Your activity: 10150 p.v.
your limit has been reached. plz Donate us to allow your ip full access, Email: [email protected]

Kidney transplantation in adults: Prevention and treatment of antibody-mediated rejection of the renal allograft

Kidney transplantation in adults: Prevention and treatment of antibody-mediated rejection of the renal allograft
Authors:
Arjang Djamali, MD, MS, FASN
Daniel C Brennan, MD, FACP
Section Editor:
Christophe Legendre, MD
Deputy Editor:
Albert Q Lam, MD
Literature review current through: Feb 2022. | This topic last updated: Feb 28, 2020.

INTRODUCTION — Antibody-mediated rejection (ABMR) is the most common cause of allograft failure after kidney transplantation [1-4]. The revised Banff 2017 classification of ABMR defines active (previously called acute) and chronic active ABMR as conditions in which histologic evidence of acute and chronic injury is associated with evidence of current/recent antibody interaction with vascular endothelium and serologic evidence of donor-specific antibodies (DSA) to human leukocyte antigen (HLA) or non-HLA antigens [5].

The cellular and molecular pathways that regulate ABMR are still under investigation. However, evidence suggests that B cell and plasma cell activation results in the generation of DSAs, which bind to HLA or non-HLA molecules expressed on endothelial cells within the renal allograft [6,7]. In active ABMR, antibodies bind to graft endothelium and activate complement-dependent and -independent mechanisms that recruit natural killer (NK) cells, polymorphonuclear neutrophils, platelets, and macrophages, which contribute to peritubular capillaritis, glomerulitis, cellular necrosis, thrombotic microangiopathy, and a relatively rapid decline in allograft function [6-8].

Chronic ABMR, on the other hand, is a distinct pathophysiological process resulting from a repetitive pattern of thrombotic events and inflammatory changes that lead to endothelial cell injury and allograft matrix remodeling [9,10]. It manifests histologically as transplant glomerulopathy and results in a slow and progressive decline in kidney function [11].

Increasing evidence suggests that the prevention and treatment of antibody-mediated injury requires a combination of strategies to inhibit B cell development, maturation, and activity. Despite a relatively large number of observational studies, it is not clear which combination therapy is the safest and most effective.

The prevention and treatment of active and chronic ABMR of the renal allograft will be reviewed here. The clinical features and diagnosis of ABMR and the treatment of acute T cell-mediated (cellular) rejection (TCMR) are discussed separately:

(See "Kidney transplantation in adults: Clinical features and diagnosis of acute renal allograft rejection".)

(See "Kidney transplantation in adults: Treatment of acute T cell-mediated (cellular) rejection of the renal allograft".)

PREDICTORS OF OUTCOME — Active and chronic ABMR are both associated with poor outcomes after kidney transplantation. Patients with active ABMR are at increased risk for subsequent rejection, chronic ABMR, and graft loss [1,12-15]. Similarly, those with chronic ABMR have a higher risk for graft loss and patient death [1-3,16,17]. However, not all patients with ABMR have poor outcomes, and many patients maintain stable allograft function for years after treatment of the initial episode of rejection. Risk factors for graft loss in patients with ABMR are discussed below.

Histologic features — Some histopathological features on the kidney biopsy at the time of rejection are associated with worse outcomes. As an example, concurrent, acute T cell-mediated (cellular) rejection (TCMR) is an independent risk factor for graft failure in patients with ABMR [18]. There is also a clear and independent association between microvascular injury and C4d staining (including focal C4d staining in post-reperfusion biopsies) with poor outcomes in active or chronic ABMR [19-22]. Similarly, transplant glomerulopathy and the degree of chronic injury (measured semiquantitatively by adding chronic interstitial, tubular, vascular, and glomerular Banff scores) are associated with worse graft survival [16,23-25].

Donor-specific antibodies — Certain characteristics of donor-specific antibodies (DSAs) have been associated with poor outcomes among patients with ABMR. As examples:

DSA strength – In a study of 402 consecutive, deceased-donor kidney transplant recipients, the risk for both ABMR and graft loss directly correlated with peak preexisting anti-human leukocyte antigen (HLA) DSA strength, as measured by mean fluorescence intensity (MFI) [26]. Patients with a peak anti-HLA DSA MFI of >6000 had a more than 100-fold higher risk for developing ABMR compared with those with an MFI of <465. Graft survival in patients with a peak anti-HLA DSA MFI of >3000 was lower than that of patients with an MFI of ≤3000. However, in the absence of biopsy-proven rejection and acute inflammation, HLA DSA may not be associated with an increased risk of graft failure [27].

DSA subclass – The immunoglobulin subclass of DSA may also predict outcomes. In a study of 125 patients with DSAs detected in the first year posttransplant, immunoglobulin G4 (IgG4) immunodominant DSA was associated with later allograft injury, increased allograft glomerulopathy, and interstitial fibrosis/tubular atrophy (IF/TA) [28]. By contrast, IgG3 immunodominant DSA was associated with a shorter time to rejection, increased microvascular injury, C4d capillary deposition, and graft failure. These findings suggest that IgG immunodominant DSA subclasses may identify distinct phenotypes of renal allograft antibody-mediated injury.

Complement-binding capacity – The ability of anti-HLA DSAs to bind complement, as determined by the C1q assay, may identify patients at high risk for renal allograft loss [29]. However, one study showed that the C1q-binding activity of DSAs largely reflects differences in antibody strength, questioning the biologic significance of the C1q assay [30].

DSA type – The type of DSA (preexisting or de novo) may also be a predictor of worse outcomes in patients with ABMR. ABMR in patients with a de novo DSA, which is thought to be mostly related to medication nonadherence or inadequate immunosuppression, has been associated with poorer outcomes compared with ABMR in patients with preexisting DSA (ie, presensitized patients) [1,25,31-33].

DSA response to treatment – There is accumulating evidence indicating that a decline in DSA strength after treatment is associated with better graft survival [25,34,35]. However, there is limited information on the definition of a validated positive DSA response.

Graft function — The degree of renal allograft dysfunction at the time of kidney biopsy appears to be directly associated with poor outcomes in patients with ABMR [16,22,31,36]. As an example, in a retrospective analysis of 205 patients with biopsy-proven ABMR, an estimated glomerular filtration rate (eGFR) of <30 mL/min per 1.73 m2 at diagnosis and a urine protein-to-creatinine ratio of ≥0.30 g/g at the time of biopsy were identified as independent determinants of allograft loss (hazard ratio [HR] 3.27 and 2.44, respectively) [31]. In another study of 123 patients with chronic ABMR, a serum creatinine of >3 mg/dL and urine protein-to-creatinine ratio of >1 g/g at the time of diagnosis were independently associated with graft loss [16]. Other observational studies have reported similar findings among patients with active and chronic ABMR [22,36].

Other risk factors — The expression of endothelial cell-associated transcripts (ENDATs) and DSA-selective transcripts has been shown to be a biomarker of active antibody-mediated injury and may predict worse graft outcomes [23,37]. These molecular assays, which are not yet in mainstream clinical practice, reflect changes in microcirculatory endothelium not normally detected by routine histopathology and DSA testing and may improve risk stratification and prognostication in patients with ABMR.

Prediction models — Novel prediction models are being developed to predict long-term kidney allograft failure, including after the treatment of rejection [38]. Using an international cohort study including 7557 kidney transplant recipients from 10 academic medical centers from Europe and the United States, 32 candidate prognostic factors for kidney allograft survival were assessed. Of these, eight functional, histologic, and immunological prognostic factors were independently associated with allograft failure and were then combined into a risk prediction score (iBox). This score showed accurate calibration and discrimination (C index 0.81, 95% CI 0.79-0.83). The iBox system showed accuracy when assessed at different times of evaluation posttransplant and was validated in different clinical scenarios including response to rejection therapy, suggesting that the iBox risk prediction score may help to guide monitoring of patients and further improve the design and development of a valid and early surrogate endpoint for clinical trials [38].

PREVENTION — Our approach to the prevention of ABMR depends upon the detection of donor-specific antibody (DSA) prior to (preexisting DSA) or after (de novo DSA) transplant.

Patients with preexisting DSA before transplant — Patients with a preexisting DSA prior to transplant have a greater risk for ABMR and graft loss compared with nonsensitized patients [39-41]. This risk is proportional to the strength of DSA as patients with a positive complement-dependent cytotoxicity (CDC) crossmatch have a higher risk of ABMR and graft loss than those with a positive flow crossmatch, who in turn have a higher risk than patients with a positive virtual crossmatch [39,40,42].

Although a common approach to prevent ABMR has been to avoid transplanting highly sensitized patients, this option renders chronic dialysis the only therapeutic option, with significant implications for patient health and quality of life and health care costs. Long-term survival in kidney transplant recipients has improved considerably with desensitization [43,44], which can be used to reduce the level of DSAs pretransplant. In addition, the enrollment of patients in special programs to optimize matching can lead to timely transplants with better outcomes. (See "Kidney transplantation in adults: HLA desensitization" and "Kidney transplantation in adults: Living unrelated donors", section on 'Kidney paired donation'.)

Our general approach to the prevention of ABMR in patients with a preexisting DSA prior to transplant is as follows:

In patients with a potential living donor, the approach depends upon the results of the most recent crossmatch:

In patients with a positive CDC crossmatch or a strongly positive flow crossmatch, we, and many other transplant centers, prefer to use kidney paired donation (KPD) programs, rather than desensitization, given the high risk of ABMR and graft loss in such patients [45-47]. Such KPD programs (including the National Kidney Registry, the Alliance for Paired Donation, and the United Network for Organ Sharing [UNOS] Kidney Paired Donations Pilot Program) enable sensitized patients with immunologically incompatible living donors to be transplanted with high-quality grafts from other living donors in similar situations who are willing to exchange organs. Although cost has been a concern for kidney exchange registries in the United States, KPD could help participating centers to avoid complex desensitization protocols while improving long-term outcomes. Mathematical modeling has predicted that an optimized matching algorithm and a national KPD program would improve graft outcomes and reduce health care costs for highly sensitized patients [48]. Some transplant centers combine desensitization and KPD.

In patients with a positive virtual crossmatch or a mild-to-moderate flow crossmatch (ie, median channel shift of <250), we employ human leukocyte antigen (HLA) desensitization strategies, which include treatment with plasmapheresis, rabbit antithymocyte globulin (rATG)-Thymoglobulin, and rituximab [20,42]. This is discussed in more detail elsewhere. (See "Kidney transplantation in adults: HLA desensitization".)

In patients without a potential living donor, we employ HLA desensitization strategies. (See "Kidney transplantation in adults: HLA desensitization".)

In all patients with a preexisting DSA before transplant who undergo kidney transplantation, we use induction and maintenance immunosuppression therapies that are appropriate for patients at high risk for the development of acute rejection [49]. The selection of induction and maintenance immunosuppression in high-risk kidney transplant recipients is discussed elsewhere. (See "Kidney transplantation in adults: Maintenance immunosuppressive therapy", section on 'Initial maintenance immunosuppression in high-risk patients' and "Kidney transplantation in adults: Induction immunosuppressive therapy", section on 'Patients at high risk of rejection'.)

Monitoring after transplant — The monitoring of renal allograft function in patients with a preexisting DSA before transplant is similar to that performed in nonsensitized patients (see "Kidney transplantation in adults: Overview of care of the adult kidney transplant recipient", section on 'Monitoring kidney allograft function'). In addition, we routinely monitor DSA levels at months 1, 3, 6, and 12 posttransplant and then annually [50]. In patients with a significant rise in DSA or who develop a de novo DSA within the first three months, we perform a renal allograft biopsy. This practice is largely consistent with the recommendations of the Consensus Guidelines on the Testing and Clinical Management Issues Associated with HLA and Non-HLA Antibodies in Transplantation [50]. Patients with a pretransplant DSA undergo protocol kidney biopsies at months 3 and 12 posttransplant.

Some UpToDate contributors to this topic perform a post-reperfusion renal allograft biopsy at the time of transplantation to identify patients at risk for ABMR [20]. In patients who are found to have evidence of positive C4d staining, plasmapheresis (two to three sessions) and a single dose of rituximab 375 mg/m2 (administered after the last session of pheresis) would be added to the induction immunosuppression regimen [20].

Patients with de novo DSA after transplant — Kidney transplant recipients who develop a de novo DSA after transplantation can present with late-onset ABMR. As mentioned above, ABMR in patients with a de novo DSA has been associated with poorer outcomes compared with ABMR in patients with preexisting DSA (see 'Donor-specific antibodies' above). The two most common causes of ABMR due to a de novo DSA are medication nonadherence and inadequate immunosuppression, the latter of which is frequently attributed to the use of minimization strategies. In addition, acute T cell-mediated (cellular) rejection (TCMR), malignancy, and opportunistic infections (such as BK polyomavirus and cytomegalovirus [CMV] infection) that require a reduction in immunosuppression may also influence the development of late-onset ABMR [1,2,51,52].

Prevention of ABMR should focus on addressing nonadherence and under-immunosuppression while balancing the safety and efficacy of long-term immunosuppression. We maintain the majority of our patients on a triple therapy immunosuppression regimen (tacrolimus, mycophenolate, and prednisone) and monitor whole blood tacrolimus levels monthly in the first three years posttransplant and every three months thereafter. In patients who do not tolerate tacrolimus, we switch to belatacept, rather than sirolimus or everolimus. We monitor DSA annually and perform renal allograft biopsies in all patients who develop a de novo DSA.

(See "Kidney transplantation in adults: Maintenance immunosuppressive therapy", section on 'Patients who are not compliant with therapy'.)

(See "Psychiatric aspects of organ transplantation", section on 'Nonadherence'.)

(See "Kidney transplantation in adults: Maintenance immunosuppressive therapy", section on 'Patients who develop toxicity from calcineurin inhibitors'.)

An analysis of two randomized trials showed that the conversion of cyclosporine to everolimus at 3 to 4.5 months after transplant was associated with significantly higher rates of de novo DSA (10.8 versus 23 percent) and ABMR (3 versus 13 percent) [53]. By contrast, treatment with belatacept, a selective costimulation blocker that targets the CD80/CD86-CD28 interaction to prevent T cell activation, was associated with a low rate of de novo DSA over seven years of treatment in phase III trials, although this was not an initial endpoint of those studies [54]. Although there has been no head-to-head comparison between tacrolimus and belatacept, these data suggest that costimulation blockade may be safe and effective in preventing de novo DSA and late ABMR. Similarly, glucocorticoid withdrawal or avoidance may not increase the risk of de novo DSA if adequate immunosuppression is otherwise maintained, although this is difficult to standardize. In a five-year, longitudinal study, 37 kidney transplant recipients were randomly assigned to chronic glucocorticoid therapy or early glucocorticoid withdrawal at day 7 posttransplant; all patients received rATG-Thymoglobulin for induction and tacrolimus and mycophenolate as maintenance immunosuppression [55]. Only one patient in the chronic glucocorticoid treatment arm and none in the glucocorticoid withdrawal arm developed a de novo DSA.

TREATMENT OF ACTIVE ANTIBODY-MEDIATED REJECTION — The primary goal of treating ABMR is to remove existing donor-specific antibodies (DSAs) and to eradicate the clonal population of B cells or plasma cells that is responsible for their production. In general, we treat all patients who have evidence of active ABMR on biopsy. Although we do not routinely perform surveillance or protocol allograft biopsies at our institution, we would treat patients who are discovered to have subclinical ABMR by surveillance biopsy. In addition, we treat patients with C4d-negative ABMR with the same approach that we use in patients with C4d-positive ABMR.

(See 'Patients with subclinical rejection' below.)

(See "Kidney transplantation in adults: Clinical features and diagnosis of acute renal allograft rejection", section on 'Subclinical rejection'.)

(See "Kidney transplantation in adults: Clinical features and diagnosis of acute renal allograft rejection", section on 'C4d-negative antibody-mediated rejection'.)

(See 'Patients with C4d-negative ABMR' below.)

The optimal treatment of active ABMR is unclear, and there have been no randomized, controlled trials with adequate statistical power to compare the safety and efficacy of different therapeutic strategies [56]. Our recommendations for the treatment of ABMR are primarily based upon available, low-quality evidence and are largely consistent with the 2009 Kidney Disease: Improving Global Outcomes (KDIGO) clinical practice guidelines and the 2019 Transplantation Society Working Group Expert Consensus [57,58].

Approach to initial therapy — Although patients with active ABMR could possibly be treated on an outpatient basis, we typically advocate inpatient admission for patients because of the complexity of the treatment regimen. Our approach to the initial treatment of active ABMR depends upon the timing of the diagnosis of ABMR (algorithm 1):

In patients who are diagnosed with active ABMR within the first year posttransplant, we treat with a combination of glucocorticoids, plasmapheresis and intravenous (IV) immune globulin (IVIG), and, in some patients, rituximab as follows:

We give IV methylprednisolone at a dose of 300 to 500 mg daily for three to five days, followed by a rapid oral prednisone taper to the patient's previous maintenance dose of prednisone. If there are no concerns for nonadherence, we augment the maintenance prednisone dose. As an example, if the rejection occurred while the patient was taking 5 mg/day, we would increase the maintenance prednisone to 7.5 to 10 mg/day.

Plasmapheresis is performed daily or every other day for a maximum of six sessions or until the serum creatinine is within 20 to 30 percent of the baseline. The initial treatment is typically a one-and-one-half-volume exchange with albumin, and subsequent treatments are a one-volume exchange with albumin. We prefer an every-other-day plasmapheresis schedule as albumin alone can often be administered for replacement with interval recovery of the prothrombin time (PT), partial thromboplastin time (PTT), and fibrinogen to acceptable levels without the need to administer fresh frozen plasma. This avoids the risk of antigen sensitization; however, one to two units of fresh frozen plasma may be used for replacement at the end of a plasmapheresis treatment if indicated by preprocedure laboratory values or in the appropriate clinical setting, such as a same-day renal allograft biopsy. (See "Therapeutic apheresis (plasma exchange or cytapheresis): Indications and technology".)

We administer IVIG at a dose of 100 mg/kg after each session of plasmapheresis. We typically give 500 mg/kg per day for one to two days after the final session of plasmapheresis, with a total cumulative target dose of at least 1000 mg/kg of IVIG. In obese patients, some centers determine the IVIG dose based upon the patient's ideal body weight (calculator 1). Sucrose-free IVIG solutions that are no more than 5 percent concentrated are preferred to decrease the risk of acute kidney injury. (See "Overview of intravenous immune globulin (IVIG) therapy" and "Intravenous immune globulin: Adverse effects", section on 'Acute kidney injury'.)

In patients with evidence of microvascular inflammation on biopsy (ie, Banff glomerulitis score [g] + peritubular capillary score [ptc] >0), we administer rituximab as a single dose of 200 to 375 mg/m2 after completion of plasmapheresis and IVIG [59].

In addition, we augment other components of maintenance immunosuppression as needed. This is discussed elsewhere. (See "Kidney transplantation in adults: Maintenance immunosuppressive therapy", section on 'Patients with acute rejection'.)

In patients who are diagnosed with active ABMR after the first year posttransplant, we treat with glucocorticoids using the same approach as described above in patients diagnosed with ABMR within the first year posttransplant. However, we do not perform plasmapheresis in such patients because of the lack of evidence supporting the safety and efficacy of plasmapheresis in late-onset ABMR. We administer IVIG at a dose of 200 mg/kg every two weeks for three doses. In obese patients, some centers determine the IVIG dose based upon the patient's ideal body weight (calculator 1). Sucrose-free IVIG solutions that are no more than 5 percent concentrated are preferred to decrease the risk of acute kidney injury. (See "Overview of intravenous immune globulin (IVIG) therapy" and "Intravenous immune globulin: Adverse effects", section on 'Acute kidney injury'.)

In patients with evidence of microvascular inflammation on biopsy, we administer rituximab as a single dose of 375 mg/m2 after completion of IVIG. We also augment maintenance immunosuppression. (See "Kidney transplantation in adults: Maintenance immunosuppressive therapy", section on 'Patients with acute rejection'.)

In all patients who are treated for active ABMR, we recommence antimicrobial and antiviral prophylaxis with a regimen that is identical to that administered in the immediate posttransplant period. This includes prophylaxis against Pneumocystis pneumonia (PCP), cytomegalovirus (CMV) infection and disease, and herpes simplex infection (in patients who are at low CMV risk) for three months. In addition, we also administer antifungal prophylaxis and a prophylactic histamine-2 (H2) blocker for prevention of peptic ulcer disease, although this practice may vary by transplant center. A detailed discussion of the different prophylactic regimens is presented separately.

(See "Prophylaxis of infections in solid organ transplantation", section on 'Pneumocystis pneumonia'.)

(See "Kidney transplantation in adults: Prevention of cytomegalovirus disease in kidney transplant recipients", section on 'CMV prophylaxis'.)

(See "Prophylaxis of infections in solid organ transplantation", section on 'Antifungal prophylaxis'.)

We do not routinely use immunoadsorption, bortezomib, tocilizumab, eculizumab, or splenectomy in the initial treatment of patients with ABMR. However, some of these therapies can be considered in patients who do not respond to initial treatment. (See 'Second-line agents in patients who have failed initial therapy' below and 'Less frequently used therapies' below.)

As mentioned above, there is a lack of high-quality evidence in the form of large, randomized, controlled trials to guide the optimal therapy of patients with active ABMR. The best data come from the following studies:

A systematic review that included five randomized and seven nonrandomized, controlled trials evaluated the effects of different treatments on graft survival among kidney transplant recipients with active ABMR [60]. All of the randomized trials were small (median of 13 patients per treatment arm), and most were conducted using outdated diagnostic criteria for ABMR. Four randomized, controlled trials assessed the benefit of plasmapheresis; one study reported a benefit, one suggested potential harm, and two showed no effect. However, the plasmapheresis regimen differed in dose, frequency, and treatment interval among the studies, and IVIG was not administered. One randomized, controlled trial found a benefit with the use of protein immunoadsorption (see 'Immunoadsorption' below). The nonrandomized, controlled studies suggested a potential benefit from treatment with rituximab, plasmapheresis, and bortezomib; however, because some studies used a combination of these therapies, the effect of individual therapies could not be distinguished.

A phase III, multicenter, randomized, placebo-controlled trial (Impact of Treatment With Rituximab on the Progression of Humoral Acute Rejection After Renal Transplantation [RITUX ERAH]) examined the effect of rituximab among 38 kidney transplant recipients with biopsy-proven, active ABMR [61]. Patients were randomly assigned to rituximab (375 mg/m2) or placebo at day 5 of treatment; all patients were treated with plasmapheresis, IVIG, and glucocorticoids. There was no difference between the two groups in the frequency of the primary endpoint, defined as a composite measure of graft loss or absence of improvement in kidney function at day 12 (52.6 and 57.9 percent of patients in the rituximab and placebo groups, respectively). Both groups showed an improvement in histologic features of ABMR and Banff scores at one and six months, with a trend in favor of the rituximab group. Although both groups showed a decrease in DSA intensity as early as day 12, there was no difference between the groups at 12 months. However, it should be noted that this study was underpowered, and significant differences between the groups may have been missed.

The benefit of a combination therapy including plasmapheresis, IVIG, and rituximab was suggested by an observational study from France that compared the efficacy of plasmapheresis/IVIG/rituximab versus high-dose IVIG alone in the treatment of ABMR [62]. Graft survival at 36 months was 92 percent among patients treated with plasmapheresis/IVIG/rituximab versus 50 percent of those treated with IVIG alone. At three months posttreatment, DSAs were significantly lower in the plasmapheresis/IVIG/rituximab group. Another study confirmed that patients with active clinical or subclinical ABMR in the first year posttransplant have better outcomes if treated with a combination strategy including plasmapheresis [63].

Monitoring the response to therapy — Our approach to monitoring patients during therapy for active ABMR depends upon whether the patient is hospitalized for treatment or treated as an outpatient. As mentioned above, patients with active ABMR can be treated on an outpatient basis, but we typically advocate inpatient admission for patients given the complexity of the treatment regimen.

In patients who are treated in the outpatient setting, we monitor serum creatinine, electrolytes, and a complete blood count prior to each plasmapheresis session or on a weekly basis for four weeks if plasmapheresis is not performed. Patients who receive plasmapheresis are seen and evaluated during plasmapheresis sessions and then in the clinic at four weeks from the start of therapy; those who do not receive plasmapheresis undergo weekly labs for one month and monthly thereafter. All patients are seen for follow-up in the clinic at three months from the start of treatment, at which time we measure a DSA level and repeat a renal allograft biopsy if a biopsy has not been performed earlier in the course of treatment [59].

Data on the reversal of ABMR are limited [4,59,63]. In general, one-year graft survival after treatment of clinical and subclinical ABMR is approximately 80 and 95 percent, respectively [63]. Patients are considered to have a successful reversal of ABMR if they meet all of the following parameters within three months of treatment:

Decrease in serum creatinine to within 20 to 30 percent of the baseline level

Decrease in proteinuria to the baseline level

Decrease in immunodominant DSA by >50 percent

Resolution of changes associated with ABMR on repeat kidney biopsy (see "Kidney transplantation in adults: Clinical features and diagnosis of acute renal allograft rejection", section on 'Histologic findings')

Most patients with ABMR with a successful response to anti-rejection therapy will demonstrate an improvement in serum creatinine within seven days of treatment. Our subsequent approach to treatment is based upon the response to initial therapy:

In patients with a decrease in serum creatinine in response to therapy, we increase the maintenance tacrolimus dose to achieve a trough level 20 to 25 percent above the level at the time of rejection and resume routine monitoring of allograft function. For patients who are taking the immediate-release formulation of tacrolimus and cannot tolerate higher doses, extended-release tacrolimus, which has fewer side effects and may allow for a higher and therapeutic trough to be obtained, is an alternative option [64]. Typically, maintenance immunosuppression is also augmented by increasing the daily dose of oral prednisone. (See "Kidney transplantation in adults: Overview of care of the adult kidney transplant recipient", section on 'Monitoring kidney allograft function' and "Pharmacology of cyclosporine and tacrolimus", section on 'Switching formulations'.)

Patients without any decrease in serum creatinine after seven days of rejection treatment are considered to have failed initial treatment. In such patients, ongoing rejection and/or another cause of renal allograft dysfunction should be suspected, and a repeat renal allograft biopsy should be performed. Our subsequent approach depends upon the histopathological and clinical findings of the patient. If the biopsy reveals no evidence of an acute, reversible process or reveals extensive fibrosis (indicating nonviable kidney tissue), we typically discontinue treatment of acute rejection. If the biopsy demonstrates evidence of persistent active ABMR, second-line agents for the treatment of ABMR can be used as rescue therapy (see 'Second-line agents in patients who have failed initial therapy' below and 'Less frequently used therapies' below). However, the intensity of additional therapy with plasmapheresis and other agents such as bortezomib and anti-complement therapy should be weighed against preexisting comorbidities and the risk of infectious and malignant complications.

Second-line agents in patients who have failed initial therapy — Most patients with active ABMR will respond to a combination of glucocorticoids, plasmapheresis, IVIG, and rituximab. However, in patients who do not respond to initial treatment with this combination, the following agents can be considered as rescue therapy.

Bortezomib — Bortezomib is a potent, reversible proteasome inhibitor that has been approved by the US Food and Drug Administration (FDA) as first-line therapy for multiple myeloma since 2008. Bortezomib reduces intracellular protein degradation by inhibiting proteasomal activity and results in apoptosis, mainly via inhibition of nuclear factor kappa-B (NFkB)-induced survival signals. The drug is particularly effective against differentiated plasma cells because of the high rate of protein synthesis in these cells [65,66]. (See "Multiple myeloma: Selection of initial chemotherapy for symptomatic disease".)

Several case reports/series have demonstrated the effectiveness of bortezomib in treating ABMR, successfully reversing acute rejection, and/or reducing DSAs [67-72]. In one study, two patients treated with bortezomib for active ABMR showed a transient decrease in bone marrow plasma cells in vivo and persistent alterations in alloantibody specificities. Total immunoglobulin G (IgG) levels were unchanged, suggesting that proteasome activity is important for plasma cell longevity and its inhibition may control antibody production in vivo [67]. In another study, two patients underwent bortezomib-based therapy for active ABMR occurring within the first two weeks posttransplant [71]. Both patients experienced prompt reversal of ABMR and elimination of detectable DSA within 14 days of treatment. However, a follow-up study of 28 patients with active ABMR found that bortezomib therapy was associated with better DSA and histologic response in patients with early (within six months of transplant) rejection but not late ABMR [72].

The use of bortezomib in patients with late ABMR was examined in a randomized, placebo-controlled trial of 44 kidney transplant recipients who were ≥6 months posttransplant, had a positive DSA, and had histologic evidence of active or chronic ABMR [73]. Patients were randomly assigned to treatment with two cycles of bortezomib (1.3 mg/m2 intravenously on days 1, 4, 8, and 11) or placebo; baseline immunosuppression was adjusted as needed according to a predefined protocol. At 24 months posttreatment, there was no significant difference between the groups in the slope of estimated glomerular filtration rate (eGFR) per year (-4.7 versus -5.2 mL/min/1.73m2 per year in the bortezomib- and placebo-treated groups, respectively). Patient and graft survival were comparable between the groups, and there were no differences in median measured GFR, proteinuria, DSA levels, or morphologic or molecular rejection phenotypes in 24-month allograft biopsy samples. However, bortezomib-treated patients experienced higher rates of gastrointestinal and hematologic toxicity.

Eculizumab — Eculizumab is a fully humanized, monoclonal antibody directed against the C5 fragment of the complement cascade and inhibits the generation of the membrane attack complex (MAC). It has received US FDA approval for treatment of paroxysmal nocturnal hemoglobinuria (PNH) and atypical hemolytic uremic syndrome (aHUS). (See "Complement-mediated hemolytic uremic syndrome in children", section on 'Complement blockade (eculizumab)' and "Treatment and prognosis of paroxysmal nocturnal hemoglobinuria", section on 'Eculizumab'.)

In kidney transplantation, eculizumab has been used to prevent ABMR in highly sensitized recipients who undergo desensitization [9]. There are also reports of its successful use as a salvage agent in treating refractory active ABMR [74-80]. The doses used have been similar to those used for the treatment of aHUS, and the duration of dosing has been variable. The lack of randomized, controlled trials proving its efficacy and safety has limited its use in kidney transplant recipients. In addition, ABMR has occurred in kidney transplant recipients who were receiving eculizumab for other indications such as receipt of a positive crossmatch kidney or HUS [81,82]. Eculizumab has not been shown to be effective for the treatment of C4d-negative active and chronic ABMR, suggesting that its efficacy may be limited to acute, complement-mediated processes [9,81,83].

Special populations

Patients with mixed acute rejection — Patients with mixed acute rejection (ie, concurrent histologic evidence of both ABMR and acute T cell-mediated (cellular) rejection [TCMR; Banff grade 2A or greater]) should be treated for both ABMR and TCMR. We treat with the combination of glucocorticoids, plasmapheresis, and IVIG, as detailed above (see 'Approach to initial therapy' above), and add rabbit antithymocyte globulin (rATG)-Thymoglobulin to the treatment regimen. In this setting, we generally perform plasmapheresis and administer IVIG on an alternate-day schedule (eg, Monday, Wednesday, Friday, and Sunday) for a minimum of four treatments. We administer rATG-Thymoglobulin (1.5 to 3 mg/kg) on an alternate-day schedule on the intervening days (eg, Tuesday, Thursday, and Saturday), for a total of three doses.

A discussion of the evidence for the use of rATG-Thymoglobulin in patients with acute TCMR is presented elsewhere. (See "Kidney transplantation in adults: Treatment of acute T cell-mediated (cellular) rejection of the renal allograft", section on 'Banff grade II or III rejection'.)

Patients with subclinical rejection — Subclinical rejection is defined as the presence of histologic evidence of acute rejection on biopsy without an elevation in the serum creatinine concentration. This diagnosis is typically established by a protocol, or surveillance, biopsy, which is obtained at a protocol-driven, prespecified time after transplant rather than for a clinical indication. (See "Kidney transplantation in adults: Clinical features and diagnosis of acute renal allograft rejection", section on 'Subclinical rejection'.)

In all patients who are found to have evidence of subclinical ABMR, we use the same therapeutic approach as that used to treat patients with clinical ABMR (see 'Approach to initial therapy' above), based upon evidence from retrospective studies that suggest that treatment of subclinical ABMR may be associated with improved graft outcomes. One study compared graft outcomes of 219 kidney transplant recipients with ABMR (77 subclinical, 142 clinical) with matched controls without ABMR [63]. One- and five-year graft survival among patients with subclinical ABMR were 95.9 and 75.7 percent, respectively, compared with 96.8 and 88.4 percent in controls. Overall, the risk of graft loss in patients with subclinical ABMR was 2.15-fold greater than that in matched controls. However, there was no significant difference in graft loss between those with treated subclinical ABMR and the controls.

Patients with C4d-negative ABMR — Some patients have histologic evidence of ABMR and a positive DSA but have little or no C4d staining in the peritubular capillaries, an entity recognized as C4d-negative ABMR. Such patients should be treated using the same approach as that for patients with C4d-positive ABMR [84]. (See "Kidney transplantation in adults: Clinical features and diagnosis of acute renal allograft rejection", section on 'C4d-negative antibody-mediated rejection' and 'Approach to initial therapy' above.)

Patients with a non-HLA DSA — ABMR can also occur in patients with non-human leukocyte antigen (HLA) donor-specific antibodies (DSAs), such as anti-angiotensin II type 1 (AT1) receptor antibodies [85] and antiendothelial antibodies [86]. The immunosuppressive treatment of ABMR in such patients is generally the same as that in patients with ABMR and an anti-HLA DSA. Patients who are found to have an anti-AT1 receptor antibody should receive, in addition to immunosuppressive therapy, an angiotensin II receptor blocker, which may inhibit AT1-receptor antibody-mediated effects [85,87]. (See "Kidney transplantation in adults: Clinical features and diagnosis of acute renal allograft rejection", section on 'Detection of donor-specific antibodies'.)

Less frequently used therapies

Immunoadsorption — Immunoadsorption with protein A (IA) has been used to reverse ABMR [88,89]. In the only controlled, open-label trial, 10 patients with severe ABMR were randomly assigned to IA or no IA (with the option of rescue IA after three weeks) [89]. All IA-treated patients responded to therapy (although one death occurred independent of IA), while four control patients remained dialysis dependent. Rescue IA was not successful.

While not available in the United States, selective IA treatment is an attractive alternative to the nonselective combination of plasmapheresis and IVIG. Given the improved outcomes in treating ABMR with plasmapheresis and IVIG, as compared with historical controls, and with IA, as compared with a control group in this small study, further analysis would ideally compare the two treatment methods in a larger number of patients. Although both forms of treatment are expensive, the selective modality of IA without a requirement for IVIG administration would appear preferable if similar outcomes are evident. As previously described, the relative contribution of IVIG or plasmapheresis in treating ABMR is unclear, and further analysis of IA may assist in clarifying this issue.

Splenectomy — We do not routinely perform splenectomy in patients with ABMR, given the lack of evidence that this intervention is safer or more efficacious than available medical therapy. However, some centers consider splenectomy in treating ABMR refractory to plasmapheresis and/or IVIG [90,91].

Four kidney transplant recipients (two ABO incompatible, one crossmatch positive, one with known risk factors) who were diagnosed with an ABMR and failed standard therapy (average 11 days) with steroid, plasmapheresis, IVIG, rATG-Thymoglobulin, and rituximab (three patients) or alemtuzumab (one patient) were treated with laparoscopic splenectomy [90]. Urine output improved immediately, and serum creatinine decreased within 48 hours.

Five patients who underwent a living-donor kidney transplant after desensitization for a positive crossmatch had an ABMR [91]. After rescue attempts with plasmapheresis and IVIG failed, they underwent splenectomy followed by plasmapheresis and IVIG. Allograft function returned within 48 hours of the procedure.

Experimental therapies

C1 inhibitors — Activation of the complement pathway is an important step in the pathogenesis of ABMR. Binding of anti-human leukocyte antigen (HLA) DSAs to complement fraction C1q, the first component in the activation of the complement cascade, has been associated with poor graft outcomes and severe phenotypes of ABMR [29]. These findings have provided the rationale for the use of proximal complement inhibition using C1 inhibitors (C1 INHs) in the treatment of ABMR. C1 INHs have been approved by the US FDA for use in patients with hereditary angioedema. (See "Hereditary angioedema: Acute treatment of angioedema attacks", section on 'First-line agents: Dosing, efficacy, and adverse reactions'.)

The use of a plasma-derived C1 INH in the treatment of active ABMR was evaluated in a phase IIb, multicenter, randomized, controlled trial of 18 kidney transplant recipients with biopsy-proven, active ABMR [92]. Patients were randomly assigned to receive C1 INH 20,000 units or placebo every other day for two weeks (total of seven doses) as adjunct therapy to standard-of-care treatment with plasmapheresis, IVIG, and rituximab. Resolution of ABMR occurred in 78 and 67 percent of patients treated with C1 INH and placebo, respectively. There was no significant difference between the groups in posttreatment renal histopathology or graft survival on day 20; however, a trend toward sustained improvement in graft function at day 90 was observed in the C1 INH group.

Similar findings were reported in a prospective pilot study of six kidney transplant recipients with active ABMR and acute allograft dysfunction that were unresponsive to treatment with plasmapheresis, IVIG, and rituximab [93]. All patients received the C1 INH Berinert (20 units/kg on days 1, 2, and 3 and then twice weekly) and high-dose IVIG (2 g/kg once per month) for six months; maintenance immunosuppression consisted of mycophenolate mofetil, tacrolimus, and prednisone. At six months, all patients showed an improvement in estimated glomerular filtration rate (eGFR) compared with baseline at the time of inclusion in the study. Renal allograft biopsies at six months revealed no significant change in histologic features; however, C4d deposition was observed in only one of six patients compared with five of six patients at baseline. In addition, of the six patients who were positive for a C1q-binding circulating DSA at the start of the study, only one had a positive DSA at six months.

Further studies are needed to determine the efficacy and safety of proximal complement inhibition in the treatment of active ABMR.

TREATMENT OF CHRONIC ANTIBODY-MEDIATED REJECTION — Chronic ABMR, the most common cause of graft failure, is more difficult to treat than active ABMR since irreversible tissue damage has already occurred to the renal allograft. Although evidence suggests that the treatment of antibody-mediated injury requires a combination of strategies to inhibit B cell development, maturation, and activity, it is not clear which combination therapy is safe and effective in patients with chronic ABMR [4,16,83,94-96].

We treat all patients with evidence of chronic ABMR using a combination of glucocorticoids and intravenous (IV) immune globulin (IVIG). We add rituximab to the treatment regimen if there is evidence of active microvascular inflammation on kidney biopsy. We do not use eculizumab or bortezomib in the treatment of chronic ABMR. Our approach in patients with chronic ABMR is similar to that used in patients with active ABMR that occurs after the first year posttransplant. (See 'Approach to initial therapy' above.)

Similar to active ABMR, there is no high-quality evidence to guide the optimal treatment of chronic ABMR, and our approach is based primarily upon the following observational studies:

In a large observational study of 123 consecutive kidney transplant recipients with biopsy-proven, chronic ABMR, 76 percent of patients lost their grafts with a median survival of 1.9 years after the diagnosis of chronic ABMR [16]. Treatment with glucocorticoids and IVIG was associated with a lower risk of graft loss (hazard ratio [HR] 0.44, 95% CI 0.20-0.96). Patients with surviving grafts had a more significant reduction in donor-specific antibodies (DSAs), suggesting the need for more mechanistic interventional clinical trials targeting DSA in patients with chronic ABMR.

In one study of four kidney transplant recipients with chronic ABMR diagnosed between 1 and 27 years posttransplant, treatment with a combination of rituximab and IVIG improved graft function in all patients; DSAs were reduced in two of the four patients [94].

In a retrospective review of 31 patients with chronic ABMR, the median graft survival time was greater among patients treated with rituximab compared with those treated without rituximab (685 versus 439 days) [96].

One randomized, placebo-controlled trial of 25 patients with chronic ABMR found no difference in the decline in estimated glomerular filtration rate (eGFR) at one year between patients treated with the combination of IVIG (four doses of 500 mg/kg) with rituximab (375 mg/m2) and those treated with placebo [97]. However, the study only achieved one-half of its targeted patient enrollment and was therefore underpowered to identify a significant difference in the primary endpoint.

The use of eculizumab in the treatment of chronic ABMR was evaluated in a pilot, randomized, controlled trial of 15 transplant recipients with positive DSA, deteriorating kidney function, and histologic evidence of ABMR [83]. Patients were randomly assigned in a 2:1 ratio to treatment with eculizumab (10 patients) or no eculizumab (5 patients) for six months, followed by six months of observation. At 12 months, there were no differences in graft function or expression of endothelial cell-associated transcripts (ENDATs), a molecular signature predictive of ABMR, between the two groups.

Bortezomib has not been shown to be effective in the treatment of patients with chronic ABMR. (See 'Bortezomib' above.)

Tocilizumab is a monoclonal antibody directed against the interleukin (IL)-6 receptor that has been used for the treatment of rheumatoid arthritis and systemic juvenile idiopathic arthritis (see "Treatment of rheumatoid arthritis in adults resistant to initial biologic DMARD therapy", section on 'Tocilizumab' and "Systemic juvenile idiopathic arthritis: Treatment", section on 'Interleukin 6 inhibitors'). One study has assessed the use of tocilizumab as rescue therapy in 36 kidney transplant patients with chronic ABMR who failed standard-of-care treatment with IVIG and rituximab, with or without plasma exchange [98]. Tocilizumab was administered as 8 mg/kg monthly, with a maximal dose of 800 mg for 6 to 25 months. Graft- and patient-survival rates in tocilizumab-treated patients were 80 and 91 percent at six years posttreatment, respectively. Significant reductions in DSAs and stabilization of renal allograft function were observed at two years. No significant adverse events or severe adverse events were reported. Tocilizumab is not routinely used in the treatment of chronic ABMR but may represent a potential alternative approach to stabilize graft function.

The diagnosis of chronic ABMR in kidney transplant recipients is discussed elsewhere. (See "Kidney transplantation in adults: Clinical features and diagnosis of acute renal allograft rejection", section on 'Chronic active antibody-mediated rejection'.)

SOCIETY GUIDELINE LINKS — Links to society and government-sponsored guidelines from selected countries and regions around the world are provided separately. (See "Society guideline links: Kidney transplantation".)

SUMMARY AND RECOMMENDATIONS

Antibody-mediated rejection (ABMR) is the most common cause of allograft failure after kidney transplantation. Increasing evidence suggests that the prevention and treatment of antibody-mediated injury requires a combination of strategies to inhibit B cell development, maturation, and activity. Despite a relatively large number of observational studies, it is not clear which combination therapy is the safest and most effective. (See 'Introduction' above.)

Active and chronic ABMR are both associated with poor outcomes after kidney transplantation. Patients with active ABMR are at increased risk for subsequent rejection, chronic ABMR, and graft loss. Similarly, those with chronic ABMR have a higher risk for graft loss and patient death. However, not all patients with ABMR have poor outcomes, and many patients maintain stable allograft function for years after treatment of the initial episode of rejection. (See 'Predictors of outcome' above.)

Our approach to the prevention of ABMR depends upon the detection of donor-specific antibody (DSA) prior to (preexisting DSA) or after (de novo DSA) transplant:

Our general approach to the prevention of ABMR in patients with a preexisting DSA prior to transplant is as follows:

-In patients with a potential living donor, the approach depends upon the results of the most recent crossmatch. In patients with a positive complement-dependent cytotoxicity (CDC) crossmatch or a strongly positive flow crossmatch, we, and many other transplant centers, prefer to use kidney paired donation (KPD) programs, rather than desensitization, given the high risk of ABMR and graft loss in such patients. Some transplant centers combine desensitization and KPD. In patients with a positive virtual crossmatch or a mild-to-moderate flow crossmatch (ie, median channel shift of <200), we employ human leukocyte antigen (HLA) desensitization strategies, which include treatment with plasmapheresis, rabbit antithymocyte globulin (rATG)-Thymoglobulin, and rituximab.

-In patients without a potential living donor, we employ HLA desensitization strategies.

-In all patients with a preexisting DSA before transplant who undergo kidney transplantation, we use induction and maintenance immunosuppression therapies that are appropriate for patients at high risk for the development of acute rejection. (See 'Patients with preexisting DSA before transplant' above.)

In patients with a de novo DSA after transplant, prevention of ABMR should focus on addressing nonadherence and under-immunosuppression while balancing the safety and efficacy of long-term immunosuppression. (See 'Patients with de novo DSA after transplant' above.)

The primary goal of treating ABMR is to remove existing DSAs and to eradicate the clonal population of B cells or plasma cells that is responsible for their production. In general, we treat all patients who have evidence of active ABMR on biopsy. Although we do not routinely perform surveillance or protocol allograft biopsies at our institution, we would treat patients who are discovered to have subclinical ABMR by surveillance biopsy. In addition, we treat patients with C4d-negative ABMR with the same approach that we use in patients with C4d-positive ABMR.

(See 'Treatment of active antibody-mediated rejection' above.)

(See 'Patients with subclinical rejection' above.)

(See 'Patients with C4d-negative ABMR' above.)

Although patients with active ABMR can be treated on an outpatient basis, we typically advocate inpatient admission for patients because of the complexity of the treatment regimen. Our approach to the initial treatment of active ABMR depends upon the timing of the diagnosis of ABMR (algorithm 1):

In patients who are diagnosed with active ABMR within the first year posttransplant, we treat with a combination of glucocorticoids, plasmapheresis and intravenous immune globulin (IVIG), and, in some patients, rituximab. (See 'Approach to initial therapy' above.)

In patients who are diagnosed with active ABMR after the first year posttransplant, we treat with glucocorticoids using the same approach as in patients diagnosed with ABMR within the first year posttransplant. However, we do not perform plasmapheresis in such patients because of the lack of evidence supporting the safety and efficacy of plasmapheresis in late-onset ABMR.

In all patients who are treated for active ABMR, we recommence antimicrobial and antiviral prophylaxis with a regimen that is identical to that administered in the immediate posttransplant period. We do not routinely use immunoadsorption, bortezomib, eculizumab, or splenectomy in the initial treatment of patients with ABMR. However, some of these therapies can be considered in patients who do not respond to initial treatment. (See 'Second-line agents in patients who have failed initial therapy' above and 'Less frequently used therapies' above.)

Patients with mixed acute rejection (ie, concurrent histologic evidence of both ABMR and acute T cell-mediated (cellular) rejection [TCMR; Banff grade 2A or greater]) should be treated for both ABMR and TCMR. We treat with the combination of glucocorticoids, plasmapheresis, and IVIG and add rATG-Thymoglobulin to the treatment regimen. (See 'Patients with mixed acute rejection' above.)

Patients are considered to have a successful reversal of ABMR if they meet all of the following parameters within three months of treatment: decrease in serum creatinine to within 20 to 30 percent of the baseline level, decrease in proteinuria to the baseline level, decrease in immunodominant DSA by >50 percent, and resolution of changes associated with ABMR on repeat kidney biopsy.

Most patients with ABMR with a successful response to anti-rejection therapy will demonstrate an improvement in serum creatinine within seven days of treatment. Our subsequent approach to treatment is based upon the response to initial therapy:

In patients with a decrease in serum creatinine in response to therapy, we increase the maintenance tacrolimus dose to achieve a trough level 20 to 25 percent above the level at the time of rejection and resume routine monitoring of allograft function. For patients who are taking the immediate-release formulation of tacrolimus and cannot tolerate higher doses, extended-release tacrolimus, which has fewer side effects and may allow for a higher and therapeutic trough to be obtained, is an alternative option. Typically, maintenance immunosuppression is also augmented by increasing the daily dose of oral prednisone.

Patients without any decrease in serum creatinine after seven days of rejection treatment are considered to have failed initial treatment. In such patients, ongoing rejection and/or another cause of renal allograft dysfunction should be suspected, and a repeat renal allograft biopsy should be performed. Our subsequent approach depends upon the histopathological and clinical findings of the patient. (See 'Monitoring the response to therapy' above.)

Chronic ABMR, the most common cause of graft failure, is more difficult to treat than active ABMR since irreversible tissue damage has already occurred to the renal allograft. It is not clear which combination therapy is safe and effective in patients with chronic ABMR. We treat all patients with evidence of chronic ABMR using a combination of glucocorticoids and IVIG. We add rituximab to the treatment regimen if there is evidence of active microvascular inflammation on kidney biopsy. We do not use eculizumab or bortezomib in the treatment of chronic ABMR. (See 'Treatment of chronic antibody-mediated rejection' above.)

ACKNOWLEDGMENT — The UpToDate editorial staff acknowledges Christina Klein, MD, who contributed to an earlier version of this topic review.

REFERENCES

  1. Wiebe C, Gibson IW, Blydt-Hansen TD, et al. Evolution and clinical pathologic correlations of de novo donor-specific HLA antibody post kidney transplant. Am J Transplant 2012; 12:1157.
  2. Sellarés J, de Freitas DG, Mengel M, et al. Understanding the causes of kidney transplant failure: the dominant role of antibody-mediated rejection and nonadherence. Am J Transplant 2012; 12:388.
  3. El-Zoghby ZM, Stegall MD, Lager DJ, et al. Identifying specific causes of kidney allograft loss. Am J Transplant 2009; 9:527.
  4. Djamali A, Kaufman DB, Ellis TM, et al. Diagnosis and management of antibody-mediated rejection: current status and novel approaches. Am J Transplant 2014; 14:255.
  5. Haas M, Loupy A, Lefaucheur C, et al. The Banff 2017 Kidney Meeting Report: Revised diagnostic criteria for chronic active T cell-mediated rejection, antibody-mediated rejection, and prospects for integrative endpoints for next-generation clinical trials. Am J Transplant 2018; 18:293.
  6. Farkash EA, Colvin RB. Diagnostic challenges in chronic antibody-mediated rejection. Nat Rev Nephrol 2012; 8:255.
  7. Sis B, Halloran PF. Endothelial transcripts uncover a previously unknown phenotype: C4d-negative antibody-mediated rejection. Curr Opin Organ Transplant 2010; 15:42.
  8. Hidalgo LG, Sis B, Sellares J, et al. NK cell transcripts and NK cells in kidney biopsies from patients with donor-specific antibodies: evidence for NK cell involvement in antibody-mediated rejection. Am J Transplant 2010; 10:1812.
  9. Cornell LD, Schinstock CA, Gandhi MJ, et al. Positive crossmatch kidney transplant recipients treated with eculizumab: outcomes beyond 1 year. Am J Transplant 2015; 15:1293.
  10. Mauiyyedi S, Pelle PD, Saidman S, et al. Chronic humoral rejection: identification of antibody-mediated chronic renal allograft rejection by C4d deposits in peritubular capillaries. J Am Soc Nephrol 2001; 12:574.
  11. Drachenberg CB, Papadimitriou JC. Endothelial injury in renal antibody-mediated allograft rejection: a schematic view based on pathogenesis. Transplantation 2013; 95:1073.
  12. Jordan SC, Pescovitz MD. Presensitization: the problem and its management. Clin J Am Soc Nephrol 2006; 1:421.
  13. Dunn TB, Noreen H, Gillingham K, et al. Revisiting traditional risk factors for rejection and graft loss after kidney transplantation. Am J Transplant 2011; 11:2132.
  14. Loupy A, Haas M, Solez K, et al. The Banff 2015 Kidney Meeting Report: Current Challenges in Rejection Classification and Prospects for Adopting Molecular Pathology. Am J Transplant 2017; 17:28.
  15. Amico P, Hönger G, Mayr M, et al. Clinical relevance of pretransplant donor-specific HLA antibodies detected by single-antigen flow-beads. Transplantation 2009; 87:1681.
  16. 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; 77:346.
  17. Gaston RS, Cecka JM, Kasiske BL, et al. Evidence for antibody-mediated injury as a major determinant of late kidney allograft failure. Transplantation 2010; 90:68.
  18. Matignon M, Muthukumar T, Seshan SV, et al. Concurrent acute cellular rejection is an independent risk factor for renal allograft failure in patients with C4d-positive antibody-mediated rejection. Transplantation 2012; 94:603.
  19. Feucht HE, Schneeberger H, Hillebrand G, et al. Capillary deposition of C4d complement fragment and early renal graft loss. Kidney Int 1993; 43:1333.
  20. Djamali A, Muth BL, Ellis TM, et al. Increased C4d in post-reperfusion biopsies and increased donor specific antibodies at one-week post transplant are risk factors for acute rejection in mild to moderately sensitized kidney transplant recipients. Kidney Int 2013; 83:1185.
  21. Loupy A, Hill GS, Suberbielle C, et al. Significance of C4d Banff scores in early protocol biopsies of kidney transplant recipients with preformed donor-specific antibodies (DSA). Am J Transplant 2011; 11:56.
  22. Einecke G, Sis B, Reeve J, et al. Antibody-mediated microcirculation injury is the major cause of late kidney transplant failure. Am J Transplant 2009; 9:2520.
  23. Patri P, Seshan SV, Matignon M, et al. Development and validation of a prognostic index for allograft outcome in kidney recipients with transplant glomerulopathy. Kidney Int 2016; 89:450.
  24. Kamal L, Broin PÓ, Bao Y, et al. Clinical, Histological, and Molecular Markers Associated With Allograft Loss in Transplant Glomerulopathy Patients. Transplantation 2015; 99:1912.
  25. Haas M, Mirocha J, Reinsmoen NL, et al. Differences in pathologic features and graft outcomes in antibody-mediated rejection of renal allografts due to persistent/recurrent versus de novo donor-specific antibodies. Kidney Int 2017; 91:729.
  26. Lefaucheur C, Loupy A, Hill GS, et al. Preexisting donor-specific HLA antibodies predict outcome in kidney transplantation. J Am Soc Nephrol 2010; 21:1398.
  27. Parajuli S, Joachim E, Alagusundaramoorthy S, et al. Donor-Specific Antibodies in the Absence of Rejection Are Not a Risk Factor for Allograft Failure. Kidney Int Rep 2019; 4:1057.
  28. Lefaucheur C, Viglietti D, Bentlejewski C, et al. IgG Donor-Specific Anti-Human HLA Antibody Subclasses and Kidney Allograft Antibody-Mediated Injury. J Am Soc Nephrol 2016; 27:293.
  29. Loupy A, Lefaucheur C, Vernerey D, et al. Complement-binding anti-HLA antibodies and kidney-allograft survival. N Engl J Med 2013; 369:1215.
  30. Yell M, Muth BL, Kaufman DB, et al. C1q Binding Activity of De Novo Donor-specific HLA Antibodies in Renal Transplant Recipients With and Without Antibody-mediated Rejection. Transplantation 2015; 99:1151.
  31. Aubert O, Loupy A, Hidalgo L, et al. Antibody-Mediated Rejection Due to Preexisting versus De Novo Donor-Specific Antibodies in Kidney Allograft Recipients. J Am Soc Nephrol 2017; 28:1912.
  32. Halloran PF, de Freitas DG, Einecke G, et al. An integrated view of molecular changes, histopathology and outcomes in kidney transplants. Am J Transplant 2010; 10:2223.
  33. Haas M. The Revised (2013) Banff Classification for Antibody-Mediated Rejection of Renal Allografts: Update, Difficulties, and Future Considerations. Am J Transplant 2016; 16:1352.
  34. Everly MJ, Everly JJ, Arend LJ, et al. Reducing de novo donor-specific antibody levels during acute rejection diminishes renal allograft loss. Am J Transplant 2009; 9:1063.
  35. Everly MJ, Terasaki PI, Trivedi HL. Durability of antibody removal following proteasome inhibitor-based therapy. Transplantation 2012; 93:572.
  36. Banfi G, Villa M, Cresseri D, Ponticelli C. The clinical impact of chronic transplant glomerulopathy in cyclosporine era. Transplantation 2005; 80:1392.
  37. Loupy A, Lefaucheur C, Vernerey D, et al. Molecular microscope strategy to improve risk stratification in early antibody-mediated kidney allograft rejection. J Am Soc Nephrol 2014; 25:2267.
  38. Loupy A, Aubert O, Orandi BJ, et al. Prediction system for risk of allograft loss in patients receiving kidney transplants: international derivation and validation study. BMJ 2019; 366:l4923.
  39. Orandi BJ, Garonzik-Wang JM, Massie AB, et al. Quantifying the risk of incompatible kidney transplantation: a multicenter study. Am J Transplant 2014; 14:1573.
  40. Mohan S, Palanisamy A, Tsapepas D, et al. Donor-specific antibodies adversely affect kidney allograft outcomes. J Am Soc Nephrol 2012; 23:2061.
  41. 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; 15:3256.
  42. Niederhaus SV, Muth B, Lorentzen DF, et al. Luminex-based desensitization protocols: the University of Wisconsin initial experience. Transplantation 2011; 92:12.
  43. Orandi BJ, Luo X, Massie AB, et al. Survival Benefit with Kidney Transplants from HLA-Incompatible Live Donors. N Engl J Med 2016; 374:940.
  44. Montgomery RA, Lonze BE, King KE, et al. Desensitization in HLA-incompatible kidney recipients and survival. N Engl J Med 2011; 365:318.
  45. Melcher ML, Leeser DB, Gritsch HA, et al. Chain transplantation: initial experience of a large multicenter program. Am J Transplant 2012; 12:2429.
  46. Roodnat JI, Kal-van Gestel JA, Zuidema W, et al. Successful expansion of the living donor pool by alternative living donation programs. Am J Transplant 2009; 9:2150.
  47. Pham TA, Lee JI, Melcher ML. Kidney paired exchange and desensitization: Strategies to transplant the difficult to match kidney patients with living donors. Transplant Rev (Orlando) 2017; 31:29.
  48. Montgomery RA, Zachary AA, Ratner LE, et al. Clinical results from transplanting incompatible live kidney donor/recipient pairs using kidney paired donation. JAMA 2005; 294:1655.
  49. 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; 101:2440.
  50. Tait BD, Süsal C, Gebel HM, et al. Consensus guidelines on the testing and clinical management issues associated with HLA and non-HLA antibodies in transplantation. Transplantation 2013; 95:19.
  51. Al Meshari K, Pall A, Elgamal H, et al. Bortezomib as an adjuvant to conventional therapy in the treatment of antibody mediated rejection (AMR): the full spectrum. Clin Transpl 2010; :383.
  52. OʼLeary JG, Samaniego M, Barrio MC, et al. The Influence of Immunosuppressive Agents on the Risk of De Novo Donor-Specific HLA Antibody Production in Solid Organ Transplant Recipients. Transplantation 2016; 100:39.
  53. Liefeldt L, Brakemeier S, Glander P, et al. Donor-specific HLA antibodies in a cohort comparing everolimus with cyclosporine after kidney transplantation. Am J Transplant 2012; 12:1192.
  54. Vincenti F, Rostaing L, Grinyo J, et al. Belatacept and Long-Term Outcomes in Kidney Transplantation. N Engl J Med 2016; 374:333.
  55. Delgado JC, Fuller A, Ozawa M, et al. No occurrence of de novo HLA antibodies in patients with early corticosteroid withdrawal in a 5-year prospective randomized study. Transplantation 2009; 87:546.
  56. Velidedeoglu E, Cavaillé-Coll MW, Bala S, et al. Summary of 2017 FDA Public Workshop: Antibody-mediated Rejection in Kidney Transplantation. Transplantation 2018; 102:e257.
  57. Kidney Disease: Improving Global Outcomes (KDIGO) Transplant Work Group. KDIGO clinical practice guideline for the care of kidney transplant recipients. Am J Transplant 2009; 9 Suppl 3:S1.
  58. Schinstock CA, Mannon RB, Budde K, et al. Recommended Treatment for Antibody-mediated Rejection After Kidney Transplantation: The 2019 Expert Consensus From the Transplantion Society Working Group. Transplantation 2020; 104:911.
  59. Parajuli S, Mandelbrot DA, Muth B, et al. Rituximab and Monitoring Strategies for Late Antibody-Mediated Rejection After Kidney Transplantation. Transplant Direct 2017; 3:e227.
  60. Roberts DM, Jiang SH, Chadban SJ. The treatment of acute antibody-mediated rejection in kidney transplant recipients-a systematic review. Transplantation 2012; 94:775.
  61. Sautenet B, Blancho G, Büchler M, et al. One-year Results of the Effects of Rituximab on Acute Antibody-Mediated Rejection in Renal Transplantation: RITUX ERAH, a Multicenter Double-blind Randomized Placebo-controlled Trial. Transplantation 2016; 100:391.
  62. Lefaucheur C, Nochy D, Andrade J, et al. Comparison of combination Plasmapheresis/IVIg/anti-CD20 versus high-dose IVIg in the treatment of antibody-mediated rejection. Am J Transplant 2009; 9:1099.
  63. Orandi BJ, Chow EH, Hsu A, et al. Quantifying renal allograft loss following early antibody-mediated rejection. Am J Transplant 2015; 15:489.
  64. Alhamad T, Venkatachalam K, Daloul R, et al. Targeting High Calcineurin Inhibitor Levels After Acute Rejection With Less Tremor: A New Strategy. Transplantation 2017; 101:e287.
  65. San Miguel JF, Schlag R, Khuageva NK, et al. Bortezomib plus melphalan and prednisone for initial treatment of multiple myeloma. N Engl J Med 2008; 359:906.
  66. Jagannath S, Barlogie B, Berenson JR, et al. Bortezomib in recurrent and/or refractory multiple myeloma. Initial clinical experience in patients with impared renal function. Cancer 2005; 103:1195.
  67. Perry DK, Burns JM, Pollinger HS, et al. Proteasome inhibition causes apoptosis of normal human plasma cells preventing alloantibody production. Am J Transplant 2009; 9:201.
  68. Nigos JG, Arora S, Nath P, et al. Treatment of antibody-mediated rejection in kidney transplant recipients: a single-center experience with a bortezomib-based regimen. Exp Clin Transplant 2012; 10:609.
  69. Everly MJ, Everly JJ, Susskind B, et al. Bortezomib provides effective therapy for antibody- and cell-mediated acute rejection. Transplantation 2008; 86:1754.
  70. Flechner SM, Fatica R, Askar M, et al. The role of proteasome inhibition with bortezomib in the treatment of antibody-mediated rejection after kidney-only or kidney-combined organ transplantation. Transplantation 2010; 90:1486.
  71. Walsh RC, Everly JJ, Brailey P, et al. Proteasome inhibitor-based primary therapy for antibody-mediated renal allograft rejection. Transplantation 2010; 89:277.
  72. Walsh RC, Brailey P, Girnita A, et al. Early and late acute antibody-mediated rejection differ immunologically and in response to proteasome inhibition. Transplantation 2011; 91:1218.
  73. Eskandary F, Regele H, Baumann L, et al. A Randomized Trial of Bortezomib in Late Antibody-Mediated Kidney Transplant Rejection. J Am Soc Nephrol 2018; 29:591.
  74. Orandi BJ, Zachary AA, Dagher NN, et al. Eculizumab and splenectomy as salvage therapy for severe antibody-mediated rejection after HLA-incompatible kidney transplantation. Transplantation 2014; 98:857.
  75. Fan J, Tryphonopoulos P, Tekin A, et al. Eculizumab Salvage Therapy for Antibody-Mediated Rejection in a Desensitization-Resistant Intestinal Re-Transplant Patient. Am J Transplant 2015; 15:1995.
  76. Yamamoto T, Watarai Y, Futamura K, et al. Efficacy of Eculizumab Therapy for Atypical Hemolytic Uremic Syndrome Recurrence and Antibody-Mediated Rejection Progress After Renal Transplantation With Preformed Donor-Specific Antibodies: Case Report. Transplant Proc 2017; 49:159.
  77. 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; 9:231.
  78. Kocak B, Arpali E, Demiralp E, et al. Eculizumab for salvage treatment of refractory antibody-mediated rejection in kidney transplant patients: case reports. Transplant Proc 2013; 45:1022.
  79. Ghirardo G, Benetti E, Poli F, et al. Plasmapheresis-resistant acute humoral rejection successfully treated with anti-C5 antibody. Pediatr Transplant 2014; 18:E1.
  80. Eskandary F, Wahrmann M, Mühlbacher J, Böhmig GA. Complement inhibition as potential new therapy for antibody-mediated rejection. Transpl Int 2016; 29:392.
  81. Burbach M, Suberbielle C, Brochériou I, et al. Report of the inefficacy of eculizumab in two cases of severe antibody-mediated rejection of renal grafts. Transplantation 2014; 98:1056.
  82. Bentall A, Tyan DB, Sequeira F, et al. Antibody-mediated rejection despite inhibition of terminal complement. Transpl Int 2014; 27:1235.
  83. Kulkarni S, Kirkiles-Smith NC, Deng YH, et al. Eculizumab Therapy for Chronic Antibody-Mediated Injury in Kidney Transplant Recipients: A Pilot Randomized Controlled Trial. Am J Transplant 2017; 17:682.
  84. Orandi BJ, Alachkar N, Kraus ES, et al. Presentation and Outcomes of C4d-Negative Antibody-Mediated Rejection After Kidney Transplantation. Am J Transplant 2016; 16:213.
  85. Dragun D, Müller DN, Bräsen JH, et al. Angiotensin II type 1-receptor activating antibodies in renal-allograft rejection. N Engl J Med 2005; 352:558.
  86. Sun Q, Liu Z, Yin G, et al. Detectable circulating antiendothelial cell antibodies in renal allograft recipients with C4d-positive acute rejection: a report of three cases. Transplantation 2005; 79:1759.
  87. Fuss A, Hope CM, Deayton S, et al. C4d-negative antibody-mediated rejection with high anti-angiotensin II type I receptor antibodies in absence of donor-specific antibodies. Nephrology (Carlton) 2015; 20:467.
  88. Böhmig GA, Exner M, Watschinger B, et al. C4d deposits in renal allografts are associated with inferior graft outcome. Transplant Proc 2001; 33:1151.
  89. Böhmig GA, Wahrmann M, Regele H, et al. Immunoadsorption in severe C4d-positive acute kidney allograft rejection: a randomized controlled trial. Am J Transplant 2007; 7:117.
  90. Kaplan B, Gangemi A, Thielke J, et al. Successful rescue of refractory, severe antibody mediated rejection with splenectomy. Transplantation 2007; 83:99.
  91. Locke JE, Zachary AA, Haas M, et al. The utility of splenectomy as rescue treatment for severe acute antibody mediated rejection. Am J Transplant 2007; 7:842.
  92. Montgomery RA, Orandi BJ, Racusen L, et al. Plasma-Derived C1 Esterase Inhibitor for Acute Antibody-Mediated Rejection Following Kidney Transplantation: Results of a Randomized Double-Blind Placebo-Controlled Pilot Study. Am J Transplant 2016; 16:3468.
  93. Viglietti D, Gosset C, Loupy A, et al. C1 Inhibitor in Acute Antibody-Mediated Rejection Nonresponsive to Conventional Therapy in Kidney Transplant Recipients: A Pilot Study. Am J Transplant 2016; 16:1596.
  94. Fehr T, Rüsi B, Fischer A, et al. Rituximab and intravenous immunoglobulin treatment of chronic antibody-mediated kidney allograft rejection. Transplantation 2009; 87:1837.
  95. Fehr T, Gaspert A. Antibody-mediated kidney allograft rejection: therapeutic options and their experimental rationale. Transpl Int 2012; 25:623.
  96. Smith RN, Malik F, Goes N, et al. Partial therapeutic response to Rituximab for the treatment of chronic alloantibody mediated rejection of kidney allografts. Transpl Immunol 2012; 27:107.
  97. Moreso F, Crespo M, Ruiz JC, et al. Treatment of chronic antibody mediated rejection with intravenous immunoglobulins and rituximab: A multicenter, prospective, randomized, double-blind clinical trial. Am J Transplant 2018; 18:927.
  98. Choi J, Aubert O, Vo A, et al. Assessment of Tocilizumab (Anti-Interleukin-6 Receptor Monoclonal) as a Potential Treatment for Chronic Antibody-Mediated Rejection and Transplant Glomerulopathy in HLA-Sensitized Renal Allograft Recipients. Am J Transplant 2017; 17:2381.
Topic 7326 Version 29.0

References

1 : Evolution and clinical pathologic correlations of de novo donor-specific HLA antibody post kidney transplant.

2 : Understanding the causes of kidney transplant failure: the dominant role of antibody-mediated rejection and nonadherence.

3 : Identifying specific causes of kidney allograft loss.

4 : Diagnosis and management of antibody-mediated rejection: current status and novel approaches.

5 : The Banff 2017 Kidney Meeting Report: Revised diagnostic criteria for chronic active T cell-mediated rejection, antibody-mediated rejection, and prospects for integrative endpoints for next-generation clinical trials.

6 : Diagnostic challenges in chronic antibody-mediated rejection.

7 : Endothelial transcripts uncover a previously unknown phenotype: C4d-negative antibody-mediated rejection.

8 : NK cell transcripts and NK cells in kidney biopsies from patients with donor-specific antibodies: evidence for NK cell involvement in antibody-mediated rejection.

9 : Positive crossmatch kidney transplant recipients treated with eculizumab: outcomes beyond 1 year.

10 : Chronic humoral rejection: identification of antibody-mediated chronic renal allograft rejection by C4d deposits in peritubular capillaries.

11 : Endothelial injury in renal antibody-mediated allograft rejection: a schematic view based on pathogenesis.

12 : Presensitization: the problem and its management.

13 : Revisiting traditional risk factors for rejection and graft loss after kidney transplantation.

14 : The Banff 2015 Kidney Meeting Report: Current Challenges in Rejection Classification and Prospects for Adopting Molecular Pathology.

15 : Clinical relevance of pretransplant donor-specific HLA antibodies detected by single-antigen flow-beads.

16 : Current outcomes of chronic active antibody mediated rejection - A large single center retrospective review using the updated BANFF 2013 criteria.

17 : Evidence for antibody-mediated injury as a major determinant of late kidney allograft failure.

18 : Concurrent acute cellular rejection is an independent risk factor for renal allograft failure in patients with C4d-positive antibody-mediated rejection.

19 : Capillary deposition of C4d complement fragment and early renal graft loss.

20 : Increased C4d in post-reperfusion biopsies and increased donor specific antibodies at one-week post transplant are risk factors for acute rejection in mild to moderately sensitized kidney transplant recipients.

21 : Significance of C4d Banff scores in early protocol biopsies of kidney transplant recipients with preformed donor-specific antibodies (DSA).

22 : Antibody-mediated microcirculation injury is the major cause of late kidney transplant failure.

23 : Development and validation of a prognostic index for allograft outcome in kidney recipients with transplant glomerulopathy.

24 : Clinical, Histological, and Molecular Markers Associated With Allograft Loss in Transplant Glomerulopathy Patients.

25 : Differences in pathologic features and graft outcomes in antibody-mediated rejection of renal allografts due to persistent/recurrent versus de novo donor-specific antibodies.

26 : Preexisting donor-specific HLA antibodies predict outcome in kidney transplantation.

27 : Donor-Specific Antibodies in the Absence of Rejection Are Not a Risk Factor for Allograft Failure.

28 : IgG Donor-Specific Anti-Human HLA Antibody Subclasses and Kidney Allograft Antibody-Mediated Injury.

29 : Complement-binding anti-HLA antibodies and kidney-allograft survival.

30 : C1q Binding Activity of De Novo Donor-specific HLA Antibodies in Renal Transplant Recipients With and Without Antibody-mediated Rejection.

31 : Antibody-Mediated Rejection Due to Preexisting versus De Novo Donor-Specific Antibodies in Kidney Allograft Recipients.

32 : An integrated view of molecular changes, histopathology and outcomes in kidney transplants.

33 : The Revised (2013) Banff Classification for Antibody-Mediated Rejection of Renal Allografts: Update, Difficulties, and Future Considerations.

34 : Reducing de novo donor-specific antibody levels during acute rejection diminishes renal allograft loss.

35 : Durability of antibody removal following proteasome inhibitor-based therapy.

36 : The clinical impact of chronic transplant glomerulopathy in cyclosporine era.

37 : Molecular microscope strategy to improve risk stratification in early antibody-mediated kidney allograft rejection.

38 : Prediction system for risk of allograft loss in patients receiving kidney transplants: international derivation and validation study.

39 : Quantifying the risk of incompatible kidney transplantation: a multicenter study.

40 : Donor-specific antibodies adversely affect kidney allograft outcomes.

41 : 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.

42 : Luminex-based desensitization protocols: the University of Wisconsin initial experience.

43 : Survival Benefit with Kidney Transplants from HLA-Incompatible Live Donors.

44 : Desensitization in HLA-incompatible kidney recipients and survival.

45 : Chain transplantation: initial experience of a large multicenter program.

46 : Successful expansion of the living donor pool by alternative living donation programs.

47 : Kidney paired exchange and desensitization: Strategies to transplant the difficult to match kidney patients with living donors.

48 : Clinical results from transplanting incompatible live kidney donor/recipient pairs using kidney paired donation.

49 : Long-term Outcomes of Kidney Transplantation in Patients With High Levels of Preformed DSA: The Necker High-Risk Transplant Program.

50 : Consensus guidelines on the testing and clinical management issues associated with HLA and non-HLA antibodies in transplantation.

51 : Bortezomib as an adjuvant to conventional therapy in the treatment of antibody mediated rejection (AMR): the full spectrum.

52 : The Influence of Immunosuppressive Agents on the Risk of De Novo Donor-Specific HLA Antibody Production in Solid Organ Transplant Recipients.

53 : Donor-specific HLA antibodies in a cohort comparing everolimus with cyclosporine after kidney transplantation.

54 : Belatacept and Long-Term Outcomes in Kidney Transplantation.

55 : No occurrence of de novo HLA antibodies in patients with early corticosteroid withdrawal in a 5-year prospective randomized study.

56 : Summary of 2017 FDA Public Workshop: Antibody-mediated Rejection in Kidney Transplantation.

57 : KDIGO clinical practice guideline for the care of kidney transplant recipients.

58 : Recommended Treatment for Antibody-mediated Rejection After Kidney Transplantation: The 2019 Expert Consensus From the Transplantion Society Working Group.

59 : Rituximab and Monitoring Strategies for Late Antibody-Mediated Rejection After Kidney Transplantation.

60 : The treatment of acute antibody-mediated rejection in kidney transplant recipients-a systematic review.

61 : One-year Results of the Effects of Rituximab on Acute Antibody-Mediated Rejection in Renal Transplantation: RITUX ERAH, a Multicenter Double-blind Randomized Placebo-controlled Trial.

62 : Comparison of combination Plasmapheresis/IVIg/anti-CD20 versus high-dose IVIg in the treatment of antibody-mediated rejection.

63 : Quantifying renal allograft loss following early antibody-mediated rejection.

64 : Targeting High Calcineurin Inhibitor Levels After Acute Rejection With Less Tremor: A New Strategy.

65 : Bortezomib plus melphalan and prednisone for initial treatment of multiple myeloma.

66 : Bortezomib in recurrent and/or refractory multiple myeloma. Initial clinical experience in patients with impared renal function.

67 : Proteasome inhibition causes apoptosis of normal human plasma cells preventing alloantibody production.

68 : Treatment of antibody-mediated rejection in kidney transplant recipients: a single-center experience with a bortezomib-based regimen.

69 : Bortezomib provides effective therapy for antibody- and cell-mediated acute rejection.

70 : The role of proteasome inhibition with bortezomib in the treatment of antibody-mediated rejection after kidney-only or kidney-combined organ transplantation.

71 : Proteasome inhibitor-based primary therapy for antibody-mediated renal allograft rejection.

72 : Early and late acute antibody-mediated rejection differ immunologically and in response to proteasome inhibition.

73 : A Randomized Trial of Bortezomib in Late Antibody-Mediated Kidney Transplant Rejection.

74 : Eculizumab and splenectomy as salvage therapy for severe antibody-mediated rejection after HLA-incompatible kidney transplantation.

75 : Eculizumab Salvage Therapy for Antibody-Mediated Rejection in a Desensitization-Resistant Intestinal Re-Transplant Patient.

76 : Efficacy of Eculizumab Therapy for Atypical Hemolytic Uremic Syndrome Recurrence and Antibody-Mediated Rejection Progress After Renal Transplantation With Preformed Donor-Specific Antibodies: Case Report.

77 : The use of antibody to complement protein C5 for salvage treatment of severe antibody-mediated rejection.

78 : Eculizumab for salvage treatment of refractory antibody-mediated rejection in kidney transplant patients: case reports.

79 : Plasmapheresis-resistant acute humoral rejection successfully treated with anti-C5 antibody.

80 : Complement inhibition as potential new therapy for antibody-mediated rejection.

81 : Report of the inefficacy of eculizumab in two cases of severe antibody-mediated rejection of renal grafts.

82 : Antibody-mediated rejection despite inhibition of terminal complement.

83 : Eculizumab Therapy for Chronic Antibody-Mediated Injury in Kidney Transplant Recipients: A Pilot Randomized Controlled Trial.

84 : Presentation and Outcomes of C4d-Negative Antibody-Mediated Rejection After Kidney Transplantation.

85 : Angiotensin II type 1-receptor activating antibodies in renal-allograft rejection.

86 : Detectable circulating antiendothelial cell antibodies in renal allograft recipients with C4d-positive acute rejection: a report of three cases.

87 : C4d-negative antibody-mediated rejection with high anti-angiotensin II type I receptor antibodies in absence of donor-specific antibodies.

88 : C4d deposits in renal allografts are associated with inferior graft outcome.

89 : Immunoadsorption in severe C4d-positive acute kidney allograft rejection: a randomized controlled trial.

90 : Successful rescue of refractory, severe antibody mediated rejection with splenectomy.

91 : The utility of splenectomy as rescue treatment for severe acute antibody mediated rejection.

92 : Plasma-Derived C1 Esterase Inhibitor for Acute Antibody-Mediated Rejection Following Kidney Transplantation: Results of a Randomized Double-Blind Placebo-Controlled Pilot Study.

93 : C1 Inhibitor in Acute Antibody-Mediated Rejection Nonresponsive to Conventional Therapy in Kidney Transplant Recipients: A Pilot Study.

94 : Rituximab and intravenous immunoglobulin treatment of chronic antibody-mediated kidney allograft rejection.

95 : Antibody-mediated kidney allograft rejection: therapeutic options and their experimental rationale.

96 : Partial therapeutic response to Rituximab for the treatment of chronic alloantibody mediated rejection of kidney allografts.

97 : Treatment of chronic antibody mediated rejection with intravenous immunoglobulins and rituximab: A multicenter, prospective, randomized, double-blind clinical trial.

98 : Assessment of Tocilizumab (Anti-Interleukin-6 Receptor Monoclonal) as a Potential Treatment for Chronic Antibody-Mediated Rejection and Transplant Glomerulopathy in HLA-Sensitized Renal Allograft Recipients.