Download

REVIEW ARTICLE

Therapeutic Apheresis in Glomerular Diseases after Kidney Transplantation

Salvadori Maurizio1*, Tsalouchos Aris2

1Renal Unit, Careggi University Hospital, Florence, Italy

2Division of Nephrology and Dialysis Unit, Saints Cosmas and Damian Hospital, Pescia, Italy

Abstract

Therapeutic apheresis is an extracorporeal treatment that selectively separates abnormal cells or substances from the blood that are linked with or cause certain disease states. It is widely used in transplantation medicine as an adjunctive therapeutic option. In kidney transplantation (KT), recurrent and de novo glomerular diseases represent the third most common cause of graft failure beyond the first year after transplantation, as current therapeutic options are limited. Evidence to support the use of therapeutic apheresis in these conditions is scarce, as it is only supported by observational studies. The purpose of this review was to examine and clarify the potential role of therapeutic apheresis and describe current evidence in the treatment of recurrent and de novo glomerular diseases after KT.

Key words: glomerular diseases, kidney transplantation, therapeutic apheresis

Received: 1 March 2020; Accepted after Revision: 6 May 2020; Published: 13 May 2020

Author for correspondence: Maurizio Salvadori, Renal Unit, Careggi University Hospital, Viale Pieraccini, Florence, Italy. Email: maurizio.salvadori1@gmail.com

How to cite: Salvadori M and Tsalouchos A. Therapeutic Apheresis in Glomerular Diseases after Kidney Transplantation. J Ren Hepat Diord. 2020; 4(1):10–17.

Doi: https://doi.org/10.15586/jrenhep.v4i1.65

Copyright: Salvadori M and Tsalouchos A

License: This open access article is licensed under Creative Commons Attribution 4.0 International (CC BY 4.0). http://creativecommons.org/licenses/by/4.0

Introduction

Recurrent and de novo glomerular diseases after kidney transplantation (KT) have a powerful impact on transplant survival. After death with a functioning graft and chronic rejection represent the third most common cause of graft failure beyond the first year from KT (1). The Renal Allograft Disease Registry (RADR) data, a consortium of six American transplant centers, examining the rate of recurrent and de novo glomerular diseases after KT, showed a prevalence of 2.8% after 2 years, 9.8% after 5 years, and 18.5% after 8 years of follow-up. In addition, the occurrence of graft failure in recipients with recurrent or de novo glomerular disease was double that of those who did not experience these clinical conditions after KT (2). Briganti et al., in analyzing registry data from the Australia and New Zealand Dialysis and Transplant Registry (ANZDATA), found that 0.5% of grafts within 1 year after KT, 3.7% within 5 years, and 8.4% within 10 years were lost due to recurrent glomerular disease (1). However, the incidence and outcomes of recurrence vary to a great extent. For example, primary focal segmental glomerulosclerosis (FSGS) and C3 glomerulopathy recur frequently; however, the disease tends to have a slow progression in terms of graft loss, and graft survival beyond 10 years is typical (1). On the other hand, anti-glomerular basement membrane (GBM) disease and anti-neutrophil cytoplasmic antibody (ANCA)-associated vasculitis recur only rarely, but when it does, it is likely to cause rapid graft loss. In the case of complement-mediated atypical hemolytic uremic syndrome (aHUS), recurrence and its impact on graft survival are dependent on the complement pathway mutation responsible.

The treatment of these conditions is often challenging for transplant nephrologists. Therapeutic apheresis (TA) techniques are widely used in KT in desensitization protocols for ABO-incompatible KT, in patients with preformed HLA-antibodies and those undergoing treatment for antibody-mediated rejection (AMR). Although evidence from adequately powered randomized controlled trials supporting the use of TA to treat recurrent and de novo glomerular diseases after KT is lacking, because of the rarity of these diseases, the use of TA, mainly therapeutic plasma exchange (TPE), is widespread in transplant centers. This review examines the evidence supporting the application of TA in treating recurrent and de novo glomerular diseases after KT.

Recurrence of primary focal segmental glomerulosclerosis

Around 30% of the patients with primary FSGS experience disease recurrence after first KT, and in one series, early graft loss occurred at an average of 24 months after diagnosis of recurrence in 14 among 25 patients with recurrent FSGS (3). The possibility of the disease to recur is even higher than 75% after a second KT when the first graft failed because of recurrence (4).

Primary FSGS is probably triggered by a circulating permeability factor that targets the glomerular filtration barrier, especially podocytes. Until now, different plasma molecules have been hypothesized to be implicated in the disease pathogenesis. However, none of them have demonstrated a clear pathogenic correlation (5). Delville et al. (6) have recently described a panel of seven antibodies (CD40, PTPRO, CGB5, FAS, P2RY11, SNRPB2, and APOL2) that predict FSGS recurrence after KT, with 92% accuracy. An elevation of the titers of anti-CD40 antibody alone, in the period before transplantation, had the best prediction (78% accuracy), with the disease recurrence after KT (6). Furthermore, the administration of anti-CD40 antibodies, purified from patients with FSGS recurrence, in cultures of human podocytes has been proven to be particularly pathogenic (6).

Over the years, for removing the putative circulating permeability factor responsible for the pathogenesis of the disease, TPE and Immuno-Absorption (IA) (with either a staphylococcal protein A [SPA] or anti-IgG column), alone or in sequence with cyclophosphamide, have been employed with benefit in patients with recurrent disease after KT (711). In support of this hypothesis, Dantal et al. (11) showed that the administration to rats of material eluted from SPA columns coming from patients with recurrent FSGS after KT result in a substantial increase in albuminuria.

In a systematic review of the literature, Ponticelli highlighted that about 70% of children and 63% of adult transplant recipients with FSGS recurrence who have been treated with TPE or IA sessions had complete or partial remission of proteinuria (12). Two recent meta-analyses have published comparable outcomes (13,14).

There are currently no precise indications on the duration and frequency of TPE sessions. A therapeutic regimen of TPE sessions typically used consists of 1.5 plasma volume exchanges for three consecutive days following treatment on alternate days for a total duration of 2 weeks (15).

TPE sessions have also been applied in protocols with various immunosuppressive agents. In a series of 10 patients, Canaud et al. (16) showed favorable outcomes by combining frequent TPE sessions, slowly tapered down for 9 months, with intravenous cyclosporine plus high-dose steroids and mycophenolate.

The use of Rituximab (RTX) in the recurrence of FSGS after KT has rapidly increased with beneficial outcomes in the last 10 years (17,18). RTX, other than being a selective depleting agent of B lymphocytes, provides a direct protective effect on podocytes. Fornoni et al. showed that RTX protects sphingomyelin phosphodiesterase acid-like 3b (SMPDL-3b) and acid sphingomyelinase (ASMase) by binding to SMPDL-3b, a protein that is mainly expressed in the lipid rafts of podocytes, and could be a target of the permeability factor (19,20). As evidenced in several case reports, the use of RTX combined with TPE looks to be more effective than the sole use of TPE (21,22). Of importance, the mean percentage of RTX removal during a first TPE session range between 47 and 54% when TPE is performed between 24 and 72 h after RTX infusion (23). This pharmacokinetic observation should be taken into account for RTX re-dosing after TPE sessions.

Recently, Martin-Moreno et al. (24) presented the case of a male patient with severe recurrence of FSGS (massive proteinuria) after KT without response to TPE sessions, and partial remission to a combination of IA and cycles of RTX. The importance of IA, as an alternative to TPE, in the treatment of recurrent FSGS after KT, and in general in the case of idiopathic nephrotic syndrome, has been highlighted in a recent review by Kronbichler et al. (25). Now randomized trials are needed to draw conclusions on whether IA is superior to TPE in terms of efficacy, safety, and quality of life.

Complement-mediated atypical hemolytic uremic syndrome

Complement-mediated aHUS is a rare disease that mainly occurs due to gene mutations of complement factors. There have been identified loss-of-function mutations in regulatory proteins of the alternative pathway of the complement system (factor H, factor I, and membrane cofactor protein [MCP]) or gain-of-function mutations in activators (C3 and factor B) (26). Furthermore, complement-mediated aHUS may be the result of autoimmune processes, through the production of auto-antibodies against complement regulatory proteins (26).

Mutations affecting the protein factors H, I, B, and C3 are associated with a high risk of recurrence (75%), and an even higher risk of graft failure in the first posttransplant year (over 90%), as these are circulating proteins that persist in recipients (27). In contrast, mutations that affect MCP are associated with a low degree of recurrence (approximately 20%) and better survival of the graft since the transplanted kidney expresses the normal protein (28).

TPE is capable of removing auto-antibodies against complement proteins and mutant circulating complement regulatory proteins, replacing them with no defective components; but, its use in the prevention of recurrence, before transplantation, and in the case of recurrence of complement-mediated aHUS posttransplantation has shown poor therapeutic results (29).

The introduction of eculizumab, an anti-C5 monoclonal antibody, positively changed the outcomes and questioned the role of TPE in the treatment of aHUS. The alleged clinical benefits of TPE in prophylactic protocols before transplantation, in addition to eculizumab used by some transplant centers (30), remain unclear and controversial. TPE remains a therapeutic alternative if eculizumab is not available for patients with autoantibodies against factor H and in the case of persistent thrombocytopenia after the initial days of administration of eculizumab (31,32). Attention should be made in case of a TPE session after eculizumab infusion because of increased drug clearance TPE induced. Therefore, an extra dose of eculizumab within 60 min after a TPE is recommended (33).

De novo thrombotic microangiopathy

The occurrence of de novo thrombotic microangiopathy (TMA) posttransplantation could be the result of pathogenetic processes that induce TMA in the general population. However, the most common causes that induce TMA after KT are: i) drug-induced TMA due to calcineurin inhibitors and mammalian target of rapamycin (mTOR) inhibitors, ii) ischemia-reperfusion injury, iii) AMR, and iv) viral infections (34).

If clinical improvement does not occur after changing the immunosuppressive regimen, in the case of drug-induced TMA, or after treating the underlying infection, TPE may find application in an attempt to stop further damage to the graft even if the levels of evidence are low for such use (35). If eculizumab is available, it becomes the treatment of choice in these cases (36,37).

When TMA is associated with AMR, the combined use of TPE and intravenous immunoglobulin (IVIG) confer favorable outcomes for the survival of the graft (38). Eculizumab becomes the preferred treatment in AMR-associated TMA if hemolysis persists in spite of the use of TPE, and in the TPE-dependent patients (34).

Recurrence of C3 glomerulopathies

C3 glomerulopathies (C3G) are rare forms of glomerulonephritis that include dense deposit disease (DDD) and C3 glomerulonephritis (C3GN), distinguished by structural characteristics observed on electron microscopy (39). Both subtypes are caused by excessive activation of the alternative complement pathway, which results either from C3 convertase-stabilizing autoantibodies, called C3 nephritic factors (C3NeFs), and other antibodies directed against factor B and factor H or from mutations of factor I, MCP, C3, factor B, factor H, and mutations of the complement factor H related proteins (CFHRs) gene cluster (40).

The recurrence rate of DDD after KT is approximately 80–100%, while the reported recurrence rate of C3GN is greater than 50% (41,42). Factors associated with an increased risk of recurrence include persistently low C3 levels and living-donor KT (43). Recurrent C3G is typically manifested within 1–2 years following KT and often results in graft loss (50% of patients) (41,42). Recurrence occurs earlier and is more aggressive if associated with monoclonal gammopathy (42).

Currently, there are no controlled studies on which to base therapeutic recommendations for recurrent C3G. We suggest that patients should be switched to mycophenolate mofetil (MMF) therapy, if it is not already part of their maintenance immunosuppression regimen.

TPE represents a therapy of unclear benefit. TPE has been proved to prevent disease progression in some patients with DDD in native kidneys with circulating C3NeFs, probably by removal of the pathologic autoantibodies (44). However, McCaughan et al. (45) reported that TPE was ineffective in a patient with recurrent DDD after KT, in spite of effective removal of C3NeF. Recently, Kumar et al. (46) demonstrated response to TPE in three of four patients with early recurrent C3G (median time for posttransplant recurrence was 3 days) and circulating autoantibodies to complement pathway regulators (two cases had positive C3NeF and one had anti-complement factor-H autoantibodies). All patients underwent five sessions of alternate-day TPE (40 ml/kg/session with membrane filter) using fresh-frozen plasma (FFP) and albumin as the replacement fluid (46). If TPE is used in such patients, we suggest alternate-day TPE, at least initially. Then, TPE should be maintained until C3NeF levels decrease by at least 50%, provided that there is simultaneous stabilization of graft function.

Among patients who have a known serum factor deficiency, as an example, a genetic cause of factor H deficiency, we suggest to begin intermittent FFP infusions or TPE before KT and extend this treatment after transplantation while monitoring for clinical evidence of disease recurrence. If such patients remain free of recurrence, the FFP infusions or TPE may be tapered off with continued monitoring for signs of recurrence.

Eculizumab is an alternative to TPE, which has been used with variable success in patients with recurrent C3G (47,48). Avacopan, an orally administered selective C5a receptor inhibitor, which was recently found to be effective in the treatment of ANCA-associated vasculitis (49), is now tested in a randomized, double-blind, placebo-controlled, phase 2 study (ClinicalTrials.gov Identifier: NCT03301467), in biopsy-proven C3G, either DDD or C3GN, with or without a renal transplant.

Antiphospholipid antibody syndrome and lupus nephritis

Antiphospholipid antibody syndrome is a multisystem autoimmune disease clinically manifested by multiple thrombotic episodes in both venous and arterial circulation, and serologically characterized by the presence of antiphospholipid (aPL) antibodies. APS can represent a primary nosological entity or be secondary to other autoimmune diseases, mainly systemic lupus erythematosus (SLE) (50).

The kidney can be damaged by the occlusion of the glomerular capillaries up to that of the main artery and vein (50). Graft loss due to a thrombotic event of graft vein, graft artery, or even a TMA in the early postoperative period represents the most frequent causes of KT failure in patients with Antiphospholipid syndrome (APS) (51). In addition, several studies have highlighted the fact that hemodialysis patients, and consequently the recipients of KT, have a high prevalence of circulating aPL, potentially harmful to the graft (52,53).

Treatment of KT recipients affected by APS with long-term warfarin, for the prevention of thrombotic events, is strongly recommended. In addition, many transplant centers prefer to treat, in the peritransplant period, all patients with aPL positivity and history of thrombotic events (54,55). However, the treatment of these patients with oral anticoagulants increases the risk of bleeding, which could result in an early loss of graft in the immediate posttransplant period. Furthermore, it must be considered that thrombotic events can occur in 40% of cases even during anticoagulant therapy (56).

Prophylactic treatment with TPE for the removal of circulating APL, in addition to maximal oral anticoagulant therapy, before a living-donor KT has proven effective in a patient with primary APS (57) and in the case of a recipient with secondary APS due to SLE (58). However, in cases of catastrophic APS (CAPS), which occur with diffuse TMA (vascular occlusion affecting three or more organ systems) (59), prophylactic treatment with eculizumab to prevent recurrence after KT must be considered as the most suitable treatment after being successfully used in a KT recipient together with systemic anticoagulation and standard immunosuppression (60).

Barbour et al. (51) reported a case of acute recurrence of TMA in a recipient of KT affected by APS and lupus nephritis, which has been effectively treated with TPE even with the presence of moderate irreversible damage to the functionality of the graft. These data suggest that further studies are required.

Recurrent and de novo anti-glomerular basement membrane antibody disease

Anti-GBM antibody disease histologically recurs to the graft in up to 50% of cases if circulating anti-GBM antibodies persist at the time of transplantation (61,62). However, clinically manifested cases of recurrent anti-GBM antibody disease are limited, as most patients are asymptomatic (61). Different cohorts described that de novo anti-GBM antibody disease can occur in 0.5–10% of KT recipients with Alport syndrome, developing anti-GBM antibodies to a collagen component expressed by the graft [alpha5 (IV) NC1] that is missing in patients with Alport (6365). The explanation for the differences in the incidence may relate to different transplant immunosuppression protocols or different thresholds for diagnosis and coding of posttransplant anti-GBM antibody disease used in the various series.

The therapeutic approach is the same as that used in nontransplanted patients. TA must be used promptly for the removal of the harmful autoantibodies, in addition to corticosteroids plus cyclophosphamide or RTX, which are useful to inhibit further antibody production (62). AI and TPE have similar results (66,67).

The endopeptidase IdeS (Immunoglobulin G degrading enzyme of Streptococcus pyogenes), an enzyme that is capable of cleaving both circulating and membrane-bound human IgG subclasses into F(ab’)2 and Fc fragments, is a promising new therapeutic approach in the treatment of anti-GBM antibody disease. Soveri et al. (68) showed rapid clearance of anti-GBM antibodies (within minutes) in three no transplant patients with severe, and refractory to standard treatment, anti-GBM nephritis. Rebound of anti-GBM antibodies occurred in all three cases, even if it was mild in patients 1 and 3, and easily managed with TPE. Clinical trials are necessary to determine the clinical utility of this new treatment option. GOOD-IDES is an open-label ongoing phase II study to evaluate the efficacy and safety of IdeS in anti-GBM antibody disease (ClinicalTrials.gov identifier: NCT03157037).

Recurrence of antineutrophil cytoplasmic antibody-associated vasculitis

Recurrence of ANCA-associated vasculitis in KT recipients is rare. In a recent review of 11 studies that included a total of 441 KT recipients, the prevalence of recurrent ANCA-associated vasculitis was 10% (69). In case of relapse, the therapeutic options are the same as in nontransplanted patients. Both cyclophosphamide and RTX have shown their efficacy in the event of a posttransplant recurrence (70).

TPE is recommended in addition to corticosteroids and either cyclophosphamide or RTX in cases where recurrence occurs with alveolar hemorrhage, severe segmental necrotizing glomerulonephritis with serum creatinine above 4.0 mg/dL, and concurrent anti-GBM disease (7072). However, Walsh et al. (73), in the recently published results of the Plasma Exchange and Glucocorticoids for Treatment of ANCA-associated vasculitis (PEXIVAS) trial, did not show that the addition of plasma exchange (PLEX) to standard therapy conferred benefits in nontransplant patients with severe ANCA-associated vasculitis in terms of lower incidence of death or End-Stage Kidney Disease (ESKD). In the interim of no data in transplant patients, we recommend that PLEX be considered in any patient who is not responsive to conventional therapy.

The most promising advancement for remission induction therapy in severe organ- or life-threatening ANCA-associated vasculitis is avacopan. Recently, the CLEAR study, a phase 2, randomized, double-blind, placebo-controlled trial, met its primary endpoint, indicating that avacopan can replace high-dose corticosteroids efficiently and safely in patients with newly diagnosed or relapsing ANCA-associated vasculitis (49). The results of the ADVOCATE study, a phase 3 trial, will assess the safety and effectiveness of avacopan as an alternative to prednisone in inducing and maintaining remission in patients with ANCA-associated vasculitis; topline results are anticipated to be published by Q3 2020 (74).

Conclusion

The application of TA is currently used in several glomerular diseases after KT. However, strong evidence is scarce as the rarity of these conditions implies that a high standard of quality randomized clinical trials (RCTs) are missing. In addition, in the era of new and emerging biological immunosuppressive therapies with an increasing number of specific actions and immune targets directed against cell-surface antigens or plasma-soluble molecules, the use of TA as an adjunctive therapeutic option becomes even more challenging in the study of future therapeutic protocols, which will best address open issues for better clinical outcomes. Growing international collaboration is demanded to improve the quality of future studies in this area.

Conflicts of interest

The authors declare no potential conflicts of interest with respect to research, authorship, and/or publication of this article.

REFERENCES

1 Briganti EM, Russ GR, McNeil JJ, Atkins RC, Chadban SJ. Risk of renal allograft loss from recurrent glomerulonephritis. N Engl J Med. 2002;347(2):103–9. 10.1056/NEJMoa013036

2 Hariharan S, Adams MB, Brennan DC, Davis CL, First MR, Johnson CP, et al. Recurrent and de novo glomerular disease after renal transplantation: A report from Renal Allograft Disease Registry (RADR). Transplantation. 1999;68(5):635–41. 10.1097/00007890-199909150-00007

3 Artero M, Biava C, Amend W, Tomlanovich S, Vincenti F. Recurrent focal glomerulosclerosis: Natural history and response to therapy. Am J Med. 1992;92(4):375. 10.1016/02-9343(92)90267-F

4 Stephanian E, Matas AJ, Mauer SM, Chavers B, Nevins T, Kashtan C, et al. Recurrence of disease in patients retransplanted for focal segmental glomerulosclerosis. Transplantation. 1992;53(4):755–7. 10.1097/00007890-199204000-00009

5 Canaud G, Delville M, Legendre C. Recurrence of focal and segmental glomerulosclerosis after transplantation. Transplantation. 2016;100(2):284–7. 10.1097/TP.0000000000000902

6 Delville M, Sigdel TK, Wei C, Li J, Hsieh SC, Fornoni A, et al. A circulating antibody panel for pretransplant prediction of FSGS recurrence after kidney transplantation. Sci Transl Med. 2014;6(256):256ra136. 10.1126/scitranslmed.3008538

7 Dall’Amico R, Ghiggeri G, Carraro M, Artero M, Ghio L, Zamorani E, et al. Prediction and treatment of recurrent focal segmental glomerulosclerosis after renal transplantation in children. Am J Kidney Dis. 1999;34(6):1048–55. 10.1016/S0272-6386(99)70010-7

8 Cheong HI, Han HW, Park HW, Ha IS, Han KS, Lee HS, et al. Early recurrent nephritic syndrome after renal transplantation in children with focal segmental glomerulosclerosis. Nephrol Dial Transplant. 2000;15(1):78–81. 10.1093/ndt/15.1.78

9 Artero ML, Sharma R, Savin VJ, Vincenti F. Plasmapheresis reduces proteinuria and serum capacity to injure glomeruli in patients with recurrent focal glomerulosclerosis. Am J Kidney Dis. 1994;23(4):574–81. 10.1016/S0272-6386(12)80381-7

10 Matalon A, Markowitz GS, Joseph RE, Cohen DJ, Saal SD, Kaplan B, et al. Plasmapheresis treatment of recurrent FSGS in adult renal transplant recipients. Clin Nephrol. 2001;56(4):271–8.

11 Dantal J, Bigot E, Bogers W, Testa A, Kriaa F, Jacques Y, et al. Effect of plasma protein adsorption on protein excretion in kidney-transplant recipients with recurrent nephrotic syndrome. N Engl J Med. 1994;330(1):7–14. 10.1056/NEJM199401063300102

12 Ponticelli C. Recurrence of focal segmental glomerular sclerosis (FSGS) after renal transplantation. Nephrol Dial Transplant. 2010;25(1):25–31. 10.1093/ndt/gfp538

13 Kashgary A, Sontrop JM, Li L, Al-Jaishi AA, Habibullah ZN, Alsolaimani R, et al. The role of plasma exchange in treating post-transplant focal segmental glomerulosclerosis: A systematic review and meta-analysis of 77 case-reports and case-series. BMC Nephrol. 2016;17(1):104. 10.1186/s12882-016-0322-7

14 Vlachopanos G, Georgalis A, Gakiopoulou H. Plasma exchange for the recurrence of primary focal segmental glomerulosclerosis in adult renal transplant recipients: A meta-analysis. J Transplant. 2015;2015:639628. 10.1155/2015/639628

15 Rudnicki M. FSGS recurrence in adults after renal transplantation. Biomed Res Int. 2016;2016:3295618. 10.1155/2016/3295618

16 Canaud G, Zuber J, Sberro R, Royale V, Anglicheau D, Snanoudj R, et al. Intensive and prolonged treatment of focal and segmental glomerulosclerosis recurrence in adult kidney transplant recipients: A pilot study. Am J Transplant. 2009;9(5):1081–6. 10.1111/j.1600-6143.2009.02580.x

17 Cravedi P, Kopp JB, Remuzzi G. Recent progress in the pathophysiology and treatment of FSGS recurrence. Am J Transplant. 2013;13(2):266–74. 10.1111/ajt.12045

18 Araya CE, Dharnidharka VR. The factors that may predict response to rituximab therapy in recurrent focal segmental glomerulosclerosis: A systematic review. J Transplant. 2011;2011:374213. 10.1155/2011/374213

19 Fornoni A, Sageshima J, Wei C, Merscher-Gomez S, Aguillon-Prada R, Jauregui AN, et al. Rituximab targets podocytes in recurrent focal segmental glomerulosclerosis. Sci Transl Med. 2011;3:85ra46. 10.1126/scitranslmed.3002231

20 Reiser J, Fornoni A. Rituximab: A boot to protect the foot. J Am Soc Nephrol. 2014;25(4):647–8. 10.1681/ASN.2013121331

21 Tsagalis G, Psimenou E, Nakopoulou L, Laggouranis A. Combination treatment with plasmapheresis and rituximab for recurrent focal segmental glomerulosclerosis after renal transplantation. Artif Organs. 2011;35(4):420–5. 10.1111/j.1525-1594.2010.01068.x

22 Hristea D, Hadaya K, Marangon N, Buhler L, Villard J, Morel P, et al. Successful treatment of recurrent focal segmental glomerulosclerosis after kidney transplantation by plasmapheresis and rituximab. Transpl Int. 2007;20(1):102–5. 10.1111/j.1432-2277.2006.00395.x

23 Puisset F, White-Koning M, Kamar N, Huart A, Haberer F, Blasco H, et al. Population pharmacokinetics of rituximab with or without plasmapheresis in kidney patients with antibody-mediated disease. Br J Clin Pharmacol. 2013;76(5):734–40. 10.1111/bcp.12098

24 Martin-Moreno PL, Rifon J, Errasti P. Efficacy of the combination of immunoadsorption and rituximab for treatment in a case of severe focal and segmental glomerulosclerosis recurrence after renal transplantation. Blood Purif. 2018;46(2):90–3. 10.1159/000488638

25 Kronbichler A, Gauckler P, Lee KH, Shin JI, Malvezzi P, Mayer G. Immunoadsorption in nephrotic syndrome: Where are we now and where are we going from here? Atheroscler Suppl. 2019;40:55–60. 10.1016/j.atherosclerosissup.2019.08.027

26 Noris M, Remuzzi G. Atypical hemolytic-uremic syndrome. N Engl J Med. 2009;361(17):1676–87. 10.1056/NEJMra0902814

27 Bresin E, Daina E, Noris M, Castelletti F, Stefanov R, Hill P, et al. Outcome of renal transplantation in patients with non-Shiga toxin-associated hemolytic uremic syndrome: Prognostic significance of genetic background. Clin J Am Soc Nephrol. 2006;1(1):88–99. 10.2215/CJN.00050505

28 Sellier-Leclerc AL, Fremeaux-Bacchi V, Dragon-Durey MA, Macher MA, Niaudet P, Guest G, et al. Differential impact of complement mutations on clinical characteristics in atypical hemolytic uremic syndrome. J Am Soc Nephrol. 2007;18(8):2392–400. 10.1681/ASN.2006080811

29 Le Quintrec M, Zuber J, Moulin B, Kamar N, Jablonski M, Lionet A, et al. Complement genes strongly predict recurrence and graft outcome in adult renal transplant recipients with atypical hemolytic and uremic syndrome. Am J Transplant. 2013;13(3):663–75. 10.1111/ajt.12077

30 Nester C, Stewart Z, Myers D, Jetton J, Nair R, Reed A, et al. Pre-emptive eculizumab and plasmapheresis for renal transplant in atypical hemolytic uremic syndrome. Clin J Am Soc Nephrol. 2011;6(6):1488–94. 10.2215/CJN.10181110

31 Fakhouri F, Zuber J, Frémeaux-Bacchi V, Loirat C. Haemolytic uraemic syndrome [published correction appears in Lancet. 2017 Aug 12;390(10095):648]. Lancet. 2017;390(10095):681–96. 10.1016/S0140-6736(17)30062-4

32 Bambauer R, Latza R, Schiel R. Therapeutic apheresis in the treatment of hemolytic uremic syndrome in view of pathophysiological aspects. Ther Apher Dial. 2011;15(1):10–19. 10.1111/j.1744-9987.2010.00903.x

33 Wijnsma KL, Ter Heine R, Moes DJAR, Langemeijer S, Schols SEM, Volokhina EB, et al. Pharmacology, pharmacokinetics and pharmacodynamics of eculizumab, and possibilities for an individualized approach to eculizumab. Clin Pharmacokinet. 2019;58(7):859–74. 10.1007/s40262-019-00742-8

34 Garg N, Rennke HG, Pavlakis M, Zandi-Nejad K. De novo thrombotic microangiopathy after kidney transplantation. Transplant Rev (Orlando). 2018;32(1):58–68. 10.1016/j.trre.2017.10.001

35 Karthikeyan V, Parasuraman R, Shah V, Vera E, Venkat KK. Outcome of plasma exchange therapy in thrombotic microangiopathy after renal transplantation. Am J Transplant. 2003;3(10):1289–94. 10.1046/j.1600-6143.2003.00222.x

36 Epperla N, Hemauer K, Hamadani M, Friedman KD, Kreuziger LB. Impact of treatment and outcomes for patients with posttransplant drug-associated thrombotic microangiopathy. Transfusion. 2017;57(11):2775–81. 10.1111/trf.14263

37 Java A, Edwards A, Rossi A, Pandey R, Gaut J, Delos Santos R, et al. Cytomegalovirus-induced thrombotic microangiopathy after renal transplant successfully treated with eculizumab: Case report and review of the literature. Transpl Int. 2015;28(9):1121–5. 10.1111/tri.12582

38 Satoskar AA, Pelletier R, Adams P, Nadasdy GM, Brodsky S, Pesavento T, et al. De novo thrombotic microangiopathy in renal allograft biopsies-role of antibody-mediated rejection. Am J Transplant. 2010;10(8):1804–11. 10.1111/j.1600-6143.2010.03178.x

39 Schena FP, Esposito P, Rossini M. A narrative review on C3 glomerulopathy: A rare renal disease. Int J Mol Sci. 2020;21(2):E525. 10.3390/ijms21020525

40 Salvadori M, Bertoni E. Complement related kidney diseases: Recurrence after transplantation. World J Transplant. 2016;6(4):632–45. 10.5500/wjt.v6.i4.632

41 Braun MC, Stablein DM, Hamiwka LA, Bell L, Bartosh SM, Strife CF. Recurrence of membranoproliferative glomerulonephritis type II in renal allografts: The North American Pediatric Renal Transplant Cooperative Study experience. J Am Soc Nephrol. 2005;16(7):2225–33. 10.1681/ASN.2005020175

42 Zand L, Lorenz EC, Cosio FG, Fervenza FC, Nasr Samih H, Gandhi MJ, et al. Clinical findings, pathology, and outcomes of C3GN after kidney transplantation. J Am Soc Nephrol. 2014;25(5):1110–17. 10.1681/ASN.2013070715

43 Alasfar S, Carter-Monroe N, Rosenberg AZ, Montgomery RA, Alachkar N. Membranoproliferative glomerulonephritis recurrence after kidney transplantation: Using the new classification. BMC Nephrol. 2016;17:7. 10.1186/s12882-015-0219-x

44 Kurtz KA, Schlueter AJ. Management of membranoproliferative glomerulonephritis type II with plasmapheresis. J Clin Apher. 2002;17(3):135–7. 10.1002/jca.10026

45 McCaughan JA, O’Rourke DM, Courtney AE. Recurrent dense deposit disease after renal transplantation: An emerging role for complementary therapies. Am J Transplant. 2012;12(4):1046–51. 10.1111/j.1600-6143.2011.03923.x

46 Kumar A, Bharati J, Nada R, Singh S, Sharma A, Gupta KL, et al. Utility of plasma exchange in early recurrent C3 glomerulopathy. Indian J Transplant. 2019;13:122–6. 10.4103/ijot.ijot_78_18

47 Vivarelli M, Emma F. Treatment of C3 glomerulopathy with complement blockers. Semin Thromb Hemost. 2014;40(4):472–7. 10.1055/s-0034-1375299

48 Bomback AS, Smith RJ, Barile GR, Zhang Y, Heher EC, Herlitz L, et al. Eculizumab for dense deposit disease and C3 glomerulonephritis. Clin J Am Soc Nephrol. 2012;7(5):748–56. 10.2215/CJN.12901211

49 Jayne DRW, Bruchfeld AN, Harper L, Schaier M, Venning MC, Hamilton P, et al. Randomized trial of C5a receptor inhibitor Avacopan in ANCA-associated vasculitis. J Am Soc Nephrol. 2017;28(9):2756–67. 10.1681/ASN.2016111179

50 Sciascia S, Cuadrado MJ, Khamashta M, Roccatello D. Renal involvement in antiphospholipid syndrome. Nat Rev Nephrol. 2014;10(5):279–89. 10.1038/nrneph.2014.38

51 Barbour TD, Crosthwaite A, Chow K, Finlay MJ, Better N, Hughes PD, et al. Antiphospholipid syndrome in renal transplantation. Nephrology (Carlton). 2014;19(4):177–85. 10.1111/nep.12217

52 Phillips AO, Jones HW, Hambley H, Hillis AN, Hendry BM. Prevalence of lupus anticoagulant and anticardiolipin antibodies in haemodialysis patients. Nephron. 1993;65(3):350–3. 10.1159/000187511

53 Ducloux D, Pellet E, Fournier V, Rebibou JM, Bresson-Vautrin C, Racadot E, et al. Prevalence and clinical significance of antiphospholipid antibodies in renal transplant recipients. Transplantation. 1999;67(1):90–3. 10.1097/00007890-199901150-00015

54 Vaidya S, Sellers R, Kimball P, Shanahan T, Gitomer J, Gugliuzza K, et al. Frequency, potential risk and therapeutic intervention in end-stage renal disease patients with antiphospholipid antibody syndrome: A multicenter study. Transplantation. 2000;69(7):1348–52. 10.1097/00007890-200004150-00023

55 Forman JP, Lin J, Pascual M, Denton MD, Tolkoff-Rubin N. Significance of anticardiolipin antibodies on short and long term allograft survival and function following kidney transplantation. Am J Transplant. 2004;4(11):1786–91. 10.1046/j.1600-6143.2004.00602.x

56 Vaidya S, Gugliuzza K, Daller JA. Efficacy of anticoagulation therapy in end-stage renal disease patients with antiphospholipid antibody syndrome. Transplantation. 2004;77(7):1046–9. 10.1097/01.TP.0000119157.81765.46

57 Ruffatti A, Marson P, Valente M, Ciprian M, Tonello M, Marchini F, et al. Plasma exchange in a patient with primary antiphospholipid syndrome undergoing kidney transplantation. Transpl Int. 2007;20(5):475–7. 10.1111/j.1432-2277.2007.00454.x

58 Sofue T, Hayashida Y, Hara T, Kawakami K, Ueda N, Kushida Y, et al. Plasmapheresis in a patient with antiphospholipid syndrome before living-donor kidney transplantation: A case report. BMC Nephrol. 2014;15:167. 10.1186/1471-2369-15-167

59 Asherson RA, Cervera R, de Groot PG, Erkan D, Boffa MC, Piette JC, et al. Catastrophic Antiphospholipid Syndrome Registry Project Group. Catastrophic antiphospholipid syndrome: International consensus statement on classification criteria and treatment guidelines. Lupus. 2003;12(7):530–4. 10.1191/0961203303lu394oa

60 Lonze BE, Singer AL, Montgomery RA. Eculizumab and renal transplantation in a patient with CAPS. N Engl J Med. 2010;362(18):1744–5. 10.1056/NEJMc0910965

61 Kotanko P, Pusey CD, Levy JB. Recurrent glomerulonephritis following renal transplantation. Transplantation. 1997;63(8):1045–52. 10.1097/00007890-199704270-00001

62 McAdoo SP, Pusey CD. Anti-glomerular basement membrane disease. Clin J Am Soc Nephrol. 2017;12(7):1162–72. 10.2215/CJN.01380217

63 Göbel J, Olbricht CJ, Offner G, Helmchen U, Repp H, Koch KM, et al. Kidney transplantation in Alport’s syndrome: Long-term outcome and allograft anti-GBM nephritis. Clin Nephrol. 1992;38(6):299–304.

64 Kalluri R, Weber M, Netzer KO, Sun MJ, Neilson EG, Hudson BG. COL4A5 gene deletion and production of post-transplant anti-alpha 3(IV) collagen alloantibodies in Alport syndrome. Kidney Int. 1994;45(3):721–6. 10.1038/ki.1994.96

65 Mallett A, Tang W, Clayton PA, Stevenson S, McDonald SP, Hawley CM, et al. End-stage kidney disease due to Alport syndrome: Outcomes in 296 consecutive Australia and New Zealand Dialysis and Transplant Registry cases. Nephrol Dial Transplant. 2014;29(12):2277–86. 10.1093/ndt/gfu254

66 Biesenbach P, Kain R, Derfler K, Perkmann T, Soleiman A, Benharkou A, et al. Long-term outcome of anti-glomerular basement membrane antibody disease treated with immunoadsorption. PLoS One. 2014;9(7):e103568. 10.1371/journal.pone.0103568

67 Zhang YY, Tang Z, Chen DM, Gong DH, Ji DX, Liu ZH. Comparison of double filtration plasmapheresis with immunoadsorption therapy in patients with anti-glomerular basement membrane nephritis. BMC Nephrol. 2014;15:128. 10.1186/1471-2369-15-128

68 Soveri I, Mölne J, Uhlin F, Nilsson T, Kjellman C, Sonesson E, et al. The IgG-degrading enzyme of Streptococcus pyogenes causes rapid clearance of anti-glomerular basement membrane antibodies in patients with refractory anti-glomerular basement membrane disease. Kidney Int. 2019;96(5):1234–8. 10.1016/j.kint.2019.06.019

69 Moran S, Little MA. Renal transplantation in antineutrophil cytoplasmic antibody-associated vasculitis. Curr Opin Rheumatol. 2014;26(1):37–41. 10.1097/BOR.0000000000000006

70 Geetha D, Kant S. Renal transplantation in anti-neutrophil cytoplasmic antibody vasculitis. Expert Rev Clin Immunol. 2018;14(3):235–40. 10.1080/1744666X.2018.1440205

71 Walters G. Role of therapeutic plasmapheresis in ANCA-associated vasculitis. Pediatr Nephrol. 2016;31(2):217–25. 10.1007/s00467-014-3038-6

72 Apaydin S. The treatment of ANCA-associated rapidly-progressive glomerulonephritis and Goodpasture syndrome with therapeutic apheresis. Transfus Apher Sci. 2018;57(1):8–12. 10.1016/j.transci.2018.02.007

73 Walsh M, Merkel PA, Peh CA, Szpirt WM, Puèchal X, Fujimoto S, et al. Plasma exchange and glucocorticoids in severe ANCA-associated vasculitis. N Engl J Med. 2020;382(7):622–31. 10.1056/NEJMoa1803537

74 Merkel PA, Jayne DR, Wang C, Hillson J, Bekker P. Evaluation of the safety and efficacy of Avacopan, a C5a receptor inhibitor, in patients with antineutrophil cytoplasmic antibody-associated vasculitis treated concomitantly with rituximab or cyclophosphamide/azathioprine: Protocol for a randomized, double-blind, active-controlled, phase 3 trial. JMIR Res Protoc. 2020;9(4):e16664. 10.2196/16664