (Banff 2007 grade II or III; assessed by blinded central pathology); graft loss; death; or loss to follow-up. Eculizumab was well tolerated with no new safety concerns. No significant difference in treatment failure rate was observed between eculizumab (9.8%) and SOC (13.7%; P = .760). To determine whether data assessment assumptions affected study outcome, biopsies were reanalyzed by central pathologists using clinical information. The resulting treatment failure rates were 11.8% and 21.6% for the eculizumab and SOC groups, respectively (nominal P = .288). When reassessment included grade I AMR, the treatment failure rates were 11.8% (eculizumab) and 29.4% (SOC; nominal P = .048). This finding suggests a potential benefit for eculizumab compared with SOC in preventing acute AMR in recipients sensitized to their living-donor kidney transplants (EudraCT 2010-019630-28).

Keywords: clinical research/practice; complement biology; donors and donation: living; immunosuppressant - fusion proteins and monoclonal antibodies; kidney transplantation/nephrology; rejection: antibody-mediated (ABMR); sensitization.

© 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
Study design. AMR, antibody‐mediated rejection; IVIg, intravenous immunoglobulin; MMF, mycophenolate mofetil; PP, plasmapheresis; SOC, standard of care; TAC, tacrolimus
Figure 2
Figure 2
Patient disposition. aFailed screening (n, %): did not meet inclusion/exclusion criteria (62, 22.5%), physician decision (n = 13, 4.7%), adverse event (3, 1.1%), withdrawal by patient (3, 1.1%), death (1, 0.4%), other (56, 20.4%). bScreened but not randomized (n, %): failed desensitization (18, 6.5%), adverse event (4, 1.5%), enrollment failure (3, 1.2%), failed inclusion/exclusion criteria (1, 0.4%), withdrawal by patient (1, 0.4%), other (6, 2.2%). cDid not receive treatment (n, %): donor changed mind (1, 0.4%), issues with donor kidney (1, 0.4%)
Figure 3
Figure 3
Graft survival through 36 months. Graft survival time was defined as the time from transplantation until the date of death or date of graft loss. A patient who did not die or have graft loss was censored at the time of last contact. *P = .209 at 36 months. SOC, standard of care

References

    1. Djamali A, Kaufman DB, Ellis TM, Zhong W, Matas A, Samaniego M. Diagnosis and management of antibody‐mediated rejection: current status and novel approaches. Am J Transplant. 2014;14(2):255‐271.
    1. Garonzik Wang JM, Montgomery RA, Kucirka LM, Berger JC, Warren DS, Segev DL. Incompatible live‐donor kidney transplantation in the United States: results of a national survey. Clin J Am Soc Nephrol. 2011;6(8):2041‐2046.
    1. Segev DL, Gentry SE, Warren DS, Reeb B, Montgomery RA. Kidney paired donation and optimizing the use of live donor organs. JAMA. 2005;293(15):1883‐1890.
    1. Lefaucheur C, Loupy A, Hill GS, et al. Preexisting donor‐specific HLA antibodies predict outcome in kidney transplantation. J Am Soc Nephrol. 2010;21(8):1398‐1406.
    1. Dunn TB, Noreen H, Gillingham K, et al. Revisiting traditional risk factors for rejection and graft loss after kidney transplantation. Am J Transplant. 2011;11(10):2132‐2143.
    1. Mohan S, Palanisamy A, Tsapepas D, et al. Donor‐specific antibodies adversely affect kidney allograft outcomes. J Am Soc Nephrol. 2012;23(12):2061‐2071.
    1. Burns JM, Cornell LD, Perry DK, et al. Alloantibody levels and acute humoral rejection early after positive crossmatch kidney transplantation. Am J Transplant. 2008;8(12):2684‐2694.
    1. Stegall MD, Chedid MF, Cornell LD. The role of complement in antibody‐mediated rejection in kidney transplantation. Nat Rev Nephrol. 2012;8(11):670‐678.
    1. Montgomery RA, Cozzi E, West LJ, Warren DS. Humoral immunity and antibody‐mediated rejection in solid organ transplantation. Semin Immunol. 2011;23(4):224‐234.
    1. Cernoch M, Viklicky O. Complement in kidney transplantation. Front Med (Lausanne). 2017;4:66.
    1. Tonelli M, Wiebe N, Knoll G, et al. Systematic review: kidney transplantation compared with dialysis in clinically relevant outcomes. Am J Transplant. 2011;11(10):2093‐2109.
    1. Orandi BJ, Chow EH, Hsu A, et al. Quantifying renal allograft loss following early antibody‐mediated rejection. Am J Transplant. 2015;15(2):489‐498.
    1. Singh N, Pirsch J, Samaniego M. Antibody‐mediated rejection: treatment alternatives and outcomes. Transplant Rev (Orlando). 2009;23(1):34‐46.
    1. Redfield RR, Ellis TM, Zhong W, et al. Current outcomes of chronic active antibody mediated rejection ‐ a large single center retrospective review using the updated BANFF 2013 criteria. Hum Immunol. 2016;77(4):346‐352.
    1. Lefaucheur C, Suberbielle‐Boissel C, Hill GS, et al. Clinical relevance of preformed HLA donor‐specific antibodies in kidney transplantation. Am J Transplant. 2008;8(2):324‐331.
    1. Couzi L, Manook M, Perera R, et al. Difference in outcomes after antibody‐mediated rejection between abo‐incompatible and positive cross‐match transplantations. Transpl Int. 2015;28(10):1205‐1215.
    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. Montgomery RA, Zachary AA, Racusen LC, et al. Plasmapheresis and intravenous immune globulin provides effective rescue therapy for refractory humoral rejection and allows kidneys to be successfully transplanted into cross‐match‐positive recipients. Transplantation. 2000;70(6):887‐895.
    1. Amrouche L, Aubert O, Suberbielle C, et al. Long‐term outcomes of kidney transplantation in patients with high levels of preformed DSA: the Necker high‐risk transplant program. Transplantation. 2017;101(10):2440‐2448.
    1. Wan SS, Ying TD, Wyburn K, Roberts DM, Wyld M, Chadban SJ. The treatment of antibody‐mediated rejection in kidney transplantation: an updated systematic review and meta‐analysis. Transplantation. 2018;102(4):557‐568.
    1. Thielke JJ, West‐Thielke PM, Herren HL, et al. Living donor kidney transplantation across positive crossmatch: the University of Illinois at Chicago experience. Transplantation. 2009;87(2):268‐273.
    1. Montgomery RA, Lonze BE, King KE, et al. Desensitization in HLA‐incompatible kidney recipients and survival. N Engl J Med. 2011;365(4):318‐326.
    1. Orandi BJ, Luo X, Massie AB, et al. Survival benefit with kidney transplants from HLA‐incompatible live donors. N Engl J Med. 2016;374(10):940‐950.
    1. Manook M, Koeser L, Ahmed Z, et al. Post‐listing survival for highly sensitised patients on the UK kidney transplant waiting list: a matched cohort analysis. Lancet. 2017;389(10070):727‐734.
    1. Sethi S, Choi J, Toyoda M, Vo A, Peng A, Jordan SC. Desensitization: overcoming the immunologic barriers to transplantation. J Immunol Res. 2017;2017:6804678.
    1. Stegall MD, Diwan T, Raghavaiah S, et al. Terminal complement inhibition decreases antibody‐mediated rejection in sensitized renal transplant recipients. Am J Transplant. 2011;11(11):2405‐2413.
    1. Locke JE, Magro CM, Singer AL, et al. The use of antibody to complement protein C5 for salvage treatment of severe antibody‐mediated rejection. Am J Transplant. 2009;9(1):231‐235.
    1. Solez K, Colvin RB, Racusen LC, et al. Banff 07 classification of renal allograft pathology: updates and future directions. Am J Transplant. 2008;8(4):753‐760.
    1. Stegall MD, Gloor J, Winters JL, Moore SB, Degoey S. A comparison of plasmapheresis versus high‐dose IVIG desensitization in renal allograft recipients with high levels of donor specific alloantibody. Am J Transplant. 2006;6(2):346‐351.
    1. Magee CC, Felgueiras J, Tinckam K, Malek S, Mah H, Tullius S. Renal transplantation in patients with positive lymphocytotoxicity crossmatches: one center's experience. Transplantation. 2008;86(1):96‐103.
    1. Vo AA, Peng A, Toyoda M, et al. Use of intravenous immune globulin and rituximab for desensitization of highly HLA‐sensitized patients awaiting kidney transplantation. Transplantation. 2010;89(9):1095‐1102.
    1. Stegall MD, Tayyab D, Cornell DL, Burns J, Dean GP, Gloor MJ. Terminal complement inhibition decreases early acute humoral rejection in sensitized renal transplant recipients [abstract 1]. Am J Transplant. 2010;10(Suppl 4):39.
    1. Santner TJ, Snell MK. Small‐sample confidence intervals for p 1– p 2 and p 1/p 2 in 2 × 2 contingency tables. J Am Stat Assoc. 1980;75(370):386‐394.
    1. Fisher RA. Statistical Methods for Research Workers. Edinburgh: Oliver and Boyd; 1925.
    1. Landis JR, Koch GG. The measurement of observer agreement for categorical data. Biometrics. 1977;33(1):159‐174.
    1. Marfo K, Lu A, Ling M, Akalin E. Desensitization protocols and their outcome. Clin J Am Soc Nephrol. 2011;6(4):922‐936.
    1. Glotz D, Russ G, Rostaing L, et al. Prevention of acute antibody‐mediated rejection in sensitized deceased‐donor kidney transplant recipients: 1‐year outcomes presented at American Society of Nephrology, Kidney Week November 3-8, 2015, San Diego. 2015.
    1. Gibson IW, Gwinner W, Brocker V, et al. Peritubular capillaritis in renal allografts: prevalence, scoring system, reproducibility and clinicopathological correlates. Am J Transplant. 2008;8(4):819‐825.
    1. Batal I, Lunz JG 3rd, Aggarwal N, et al. A critical appraisal of methods to grade transplant glomerulitis in renal allograft biopsies. Am J Transplant. 2010;10(11):2442‐2452.
    1. Smith B, Cornell LD, Smith M, et al. A method to reduce variability in scoring antibody‐mediated rejection in renal allografts: implications for clinical trials ‐ a retrospective study. Transpl Int. 2018;32:173‐183.
    1. Dean PG, Park WD, Cornell LD, Gloor JM, Stegall MD. Intragraft gene expression in positive crossmatch kidney allografts: ongoing inflammation mediates chronic antibody‐mediated injury. Am J Transplant. 2012;12(6):1551‐1563.
    1. Gloor JM, Winters JL, Cornell LD, et al. Baseline donor‐specific antibody levels and outcomes in positive crossmatch kidney transplantation. Am J Transplant. 2010;10(3):582‐589.
    1. Legendre CM, Licht C, Muus P, et al. Terminal complement inhibitor eculizumab in atypical hemolytic–uremic syndrome. N Engl J Med. 2013;368(23):2169‐2181.
    1. Licht C, Greenbaum LA, Muus P, et al. Efficacy and safety of eculizumab in atypical hemolytic uremic syndrome from 2‐year extensions of phase 2 studies. Kidney Int. 2015;87(5):1061‐1073.
    1. Hillmen P, Young NS, Schubert J, et al. The complement inhibitor eculizumab in paroxysmal nocturnal hemoglobinuria. N Engl J Med. 2006;355(12):1233‐1243.
    1. Loupy A, Lefaucheur C, Vernerey D, et al. Complement‐binding anti‐HLA antibodies and kidney‐allograft survival. N Engl J Med. 2013;369(13):1215‐1226.
    1. Lefaucheur C, Viglietti D, Hidalgo LG, et al. Complement‐activating anti‐HLA antibodies in kidney transplantation: allograft gene expression profiling and response to treatment. J Am Soc Nephrol. 2018;29(2):620‐635.
    1. Bouquegneau A, Loheac C, Aubert O, et al. Complement‐activating donor‐specific anti‐HLA antibodies and solid organ transplant survival: a systematic review and meta‐analysis. PLoS Med. 2018;15(5):e1002572.

Source: PubMed

3
Suscribir