Kidney Res Clin Pract > Volume 40(3); 2021 > Article
Park, Ko, Chung, and Yang: Kidney transplantation in highly sensitized recipients

Abstract

In kidney transplantation (KT), overcoming donor shortage is particularly challenging in patients with preexisting donor-specific antibodies (DSAs) against human leukocyte antigen (HLA), called HLA-incompatible KT (HLAi KT), carrying the risk of rejection and allograft loss. Thus, it is necessary to accurately evaluate the degree of sensitization before HLAi KT, and undertake appropriate pretreatment strategies. To determine the degree of sensitization, complement-dependent cytotoxicity has been the only method employed; the development of a method using flow cytometry further improved the test sensitivity. However, these tests present disadvantages, including the need for living cells, with a solid-phase assay developed to resolve this problem. Currently, the method using Luminex (Luminex Corp.) is widely used in clinical practice. As this method measures DSAs using single antigen beads, it is possible to classify immunological risks by measuring the type and amount of DSAs. Furthermore, there have been major advances in methods that involve DSA removal before HLAi KT. In the early stages of desensitization, plasmapheresis and intravenous immunoglobulins were the main treatment methods employed; however, the introduction of CD20 monoclonal antibody and proteasome inhibitors further increased the success rate of desensitization. Currently, HLAi KT has been established as an important transplant method, but an understanding of DSAs and a novel desensitization treatment are warranted.

Introduction

Kidney transplantation (KT) is a well-known treatment strategy that best improves the quality of life and survival outcomes in patients with end-stage kidney disease (ESKD) [1]. However, although the number of patients with ESKD is markedly increasing worldwide, the number of living donors is limited, resulting in a growing number of patients on waiting lists for transplantation [2,3]. To overcome this donor shortage, KT in ABO-incompatible (ABOi) or sensitized patients has been attempted to increase the potential living donor pool. In the Republic of Korea, ABOi KT was initiated in 2007 and has been rapidly increasing [4]. Furthermore, a case of successful KT was reported in the Republic of Korea in 2002 after plasmapheresis was performed in a patient who had a positive crossmatch test with a living donor [5], and KT in highly sensitized recipients is currently being actively implemented across several centers.
KT in recipients with alloantibodies to donor human leukocyte antigens (HLAs) is termed HLA-incompatible (HLAi) KT. The presence of alloantibodies against HLAs of potential donors was previously considered a major barrier to KT [6]. However, as new technologies for measuring the characteristics and strength of these donor-specific antibodies (DSAs) have emerged since the early 2000s, immunologic risk stratification has been possible in highly sensitized recipients [7] along with advances in desensitization treatment. Accordingly, a highly sensitized status as a barrier to KT is being surpassed by these developments, which are being actively implemented to overcome donor shortage [8].
In this review, we describe the pretransplant alloantibody detection, desensitization treatment, allograft and patient outcomes, and our center’s experiences and outcomes in HLAi KT.

Alloantibody detection

To evaluate highly sensitized recipients and to implement appropriate treatment regimens, it is crucial to first understand the various tests for immunologic risk stratification. Successful KT can be achieved by identifying preformed DSAs through complement-dependent cytotoxicity (CDC) crossmatch, flow cytometry crossmatch (FCXM), and solid-phase binding assay (SPA), and reaching the appropriate target antibody range before transplantation [9]. As large differences in sensitivity and specificity exist between these tests, individualized immunologic risk stratification is generally performed by employing a combination of test results [10].

Complement-dependent cytotoxicity crossmatch

CDC crossmatch is a traditional test that was first employed as a pretransplant immunological test and remains a routinely performed test until now [6]. This test can detect whether complement-fixing immunoglobulin (Ig) M and IgG antibodies targeting donor lymphocytes are present in the recipient’s serum [11]. Typically, the test result is positive only when there are sufficient antibodies that can bind to the donor antigen and activate the complement cascade; hence, the sensitivity of CDC crossmatch is relatively inferior when compared with other tests [12].
The CDC crossmatch has several limitations. This test can only detect complement-fixing antibodies, requires viable donor lymphocytes for testing, and the sensitivity may vary depending on the rabbit complement batch. To overcome these limitations, various techniques have been employed [12]. Prolonging the incubation time to 120 minutes after adding the complement can increase the test sensitivity. The Amos wash technique is used to increase test sensitivity and detect only clinically meaningful IgG antibodies by removing anticomplementary factors and low-affinity IgM antibodies by washing the unbound antibodies from the cell suspension before adding complement [13]. The anti-human globulin (AHG) augmentation method is widely used to increase test sensitivity by adding an anti-kappa light chain. AHG enhances complement activation via the proximity of the Fc portion of the antibody. Therefore, detection of low-titer anti-HLA antibodies and non-complement-fixing antibodies is feasible [14]. Pretreatment of patient serum with dithiothreitol or dithioerythritol removes disulfide bonds from the IgM pentamer, but IgG remains relatively intact; hence, IgG and IgM antibodies can be distinguished [15].
Despite these efforts, CDC crossmatch is a reaction between a patient’s serum and donor lymphocytes; hence, the specific profile of the antibody cannot be identified, and false positives may be demonstrated by non-HLA antibodies that are not pathogenic [16]. Furthermore, this test may be affected by drugs or the patient’s underlying disease, with false positives detected owing to the patient’s autoantibodies [17]. Moreover, rituximab can be detected in a patient’s serum for more than 3 months; hence, the B-cell CDC crossmatch can produce false positives during this period [18]. Nevertheless, the CDC crossmatch test remains meaningful as it can stratify high immunologic risks and can be used to reevaluate pretransplant immunologic risks after appropriate desensitization treatment.

Flow cytometry crossmatch

Although CDC crossmatch is effective in avoiding hyperacute rejection, several transplant patients have revealed poor clinical outcomes such as primary non-function and delayed graft function. This could be attributed to low-titer DSA, which is lower than the detection threshold of conventional CDC crossmatch in sensitized patients. Garovoy et al. [19] identified low-titer IgG DSAs by the FCXM method, which was not observed in CDC crossmatch. FCXM is a more sensitive test than CDC crossmatch, and it was reported that the incidence of delayed graft function, acute rejection, and graft failure was higher in the case of FCXM-positive with CDC crossmatch-negative than that of both negatives before transplantation, suggesting that weak and sublytic DSAs play a pathogenic role [20].
FCXM is a cell-based method in which donor lymphocytes react with the recipient serum, similar to CDC crossmatch. However, unlike CDC crossmatch, in this method, the flow cytometer signal is read by adding fluorochrome-conjugated antibodies rather than by employing a cytotoxic response [21]. In some laboratories, donor lymphocytes are pretreated with pronase to remove Fc receptors and CD20 from the B-cell surface. By removing the Fc receptor on the B-cell surface, background noise that may occur due to binding of non-HLA antibodies can be removed; removal of CD20 can reduce the effect of rituximab on B-cell FCXM [22].
As a significant proportion of patients reportedly presented an uneventful clinical course, although they were FCXM-positive, FCXM is considered a test with high sensitivity and low specificity in early graft dysfunction and antibody-mediated rejection (ABMR) [23]. Furthermore, FCXM can be impacted by several variable factors, including flow cytometers, fluorochrome reagents, cell-to-serum ratio, and incubation conditions, rendering standardization difficult. It is necessary to establish individual cutoff values for each laboratory [24]. Moreover, the problem of false positives induced by non-HLA antibodies, autoantibodies, and rituximab tends to persist, which are disadvantages associated with traditional CDC crossmatch [23].
FCXM results must be interpreted together with the CDC crossmatch results (Table 1). For example, when only T-cell CDC and T-cell FCXM results are positive, this may indicate T cell-specific antibody but probably not IgG HLA class I antibody; T-cell FCXM-negative and only B-cell FCXM-positive may indicate the presence of class II DSAs, low-titer class I DSAs, autoantibodies, or antibodies against major histocompatibility complex class I-related chain A [25]. Therefore, CDC and FCXM test results must be interpreted in combination with the SPA test described later to obtain accurate information regarding immunologic risks in sensitized patients.

Solid-phase binding assays

Advances in the SPA method have greatly contributed to the increased sensitivity and specificity of HLA antibody detection [7]. The first SPA techniques were enzyme-linked immunosorbent assay and flow cytometry, but recently, these have been largely superseded by "Luminex" technology (Luminex Corp., Austin, TX, USA), which uses polystyrene microbeads with attached purified HLA proteins. The panel reactive antibody (PRA) test is a method to screen anti-HLA antibodies present in the recipient’s serum using a pooled antigen panel composed of microbeads of HLA class I or class II antigens, obtained from multiple donors. A single antigen bead (SAB) panel is the most sensitive test method because each bead is coated with only one HLA allelic antigen, presenting the advantage of being able to detect specific DSAs [26]. Therefore, it is recommended that highly sensitized recipients perform at least one SAB assay before transplantation [24].
Antigen-coated microbeads are incubated with the recipient’s serum, followed by addition of fluorescent-labeled anti-human IgG. If anti-HLA antibodies are present in the recipient’s serum, they bind to the microbeads, and as fluorescent-labeled anti-human IgG binds sequentially to anti-HLA antibodies, it can be detected using a dual-laser instrument [24]. Luminex SAB provides semiquantitative information regarding the anti-HLA antibody titer through median fluorescence intensity (MFI) values, which are used as important information in pretransplant immunologic risk assessment [27]. Luminex SAB may present false-positive or false-negative results owing to IgM or various inhibitory factors present in the recipient’s serum. However, these limitations can be overcome through hypotonic dialysis, heat inactivation, or pretreatment with dithiothreitol or ethylenediaminetetraacetic acid [28].
However, as Luminex SAB is a semiquantitative test, results should be interpreted by utilizing professional knowledge rather than absolute values. In particular, as the coefficient of variance of the MFI values exceeds 25%, a change of less than 1,000 or less than 25% of the MFI values should not be interpreted as clinically significant [27]. Additionally, the prozone phenomenon may occur at markedly high antibody titers. The high titer of antibodies interferes with the formation of the antibody-antigen complex, resulting in a false-negative result. This is clinically indicated by a strong positive CDC crossmatch result, with a negative SAB assay result. In this case, when the recipient’s serum is diluted, the SAB assay results in an increase in the MFI values, allowing antibody detection [29]. The presence of antibodies against the shared epitope in the recipient’s serum may result in false-positive results for multiple microbeads representing HLA antigens [30]. Furthermore, the SAB assay is an extremely sensitive test and does not show ABMR or poor allograft survival in all patients with detected DSAs [31,32]. Therefore, to detect cytotoxic DSAs, a modified test method (C1q binding assay, C3d binding assay) that detects antibodies capable of complement activation is currently being implemented [33].
As the SAB assay can detect specific antibodies, calculated PRA (cPRA) can be derived. cPRA is an index that evaluates the actual degree of sensitization by reflecting the HLA antigen frequencies of the population and can be extremely important in counseling the waiting time of highly sensitized recipients waiting for deceased donors [34]. Furthermore, virtual crossmatching is possible by comparing the recipient’s specific anti-HLA antibodies and donor HLA typing results. The deceased donor allocation time can be reduced by allowing the compatible donor-recipient combination to be checked in advance before the actual crossmatch test [35]. In addition, the SAB assay can be used for monitoring de novo DSA, allowing proper patient management after transplantation [36].
Individualized pretransplant immunologic risk assessment is possible by using the results of previously described CDC crossmatch, FCXM, and SAB assays collectively. By interpreting these test results together, the immunologic risk of sensitized patients can be stratified into the high-risk group (the probability of hyperacute rejection is high if antibody reduction therapy is not performed owing to high-titer DSAs), the intermediate-risk group (the probability of hyperacute rejection is low, with a higher probability of ABMR and poor allograft outcome), and the low-risk group (the incidence of rejection may be high owing to low-titer DSAs, but the evidence for poor allograft outcome is insufficient) [10] (Table 2). At our center, as immunologic risk evaluation, crossmatch and PRA are used as screening tests to identify the presence of anti-HLA antibodies; if the screening test is positive, the SAB assay is performed to identify specific DSAs (Fig. 1).

Future direction

As the development of the SAB assay allows the identification of specific DSAs, there has been great progress in evaluating the degree of sensitization. However, some challenges need to be further addressed [37].
Complement-binding assays determine whether DSA can activate the complement cascade. This has been reported as a valuable test for pretransplant immunologic risk stratification, predicting ABMR and graft loss occurrence [38,39]. Several studies have reported that complement-binding DSA has a strong correlation with high antibody titers (high MFI levels) [40,41]. However, since the antibody titer can change dynamically, the complement-binding assay can show inconsistent results [40]. In addition, in a previous study, ABMR with DSA did not show complement fixation in vitro, but 40% of them showed C4d-positive histology in vivo [42]; thus, further research is needed on this.
It is thought that the mechanism of alloimmune response is different depending on the IgG subclass. IgG1 and IgG3 subclasses are known to have stronger complement-fixing properties than IgG2 and IgG4 subclasses [43]. Furthermore, previous studies have reported that IgG1 and IgG3 subclasses are strongly related to acute rejection and graft loss [44,45]. However, there is a technical limitation that the currently used IgG subclass-specific reagents cannot classify subclasses in 10% to 20% of total IgG-positive HLA antibodies [44]. In addition, IgG2 and IgG4 are also known to have some correlation with rejection [46]; therefore, further research is warranted.
Progress is being made in uncovering the outside-in signal transduction pathway by DSA. As the mechanistic target of rapamycin signaling axis is revealed as a pathway for activation of endothelial cells by HLA class II molecules [47], research on the development of diagnostic tools and drugs using this is expected. In addition, the relationship between the Fc receptor genotype and HLA subclass in leukocyte recruitment and microcirculation inflammation has been reported [48], and the pathological mechanisms for this are expected to be uncovered in the future.
DSA is not observed, but the presence of previously sensitized memory B/T cells may induce a rapid alloimmune response after transplantation [49]. Therefore, attempts are being made to discover these memory B/T cells before transplantation. Test methods such as flow cytometric HLA-binding memory B-cell assay, in vitro differentiation of memory B-cell assay, donor-specific interferon-γ secreting T-cell assay, follicular helper T-cell assay, immunophenotypic bulk memory T-cell assay have been attempted, but there are no tests showing clinically useful predictable power to date [5054].
HLA allorecognition by B/T cells is more likely to occur as recipient HLA and donor HLA molecules are more different [55]. Currently, computational algorithms can be used to quantify the HLA molecule mismatch (mMM) between the recipient and donor. Commonly used methods are HLA Matchmaker, Electrostatic Mismatch Score, Predicted Indirectly ReCognizable HLA Epitopes Matching, and simple counting of amino acid mismatches [5658]. In previous studies, the HLA-DR or -DQ mMM score was an independent risk factor for de novo DSA, and it has been reported that both simple HLA type mismatch and mMM are important for pretransplant immunologic risk stratification [59]. However, since various mMM calculation systems currently exist, standardization is required. Moreover, some mMMs have been reported to have stronger immunogenicity than other mMMs, necessitating further research on this [57,60].

Desensitization treatment

Currently, desensitization is performed by removing preexisting antibodies and inhibiting the production of antibodies (Fig. 2). Plasmapheresis, immunoadsorption, high-dose intravenous immunoglobulin (IVIG), and plasmapheresis with low-dose IVIG methods are available to remove existing antibodies [61]. With the introduction of drugs that inhibit the production of antibodies, the success rate of desensitization treatment has increased. As such, CD20 monoclonal antibody that removes B cells or a proteasome inhibitor, which induces apoptosis of plasma cells, can be employed. The most widely used method for desensitization treatment worldwide is plasmapheresis with low-dose IVIG [62] in conjunction with rituximab [8]. Our center also uses a desensitization protocol based on plasmapheresis with low-dose IVIG with rituximab (Fig. 3A) [63]. If T-CDC-AHG is positive or if there is no appropriate DSA MFI titer reduction even after three or more plasmapheresis sessions, a bortezomib-added protocol is performed (Fig. 3B) [64]. In the case of PRA ≥ 50% alone (with crossmatch-negative and DSA MFI titer < 3,000), only rituximab is administered 7 days before transplantation without plasmapheresis [65].

Plasmapheresis or immunoadsorption

Plasmapheresis or immunoadsorption is a method that physically removes immunoglobulins from the recipient’s serum. As a desensitization method, plasmapheresis is mainly performed in the United States, and immunoadsorption is mainly performed in Europe. As in the United States, plasmapheresis is currently the main method undertaken in the Republic of Korea. Plasmapheresis is not specific to the removal of alloantibodies, and hence, all plasma proteins, including albumin and blood coagulation factors, are lost, which need to be replaced with albumin or fresh frozen plasma. Immunoadsorption utilizes columns that selectively remove the immunoglobulin, preventing excessive loss of coagulation factors [66]. Reportedly, one treatment session with plasmapheresis or immunoadsorption can result in an alloantibody reduction between 15% and 20%, and alloantibody levels are reduced by more than 90% with 3 to 6 treatment sessions. However, rebound anti-HLA antibody titers can be detected within 4 weeks after the completion of plasmapheresis or immunoadsorption [67].

Intravenous immunoglobulin

IVIG is a preparation that is produced by separating the gamma globulin fraction of pooled human plasma and is employed to treat various autoimmune diseases or hypogammaglobulinemia. The mechanism of action of IVIG in desensitization treatment remains unclear, but it is considered that IVIG modulates the immune response through various pathways [68]. These complex mechanisms could involve the neutralization of cytokines and antibodies, inhibition of B/T cells through the saturation of Fc receptors, increased regulatory T cells, inhibition of immune complex formation, and inhibition of dendritic cell activity [69]. In a retrospective study that directly compared high-dose IVIG (2 g/kg) and plasmapheresis with low-dose IVIG (100 mg/kg) as desensitization treatment in T-CDC crossmatch-positive recipients, the proportion of patients who achieved crossmatch negativity was 38% in the high-dose IVIG group and 84% in the plasmapheresis with low-dose IVIG group [70]. The incidence of ABMR was 80% in the high-dose IVIG group and 37% in the plasmapheresis with low-dose IVIG group; hence, plasmapheresis with low-dose IVIG was reported to be a better desensitization treatment method [70]. However, in another large-scale study, the comparable success rate of high-dose IVIG and rituximab combination therapy has also been reported [8], and high-dose IVIG can have an advantage over plasmapheresis, as there is no concern regarding the loss of other plasma proteins. Currently, at our center, highly sensitized patients waiting for deceased donors are undergoing high-dose IVIG therapy (1 g/kg/day for 4 days [at day 1, day 2, day 30, day 31] for the removal of anti-HLA antibodies.

Anti-CD20 monoclonal antibody

Rituximab is a monoclonal antibody against CD20 expressed on the surface of B cells and their progenitor cells. It removes B cells present in the peripheral blood and spleen and inhibits their differentiation into plasma cells, thereby suppressing antibody production. In highly sensitized patients, administration of rituximab significantly reduced re-elevation of anti-HLA antibody titers after KT [71]. However, as plasma cells do not express CD20, they cannot be removed with rituximab, and hence, the effect of rituximab may be insufficient in plasma cells that continuously generate antibodies [71]. Nevertheless, to date, rituximab is the most widely used agent to inhibit antibody production during desensitization treatment.

Proteasome inhibitor

Bortezomib is a reversible 26S proteasome inhibitor that inhibits the migration of nuclear factor-kappa B to the cell nucleus, thereby inducing apoptosis of plasma cells [72]. The bone marrow niche resident long-lived plasma cells are known to continuously produce anti-HLA antibodies, which do not express CD20 and are not removed by rituximab [73]. Therefore, desensitization treatment via additional administration of bortezomib targeting plasma cells has been attempted when conventional desensitization treatment does not respond sufficiently, or the antibody titer is markedly high [74]. Carfilzomib is a second-generation irreversible proteasome inhibitor. In a recent study, it was reported that the carfilzomib monotherapy-based desensitization protocol showed acceptable safety and toxicity with significant bone marrow plasma cell depletion and DSA reduction [75]. In highly sensitized patients awaiting a deceased donor, it has been reported that the chance of deceased donor KT was increased by lowering the DSA titer through desensitization using high-dose IVIG, rituximab, and bortezomib [76]. Our center has also been performing this desensitization protocol since 2019, and to date, three deceased donor KTs have been successfully performed.

Novel agents

Eculizumab is a monoclonal antibody against complement C5 that prevents the separation of C5a and C5b, and finally inhibits the formation of the membrane attack complex C5b–C9. Therefore, it could prevent allograft damage by inhibiting the complement cascade. In a study comparing an eculizumab treatment group and the historical control group in crossmatch-positive recipients, the incidence of acute ABMR after KT was significantly lower in the eculizumab treatment group than in the historical control group [77]. However, a large-scale study is still lacking, and further studies are needed.
C1-esterase inhibitor (C1-INH), another complement inhibitor, inhibits the classical and lectin complement pathways by inhibiting C1r and C1s. A randomized phase I/II study compared C1-INH with placebo to prevent acute ABMR in HLAi KT. Twenty recipients who underwent IVIG, rituximab, with or without plasmapheresis before KT, were compared by dividing them into C1-INH and placebo groups. There was no ABMR in the C1-INH group, but one ABMR was reported in the placebo group [78]. It is thought that C1-INH, to some extent, contributes to the prevention of ABMR in highly sensitized patients, but further studies are needed.
Interleukin-6 is a cytokine that induces inflammation through various mechanisms, including the differentiation of B cells into plasma cells. In a previous study using an anti-interleukin-6 receptor blocker (tocilizumab) as desensitization treatment, it was revealed that successful desensitization was possible when tocilizumab was administered to patients who failed to be desensitized following high-dose IVIG and rituximab [79]. Additionally, in KT recipients with chronic ABMR, a significant decrease in the DSA titer was observed when tocilizumab was administered in patients whose DSA titer did not decrease following standard therapy (IVIG + rituximab with or without plasmapheresis). Tocilizumab is expected to contribute not only to desensitization but also to chronic ABMR treatment [80].
Belatacept is a fusion protein that inhibits costimulatory signals in T-cell activation by binding to CD80/CD86 molecules of antigen-presenting cells. It is attracting attention as a new maintenance immunosuppressant [81]. In a recent human in vitro study, it was reported that belatacept inhibits plasmablast differentiation and immunoglobulin production by acting independently on B cells [82]. In a recent retrospective cohort study of highly sensitized recipients (cPRA ≥ 98%), the belatacept group showed a significant decrease in HLA class I antibodies, suggesting that belatacept may be an option for desensitization.
The IgG-degrading enzyme of Streptococcus pyogenes (IdeS) is a cysteine endopeptidase that divides IgG into F(ab’)2 and Fc to neutralize antibodies. It reportedly acts rapidly and neutralizes most of the IgG within 4 hours of administration [83]. In the United States and Sweden, 24 out of 25 highly sensitized recipients reported successful deceased donor KT after IdeS administration. One patient presented with hyperacute rejection, but it was presumed to be caused by unrecognized IgM, IgA, or non-HLA antibodies as no detectable IgG DSAs were observed after IdeS administration [84]. IdeS is currently being investigated in a clinical trial and is expected to play an important role as a desensitization tool in highly sensitized recipients in the future.

Alternative to desensitization (kidney paired donation)

Kidney paired donation (KPD) is a method of performing KT without desensitization in highly sensitized recipients through compatible recipient-donor pair matching in recipient and donor pool [85]. The KPD program has been developed in many countries over the past 30 years, and recent data from KPD registries in Australia and Canada reported that the KT match rates of recipients with cPRA between 50% and 96% and recipients with cPRA < 50% were similar [86]. In a recent study comparing the KPD network program (National Kidney Registry) and all national transplants registry (United Network for Organ Sharing) in the United States, KT performed through the KPD network program showed a higher proportion of retransplantation and highly sensitized recipients (cPRA > 80%) [87]. In a study that analyzed how highly sensitized recipients received KT in the United States from 2009 to 2017, it was observed that living donor KT through KPD gradually increased, which is thought to be due to the development of the KPD program [88].

Allograft and patient outcomes

Table 3 summarizes the studies on HLAi KT to date. As highly sensitized recipients present preexisting alloantibodies before KT, the ABMR incidence was higher than in those without alloantibodies [89]. Consequently, HLAi KT has a significantly lower allograft survival rate than HLA-compatible KT (HLAc KT), which has been reported to be significantly reduced from 1 year of transplantation (89.9% in HLAi KT vs. 97.6% in HLAc KT) [90].
HLAi KT poses concerns regarding increased rejection and worse allograft outcomes, as well as increased infection and malignancy risks due to desensitization treatment. Until now, most studies on the risk of desensitization treatment have been short-term studies; hence, the impact of the long-term risk of desensitization treatment remains unclear. Inconsistent results have been reported on the effect of desensitization treatment on infection risk [9194]. Kahwaji et al. [93] reported that there were no differences in bacterial, viral, and fungal infection rates between the rituximab with high-dose IVIG group and the non-administered group over 18 months after transplantation. Conversely, Ko et al. [92] reported that desensitization treatment was an independent risk factor for infection-related mortality (adjusted hazard ratio, 3.40; p = 0.002) in a nationwide cohort study in the Republic of Korea. In a recent phase 2 randomized controlled trial comparing the eculizumab-added group and the standard desensitization (plasmapheresis + IVIG or plasmapheresis alone) group in HLAi KT, it was reported that the overall infection rate was numerically higher in the eculizumab-added group (62.7% in the eculizumab group vs. 49.0% in the standard desensitization group) [95]. Few studies have highlighted the malignancy risk associated with desensitization treatment in HLAi KT. In previous studies, nonmelanoma skin cancer and lymphoproliferative disorder were common in the desensitization treatment group [96], with urothelial carcinoma particularly common in the Asian population [97].
DSA titers at the time of transplantation and allograft survival were found to be correlated [63,98]. In an observational study of living donor KT patients at 22 centers of United States, the overall graft survival rates at 1 year and 5 years of transplantation were highest in HLAc KT, the second in the positive SAB (Luminex) assay with negative FCXM (PLNF) group, the third in the positive FCXM with negative CDC crossmatch (PFNC) group, and the lowest in the positive CDC crossmatch (PCC) group. The authors reported that the higher the intensity of sensitization before transplantation, the higher the overall allograft loss rate (Fig. 4) [98]. It is postulated that in addition to the DSA titer, DSA characteristics can affect the allograft outcome. In a patient in whom the C1q binding DSA was converted to negative, but CDC crossmatch remained positive at the time of transplantation, a favorable outcome was observed without evidence of antibody-mediated injury after transplantation [74]. In the future, additional studies on allograft outcomes according to the characteristics of DSA (C1q binding, C3d binding, or IgG subclass) are necessary.
Although HLAi KT has a higher ABMR incidence and lower allograft survival rate than HLAc KT, HLAi KT has advantages over continuing dialysis while waiting for deceased donor KT. Montgomery et al. [62] reported that the HLAi KT group showed more than twice the survival benefit when compared with the dialysis-only group and the dialysis-or-deceased donor KT group in a single-center study. Similarly, Orandi et al. [98] reanalyzed this through the national cohort and reported that HLAi KT demonstrated a better long-term survival rate when compared with dialysis only and dialysis-or-deceased donor KT groups. Although HLAi KT costs approximately $30,000 more than HLAc KT in terms of economics [99], compared to continuing dialysis, HLAi KT increases the quality of life and patient survival and reduces the need for dialysis, making it more cost-effective than continuing dialysis [100].

Our center’s experiences and outcomes

At our center, a total of 137 HLAi KTs were performed from 2010 to 2020. Based on immunologic stratification, 41 patients (29.9%) were PCC, 46 (33.6%) were PFNC, and 50 (36.5%) were PLNF. Among these patients, 41 patients (29.9%) had concomitant ABO incompatibility. Desensitization treatment was performed with rituximab and plasmapheresis with low-dose IVIG according to our center protocol (Fig. 3A). In five patients, the bortezomib-based protocol was performed (Fig. 3B). Antithymocyte globulin was used in 62.0% (85 of 137) and basiliximab in 38.0% (52 of 137) as induction therapy before transplantation. In the posttransplant allograft outcome, de novo DSAs occurred in 10.2% (14 of 137) of patients, and preexisting DSA rebound occurred in 21.9% (30 of 137) of patients. On analyzing biopsy-proven rejection, the acute rejection rate was 30.7% (42 of 137) and the chronic rejection rate was 11.7% (16 of 137). In sub-analysis, the acute T cell-mediated rejection (TCMR) rate was 14.6% (20 of 137), acute ABMR rate was 20.4% (28 of 137), chronic active TCMR rate was 0.7% (1 of 137), and the chronic active ABMR rate was 10.9% (15 of 137). The median follow-up duration was 44.8 months, death censored graft loss rate was 9.5% (13 of 137), overall graft loss rate was 13.9% (19 of 137), and the patient death rate was 8.0% (11 of 137). Following analysis according to the immunological risk group, the overall graft loss rate was 19.5% (8 of 41) in the PCC group, 15.2% (7 of 46) in the PFNC group, and 8.0% (4 of 50) in the PLNF group. It should be considered that the median follow-up duration was 44.8 months, but the overall graft survival of HLAi KT in crossmatch-positive patients at our center was marginally improved compared to previous studies (Table 3).

Conclusion

Highly sensitized recipients are less likely to have deceased donor KT opportunities than non-sensitized recipients. Moreover, in these patients, living donor KT after desensitization treatment reportedly presents a better patient survival rate than that observed with continuing dialysis while waiting for deceased donor KT. Therefore, if a potential donor is available, it is necessary to actively perform HLAi KT. Furthermore, owing to great advances in SAB assays and desensitization treatment in the field of transplantation, it is expected that patient prognosis can be improved through appropriate immunologic risk stratification and desensitization treatment. Moreover, many researches are ongoing in the field of transplant immunity and new agents of desensitization treatment. Based on these findings, in the future, we anticipate that safe KT will be performed even in highly sensitized recipients, thereby actively improving their prognosis.

Notes

Conflict of interest

All authors have no conflicts of interest to declare.

Authors’ contributions

Investigation, Data curation: EJK, BHC

Writing–original draft: YP, CWY

Writing–review & editing: YP, CWY

All authors read and approved the final manuscript.

Figure 1.

Screening of alloantibodies in recipients in Seoul St. Mary’s Hospital.

AHG, anti-human globulin; CDC, complement-dependent cytotoxicity; DSA, donor-specific antibody; FCXM, flow cytometry crossmatch. Luminex assay, Luminex Corp., Austin, TX, USA.
j-krcp-21-012f1.jpg
Figure 2.

Basic concept of desensitization treatment in highly sensitized recipients.

IVIG, intravenous immunoglobulin.
j-krcp-21-012f2.jpg
Figure 3.

The desensitization protocol of Seoul St. Mary’s Hospital.

(A) Standard desensitization protocol. The target goals were T-CDC and T-FCXM-negative conversion, DSA MFI titer less than 3,000, and C1q binding assay negative conversion. (B) Bortezomib-based desensitization protocol. If T-CDC or T-AHG is positive, or if there is no adequate DSA reduction after three or more PP/IVIG sessions, bortezomib-based protocol is performed. The target goals were T-CDC and T-FCXM-negative conversion, DSA MFI titer less than 3,000, and C1q binding assay negative conversion.
AHG, anti-human globulin; ATG, antithymocyte globulin; CDC, complement-dependent cytotoxicity; D, day; DSA, donor-specific antibody; FCXM, flow cytometry crossmatch; KT, kidney transplantation; MFI, median fluorescence intensity; IVIG, intravenous immunoglobulin; PP, plasmapheresis; XM, crossmatch.
aBasiliximab is administered at 20 mg/day on days 0 and 4. ATG is administered at a dose of 1.5 mg/kg/day from day 0 to day 4 for 5 days.
j-krcp-21-012f3.jpg
Figure 4.

Overall allograft survival rate by crossmatch and SAB assay results at the time of transplantation.

Modified from the article of Orandi et al. (Am J Transplant 2014;14:1573-1580).
PCC, positive complement cytotoxicity; PFNC, positive flow cytometry/negative complement cytotoxicity; PLNF, positive SAB (Luminex, Luminex Corp., Austin, TX, USA) assay/negative flow cytometry; SAB, single antigen bead.
j-krcp-21-012f4.jpg
Table 1.
Interpretation of CDC crossmatch and FCXM results
CDC-AHG
FCXM
Interpretation
T cell B cell T cell B cell
+ + + + High-titer IgG HLA class I and/or IgG HLA class II DSA
High risk of hyperacute rejection
+ + Probably not IgG HLA class I DSA
Possible T cell-specific antibody
Further test needed
+ + + Low-titer IgG HLA class I and/or IgG HLA class II DSA
+ + Low-titer IgG HLA class I and/or IgG HLA class II DSA
+ + IgG HLA class II and/or low-titer IgG HLA class I DSA
Autoantibodies
–/+ + Possible IgM autoantibodies
Prior to IgG HLA class I DSA
DTT can be helpful
+ IgG HLA class II DSA and/or low-titer IgG HLA class I DSA
Autoantibodies
Not detected by crossmatch
SPA may detect low-level of DSA

AHG, anti-human globulin; CDC, complement-dependent cytotoxicity; DSA, donor-specific antibody; DTT, dithiothreitol; FCXM, flow cytometry crossmatch; HLA, human leukocyte antigen; SPA, solid-phase immunoassay.

Table 2.
Immunologic risk stratification by CDC crossmatch, FCXM, and SAB assay
CDC
FCXM
SAB assay (IgG HLA class I or class II DSA) Current or historical Immunologic risk
T cell B cell T cell B cell
+ + NA NA HLA class I Current High
+ NA NA HLA class II Current High
+ NA NA HLA class I Current Intermediate
+ + NA NA HLA class I Historical Intermediate
+ NA NA HLA class II Historical Intermediate
+ NA NA HLA class I Historical Intermediate
+ + HLA class I Current Intermediate
+ HLA class II Current Intermediate
+ HLA class I Current Intermediate
+ + HLA class I Historical Intermediate
+ HLA class II Historical Intermediate
+ HLA class I Historical Low
HLA class I Both Low
HLA class II Both Low
Negative Both Standard

High immunologic risk: This is generally contraindicated for KT. Desensitization is absolutely necessary for KT. Intermediate immunologic risk: If there are other options, consider other options, such as when the patient has a short waiting time for deceased donor KT and low calculated panel reactive antibody that makes HLAc KT possible. Appropriate desensitization treatment and active immunosuppression are required, clinical attention is required at the time of transplantation, and antibody monitoring after KT is necessary. Low immunologic risk: It has been reported that the incidence of rejection increases owing to a low donor-specific antibody titer and the allograft function at 1 year of transplantation was lower than that of HLAc KT. However, there is insufficient evidence that long-term allograft survival is inferior to that of HLAc KT.

CDC, complement-dependent cytotoxicity; FCXM, flow cytometry crossmatch; HLA, human leukocyte antigen; HLAc KT, HLA-compatible KT; IgG, immunoglobulin G; KT, kidney transplantation; NA, not applicable; SAB, single antigen beads.

Table 3.
Current studies’ outcomes of HLAi KT
Study Year of publication No. of patients Desensitization method Acute rejection (%) Graft survival (%) Patient survival (%) Follow-up (mo)
Sonnenday et al.a 2002 18 (X+) PP + LD IVIG 27.8 94.4 100 17
Thielke et al.b 2009 57 (X+) PP + LD IVIG with/without rituximab 43.0 93.0 95.0 12
Vo et al.c 2010 76 (X+) HD IVIG + rituximab 37.0 84.0 95.0 24
Montgomery et al. [11] 2011 211 (X+) PP + LD IVIG - - 90.6 12
85.7 36
80.6 60
Bentall et al.d 2013 102 (X+) PP + LD IVIG + splenectomy (n = 16) 37 70.7 83.5 60
PP + LD IVIG (n = 48)
HD IVIG (n = 21)
None (n = 17)
Riella et al. [96] 2014 39 (X+) PP + LD IVIG with/without rituximab - 94 95 12
- 88 95 36
61.5 84 86 60
Orandi et al.e 2014 304 (PCC) - - 60.1 (PCC) 80.9 (PCC) 60
536 (PFNC) 71.2 (PFNC) 87.1 (PFNC)
185 (PLNF) 79.8 (PLNF) 90.4 (PLNF)
Okada et al.f 2018 36 (X –D+) Rituximab (2 doses) (X–D+) 19.4 (X–D+)h 97.2 (X–D+) 100 (X–D+) 48
15 (X+D+) PP + LD IVIG + rituximab (X+D+) 60.0 (X+D+)h 86.7 (X+D+) 93.3 (X+D+)
Kwon et al.g 2019 176 (ABOc/X+) PP + LD IVIG + rituximab 11.7 (ABOc)i 95.5 (ABOc) 98.6 (ABOc) 60
87 (ABOi/X+) 25.5 (ABOi)i 89.9 (ABOi) 80.9 (ABOi)
Our center - 41 (PCC) PP + LD IVIG + rituximab 41.5 (PCC) 80.5 (PCC) 92.7 (PCC) 45
46 (PFNC) (with/without bortezomib) 32.6 (PFNC) 84.8 (PFNC) 89.1 (PFNC)
50 (PLNF) 20.0 (PLNF) 92.0(PLNF) 94.0 (PLNF)

a Transplant Proc 2002;1614-1616.,

b Transplantation 2009;87:268-273.,

c Transplantation 2010;89:1095-1102.,

d Am J Transplant 2013;13:76-85.,

e N Engl J Med 2016;374:940-950.,

f Transpl Int 2018;31:1008-1017.,

g Sci Rep 2019;9:18229.

h Incidence of acute rejection within 3 months of transplantation,

i Incidence of acute rejection within 1 year of transplantation.

ABOc, ABO compatible; ABOi, ABO-incompatible; D, donor-specific antibody (Luminex, Corp., Austin, TX, USA); HD, high-dose; HLA, human leukocyte antigen; HLAi KT, HLA-incompatible KT; IVIG, intravenous immunoglobulin; LD, low-dose; PCC, positive complement cytotoxicity; PFNC, positive flow cytometry/negative complement cytotoxicity; PLNF, positive single antigen bead (Luminex, Luminex Corp., Austin, TX, USA) assay/negative flow cytometry; PP, plasmapheresis; X, crossmatch.

References

1. Wolfe RA, Ashby VB, Milford EL, et al. Comparison of mortality in all patients on dialysis, patients on dialysis awaiting transplantation, and recipients of a first cadaveric transplant. N Engl J Med 1999;341:1725–1730.
crossref pmid
2. Satayathum S, Pisoni RL, McCullough KP, et al. Kidney transplantation and wait-listing rates from the international Dialysis Outcomes and Practice Patterns Study (DOPPS). Kidney Int 2005;68:330–337.
crossref pmid
3. Jeon HJ, Bae HJ, Ham YR, et al. Outcomes of end-stage renal disease patients on the waiting list for deceased donor kidney transplantation: a single-center study. Kidney Res Clin Pract 2019;38:116–123.
crossref pmid pmc
4. Korean Network for Organ Sharing (KONOS). KONOS annual report; updated September 8, 2020 [Internet]. Seoul (Korea): KONOS; c2020 [cited 2020 Dec 11]. Available from: https://www.konos.go.kr/konosis/common/bizlogic.jsp.
5. Kwon KH, Moon JI, Chang HJ, et al. The results of renal transplantation after lymphocyte cross-match negative conversion by combination therapy with plasmapheresis, intravenous gamma globulin and potent immunosuppresants in patients with positive LCM. J Korean Soc Transplant 2002;16:172–177.
6. Patel R, Terasaki PI. Significance of the positive crossmatch test in kidney transplantation. N Engl J Med 1969;280:735–739.
crossref pmid
7. Taylor CJ, Kosmoliaptsis V, Summers DM, Bradley JA. Back to the future: application of contemporary technology to long-standing questions about the clinical relevance of human leukocyte antigen-specific alloantibodies in renal transplantation. Hum Immunol 2009;70:563–568.
crossref pmid
8. Vo AA, Lukovsky M, Toyoda M, et al. Rituximab and intravenous immune globulin for desensitization during renal transplantation. N Engl J Med 2008;359:242–251.
crossref pmid
9. Reinsmoen NL, Lai CH, Vo A, et al. Acceptable donor-specific antibody levels allowing for successful deceased and living donor kidney transplantation after desensitization therapy. Transplantation 2008;86:820–825.
crossref pmid
10. Dyer PA, Claas FH, Doxiadis II, Glotz D, Taylor CJ. Minimising the clinical impact of the alloimmune response through effective histocompatibility testing for organ transplantation. Transpl Immunol 2012;27:83–88.
crossref pmid
11. Chandraker A, Sayegh MH, Singh AK. Core concepts in renal transplantation. Boston (MA): Springer Science & Business Media; 2012.
12. Hopkins KA. The basic lymphocyte microcytotoxicity test. In: Phelan D, Mickelson EM, Noreen HS, The American Society for Histocompatibility and Immunogenetics (ASHI) laboratory manual, Vol I. B. 1.1. Lexana (KS): ASHI; 1993.
13. Amos DB, Bashir H, Boyle W, MacQueen M, Tiilikainen A. A simple micro cytotoxicity test. Transplantation 1969;7:220–223.
crossref pmid
14. Fuller TC, Phelan D, Gebel HM, Rodey GE. Antigenic specificity of antibody reactive in the antiglobulin-augmented lymphocytotoxicity test. Transplantation 1982;34:24–29.
crossref pmid
15. Okuno T, Kondelis N. Evaluation of dithiothreitol (DTT) for inactivation of IgM antibodies. J Clin Pathol 1978;31:1152–1155.
crossref pmid pmc
16. Sigdel TK, Sarwal MM. Moving beyond HLA: a review of nHLA antibodies in organ transplantation. Hum Immunol 2013;74:1486–1490.
crossref pmid pmc
17. Schlaf G, Pollok-Kopp B, Schabel E, Altermann W. Artificially positive crossmatches not leading to the refusal of kidney donations due to the usage of adequate diagnostic tools. Case Rep Transplant 2013;2013:746395.
crossref pmid pmc
18. Vieira CA, Agarwal A, Book BK, et al. Rituximab for reduction of anti-HLA antibodies in patients awaiting renal transplantation: 1. Safety, pharmacodynamics, and pharmacokinetics. Transplantation 2004;77:542–548.
crossref pmid
19. Garovoy M, Rheinschmidt MA, Bigos M, et al. Flow cytometry analysis: a high technology crossmatch technique facilitating transplantation. Transplant Proc 1983;15:1939–1944.
20. Karpinski M, Rush D, Jeffery J, et al. Flow cytometric crossmatching in primary renal transplant recipients with a negative anti-human globulin enhanced cytotoxicity crossmatch. J Am Soc Nephrol 2001;12:2807–2814.
crossref pmid
21. Hamrick C, Lebeck L. Flow cytometric T and B cell crossmatching. ASHI laboratory manual. 4th ed. Philadelphia (PA): American Society for Histocompatibility and Immunogenetics; 2000. p. 41–45.
22. Hetrick SJ, Schillinger KP, Zachary AA, Jackson AM. Impact of pronase on flow cytometric crossmatch outcome. Hum Immunol 2011;72:330–336.
crossref pmid
23. Guillaume N. Improved flow cytometry crossmatching in kidney transplantation. HLA 2018;92:375–383.
crossref pmid
24. 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;95:19–47.
crossref pmid
25. Terasaki PI, Ozawa M, Castro R. Four-year follow-up of a prospective trial of HLA and MICA antibodies on kidney graft survival. Am J Transplant 2007;7:408–415.
crossref pmid
26. Lachmann N, Todorova K, Schulze H, Schönemann C. Luminex(®) and its applications for solid organ transplantation, hematopoietic stem cell transplantation, and transfusion. Transfus Med Hemother 2013;40:182–189.
crossref pmid pmc
27. Reed EF, Rao P, Zhang Z, et al. Comprehensive assessment and standardization of solid phase multiplex-bead arrays for the detection of antibodies to HLA. Am J Transplant 2013;13:1859–1870.
crossref pmid pmc
28. Tait BD. Detection of HLA antibodies in organ transplant recipients: triumphs and challenges of the solid phase bead assay. Front Immunol 2016;7:570.
crossref pmid pmc
29. Schnaidt M, Weinstock C, Jurisic M, Schmid-Horch B, Ender A, Wernet D. HLA antibody specification using single-antigen beads: a technical solution for the prozone effect. Transplantation 2011;92:510–515.
crossref pmid
30. Tambur AR, Campbell P, Claas FH, et al. Sensitization in transplantation: assessment of risk (STAR) 2017 Working Group Meeting Report. Am J Transplant 2018;18:1604–1614.
crossref pmid
31. Kamburova EG, Wisse BW, Joosten I, et al. Differential effects of donor-specific HLA antibodies in living versus deceased donor transplant. Am J Transplant 2018;18:2274–2284.
crossref pmid pmc
32. Süsal C, Ovens J, Mahmoud K, et al. No association of kidney graft loss with human leukocyte antigen antibodies detected exclusively by sensitive Luminex single-antigen testing: a Collaborative Transplant Study report. Transplantation 2011;91:883–887.
crossref pmid
33. Lee H, Han E, Choi AR, et al. Clinical impact of complement (C1q, C3d) binding de novo donor-specific HLA antibody in kidney transplant recipients. PLoS One 2018;13:e0207434.
crossref pmid pmc
34. Keith DS, Vranic GM. Approach to the highly sensitized kidney transplant candidate. Clin J Am Soc Nephrol 2016;11:684–693.
crossref pmid pmc
35. Taylor CJ, Kosmoliaptsis V, Sharples LD, et al. Ten-year experience of selective omission of the pretransplant crossmatch test in deceased donor kidney transplantation. Transplantation 2010;89:185–193.
crossref pmid
36. Crespo M, Zárraga S, Alonso Á, et al. Monitoring of donor-specific anti-HLA antibodies and management of immunosuppression in kidney transplant recipients: an evidence-based expert paper. Transplantation 2020;104(8 Suppl 2):S1–S12.
crossref
37. Tambur AR, Campbell P, Chong AS, et al. Sensitization in transplantation: Assessment of risk (STAR) 2019 Working Group Meeting Report. Am J Transplant 2020;20:2652–2668.
crossref pmid pmc
38. Sicard A, Ducreux S, Rabeyrin M, et al. Detection of C3d-binding donor-specific anti-HLA antibodies at diagnosis of humoral rejection predicts renal graft loss. J Am Soc Nephrol 2015;26:457–467.
crossref pmid
39. Loupy A, Lefaucheur C, Vernerey D, et al. Complement-binding anti-HLA antibodies and kidney-allograft survival. N Engl J Med 2013;369:1215–1226.
crossref pmid
40. Tambur AR, Herrera ND, Haarberg KM, et al. Assessing antibody strength: comparison of MFI, C1q, and titer information. Am J Transplant 2015;15:2421–2430.
crossref pmid
41. Yell M, Muth BL, Kaufman DB, Djamali A, Ellis TM. C1q binding activity of de novo donor-specific HLA antibodies in renal transplant recipients with and without antibody-mediated rejection. Transplantation 2015;99:1151–1155.
crossref pmid
42. Wiebe C, Gareau AJ, Pochinco D, et al. Evaluation of C1q status and titer of de novo donor-specific antibodies as predictors of allograft survival. Am J Transplant 2017;17:703–711.
crossref pmid
43. Vidarsson G, Dekkers G, Rispens T. IgG subclasses and allotypes: from structure to effector functions. Front Immunol 2014;5:520.
crossref pmid pmc
44. 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;27:293–304.
crossref pmid
45. Arnold ML, Ntokou IS, Doxiadis II, Spriewald BM, Boletis JN, Iniotaki AG. Donor-specific HLA antibodies: evaluating the risk for graft loss in renal transplant recipients with isotype switch from complement fixing IgG1/IgG3 to noncomplement fixing IgG2/IgG4 anti-HLA alloantibodies. Transpl Int 2014;27:253–261.
crossref pmid
46. Jackson AM, Kanaparthi S, Burrell BE, et al. IgG4 donor-specific HLA antibody profile is associated with subclinical rejection in stable pediatric liver recipients. Am J Transplant 2020;20:513–524.
crossref pmid
47. Jin YP, Valenzuela NM, Zhang X, Rozengurt E, Reed EF. HLA class II-triggered signaling cascades cause endothelial cell proliferation and migration: relevance to antibody-mediated transplant rejection. J Immunol 2018;200:2372–2390.
crossref pmid pmc
48. Arnold ML, Kainz A, Hidalgo LG, et al. Functional Fc gamma receptor gene polymorphisms and donor-specific antibody-triggered microcirculation inflammation. Am J Transplant 2018;18:2261–2273.
crossref pmid
49. Crotty S, Ahmed R. Immunological memory in humans. Semin Immunol 2004;16:197–203.
crossref pmid
50. Lúcia M, Luque S, Crespo E, et al. Preformed circulating HLA-specific memory B cells predict high risk of humoral rejection in kidney transplantation. Kidney Int 2015;88:874–887.
crossref pmid
51. Karahan GE, de Vaal YJ, Roelen DL, Buchli R, Claas FH, Heidt S. Quantification of HLA class II-specific memory B cells in HLA-sensitized individuals. Hum Immunol 2015;76:129–136.
crossref pmid
52. Heeger PS, Greenspan NS, Kuhlenschmidt S, et al. Pretransplant frequency of donor-specific, IFN-gamma-producing lymphocytes is a manifestation of immunologic memory and correlates with the risk of posttransplant rejection episodes. J Immunol 1999;163:2267–2275.
pmid
53. Cortes-Cerisuelo M, Laurie SJ, Mathews DV, et al. Increased pretransplant frequency of CD28+ CD4+ TEM predicts belatacept-resistant rejection in human renal transplant recipients. Am J Transplant 2017;17:2350–2362.
crossref pmid pmc
54. Cano-Romero FL, Laguna Goya R, Utrero-Rico A, et al. Longitudinal profile of circulating T follicular helper lymphocytes parallels anti-HLA sensitization in renal transplant recipients. Am J Transplant 2019;19:89–97.
crossref pmid
55. Copley HC, Elango M, Kosmoliaptsis V. Assessment of human leukocyte antigen immunogenicity: current methods, challenges and opportunities. Curr Opin Organ Transplant 2018;23:477–485.
crossref pmid pmc
56. Duquesnoy RJ. Clinical usefulness of HLAMatchmaker in HLA epitope matching for organ transplantation. Curr Opin Immunol 2008;20:594–601.
crossref pmid
57. Wiebe C, Kosmoliaptsis V, Pochinco D, Taylor CJ, Nickerson P. A comparison of HLA molecular mismatch methods to determine HLA immunogenicity. Transplantation 2018;102:1338–1343.
crossref pmid pmc
58. Geneugelijk K, Niemann M, Drylewicz J, et al. PIRCHE-II is related to graft failure after kidney transplantation. Front Immunol 2018;9:321.
crossref pmid pmc
59. Wiebe C, Pochinco D, Blydt-Hansen TD, et al. Class II HLA epitope matching-A strategy to minimize de novo donor-specific antibody development and improve outcomes. Am J Transplant 2013;13:3114–3122.
crossref pmid
60. Tambur AR, McDowell H, Hod-Dvorai R, Abundis MA, Pinelli DF. The quest to decipher HLA immunogenicity: telling friend from foe. Am J Transplant 2019;19:2910–2925.
crossref pmid
61. Jordan SC, Pescovitz MD. Presensitization: the problem and its management. Clin J Am Soc Nephrol 2006;1:421–432.
crossref pmid
62. Montgomery RA, Lonze BE, King KE, et al. Desensitization in HLA-incompatible kidney recipients and survival. N Engl J Med 2011;365:318–326.
crossref pmid
63. Chung BH, Choi BS, Oh EJ, et al. Clinical impact of the baseline donor-specific anti-human leukocyte antigen antibody measured by Luminex single antigen assay in living donor kidney transplant recipients after desensitization therapy. Transpl Int 2014;27:49–59.
crossref pmid
64. Yang KS, Jeon H, Park Y, et al. Use of bortezomib as anti-humoral therapy in kidney transplantation. J Korean Med Sci 2014;29:648–651.
crossref pmid pmc
65. Hwang SD, Chung BH, Oh EJ, et al. Effect of pretransplant rituximab use on posttransplant clinical outcomes in patients with high panel reactive antibody scores. Nephron 2015;130:239–244.
crossref pmid
66. Marlu R, Bennani HN, Seyve L, et al. Effect of immunoadsorption alone or combined with membrane filtration on hemostasis parameters. J Clin Apher 2020;35:444–452.
crossref pmid
67. Hakim RM, Milford E, Himmelfarb J, Wingard R, Lazarus JM, Watt RM. Extracorporeal removal of anti-HLA antibodies in transplant candidates. Am J Kidney Dis 1990;16:423–431.
crossref pmid
68. Kazatchkine MD, Kaveri SV. Immunomodulation of autoimmune and inflammatory diseases with intravenous immune globulin. N Engl J Med 2001;345:747–755.
crossref pmid
69. Schwab I, Nimmerjahn F. Intravenous immunoglobulin therapy: how does IgG modulate the immune system? Nat Rev Immunol 2013;13:176–189.
crossref pmid
70. Stegall MD, Gloor J, Winters JL, Moore SB, Degoey S. A comparison of plasmapheresis versus high-dose IVIG desensitization in renal allograft recipients with high levels of donor specific alloantibody. Am J Transplant 2006;6:346–351.
crossref pmid
71. Jackson AM, Kraus ES, Orandi BJ, Segev DL, Montgomery RA, Zachary AA. A closer look at rituximab induction on HLA antibody rebound following HLA-incompatible kidney transplantation. Kidney Int 2015;87:409–416.
crossref pmid
72. Frankland-Searby S, Bhaumik SR. The 26S proteasome complex: an attractive target for cancer therapy. Biochim Biophys Acta 2012;1825:64–76.
crossref pmid
73. DiLillo DJ, Hamaguchi Y, Ueda Y, et al. Maintenance of long-lived plasma cells and serological memory despite mature and memory B cell depletion during CD20 immunotherapy in mice. J Immunol 2008;180:361–371.
crossref pmid
74. Lee J, Park BG, Jeong HS, et al. Successful kidney transplantation across a positive complement-dependent cytotoxicity crossmatch by using C1q assay-directed, bortezomib-assisted desensitization: a case report. Medicine (Baltimore) 2017;96:e8145.
crossref pmid pmc
75. Tremblay S, Driscoll JJ, Rike-Shields A, et al. A prospective, iterative, adaptive trial of carfilzomib-based desensitization. Am J Transplant 2020;20:411–421.
crossref pmid
76. Jeong JC, Jambaldorj E, Kwon HY, et al. Desensitization using bortezomib and high-dose immunoglobulin increases rate of deceased donor kidney transplantation. Medicine (Baltimore) 2016;95:e2635.
crossref pmid pmc
77. Cornell LD, Schinstock CA, Gandhi MJ, Kremers WK, Stegall MD. Positive crossmatch kidney transplant recipients treated with eculizumab: outcomes beyond 1 year. Am J Transplant 2015;15:1293–1302.
crossref pmid
78. Vo AA, Zeevi A, Choi J, et al. A phase I/II placebo-controlled trial of C1-inhibitor for prevention of antibody-mediated rejection in HLA sensitized patients. Transplantation 2015;99:299–308.
crossref pmid
79. Vo AA, Choi J, Kim I, et al. A phase I/II trial of the interleukin-6 receptor-specific humanized monoclonal (tocilizumab) + intravenous immunoglobulin in difficult to desensitize patients. Transplantation 2015;99:2356–2363.
crossref pmid
80. Shin BH, Everly MJ, Zhang H, et al. Impact of tocilizumab (anti-IL-6R) treatment on immunoglobulins and anti-HLA antibodies in kidney transplant patients with chronic antibody-mediated rejection. Transplantation 2020;104:856–863.
crossref pmid
81. Perez CP, Patel N, Mardis CR, Meadows HB, Taber DJ, Pilch NA. Belatacept in solid organ transplant: review of current literature across transplant types. Transplantation 2018;102:1440–1452.
pmid
82. Leibler C, Thiolat A, Hénique C, et al. Control of humoral response in renal transplantation by belatacept depends on a direct effect on B cells and impaired T follicular helper-B cell crosstalk. J Am Soc Nephrol 2018;29:1049–1062.
crossref pmid pmc
83. Wenig K, Chatwell L, von Pawel-Rammingen U, Björck L, Huber R, Sondermann P. Structure of the streptococcal endopeptidase IdeS, a cysteine proteinase with strict specificity for IgG. Proc Natl Acad Sci U S A 2004;101:17371–17376.
crossref pmid pmc
84. Jordan SC, Lorant T, Choi J, et al. IgG Endopeptidase in highly sensitized patients undergoing transplantation. N Engl J Med 2017;377:442–453.
crossref pmid
85. Kwak JY, Kwon OJ, Lee KS, Kang CM, Park HY, Kim JH. Exchange-donor program in renal transplantation: a single-center experience. Transplant Proc 1999;31:344–345.
crossref pmid
86. Ferrari P, Weimar W, Johnson RJ, Lim WH, Tinckam KJ. Kidney paired donation: principles, protocols and programs. Nephrol Dial Transplant 2015;30:1276–1285.
crossref pmid
87. Flechner SM, Thomas AG, Ronin M, et al. The first 9 years of kidney paired donation through the National Kidney Registry: characteristics of donors and recipients compared with National Live Donor Transplant Registries. Am J Transplant 2018;18:2730–2738.
crossref pmid pmc
88. Jackson KR, Motter JD, Kernodle A, et al. How do highly sensitized patients get kidney transplants in the United States?: trends over the last decade. Am J Transplant 2020;20:2101–2112.
crossref pmid pmc
89. Jeong HJ. Diagnosis of renal transplant rejection: Banff classification and beyond. Kidney Res Clin Pract 2020;39:17–31.
crossref pmid pmc
90. Haririan A, Nogueira J, Kukuruga D, et al. Positive cross-match living donor kidney transplantation: longer-term outcomes. Am J Transplant 2009;9:536–542.
crossref pmid
91. Chung BH, Yun JT, Ha SE, et al. Combined use of rituximab and plasmapheresis pre-transplant increases post-transplant infections in renal transplant recipients with basiliximab induction therapy. Transpl Infect Dis 2013;15:559–568.
crossref pmid
92. Ko EJ, Yu JH, Yang CW, Chung BH; Korean Organ Transplantation Registry Study Group. Clinical outcomes of ABO- and HLA-incompatible kidney transplantation: a nationwide cohort study. Transpl Int 2017;30:1215–1225.
crossref pmid
93. Kahwaji J, Sinha A, Toyoda M, et al. Infectious complications in kidney-transplant recipients desensitized with rituximab and intravenous immunoglobulin. Clin J Am Soc Nephrol 2011;6:2894–2900.
crossref pmid pmc
94. Ko EJ, Yang J, Ahn C, et al. Clinical outcomes of kidney transplantation in older end-stage renal disease patients: a nationwide cohort study. Geriatr Gerontol Int 2019;19:392–398.
crossref pmid
95. Marks WH, Mamode N, Montgomery RA, et al. Safety and efficacy of eculizumab in the prevention of antibody-mediated rejection in living-donor kidney transplant recipients requiring desensitization therapy: a randomized trial. Am J Transplant 2019;19:2876–2888.
crossref pmid pmc
96. Riella LV, Safa K, Yagan J, et al. Long-term outcomes of kidney transplantation across a positive complement-dependent cytotoxicity crossmatch. Transplantation 2014;97:1247–1252.
crossref pmid
97. Yang CY, Lee CY, Yeh CC, Tsai MK. Renal transplantation across the donor-specific antibody barrier: graft outcome and cancer risk after desensitization therapy. J Formos Med Assoc 2016;115:426–433.
crossref pmid
98. Orandi BJ, Garonzik-Wang JM, Massie AB, et al. Quantifying the risk of incompatible kidney transplantation: a multicenter study. Am J Transplant 2014;14:1573–1580.
crossref pmid
99. Axelrod D, Lentine KL, Schnitzler MA, et al. The incremental cost of incompatible living donor kidney transplantation: a national cohort analysis. Am J Transplant 2017;17:3123–3130.
crossref pmid
100. Axelrod DA, Schnitzler MA, Xiao H, et al. An economic assessment of contemporary kidney transplant practice. Am J Transplant 2018;18:1168–1176.
crossref pmid


ABOUT
BROWSE ARTICLES
EDITORIAL POLICY
FOR CONTRIBUTORS
Editorial Office
#301, (Miseung Bldg.) 23, Apgujenog-ro 30-gil, Gangnam-gu, Seoul 06022, Korea
Tel: +82-2-3486-8736    Fax: +82-2-3486-8737    E-mail: registry@ksn.or.kr                

Copyright © 2024 by The Korean Society of Nephrology.

Developed in M2PI

Close layer