Antibody-Mediated Rejection in Kidney Transplantation: Immunopathogenesis, Innate–Adaptive Crosstalk, and Therapeutic Advances
Livia Maria Surdi
, Maribel Dagher
, Tamara Merhej
, John Choi
, Jamil R. Azzi *![]()
-
Transplantation Research Center, Renal Division, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
* Correspondence: Jamil R. Azzi![]()
Academic Editor: Rending Wang
Special Issue: Antibody Mediated Rejection in Kidney Transplantation
Received: April 17, 2025 | Accepted: August 27, 2025 | Published: September 10, 2025
OBM Transplantation 2025, Volume 9, Issue 3, doi:10.21926/obm.transplant.2503258
Recommended citation: Surdi LM, Dagher M, Merhej T, Choi J, Azzi JR. Antibody-Mediated Rejection in Kidney Transplantation: Immunopathogenesis, Innate–Adaptive Crosstalk, and Therapeutic Advances. OBM Transplantation 2025; 9(3): 258; doi:10.21926/obm.transplant.2503258.
© 2025 by the authors. This is an open access article distributed under the conditions of the Creative Commons by Attribution License, which permits unrestricted use, distribution, and reproduction in any medium or format, provided the original work is correctly cited.
Abstract
Antibody-mediated rejection (ABMR) remains a major barrier to long-term graft survival in kidney transplantation. Defined by the presence of donor-specific antibodies (DSAs) and characteristic histological changes, such as C4d deposition in peritubular capillaries, ABMR can present acutely, chronically, or subclinically, often manifesting as graft dysfunction. Recent advances in genomic profiling and diagnostic assays have improved our understanding of its pathophysiology, yet therapeutic strategies remain limited. Early detection through routine monitoring and timely intervention, particularly in subclinical ABMR, may improve outcomes. In this review, we provide an in-depth analysis of ABMR in kidney transplantation, with a particular emphasis on recent insights into its immunopathogenesis—emphasizing the dynamic crosstalk between innate and adaptive immunity and its implications for allograft injury. We also discuss how this evolving understanding is reshaping current diagnostic approaches and informing the development of innovative therapeutic strategies. Key findings from recent literature underscore the need for a more integrated approach that bridges mechanistic insight with clinical application, aiming to improve diagnostic precision and long-term graft outcomes.
Graphical abstract

Keywords
Antibody-mediated rejection; kidney transplantation; donor-specific antibodies; graft dysfunction
1. Introduction
Kidney transplantation remains the most effective treatment for patients with end-stage renal disease (ESRD), significantly improving survival rates, quality of life, and reduced treatment costs compared with hemodialysis [1]. The introduction of new potent and precise multimodal immunotherapy in recent decades has significantly reduced acute rejection rates and considerably improved 1-year graft survival following renal transplantation. Improvements in graft half-life for both living and deceased donor transplants have also been shown [2]. While graft survival within the first year exceeds 90%, more than 50% of kidney allografts are lost by the 10th year post-transplant [3]. No significant improvement has been noted when it comes to long-term kidney allograft outcomes [4]. Apart from recurrence of the original disease in the graft, antibody-mediated rejection (ABMR) has been the major cause of kidney transplant failure and is considered as one of the major obstacles to long-term outcomes [5]. Patients with ABMR, specifically those with subclinical ABMR, have a 3.5-fold increased risk of graft loss, highlighting its severe impact on outcomes [6].
The clinical presentation of ABMR in kidney transplantation includes a decline in renal function, frequently detected through routine monitoring. Diagnosis is based on a combination of serological, histological and immunopathological criteria defined by the Banff classification. these criteria include, but are not limited to the detection of donor-specific antibodies (DSAs) against HLA or other alloantigens, and histopathological findings on kidney biopsy that show evidence of microvascular inflammation such as glomerulitis (g) and peritubular capillaritis (ptc) [7]. Immunohistochemical staining for C4d in peritubular capillaries indicating complement activation, is also a supportive marker of ABMR. The Kidney Disease: Improving Global Outcomes (KDIGO) guidelines highlight the importance of these criteria for diagnosis [8].
Formerly, ABMR following renal transplantation was a devastating occurrence that predictably led to allograft loss. It has been the focus of active research in the field and has led to an increased understanding of the molecular and histologic changes that underlie this humoral type of rejection as well as potential therapeutic interventions. However, there are still several unknown pathways, which help explain the limited effectiveness of some of the currently available therapies used to treat ABMR. This review focuses on ABMR as a central cause of long-term graft failure and explores two main aspects: the immunopathogenic mechanisms underlying ABMR, with special attention to the crosstalk between innate and adaptive immunity, and the current and emerging therapeutic strategies. By connecting fundamental immunology with clinical translation, we aim to provide a comprehensive yet focused synthesis of the latest evidence.
2. Pathophysiology of ABMR
From an immunological perspective, both innate and adaptive immune responses contribute to allograft rejection [9]. While the adaptive immune system has historically been considered the main driver of ABMR pathogenesis, emerging evidence highlights a crucial role for the innate immune system. The innate immune system first recognizes donor antigens, presenting specific epitopes to the adaptive immune system. This process triggers a humoral response, leading to the production of donor-specific antibodies (DSAs), which primarily target donor human leukocyte antigen (HLA) molecules.
DSAs are classified into pre-existing and de novo DSAs. Pre-existing DSAs arise from previous sensitizing events, such as pregnancy, blood transfusion, or prior transplantation and are usually identified during pre-transplant screening with single antigen bead assay (SAB). In contrast, de novo DSAs can emerge at any time post-transplant after the recognition of the alloantigens, such as allo-HLA molecules, by the host immune cells. The development of de novo DSAs is often related to insufficient immunosuppressive therapy or patient non-adherence [10,11]. The likelihood of developing de novo DSAs during the post-transplant phase cannot be accurately estimated or predicted. However, several studies have attempted to identify predictive factors associated with an increased risk of de novo DSAs developing in kidney transplant recipients. Known risk factors include the presence of pre-transplant HLA-specific antibodies, previous transplants, graft inflammation [12], high HLA mismatches, especially at the HLA-DQ locus, inadequate immunosuppression and nonadherence to treatment [13].
Additionally, recent evidence has highlighted a correlation between de novo DSA formation, the Mean Fluorescence Intensity (MFI) of anti-HLA antibodies, and donor-derived cell-free DNA (dd-cfDNA) levels. MFI is commonly used in Luminex assays to estimate the strength of anti-HLA antibodies, while dd-cfDNA is a biomarker of graft injury, reflecting donor-derived DNA fragments released into the recipient’s circulation. This evidence suggests that dd-cfDNA monitoring, especially when combined with DSA testing, could provide a valuable minimally invasive approach for assessing the risk of de novo DSA formation and subsequent graft injury [14]. However, it's important to note that while dd-cfDNA shows promise in predicting early detection of de novo DSA, more research is needed to fully establish its predictive value and optimal use in clinical practice.
Whether the DSAs are pre-existing or de novo, their interaction with alloantigens triggers an immune response that culminates in graft injury and rejection.
2.1 The Role of HLA in Mediating Allograft Rejection
HLA is located on the short arm of chromosome 6 (position 6p21) and is inherited from both parents. It is considered one of the most polymorphic regions of the human genome and this high degree of polymorphism explains the significant variability among individuals [15]. As mentioned above, subtle structural differences between HLA alleles trigger immune responses, underscoring the important role of HLA in the immunopathogenesis of rejection [16].
The HLA gene comprises two principal regions, each encoding a distinct HLA class. Specifically, Class I HLA is expressed on the surface of all nucleated cells, including endothelial cells within the transplanted kidney. It is defined by the loci HLA-A, HLA-B, and HLA-C. The major role of Class I HLA molecules is to present segments of endogenous and other cytoplasmic proteins after they are processed by the proteasome. These peptide-Class I HLA complexes are surveyed by cytotoxic cells, such as CD8+ T cells or natural killer cells (NK cells) [17]. While self-peptides presented by HLA induce inhibitory signals, non-self peptides, such as viral peptides, lead to lymphocyte activation and cytotoxic responses. In contrast, Class II HLA, characterized by the loci HLA-DR, -DQ and -DP, is primarily found on the surface of professional antigen-presenting cells (APCs), including dendritic cells, macrophages and B cells.
One of the major roles of APCs is to recognize and phagocytose exogenous antigens. Within the intracellular environment, these antigens are internalized into endosomal and lysosomal vesicles, which are characterized by a highly acidic environment. This, together with the action of proteolytic enzymes, ensures the degradation of antigens into small peptides. Subsequently, these peptide fragments are loaded onto class II HLA molecules and transported to the cell surface, where the peptide-HLA complex is recognized by CD4+ T lymphocytes in the lymph nodes, initiating and amplifying the adaptive immune response [18].
In kidney transplant recipients, the presence of DSAs together with a pro-inflammatory milieu rich in cytokines (such as IFN-gamma, IFN-γ), may cause upregulation of HLA Class II molecules on donor endothelial cells via the JAK-STAT signaling pathway [19]. This upregulation increases the likelihood of de novo DSA formation, thereby escalating the immune response and promoting a positive feedback loop that contributes to both acute and chronic ABMR [20] (Figure 1).
Figure 1 Schematic representation of the vicious cycle initiated by DSAs in antibody-mediated rejection. DSAs bind to donor HLA (Class I and II) antigens, triggering complement activation and promoting an inflammatory response. This inflammatory response leads to the recruitment of inflammatory cells, which contribute to endothelial injury. Concurrently, inflammatory cytokines such as IFN-γ induce the up-regulation of HLA Class II expression on endothelial cells, enhancing further DSA binding and perpetuating the cycle. Moreover, the complement activation can cause a direct injury to the endothelial cells.
A primary predictor of allograft rejection in clinical practice is the degree of HLA mismatch between donor and recipient. A higher number of mismatches correlates with a greater risk of de novo DSA production and allograft failure [21]. Beyond HLA mismatch, individual variability in HLA expression also influences the risk of rejection. For instance, certain HLA-DQ alleles, such as HLA-DQ5, are associated with higher expression levels on endothelial cells and a greater risk of microvascular injury. In contrast, alleles like HLA-DQ7 and HLA-DQ8 exhibit lower expression levels and do not appear to increase this risk [22].
Considering this inter-individual and inter-locus variability, the risk of rejection may not solely depend on the presence of an allele mismatch but also on the level of allele expression on graft endothelial cells [22]. Since undetected mismatches or variations in allele expression can contribute to antibody formation and immune activation, pre-transplant compatibility assessments are essential. For instance, living donor transplants increase the likelihood of graft survival by allowing a better screening and selection of potential donors with fewer HLA mismatches, particularly when the donor is a close relative.
2.2 The Role of Non-HLA Antigens in Kidney Transplantation
While HLA molecules remain the principal alloantigens involved in the pathogenesis of kidney allograft rejections, growing evidence highlights the contribution of non-HLA antigens in graft dysfunction and rejection [23,24]. Senev et al. collected the data from a cohort of 935 kidney transplant patients who underwent kidney biopsy. They showed that more than half of the patients met the Banff histological criteria for ABMR, without the presence of circulating HLA-DSA [25]. In a subsequent study from the same group, a more extensive cohort of HLA-DSA negative ABMR cases was tested for the presence in the serum of non-HLA antibodies, including anti-AT1R and anti-MICA. The authors demonstrated a significantly higher prevalence of these antibodies in patients with histological ABMR compared to matched controls without rejection, supporting their potential pathogenic role. These findings suggest that non-HLA DSAs may contribute to the pathogenesis of ABMR in the absence of classical HLA-DSA [23].
The most significant non-HLA antigens are the Minor Histocompatibility Antigens (mHAs). These are polymorphic proteins expressed in the allograft and presented by the recipient’s antigen presenting cells (APC) to naïve T cells, thereby eliciting adaptive immune responses. Even in the presence of HLA compatibility, this interaction may lead to chronic rejection [26,27]. The genetic basis for mHAs is a non-synonymous single nucleotide polymorphism (nsSNPs) which results in mutations in the amino acid sequence of the corresponding protein, leading to variations among individuals. For instance, proteins encoded by the Y chromosome (HY antigens) have been extensively studied as a culprit of rejection in male-to-female transplant [28].
Another critical non-HLA antigen is the MHC class I chain-related protein A (MICA) expressed on endothelial cells. Different studies, including data from a French multicenter cohort study, have shown a correlation between the detection of anti-MICA antibodies and worse graft outcomes. These antibodies are associated with a higher incidence of ABMR and reduced graft survival, even in recipients with no HLA-mismatches at the time of transplantation [29,30]. Importantly, MICA and its homolog MHC class I chain-related protein B (MICB) can interact with the NKG2D receptor expressed on natural killer (NK) cells, potentially triggering NK cell-mediated cytotoxicity against the allograft, further contributing to allograft rejection [31,32].
Furthermore, anti‐endothelial cell antibodies (AECA), first identified in 1980, have been associated with an increased risk of rejection [33,34]. Among these antigens, some examples include antibodies against angiotensin II type 1 receptor (AT1R) and endothelin receptor A (ETAR), both expressed on the surface of endothelial cells. However, not every transplant recipient with positive AT1R and ETAR antibodies experiences allograft dysfunction, suggesting that their pathogenic potential may depend on additional factors, including the degree of ischemia-reperfusion injury [35], the immunologic milieu [36], genetic polymorphisms [37] or antibody titer [38,39]. Therefore, detection of these antibodies may help identify patients at risk of ABMR even in the absence of HLA-DSA, although routine clinical implementation is still lacking.
In summary, despite growing evidence supporting the pathogenic role of non-HLA antibodies, their use in clinical practice remains limited and not routinely incorporated into transplant immunological screening. The findings from Senev et al. and others reinforce the need to reconsider current diagnostic criteria and explore the integration of non-HLA antibody screening, especially in patients with histological evidence of ABMR but no detectable HLA-DSA [23,25]. However, the absence of standardized testing protocols and consensus thresholds for positivity still represents a major barrier to routine clinical implementation. Prospective multicenter studies will be critical to define the true clinical utility of these biomarkers.
2.2.1 HLA and Non-HLA: Pre-Transplant Screening
HLA and non-HLA antibody screening play a critical role in transplant immunology, influencing both pre-transplant compatibility assessments and early risk stratification.
Pre-transplant HLA screening is typically performed through three main approaches: HLA typing, Luminex-based single antigen bead (SAB) assays, and physical or virtual crossmatch.
HLA Typing identifies the recipient's HLA alleles and compares them to the donor's HLA alleles to estimate the risk of mismatch [40].
However, crossmatching is a mandatory pre-transplant test, which determines whether a recipient has pre-existing antibodies against donor HLA or non-HLA molecules. Traditionally, this is done through physical crossmatch, which involves mixing recipient serum with live donor lymphocytes to detect complement-dependent cytotoxicity or flow cytometry-based crossmatch [41], but its main limitation is the need for viable donor cells and the time required to obtain results [42].
To improve sensitivity and specificity, Luminex-based SAB assays have been introduced. These assays detect and quantify DSAs with high resolution by exposing recipient serum to fluorescent beads coated with individual HLA antigens [43]. This approach allows for a more precise characterization of alloimmune risk.
Nevertheless, virtual crossmatch (vXM) has become an essential tool in transplantation, as it allows the evaluation of immunological compatibility between a recipient and a potential donor [44]. This is done by analyzing the recipient’s pre-transplant HLA antibody profile using data from SAB assays and donor HLA typing. vXM enables early risk stratification and helps predict the likelihood of antibody-mediated rejection before transplantation [44]. Unlike traditional physical crossmatch, which requires actual donor cells, vXM facilitates more rapid and efficient organ allocation while minimizing unexpected immunologic incompatibilities and reducing rejection rates [40].
Detection of non-HLA antibody in kidney transplantation can also be performed using both cell-based and solid-phase assays. Early cell-based crossmatch assays use primary endothelial cells, such as Human Umbilical Vein Endothelial Cells (HUVEC) or donor-derived endothelial cells, to screen for antibodies against targets such as AT1R and MICA [45]. In parallel, solid-phase methods, such as ELISAs and Luminex-based assays, provide rapid, high-throughput detection using native antigen preparations. These methods are especially useful for identifying antibodies like anti-AT1R and anti-ETAR [46,47]. Recent advances also include protein microarrays and genome-wide analyses to uncover novel antigenic targets linked to graft outcomes [48,49].
Despite these advancements, limitations remain, particularly in the detection of non-HLA antibodies that may contribute to graft dysfunction. Moreover, the lack of standardized thresholds remains challenges for clinical implementation.
2.2.2 The Role of ABO Antibodies in Kidney Transplantation
Historically considered a major barrier to transplantation, ABO-incompatible (ABOi) kidney transplants have become a viable option over the past few decades, particularly in Asia and Europe, thanks to advances in immunosuppressive protocols and desensitization strategies [50]. ABO incompatibility is caused by the presence of preformed antibodies against A and B antigens, primarily IgM and IgG, which can cause hyperacute rejection through complement activation and rapid graft destruction [51]. These antibodies are produced by B lymphocytes in response to blood group antigens, which are expressed not only on red blood cells but also on endothelial and distal tubular cells of the transplanted kidney [52]. In this context, rejection occurs via complement-mediated cytotoxicity, leading to endothelial damage, thrombosis and rapid allograft necrosis. These mechanisms are similar to those observed with donor-specific anti-HLA antibodies. The main desensitization strategies include plasmapheresis, intravenous immunoglobulins (IVIG), and anti-CD20 monoclonal antibodies, such as rituximab.
A recent study by a Swiss group examined the impact of pre-transplant DSA on the risk of developing ABMR in living donor kidney transplants, comparing ABOi and ABO-compatible (ABOc) recipients [53]. They observed a higher incidence of ABMR in ABOi transplants compared to ABOc transplants; however, this did not significantly affect graft or overall survival, which remained similar between the two groups. Additionally, the presence of pre-transplant DSA was associated with significantly worse long-term outcomes in both ABOi and ABOc living donor kidney transplants. This may suggest that the risk associated with pre-transplant DSA could be comparable, regardless of ABO compatibility [53].
However, ABOi transplantation seems to be associated with an increased risk of infections and thrombotic complications, particularly in the early post-transplant period [51]. The development of novel therapeutic approaches, such as complement inhibitors, could further improve the safety and efficacy of this type of transplant [54,55].
2.3 Alloantigen Recognition and Immune Activation: The Driving Forces of Transplant Immunity
In the absence of prior sensitization to donor antigens, the recognition of non-self-epitopes is initially mediated by the innate immune system, exerting its function through antigen-presenting cells (APCs), including dendritic cells (DCs), macrophages and B cells.
In fact, three different pathways of alloantigen recognition have been identified as contributing to graft rejection. The first pathway known as “direct”, involves donor’s APCs, specifically DCs, which migrate from the graft to the recipient’s lymph nodes. There, they present donor-derived antigens to naïve T cells, notably donor’s HLA. This pathway is predominant in the early post-transplant phase, since donor APCs are short-lived and decline over time [56]. The second pathway, known as “indirect”, relies on recipient-derived APCs, which capture and process donor alloantigens from the graft. These phagocytic APCs degrade the alloantigens into smaller peptides and present them on their own HLA Class II molecules. The alloantigen-HLA complex is then recognized by the recipient’s CD4+ T cells, eliciting an adaptive immune response that become more prominent in the last phase of rejection and is strongly associated with chronic graft injury [57]. Finally, the third pathway also called “semi-direct”, consists of an interaction between recipient and donors APCs which leads to an exchange of part of the donor APCs membrane, including the intact peptide-HLA complex, to the recipients’ APCs. Consequently, recipient APCs can simultaneously present both self- and donor-derived HLA to naïve T cells [58].
In all three pathways, the DCs expressing CC-chemokine receptor 7 (CCR7) migrate to the T-cell zone of lymph nodes. This migration is facilitated by a chemokine concentration gradient consisting of C-C motif 19 and 21 (CCL19 and CCL21), secreted by high endothelial venules (HEVs) and fibroblastic reticular cells [59].
The migration of DCs into the T zone of the lymph node with subsequent alloantigen presentation of HLA II complex to TCR of naïve CD4+ T cells represent the first stimulatory signal for T cell activation. Additional costimulatory signals will follow; the second signal for activation is the interaction of CD80/CD86 expressed on DCs and CD28 on naïve T cells. Finally, a third signal consists of cytokine-driven proliferation and survival, primarily mediated by IL-2 secretion from naïve T cells and CD25 upregulation, culminating in full T-cell activation.
A critical step in ABMR is the differentiation of CD4 T cells into T follicular helper cell (Tfh), a specialized subset of T cells. This differentiation is promoted by a pro-inflammatory environment, mainly consisting in high concentration of IL-6 produced by DCs [60,61] (Figure 2).
Figure 2 Activation of naïve CD4⁺ T cells and their differentiation into T follicular helper (Tfh) cells. (A) Dendritic cells (DCs) migrate into the lymph node following CCL19/CCL21 gradients through high endothelial venules (HEVs), reaching the T cell zone. There, DCs interact with naïve CD4⁺ T cells, providing three signals: antigen presentation via HLA-II/TCR (signal 1), co-stimulation through CD80/CD86–CD28 (signal 2), and cytokines such as IL-2 (signal 3). (B) Upon activation, T cells are further stimulated by IL-6 and IL-21, which promote Bcl-6 expression and drive commitment to the Tfh lineage. Differentiated Tfh cells upregulate CXCR5, ICOS, and CD40L, enabling migration to the B cell follicles where they interact with B lymphocytes to support germinal center responses.
Tfh are characterized by the expression of CXC-chemokine receptor 5 (CXCR5), B-cell lymphoma 6 (bcl-6) and Inducible T-Cell Co-Stimulator (ICOS), all of which are essential for their survival, differentiation, and germinal center formation [62,63]. Particularly, ICOS is a co-stimulatory molecule belonging to the CD28/B7 family, essential for Tfh cell survival and proliferation; Bcl-6 is a repressor transcriptional factor crucial for the differentiation in Tfh cell and for the germinal center formation [60,64].
Finally, Tfh guided by the CCL13 gradient, which is a CXCR5 ligand, migrate from the T-cell zone to the B-cell follicles, where they interact with B cells [65,66]. This interaction is reinforced by different co-stimulatory signals, such as ICOS-ICOSL and CD40-CD40L interactions, as well as IL-21 signaling.
These exchanges contribute to the formation of germinal centers within the secondary follicles of lymph nodes, where B cells undergo the “germinal center reaction”. This reaction generates memory B cells and plasma cells, but it also enables somatic hypermutation and affinity maturation, ensuring high specificity for donor HLA antigens [67,68]. Long-lived plasma cells migrate to the bone marrow and continuously produce DSAs, while memory B cells persist in secondary lymphoid organs, providing a rapid source of antibody production upon antigen re-exposure [69].
Circulating DSAs bind to donor HLA on graft endothelial cells, activating the classical complement cascade and leading to C4d deposition as well as formation of the membrane attack complex (MAC) formed by C5b-C9 proteins [70]. The MAC causes direct endothelial and parenchymal injury through pore formation, increased membrane permeability and subsequent inflammation [12]. This injury further promotes the release of pro-inflammatory cytokines and the upregulation of adhesion molecules, including ICAM-1, VCAM-1 and E-selectin [71]. These molecules facilitate leukocyte adhesion and infiltration into the graft, thereby amplifying inflammation and driving progressive ischemia and fibrosis [72,73].
Beyond complement activation, DSAs can also cause graft injury through another toxic pathway known as antibody-dependent cell-mediated cytotoxicity (ADCC). In this pathway, the Fc portion of DSAs engages Fcγ receptors on immune effector cells, including NK cells and cytotoxic T cells, triggering endothelial damage even in the absence of C4d deposition [74].
Clinically, ABMR can manifest as either C4d-positive or C4d-negative rejection. While C4d-negative ABMR has been considered less severe, evidence suggests that it is still associated with poor graft outcomes. Orandi et al. [75] compared graft survival in C4d-negative versus C4d-positive ABMR, reporting 1- and 2-year survival rates of 93.4% and 90.2% in the C4d-negative group, versus 86.8% and 82.6% in the C4d-positive group. These findings underscore that both complement- and non-complement-mediated mechanisms substantially contribute to graft injury, highlighting the importance of recognizing and managing all forms of ABMR.
2.4 The Innate Immune System: A Key Player in ABMR
The adaptive immune system is the principal effector in the pathophysiology of ABMR; however, emerging evidence highlights that the innate immune system is actively involved in the rejection process. NK cells, macrophages, the complement system and other innate immune components interact with DSAs to initiate and exacerbate inflammation and cytotoxic cascades that worsen graft injury. Innate immune cells are further recruited into the graft by the increased concentration of C3a and C5a, complement fragments [76]. As described previously, the complement system is a central component of innate immunity that is activated during ABMR, and the classical pathway is activated primarily through the binding of DSAs. In fact, the classical pathway of complement cascade is initiated by the interaction between DSAs and C1q, triggering the activation of the C1r and C1s proteins. The formation of the C3 convertase (C4b2a) and subsequently C5 convertase culminates in the assembly of the membrane attack complex (MAC, C5b-C9), which directly damages endothelial cells and increases vascular permeability [77]. A comprehensive review of the complement cascade and its role in ABMR has been summarized in multiple articles [78,79]. A hallmark of ABMR is C4d deposition, a fragment of C4, in peritubular capillaries detected in the kidney biopsy [80].
Macrophages and monocytes are a critical component of the innate immune response in ABMR. They amplify graft injury by promoting inflammation, tissue damage, and fibrosis [81]. Monocytes originate in the bone marrow from common myeloid progenitors, circulate in the blood and migrate to peripheral tissues in response to inflammation, where they differentiate into resident macrophages. Notably, two main tissue-resident macrophages (TRMs) exist within tissues. The first type, mentioned previously, originates from circulating monocytes, while the second type emerges from the yolk sac progenitors established during embryogenesis, capable of self-renewal and persistence. As a result, the transplanted organ contains both types of cells [82].
After transplantation, both donor TRMs and recipient-derived macrophages coexist within the graft [83]. The role of donor TRMs in transplant outcomes is not yet clear, but different groups are exploring their role in graft rejection. For instance, a recent study by Kopecky et al demonstrated in a murine heart transplant model that donor monocyte-derived CCR2+ macrophages promote inflammation and drive allograft rejection. In contrast, the selective depletion of donor CCR2- macrophages originating from the yolk sac decreases allograft survival. This study also included single-cell analysis, highlighting significant heterogeneity among recipient monocytes, macrophages and dendritic cells [83]. These findings suggest that macrophages may either promote or suppress rejection depending on their phenotype.
In addition to donor macrophages, recipient macrophages expressing Fcγ receptors can interact with the Fc region of DSAs. This interaction stimulates the release of pro-inflammatory cytokines that contribute to the tissue damage within the graft [84].
Distinct macrophage subsets exert distinct roles during various stages of ABMR: pro-inflammatory macrophages (M1) predominate in early stages and release cytokines such as IL-6 and TNF-α. In contrast, M2 macrophages (M2) may play a role in chronic ABMR, promoting fibrosis via TGF-β and IL-10 [85]. The diverse roles of macrophages highlight the need to understand their dynamics in the pathophysiology of transplant rejection.
NK cells are another crucial innate immune population involved in ABMR. Originating from the common lymphoid progenitor in the bone marrow and are released into the lymphatic system. These cells play a remarkable role in the pathogenesis of ABMR [86]. Their involvement is primarily through the interaction between the CD16 (FcγRIII) on their surface and the Fc portion of DSAs bound to donor endothelial cells. This interaction activates NK cells, leading to a subsequent release of granzyme and perforin, causing direct damage to endothelial cells. This process is known as antibody-dependent cellular cytotoxicity (ADCC) [87]. NK cells also produce large amounts of interferon-gamma (IFN-γ), further activating macrophages and endothelial cells. This process intensifies the inflammatory response and exacerbates tissue damage within the graft.
In addition, numerous studies have emphasized the involvement of NK cells and macrophages in the pathophysiology of kidney rejection, demonstrated the increased infiltration in peritubular capillaries in both C4d-positive and C4d-negative ABMR lesions [88]. Notably, in C4d-negative ABMR, where complement activation is absent, NK cell-mediated cytotoxicity still represents a major mechanism of endothelial damage [89]. All this evidence should encourage researchers to focus more on the role of NK cells in the pathophysiology of rejection, with the aim of using these cells as a therapeutic target [90].
2.5 Role of Ischemia-Reperfusion Injury (IRI) and DAMP Expression
Ischemia-reperfusion injury (IRI) significantly contributes to delayed graft function (DGF), acute rejection, and chronic graft dysfunction [91]. Following the donor’s death, particularly in cases of cardiac arrest (donation after cardiac death - DCD), hemodynamic instability reduces renal perfusion, triggering complement activation and innate immune responses. This injury is exacerbated during kidney retrieval, as clamping the renal artery induces significant ischemia. Following retrieval, the organ undergoes a storage period known as “cold ischemia”, during which it is usually preserved on ice. While this method extends the organ’s viability, it can also cause additional damage, increasing DGF rates [92].
Various endogenous molecules, released or exposed by stressed or dying cells during IRI, have been implicated in the pathophysiology of kidney rejection [93]. These molecules, known as damage-associated molecular patterns (DAMPs) and pathogen-associated molecular patterns (PAMPs), are recognized by pattern recognition receptors (PRRs) like TLRs, expressed on innate immune cells [94,95]. The interaction between DAMPs and PRRs trigger pro-inflammatory responses and endothelial activation [96]. In recent years, several DAMPs have been identified as key drivers of immune activation leading to graft rejection [93]. Well-characterized DAMPs include heat-Shock protein (HSP), high-mobility group box 1 (HMGB-1) and ATP [56,97]. Injured cells can also release mitochondrial components such as mitochondrial DNA and N-formyl peptides, which mimic bacterial signals and activate immune responses [98,99]. Additionally, during IRI, components of the extracellular matrix – such as hyaluronate and heparan sulfate – can break down into smaller fragments, acting as potent extracellular DAMPs, further promoting activation of the innate immune system [97].
In recent decades, the rising demand for kidney transplantation, coupled with a limited supply of available organs, has prompted clinicians to explore strategies for increasing the donor pool [100]. This includes broadening donor criteria to include extended criteria donors (ECD) and DCD donors [101]. However, these donor categories, particularly DCD donors who experience more severe ischemia, are associated with increased risk of DGF and subsequent rejection [102,103].
Although preventing cold ischemia-induced damage is important to reduce the likelihood of rejection, further attention should be directed to reperfusion damage [93,97]. Indeed, once recipient vasculature is connected to graft renal artery, reperfusion typically induces pro-inflammatory genes upregulation [103]. These pathways are mediated by an enhanced expression of transcription factors such as Nuclear Factor Kappa B (NF-κB) and Toll-like receptors (TLRs) [104]. In addition, multiple mechanisms including apoptosis, necroptosis, autophagy, microvascular dysfunction, and transcriptional reprogramming can cause additional damage to the kidney [105,106]. This results in the activation of both the innate and adaptive immune systems characterized by inflammatory cell infiltration and heightened immune activation [92].
The introduction of kidney machine perfusion techniques, which allow organ perfusion prior to implantation, has contributed to reducing the risk of DGF and subsequent immune activation [107]. A recent meta-analysis comparing different perfusion techniques highlighted a significant reduction in DGF rates when hypothermic machine perfusion (HMP) was used instead of static cold storage (SCS) [108]. Nevertheless, no significant differences were observed between HMP and oxygenated HMP (HMP + O2). Regarding rejection rates, no substantial differences were observed between SCS and HMP; however, HMP + O2 appears to be associated with a reduction in acute rejection episodes, as confirmed by histological analysis kidney biopsies [108].
In conclusion, the interplay between DAMPs, PRRs, and innate immune cells represents a pivotal mechanism driving IRI-induced injury.
2.6 Immunoregulation in Kidney Transplantation: A Matter of Tolerance
The immunoregulatory system is a key component of immunity that maintains a balance between immune activation and suppression, ultimately promoting immunotolerance. In ABMR, this serves as a crucial mechanism to foster tolerance and mitigate harmful stimuli. The main protagonists of the immunoregulatory response are regulatory macrophages (Mregs) and regulatory T cells (Tregs). Together, they work to minimize tissue damage and regulate the immune response, with the ultimate goal of preserving graft function and enhancing long-term graft survival [109].
2.6.1 Regulatory T Cells (Tregs)
Tregs are a specialized subset of CD4+ T cells that play a crucial role in maintaining immunotolerance in transplantation. The concept of immunotolerance was introduced in 1970, when Gershon and Kondo demonstrated that exposing immune cells to a non-self-antigen could induce immune tolerance [110]. This mechanism was not initially attributed to a specific subpopulation; however, it introduced the concept of immune tolerance. In 1995, Sakaguchi et al. first identified CD4+ CD25+ cells and demonstrated that depleting CD25 (the α-chain of IL-2 receptor) and transferring them into mice could spontaneously induce autoimmune diseases [111]. Less than a decade later, in 2003, two independent research groups described forkhead box P3 (FOXP3) expression in CD4+ CD25+ T cells and elucidated its role in immunoregulation [112,113]. FOXP3 was found to be a critical transcription factor for Treg function, along with another transcription factor, Helios, which is essential for maintaining their immunoregulatory activity [114].
Tregs modulate immune responses through various mechanisms, including the secretion of immunoregulatory cytokines such as IL-10, IL-35 and TGF-β, direct cell-to-cell interactions, and the up-regulation of inhibitory receptors like cytotoxic T-lymphocyte associated antigen-4 (CTLA-4). The interaction between CTLA-4 and CD80/CD86 on APCs stimulate the production of indoleamine-2,3-dioxygenase (IDO), which inhibits T cell activation and proliferation, thereby reducing effector T cell activity and limiting B cell responses [115]. Additionally, activated Tregs release cytolytic enzymes, such as perforin and granzymes A and B, which target effector T cells, inducing cytotoxicity and apoptosis [116].
Finally, Tregs can suppress T follicular helper (Tfh) cells, thereby limiting their ability to activate B cells. However, upon TCR activation, Tregs up-regulate CXCR5, enabling their migration to the border between T and B cell zones, as well as into germinal centers (GCs), guided by the high concentration of CXCL13. Simultaneously, they downregulate CCR7, which typically retains cells in the T-cell zone. In this region, they can interact directly with and inhibit B cells [117] (Figure 3).
Figure 3 (A-B) The role of Tregs in immune regulation within the lymph node. (A) Representation of the lymph node microenvironment. The left side shows the T cell zone, where naïve T cells interact with dendritic cells (DCs), and Tregs are present. The right side shows the immune regulation, where Tregs inhibit naïve T cell activation via CTLA-4/CD86 interaction, the production of indoleamine 2,3-dioxygenase (IDO), and the secretion of immunosuppressive factors such as IL-10 and TGF-β. (B) Interactions of Tregs in the T-B border. The left side highlights Tregs’ cytotoxic effect on naïve T cells through granzyme B release, inducing apoptosis. The right side shows Tregs migrating to the B cell zone, driven by CXCL13, and inhibiting B cell differentiation into plasma cells through regulatory cytokines (IL-10, TGF-β) and CTLA-4/CD86 signaling.
While CD4+ Tregs are the most extensively studied regulatory T-cell population, recent years have seen growing scientific interest in the role of the CD8+ Treg subset in autoimmune diseases and kidney transplant tolerance. This population has been well-characterized in mice since the early 2000s. CD8+ Tregs are distinguished by high expression of CD122 (also known as IL-2 receptor β chain, IL-2Rβ) and inhibitory Ly49 receptor (Ly49A, Ly49 C/I, Ly49F, Ly49G). The human equivalent of Ly49 is the killer cell immunoglobulin-like receptor (KIR) gene.
Unlike CD4+ Tregs, CD8+ Tregs express FOXP3 only under inducible conditions but exhibit constitutive expression of Helios. Over the past two decades, numerous studies have highlighted the critical role of Helios in maintaining the immune homeostasis. A defect in this transcription factor can disrupt tolerance, leading to the development of autoimmune diseases [114].
Similar to CD4+ Tregs, CD8+ Tregs exert their functions through by secreting cytotoxic molecules such as perforin, which targets and kills alloreactive CD4+ T cells and Tfh cells [118].
In recent years, a subset of CD8+ Tregs has been identified that specifically recognizes a particular type of MHC class Ib molecule, Qa-1 in mice and HLA-E in humans, expressed on CD4+ effector T cells. Research by Hye-Jung Kim and colleagues has demonstrated the crucial role of this interaction in maintaining immunotolerance. In fact, blocking the interaction between these two cell populations resulted in the development of a severe systemic lupus erythematosus–like autoimmune disease, highlighting its essential role in immune regulation [119].
Another important discovery, observed in a murine heart transplant model, emphasizes the interaction between Qa-1-restricted CD8+ Tregs and CD4+ Tfh cells. This interaction is linked to the suppression of the Tfh cell activity, which in turn suppresses B cell function and the production of DSAs, ultimately reducing the risk of ABMR [120].
These findings have motivated researchers to explore therapeutic strategies aimed at enhancing Treg function in solid organ transplantation, reducing the incidence of ABMR and improving allograft survival.
Emerging technologies, such as cell therapies currently applied in onco-hematology, are increasingly being explored for potential applications in different medical fields, including autoimmune diseases and transplantation. Recent studies have reported promising results on the use of CD19-targeting CAR-T cells in autoimmune diseases, such as systemic lupus erythematosus (SLE), demonstrating their ability to deplete pathogenic B cells and reduce autoantibody production [121]. Given the critical role of B cells and donor-specific antibodies in ABMR, CD19 CAR-T cell therapy could similarly modulate the alloimmune response, potentially mitigating ABMR and improving graft survival. Moreover, the development of alternative and more effective therapeutic strategies could enable a reduction in conventional immunosuppressive treatments, minimizing their associated adverse effects.
2.6.2 Regulatory Macrophages
A specialized subset of cells contributing to the immune regulation are the regulatory macrophages (Mregs). These cells exert their functions by the secretion of cytokines such as interleukin-10 (IL-10) and TGF-β which play a crucial role in promoting immune tolerance and facilitating tissue repair [109,122]. An interesting feature emerges when Mregs are activated by IFN-γ: in this context, they release TGF-β, which inhibits T-cell proliferation and promotes the expansion of Tregs. In addition, Mregs can induce the expression of T-cell immunoreceptors with Ig and ITIM domains (TIGIT) on CD4+ T cells. These TIGIT+ CD4+ T cells can suppress effector T cells and inhibit dendritic cell maturation, thereby enhancing immune regulation [123].
Over the past few decades, Mregs have been subject to various clinical trials investigating their role in immune tolerance induction in kidney transplantation. In the Transplant Acceptance-Inducing Cell trial I (TAIC I) trial [124], Hutchinson et al. investigated the use of donor-derived TAICs, a type of immunoregulatory macrophage, in kidney transplant recipients. Although the trial primarily focused on tapering immunosuppressive therapy and acute cellular rejection, it provided insight into the ability of regulatory macrophages to modulate alloimmune responses. While these results highlight the potential of Mregs in immune regulation, their specific role in preventing ABMR remains unclear. Further research is needed to determine whether Mreg-based therapies can reduce the incidence or the severity of ABMR.
3. Clinical Presentation and Diagnosis of ABMR
The clinical presentation of ABMR is associated with a deterioration in kidney function, often evidenced by a rise in serum creatinine levels. This can occur acutely or chronically. Symptoms may be nonspecific, including malaise, fever, or graft tenderness, but many cases are asymptomatic and detected through routine monitoring [8].
An early detection of kidney allograft dysfunction, and a timely diagnosis of ABMR is crucial, as ABMR can result in irreversible damage to the allograft if not properly managed with full functional recovery [4]. Kidney transplant recipients should be monitored with signs of ABMR with a frequency that balances early detection and clinical pragmatism. The Kidney Disease: Improving Global Outcomes (KDIGO) guidelines recommend frequent monitoring in the early post-transplant period, with visits 2-3 times per week during the first month, every 1-3 weeks during months 2-3, every 4-8 weeks during months 4-12, and every 2-4 months after the first year [125]. This schedule allows for the early detection of graft dysfunction and the presence of de novo DSAs, which are critical for detecting ABMR. For high-risk patients, with preexisting DSAs or those who develop de novo DSAs, protocol biopsies are recommended within the first-year post-transplant to detect subclinical ABMR [126].
The gold standard for diagnosis of kidney allograft rejection or lack thereof is based on kidney pathology acquired through allograft biopsies and relies on the Banff classification to segregate the findings [127].
The Banff classification is a standardized framework that classifies and rates kidney transplant rejection depending on histological features. It was created in 1991 and has since been updated numerous times to improve its precision and clinical applicability [128], with the last Banff meeting taking place in 2022 [80].
This classification divides antibody-mediated rejection into acute antibody-mediated rejection (aABMR), chronic active antibody-mediated rejection (caABMR), and chronic (inactive) antibody-mediated rejection (cABMR). Nevertheless, patients often manifest multiple types of rejection concomitantly. Several acute and chronic histological findings are taken into consideration in this classification to confirm rejection.
In acute rejection, modifications of this form of rejection include i for interstitial inflammation, t for tubulitis, v for arteritis, g for glomerulitis, PTC for peritubular capillary inflammation and c4d representing complement 4d product [7,127]; whereas, chronic rejection variations are ct for tubular atrophy, ci for interstitial fibrosis, cv for arteriolopathy, cg for chronic glomerulopathy (or transplant glomerulopathy), and peritubular capillary basement membrane thickening [127].
Light microscopy is generally used to diagnose the majority of these changes, however, c4d, which is a histologic indicator of complement activation, can be diagnosed through either immunofluorescence or immunohistochemistry, making it a unique histological component [127]. Also, electron microscopy helps in depicting peritubular capillary basement membrane thickening [128,129].
For each variation, a grade ranging from 0 to 3 is given, and the degree of rejection is established on the gravity of one or a combination of the findings above [127].
There are three criteria that should be met to diagnose aABMR (Figure 4) [127,130]. caABMR is a pathological process resulting from continuous antibody-mediated damage, which causes chronic harm to endothelial cells and remodeling of the allograft matrix [131]. In addition to the same diagnostic criteria of aABMR, in caABMR, there is also chronic changes of cg, peritubular capillary basement membrane thickening, or chronic thickening of the arteriolar intima cv. In chronic (inactive) caABMR, we find these similar chronic changes without the presence of active inflammation. During the 2022 Banff meeting, two supplemental ABMR subtypes were added to the profiles cited in the table, which are: probable ABMR marked by circulating DSA and individual microvascular inflammation (MVI) lesions that do not meet the histological threshold for MVI (g + ptc < 2) and which doesn’t require treatment unless in certain situations such as high-risk crossmatch positive transplants which aren’t correlated yet with significant MVI superior to the threshold, or if there is a rapid deterioration of kidney function among patients with de novo HLA-DSA [7]; and MVI which exceeds the histological threshold but is deficient of circulating DSA and shows negative C4d staining in the peritubular capillaries. Also, during this meeting new diagnostic techniques relying on computerized reporting algorithms were presented [80]. These systems insert “raw” histological and other data into a software program which then generates classifications based on Banff guidelines or alternative algorithms. Among these presentations, the “Software-assisted sign out” (SAS) designed at Pittsburgh, the Banff automation system from the Paris Transplant Institute [132], and machine-learning classifiers designed to analyze kidney graft scores in conjunction with clinical data [133].
Figure 4 Diagnostic criteria for active ABMR (all 3 criteria must be met) [127,130].
4. Treatment
Management strategies for both clinical and subclinical forms of ABMR differ across transplant centers and often involve a combination of treatments such as high-dose steroids, plasmapheresis, and intravenous immunoglobulin (IVIG). However, the use of additional therapies varies widely. The Kidney Disease: Improving Global Outcomes (KDIGO) guidelines, as cited by the National Kidney Foundation Kidney Disease Outcomes Quality Initiative, suggest plasmapheresis, IVIG and Rituximab as therapeutic options, but only with a weak recommendation (2C), reflecting the low level of evidence and reliance on expert consensus rather than high-quality randomized data [125]. A 2018 systematic review highlighted that, despite limited evidence, plasmapheresis and IVIG have become standard treatments for ABMR. Rituximab has been deemed ineffective, and the roles of bortezomib and complement inhibitors remain uncertain [134].
Additionally, the timing and presence of chronic changes in ABMR play a critical role in determining the course of treatment (Table 1). Notably, plasmapheresis, while occasionally utilized, does not directly address the root cause of ABMR and should be used judiciously [135].
Table 1 Therapeutic options in acute and late ABMR based on the 2019 consensus [135].

In addition to the standard therapeutic options, other, less used, alternatives exist. We cite the ones below.
4.1 Complement Inhibitors
The activation of the classical complement pathway plays a critical role in the downstream effects of DSA and is a key contributor to the manifestations of ABMR [136]. Complement inhibitors, such as Eculizumab, a monoclonal antibody targeting complement protein C5 activation, and complement 1 esterase inhibitors (C1 INHs), have been employed for the treatment of ABMR with varying levels of success. This molecule can cause a variety of side effects, including infection with encapsulated bacteria (ie. Streptococcus pneumonia, Neisseria meningitidis, Haemophilus influenza etc.) [54]. Therefore, vaccination against these pathogens and/or prophylactic antibiotics are required before treatment initiation. Eculizumab showed success when treating grave or refractory cases of ABMR in HLA- or ABO-incompatible kidney transplants [137,138,139,140]. However, it is not routinely used in the treatment of ABMR.
4.2 Proteasome Inhibitors
An example of proteasome inhibitors originally used to treat multiple myeloma is Bortezomib. This molecule has also been utilized in the management of ABMR, particularly due to its ability to target protein synthesis rate which is characteristic of plasma cells and leading to their apoptosis [141]. Since its use in 2008, several case reports and retrospective studies have demonstrated Bortezomib’s efficacy in treating ABMR, especially in conjunction with other treatment options [17,142,143,144,145]. However, the BORTEJECT trial which is the sole study assessing Bortezomib's potential to prevent GFR decline in late DSA-positive ABMR among kidney transplant recipients, revealed no significant improvement in the eGFR slope, two-year graft survival, urinary protein levels, DSA concentrations, or rejection phenotypes compared to placebo. Additionally, Bortezomib was linked to notable gastrointestinal and hematologic toxicities [146].
4.3 Splenectomy
This surgical procedure is considered a rescue intervention when cases are refractory to plasmapheresis and/or IVIG. In fact, a case series of four patients with severe refractory ABMR showed improvement of urine output and a decrease of creatinine 48 hours following laparoscopic splenectomy [147]. However, this technique is not yet routinely recommended for the treatment of ABMR [135].
5. Novel Emerging Therapies
5.1 Carfilzomib
Carfilzomib is a second-generation irreversible proteasome inhibitor with a comparable mode of action to Bortezomib. This molecule was tested on six animal kidney transplant recipients along with Belatacept as a desensitizing agent. These two agents demonstrated a notably extended survival of the graft and decreased ABMR scores on kidney biopsy at 1 month. However, a progressive rebound of DSA and ABMR were then noticed in four of five animals with long-term graft survival [148]. Carfilzomib has only been used in the management of ABMR in patients with lung and heart transplants and showed promising outcomes [149,150]. No data exists yet for its use in treating ABMR in human kidney transplant recipients. Nevertheless, clinical trials in organ transplantation (CTOT-42) are ongoing to study the efficacy of Carfilzomib in the management of chronic active ABMR. Finally, some of the reported adverse events included fatigue, diarrhea, mild peripheral neuropathy, cytopenias such as thrombocytopenia and neutropenia, as well as both viral and bacterial infections. In some cases, septic shock led to fatal outcomes [151].
5.2 Interleukin-6 Antagonist Treatments
Interleukin-6 (IL-6) is an inflammatory cytokine with an established role in B-cell and T-cell stimulation, multiplication and function. Moreover, IL-6 has been identified as a critical component in key signaling pathways linked to rejection in transplant patients [152]. Many molecules have been developed that target either IL-6 or its receptor; from these molecules, we mention the ones below.
5.2.1 Tocilizumab
Tocilizumab is the pioneering recombinant humanized monoclonal antibody designed to target the interleukin-6 receptor (IL-6R). It is FDA-approved for the management of rheumatoid arthritis and juvenile idiopathic arthritis [151]. In kidney transplant, Tocilizumab has been used, alongside IVIG, as a desensitizing agent in a phase I/II trial and showed efficacy and safety in decreasing DSA levels and prohibiting the development of ABMR [153]. Not many published studies evaluating the use of Tocilizumab in acute ABMR exist; however, Pottebaum et al. examined using both Tocilizumab and standard therapy for acute ABMR in seven kidney transplant patients. The authors noticed either a stabilization or improvement in kidney function, along with a reduction of DSA concentrations by ≥50% observed in four patients. However, when Tocilizumab was administered for less than six months, many patients had recurrent rejection [154]. On the other hand, in chronic active ABMR, Tocilizumab has been broadly studied in multiple studies with encouraging outcomes [155,156]. In the most recent study published in 2021 by Noble et al., the authors conducted a retrospective study in 40 kidney transplant patients receiving Tocilizumab monthly for the management of chronic active ABMR. After 12 months of follow-up, there was no evidence of clinical or histological deterioration, except in patients who presented with more advanced clinical severity at the start of treatment [157]. In contrast, conflicting results were published by Kumar et al. regarding Tocilizumab’s safety and efficacy in 10 kidney transplant recipients with chronic active ABMR. Tocilizumab was given with a median of five doses. At a median 12-month follow-up, no improvements were observed in renal function, the trajectory of eGFR decline, or histological findings on protocol biopsies. In addition, one patient passed away due to a complicated hip infection [158].
5.2.2 Clazakizumab
Clazakizumab is a genetically engineered humanized monoclonal IgG1 antibody with high affinity for IL-6 [159]. Its use remains under careful investigation and has also been explored in conditions such as rheumatoid and psoriatic arthritis.
In a phase 2 single-center open-label study, Clazakizumab was given monthly to 10 kidney transplant recipients with refractory active ABMR for 12 months. The treatment demonstrated a trend toward stabilizing eGFR, lowering DSA titers, and reducing graft inflammation [160]. These findings have led to the initiation of a large phase 3 placebo-controlled clinical trial (IMAGINE), currently recruiting participants from North America, Europe, Asia, and Australia.
Additionally, Doberer et al. conducted a pilot randomized controlled trial to assess the safety and efficacy of Clazakizumab in late ABMR. In part A of the study, 20 kidney transplant recipients with positive DSA at least one year post-transplant were randomized to receive Clazakizumab or a placebo weekly for 12 weeks. Clazakizumab demonstrated a slower decline in eGFR and significantly reduced DSA titers compared to the placebo group. In part B, where all patients received Clazakizumab for an additional 40 weeks, the eGFR decline rate improved further. However, 25% of participants experienced serious adverse events, including diverticulitis, pleural effusions, and acute kidney injury [161].
Finally, Sitluximab and Sirukumab are additional interleukin-6 inhibitors that haven’t yet been evaluated in kidney transplantation [162,163].
5.2.3 Daratumumab
Daratumumab is a human immunoglobulin G1K monoclonal antibody. It is the first human-specific therapy in its class to target CD38-expressing plasma cells. This FDA-approved antibody is used for the treatment of newly diagnosed or relapsed/refractory multiple myeloma [151]. Its immunomodulatory effects are mediated through several Fc-dependent immune effector mechanisms, including antibody-dependent cellular phagocytosis, antibody-dependent cell-mediated cytotoxicity, complement-dependent cytotoxicity, and direct cellular apoptosis [164].
Notably, Daratumumab can interfere with cross-matching and red blood cell (RBC) antibody screening by binding to CD38 on RBCs, leading to a positive indirect Coombs test. Another key consideration is the potential for resistance during therapy, often attributed to complement-inhibitory proteins, such as CD55 and CD59, or the development of antidrug antibodies [165].
Kwun et al. investigated the use of Daratumumab as a desensitizing agent in a sensitized nonhuman primate model and in two clinical cases of combined heart–kidney transplantation for managing ABMR. The study demonstrated a significant reduction in DSA titers in both the animal model and clinical cases when Daratumumab was used for desensitization and ABMR management. However, in the nonhuman primate model, this reduction in DSA titers was not sustained, as all recipients experienced a rapid rebound of antibodies [166]. Additionally, numerous recent case reports have showed hopeful outcomes of Daratumumab in managing ABMR. One example is a case report published by Spica et al. in which Daratumumab was administered as a rescue therapy, after standard therapy failed, to a patient with ABO-incompatible kidney transplant and refractory ABMR. The blood group antibody concentration was reduced and sustained at low levels, facilitating the recovery of graft function [167]. To date, more than 304 clinical trials are being conducted to evaluate Daratumumab’s efficacy in numerous pathological conditions, one of which is its use as a desensitizing agent in kidney transplant [151].
Finally, Isatuximab is another example of a CD-38 inhibitor, approved for the management of multiple myeloma, but has not been tested yet in kidney transplant recipients [168].
5.2.4 Belimumab
Belimumab is a humanized IgG1 monoclonal antibody targeting B lymphocyte stimulator (BLyS). BLyS, also referred to as B-cell activating factor (BAFF), is a tumor necrosis factor (TNF) family cytokine that supports B-cell maturation, survival, and activation. Excessive BLyS expression is linked to autoimmunity. In kidney transplant recipients, elevated serum BLyS levels have been correlated with the development of de novo donor-specific antibodies (DSA) and an increased risk of antibody-mediated rejection (ABMR) [169,170]. Belimumab is FDA approved for the treatment of systemic lupus erythematosus and has been used in the management of lupus nephritis with proven efficacy [171]. Despite its use in lupus nephritis, Belimumab has not been widely investigated in the management of active ABMR. A case report described the use of Belimumab in a combined kidney–pancreas transplant recipient experiencing mixed rejection of the kidney allograft. Despite conventional treatments with TPE, Rituximab, and IVIG, persistent DSA and inflammation were observed. Belimumab administration resulted in a reduction of class II DSA levels and an improvement in graft function [172]. On the contrary, in a phase 2 randomized controlled trial, Belimumab was evaluated for the prevention of ABMR [173]. In this trial, 28 kidney transplant recipients were divided into 2 groups receiving either Belimumab or placebo, in addition to the conventional immunosuppressive drugs Basilixmab, mycophenolate, prednisone and tacrolimus. In conclusion, when combined to the standard therapy, Belimumab is effective and did not pose a higher risk of infection compared to the placebo.
5.2.5 Imlifidase
Imlifidase is an innovative recombinant cysteine protease produced by Streptococcus pyogenes that fragments human IgG antibodies at the lower hinge region of their heavy chains. This breaking disrupts complement-dependent cytotoxicity and antibody-dependent cellular cytotoxicity, impairs B-cell receptor function, and reduces natural killer (NK) cell activity [174]. This molecule was initially studied and continues to be investigated in ongoing research as a component of desensitization protocols [175]. Imlifidase shows promise as an innovative treatment option for managing ABMR. However, its use may be constrained by the risk of antibody rebound, including anti-imlifidase antibodies, DSAs, and total IgG levels. While our understanding of these risks remains limited, they may be mitigated by administering anti-CD20 antibodies and/or IVIg. Additionally, several significant drug interactions must be considered. Since imlifidase cleaves IgG antibodies, co-administered antibody-based therapies could be affected. To preserve the efficacy of concurrent treatments, it is recommended to separate the administration of IVIG by 12 hours, Rituximab, Basiliximab, rabbit antithymocyte globulin, and Alemtuzumab by 4 days, and Belatacept by 1 week [174].
In summary, while targeting IL-6 seems like a promising novel therapeutic option, recent studies have shown limited efficacy and disappointing results, particularly for Clazakizumab. While these molecules remain in clinical development, they will not be in routine practice for the management of ABMR in the foreseeable future. It is finally important to note that, given Tocilizumab’s proven nephroprotective effects in auto-immune kidney disease such as patients with ANCA vasculitis and glomerulonephritis with or without underlying rheumatoid arthritis and Tofacitinib’s high immunosuppressive efficacy, these anti-IL-6 therapies could be considered an alternative for the highly nephrotoxic calcineurin inhibitors [176].
5.3 Novel CD-20 Therapies
Rituximab, among the anti-CD20 antibody therapies, is a widely recognized treatment in solid organ transplantation and is frequently incorporated into contemporary strategies for managing ABMR in kidney transplant recipients. Currently, several studies have been published evaluating the effectiveness of other anti-CD20 monoclonal antibodies in treating antibody-mediated diseases. One example is Ofatumumab, a substitute anti-CD20 therapy approved for managing relapsing multiple sclerosis. In a published case report, this drug was given to a sensitized combined heart-kidney transplant patient along with TPE, IVIG and Tocilizumab as a desensitizing protocol. The patient did not exhibit rejection one year after the transplant with adequate kidney and heart allograft performance [177]. Additionally, Obinutuzumab, a glyco-engineered type II anti-CD20 monoclonal antibody, has demonstrated enhanced B-cell depletion compared to Rituximab, a type I anti-CD20 antibody. In the phase 1 THEORY trial, Obinutuzumab showed potential for B-cell depletion in desensitization protocols for highly sensitized kidney transplant candidates. However, its impact on reducing anti-HLA antibodies has been unreliable [178]. Finally, Inebilizumab is a therapeutic agent targeting CD19 expressed on B cells, leading to their depletion. An attempt to study this molecule in highly sensitized patients awaiting first or second kidney transplants failed due to insufficient participant recruitment in the study [179].
5.4 Adjustment of Immunosuppressive Regimens in ABMR
The addition of therapies targeting antibody-mediated rejection (AMR), such as plasmapheresis, intravenous immunoglobulin, anti-CD20 antibodies, or proteasome inhibitors, to standard immunosuppression increases the cumulative risk of infection and malignancy, particularly in the context of already potent maintenance regimens [85,180,181,182]. This risk is especially pronounced in patients with prior exposure to lymphocyte-depleting agents or those with comorbidities. Therefore, individualized adjustment of immunosuppressive protocols is necessary to balance the efficacy of AMR treatment with the minimization of adverse effects [182,183]. One of the adopted approaches is the minimization, withdrawal, or substitution of calcineurin inhibitors (CNIs), all while considering the increased risk of acute rejection [184,185]. Practically, protocol modifications in response to AMR may include switching from azathioprine to mycophenolate mofetil (MMF), from MMF to Sirolimus, or from Cyclosporine to Tacrolimus, as well as increasing the dose of MMF or corticosteroids [180,182]. In patients with infection, malignancy, or post-transplant lymphoproliferative disorder, immunosuppression is often reduced or modified, with mammalian target of rapamycin (mTOR) inhibitors-based regimens considered in certain malignancy scenarios and CNI minimization or withdrawal in the setting of viral-driven lymphoproliferative disease [182,183,186].
Finally, while several fundamental features that define antibody-mediated rejection are shared between the Banff and KDIGO frameworks, important differences remain in their diagnostic and therapeutic emphasis. This is highlighted in Table 2.
Table 2 Summary Table comparing KDIGO and Banff Guidelines on ABMR.

6. Limitations
Our review provides a comprehensive overview of the current understanding of ABMR in kidney transplantation, yet several limitations must be acknowledged. First, while the pathophysiology of ABMR is increasingly understood, many mechanistic insights remain based on preclinical models or limited patient cohorts, which may not capture the full heterogeneity of clinical presentations. Additionally, diagnostic criteria continue to evolve, and while the Banff classification provides a structured framework, its reliance on histopathology and DSA detection may result in diagnostic ambiguity, especially in borderline or mixed rejection phenotypes. Also, treatment recommendations, both standard and emerging, are largely informed by small-scale studies or expert consensus, with a relative scarcity of large randomized controlled trials (RCTs) directly comparing therapeutic strategies. Finally, the rapidly evolving landscape of novel immunomodulatory therapies introduces both promise and uncertainty, as long-term safety and efficacy data remain limited. These factors underscore the need for ongoing research, collaborative registries, and more robust clinical trials to refine the management of ABMR and improve transplant outcomes.
7. Conclusion
ABMR in clinical practice is a complex process with a wide gray zone area. The histologic criteria, primarily guided by the Banff classification, have played an invaluable role in standardizing the diagnosis of ABMR. Evidence underscores the significant role of DSA in both acute and chronic ABMR. However, histologic assessment of biopsy samples has low specificity and high interobserver variability, moreover, not all DSA detected by current assays cause injury to the allograft and ABMR can often come with undetectable DSAs. Likewise, C4d has significant limitations as a marker of ABMR. It also remains unclear which combination therapy is the most effective all while considering drug toxicity. While much progress has been made in advancing our understanding, research attempts are ongoing to resolve the ambiguous events and to define risk stratification strategies for ABMR phenotypes to guide prevention and therapy. Modern tools to improve diagnosis, especially when facing increasingly complex phenotypes must be available moving forward. Reliable biomarkers for diagnosis and prognosis as well as prediction models have shown promising findings however, there remains significant potential for further optimization and improvement.
Author Contributions
LMS and MD contributed equally to writing the review and would like to share first co-authorship. LMS, MD and TM wrote the manuscript under the supervision of JA and JC.
Funding
The authors received no financial support for the research, authorship and publication of this article.
Competing Interests
The author declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as potential conflict of interest.
References
- Kramer A, Boenink R, Stel VS, Santiuste de Pablos C, Tomović F, Golan E, et al. The ERA-EDTA registry annual report 2018: A summary. Clin Kidney J. 2021; 14: 107-123. [CrossRef] [Google scholar] [PubMed]
- Matas AJ, Smith JM, Skeans MA, Lamb KE, Gustafson SK, Samana CJ, et al. OPTN/SRTR 2011 annual data report: Kidney. Am J Transplant. 2013; 13: 11-46. [CrossRef] [Google scholar] [PubMed]
- Matas AJ, Smith JM, Skeans MA, Thompson B, Gustafson SK, Stewart DE, et al. OPTN/SRTR 2013 annual data report: Kidney. Am J Transplant. 2015; 15: 1-34. [CrossRef] [Google scholar] [PubMed]
- Meier-Kriesche HU, Schold JD, Srinivas TR, Kaplan B. Lack of improvement in renal allograft survival despite a marked decrease in acute rejection rates over the most recent era. Am J Transplant. 2004; 4: 378-383. [CrossRef] [Google scholar] [PubMed]
- Parajuli S, Aziz F, Garg N, Panzer SE, Joachim E, Muth B, et al. Histopathological characteristics and causes of kidney graft failure in the current era of immunosuppression. World J Transplant. 2019; 9: 123-133. [CrossRef] [Google scholar] [PubMed]
- Loupy A, Vernerey D, Tinel C, Aubert O, van Huyen JP, Rabant M, et al. Subclinical rejection phenotypes at 1 year post-transplant and outcome of kidney allografts. J Am Soc Nephrol. 2015; 26: 1721-1731. [CrossRef] [Google scholar] [PubMed]
- Cornell LD. Histopathologic features of antibody mediated rejection: The Banff classification and beyond. Front Immunol. 2021; 12: 718122. [CrossRef] [Google scholar] [PubMed]
- Eckardt KU, Kasiske BL, Zeier MG. KDIGO clinical practice guideline for the care of kidney transplant recipients. Am J Transplant. 2009; 9: S1-S155. [CrossRef] [Google scholar] [PubMed]
- Tamara F, Fajar JK, Gersom C, Wicaksono RS, Tupamahu AR, Huda FN, et al. Global prevalence and contributing factors of transplant renal artery stenosis in renal transplant recipients: A systematic review and meta-analysis. Narra J. 2024; 4: e1782. [CrossRef] [Google scholar] [PubMed]
- Zhang X, Jordan SC. Assessment of IgM DSAs in transplant recipients: Relationship to de novo IgG DSAs and risk for antibody rejection. Transplant Direct. 2024; 10: e1583. [CrossRef] [Google scholar] [PubMed]
- Willicombe M, Brookes P, Sergeant R, Santos-Nunez E, Steggar C, Galliford J, et al. De novo DQ donor-specific antibodies are associated with a significant risk of antibody-mediated rejection and transplant glomerulopathy. Transplantation. 2012; 94: 172-177. [CrossRef] [Google scholar] [PubMed]
- Zhang R. Donor-specific antibodies in kidney transplant recipients. Clin J Am Soc Nephrol. 2018; 13: 182-192. [CrossRef] [Google scholar] [PubMed]
- López del Moral C, Wu K, Naik M, Osmanodja B, Akifova A, Lachmann N, et al. The natural history of de novo donor-specific HLA antibodies after kidney transplantation. Front Med. 2022; 9: 943502. [CrossRef] [Google scholar] [PubMed]
- Tian Y, Frischknecht L, Mallone A, Rössler F, Schachtner T, Nilsson J. Evaluation of de novo donor specific antibodies after kidney transplantation in the era of donor-derived cell-free DNA. Front Immunol. 2025; 15: 1530065. [CrossRef] [Google scholar] [PubMed]
- Pierini F, Nutsua M, Böhme L, Özer O, Bonczarowska J, Susat J, et al. Targeted analysis of polymorphic loci from low-coverage shotgun sequence data allows accurate genotyping of HLA genes in historical human populations. Sci Rep. 2020; 10: 7339. [CrossRef] [Google scholar] [PubMed]
- Montgomery RA, Tatapudi VS, Leffell MS, Zachary AA. HLA in transplantation. Nat Rev Nephrol. 2018; 14: 558-570. [CrossRef] [Google scholar] [PubMed]
- Flechner SM, Fatica R, Askar M, Stephany BR, Poggio E, Koo A, 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: 1486-1492. [CrossRef] [Google scholar] [PubMed]
- Blum JS, Wearsch PA, Cresswell P. Pathways of antigen processing. Annu Rev Immunol. 2013; 31: 443-473. [CrossRef] [Google scholar] [PubMed]
- Cross AR, Lion J, Poussin K, Glotz D, Mooney N. Inflammation determines the capacity of allogenic endothelial cells to regulate human Treg expansion. Front Immunol. 2021; 12: 666531. [CrossRef] [Google scholar] [PubMed]
- Steinhoff G, Wonigeit K, Pichlmayr R. Analysis of sequential changes in major histocompatibility complex expression in human liver grafts after transplantation. Transplantation. 1988; 45: 394-401. [CrossRef] [Google scholar] [PubMed]
- Zachary AA, Leffell MS. HLA mismatching strategies for solid organ transplantation–A balancing act. Front Immunol. 2016; 7: 575. [CrossRef] [Google scholar] [PubMed]
- Béland S, Désy O, El Fekih R, Marcoux M, Thivierge MP, Desgagné JS, et al. Expression of class II human leukocyte antigens on human endothelial cells shows high interindividual and intersubclass heterogeneity. J Am Soc Nephrol. 2023; 34: 846-856. [CrossRef] [Google scholar] [PubMed]
- Senev A, Ray B, Lerut E, Hariharan J, Heylen C, Kuypers D, et al. The pre-transplant non-HLA antibody burden associates with the development of histology of antibody-mediated rejection after kidney transplantation. Front Immunol. 2022; 13: 809059. [CrossRef] [Google scholar] [PubMed]
- Alelign T, Ahmed MM, Bobosha K, Tadesse Y, Howe R, Petros B. Kidney transplantation: The challenge of human leukocyte antigen and its therapeutic strategies. J Immunol Res. 2018; 2018: 5986740. [CrossRef] [Google scholar] [PubMed]
- Senev A, Coemans M, Lerut E, Van Sandt V, Daniëls L, Kuypers D, et al. Histological picture of antibody-mediated rejection without donor-specific anti-HLA antibodies: Clinical presentation and implications for outcome. Am J Transplant. 2019; 19: 763-780. [CrossRef] [Google scholar] [PubMed]
- Zorn E, See SB. Antibody responses to minor histocompatibility antigens after solid organ transplantation. Transplantation. 2022; 106: 749-753. [CrossRef] [Google scholar] [PubMed]
- Spierings E. Minor histocompatibility antigens: Past, present, and future. Tissue Antigens. 2014; 84: 374-360. [CrossRef] [Google scholar] [PubMed]
- Tan JC, Wadia PP, Coram M, Grumet FC, Kambham N, Miller K, et al. HY antibody development associates with acute rejection in female patients with male kidney transplants. Transplantation. 2008; 86: 75-81. [CrossRef] [Google scholar] [PubMed]
- Carapito R, Aouadi I, Verniquet M, Untrau M, Pichot A, Beaudrey T, et al. The MHC class I MICA gene is a histocompatibility antigen in kidney transplantation. Nat Med. 2022; 28: 989-998. [CrossRef] [Google scholar] [PubMed]
- Zou Y, Stastny P, Süsal C, Döhler B, Opelz G. Antibodies against MICA antigens and kidney-transplant rejection. N Engl J Med. 2007; 357: 1293-1300. [CrossRef] [Google scholar] [PubMed]
- Risti M, Bicalho MD. MICA and NKG2D: Is there an impact on kidney transplant outcome? Front Immunol. 2017; 8: 179. [CrossRef] [Google scholar] [PubMed]
- Baranwal AK, Mehra NK. Major histocompatibility complex class I chain-related A (MICA) molecules: Relevance in solid organ transplantation. Front Immunol. 2017; 8: 182. [CrossRef] [Google scholar] [PubMed]
- Claas FH, Paul LC, Van Es LA, Van Rood JJ. Antibodies against donor antigens on endothelial cells and monocytes in eluates of rejected kidney allografts. Tissue Antigens. 1980; 15: 19-24. [CrossRef] [Google scholar] [PubMed]
- Breimer ME, Rydberg L, Jackson AM, Lucas DP, Zachary AA, Melancon JK, et al. Multicenter evaluation of a novel endothelial cell crossmatch test in kidney transplantation. Transplantation. 2009; 87: 549-556. [CrossRef] [Google scholar] [PubMed]
- Pearl MH, Zhang Q, Diaz MF, Grotts J, Rossetti M, Elashoff D, et al. Angiotensin II Type 1 receptor antibodies are associated with inflammatory cytokines and poor clinical outcomes in pediatric kidney transplantation. Kidney Int. 2018; 93: 260-269. [CrossRef] [Google scholar] [PubMed]
- Moreso F, Crespo M, Ruiz JC, Torres A, Gutierrez-Dalmau A, Osuna A, 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: 927-935. [CrossRef] [Google scholar] [PubMed]
- Philogene MC, Johnson T, Vaught AJ, Zakaria S, Fedarko N. Antibodies against angiotensin II type 1 and endothelin A receptors: Relevance and pathogenicity. Hum Immunol. 2019; 80: 561-567. [CrossRef] [Google scholar] [PubMed]
- Reindl‐Schwaighofer R, Heinzel A, Gualdoni GA, Mesnard L, Claas FH, Oberbauer R. Novel insights into non‐HLA alloimmunity in kidney transplantation. Transpl Int. 2020; 33: 5-17. [CrossRef] [Google scholar] [PubMed]
- Deltombe C, Gillaizeau F, Anglicheau D, Morelon E, Trébern-Launay K, Le Borgne F, et al. Is pre-transplant sensitization against angiotensin II type 1 receptor still a risk factor of graft and patient outcome in kidney transplantation in the anti-HLA Luminex era? A retrospective study. Transpl Int. 2017; 30: 1150-1160. [CrossRef] [Google scholar] [PubMed]
- Althaf MM, El Kossi M, Jin JK, Sharma A, Halawa AM. Human leukocyte antigen typing and crossmatch: A comprehensive review. World J Transplant. 2017; 7: 339-348. [CrossRef] [Google scholar] [PubMed]
- Morris AB, Sullivan HC, Krummey SM, Gebel HM, Bray RA. Out with the old, in with the new: Virtual versus physical crossmatching in the modern era. HLA. 2019; 94: 471-481. [CrossRef] [Google scholar] [PubMed]
- Rocha Y, Jaramillo A, Neumann J, Hacke K, Palou E, Torres J. Crossmatch assays in transplantation: Physical or virtual?: A review. Medicine. 2023; 102: e36527. [CrossRef] [Google scholar] [PubMed]
- 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] [Google scholar] [PubMed]
- Bhaskaran MC, Heidt S, Muthukumar T. Principles of virtual crossmatch testing for kidney transplantation. Kidney Int Rep. 2022; 7: 1179-1188. [CrossRef] [Google scholar] [PubMed]
- Pontes LF, Carvalho L, Stumbo AC, Porto LC. Detection and localization of non-HLA-ABC antigenic sites relevant to kidney rejection on endothelial cells. J Immunol Methods. 2001; 251: 73-80. [CrossRef] [Google scholar] [PubMed]
- Kamburova EG, Kardol-Hoefnagel T, Wisse BW, Joosten I, Allebes WA, Van der Meer A, et al. Development and validation of a multiplex non-HLA antibody assay for the screening of kidney transplant recipients. Front Immunol. 2018; 9: 3002. [CrossRef] [Google scholar] [PubMed]
- Kamburova EG, Gruijters ML, Kardol-Hoefnagel T, Wisse BW, Joosten I, Allebes WA, et al. Antibodies against ARHGDIB are associated with long-term kidney graft loss. Am J Transplant. 2019; 19: 3335-3344. [CrossRef] [Google scholar] [PubMed]
- Reindl-Schwaighofer R, Heinzel A, Kainz A, van Setten J, Jelencsics K, Hu K, et al. Contribution of non-HLA incompatibility between donor and recipient to kidney allograft survival: Genome-wide analysis in a prospective cohort. Lancet. 2019; 393: 910-917. [CrossRef] [Google scholar] [PubMed]
- Li L, Wadia P, Chen R, Kambham N, Naesens M, Sigdel TK, et al. Identifying compartment-specific non-HLA targets after renal transplantation by integrating transcriptome and “antibodyome” measures. Proc Natl Acad Sci USA. 2009; 106: 4148-4153. [CrossRef] [Google scholar] [PubMed]
- Mohamed M, Sweeney T, Alkhader D, Nassar M, Alqassieh A, Lakhdar S, et al. ABO incompatibility in renal transplantation. World J Transplant. 2021; 11: 388-399. [CrossRef] [Google scholar] [PubMed]
- Morath C, Zeier M, Döhler B, Opelz G, Süsal C. ABO-incompatible kidney transplantation. Front Immunol. 2017; 8: 234. [CrossRef] [Google scholar] [PubMed]
- Breimer ME, Mölne J, Nordén G, Rydberg L, Thiel G, Svalander CT. Blood group A and B antigen expression in human kidneys correlated to A1/A2/B, Lewis, and secretor status. Transplantation. 2006; 82: 479-485. [CrossRef] [Google scholar] [PubMed]
- Deng Y, Frischnknecht L, Wehmeier C, de Rougemont O, Villard J, Ferrari-Lacraz S, et al. Pre-transplant donor specific antibodies in ABO incompatible kidney transplantation–data from the Swiss transplant cohort study. Front Immunol. 2024; 15: 1355128. [CrossRef] [Google scholar] [PubMed]
- Benamu E, Montoya JG. Infections associated with the use of eculizumab: Recommendations for prevention and prophylaxis. Curr Opin Infect Dis. 2016; 29: 319-329. [CrossRef] [Google scholar] [PubMed]
- Heo GY, Jung M, Piao H, Kim HJ, Kim HW, Lee J, et al. Successful eculizumab treatment as an adjunctive therapy to desensitization in ABO-incompatible living donor kidney transplantation and its molecular phenotypes. Front Immunol. 2024; 15: 1465851. [CrossRef] [Google scholar] [PubMed]
- Li Q, Lan P. Activation of immune signals during organ transplantation. Signal Transduct Target Ther. 2023; 8: 110. [CrossRef] [Google scholar] [PubMed]
- Siu JH, Surendrakumar V, Richards JA, Pettigrew GJ. T cell allorecognition pathways in solid organ transplantation. Front Immunol. 2018; 9: 2548. [CrossRef] [Google scholar] [PubMed]
- Cucchiari D, Podestà MA, Ponticelli C. Pathophysiology of rejection in kidney transplantation. Expert Rev Clin Immunol. 2024; 20: 1471-1481. [CrossRef] [Google scholar] [PubMed]
- Liu J, Zhang X, Cheng Y, Cao X. Dendritic cell migration in inflammation and immunity. Cell Mol Immunol. 2021; 18: 2461-2471. [CrossRef] [Google scholar] [PubMed]
- Crotty S. T follicular helper cell differentiation, function, and roles in disease. Immunity. 2014; 41: 529-542. [CrossRef] [Google scholar] [PubMed]
- van Besouw NM, Mendoza Rojas A, Baan CC. The role of follicular T helper cells in the humoral alloimmune response after clinical organ transplantation. HLA. 2019; 94: 407-414. [CrossRef] [Google scholar] [PubMed]
- Zhang H, Sage PT. Role of T follicular helper and T follicular regulatory cells in antibody-mediated rejection: New therapeutic targets? Curr Opin Organ Transplant. 2022; 27: 371-375. [CrossRef] [Google scholar] [PubMed]
- Hale JS, Ahmed R. Memory T follicular helper CD4 T cells. Front Immunol. 2015; 6: 16. [CrossRef] [Google scholar] [PubMed]
- Sharpe AH, Freeman GJ. The B7–CD28 superfamily. Nat Rev Immunol. 2002; 2: 116-126. [CrossRef] [Google scholar] [PubMed]
- Lakkis FG, Lechler RI. Origin and biology of the allogeneic response. Cold Spring Harb Perspect Med. 2013; 3: a014993. [CrossRef] [Google scholar] [PubMed]
- Marino J, Paster J, Benichou G. Allorecognition by T lymphocytes and allograft rejection. Front Immunol. 2016; 7: 582. [CrossRef] [Google scholar] [PubMed]
- Chong AS. B cells as antigen-presenting cells in transplantation rejection and tolerance. Cell Immunol. 2020; 349: 104061. [CrossRef] [Google scholar] [PubMed]
- Gatto D, Brink R. The germinal center reaction. J Allergy Clin Immunol. 2010; 126: 898-907. [CrossRef] [Google scholar] [PubMed]
- Luque S, Lúcia M, Bestard O. Refinement of humoral immune monitoring in kidney transplantation: The role of “hidden” alloreactive memory B cells. Transpl Int. 2017; 30: 955-968. [CrossRef] [Google scholar] [PubMed]
- Bouquegneau A, Loheac C, Aubert O, Bouatou Y, Viglietti D, Empana JP, et al. Complement-activating donor-specific anti-HLA antibodies and solid organ transplant survival: A systematic review and meta-analysis. PLoS Med. 2018; 15: e1002572. [CrossRef] [Google scholar] [PubMed]
- Tedesco F, Pausa M, Nardon E, Introna M, Mantovani A, Dobrina A. The cytolytically inactive terminal complement complex activates endothelial cells to express adhesion molecules and tissue factor procoagulant activity. J Exp Med. 1997; 185: 1619-1628. [CrossRef] [Google scholar] [PubMed]
- Song G, Wang S, Barkestani MN, Mullan C, Fan M, Jiang B, et al. Membrane attack complexes, endothelial cell activation, and direct allorecognition. Front Immunol. 2022; 13: 1020889. [CrossRef] [Google scholar] [PubMed]
- Stites E, Le Quintrec M, Thurman JM. The complement system and antibody-mediated transplant rejection. J Immunol. 2015; 195: 5525-5531. [CrossRef] [Google scholar] [PubMed]
- Legris T, Picard C, Todorova D, Lyonnet L, Laporte C, Dumoulin C, et al. Antibody-dependent NK cell activation is associated with late kidney allograft dysfunction and the complement-independent alloreactive potential of donor-specific antibodies. Front Immunol. 2016; 7: 288. [CrossRef] [Google scholar] [PubMed]
- Orandi BJ, Alachkar N, Kraus ES, Naqvi F, Lonze BE, Lees L, et al. Presentation and outcomes of C4d‐negative antibody‐mediated rejection after kidney transplantation. Am J Transplant. 2016; 16: 213-220. [CrossRef] [Google scholar] [PubMed]
- Strainic MG, Liu J, Huang D, An F, Lalli PN, Muqim N, et al. Locally produced complement fragments C5a and C3a provide both costimulatory and survival signals to naive CD4+ T cells. Immunity. 2008; 28: 425-435. [CrossRef] [Google scholar] [PubMed]
- Dunkelberger JR, Song WC. Complement and its role in innate and adaptive immune responses. Cell Res. 2010; 20: 34-50. [CrossRef] [Google scholar] [PubMed]
- Stegall MD, Chedid MF, Cornell LD. The role of complement in antibody-mediated rejection in kidney transplantation. Nat Rev Nephrol. 2012; 8: 670-678. [CrossRef] [Google scholar] [PubMed]
- Anwar IJ, DeLaura I, Ladowski J, Gao Q, Knechtle SJ, Kwun J. Complement-targeted therapies in kidney transplantation-insights from preclinical studies. Front Immunol. 2022; 13: 984090. [CrossRef] [Google scholar] [PubMed]
- Naesens M, Roufosse C, Haas M, Lefaucheur C, Mannon RB, Adam BA, et al. The Banff 2022 Kidney Meeting Report: Reappraisal of microvascular inflammation and the role of biopsy-based transcript diagnostics. Am J Transplant. 2024; 24: 338-349. [CrossRef] [Google scholar] [PubMed]
- Ordikhani F, Pothula V, Sanchez-Tarjuelo R, Jordan S, Ochando J. Macrophages in organ transplantation. Front Immunol. 2020; 11: 582939. [CrossRef] [Google scholar] [PubMed]
- Wu Y, Hirschi KK. Tissue-resident macrophage development and function. Fronti Cell Dev Biol. 2021; 8: 617879. [CrossRef] [Google scholar] [PubMed]
- Kopecky BJ, Dun H, Amrute JM, Lin CY, Bredemeyer AL, Terada Y, et al. Donor macrophages modulate rejection after heart transplantation. Circulation. 2022; 146: 623-638. [CrossRef] [Google scholar] [PubMed]
- Guilliams M, Bruhns P, Saeys Y, Hammad H, Lambrecht BN. The function of Fcγ receptors in dendritic cells and macrophages. Nat Rev Immunol. 2014; 14: 94-108. [CrossRef] [Google scholar] [PubMed]
- Loupy A, Lefaucheur C. Antibody-mediated rejection of solid-organ allografts. N Engl Med. 2018; 379: 1150-1160. [CrossRef] [Google scholar] [PubMed]
- Pontrelli P, Rascio F, Castellano G, Grandaliano G, Gesualdo L, Stallone G. The role of natural killer cells in the immune response in kidney transplantation. Front Immunol. 2020; 11: 1454. [CrossRef] [Google scholar] [PubMed]
- Turner JE, Rickassel C, Healy H, Kassianos AJ. Natural killer cells in kidney health and disease. Front Immunol. 2019; 10: 587. [CrossRef] [Google scholar] [PubMed]
- Hidalgo LG, Sis B, Sellares J, Campbell PM, Mengel M, Einecke G, 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: 1812-1822. [CrossRef] [Google scholar] [PubMed]
- Kenta I, Takaaki K. Molecular mechanisms of antibody-mediated rejection and accommodation in organ transplantation. Nephron. 2020; 144: 2-6. [CrossRef] [Google scholar] [PubMed]
- Venner JM, Hidalgo LG, Famulski KS, Chang J, Halloran PF. The molecular landscape of antibody-mediated kidney transplant rejection: Evidence for NK involvement through CD16a Fc receptors. Am J Transplant. 2015; 15: 1336-1348. [CrossRef] [Google scholar] [PubMed]
- Zhao H, Alam A, Soo AP, George AJ, Ma D. Ischemia-reperfusion injury reduces long term renal graft survival: Mechanism and beyond. EBioMedicine. 2018; 28: 31-42. [CrossRef] [Google scholar] [PubMed]
- Salvadori M, Rosso G, Bertoni E. Update on ischemia-reperfusion injury in kidney transplantation: Pathogenesis and treatment. World J Transplant. 2015; 5: 52-67. [CrossRef] [Google scholar] [PubMed]
- Nieuwenhuijs-Moeke GJ, Pischke SE, Berger SP, Sanders JS, Pol RA, Struys MM, et al. Ischemia and reperfusion injury in kidney transplantation: Relevant mechanisms in injury and repair. J Clin Med. 2020; 9: 253. [CrossRef] [Google scholar] [PubMed]
- Gui Z, Al Moussawy M, Sanders SM, Abou-Daya KI. Innate allorecognition in transplantation: Ancient mechanisms with modern impact. Transplantation. 2024; 108: 1524-1531. [CrossRef] [Google scholar] [PubMed]
- Land WG, Agostinis P, Gasser S, Garg AD, Linkermann A. Transplantation and damage-associated molecular patterns (DAMPs). Am J Transplant. 2016; 16: 3338-3361. [CrossRef] [Google scholar] [PubMed]
- Schröppel B, Legendre C. Delayed kidney graft function: From mechanism to translation. Kidney Int. 2014; 86: 251-258. [CrossRef] [Google scholar] [PubMed]
- Lasorsa F, Rutigliano M, Milella M, d’Amati A, Crocetto F, Pandolfo SD, et al. Ischemia-reperfusion injury in kidney transplantation: Mechanisms and potential therapeutic targets. Int J Mol Sci. 2024; 25: 4332. [CrossRef] [Google scholar] [PubMed]
- Zhang Q, Raoof M, Chen Y, Sumi Y, Sursal T, Junger W, et al. Circulating mitochondrial DAMPs cause inflammatory responses to injury. Nature. 2010; 464: 104-107. [CrossRef] [Google scholar] [PubMed]
- Kim K, Moon H, Lee YH, Seo JW, Kim YG, Moon JY, et al. Clinical relevance of cell-free mitochondrial DNA during the early postoperative period in kidney transplant recipients. Sci Rep. 2019; 9: 18607. [CrossRef] [Google scholar] [PubMed]
- Cron DC, Patzer RE, Adler JT. Supply, demand, and a growing US kidney transplant waiting list. JAMA Netw Open. 2025; 8: e251675. [CrossRef] [Google scholar] [PubMed]
- Alsolami E, Lotfy K, Alkhunaizi A, Cuerden M, Weir MA, House AA. Impact of donation after circulatory death on outcomes of expanded criteria donor kidney transplants. Transplant Proc. 2024; 56: 50-57. [CrossRef] [Google scholar] [PubMed]
- Urbanellis P, Mazilescu L, Kollmann D, Linares-Cervantes I, Kaths JM, Ganesh S, et al. Prolonged warm ischemia time leads to severe renal dysfunction of donation-after-cardiac death kidney grafts. Sci Rep. 2021; 11: 17930. [CrossRef] [Google scholar] [PubMed]
- Jiang S, Su H. Exploration of the shared gene signatures and biological mechanisms between ischemia-reperfusion injury and antibody-mediated rejection in renal transplantation. Transpl Immunol. 2024; 83: 102001. [CrossRef] [Google scholar] [PubMed]
- Molinero LL, Alegre ML. Role of T cell-nuclear factor κB in transplantation. Transplant Rev. 2012; 26: 189-200. [CrossRef] [Google scholar] [PubMed]
- Kalogeris T, Baines CP, Krenz M, Korthuis RJ. Cell biology of ischemia/reperfusion injury. Int Rev Cell Mol Biol. 2012; 298: 229-317. [CrossRef] [Google scholar] [PubMed]
- Bonventre JV, Yang L. Cellular pathophysiology of ischemic acute kidney injury. J Clin Invest. 2011; 121: 4210-4221. [CrossRef] [Google scholar] [PubMed]
- Montenovo MI, Perkins JD, Kling CE, Sibulesky L, Dick AA, Reyes JD. Machine perfusion decreases delayed graft function in donor grafts with high kidney donor profile index. Exp Clin Transplant. 2020; 19: 8-13. [CrossRef] [Google scholar] [PubMed]
- Ghoneima AS, Sousa Da Silva RX, Gosteli MA, Barlow AD, Kron P. Outcomes of kidney perfusion techniques in transplantation from deceased donors: A systematic review and meta-analysis. J Clin Med. 2023; 12: 3871. [CrossRef] [Google scholar] [PubMed]
- Ott LC, Cuenca AG. Innate immune cellular therapeutics in transplantation. Front Transplant. 2023; 2: 1067512. [CrossRef] [Google scholar] [PubMed]
- Gershon RK, Kondo K. Infectious immunological tolerance. Immunology. 1971; 21: 903-914. [Google scholar]
- Sakaguchi S, Sakaguchi N, Asano M, Itoh M, Toda M. Immunologic self-tolerance maintained by activated T cells expressing IL-2 receptor alpha-chains (CD25). Breakdown of a single mechanism of self-tolerance causes various autoimmune diseases. J Immunol. 1995; 155: 1151-1164. [CrossRef] [Google scholar]
- Hori S, Nomura T, Sakaguchi S. Control of regulatory T cell development by the transcription factor Foxp3. Science. 2003; 299: 1057-1061. [CrossRef] [Google scholar] [PubMed]
- Fontenot JD, Gavin MA, Rudensky AY. Foxp3 programs the development and function of CD4+CD25+ regulatory T cells. Nat Immunol. 2003; 4: 330-336. [CrossRef] [Google scholar] [PubMed]
- Kim HJ, Barnitz RA, Kreslavsky T, Brown FD, Moffett H, Lemieux ME, et al. Stable inhibitory activity of regulatory T cells requires the transcription factor Helios. Science. 2015; 350: 334-339. [CrossRef] [Google scholar] [PubMed]
- Cassano A, Chong AS, Alegre ML. Tregs in transplantation tolerance: Role and therapeutic potential. Front Transplant. 2023; 2: 1217065. [CrossRef] [Google scholar] [PubMed]
- Sula Karreci E, Eskandari SK, Dotiwala F, Routray SK, Kurdi AT, Assaker JP, et al. Human regulatory T cells undergo self-inflicted damage via granzyme pathways upon activation. JCI Insight. 2017; 2: e91599. [CrossRef] [Google scholar] [PubMed]
- Lim HW, Hillsamer P, Banham AH, Kim CH. Cutting edge: Direct suppression of B cells by CD4+ CD25+ regulatory T cells. J Immunol. 2005; 175: 4180-4183. [CrossRef] [Google scholar] [PubMed]
- Mishra S, Srinivasan S, Ma C, Zhang N. CD8+ regulatory T cell–a mystery to be revealed. Front Immunol. 2021; 12: 708874. [CrossRef] [Google scholar] [PubMed]
- Kim HJ, Verbinnen B, Tang X, Lu L, Cantor H. Inhibition of follicular T-helper cells by CD8+ regulatory T cells is essential for self tolerance. Nature. 2010; 467: 328-332. [CrossRef] [Google scholar] [PubMed]
- Choi JY, Eskandari SK, Cai S, Sulkaj I, Assaker JP, Allos H, et al. Regulatory CD8 T cells that recognize Qa-1 expressed by CD4 T-helper cells inhibit rejection of heart allografts. Proc Natl Acad Sci USA. 2020; 117: 6042-6046. [CrossRef] [Google scholar] [PubMed]
- Müller F, Taubmann J, Bucci L, Wilhelm A, Bergmann C, Völkl S, et al. CD19 CAR T-Cell Therapy in Autoimmune Disease - A Case Series with Follow-up. N Engl J Med. 2024; 390: 687-700. [CrossRef] [Google scholar] [PubMed]
- Zhang F, Zhang J, Cao P, Sun Z, Wang W. The characteristics of regulatory macrophages and their roles in transplantation. Int Immunopharmacol. 2021; 91: 107322. [CrossRef] [Google scholar] [PubMed]
- Riquelme P, Haarer J, Kammler A, Walter L, Tomiuk S, Ahrens N, et al. TIGIT+ iTregs elicited by human regulatory macrophages control T cell immunity. Nat Commun. 2018; 9: 2858. [CrossRef] [Google scholar] [PubMed]
- Hutchinson JA, Riquelme P, Brem-Exner BG, Schulze M, Matthäi M, Renders L, et al. Transplant acceptance-inducing cells as an immune-conditioning therapy in renal transplantation. Transpl Int. 2008; 21: 728-741. [CrossRef] [Google scholar] [PubMed]
- Bia M, Adey DB, Bloom RD, Chan L, Kulkarni S, Tomlanovich S. KDOQI US commentary on the 2009 KDIGO clinical practice guideline for the care of kidney transplant recipients. Am J Kidney Dis. 2010; 56: 189-218. [CrossRef] [Google scholar] [PubMed]
- Filippone EJ, Farber JL. The problem of subclinical antibody-mediated rejection in kidney transplantation. Transplantation. 2021; 105: 1176-1187. [CrossRef] [Google scholar] [PubMed]
- Alasfar S, Kodali L, Schinstock CA. Current therapies in kidney transplant rejection. J Clin Med. 2023; 12: 4927. [CrossRef] [Google scholar] [PubMed]
- Solez KI, Axelsen RA, Benediktsson H, Burdick JF, Cohen AH, Colvin RB, et al. International standardization of criteria for the histologic diagnosis of renal allograft rejection: The Banff working classification of kidney transplant pathology. Kidney Int. 1993; 44: 411-422. [CrossRef] [Google scholar] [PubMed]
- Loupy A, Haas M, Roufosse C, Naesens M, Adam B, Afrouzian M, et al. The Banff 2019 Kidney Meeting Report (I): Updates on and clarification of criteria for T cell– and antibody-mediated rejection. Am J Transplant. 2020; 20: 2318-2831. [CrossRef] [Google scholar] [PubMed]
- Loupy A, Suberbielle-Boissel C, Hill GS, Lefaucheur C, Anglicheau D, Zuber J, et al. Outcome of subclinical antibody-mediated rejection in kidney transplant recipients with preformed donor-specific antibodies. Am J Transplant. 2009; 9: 2561-2570. [CrossRef] [Google scholar] [PubMed]
- Mauiyyedi S, Pelle PD, Saidman S, Collins AB, Pascual M, Tolkoff-Rubin NE, 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: 574-582. [CrossRef] [Google scholar] [PubMed]
- Yoo D, Goutaudier V, Divard G, Gueguen J, Astor BC, Aubert O, et al. An automated histological classification system for precision diagnostics of kidney allografts. Nat Med. 2023; 29: 1211-1220. [CrossRef] [Google scholar] [PubMed]
- Labriffe M, Woillard JB, Gwinner W, Braesen JH, Anglicheau D, Rabant M, et al. Machine learning-supported interpretation of kidney graft elementary lesions in combination with clinical data. Am J Transplant. 2022; 22: 2821-2833. [CrossRef] [Google scholar] [PubMed]
- Wan SS, Ying TD, Wyburn K, Roberts DM, Wyld M, Chadban SJ. The treatment of antibody-mediated rejection in kidney transplantation: An updated systematic review and meta-analysis. Transplantation. 2018; 102: 557-568. [CrossRef] [Google scholar] [PubMed]
- Schinstock CA, Mannon RB, Budde K, Chong AS, Haas M, Knechtle S, et al. Recommended treatment for antibody-mediated rejection after kidney transplantation: The 2019 expert consensus from the Transplantion Society Working Group. Transplantation. 2020; 104: 911-922. [CrossRef] [Google scholar] [PubMed]
- Bhalla A, Alachkar N, Alasfar S. Complement-based therapy in the management of antibody-mediated rejection. Adv Chronic Kidney Dis. 2020; 27: 138-148. [CrossRef] [Google scholar] [PubMed]
- Biglarnia AR, Nilsson B, Nilsson T, Von Zur-Mühlen B, Wagner M, Berne C, et al. Prompt reversal of a severe complement activation by eculizumab in a patient undergoing intentional ABO-incompatible pancreas and kidney transplantation: Complement reversal by eculizumab during ABO reactivity. Transpl Int. 2011; 24: e61-e66. [CrossRef] [Google scholar] [PubMed]
- Stewart ZA, Collins TE, Schlueter AJ, Raife TI, Holanda DG, Nair R, et al. Case report: Eculizumab rescue of severe accelerated antibody-mediated rejection after ABO-incompatible kidney Transplant. Transplant Proc. 2012; 44: 3033-3036. [CrossRef] [Google scholar] [PubMed]
- Smith B, Kumar V, Mompoint-Williams D, Reed RD, MacLennan PA, Stegner K, et al. Dosing eculizumab for antibody-mediated rejection in kidney transplantation: A case report. Transplant Proc. 2016; 48: 3099-3105. [CrossRef] [Google scholar] [PubMed]
- Tan EK, Bentall A, Dean PG, Shaheen MF, Stegall MD, Schinstock CA. Use of eculizumab for active antibody-mediated rejection that occurs early post–kidney transplantation: A consecutive series of 15 cases. Transplantation. 2019; 103: 2397-2404. [CrossRef] [Google scholar] [PubMed]
- Perry DK, Burns JM, Pollinger HS, Amiot BP, Gloor JM, Gores GJ, et al. Proteasome inhibition causes apoptosis of normal human plasma cells preventing alloantibody production. Am J Transplant. 2009; 9: 201-209. [CrossRef] [Google scholar] [PubMed]
- Walsh RC, Everly JJ, Brailey P, Rike AH, Arend LJ, Mogilishetty G, et al. Proteasome inhibitor-based primary therapy for antibody-mediated renal allograft rejection. Transplantation. 2010; 89: 277-284. [CrossRef] [Google scholar] [PubMed]
- Walsh RC, Brailey P, Girnita A, Alloway RR, Shields AR, Wall GE, et al. Early and late acute antibody-mediated rejection differ immunologically and in response to proteasome inhibition. Transplantation. 2011; 91: 1218-1226. [CrossRef] [Google scholar] [PubMed]
- Everly MJ, Everly JJ, Susskind B, Brailey P, Arend LJ, Alloway RR, et al. Bortezomib provides effective therapy for antibody- and cell-mediated acute rejection. Transplantation. 2008; 86: 1754-1761. [CrossRef] [Google scholar] [PubMed]
- Nigos JG, Arora S, Nath P, Hussain SM, Marcus RJ, Ko TY, 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: 609-613. [CrossRef] [Google scholar] [PubMed]
- Eskandary F, Regele H, Baumann L, Bond G, Kozakowski N, Wahrmann M, et al. A randomized trial of bortezomib in late antibody-mediated kidney transplant rejection. J Am Soc Nephrol. 2018; 29: 591-605. [CrossRef] [Google scholar] [PubMed]
- Kaplan B, Gangemi A, Thielke J, Oberholzer J, Sankary H, Benedetti E. Successful rescue of refractory, severe antibody mediated rejection with splenectomy: Transplantation. 2007; 83: 9-100. [CrossRef] [Google scholar] [PubMed]
- Ezekian B, Schroder PM, Mulvihill MS, Barbas A, Collins B, Freischlag K, et al. Pretransplant desensitization with costimulation blockade and proteasome inhibitor reduces DSA and delays antibody-mediated rejection in highly sensitized nonhuman primate kidney transplant recipients. J Am Soc Nephrol. 2019; 30: 2399-2411. [CrossRef] [Google scholar] [PubMed]
- Sacha L, Teuteberg JJ, Zeevi A, Bermudez C, Kormos R, Ensor C, et al. Carfilzomib for refractory antibody mediated rejection and allosensitization in heart transplantation. J Heart Lung Transplant. 2014; 33: S31. [CrossRef] [Google scholar]
- Pham C, Pierce BJ, Nguyen DT, Graviss EA, Huang HJ. Assessment of carfilzomib treatment response in lung transplant recipients with antibody-mediated rejection. Transplant Direct. 2021; 7: e680. [CrossRef] [Google scholar] [PubMed]
- Abuazzam F, Dubrawka C, Abdulhadi T, Amurao G, Alrata L, Yaseen Alsabbagh D, et al. Emerging therapies for antibody-mediated rejection in kidney transplantation. J Clin Med. 2023; 12: 4916. [CrossRef] [Google scholar] [PubMed]
- Jordan SC, Choi J, Kim I, Wu G, Toyoda M, Shin B, et al. Interleukin-6, a cytokine critical to mediation of inflammation, autoimmunity and allograft rejection: Therapeutic implications of IL-6 receptor blockade. Transplantation. 2017; 101: 32-44. [CrossRef] [Google scholar] [PubMed]
- Vo AA, Choi J, Kim I, Louie S, Cisneros K, Kahwaji J, 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] [Google scholar] [PubMed]
- Pottebaum AA, Venkatachalam K, Liu C, Brennan DC, Murad H, Malone AF, et al. Efficacy and safety of tocilizumab in the treatment of acute active antibody-mediated rejection in kidney transplant recipients. Transplant Direct. 2020; 6: e543. [CrossRef] [Google scholar] [PubMed]
- Choi J, Aubert O, Vo A, Loupy A, Haas M, Puliyanda D, 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: 2381-2389. [CrossRef] [Google scholar] [PubMed]
- Lavacca A, Presta R, Gai C, Mella A, Gallo E, Camussi G, et al. Early effects of first‐line treatment with anti‐interleukin‐6 receptor antibody tocilizumab for chronic active antibody‐mediated rejection in kidney transplantation. Clin Transplant. 2020; 34: e13908. [CrossRef] [Google scholar] [PubMed]
- Noble J, Giovannini D, Laamech R, Imerzoukene F, Janbon B, Marchesi L, et al. Tocilizumab in the treatment of chronic antibody-mediated rejection post kidney transplantation: Clinical and histological monitoring. Front Med. 2021; 8: 790547. [CrossRef] [Google scholar] [PubMed]
- Kumar D, Yakubu I, Safavi F, Levy M, Moinuddin I, Kimball P, et al. Lack of histological and molecular signature response to tocilizumab in kidney transplants with chronic active antibody mediated rejection: A case series. Kidney360. 2020; 1: 663-670. [CrossRef] [Google scholar] [PubMed]
- Nickerson PW, Böhmig GA, Chadban S, Kumar D, Mannon RB, Van Gelder T, et al. Clazakizumab for the treatment of chronic active antibody-mediated rejection (AMR) in kidney transplant recipients: Phase 3 IMAGINE study rationale and design. Trials. 2022; 23: 1042. [CrossRef] [Google scholar] [PubMed]
- Jordan SC, Ammerman N, Choi J, Huang E, Najjar R, Peng A, et al. Evaluation of Clazakizumab (Anti–Interleukin-6) in patients with treatment-resistant chronic active antibody-mediated rejection of kidney allografts. Kidney Int Rep. 2022; 7: 720-731. [CrossRef] [Google scholar] [PubMed]
- Doberer K, Duerr M, Halloran PF, Eskandary F, Budde K, Regele H, et al. A Randomized clinical trial of anti–IL-6 antibody Clazakizumab in late antibody-mediated kidney transplant rejection. J Am Soc Nephrol. 2021; 32: 708-722. [CrossRef] [Google scholar] [PubMed]
- Sarosiek S, Shah R, Munshi NC. Review of siltuximab in the treatment of multicentric Castleman’s disease. Ther Adv Hematol. 2016; 7: 360-366. [CrossRef] [Google scholar] [PubMed]
- Thanarajasingam U, Niewold TB. Sirukumab: A novel therapy for lupus nephritis? Expert Opin Investig Drugs. 2014; 23: 1449-1455. [CrossRef] [Google scholar] [PubMed]
- Saltarella I, Desantis V, Melaccio A, Solimando AG, Lamanuzzi A, Ria R, et al. Mechanisms of Resistance to Anti-CD38 Daratumumab in Multiple Myeloma. Cells. 2020; 9: 167. [CrossRef] [Google scholar] [PubMed]
- Nooka AK, Kaufman JL, Hofmeister CC, Joseph NS, Heffner TL, Gupta VA, et al. Daratumumab in multiple myeloma. Cancer. 2019; 125: 2364-2382. [CrossRef] [Google scholar] [PubMed]
- Kwun J, Matignon M, Manook M, Guendouz S, Audard V, Kheav D, et al. Daratumumab in sensitized kidney transplantation: Potentials and limitations of experimental and clinical use. J Am Soc Nephrol. 2019; 30: 1206-1219. [CrossRef] [Google scholar] [PubMed]
- Spica D, Junker T, Dickenmann M, Schaub S, Steiger J, Rüfli T, et al. Daratumumab for treatment of antibody-mediated rejection after ABO-incompatible kidney transplantation. Case Rep Nephrol Dial. 2019; 9: 149-157. [CrossRef] [Google scholar] [PubMed]
- Frampton JE. Isatuximab: A review of its use in multiple myeloma. Target Oncol. 2021; 16: 675-686. [CrossRef] [Google scholar] [PubMed]
- Banham G, Prezzi D, Harford S, Taylor CJ, Hamer R, Higgins R, et al. Elevated pretransplantation soluble baff is associated with an increased risk of acute antibody-mediated rejection. Transplantation. 2013; 96: 413-420. [CrossRef] [Google scholar] [PubMed]
- Thibault-Espitia A, Foucher Y, Danger R, Migone T, Pallier A, Castagnet S, et al. BAFF and BAFF-R levels are associated with risk of long-term kidney graft dysfunction and development of donor-specific antibodies. Am J Transplant. 2012; 12: 2754-2762. [CrossRef] [Google scholar] [PubMed]
- Mackay F, Schneider P, Rennert P, Browning J. BAFF and APRIL: A tutorial on B cell survival. Annu Rev Immunol. 2003; 21: 231-264. [CrossRef] [Google scholar] [PubMed]
- Miller CL, Madsen JC. IL-6 Directed therapy in transplantation. Curr Transplant Rep. 2021; 8: 191-204. [CrossRef] [Google scholar] [PubMed]
- Banham GD, Flint SM, Torpey N, Lyons PA, Shanahan DN, Gibson A, et al. Belimumab in kidney transplantation: An experimental medicine, randomised, placebo-controlled phase 2 trial. Lancet. 2018; 391: 2619-2630. [CrossRef] [Google scholar] [PubMed]
- Huang E, Maldonado AQ, Kjellman C, Jordan SC. Imlifidase for the treatment of anti-HLA antibody-mediated processes in kidney transplantation. Am J Transplant. 2022; 22: 691-697. [CrossRef] [Google scholar] [PubMed]
- Kjellman C, Maldonado AQ, Sjöholm K, Lonze BE, Montgomery RA, Runström A, et al. Outcomes at 3 years posttransplant in imlifidase-desensitized kidney transplant patients. Am J Transplant. 2021; 21: 3907-3918. [CrossRef] [Google scholar] [PubMed]
- Gubernatorova EO, Samsonov MY, Drutskaya MS, Lebedeva S, Bukhanova D, Materenchuk M, et al. Targeting inerleukin-6 for renoprotection. Front Immunol. 2024; 15: 1502299. [CrossRef] [Google scholar] [PubMed]
- Hoang J, Guha A, Bhimaraj A, Kim J, Suarez E, Trachtenberg B, et al. Ofatumumab and tocilizumab desensitization in a highly sensitized recipient: A case report. J Heart Lung Transplant. 2021; 40: S493. [CrossRef] [Google scholar]
- Redfield RR, Jordan SC, Busque S, Vincenti F, Woodle ES, Desai N, et al. Safety, pharmacokinetics, and pharmacodynamic activity of obinutuzumab, a type 2 anti-CD20 monoclonal antibody for the desensitization of candidates for renal transplant. Am J Transplant. 2019; 19: 3035-3045. [CrossRef] [Google scholar] [PubMed]
- A phase 2 open-label, prospective, randomized study of Inebilizumab, VIB4920, or the combination to evaluate safety and tolerability in highly sensitized candidates awaiting kidney transplantation from a deceased donor [Internet]. 2024 [cited date 2024 December 3]. Available from: https://adisinsight.springer.com/trials/700315205.
- Colvin MM, Cook JL, Chang P, Francis G, Hsu DT, Kiernan MS, et al. Antibody-mediated rejection in cardiac transplantation: Emerging knowledge in diagnosis and management: A scientific statement from the American Heart Association. Circulation. 2015; 131: 1608-1639. [CrossRef] [Google scholar] [PubMed]
- Montgomery RA, Loupy A, Segev DL. Antibody-mediated rejection: New approaches in prevention and management. Am J Transplant. 2018; 18: 3-17. [CrossRef] [Google scholar] [PubMed]
- Russo MW, Wheless W, Vrochides D. Management of long-term complications from immunosuppression. Liver Transpl. 2024; 30: 647-658. [CrossRef] [Google scholar] [PubMed]
- Wojciechowski D, Wiseman A. Long-term immunosuppression management: Opportunities and uncertainties. Clin J Am Soc Nephrol. 2021; 16: 1264-1271. [CrossRef] [Google scholar] [PubMed]
- Karpe KM, Talaulikar GS, Walters GD. Calcineurin inhibitor withdrawal or tapering for kidney transplant recipients. Cochrane Database Syst Rev. 2017. doi: 10.1002/14651858.CD006750.pub2. [CrossRef] [Google scholar] [PubMed]
- Hariharan S, Israni AK, Danovitch G. Long-term survival after kidney transplantation. N Engl J Med. 2021; 385: 729-743. [CrossRef] [Google scholar] [PubMed]
- Thieme CJ, Schulz M, Wehler P, Anft M, Amini L, Blàzquez-Navarro A, et al. In vitro and in vivo evidence that the switch from calcineurin to mTOR inhibitors may be a strategy for immunosuppression in Epstein–Barr virus–associated post-transplant lymphoproliferative disorder. Kidney Int. 2022; 102: 1392-1408. [CrossRef] [Google scholar] [PubMed]
- Chadban SJ, Ahn C, Axelrod DA, Foster BJ, Kasiske BL, Kher V, et al. KDIGO clinical practice guideline on the evaluation and management of candidates for kidney transplantation. Transplantation. 2020; 104: S11-S103. [CrossRef] [Google scholar] [PubMed]






