pISSN 3022-6783
eISSN 3022-7712

View

Article View

Clin Transplant Res 2024; 38(4): 341-353

Published online December 31, 2024

https://doi.org/10.4285/ctr.24.0047

© The Korean Society for Transplantation

Contribution of long-lived plasma cells to antibody-mediated allograft rejection

Eunkyeong Jang1 , Jeehee Youn1,2

1Laboratory of Autoimmunology, Department of Anatomy and Cell Biology, Hanyang University College of Medicine, Seoul, Korea
2Department of Biomedical Science, Graduate School of Biomedical Science and Engineering, Hanyang University, Seoul, Korea

Correspondence to: Jeehee Youn
Laboratory of Autoimmunology, Department of Anatomy and Cell Biology, Hanyang University College of Medicine, 222 Wangsimni-ro, Seongdong-gu, Seoul 04763, Korea
E-mail: jhyoun@hanyang.ac.kr

Received: October 17, 2024; Revised: November 15, 2024; Accepted: November 15, 2024

This is an Open Access article distributed under the terms of the Creative Commons Attribution Non-Commercial License (http://creativecommons.org/licenses/by-nc/4.0/) which permits unrestricted non-commercial use, distribution, and reproduction in any medium, provided the original work is properly cited.

Persistent alloantigens derived from allograft tissues can be recognized by the host’s alloreactive immune system. This process enables cognate B cells to differentiate into plasma cells, which secrete donor-specific antibodies that are key drivers of antibody-mediated allograft rejection. A subset of these plasma cells can survive for extended periods in a suitable survival niche and mature into long-lived plasma cells (LLPCs), which are a cellular component of humoral memory. The current understanding of LLPCs is limited due to their scarcity, heterogeneity, and absence of unique markers. However, accumulating evidence indicates that LLPCs, unlike conventional short-lived plasma cells, can respond to extrinsic signals from their survival niches and can resist cell death associated with intracellular stress through cell-intrinsic mechanisms. Notably, they are refractory to traditional immunosuppressants and B cell depletion therapies. This resistance, coupled with their longevity, may explain why current treatments targeting antibody-mediated rejection are often ineffective. This review offers insights into the biology of LLPCs and discusses ongoing therapeutic trials that target LLPCs in the context of antibody-mediated allograft rejection.

Keywords: Allografts, Graft rejection, Antibody-producing cells, Plasma cells

HIGHLIGHTS
  • Long-lived plasma cells (LLPCs) contribute to long-term humoral memory.

  • Alloreactive LLPCs are a cellular source of preexisting or posttransplant alloantibodies.

  • LLPCs are refractory to immunosuppressants and B cell-depleting therapies.

  • Immunotherapies targeting LLPCs are necessary to promote successful transplantation.

The immune system has evolved to discriminate between “self” and “nonself,” thereby controlling alternative reactions: self-tolerance in the case of the former and aggressive responses in that of the latter. This principle appears to be consistently upheld in allogeneic transplantation, where highly polymorphic proteins derived from allografts, such as human leukocyte antigens (HLAs) and minor histocompatibility proteins, are perceived as “nonself” molecules by the recipient. When these donor-specific antigens are recognized and presented by dendritic cells and B cells, antigen-cognate T cell clones are activated and differentiate into effector cells, including follicular helper T (Tfh) cells, proinflammatory Th17 cells, and cytotoxic T lymphocytes. Some activated B cells give rise to plasma cells (PCs) that secrete potentially pathogenic alloantibodies, in a process that may be dependent on or independent of Tfh cells. In kidney transplantation, approximately 7% of adult recipients reportedly develop donor-specific antibodies (DSAs; primarily donor HLA-specific antibodies) within 5 years of transplantation, and this proportion rises to 20% after 10 years [14]. Posttransplant DSAs have also been identified in significant numbers of recipients of other solid organ transplants [58].

In addition to being produced de novo during the posttransplantation period, alloantibodies preexist in many individuals on transplant waiting lists [911]. This preexistence can result from prior exposure to allogeneic tissues via pregnancy, blood transfusion, or earlier transplantation [12]. In addition, natural antibodies, which are established independently of such exposures, contribute to presensitization to alloantigens. This is likely because they are polyreactive and can cross-react with diverse epitopes from microbes, alloantigens, and autoantigens [13,14]. Indeed, transplant recipients with preformed DSAs are known to face a higher risk of antibody-mediated rejection (AMR) and subsequent allograft loss across all solid organ transplants. This evidence supports the potential critical role of preformed DSAs in the promotion of AMR [6,8,1519].

Both preexisting and posttransplant DSAs can trigger pathological events associated with AMR. These events are characterized by swelling of endothelial cells, microvascular inflammation, and the formation of intravascular immune cell infiltrates, which may occur with or without complement deposition. Accumulating evidence indicates that AMR is a major contributor to decreased long-term graft survival. For instance, 40% of patients lose their renal allografts within 5 years following the emergence of de novo DSAs—a rate double that observed in patients without DSAs [4]. Moreover, over half of late allograft failures can be attributed to HLA DSA-mediated chronic rejection [2,2024]. Consequently, to improve transplant outcomes, it is crucial to understand the immunopathological mechanisms underpinning DSA production and to develop strategies targeting DSAs.

Despite decades of attempts to understand the biology of antibody-secreting cells and to apply this knowledge in developing treatments, AMR remains a formidable obstacle to successful transplantation. Current desensitization strategies, such as B cell depletion protocols, are often ineffective. One potential explanation is the inadequate depletion of alloreactive memory B cells and long-lived PCs (LLPCs), which together form the basis of humoral memory. Although the characteristics of LLPCs have not been fully elucidated, they possess unique features that set them apart from other B cell subsets, including conventional PCs. Furthermore, they have been implicated in AMR, especially chronic AMR.

Here, we present a contemporary perspective on the biology of antibody-secreting B cells, focusing on LLPCs. We explore their developmental pathways, survival mechanisms, and functional characteristics. Given the increasingly recognized importance of LLPCs in AMR, we also discuss potential strategies for targeting these cells.

AMR is initiated by the binding of alloantibodies to alloantigens, predominantly HLA molecules, on the surface of vascular endothelial cells. This interaction induces inflammatory responses mediated by the complement system and/or Fc receptors [2527]. Local complement activation results in cell lysis, the recruitment and activation of neutrophils, and thrombus formation. Concurrently, the engagement of Fc receptors by alloantibodies authorizes neutrophils and natural killer cells to attack and destroy endothelial cells. These combined mechanisms contribute to endothelial injury and the development of intravascular thromboses, leading to graft dysfunction.

The processes that underlie chronic AMR are more complex than those associated with acute AMR. They develop insidiously over months to years and may occur with or without clinically evident episodes of acute rejection. In the majority of vascularized grafts, chronic inflammatory responses induce the proliferation of intimal smooth muscle cells within blood vessels and fibroblasts in the interstitial tissue, leading to vascular occlusion and interstitial fibrosis [28,29]. Ultimately, the grafts fail, predominantly due to the resulting ischemic damage [30].

The adaptive immune response to an allograft begins in the peripheral lymphoid organs that drain the graft (Fig. 1). Donor dendritic cells within the allograft migrate to the lymphoid tissue, where they can directly present allogeneic HLA molecules to host T cells. Alternatively, in the indirect pathway, recipient dendritic cells that have infiltrated the allograft capture and present HLA peptides to alloreactive host T cells. Upon priming by both direct and indirect allorecognition pathways, the alloantigen-cognate T cells become activated and differentiate into effector cells. A subset of activated CD4+ T cells, known as extrafollicular helper T cells, migrate toward the B cell follicles [31]. Concurrently, antigen-engaged B cells move to the interface of T and B cell zones to interact with the extrafollicular helper T cells. This interaction triggers the initial T cell-dependent B cell activation, leading to the formation of extrafollicular foci. Within these foci, B cells proliferate and differentiate into PCs, which are predominantly short-lived [32,33].

Figure 1. Humoral alloimmune responses in the context of solid organ transplantation. Adaptive immune responses to alloantigens are initiated by the activation of alloreactive dendritic cells. Within secondary lymphoid organs, B cells that have engaged with alloantigens interact with cognate CD4+ T cells at the interface of the T cell zone and the follicle. This interaction prompts the B cells to proliferate and differentiate into either extrafollicular PCs or GC B cells. The latter undergo a mutation-prone proliferation process and eventually give rise to memory B cells and affinity-matured PCs. Whether generated by the extrafollicular or GC responses, most PCs are short-lived. However, a subset migrates to the bone marrow, which provides survival niches. In the bone marrow, they further differentiate into LLPCs. LLPCs can also be found in intragraft tissues, where they may develop in situ within tertiary lymphoid clusters that form in the graft, or they may migrate from secondary lymphoid organs. DC, dendritic cell; SLPC, short-lived plasma cell; eTFH, extrafollicular helper T cell; TFH, follicular helper T cell; GC, germinal center; FDC, follicular dendritic cell; SHM, somatic hypermutation; PC, plasma cell; TLC, tertiary lymphoid cluster; LLPC, long-lived PC.

Of the T and B cells activated in the extrafollicular foci, some return to the follicles to initiate germinal center (GC) reactions [3436]. Within the GCs, B cells undergo a repetitive cycle, moving between the light and dark zones. They experience mutation-prone proliferation in the dark zone before shuttling to the light zone, where high-affinity GC B cells are positively selected under the control of Tfh cells. Ultimately, affinity-matured B cells exit the GC reaction by differentiating into either memory B cells or PCs.

The stepwise differentiation of naive B cells into PCs is governed by a genetic program. The transcription factors PAX5, BCL6, and BLIMP1 are relatively well-established master regulators that delineate the identities of naive B cells, GC B cells, and PCs, respectively [3739]. These molecules, along with other transcription factors including IRF4, BACH2, and XBP1, form a complex genetic regulatory network that orchestrates the elaborate differentiation program [4042].

Most PCs generated via the GC reaction are proliferating cells that undergo apoptotic cell death in situ within a week, as these cells appear to possess few survival niches. To extend their lifespan, PCs migrate out of the organs in which they originated, seeking survival niches that are primarily found in the bone marrow (BM) [43,44]. The homing of PCs to the BM is mediated by the CXCL12-CXCR4 axis; BM stromal cells release CXCL12, and PCs expressing the CXCR4 receptor follow the chemokine gradient to their destination. Upon arrival in the BM, they differentiate further into mature antibody-secreting cells and can survive for years or even decades. Consequently, LLPCs, which secrete high-affinity isotype-switched antibodies, are predominantly generated during the GC reaction and are mainly located in the BM. However, recent studies have revealed that a substantial proportion of LLPCs are found in various anatomical locations that provide their own survival niches. These tissue-resident PCs display a transcriptional signature specific to their tissue of origin [45]. Intriguingly, analysis of the transcriptional signatures indicates that the BM contains a diverse collection of PCs from all tissues, suggesting that tissue-resident PCs may not be sessile but instead migrate to the BM.

Antibody-secreting cells can be categorized into at least three subsets: plasmablasts, short-lived PCs (SLPCs), and LLPCs. These subsets appear to represent stages in a maturation continuum, as evidenced by their shared PC molecular markers, which include CD138 (syndecan-1), BLIMP1, XBP1, and IRF4. The initial PC precursors, which arise from either extrafollicular or GC responses, are rapidly proliferating plasmablasts. As they differentiate into mature PCs, they lose B cell markers such as CD19, CD20, B220, major histocompatibility complex class II, and SLAMF6 [46]. These mature PCs continue to proliferate and are turned over within a few days. While most of these cells end their lifecycle as SLPCs, a subset acquires the necessary competence to become LLPCs. These LLPCs can secrete antibodies for decades without requiring antigenic stimulation.

LLPCs are a rare and diverse group of antibody-secreting cells that lack unique markers [47]. While most LLPCs are found in the BM, they do not represent all BM PCs. In naive mice and healthy humans, LLPCs make up less than 25% of the BM PC population, which itself is a minor fraction, comprising only 0.1%–5% of the total immune cell population [46,48,49]. Following deliberate immunization, the proportion of LLPCs in the BM can increase markedly to about 65%, but it never approaches 100% [50]. A previous study has shown that the BM-resident CD38hi antibody-secreting cell population can be divided into four subsets based on the presence or absence of CD19 and CD138 on the cell surface [48]. While LLPCs have been found to be enriched among CD38hiCD19CD138hi cells, this subset includes both LLPCs and SLPCs. In mice, LLPCs were identified within the B220BLIMP1+CD138hi cell fraction, which similarly included SLPCs. Transcriptome profiling and biochemical analyses have revealed several genetic markers that are differentially expressed in LLPCs compared to SLPCs, including BLIMP1, XBP1, CD138, B cell maturation antigen (BCMA), CD28, and MCL-1 (Fig. 2) [44,5157]. However, these markers are not exclusive to LLPCs and are also present in SLPCs at varying levels. Consequently, the physiology of LLPCs remains incompletely understood.

Figure 2. Characteristics of LLPCs. LLPCs exhibit numerous characteristics that distinguish them from SLPCs. Molecules associated with plasma cell survival and function are upregulated in LLPCs, whereas B cell markers are downregulated. These molecular changes not only contribute to the resistance of LLPCs to traditional cytostatic treatments and B cell depletion therapies but also inform the mechanisms of action for ongoing therapeutic trials. SLPC, short-lived plasma cell; LLPC, long-lived plasma cell; ATG, autophagy gene; BCMA, B cell maturation antigen.

What determines the difference in cell fate between SLPCs and LLPCs, as well as the longevity of PCs? It appears to hinge on a variation in responsiveness to prosurvival signals from the microenvironment, rather than on distinct genetic programs. For extended survival, PCs exit the differentiation site and migrate toward their survival niches, predominantly located in the BM. These niches are structured by BM stromal cells that secrete CXCL12 and attract CXCR4+ PCs [58]. Additional niche components include megakaryocytes, eosinophils, and osteoclasts, which produce prosurvival factors such as BAFF, APRIL, tumor necrosis factor, and interleukin-6 (IL-6) [5961]. These factors are also concentrated in inflamed tissues, drawing CXCR3-expressing PCs that persist until the inflammation subsides [6264]. Our research, along with that of others, has demonstrated that in certain severe autoimmune diseases in both mice and humans, a substantial proportion (over 30%) of splenic PCs are LLPCs [6569]. These ectopic LLPC populations are sustained by myeloid-derived suppressor cells (MDSCs) originating from the spleen, which secrete BAFF [70]. Notably, these cells have recently been found to promote Tfh cell development [71]. This cellular network, termed the Tfh-LLPC-MDSC axis, may be pivotal in the in situ perpetuation of autoimmune responses.

Only PCs that respond effectively to these survival factors can persist as LLPCs. The PC surface molecules BCMA and CD138 are well-established factors responsible for this survival. Among the three receptors for BAFF and APRIL, only BCMA, not BAFF-R or TACI, is elevated in BM PCs [56]. The survival of BM PCs is notably compromised by the deletion of BCMA, indicating its critical role in LLPC survival. Supporting this, signals associated with BCMA engagement have been shown to induce the expression of MCL-1, a crucial prosurvival factor for PCs [57]. CD138 is a canonical surface marker of PCs that is upregulated upon maturation [54]. It increases heparan sulfate levels on the PC surface, which in turn promotes IL-6 and APRIL signaling. Consequently, mature CD138hi PCs gain a selective survival advantage over their less mature CD138lo PCs counterparts due to this enhanced prosurvival cytokine signaling. Certain adhesion molecules expressed on BM PCs, including VLA4, LFA-1, CD44, and PSGL-1, also contribute to PC survival by interacting with molecules in their niche [72,73].

The acquisition of the molecular competence mentioned above appears to largely rely on the increased activity of transcription factors that are essential for PC development. The transcription repressor BLIMP1 serves as a master regulator of PC differentiation, as its ectopic expression is sufficient to drive this process [74,75]. Utilizing a BLIMP1-GFP reporter system, Kallies et al. [52] demonstrated that LLPCs express higher levels of BLIMP1 compared to SLPCs. The induced conditional deletion of Prdm1, the gene encoding BLIMP1, results in the loss of previously formed PCs, likely due to endoplasmic reticulum (ER) stress-induced apoptosis. This indicates that BLIMP1 is not only necessary for PC formation but also for the maintenance of LLPC populations [53].

The influence of BLIMP1 on PC maintenance appears to be partially mediated by XBP1, which is itself an essential factor for PC development [39,76,77]. BLIMP1 upregulates XBP1 expression by directly suppressing the transcription of Pax5, which in turn derepresses genes, including Xbp1, that are normally repressed by PAX5. The spliced, active form of XBP1 can initiate the unfolded protein response (UPR) in PCs and thereby prevent ER stress-associated cell death. This process is crucial for PC survival due to the high levels of ER stress these cells experience from their extensive antibody production and secretion. ER stress activates the UPR to maintain cell viability and reestablish protein homeostasis. The XBP1 pathway of the UPR primarily serves to facilitate ER expansion, enabling PCs to secrete large quantities of immunoglobulins (Ig). XBP1 induces the transcription of numerous target genes, including those associated with molecular chaperones; this improves protein folding capacity and ER-associated degradation, which ultimately delivers ubiquitinated substrates to proteasomes. Based on our research, one target of XBP1 is the molecular chaperone FKBP13, which plays a pivotal role in the quality control of Ig within the ER [78]. FKBP13 levels are higher in LLPCs than in SLPCs, and inhibiting FKBP13 with rapamycin treatment reduces the PC lifespan. These findings indicate that proteostatic mechanisms involving the XBP1-FKBP13 axis are key to the prolonged survival of LLPCs. Additionally, this work highlights a previously unappreciated effect of rapamycin in the depletion of LLPCs.

A second mechanism implicated in the adaptation of cells to protein stress is autophagy. Autophagy is a natural, conserved process that eliminates unnecessary or dysfunctional components via a lysosome-dependent regulated pathway [79]. Pengo et al. [80] have reported that autophagy is critical for LLPC survival. They showed that the number of LLPCs was significantly reduced in mice immunized with a T cell-dependent antigen after deletion of the autophagy gene Atg5.

PCs have a higher metabolic demand than naive B cells because they require ample energy and nutrient resources to secrete large amounts of glycosylated Ig [81]. As a cell’s metabolic program is closely associated with its differentiation status, it is reasonable to anticipate that LLPCs would exhibit metabolic profiles distinct from those of SLPCs. The results of one study support this notion [82], demonstrating that LLPCs import more glucose than SLPCs, and when faced with metabolic stress, LLPCs—but not SLPCs—redirect glucose metabolism towards the production of pyruvate. Consistent with these observations, the genetic ablation of Mpc2, a gene encoding a component of the mitochondrial pyruvate carrier, resulted in a reduced lifespan for LLPCs. Thus, mitochondrial pyruvate import may represent a critical pathway for the survival of LLPCs.

However, there are some important exceptions to the traditional paradigm describing the identity of LLPCs. First, while this paradigm limits their origin to GC B cells, recent studies have shown that early-wave extrafollicular plasmablasts and B-1 cell-derived PCs can also persist for extended periods in the BM [47,8385]. Second, not all BM-residing PCs exhibit long-term survival [51]. Third, the longstanding belief that most PCs in the spleen are short-lived is now being questioned, following the identification of substantial numbers of splenic LLPCs in both mouse models and patients with autoimmune diseases [6569].

Despite these caveats, resistance to traditional therapies remains a defining characteristic of LLPCs. Numerous studies have demonstrated that LLPCs are not eliminated by treatments that target B lineage cells, including irradiation, prednisone, cyclophosphamide, and anti-CD20 antibodies [65,67,8688]. This resistance, coupled with their sustained antibody production, contributes to the challenge of treating LLPC-mediated pathologies.

LLPCs contribute to AMR by producing a diverse array of alloantibodies. They appear to be the primary source of panel-reactive antibodies, including preformed DSAs, which can result in a positive serum cross-match prior to kidney transplantation. Furthermore, alloreactive LLPCs may emerge after transplantation and continue to produce de novo DSAs over extended periods.

As in autoimmune spleens, alloreactive LLPCs can accumulate in grafted organs. The microenvironment of the transplant organ may induce PCs to migrate to the graft, where they subsequently differentiate into LLPCs. Alternatively, the inflammatory milieu within the graft could enable circulating or tissue-resident immune cells to initiate lymphoid organogenesis within the graft niche. Subsequently, some memory B cells and GC B cells may transform into PCs and ultimately LLPCs. This possibility is supported by a study showing that chronically rejected human kidneys contain tertiary lymphoid foci where GC B cells, memory B cells, and PCs are concentrated [8992]. Regardless of the initial site of PC formation, the presence of LLPCs in a graft can contribute to organ rejection through the continuous local production of alloantibodies.

Current therapies commonly used to treat AMR include plasmapheresis and intravenous Ig (IVIg). Plasmapheresis works by physically removing antibodies from the circulation. In turn, IVIg has multiple mechanisms of action: it neutralizes the effects of pathogenic antibodies, inhibits complement activation, blocks neonatal Fc receptor to reduce antibody recycling, and interacts with FcγRIIb to send inhibitory signals to immune cells [93]. Although these protocols are considered first-line treatments for AMR, their efficacy is limited [9397]. Consequently, it is necessary to establish more aggressive protocols that target antibody-secreting cells, rather than simply removing or inhibiting the antibodies produced.

Anti-CD20 therapy has been developed to deplete B cells. Rituximab, a type 1 anti-CD20 monoclonal antibody, is utilized for desensitization in HLA-incompatible and ABO-incompatible transplantation, typically in conjunction with plasmapheresis and IVIg administration [98]. However, in multiple clinical trials, its efficacy has not been demonstrated [99,100]. Other mechanistic studies have revealed that while rituximab depletes circulating B cells, primarily through complement-dependent cytotoxicity, it does not effectively reduce tissue-resident B cells and mature PCs [101,102]. The mechanism of memory B cell resistance is unclear, but a gradual loss of CD20 expression on the surface of PCs during their maturation may underlie the resistance observed in mature PCs, including LLPCs. Notably, rituximab treatment has been found to exacerbate primary immune thrombocytopenia by increasing the number of LLPCs [68]. This suggests that the milieu generated by B cell depletion may actually encourage the development and local tissue settlement of LLPCs, offering insights from an autoimmune disease into the challenges of transplantation. Obinutuzumab, a glycoengineered type 2 anti-CD20 monoclonal antibody, has shown greater efficacy in depleting various B cell subsets, including memory B cells, plasmablasts, and PCs, via antibody-dependent cellular cytotoxicity [103]. Despite its beneficial effects in this context and its efficacy in the treatment of systemic lupus erythematosus, the combination of obinutuzumab with IVIg has not led to a reduction in HLA antibody levels [104]. The presence of CD20 LLPCs likely contributed to the lack of effectiveness of this treatment approach.

Inhibiting proteasomes could be a promising strategy for eliminating LLPCs, as proteasome activity is essential for the survival of all PCs, suggesting that proteasome inhibitors would likely not discriminate between LLPCs and SLPCs. However, the proteasome inhibitor bortezomib did not significantly reduce DSA levels or improve the histological or molecular signs of AMR in certain clinical settings [105,106]. Furthermore, bortezomib therapy has been associated with significant toxicity [107]. Another proteasome inhibitor, carfilzomib, has been shown to be more effective than bortezomib in lung transplant recipients; those who responded to carfilzomib were less prone to experiencing chronic lung allograft dysfunction or progression compared to nonresponders [108]. Nevertheless, concerns have arisen regarding rebound DSA and AMR-driven allograft failure following the discontinuation of treatment. Consequently, despite their theoretical potential, the current evidence seems insufficient to endorse the widespread use of proteasome inhibitors for the treatment of AMR.

Since BCMA is essential for the survival of LLPCs, it is considered an attractive therapeutic target. Strategies aimed at BCMA were initially developed for B cell malignancies and have since been expanded to include desensitization to DSAs in allograft transplants. A monoclonal antibody specific for both BCMA and CD3 has been developed to target BCMA on myeloma cells and PCs, as well as CD3 on T cells [109,110]. This bispecific antibody functions by recruiting and activating cytotoxic T cells to kill BCMA+ cells. Although anti-BCMA-CD3 bispecific antibodies have shown promising efficacy in the treatment of multiple myeloma, these antibodies—like other T cell-engaging therapies—may cause adverse effects, including cytokine release syndrome and T cell depletion [111]. To mitigate such adverse effects, a novel monoclonal antibody with low affinity for CD3 (REGN 5458) has been developed and is currently under investigation as a means to decrease HLA antibody levels and improve transplant rates in patients who are highly HLA-sensitized and awaiting kidney transplantation [110].

Chimeric antigen receptor (CAR) T cell therapy has been employed to deplete BCMA+ PCs. CAR T cells are autologous T cells that have been genetically modified to express a synthetic receptor specific to a target antigen. Upon encountering their designated antigen, CAR T cells are activated and initiate cytotoxic attacks on cells expressing that antigen. Currently, CAR T cells that react to the pan-B cell antigen CD19 and to BCMA are being employed as secondary treatments for various B cell malignancies, including multiple myeloma [112]. The potential for CAR T cells to deplete LLPCs has also been explored in the field of transplantation [113]. Zhang et al. [114] demonstrated that CAR T cells targeting CD19 and BCMA efficiently eliminated DSAs, memory B cells, and LLPCs and prevented AMR in allosensitized mice. They also observed a clinically meaningful reduction in alloantibodies in a human trial (ClinicalTrials.gov: NCT03549442). These findings provide evidence that LLPCs are a key cellular source of alloantibodies and suggest that immunotherapy targeting LLPCs holds promise for desensitization and/or treatment of allograft rejection.

Because preexisting and posttransplant alloantibodies are major contributors to AMR, it is essential to understand the biology of antibody-secreting cells to inform the development of immunotherapies with curative potential. LLPCs are a mature subset of antibody-secreting cells that can survive for decades without dividing. Emerging through diverse pathways of B cell differentiation, they are found not only in the BM but also in transplanted tissues that provide the necessary survival factors. The capacity of LLPCs to respond to external survival signals and to manage intracellular protein stress allows them to have extended lifespans accompanied by the constitutive production of antibodies. Chronic and refractory AMR may be explained by the resistance of LLPCs to traditional immunosuppressive and B cell depletion therapies. Therefore, developing a method to eliminate alloreactive LLPCs is a critical element in any strategy aimed at preventing AMR.

Conflict of Interest

No potential conflict of interest relevant to this article was reported.

Author Contributions

All the work was done by Eunkyeong Jang and Jeehee Youn. All authors read and approved the final manuscript.

  1. Schinstock CA, Cosio F, Cheungpasitporn W, Dadhania DM, Everly MJ, Samaniego-Picota MD, et al. The value of protocol biopsies to identify patients with de novo donor-specific antibody at high risk for allograft loss. Am J Transplant 2017;17:1574-84.
    Pubmed KoreaMed CrossRef
  2. Everly MJ, Rebellato LM, Haisch CE, Ozawa M, Parker K, Briley KP, et al. Incidence and impact of de novo donor-specific alloantibody in primary renal allografts. Transplantation 2013;95:410-7.
    Pubmed CrossRef
  3. Viglietti D, Loupy A, Vernerey D, Bentlejewski C, Gosset C, Aubert O, et al. Value of donor-specific anti-HLA antibody monitoring and characterization for risk stratification of kidney allograft loss. J Am Soc Nephrol 2017;28:702-15.
    Pubmed KoreaMed CrossRef
  4. Wiebe C, Gibson IW, Blydt-Hansen TD, Pochinco D, Birk PE, Ho J, et al. Rates and determinants of progression to graft failure in kidney allograft recipients with de novo donor-specific antibody. Am J Transplant 2015;15:2921-30.
    Pubmed CrossRef
  5. Smith JD, Banner NR, Hamour IM, Ozawa M, Goh A, Robinson D, et al. De novo donor HLA-specific antibodies after heart transplantation are an independent predictor of poor patient survival. Am J Transplant 2011;11:312-9.
    Pubmed CrossRef
  6. Morrell MR, Pilewski JM, Gries CJ, Pipeling MR, Crespo MM, Ensor CR, et al. De novo donor-specific HLA antibodies are associated with early and high-grade bronchiolitis obliterans syndrome and death after lung transplantation. J Heart Lung Transplant 2014;33:1288-94.
    Pubmed CrossRef
  7. Kaneku H, O'Leary JG, Banuelos N, Jennings LW, Susskind BM, Klintmalm GB, et al. De novo donor-specific HLA antibodies decrease patient and graft survival in liver transplant recipients. Am J Transplant 2013;13:1541-8.
    Pubmed KoreaMed CrossRef
  8. Cheng EY, Everly MJ, Kaneku H, Banuelos N, Wozniak LJ, Venick RS, et al. Prevalence and clinical impact of donor-specific alloantibody among intestinal transplant recipients. Transplantation 2017;101:873-82.
    Pubmed KoreaMed CrossRef
  9. Lentine KL, Smith JM, Lyden GR, Miller JM, Dolan TG, Bradbrook K, et al. OPTN/SRTR 2022 annual data report: kidney. Am J Transplant 2024;24(2 Suppl 1):S19-118.
    Pubmed CrossRef
  10. Kwong AJ, Kim WR, Lake JR, Schladt DP, Schnellinger EM, Gauntt K, et al. OPTN/SRTR 2022 annual data report: liver. Am J Transplant 2024;24(2 Suppl 1):S176-265.
    Pubmed CrossRef
  11. Valapour M, Lehr CJ, Schladt DP, Smith JM, Swanner K, Weibel CJ, et al. OPTN/SRTR 2022 annual data report: lung. Am J Transplant 2024;24(2 Suppl 1):S394-456.
    Pubmed CrossRef
  12. Butler CL, Valenzuela NM, Thomas KA, Reed EF. Not all antibodies are created equal: factors that influence antibody mediated rejection. J Immunol Res 2017;2017:7903471.
    Pubmed KoreaMed CrossRef
  13. Zorn E, See SB. Polyreactive natural antibodies in transplantation. Curr Opin Organ Transplant 2017;22:8-13.
    Pubmed KoreaMed CrossRef
  14. Notkins AL. Polyreactivity of antibody molecules. Trends Immunol 2004;25:174-9.
    Pubmed CrossRef
  15. Alba AC, Tinckam K, Foroutan F, Nelson LM, Gustafsson F, Sander K, et al. Factors associated with anti-human leukocyte antigen antibodies in patients supported with continuous-flow devices and effect on probability of transplant and post-transplant outcomes. J Heart Lung Transplant 2015;34:685-92.
    Pubmed CrossRef
  16. Bielmann D, Hönger G, Lutz D, Mihatsch MJ, Steiger J, Schaub S. Pretransplant risk assessment in renal allograft recipients using virtual crossmatching. Am J Transplant 2007;7:626-32.
    Pubmed CrossRef
  17. Irving CA, Carter V, Gennery AR, Parry G, Griselli M, Hasan A, et al. Effect of persistent versus transient donor-specific HLA antibodies on graft outcomes in pediatric cardiac transplantation. J Heart Lung Transplant 2015;34:1310-7.
    Pubmed CrossRef
  18. Roux A, Bendib Le Lan I, Holifanjaniaina S, Thomas KA, Hamid AM, Picard C, et al. Antibody-mediated rejection in lung transplantation: clinical outcomes and donor-specific antibody characteristics. Am J Transplant 2016;16:1216-28.
    Pubmed CrossRef
  19. Tran A, Fixler D, Huang R, Meza T, Lacelle C, Das BB. Donor-specific HLA alloantibodies: impact on cardiac allograft vasculopathy, rejection, and survival after pediatric heart transplantation. J Heart Lung Transplant 2016;35:87-91.
    Pubmed CrossRef
  20. de Kort H, Munivenkatappa RB, Berger SP, Eikmans M, van der Wal A, de Koning EJ, et al. Pancreas allograft biopsies with positive c4d staining and anti-donor antibodies related to worse outcome for patients. Am J Transplant 2010;10:1660-7.
    Pubmed CrossRef
  21. Einecke G, Sis B, Reeve J, Mengel M, Campbell PM, Hidalgo LG, et al. Antibody-mediated microcirculation injury is the major cause of late kidney transplant failure. Am J Transplant 2009;9:2520-31.
    Pubmed CrossRef
  22. Loupy A, Toquet C, Rouvier P, Beuscart T, Bories MC, Varnous S, et al. Late failing heart allografts: pathology of cardiac allograft vasculopathy and association with antibody-mediated rejection. Am J Transplant 2016;16:111-20.
    Pubmed CrossRef
  23. Wozniak LJ, Hickey MJ, Venick RS, Vargas JH, Farmer DG, Busuttil RW, et al. Donor-specific HLA antibodies are associated with late allograft dysfunction after pediatric liver transplantation. Transplantation 2015;99:1416-22.
    Pubmed KoreaMed CrossRef
  24. Iacob S, Cicinnati VR, Lindemann M, Heinemann FM, Radtke A, Kaiser GM, et al. Donor-specific anti-HLA antibodies and endothelial C4d deposition-association with chronic liver allograft failure. Transplantation 2015;99:1869-75.
    Pubmed CrossRef
  25. Smith RN, Brousaides N, Grazette L, Saidman S, Semigran M, Disalvo T, et al. C4d deposition in cardiac allografts correlates with alloantibody. J Heart Lung Transplant 2005;24:1202-10.
    Pubmed CrossRef
  26. Coxon A, Cullere X, Knight S, Sethi S, Wakelin MW, Stavrakis G, et al. Fc gamma RIII mediates neutrophil recruitment to immune complexes: a mechanism for neutrophil accumulation in immune-mediated inflammation. Immunity 2001;14:693-704.
    Pubmed CrossRef
  27. 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;10:1812-22.
    Pubmed CrossRef
  28. Mehra MR, Ventura HO, Stapleton DD, Smart FW, Collins TC, Ramee SR. Presence of severe intimal thickening by intravascular ultrasonography predicts cardiac events in cardiac allograft vasculopathy. J Heart Lung Transplant 1995;14:632-9.
  29. Miyagawa-Hayashino A, Yoshizawa A, Uchida Y, Egawa H, Yurugi K, Masuda S, et al. Progressive graft fibrosis and donor-specific human leukocyte antigen antibodies in pediatric late liver allografts. Liver Transpl 2012;18:1333-42.
    Pubmed CrossRef
  30. Ioannou A, Dalle Lucca J, Tsokos GC. Immunopathogenesis of ischemia/reperfusion-associated tissue damage. Clin Immunol 2011;141:3-14.
    Pubmed CrossRef
  31. Jin H, Yang K, Zhang H, Chen Y, Qi H, Fan Z, et al. Expansion of circulating extrafollicular helper T-like cells in patients with chronic graft-versus-host disease. J Autoimmun 2019;100:95-104.
    Pubmed CrossRef
  32. Deng R, Hurtz C, Song Q, Yue C, Xiao G, Yu H, et al. Extrafollicular CD4+ T-B interactions are sufficient for inducing autoimmune-like chronic graft-versus-host disease. Nat Commun 2017;8:978.
    Pubmed KoreaMed CrossRef
  33. MacLennan IC, Toellner KM, Cunningham AF, Serre K, Sze DM, Zúñiga E, et al. Extrafollicular antibody responses. Immunol Rev 2003;194:8-18.
    Pubmed CrossRef
  34. Allen CD, Okada T, Cyster JG. Germinal-center organization and cellular dynamics. Immunity 2007;27:190-202.
    Pubmed KoreaMed CrossRef
  35. Batista FD, Harwood NE. The who, how and where of antigen presentation to B cells. Nat Rev Immunol 2009;9:15-27.
    Pubmed CrossRef
  36. Srinivasan M, Flynn R, Price A, Ranger A, Browning JL, Taylor PA, et al. Donor B-cell alloantibody deposition and germinal center formation are required for the development of murine chronic GVHD and bronchiolitis obliterans. Blood 2012;119:1570-80.
    Pubmed KoreaMed CrossRef
  37. Basso K, Dalla-Favera R. BCL6: master regulator of the germinal center reaction and key oncogene in B cell lymphomagenesis. Adv Immunol 2010;105:193-210.
    Pubmed CrossRef
  38. Pridans C, Holmes ML, Polli M, Wettenhall JM, Dakic A, Corcoran LM, et al. Identification of Pax5 target genes in early B cell differentiation. J Immunol 2008;180:1719-28.
    Pubmed CrossRef
  39. Shaffer AL, Shapiro-Shelef M, Iwakoshi NN, Lee AH, Qian SB, Zhao H, et al. XBP1, downstream of Blimp-1, expands the secretory apparatus and other organelles, and increases protein synthesis in plasma cell differentiation. Immunity 2004;21:81-93.
    Pubmed CrossRef
  40. Iwakoshi NN, Lee AH, Vallabhajosyula P, Otipoby KL, Rajewsky K, Glimcher LH. Plasma cell differentiation and the unfolded protein response intersect at the transcription factor XBP-1. Nat Immunol 2003;4:321-9.
    Pubmed CrossRef
  41. Ochiai K, Katoh Y, Ikura T, Hoshikawa Y, Noda T, Karasuyama H, et al. Plasmacytic transcription factor Blimp-1 is repressed by Bach2 in B cells. J Biol Chem 2006;281:38226-34.
    Pubmed CrossRef
  42. Ochiai K, Maienschein-Cline M, Simonetti G, Chen J, Rosenthal R, Brink R, et al. Transcriptional regulation of germinal center B and plasma cell fates by dynamical control of IRF4. Immunity 2013;38:918-29.
    Pubmed KoreaMed CrossRef
  43. Brynjolfsson SF, Persson Berg L, Olsen Ekerhult T, Rimkute I, Wick MJ, Mårtensson IL, et al. Long-lived plasma cells in mice and men. Front Immunol 2018;9:2673.
    Pubmed KoreaMed CrossRef
  44. Lightman SM, Utley A, Lee KP. Survival of long-lived plasma cells (LLPC): piecing together the puzzle. Front Immunol 2019;10:965.
    Pubmed KoreaMed CrossRef
  45. Tellier J, Tarasova I, Nie J, Smillie CS, Fedele PL, Cao WH, et al. Unraveling the diversity and functions of tissue-resident plasma cells. Nat Immunol 2024;25:330-42.
    Pubmed CrossRef
  46. Robinson MJ, Ding Z, Dowling MR, Hill DL, Webster RH, McKenzie C, et al. Intrinsically determined turnover underlies broad heterogeneity in plasma-cell lifespan. Immunity 2023;56:1596-12.
    Pubmed CrossRef
  47. Liu X, Yao J, Zhao Y, Wang J, Qi H. Heterogeneous plasma cells and long-lived subsets in response to immunization, autoantigen and microbiota. Nat Immunol 2022;23:1564-76.
    Pubmed CrossRef
  48. Halliley JL, Tipton CM, Liesveld J, Rosenberg AF, Darce J, Gregoretti IV, et al. Long-lived plasma cells are contained within the CD19(-)CD38(hi)CD138(+) subset in human bone marrow. Immunity 2015;43:132-45.
    Pubmed KoreaMed CrossRef
  49. Koike T, Fujii K, Kometani K, Butler NS, Funakoshi K, Yari S, et al. Progressive differentiation toward the long-lived plasma cell compartment in the bone marrow. J Exp Med 2023;220:e20221717.
    Pubmed KoreaMed CrossRef
  50. Manz RA, Thiel A, Radbruch A. Lifetime of plasma cells in the bone marrow. Nature 1997;388:133-4.
    Pubmed CrossRef
  51. Fooksman DR, Jing Z, Park R. New insights into the ontogeny, diversity, maturation and survival of long-lived plasma cells. Nat Rev Immunol 2024;24:461-70.
    Pubmed CrossRef
  52. Kallies A, Hasbold J, Tarlinton DM, Dietrich W, Corcoran LM, Hodgkin PD, et al. Plasma cell ontogeny defined by quantitative changes in blimp-1 expression. J Exp Med 2004;200:967-77.
    Pubmed KoreaMed CrossRef
  53. Shapiro-Shelef M, Lin KI, Savitsky D, Liao J, Calame K. Blimp-1 is required for maintenance of long-lived plasma cells in the bone marrow. J Exp Med 2005;202:1471-6.
    Pubmed KoreaMed CrossRef
  54. McCarron MJ, Park PW, Fooksman DR. CD138 mediates selection of mature plasma cells by regulating their survival. Blood 2017;129:2749-59.
    Pubmed KoreaMed CrossRef
  55. Rozanski CH, Arens R, Carlson LM, Nair J, Boise LH, Chanan-Khan AA, et al. Sustained antibody responses depend on CD28 function in bone marrow-resident plasma cells. J Exp Med 2011;208:1435-46.
    Pubmed KoreaMed CrossRef
  56. O'Connor BP, Raman VS, Erickson LD, Cook WJ, Weaver LK, Ahonen C, et al. BCMA is essential for the survival of long-lived bone marrow plasma cells. J Exp Med 2004;199:91-8.
    Pubmed KoreaMed CrossRef
  57. Peperzak V, Vikström I, Walker J, Glaser SP, LePage M, Coquery CM, et al. Mcl-1 is essential for the survival of plasma cells. Nat Immunol 2013;14:290-7.
    Pubmed KoreaMed CrossRef
  58. Tokoyoda K, Egawa T, Sugiyama T, Choi BI, Nagasawa T. Cellular niches controlling B lymphocyte behavior within bone marrow during development. Immunity 2004;20:707-18.
    Pubmed CrossRef
  59. Chu VT, Fröhlich A, Steinhauser G, Scheel T, Roch T, Fillatreau S, et al. Eosinophils are required for the maintenance of plasma cells in the bone marrow. Nat Immunol 2011;12:151-9.
    Pubmed CrossRef
  60. Winter O, Moser K, Mohr E, Zotos D, Kaminski H, Szyska M, et al. Megakaryocytes constitute a functional component of a plasma cell niche in the bone marrow. Blood 2010;116:1867-75.
    Pubmed CrossRef
  61. Geffroy-Luseau A, Jégo G, Bataille R, Campion L, Pellat-Deceunynck C. Osteoclasts support the survival of human plasma cells in vitro. Int Immunol 2008;20:775-82.
    Pubmed CrossRef
  62. Cassese G, Lindenau S, de Boer B, Arce S, Hauser A, Riemekasten G, et al. Inflamed kidneys of NZB /W mice are a major site for the homeostasis of plasma cells. Eur J Immunol 2001;31:2726-32.
    Pubmed CrossRef
  63. Tsubaki T, Takegawa S, Hanamoto H, Arita N, Kamogawa J, Yamamoto H, et al. Accumulation of plasma cells expressing CXCR3 in the synovial sublining regions of early rheumatoid arthritis in association with production of Mig/CXCL9 by synovial fibroblasts. Clin Exp Immunol 2005;141:363-71.
    Pubmed KoreaMed CrossRef
  64. van Laar JM, Melchers M, Teng YK, van der Zouwen B, Mohammadi R, Fischer R, et al. Sustained secretion of immunoglobulin by long-lived human tonsil plasma cells. Am J Pathol 2007;171:917-27.
    Pubmed KoreaMed CrossRef
  65. Hoyer BF, Moser K, Hauser AE, Peddinghaus A, Voigt C, Eilat D, et al. Short-lived plasmablasts and long-lived plasma cells contribute to chronic humoral autoimmunity in NZB/W mice. J Exp Med 2004;199:1577-84.
    Pubmed KoreaMed CrossRef
  66. Erickson LD, Lin LL, Duan B, Morel L, Noelle RJ. A genetic lesion that arrests plasma cell homing to the bone marrow. Proc Natl Acad Sci U S A 2003;100:12905-10.
    Pubmed KoreaMed CrossRef
  67. Jang E, Cho WS, Cho ML, Park HJ, Oh HJ, Kang SM, et al. Foxp3+ regulatory T cells control humoral autoimmunity by suppressing the development of long-lived plasma cells. J Immunol 2011;186:1546-53.
    Pubmed CrossRef
  68. Mahévas M, Patin P, Huetz F, Descatoire M, Cagnard N, Bole-Feysot C, et al. B cell depletion in immune thrombocytopenia reveals splenic long-lived plasma cells. J Clin Invest 2013;123:432-42.
    Pubmed KoreaMed CrossRef
  69. Mahévas M, Michel M, Vingert B, Moroch J, Boutboul D, Audia S, et al. Emergence of long-lived autoreactive plasma cells in the spleen of primary warm auto-immune hemolytic anemia patients treated with rituximab. J Autoimmun 2015;62:22-30.
    Pubmed CrossRef
  70. Jang E, Cho S, Pyo S, Nam JW, Youn J. An inflammatory loop between spleen-derived myeloid cells and CD4+ t cells leads to accumulation of long-lived plasma cells that exacerbates lupus autoimmunity. Front Immunol 2021;12:631472.
    Pubmed KoreaMed CrossRef
  71. Jang E, Cho WS, Oh YK, Cho ML, Kim JM, Paik DJ, et al. Splenic long-lived plasma cells promote the development of follicular helper T cells during autoimmune responses. J Immunol 2016;196:1026-35.
    Pubmed CrossRef
  72. Cassese G, Arce S, Hauser AE, Lehnert K, Moewes B, Mostarac M, et al. Plasma cell survival is mediated by synergistic effects of cytokines and adhesion-dependent signals. J Immunol 2003;171:1684-90.
    Pubmed CrossRef
  73. Underhill GH, Minges Wols HA, Fornek JL, Witte PL, Kansas GS. IgG plasma cells display a unique spectrum of leukocyte adhesion and homing molecules. Blood 2002;99:2905-12.
    Pubmed CrossRef
  74. Turner CA Jr, Mack DH, Davis MM. Blimp-1, a novel zinc finger-containing protein that can drive the maturation of B lymphocytes into immunoglobulin-secreting cells. Cell 1994;77:297-306.
    Pubmed CrossRef
  75. Messika EJ, Lu PS, Sung YJ, Yao T, Chi JT, Chien YH, et al. Differential effect of B lymphocyte-induced maturation protein (Blimp-1) expression on cell fate during B cell development. J Exp Med 1998;188:515-25.
    Pubmed KoreaMed CrossRef
  76. Shapiro-Shelef M, Lin KI, McHeyzer-Williams LJ, Liao J, McHeyzer-Williams MG, Calame K. Blimp-1 is required for the formation of immunoglobulin secreting plasma cells and pre-plasma memory B cells. Immunity 2003;19:607-20.
    Pubmed CrossRef
  77. Iwakoshi NN, Lee AH, Glimcher LH. The X-box binding protein-1 transcription factor is required for plasma cell differentiation and the unfolded protein response. Immunol Rev 2003;194:29-38.
    Pubmed CrossRef
  78. Jeong M, Jang E, Choi SS, Ji C, Lee K, Youn J. The function of FK506-binding protein 13 in protein quality control protects plasma cells from endoplasmic reticulum stress-associated apoptosis. Front Immunol 2017;8:222.
    Pubmed KoreaMed CrossRef
  79. Deretic V, Saitoh T, Akira S. Autophagy in infection, inflammation and immunity. Nat Rev Immunol 2013;13:722-37.
    Pubmed KoreaMed CrossRef
  80. Pengo N, Scolari M, Oliva L, Milan E, Mainoldi F, Raimondi A, et al. Plasma cells require autophagy for sustainable immunoglobulin production. Nat Immunol 2013;14:298-305.
    Pubmed CrossRef
  81. Aronov M, Tirosh B. Metabolic control of plasma cell differentiation: what we know and what we don't know. J Clin Immunol 2016;36(Suppl 1):12-7.
    Pubmed CrossRef
  82. Lam WY, Becker AM, Kennerly KM, Wong R, Curtis JD, Llufrio EM, et al. Mitochondrial pyruvate import promotes long-term survival of antibody-secreting plasma cells. Immunity 2016;45:60-73.
    Pubmed KoreaMed CrossRef
  83. Bortnick A, Chernova I, Quinn WJ 3rd, Mugnier M, Cancro MP, Allman D. Long-lived bone marrow plasma cells are induced early in response to T cell-independent or T cell-dependent antigens. J Immunol 2012;188:5389-96.
    Pubmed KoreaMed CrossRef
  84. Taillardet M, Haffar G, Mondière P, Asensio MJ, Gheit H, Burdin N, et al. The thymus-independent immunity conferred by a pneumococcal polysaccharide is mediated by long-lived plasma cells. Blood 2009;114:4432-40.
    Pubmed CrossRef
  85. Reynolds AE, Kuraoka M, Kelsoe G. Natural IgM is produced by CD5- plasma cells that occupy a distinct survival niche in bone marrow. J Immunol 2015;194:231-42.
    Pubmed KoreaMed CrossRef
  86. Miller JJ 3rd, Cole LJ. Resistance of long-lived lymphocytes and plasma cells in rat lymph nodes to treatment with prednisone, cyclophosphamide, 6-mercaptopurine, and actinomycin D. J Exp Med 1967;126:109-25.
    Pubmed KoreaMed CrossRef
  87. Slifka MK, Antia R, Whitmire JK, Ahmed R. Humoral immunity due to long-lived plasma cells. Immunity 1998;8:363-72.
    Pubmed CrossRef
  88. Silverman GJ, Weisman S. Rituximab therapy and autoimmune disorders: prospects for anti-B cell therapy. Arthritis Rheum 2003;48:1484-92.
    Pubmed CrossRef
  89. Thaunat O, Patey N, Caligiuri G, Gautreau C, Mamani-Matsuda M, Mekki Y, et al. Chronic rejection triggers the development of an aggressive intragraft immune response through recapitulation of lymphoid organogenesis. J Immunol 2010;185:717-28.
    Pubmed CrossRef
  90. Kerjaschki D, Regele HM, Moosberger I, Nagy-Bojarski K, Watschinger B, Soleiman A, et al. Lymphatic neoangiogenesis in human kidney transplants is associated with immunologically active lymphocytic infiltrates. J Am Soc Nephrol 2004;15:603-12.
    Pubmed CrossRef
  91. Baddoura FK, Nasr IW, Wrobel B, Li Q, Ruddle NH, Lakkis FG. Lymphoid neogenesis in murine cardiac allografts undergoing chronic rejection. Am J Transplant 2005;5:510-6.
    Pubmed CrossRef
  92. Thaunat O, Field AC, Dai J, Louedec L, Patey N, Bloch MF, et al. Lymphoid neogenesis in chronic rejection: evidence for a local humoral alloimmune response. Proc Natl Acad Sci U S A 2005;102:14723-8.
    Pubmed KoreaMed CrossRef
  93. Perez EE, Orange JS, Bonilla F, Chinen J, Chinn IK, Dorsey M, et al. Update on the use of immunoglobulin in human disease: a review of evidence. J Allergy Clin Immunol 2017;139(3 Suppl):S1-46.
    Pubmed CrossRef
  94. Bonomini V, Vangelista A, Frascà GM, Di Felice A, Liviano D'Arcangelo G. Effects of plasmapheresis in renal transplant rejection: a controlled study. Trans Am Soc Artif Intern Organs 1985;31:698-703.
  95. Allen NH, Dyer P, Geoghegan T, Harris K, Lee HA, Slapak M. Plasma exchange in acute renal allograft rejection: a controlled trial. Transplantation 1983;35:425-8.
    Pubmed CrossRef
  96. Kirubakaran MG, Disney AP, Norman J, Pugsley DJ, Mathew TH. A controlled trial of plasmapheresis in the treatment of renal allograft rejection. Transplantation 1981;32:164-5.
    Pubmed CrossRef
  97. Lefaucheur C, Viglietti D, Hidalgo LG, Ratner LE, Bagnasco SM, Batal I, et al. Complement-activating anti-HLA antibodies in kidney transplantation: allograft gene expression profiling and response to treatment. J Am Soc Nephrol 2018;29:620-35.
    Pubmed KoreaMed CrossRef
  98. Vo AA, Lukovsky M, Toyoda M, Wang J, Reinsmoen NL, Lai CH, et al. Rituximab and intravenous immune globulin for desensitization during renal transplantation. N Engl J Med 2008;359:242-51.
    Pubmed CrossRef
  99. Sautenet B, Blancho G, Büchler M, Morelon E, Toupance O, Barrou B, et al. One-year results of the effects of rituximab on acute antibody-mediated rejection in renal transplantation: RITUX ERAH, a multicenter double-blind randomized placebo-controlled trial. Transplantation 2016;100:391-9.
    Pubmed CrossRef
  100. Bailly E, Ville S, Blancho G, Morelon E, Bamoulid J, Caillard S, et al. An extension of the RITUX-ERAH study, multicenter randomized clinical trial comparing rituximab to placebo in acute antibody-mediated rejection after renal transplantation. Transpl Int 2020;33:786-95.
    Pubmed CrossRef
  101. Huang H, Benoist C, Mathis D. Rituximab specifically depletes short-lived autoreactive plasma cells in a mouse model of inflammatory arthritis. Proc Natl Acad Sci U S A 2010;107:4658-63.
    Pubmed KoreaMed CrossRef
  102. Kamburova EG, Koenen HJ, Borgman KJ, ten Berge IJ, Joosten I, Hilbrands LB. A single dose of rituximab does not deplete B cells in secondary lymphoid organs but alters phenotype and function. Am J Transplant 2013;13:1503-11.
    Pubmed CrossRef
  103. Looney CM, Schroeder A, Tavares E, Garg J, Schindler T, Vincenti F, et al. Obinutuzumab effectively depletes key B-cell subsets in blood and tissue in end-stage renal disease patients. Transplant Direct 2023;9:e1436.
    Pubmed KoreaMed CrossRef
  104. Arnold J, Dass S, Twigg S, Jones CH, Rhodes B, Hewins P, et al. Efficacy and safety of obinutuzumab in systemic lupus erythematosus patients with secondary non-response to rituximab. Rheumatology (Oxford) 2022;61:4905-9.
    Pubmed CrossRef
  105. Kwun J, Burghuber C, Manook M, Iwakoshi N, Gibby A, Hong JJ, et al. Humoral compensation after bortezomib treatment of allosensitized recipients. J Am Soc Nephrol 2017;28:1991-6.
    Pubmed KoreaMed CrossRef
  106. 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.
    Pubmed KoreaMed CrossRef
  107. Woodle ES, Shields AR, Ejaz NS, Sadaka B, Girnita A, Walsh RC, et al. Prospective iterative trial of proteasome inhibitor-based desensitization. Am J Transplant 2015;15:101-18.
    Pubmed CrossRef
  108. Ensor CR, Yousem SA, Marrari M, Morrell MR, Mangiola M, Pilewski JM, et al. Proteasome inhibitor carfilzomib-based therapy for antibody-mediated rejection of the pulmonary allograft: use and short-term findings. Am J Transplant 2017;17:1380-8.
    Pubmed CrossRef
  109. van de Donk NW, Zweegman S. T-cell-engaging bispecific antibodies in cancer. Lancet 2023;402:142-58.
    Pubmed CrossRef
  110. Study for desensitization of chronic kidney disease in adult patients in need of a kidney transplant who are highly sensitized to human leukocyte antigen. ClinicalTrials.gov identifier: NCT05092347. Updated 2024 Jul 31. Accessed 2024 Dec 6. https://clinicaltrials.gov/study/NCT05092347
  111. Tacchetti P, Barbato S, Mancuso K, Zamagni E, Cavo M. Bispecific antibodies for the management of relapsed/refractory multiple myeloma. Cancers (Basel) 2024;16:2337.
    Pubmed KoreaMed CrossRef
  112. Du Z, Zhu S, Zhang X, Gong Z, Wang S. Non-conventional allogeneic anti-BCMA chimeric antigen receptor-based immune cell therapies for multiple myeloma treatment. Cancers (Basel) 2023;15:567.
    Pubmed KoreaMed CrossRef
  113. Hill JA, Kiem ES, Bhatti A, Liu W, Keane-Candib J, Fitzpatrick KS, et al. Anti-HLA antibodies in recipients of CD19 versus BCMA-targeted CAR T-cell therapy. Am J Transplant 2023;23:416-22.
    Pubmed KoreaMed CrossRef
  114. Zhang Z, Markmann C, Yu M, Agarwal D, Rostami S, Wang W, et al. Immunotherapy targeting B cells and long-lived plasma cells effectively eliminates pre-existing donor-specific allo-antibodies. Cell Rep Med 2023;4:101336.
    Pubmed KoreaMed CrossRef