Outcomes at 3 years posttransplant in imlifidase-desensitized kidney transplant patients

Christian Kjellman, Angela Q Maldonado, Kristoffer Sjöholm, Bonnie E Lonze, Robert A Montgomery, Anna Runström, Tomas Lorant, Niraj M Desai, Christophe Legendre, Torbjörn Lundgren, Bengt von Zur Mühlen, Ashley A Vo, Håkan Olsson, Stanley C Jordan, Christian Kjellman, Angela Q Maldonado, Kristoffer Sjöholm, Bonnie E Lonze, Robert A Montgomery, Anna Runström, Tomas Lorant, Niraj M Desai, Christophe Legendre, Torbjörn Lundgren, Bengt von Zur Mühlen, Ashley A Vo, Håkan Olsson, Stanley C Jordan

Abstract

Imlifidase is a cysteine proteinase which specifically cleaves IgG, inhibiting Fc-mediated effector function within hours of administration. Imlifidase converts a positive crossmatch to a potential donor (T cell, B cell, or both), to negative, enabling transplantation to occur between previously HLA incompatible donor-recipient pairs. To date, 39 crossmatch positive patients received imlifidase prior to a kidney transplant in four single-arm, open-label, phase 2 studies. At 3 years, for patients who were AMR+ compared to AMR-, death-censored allograft survival was 93% vs 77%, patient survival was 85% vs 94%, and mean eGFR was 49 ml/min/1.73 m2 vs 61 ml/min/1.73 m2 , respectively. The incidence of AMR was 38% with most episodes occurring within the first month post-transplantation. Sub-analysis of patients deemed highly sensitized with cPRA ≥ 99.9%, and unlikely to be transplanted who received crossmatch-positive, deceased donor transplants had similar rates of patient survival, graft survival, and eGFR but a higher rate of AMR. These data demonstrate that outcomes and safety up to 3 years in recipients of imlifidase-enabled allografts is comparable to outcomes in other highly sensitized patients undergoing HLA-incompatible transplantation. Thus, imlifidase is a potent option to facilitate transplantation among patients who have a significant immunologic barrier to successful kidney transplantation. Clinical Trial: ClinicalTrials.gov (NCT02790437), EudraCT Number: 2016-002064-13.

Keywords: alloantibody; clinical research/practice; crossmatch; desensitization; immunosuppressant - other; immunosuppression/immune modulation; kidney transplantation/nephrology; sensitization.

Conflict of interest statement

The authors of this manuscript have conflicts of interest to disclose as described by the American Journal of Transplantation. Christian Kjellman, Angela Q. Maldonado, Kristoffer Sjöholm, Håkan Olsson, Anna Runström, and Tomas Lorant receive employment funds from and have a personal financial interest in Hansa Biopharma AB. Bonnie E. Lonze, Robert A. Montgomery, Tomas Lorant, Niraj M. Desai, Christophe Legendre, Torbjörn Lundgren, Bengt von Zur Mühlen, Ashley A. Vo, and Stanley C. Jordan receive research funds from Hansa Biopharma AB.

© 2021 The Authors. American Journal of Transplantation published by Wiley Periodicals LLC on behalf of The American Society of Transplantation and the American Society of Transplant Surgeons.

Figures

FIGURE 1
FIGURE 1
Imlifidase 46 patient enrollment and follow up. Flow chart of the enrolment in feeder studies and in the long‐term follow‐up study (17‐HMedIdeS‐14), and what data is analyzed in the patients
FIGURE 2
FIGURE 2
Patient survival estimated with Kaplan‐Meier. Alive patents are censored at last known visit, indicated in graphics as a thin vertical line. (A) XM+ patients. (B) XM+ patients separated by AMR or No AMR
FIGURE 3
FIGURE 3
Graft survival estimated with Kaplan–Meier. Patients without graft loss are censored last known visit or death, indicated in graphics as a thin vertical line. (A) XM+ patients. (B) XM+ patients separated by AMR or No AMR
FIGURE 4
FIGURE 4
Estimated GFR with MDRD. eGFR with individual patient data (thin gray lines), mean (black line), standard deviation (error bars), and red line is a linear regression from 30 days up to 3 years (A) XM+ patients. (B) XM+ patients separated by AMR (solid lines) or No AMR (dashed lines)
FIGURE 5
FIGURE 5
Immunodominant DSA with individual patient data. Immunodominant DSA with individual patient data (thin gray lines), mean (colored lines with points and SD as error bars), and linear regression (dashed colored lines) from 14 days up to 3 years for HLA class I and II. (A) XM+ patients. (B) XM+ patients with AMR. (C) XM+ patients without AMR
FIGURE 6
FIGURE 6
Time to first AMR. Visualized using reverse Kaplan–Meier estimate for cumulative incidence of AMR for XM+ patients. Patients without AMR are censored last known visit or death, indicated in graphics as a thin vertical line
FIGURE 7
FIGURE 7
Intensity of 6 patient's individual DSA over time. DSA (MFI ≥ 1000 at any time) for each patient visualized as a line, color depending on HLA‐type, if multiple DSA for one type one line is dashed. (A,B) Patients without AMR, (C–F) patients with AMR
FIGURE 8
FIGURE 8
Outcome of the group with XM+, DD, and cPRA ≥ 99.9%. (A) Patient survival. (B) Graft survival, death censored. (C) Time to first AMR. (D) eGFR with individual patient data (thin gray lines), mean (black line with points), standard deviation (gray ribbon) and red line is a linear regression from 30 days up to 3 years. (E) Immunodominant DSA with individual patient data (thin gray lines), mean (colored lines with points and SD as error bars) and linear regression (dashed colored lines) from 14 days up to 2 years for HLA class I and II

References

    1. Habal MV, Farr M, Restaino S, Chong A. Desensitization in the era of precision medicine: moving from the bench to bedside. Transplantation. 2019;103(8):1574‐1581.
    1. Jackson KR, Holscher C, Motter JD, et al. Posttransplant outcomes for CPRA‐100% recipients under the new kidney allocation system. Transplantation. 2020;104(7):1456‐1461.
    1. Schinstock CA, Smith BH, Montgomery RA, et al. Managing highly sensitized renal transplant candidates in the era of kidney paired donation and the new kidney allocation system: is there still a role for desensitization? Clin Transplant. 2019;33(12):e13751.
    1. Vo AA, Petrozzino J, Yeung K, et al. Efficacy, outcomes, and cost‐effectiveness of desensitization using IVIG and rituximab. Transplantation. 2013;95(6):852‐858.
    1. Jackson AM, Kraus ES, Orandi BJ, Segev DL, Montgomery RA, Zachary AA. A closer look at rituximab induction on HLA antibody rebound following HLA‐incompatible kidney transplantation. Kidney Int. 2015;87(2):409‐416.
    1. Jackson KR, Covarrubias K, Holscher CM, et al. The national landscape of deceased donor kidney transplantation for the highly sensitized: transplant rates, waitlist mortality, and posttransplant survival under KAS. Am J Transplant. 2018;4:1129‐1138.
    1. Jackson KR, Motter JD, Kernodle A, et al. How do highly sensitized patients get kidney transplants in the United States? Trends over the last decade. Am J Transplant. 2020;20:2101‐2112.
    1. EUROSTAM . Final Report Summary ‐ EUROSTAM (A Europe‐wide Strategy to enhance Transplantation of highly sensitized patients on basis of Acceptable HLA Mismatches.). 2018. . Accessed 10 January 2018.
    1. Ge S, Chu M, Choi J, et al. Imlifidase inhibits HLA antibody‐mediated NK cell activation and antibody‐dependent cell‐mediated cytotoxicity (ADCC) in vitro. Transplantation. 2020;104(8):1574‐1579.
    1. Lorant T, Bengtsson M, Eich T, et al. Safety, immunogenicity, pharmacokinetics, and efficacy of degradation of anti‐HLA antibodies by IdeS (imlifidase) in chronic kidney disease patients. Am J Transplant. 2018;18(11):2752‐2762.
    1. Jordan SC, Lorant T, Choi J, et al. IgG endopeptidase in highly sensitized patients undergoing transplantation. N Engl J Med. 2017;377(5):442‐453.
    1. Lonze BE, Tatapudi VS, Weldon EP, et al. IdeS (Imlifidase): a novel agent that cleaves human IgG and permits successful kidney transplantation across high‐strength donor‐specific antibody. Ann Surg. 2018;268:488‐496.
    1. Jordan SC, Legendre C, Desai N, et al. Imlifidase desensitization in crossmatch‐positive, highly‐sensitized kidney transplant recipients: results of an international phase 2 trial (Highdes). Transplantation. 2020;268:488‐496.
    1. Stewart DE, Kucheryavaya AY, Klassen DR, Turgeon NA, Formica RN, Aeder MI. Changes in deceased donor kidney transplantation one year after KAS implementation. Am J Transplant. 2016;16:1834‐1847.
    1. Hahn AB, Mackey M, Constantino D, et al. The new kidney allocation system does not equally advantage all very high CPRA candidates – a single center analysis. Hum Immunol. 2017;78:37‐40.
    1. Stewart DE, Wilk AR, Toll AE, et al. Measuring and monitoring equity in access to deceased donor kidney transplantation. Am J Transplant. 2018;18:1924‐1935.
    1. Stewart DE, Wilk AR, Klassen DK. KAS turns four: the state of deceased donor kidney allocation in the U.S. OBM Transplantation. 2019;3:1. 10.21926/obm.transplant.1901041.
    1. Meier‐Kriesche HU, Schold JD, Srinivas TR, Reed A, Kaplan B. Kidney transplantation halts cardiovascular disease progression in patients with end‐stage renal disease. Am J Transplant. 2004;4(10):1662‐1668.
    1. Meier‐Kriesche HU, Baliga R, Kaplan B. Decreased renal function is a strong risk factor for cardiovascular death after renal transplantation. Transplantation. 2003;75(8):1291‐1295.
    1. Heidt S, Haasnoot GW, Witvliet MD, et al. Allocation to highly sensitized patients based on acceptable mismatches results in low rejection rates comparable to nonsensitized patients. Am J Transplant. 2019;19(10):2926‐2933.
    1. Orandi BJ, Luo X, Massie AB, et al. Surival benefit of kidney transplants from HLA‐incomptable live donors. N Engl J Med. 2016;374:940‐950.
    1. Kasiske BL, Israni AK, Snyder JJ, Skeans MA. Patient outcomes in renal transplantation I. The relationship between kidney function and long‐term graft survival after kidney transplant. Am J Kidney Dis. 2011;57(3):466‐475.
    1. Duquesnoy R, Marrari M. Detection of antibodies against HLA‐C epitopes in patients with rejected kidney transplants. Transpl Immunol. 2011;24(3):164‐171.
    1. Tinckam K, Rose C, Hariharan S, Gill J. Re‐examining risk of repeated HLA mismatch in kidney transplantation. J Am Soc Nephrol. 2016;27(9):2833‐2841.
    1. Heidt S, Roelen D, de Vaal Y, et al. A novel ELISPOT assay to quantify HLA‐specific B cells in HLA‐immunized individuals. Am J Transplant. 2012;12(6):1469‐1478.
    1. Gaston RS, Fieberg A, Helgeson ES, et al. Late graft loss after kidney transplantation: is "death with function" really death with a functioning allograft? Transplantation. 2020;104(7):1483‐1490.

Source: PubMed

3
Předplatit