Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2025 Aug 1.
Published in final edited form as: Transplantation. 2024 Feb 16;108(8):1715–1729. doi: 10.1097/TP.0000000000004911

Negative Vaccination Strategies for Promotion of Transplant Tolerance

Matthew J Tunbridge 1,2, Xunrong Luo 2, Angus W Thomson 3,4
PMCID: PMC11265982  NIHMSID: NIHMS1953142  PMID: 38361234

Abstract

Organ transplantation requires the use of immunosuppressive medications that lack antigen specificity, have many adverse side effects, and fail to induce immunological tolerance to the graft. The safe induction of tolerance to allogeneic tissue without compromising host responses to infection or enhancing the risk of malignant disease is a major goal in transplantation. One promising approach to achieve this goal is based on the concept of “negative vaccination”. Vaccination (or actively-acquired immunity) involves the presentation of both a foreign antigen and immunostimulatory adjuvant to the immune system to induce antigen-specific immunity. By contrast, negative vaccination, in the context of transplantation, involves the delivery of donor antigen prior to or after transplantation, together with a “negative adjuvant” to selectively inhibit the allo-immune response. This review will explore established and emerging negative vaccination strategies for promotion of organ or pancreatic islet transplant tolerance. These include donor regulatory myeloid cell infusion, that has progressed to early phase clinical trials, apoptotic donor cell infusion that has advanced to nonhuman primate models, and novel nanoparticle antigen-delivery systems.

Introduction

Clinical transplantation provides life-saving therapy to patients with organ system failure. However, allogeneic transplantation requires the use of immunosuppressive (IS) medications that lack antigen (Ag) specificity, have a multitude of adverse side effects, and fail to induce tolerance to the transplanted organ. The safe induction of immunologic tolerance to allogeneic tissue, without compromising host responses to infection or enhancing the risk of malignant disease, is a major goal in transplantation. Observations of naturally-occurring hematopoietic cell chimerism and tolerance were first made in cattle by Owen in 1945.1 Later experiments in mice by Medawar et al published in 1953, revealed that fetal introduction of donor hematopoietic cells led to tolerance of foreign skin grafts in adulthood.2 Over the past 70 years, numerous experimental approaches, including induction of chimerism, or use of diversely-acting pharmacologic or biologic agents, have been shown to induce transplant tolerance in adult rodents. However, identification of a therapeutic regimen that can safely and predictably induce organ transplant tolerance in humans has proved elusive.

Multiple strategies to promote tolerance have been tested in humans. These include the induction of donor hematopoietic cell chimerism that, despite limited success,3 cannot currently be generalized due to risks associated with the conditioning regimens required to promote chimerism. They also include adoptive cell therapies such a T regulatory cells (T regs) and chimeric antigen receptor (CAR) T regs, for which unequivocal evidence of efficacy has yet to be documented in organ transplantation.4 An approach that holds considerable promise based on extensive preclinical testing in recent years is based on the concept of “negative vaccination”. On the one hand, vaccination (or actively-acquired immunity) involves the presentation of both foreign Ag and an immunostimulatory adjuvant to the immune system to induce Ag-specific immunity. On the other, negative vaccination to promote tolerance, involves the delivery of donor Ag together with a “negative adjuvant” that in combination, selectively and durably inhibit the alloimmune response. Instigation of either response is dependent on specialized Ag-presenting cells (APCs) of the innate immune system with the capacity to induce or regulate allo-immunity.5,6

Antigen-presenting myeloid cells as key to “negative vaccination” – the roles of dendritic cells and macrophages

A potential tolerance vaccine must interact with endogenous Ag-presentation systems. The most proficient Ag-acquiring, processing, and presenting cell of the immune system is the dendritic cell (DC). DCs were first described by Steinman and Cohn in 19737 and are a heterogeneous population of uniquely well-equipped “professional” APCs derived from bone marrow precursors, that play critical roles in both inducing and modulating innate and acquired immune responses.8,9

Most DCs are ultimately derived from common myeloid/lymphoid bone-marrow progenitors. As immature DCs, they migrate via blood to tissues throughout the body where they act as sentinels. As such, they sample the local microenvironment and recognize pathogen-associated molecular patterns (PAMPs) and danger-associated molecular patterns (DAMPs) that trigger their maturation. Consequently, they become well-equipped to present Ag to and activate immune effector cells in secondary lymphoid tissue. While all conventional (c) DCs in blood and tissue inherently express major histocompatibility complex (MHC) class II Ag and cluster of differentiation (CD) 11c to varying degrees, multiple distinct subsets of DCs with different ontogeny, cell surface phenotypes and function have been described. In addition to cDC, these include nonconventional plasmacytoid DCs (pDC), epidermal Langerhans cells (LC), and inflammatory monocyte-derived DCs (MoDC).10,11

cDCs are distributed ubiquitously throughout all lymphoid and nonlymphoid tissues, including all commonly-transplanted organs. They present endogenous Ag through MHC class I and exogenous Ag through MHC class II molecules. Two principal subsets of cDC: cDC1 and cDC2, are distinguished in humans by the presence of cell-surface molecules CD141 and CD1c respectively. cDC1s are proficient in cross-presentation and priming of CD8+ T cells against extracellular Ags, while cDC2 are potent stimulators of naïve T cells, with ability to induce either T helper (Th) cell polarization or regulatory T cells (T-regs). pDC (CD11c+CD123+) are found in peripheral organs, or as circulating cells that express interferon (IFN) regulatory factor (IRF) 7 and secrete type-1 IFN upon viral infection.12 They also exhibit tolerogenic properties, favoring a T-reg response.12 MoDC are monocyte-derived inflammatory cells that infiltrate local tissues as a consequence of inflammation or infection. They release tumor necrosis factor (TNF) and inducible nitric oxide synthase (iNOS) in response to pathogens. Unlike the other DC populations, epidermal LCs have an embryonic origin in hematopoietic precursors and migrate following activation to skin-draining lymph nodes where they present Ags and exhibit immune regulatory functions.13,14 Harnessing the immunoregulatory properties of these diverse DC populations offers a key approach to tolerance induction.

Macrophages are versatile phagocytic innate immune cells derived from circulating monocytes, with considerable phenotypic/functional plasticity and functional diversity in response to the local microenvironment factors and immune cell signaling. They can act as APCs and drive a spectrum of immune responses dependent on local environmental factors, with inflammatory phenotypes that drive Th1 and Th17 cell responses, to anti-inflammatory/tissue repair phenotypes.15

DC-regs and M-regs

DCs maintain self-tolerance in the healthy steady-state.16 Each DC subset appears to have the capacity to promote immune regulation or tolerance through several mechanisms, T cell anergy, apoptosis/deletion and the induction of T-regs. So-called “tolerogenic” or “regulatory” DCs (DC-regs) with capacity to inhibit naïve or memory T cell responses are not defined by a specific cell-surface marker, but in general exhibit characteristic upregulation of negative T cell coregulatory or suppressive molecules such as program death ligand (PD-L) 1 and Fas ligand, and release anti-inflammatory cytokines such as IL-10, indoleamine 2,3-dioxygenase (IDO), adenosine, and other factors. DC-reg are weak T cell stimulators and inhibit their responses by inducing anergy or apoptosis.17

A subset of regulatory macrophages (M-regs) with regulatory and suppressive activity is found in vivo, or can be induced in culture by a variety of signals including monocyte colony-stimulating factor (M-CSF) and interleukin (IL)-10.18 Thus, macrophages are also key agents/targets for promotion of tolerance. They can also spare or induce T-regs.19 M-regs appear to inhibit allo-Ag responses by suppressing T cell proliferation via induction of IL-10-producing T-regs mediated by transforming growth factor- β (TGF-β), retinoic acid and IDO, and by eliminating allo-reactive T cells.20,21

Production and dosing of DC-regs and M-regs for therapeutic applications

Numerous protocols have been described for generation of DC-regs in animal models and humans.22 Processes for manufacturing DC-regs for early phase clinical testing in transplantation and autoimmunity have been described in detail,18,23,24 without consensus regarding an optimal protocol. Usually, peripheral blood monocytes are isolated from leukapheresis products by elutriation and cultured with granulocyte macrophage-colony stimulating factor (GM-CSF) and IL-4, as well as biological agents such as rapamycin, dexamethasone, or Vitamin D3, or anti-inflammatory cytokines such as IL-10.18,22 Upon generation, manufactured DC-regs have low expression of co-stimulatory molecules (CD80 and CD86) but comparatively high levels of PD-L1, and produce minimal levels of IL-12 but high levels of IL-10 in response to CD40 ligation.25 In addition and importantly, unlike immature DC in vivo, they also resist potent maturation signals such as Toll-like receptor (TLR)-4 ligation, or pro-inflammatory cytokines.18,22,25

Guidelines for manufacturing M-regs have also been developed.26 Similarly to DC-reg generation, CD14+ monocytes are isolated from PBMC and cultured in growth factor (M-CSF). After this initial induction, M-regs are stimulated with IFN-γ and express high IDO activity.27 Donor-derived M-regs have been tested safely in human renal transplantation.27,28

In experimental transplantation and in patients, adoptively-transferred regulatory myeloid cells can be either donor- or recipient-derived. Donor cells can immediately present donor Ag to host T cells via the direct pathway of allo-recognition. However, it is unlikely that these allogeneic cells survive for significant periods following their infusion. Indeed, donor MHC Ags and other molecules are soon conveyed to recipient APCs via donor-derived small extracellular vesicles29 that appear to mediate the immune-modifying effect of the donor cell infusion.30,31 Manufactured recipient-derived regulatory cells can either be pulsed with donor allo-Ag or infused unpulsed a day before transplantation, whereupon they acquire and process allograft-derived Ag in vivo. Practically, generation of donor regulatory myeloid cells is not feasible for infusion before deceased donor transplantation due to the time required for their production, although this approach has been documented in nonhuman primate (NHP) kidney graft recipients3235 and is feasible in human live donor transplant recipients.29,36 Pretransplant infusion of autologous DC-regs can extend cardiac allograft survival and induce donor-specific tolerance in rats.37 Moreover, an early phase live-donor human kidney transplant trial of peritransplant autologous DC-reg infusion has been undertaken and shown to be feasible and safe, with some preliminary evidence of DC-reg function.24,28 Mechanistically, it appears that the immunomodulatory effect of autologous DC-reg infusion can be shaped to induce donor-specific tolerance in rats when the cells are co-administered with suboptimal levels of IS agents. However, it is unclear whether this represents indirect Ag presentation in vivo, or an alternative mechanism.37 An additional unanswered question is whether myeloid APCs generated ex vivo from patients with preexisting end-stage organ disease retain the same functional properties and potential as those from healthy individuals.

In terms of dosage, a single donor-derived DC-reg infusion of 2.5–10 ×106 cells/kg has been administered pretransplant in NHP renal transplantation3335 with a similar dose range in phase 1/2a clinical trials in live donor liver transplantation.29 Similar single doses have been used in early phase human trials of donor-derived M-regs (8 × 106 cells/kg,27) and autologous DC-regs (1 × 106 cells/kg,24) in kidney transplantation.

Efficacy of ex vivo-generated regulatory myeloid APCs in animal models

In mice, donor DC-reg infusion without IS therapy prior to islet allografting doubles graft survival time from 15 to 30 days.38 Similarly, recipient DC-reg infusion prior to sex-mismatched skin grafting significantly prolonged graft survival time.39 However, when directly compared in a heart allograft model, rapamycin-treated donor DC-reg infusion 1 week prior to transplant resulted in superior, although not indefinite graft survival.40 Untreated, immature bone-marrow-derived donor DC-regs have been investigated for therapeutic effect, with a modest increase in median survival of murine cardiac transplants from 9.5 to 22 days when infused without IS therapy 1 week prior to transplantation.41 Lutz et al however showed that donor-derived DC-regs that induced T cell unresponsiveness in vitro and in vivo prolonged haplotype-specific cardiac allograft survival (from 8 days to >100 days median survival time) when they were administered 7 days (but not 3, 14, or 28 days) before transplantation.42 Similar results have been obtained in mice using a protocol to generate DC-regs from pluripotent stem cells,30,43 or with monocyte-derived M-reg infusion.20 Alternative approaches with donor-derived M-reg infusion alone prior to heterotopic heart transplantation in mice have also demonstrated significant extension of graft survival from <10 days to >30 days.20

In a NHP model of kidney transplantation, DC-reg infusion has shown significant promise. Pretransplant (day −7) donor DC-reg infusion combined with a minimal IS regimen of co-stimulatory CD28 blockade using CTLA-4 Ig and tapered rapamycin therapy safely led to prolongation of median graft survival time from 39.5 to 113.5 days, with no adverse events or evidence of host sensitization.32,33,35 Prolongation of renal graft survival was also observed when recipient-derived DC-regs pulsed with donor allo-Ag were administered 1 day before transplant.34

In summary, regulatory myeloid APC infusions given preemptively before transplant can effectively prolong graft survival and promote donor-specific tolerance in rodents, principally through 1) induction of Foxp3+ T-regs, 2) induction of T cell anergy, 3) elimination of allo-reactive T cells.44,45 Donor-derived DC-reg infusion also safely prolongs graft survival in NHP when combined with minimal IS drug therapy. A summary of regulatory myeloid cell infusion studies in preclinical models and early-phase clinical trials can be found in Table 1.

Table 1 –

Preclinical and clinical studies of regulatory myeloid cell infusion prior to transplantation

Reference Model Treatment Outcomes
Rodent models
Rastellini et al 199538 Islet allograft
Mouse
Treatment: Donor DCreg infusion 7 days prior to islet transplantation without other IS
Methods: Hepatic DC progenitors cultured with GM-CSF
Control: Syngeneic cell infusion
Mean allograft survival 15.3 days in control group vs 30.3 days in donor cell infusion group (p < 0.001)
Fu et al 199641 Heart allograft
Mouse
Treatment: Donor DCreg infusion 7 days prior to heart transplantation without other IS
Methods: Bone marrow-derived DC cultured with GM-CSF
Control: GM-CSF + IL-4 stimulated donor DC infusion
Mean allograft survival 7 days in control group vs 22 days in donor cell infusion group (p < 0.001)
Lutz et al 200042 Heart allograft
Mouse
Treatment: Donor DCreg infusion 7 days prior to transplantation without other IS
Methods: Bone marrow-derived DC cultured with low concentration GM-CSF
Control: Multiple groups, primary comparator no infusion
Mean allograft survival 8 days in control vs >100 days in donor cell infusion group (p < 0.01)
Taner et al 200540 Heart allograft
Mouse
Treatment: Recipient (syngeneic) DCreg infusion 10, 3, and 0 days prior to transplantation without other IS
Methods: Bone marrow-derived DC cultured with GM-CSF, IL-4, and rapamycin, pulsed with allogeneic donor antigen
Control: Multiple groups, primary comparator no infusion
Mean allograft survival 9 days in control vs >59 days in recipient cell infusion group (p = 0.0014)
Turnquist et al 200784 Heart allograft
Mouse
Treatment: Recipient (syngeneic) DCreg infusion 7 days prior to transplantation with concomitant rapamycin administration
Methods: Bone marrow-derived DC cultured with GM-CSF and rapamycin, pulsed with allogeneic antigens
Control: Multiple groups, primary comparator recipient DCreg infusion without rapamycin co-administration
Mean allograft survival 12 days in control vs >100 days in recipient cell infusion plus rapamycin group (p = 0.0013)
Li et al 2008107 Heart allograft
Mouse
Treatment: Recipient (syngeneic) DCreg infusion 3 days prior and 1 day post transplantation with whole body irradiation
Methods: Syngeneic DCregs isolated from previously tolerised syngeneic recipients using CD45RA Ab and a deoxyspergualin analog.
Control: Control DCs isolated from syngeneic rejecting recipients
Mean allograft survival 13 days in control vs 39.4 days in DCreg infusion group (p < 0.001)
Divito et al 201030 Heart allograft
Mouse
Treatment: Donor DCreg infusion 7 days prior to transplantation without other IS
Methods: Bone marrow-derived DC cultured with GM-CSF, IL-4, and Vitamin D3.
Control: Multiple groups, primary comparator no infusion
Mean allograft survival 11.1 days in control vs 52.2 days in DCreg infusion group (p < 0.001)
Riquelme et al 201320 Heart allograft
Mouse
Treatment: Donor Mreg infusion 8 days prior to transplantation without other IS
Methods: Bone marrow-derived monocytes cultured with M-CSF and pulsed with IFNγ
Control: Multiple groups, primary comparator no infusion
Mean allograft survival 8.7 days in control vs 32.9 days in Mreg infusion group (p < 0.001)
Segovia et al 201439 Skin allograft
Mouse
Treatment: Recipient (syngeneic) DCreg infusion 1 day prior to transplantation with 5 doses anti-CD3 antibody between day −1 to day +7
Methods: Bone marrow-derived DC cultured with low-dose GM-CSF
Control: Multiple groups, primary comparator anti-CD3 alone
Median allograft survival approximately 50 days in control vs >75 days in DCreg infusion plus anti-CD3 group (exact values and p values not reported)
Cai et al 201743 Heart allograft
Mouse
Treatment: Donor DCreg infusion 7 days prior to transplantation without other IS
Methods: Induced pluripotent stem cells cultured with GM-CSF, TGF-β, IL-10, with late IFN-γ stimulation
Control: Multiple groups, primary comparator no infusion
Mean allograft survival 8 days in control vs >100 days in DC-reg infusion group (p < 0.001)
Bériou et al 200537 Heart allograft
Rat
Treatment: Recipient (syngeneic) DCreg infusion ± a deoxyspergualin analog as additional IS
Methods: Bone marrow-derived DC cultured with GM-CSF and IL-4
Control: Multiple groups, primary comparator no treatment
Median allograft survival 6 days in control vs 21.5 days in DCreg infusion alone (p < 0.01) vs 100 days in DCreg infusion plus the deoxyspergualin analog (p < 0.01)
Pêche et al 2005108 Heart allograft
Rat
Treatment: Recipient (syngeneic) DCreg infusion 1 day prior to transplantation without other IS
Methods: Bone marrow-derived DC cultured with GM-CSF and IL-4
Control: Multiple groups, primary comparator no treatment, second comparator donor DCreg infusion 1 day prior to transplantation
Median allograft survival 6 days in no treatment group vs 16.5 days in donor DCreg infusion control (p < 0.05 vs no treatment) vs 22.5 days in recipient DCreg infusion group (p < 0.01 vs no treatment)
Zheng et al 201075 Heart allograft
Rat
Treatment: Recipient (syngeneic) DCreg infusion 7 days prior to transplantation without other IS
Methods: Recipient (syngeneic) bone marrow-derived DCs co-cultured with apoptotic donor splenocytes treated with UV-A irradiation
Control: No treatment, or recipient DCs cultured with nonapoptotic donor splenocytes
Median allograft survival approximately <10 days in control groups vs >25 days in DC infusion group (p < 0.01)
Nonhuman primate models
Ezzelarab et al 201333,35 Renal allograft Treatment: Donor DCreg infusion 7 days prior to transplantation with CTLA4Ig between days −7 and 10, and daily low dose rapamycin for 6 months
Methods: Donor leukapheresis pretransplantation, DC culture with GM-CSF, IL-4, IL-10 and Vitamin D3
Control: No DC-reg infusion, but other IS (CTLA4 Ig and tapering rapamycin) identical
Median allograft survival 39.5 days in control vs 113.5 days in donor DCreg infusion (p = 0.0138)
Ezzelarab et al 201734 Renal allograft Treatment: Recipient (autologous) DCreg infusion 1 day prior to transplantation with CTLA4 Ig between days −7 and 10, and daily low dose rapamycin for 6 months
Methods: Recipient leukapheresis pretransplantation, DC culture with GM-CSF, IL-4, IL-10 and Vitamin D3. DCregs pulsed with donor Ag prior to infusion.
Control: No DCreg infusion, but other IS (CTLA4Ig and tapering rapamycin) identical (shared control group with Ezzelarab et al 201335)
Median allograft survival 39.5 days in control vs 56 days in recipient DCreg infusion (p = 0.1133; NS)
Clinical trials
Hutchinson et al 2008104 Renal allograft (live donor) Treatment: Donor Mreg infusion 5 days prior to transplantation with ATG, tacrolimus, mycophenolate, and weaning steroid
Methods: Donor leukapheresis pretransplantation, Mreg culture with M-CSF, co-cultured with recipient PBMC
Control: No control, single-arm study
Clinical safety, no immediate adverse outcomes (n = 5)
4/5 weaned to tacrolimus monotherapy
1/5 tolerated 8 months of no immunosuppression
2/5 acute rejection episode year 1
Hutchinson et al 2008105 Renal allograft (deceased donor) Treatment: Donor Mreg infusion 5 days posttransplantation with tacrolimus, tapered sirolimus, and tapered steroid
Methods: Donor leukapheresis pretransplantation, Mreg culture with M-CSF, co-cultured with recipient PBMC
Control: No control, single-arm study
Clinical safety, no immediate adverse outcomes (n = 10)
7/10 acute rejection episodes year 1
Hutchinson et al 201127 Renal allograft (live donor) Treatment: Donor Mreg infusion 6–7 days prior to transplantation with tacrolimus, azathioprine, and weaning steroids
Methods: Donor leukapheresis pretransplantation, Mreg culture with M-CSF and IFNγ
Control: No control, single-arm study
Clinical safety, no immediate adverse outcomes (n = 2)
Macedo et al 202129
Tran et al 202336
Liver allograft (live donor) Treatment: Donor DCreg infusion 7 days prior to transplantation with tacrolimus, mycophenolate mofetil, and weaning steroids
Methods: Donor leukapheresis pretransplantation, DCreg culture with GM-CSF, IL-4, IL-10 and Vitamin D3
Control: No control; single-arm study
Clinical safety, no immediate adverse outcomes (n = 14)
Clinical observations and detailed mechanistic studies up to 1 year posttransplant (follow-up to Macedo et al, 2021)
Sawitzki et al 202028
Follow-up reported in Moreau et al 202324
Renal allograft (live donor) Treatment: Recipient (autologous; no Ag) DCreg infusion OR donor Mreg infusion 1 day prior to transplantation plus tacrolimus, mycophenolate mofetil, and standard steroids
Methods: Recipient leukapheresis pretransplantation, DCreg culture with GM-CSF
Control: Basiliximab induction plus tacrolimus, mycophenolate mofetil, and standard steroids
Clinical safety, no immediate adverse outcomes (n = 8 in treatment group)
100% graft survival at 3 years in all groups with 5/8 DCreg infusion patients able to reduce immunosuppression

In vivo targeting of DCs: apoptotic donor cells as negative vaccines

The immune system removes billions of circulating apoptotic cell fragments daily while maintaining self-tolerance.46 This is mediated through a process termed “efferocytosis”, involving phagocytosis of apoptotic fragments by immune cells. Apoptotic cells present pro-phagocytic signals to the immune system in a manner that necrotic cells do not.47 Phagocytic tyrosine kinase receptors mediate this process in an immunologically silent manner with triggering of anti-inflammatory signaling pathways, the release of anti-inflammatory cytokines, and suppression of pro-inflammatory cytokines by inhibition of the nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) signaling pathway.47,48

These host anti-inflammatory pathways can be harnessed to induce donor-specific tolerance. Thus, apoptotic donor cells generated using a variety of methods have been used in experimental models of transplant tolerance. To present the full complement of donor class I and II MHC Ags, donor splenocytes or ex vivo-expanded donor B cells have been used.49 In mice, donor apoptotic cells localize to the recipient’s splenic marginal zone within hours50 and are phagocytosed by recipient DCs and macrophages through host efferocytic receptor tyrosine kinase pathways.51,52 Recipient DCs and macrophages that phagocytose apoptotic donor cells broadly cause an increase in anti-inflammatory cytokines,50,53,54 a reduction in pro-inflammatory cytokines,55 and subsequent induction of donor Ag-specific Foxp3+ T-regs.55,56

Murine models show marked increases in anti-inflammatory cytokines such as IL-10, IL-13, and TGF-β following apoptotic donor leukocyte infusion.53,54 The effect is rapid, with IL-10 production by splenic macrophages within 10 minutes of infusion rapidly altering the phenotype of F4/80+ macrophages to favor high PD-L1 expression.50 In mouse heterotopic heart transplantation, expression of anti-inflammatory cytokines is also increased within allograft tissue.54 Concomitantly, there is a reduction in circulating pro-inflammatory cytokines, including IFN-γ, TNF-α, IL-1β, IL-6, IL-17, and IL-23.54 The direct and immediate effect of the apoptotic body response is observed in mixed leukocyte reactions, where co-culture with apoptotic donor cells results in a reduction in IFN-γ production by allogeneic T cells, and a corresponding increase in cardiac allograft survival.57

Another mechanism by which donor apoptotic cells may induce tolerance is via upregulation of the negative coregulatory molecules PD-L1 and 2 by recipient CD11c+ DCs,52 – pathways that appear crucial for downregulation of T effector cells.58 CD8α+ DC that capture apoptotic cells remain immature in a DC-reg-like state, activating anti-donor CD4+ T cells that are deficiently responsive to IL-7 and IL-15, and cannot upregulate the anti-apoptotic protein B-cell lymphoma-extras large (BCL-XL) or secrete pro-inflammatory cytokines.47 Simultaneously, CD4+ T cells with indirect allo-specificity undergo an initial expansion, followed by profound clonal contraction and sequestration in the spleen without trafficking to the allograft or draining lymphatic system.52 This inhibits CD8+ T cell responses as the remaining CD4+ T cell population suppresses CD40 signaling.59

While allo-specific T cells are downregulated, there is an increase in CD4+Foxp3+ T-regs following apoptotic donor cell administration.55,56 Splenic macrophages are polarized towards the alternatively-activated M2 phenotype, leading to expansion of T-regs in the spleen and allograft-draining lymph nodes.53 Direct in vitro culture of apoptotic allogeneic APCs with naïve CD4+ and CD4+CD25 T cells induces a Treg phenotype with upregulation of Foxp3, high expression of CD62L and CD2, and low expression of co-stimulatory signaling molecules that do not stimulate proliferation or inflammatory cytokine secretion by co-cultured CD4+ T cells.60 Similarly, there appears to be an increase in circulating regulatory B cell phenotypes, with a simultaneous reduction in allo-specific effector B cells.61

Myeloid-derived suppressor cells (MDSCs) are a heterogeneous suppressor cell population that express the myeloid marker glucocorticoid receptor 1 (Gr1) and appear to have an important role in immune tolerance, infiltrating the allograft and suppressing local inflammatory cell responses. In mice, apoptotic donor cell infusions increase CD11b+ MDSCs that maintain the ability to suppress T cell proliferation in vitro. In a mouse cardiac allograft model, these MDSCs migrate to the allograft,62 mediating a protective effect by local inhibition of CD8+ T effector cells and potential induction of Treg in an IFN-y dependent process.62,63 Adoptive transfer of MDSCs in mice has also shown promise by prolonging cardiac and skin allograft survival.64,65

In summary, apoptotic donor leukocytes package the entire spectrum of major and minor donor Ags in a fashion that induces donor-specific tolerance through (1) rapid Ag uptake by recipient APCs; (2) an increase in anti-inflammatory cytokines and concomitant decrease in inflammatory cytokines that drive the allo-immune response; (3) phenotypic alteration of DCs to promote a regulatory response; (4) downregulation of allo-specific CD4+ T cells, inhibition of CD8+ effector T cell responses; (5) induction of Foxp3+ T-regs; and (6) activation of MDSCs.

Production and dosing of apoptotic cells for therapeutic application

Apoptotic cells for Ag-specific tolerance have generally been manufactured using either radiation- or chemically-induced apoptosis. UV radiation combined with freezing/thawing cycles induces apoptosis47,66 though generally, chemically-induced apoptosis has been preferred using the cross-linker 1-ethyl-3-(3-dimethylaminopropyl) carbodiimide (ECDI). ECDI was first synthesized in 1961,67 later finding use as a coupling agent for Ag presentation68 and as an apoptotic agent for Ag-specific tolerance.69

Donor leukocytes are the most common cell source, and both splenocytes and PBMCs have been used. Donor spleens can be processed to a single cell suspension, providing a sufficient number of MHC class I and II positive cells for infusion. However, numbers may not be sufficient for the provision of multiple infusions. Donor B cells can also be isolated from PBMCs and expanded ex vivo to achieve similar cell doses for infusion.49 An alternative approach is ECDI-coupling of recipient leukocytes with donor allo-Ag.50,59 It is important to avoid use of necrotic or late apoptotic cells as they interact differently with the immune system and can actually lead to DC maturation and sensitization.66

With regards to an appropriate dose of apoptotic cells, the aim is to induce tolerance, while avoiding theoretical saturation of efferocytic systems and causing paradoxical sensitization. Historical studies have shown significant variation in immune responsiveness to foreign HLA antigens with different Ag loads.70 A total of 1×108 ECDI-splenocytes can provide indefinite islet allograft survival in mice.58 Further dose-finding studies showed doses as low as 5×106 cells compromised graft protection, but 1×107 cells provided similar protection and may be an appropriate target dose.71 In translational NHP studies, a similar threshold calculated to 0.25 ×109 cells/kg demonstrated efficacy in islet transplantation.61

Efficacy of apoptotic donor cells in animal models

Transplant tolerance using apoptotic cells has been demonstrated in a variety of animal models (mouse, rat, NHP), and with a variety of allografts (islet, heart, skin).

In murine skin allograft models, donor ECDI-splenocyte infusions together with concurrent rapamycin administration significantly extended graft survival time for both full-thickness and vascularized grafts.53,55 Long-term mouse cardiac allograft survival (>100 days) has also been achieved following donor ECDI-splenocyte infusion, including full MHC mismatch models and (together with OX40-OX40L blockade) in presensitized recipients.47,57,62,72 Similarly, donor ECDI-splenocyte infusion alone can induce tolerance of islet allografts,52,56,58 with co-administration of B cell blockade using anti-CD20 inducing tolerance in a rat-to-mouse islet xenograft model.73

An alternative method of activating apoptotic mechanisms is through extracorporeal photopheresis (ECP) of donor leukocytes and pretransplant infusion that induces recipient DC-regs and T-regs for long-term graft survival in mouse models.74 Also, in a rat cardiac allograft model, infusion of donor lymphocytes treated with 8-methoxypsoralen and UV A light leads to uptake by recipient DCs, inducing T-regs and suppression of heart allograft rejection.75

Importantly, it does appear that apoptotic donor cell-induced tolerance can be disrupted in the experimental setting by specific immunological challenges. CMV infection breaks established tolerance in a model of murine islet allotransplantation,76 a mechanism that appears dependent on conversion of anergic T cells to IFN-γ-producing effector T cells.77 Other similar models of tolerance using donor-specific transfusion have shown that disruption of tolerance also occurs with TLR engagement following CpG administration.78

NHPs are valuable preclinical models for transplant immunology research, with highly-conserved MHC proteins between NHP and humans.79,80 Two major studies have investigated the use of apoptotic cells for tolerance induction following islet allo-transplantation in monkeys. One study in cynomolgus monkeys combined ECDI-donor lymphocyte infusion with induction IS comprising anti-thymocyte globulin (ATG), anti-IL-6 antibody (Ab), and 30 days rapamycin administration. The experimental group displayed prolonged graft survival (85.5 versus 13.5 days), with the longest graft survival 133 days.81 Another study in rhesus macaques combined ECDI-donor lymphocyte infusion with induction IS comprising ATG and 30 days co-stimulatory pathway (CD40/CD154) blockade, anti-IL-6 Ab, soluble TNF receptor, and rapamycin. This led to long-term graft tolerance >1 year with no further IS after 21 days posttransplant.61 A broad array of mechanisms were implicated in these studies, including increased circulating T-regs, B regulatory cells (B-regs), and MDSCs following apoptotic donor cell infusion.61 A summary of donor apoptotic cell infusion studies in preclinical models and clinical transplantation can be found in Table 2.

Table 2 –

Preclinical and clinical studies of donor apoptotic cell infusion prior to transplantation

Reference Model Treatment Outcomes
Rodent models
Kaneko et al 200357 Heart allograft
Mouse
Treatment: ECDI-treated donor DC infusion 7 days prior to transplantation without other IS
Methods: Bone-marrow derived DC cultured with GM-CSF and IL-4, pulsed with donor cell lysate and ECDI
Control: Control donor DCs or no treatment
Median survival approximately 10 days in control groups vs >25 days in ECDI-DC infusion group (exact values not reported; p < 0.01)
Wang et al 200647 Heart allograft
Mouse
Treatment: Apoptotic donor splenocyte infusion 7 days prior to transplantation without other IS
Methods: Donor splenocytes with apoptosis induced by UV-B irradiation
Control: Necrotic donor splenocytes or no treatment
Median survival approximately 20 days in control groups vs 40 days in apoptotic donor splenocyte infusion group (exact values not reported, p = 0.0001)
Luo et al 200858 Islet allograft
Mouse
Treatment: ECDI-treated donor splenocyte infusion 7 days prior and 1 day after transplantation without other IS
Methods: Donor splenocytes with apoptosis induced by ECDI
Control: Multiple groups, primary comparator no treatment
Median survival 20 days in control groups vs >100 days in ECDI-splenocyte infusion group (p < 0.0036)
Martin et al 201059 Sex-mismatched skin allograft
Mouse
Treatment: ECDI-treated recipient splenocyte infusion 7 days prior and 0 days after transplantation without other IS
Methods: Recipient splenocytes ECDI-cross-linked to Y chromosomal CD4 antigen
Control: Alternative non-CD4 Ag ECDI-cross linked to recipient splenocytes, or no treatment
Median survival approximately 20 days in control groups vs >100 days in ECDI-splenocyte infusion group (p < 0.001)
Kheradmand et al 201156 Islet allograft
Mouse
Treament: ECDI-donor splenocyte infusion 7 days prior and 1 day after transplantation without other IS
Methods: Donor splenocytes with apoptosis induced by ECDI
Control: No infusion
Median survival <20 days in control group vs >150 days in ECDI-splenocyte infusion group (p value not reported)
Kheradmand et al 201252 Islet allograft
Mouse
Treatment: ECDI-donor splenocyte infusion 7 days prior and 1 day after transplantation ± anti-CD20 antibody or macrophage depletion or CD11c depletion
Methods: Donor splenocytes with apoptosis induced by ECDI, macrophage depletion with lipo-clodronate
Control: No infusions
Median survival <10 days in control group vs <20 days in ECDI-splenocyte infusion plus CD11c depletion group vs >100 days in ECDI-splenocyte (p = 0.008) ± anti-CD20 ± macrophage depletion groups
Chen et al 201262 Heart allograft
Mouse
Treatment: ECDI-donor splenocyte infusions 7 days prior and 1 day after; ± 7 / 14 days after transplantation; ± rapamycin from 1 day prior to 8 days posttransplant
Methods: Donor splenocytes with apoptosis induced by ECDI
Control: No infusions
Mean survival 9 days in control vs 45 days in 2 infusion group (p = 0.0005) vs 85 days in 4 infusion group (p = 0.012) vs >150 days in 2 infusion group with rapamycin (p = 0.0018)
Wang et al 201373 Islet xenograft
Rat >Mouse
Treatment: ECDI-donor splenocyte infusions 7 days prior and 1 day after transplantation plus depleting anti-CD20 antibody
Methods: Donor splenocytes with apoptosis induced by ECDI
Control: No infusions
Median survival approximately <20 days in control vs >40 days in ECDI-splenocyte infusion group (p = 0.0026; exact values not reported). Indefinite graft survival when combined with anti-CD20 Ab
Bryant et al 201463 Heart allograft
Mouse
Treatment: ECDI-donor splenocytes infusions 7 days prior and 1 day after transplantation without other IS
Methods: Donor splenocytes with apoptosis induced by ECDI
Control: No infusion
Median survival approximately 10 days in control vs >20 days in ECDI-splenocyte infusion group (p < 0.05, exact values not reported)
Wang et al 201571 Islet allograft
Mouse
Treatment: ECDI-donor splenocytes infusions 7 days prior and 1 day after transplantation without other IS
Methods: Donor splenocytes with apoptosis induced by ECDI, or recipient splenocytes ECDI-coupled to donor splenocyte lysate
Control: Multiple groups, primary comparator no infusion or recipient splenocytes infusions 7 days prior and 1 day after transplantation
Median survival approximately <20 days in control vs >100 days for either donor or antigen-pulsed recipient ECDI-splenocyte infusion (p = 0.0017)
Lai et al 201754 Heart allograft
Mouse
Treatment: ECDI-donor splenocytes infusions 7 days prior and 1 day after transplantation ± α1-antitrypsin from 1 day prior to 8 days after transplantation
Methods: Donor splenocytes with apoptosis induced by ECDI
Control: No infusion
Mean survival 7 days in control vs 42 days in ECDI-splenocyte infusion group (p = 0.0006) vs >90 days in ECDI-splenocyte infusion plus α1-antitrypsin infusion group (p = 0.0005)
Ding et al 201855 Skin allograft
Mouse
Treatment: ECDI-donor splenocytes 1–3 infusions pre- and posttransplantation ± low-dose rapamycin
Methods: Donor splenocytes with apoptosis induced by ECDI
Control: No infusion
Median survival approximately <20 days in control vs >20 days in 2x ECDI-splenocyte infusions plus rapamycin (p < 0.01, exact values not reported)
Zhang et al 201951 Islet OR heart allografts
Mouse
Treatment: ECDI-donor splenocytes infusions 7 days prior and 1 day after transplantation without other IS
Methods: Donor splenocytes with apoptosis induced by ECDI
Control: No infusion
Islet allograft: Median survival approximately < 20 days in control vs > 100 days in ECDI-splenocyte infusion group (p < 0.001, exact values not reported)
Heart allograft: Median survival approximately <10 days in control vs 15 days in ECDI-splenocyte infusion group (p < 0.01; exact values not reported)
Zhou et al 201953 Skin allograft
Mouse
Treatment: ECDI-donor splenocytes infusion 7 days prior and 1 day after transplantation plus rapamycin
Methods: Donor splenocytes with apoptosis induced by ECDI
Control: ECDI-donor splenocytes infusions alone
Median survival 15 days in control vs 20 days in ECDI-splenocyte infusion plus rapamycin group (no p value reported)
Xingqiang et al 201988 Heart allograft
Mouse
Treatment: ECDI-donor splenocyte infusions 7 days prior and 1 day after transplantation plus cordycepin daily from day 0 to day 7
Methods: Donor splenocytes with apoptosis induced by ECDI
Control: Multiple groups, primary comparator ECDI-donor splenocytes infusions alone, and no infusion
Median survival approximately 10 days in no infusion group vs > 40 days in ECDI-donor splenocyte infusion alone (p = 0.0006) vs 80 days in ECDI-donor splenocyte infusion with cordycepin infusion (p = 0.0001; exact values not reported)
Dangi et al202085 Islet allograft
Sensitized mouse
Treatment: ECDI-donor splenocyte infusion 7 days prior and 1 day after transplantation plus rapamycin daily from 7 days prior to 10 days after transplantation with 4 doses of anti-CD40 Ab.
Methods: Donor splenocytes with apoptosis induced by ECDI
Control: Multiple groups, primary comparator no anti-CD40 Ab, and no infusion
Median survival approximately 5 days in no infusion group vs 10 days in ECDI-donor splenocyte infusion no anti-CD40 group vs 30 days in anti-CD40 (p < 0.05, exact values not reported)
Lai et al 202072 Heart allograft
Mouse
Treatment: ECDI-donor splenocytes + OX40L Ab
Methods: Donor splenocytes with apoptosis induced by ECDI
Control: Multiple groups, primary comparator no anti-OX40L antibody, and no infusion
Median survival approximately < 10 days in no infusion group vs > 20 days in ECDI-donor splenocyte infusion alone group vs >100 days in ECDI-donor splenocyte infusion plus anti-OX40L Ab group (p < 0.001; exact values not reported)
Schneiderman et al 202274 Heart, renal, or liver allograft
Mouse
Treatment: Apoptotic donor leukocyte infusion 7 days prior to transplantation ± rapamycin ± tacrolimus
Methods: Donor splenocyte extracorporeal photopheresis by incubating with methoxypsoralen and UV-A irradiation
Control: Multiple groups, primary comparator infusion of untreated cells, and no infusion group
Heart allograft: Median survival < 10 days in no infusion group vs > 15 days in apoptotic cell infusion group (p < 0.0001) vs >50 days in apoptotic cell infusion plus rapamycin group (p < 0.05)

Liver allograft: Median survival <20 days in no infusion group vs >200 days in apoptotic cell infusion group (p < 0.0001)
Chen et al 201587 Renal allograft
Rat
Treatment: ECDI-donor splenocyte infusions 7 days prior and 1 day after transplantation, ± α1-antitrypsin 1 day prior and 1/3 days after transplantation
Methods: Donor splenocytes with apoptosis induced by ECDI
Control: No infusion
Median survival approximately <10 days in control group vs 50 days in ECDI-splenocyte infusion group vs >200 days in ECDI-splenocyte with α1-antitrypsin infusion (p < 0.05; exact values not reported)
Nonhuman primate models
Lei et al 201581 Islet allograft Treatment: ECDI-donor lymphocyte infusion day 0 after transplantation plus pretransplant ATG 2 doses, anti-IL-6 Ab 4 doses, and rapamycin for 30 days posttransplant
Methods: Lymphoid cells isolated from donor spleen and lymph nodes, incubated with ECDI prior to infusion
Control: Identical immunosuppression without ECDI-donor lymphocyte infusion
Mean survival 13.5 days in control vs 85.5 days in ECDI-donor lymphocyte infusion group (p < 0.0177)
Singh et al 201961 Islet allograft (5/6 HLA mismatch model) Treatment: ECDI-donor lymphocyte infusions 7 days prior and 1 day after transplantation plus anti-CD40 Ab 3 doses, soluble TNF receptor 4 doses, anti-IL-6 Ab 3 doses, and rapamycin for 14 days
Methods: Donor blood draw with B cell isolation, B cell culture and expansion; ECDI treatment of donor B cells prior to infusion.
Control: Identical immunosuppression without ECDI-donor lymphocyte infusion
Median survival approximately <180 days in control group vs >365 days in ECDI-donor lymphocyte infusion group (p = 0.021)
Clinical trials
Mevorach et al 2014106 Allogeneic hematopoietic stem cell transplant Treatment: Single infusion of donor mononuclear apoptotic cells plus usual induction therapy with either busulfan or total body irradiation
Methods: Donor leukapheresis with cellular apoptosis induced with freezing/thawing cycle followed by cell culture with methylprednisolone
Control: No infusion, usual induction therapy
Clinical safety, no immediate adverse outcomes (n = 13)
No episodes of GVHD in high dose group (0/6) compared to 43% in low dose group (3/7)

The “negative adjuvant” effect: leveraging concomitant immunosuppression strategies to enhance Ag-specific tolerance

Infusion of regulatory myeloid cells or apoptotic donor cells prolongs allograft survival and can induce donor-specific tolerance in rodent models. However, these strategies alone are usually insufficient to induce permanent graft tolerance. An adjuvant is a substance that helps and enhances the effect of a drug or biologic system. “Negative adjuvants” that promote a tolerogenic milieu have thus been used to enhance the immune-modifying effect of DC-reg or apoptotic cell infusions.

Mechanistic target of rapamycin (mTOR) inhibitors have immunoregulatory and tolerogenic properties,82 -they inhibit maturation of DCs, enhance Foxp3+ T-reg responses and activity, and inhibit alloreactive T effector cells.60,61,83 Rapamycin co-administered during the peritransplant period has been used as an adjuvant to DC-reg or apoptotic cell infusions, with successful establishment of tolerance in murine models.37,40,62,84

Co-stimulation blockade co-administered at the time of DC-reg infusion reduces the possibility of signal 2-dependent allo-stimulation. Thus DC-reg infusion with CD28 pathway blockade using cytotoxic T lymphocyte-associated antigen (CTLA)-4 immunoglobulin (Ig) in combination with rapamycin prolongs renal allograft survival in NHP models.32,33,35

Co-stimulation blockade strategies, together with apoptotic cell infusion have focused on blockade of CD40/CD15447 and late co-stimulatory molecule OX40-OX40L signaling,72 rather than on B7/CD28 co-stimulation. Apoptotic donor DC infusion 7 days after transplant, together with CD40/CD154 blockade, significantly prolonged murine heart allograft survival from <20 to >100 days.47 Notably, while it appeared that ECDI-treated splenocyte infusion alone was insufficient for the induction of tolerance in a sensitized mouse islet allograft model, addition of CD40 blockade and anti-CD20 therapy facilitated tolerance induction and long-term allograft survival >180 days.85 Similarly, administration of OX40/OX40L blockade with multiple infusions of ECDI-treated splenocytes from day −7 to day +14 posttransplant induced tolerance in a presensitized mouse cardiac allograft model when ECDI-treated cell infusion alone was insufficient.72

Anti-inflammatory molecules have been used in combination with apoptotic donor leukocyte infusions to reduce potential activation of allo-immune responses. α1-antitrypsin is a serine protease inhibitor with immunoregulatory properties mediated via inhibition of inflammatory cytokine production and complement activation.86 It has been used in combination with apoptotic (ECDI-fixed) cell infusion to induce mouse heart allograft tolerance54 and to prolong rat renal allograft survival.87 Cordycepin is a fungal compound with anti-inflammatory properties that promotes an M2 regulatory macrophage phenotype, reduces pro-inflammatory cytokine release and increases IL-10 production by PBMCs.88 Combining cordycepin with donor apoptotic cell infusion significantly prolonged mouse heart allograft survival from approximately 21 days with apoptotic cell infusion alone, to > 40 days with combined treatment.88

Lymphodepletion represents an opportunity to reshape a newly “naïve” immune system. This concept has been used in NHPs, where leukodepletion followed by apoptotic donor cell infusions and a regimen of rapamycin, co-stimulation blockade, and anti-inflammatory molecules (anti-IL-6 and soluble TNF receptor) prolonged islet allograft survival.61,81 Thus overall, a variety of pharmacologic and biologic agents have proven effective as adjuvants to promote the tolerogenicity of donor apoptotic cells.

Other conventional immunosuppressive agents, including calcineurin inhibitors and mycophenolate do not potentiate or interfere with apoptotic cell infusion-induced tolerance in murine models.71 DC-reg infusions in human clinical trials have been combined with conventional immunosuppression using tacrolimus, mycophenolate, and steroids,24,36 although there is concern that calcineurin inhibitors could potentially impair T-reg induction.89 Clinical translation remains focused on the safe introduction of negative vaccination strategies to existing clinical practice, and it is likely that future studies will combine these approaches with triple immunosuppression as standard of care, while acknowledging that the most promising animal models have used combinations of co-stimulation blockade and mTOR inhibition.

Developing alternative methods of negative vaccination via targeting of APCs in vivo

Apoptotic cell infusions are a well-developed approach to target the in vivo Ag-presentation system. However, alternative methods of Ag delivery to inhibit anti-donor immune reactivity and promote tolerance are under investigation.

Exosomes are small extracellular membrane vesicles secreted/released by hematopoietic cells, including DCs that express MHC class II. Pretransplant infusion of exosomes secreted by immature donor DCs prolongs MHC-mismatched heart graft survival in rats.90 Moreover, combined with IS or additional donor Ag-specific regulatory cell therapy (T-regs), exosome administration induces tolerance in a rat liver transplant model.91 Exosome-based methods avoid any potential risk of regulatory cell maturation in vivo and host sensitization.

Donor Ag coupling to specialized drug-delivery nanoparticles represents a promising technology for targeting APCs in situ that does not require isolation and culture of donor or recipient cells ex vivo. Delivery platforms such as poly(lactide-co-glycolide) (PLG) in ovalbumin (OVA)-sensitized mice,92,93 and β sheets of proteinaceous aggregates in murine skin transplant models94 have shown promise. These nanoparticles can also encapsulate immune-modifying drugs such as rapamycin,93,95,96 and be used to target myeloid cells in vivo at the time of transplant to promote tolerance.97,98

DCs sample their environment through an array of pattern recognition receptors (PRRs) including C-type lectin receptors (CLR). CLRs can act as adhesion molecules and as endocytic receptors, recognising a wide array of carbohydrate-containing molecules. The nature of the ligand and local microenvironment appears to determine whether the immune response triggered by receptor activation is inflammatory or tolerogenic. Approaches to activate these receptors use Ag fused to either anti-receptor antibodies or receptor-activating carbohydrate moieties. One molecule of interest expressed on cDC1s is the endocytic receptor DEC-205 (CD205).99 Anti-CD205 fusion antibodies bound to specific Ag have shown promise in experimental models of autoimmune disease,100 but their complex distribution across different cell types in humans may limit translation to clinical studies.9 CD-209 (the C-type lectin DC-SIGN) expressed on DCs has also been targeted using porous silicon nanoparticles loaded with rapamycin, and in vitro experiments have demonstrated phenotypic change towards a regulatory phenotype in DCs, with an increase in Ag-specific splenic T-regs when administered in OVA-sensitized mice.95,96 Other preclinical models have used glycan-modified and sialylated Ags to target other CLRs and sialic-acid binding immunoglobulin-type lectins (Siglecs) with some success in murine models of experimental autoimmune encephalomyelitis and allergy.101,102 It is unclear how applicable these methods will be in transplantation where tolerance is desired across a broad array of polymorphic MHC molecules. A summary of alternative antigen delivery approaches in preclinical models can be found in Table 3.

Table 3.

Preclinical studies of alternative forms of donor antigen delivery

Reference Model Treatment Outcomes
Rodent models
Bryant et al 2014109 Islet allograft
Mouse
Treatment: Donor antigen-coupled poly(d,l-lactide-co-glycolide) (PLG) particles infused 7 days prior and 1 day after transplantation without other IS
Methods: PLG coupled by ECDI to donor spleen lysate to create donor antigen-couple PLG particles
Control: Multiple dose groups, primary comparator sham injection of carrier molecule without donor antigen coupling
Median survival <20 days in control group vs >20 days in nanoparticle-treated group (p = 0.0099; exact values not reported)
Shah et al 201992 Skin allograft
Mouse
Treatment: Donor antigen-coupled PLG infused 7 days prior and 1 day after transplantation without other IS
Methods: PLG coupled by ECDI to donor protein to create donor antigen-coupled PLG particles
Control: Multiple dose groups, primary comparator sham injection of carrier molecule without donor antigen coupling
Median survival <20 days in control group vs >20 days in nanoparticle-treated group (p < 0.001; exact values not reported)
Luo et al 2023110 Heart allograft
Mouse
Treatment: Donor BMDC-derived exosomes injected daily from 0 days to 3 days after transplantation without other IS
Methods: BM-derived donor DCs genetically altered to create high PD-L1 expression, with supernatant collected when cell fusion rate reached 80%
Control: Multiple groups, primary comparator no injection of exosomes
Mean survival approximately 6 days in control group vs 12 days in exosome-treated group (p = 0.001; exact values not reported)
Pêche et al2003111 Heart allograft
Rat
Treatment: Donor BMDC-derived exosomes 10μg injected 14 days and 7 days prior to transplantation without other IS
Methods: BM-derived donor cells co-cultured with GM-CSF and IL-4 with supernatant collected at day 10
Control: Multiple dose groups, primary comparator no injection of exosomes
Median survival 6 days in control group vs 25 days in exosome-treated group (p < 0.01)
Pêche et al 200690 Heart allograft
Rat
Treatment: Donor BMDC-derived exosomes 25μg injected 0 days and 6 days after transplantation without other IS
Methods: BM-derived donor cells co-cultured with GM-CSF and IL-4 with supernatant collected at day 10
Control: Multiple dose groups, primary comparator no injection of exosomes
Median survival 6 days in control group vs 25.5 days in exosome-treated group (p < 0.05)
Ma et al 201691 Liver allograft
Rat
Treatment: Donor BMDC-derived exosomes 25μg injected 7 days prior, 0 days and 7 days after transplantation without other IS
Methods: BM-derived donor (syngeneic) cells co-cultured with GM-CSF and IL-4 with supernatant collected at days 6 and 11
Control: Multiple dose groups, primary comparator no injection of exosomes
Median survival 10 days in control group vs 37 days in exosome-treated group (p < 0.0001)

Clinical translation

The 2022 Fifth International Sam Strober Workshop on clinical transplant tolerance described ongoing efforts at clinical translation of tolerance-promoting strategies in organ transplantation.103 Preclinical NHP studies have demonstrated the feasibility, safety, and efficacy of donor-derived DC-reg infusion prior to transplant in prolonging renal allograft survival,35,79,81 and of 2-dose pre- and posttransplant apoptotic donor cell treatment in promoting islet allograft tolerance.61

Early phase clinical trials of DC-regs and M-regs have been undertaken internationally. The ONE study was a concerted evaluation of 7, small, single-center phase 1/2a clinical trials of regulatory cell-based therapy in live-donor kidney transplantation using autologous T-regs or DC-regs, or donor-derived M-regs.28 Autologous DC-reg infusion 1 day prior to transplantation was compared to standard-of-care IS with basiliximab induction, tacrolimus, mycophenolate, and tapering steroid. DC-reg infusions were safe, with acute rejection in 2/8 patients in the treatment group and 1/9 in the reference group – all were treated with glucocorticoids and graft survival at 3 years was 100%. There were no differences in infectious complications. IS was reduced with mycophenolate tapering in 5/8, and tacrolimus monotherapy achieved in 2/8 patients.24

Two earlier small phase 1/2a studies demonstrated the safety of either donor- or recipient-derived M-reg infusion prior to kidney transplantation. These cell infusions in combination with standard induction IS appeared to facilitate periods of reduced or no IS within the first year of transplant. However, rates of acute rejection were high at 4/10 and long-term IS was universally required.104,105 An ongoing phase 1/2a trial at the University of Pittsburgh will report on 13 live donor liver transplant recipients given donor-derived DC-regs (within the dose range that proved efficacious in NHP) a week before transplant, and who received a protocol graft biopsy at 1 year. Patients with “permissive” biopsies at 1 year then began early withdrawal of IS therapy. In a second trial at the same center, 14 live donor liver transplant recipients with a permissive biopsy at 1–2 years posttransplant have received donor-derived DC-regs within the same dose range as in the initial trial, followed by weaning of IS therapy. In a further trial at the University of Pittsburgh dose escalation of donor-derived DC-reg infusion pretransplant in live donor kidney recipients is being conducted in a total of 14 donor-recipient pairs. These DC-reg trials incorporate detailed mechanistic studies to ascertain the fate of the infused regulatory cells, and their influence on allo-immune responses.29,36

Apoptotic donor cell therapy has not yet been studied in human organ transplantation. However, apoptotic donor mononuclear cell infusion in 13 patients receiving HLA-matched allogeneic bone marrow transplants for the prevention of GVHD was successful in a small phase 1/2a trial.106 Those in the high-dose group had no episodes of GVHD as compared to 23% in the low-dose group.106 Trials in solid organ transplantation are likely to be undertaken once feasible and effective treatment protocols for live and deceased donor transplantation have been established in NHP models.

Conclusions

Induction of tolerance in humans to allogeneic tissue expressing the full complement of donor MHC and non-HLA Ags remains a challenging task. “Negative vaccination” approaches that deliver donor Ag together with a negative adjuvant have progressed significantly (Fig 1). Regulatory myeloid APC therapy has progressed to human trials, while the use of donor apoptotic cells shows significant promise in NHP models. It remains to be seen whether true tolerance will be achieved in clinical translation, or whether using the promising strategies described herein, a reduced burden of IS - which would in itself be of significant benefit – can be achieved, especially if accomplished early posttransplant.

Figure 1:

Figure 1:

Diagrammatic representation of strategies for tolerance induction using negative vaccination in solid organ transplantation. Created using biorender.com.

Financial disclosure:

MT is supported by a Jacquot Research Entry Scholarship from the Don and Lorraine Jacquot Foundation. He has previously received travel sponsorship from Amgen.

XL is in receipt of National Institutes of Health research grants R01 DK 132889, U01 AI 090956 and U19 AI 131471.

AWT is in receipt of National Institutes of Health research grants R01 AI 118777, U01 AI 136779 and U19 AI 131453.

Abbreviations

Ab

antibody

Ag

antigen

APC(s)

antigen-presenting cell(s)

ATG

anti-thymocyte globulin

BCL-XL

B-cell lymphoma – extra large

CAR

chimeric antigen receptor

CD

cluster of differentiation

CTLA-4 Ig

cytotoxic T lymphocyte associated protein 4 immunoglobulin

DAMP

danger associated molecular pattern

DC(s)

dendritic cell(s)

DC-reg(s)

regulatory dendritic cells

ECDI

1-ethyl-3-(3-dimethylaminopropyl) carbodiimide

Foxp3

forkhead boxp3

GM-CSF

granulocyte macrophage-colony stimulating factor

Gr1

glucocorticoid receptor 1

GVHD

graft versus host disease

IL

interleukin

IS

immunosuppressive/immunosuppression

IFN

interferon

Inos

inducible nitric oxide synthase

M-CSF

macrophage-colony stimulating factor

MDSC(s)

myeloid-derived suppressor cells

MHC

major histocompatibility complex

M-reg(s)

regulatory macrophage(s)

NF-κB

nuclear factor kappa-light-chain-enhancer of activated B cells

NHP

nonhuman primate

OVA

ovalbumin

PAMP

pathogen-associated molecular pattern

PBMC

peripheral blood mononuclear cell

PD-L1

programmed death ligand-1

PLG

polylactide-co-glycolide

Th cell

T helper cell

TGF-β

transforming growth factor β

TLR

Toll-like receptor

TNF

tumor necrosis factor

T-reg(s)

regulatory T cell(s)

Footnotes

Disclosures: AWT is co-inventor of University of Pittsburgh invention disclosures and a provisional patent application that concern protocols for generation and clinical evaluation of regulatory dendritic cells. XL is co-inventor of use of ECDI-fixed cell tolerance as a method for preventing allograft rejection.

References

  • 1.Owen RD. Immunogenetic Consequences of Vascular Anastomoses Between Bovine Twins. Science. 1945;102(2651):400–401. [DOI] [PubMed] [Google Scholar]
  • 2.Billingham RE, Brent L, Medawar PB. Actively acquired tolerance of foreign cells. Nature. 1953;172(4379):603–606. [DOI] [PubMed] [Google Scholar]
  • 3.Spitzer TR, Sachs DH. Transplantation tolerance through hematopoietic chimerism. N Engl J Med. 2022;386(24):2332–2333. [DOI] [PubMed] [Google Scholar]
  • 4.Pilat N, Steiner R, Sprent J. Treg therapy for the induction of immune tolerance in transplantation—not lost in translation? Int J Mol Sci. 2023;24(2):1752. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Azimzadeh AM, Bromberg JS. Transplantation: negative vaccination to modulate transplant immunity. Nat Rev Nephrol. 2013;9(10):557–559. [DOI] [PubMed] [Google Scholar]
  • 6.Turnquist HR, Thomson AW. Taming the lions: manipulating dendritic cells for use as negative cellular vaccines in organ transplantation. Curr Opin Organ Transplant. 2008;13(4):350–357. [DOI] [PubMed] [Google Scholar]
  • 7.Steinman RM, Cohn ZA. Identification of a novel cell type in peripheral lymphoid organs of mice. I. Morphology, quantitation, tissue distribution. J Exp Med. 1973;137(5):1142–1162. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Obregon C, Kumar R, Pascual MA, et al. Update on dendritic cell-induced immunological and clinical tolerance. Front Immunol. 2017;8:1514. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Castenmiller C, Keumatio-Doungtsop BC, van Ree R, et al. Tolerogenic immunotherapy: targeting DC surface receptors to induce antigen-specific tolerance. Front Immunol. 2021;12:643240. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Naik SH, Sathe P, Park HY, et al. Development of plasmacytoid and conventional dendritic cell subtypes from single precursor cells derived in vitro and in vivo. Nat Immunol. 2007;8(11):1217–1226. [DOI] [PubMed] [Google Scholar]
  • 11.Wu L, Liu YJ. Development of dendritic-cell lineages. Immunity. 2007;26(6):741–750. [DOI] [PubMed] [Google Scholar]
  • 12.Rogers NM, Isenberg JS, Thomson AW. Plasmacytoid dendritic cells: no longer an enigma and now key to transplant tolerance? Am J Transplant. 2013;13(5):1125–1133. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Lutz MB, Döhler A, Azukizawa H. Revisiting the tolerogenicity of epidermal Langerhans cells. Immunol Cell Biol. 2010;88(4):381–386. [DOI] [PubMed] [Google Scholar]
  • 14.Nirschl CJ, Anandasabapathy N. Duality at the gate: skin dendritic cells as mediators of vaccine immunity and tolerance. Hum Vaccin Immunother. 2016;12(1):104–116. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Broichhausen C, Riquelme P, Geissler EK, et al. Regulatory macrophages as therapeutic targets and therapeutic agents in solid organ transplantation. Curr Opin Organ Transplant. 2012;17(4):332–342. [DOI] [PubMed] [Google Scholar]
  • 16.Ohnmacht C, Pullner A, King SB, et al. Constitutive ablation of dendritic cells breaks self-tolerance of CD4 T cells and results in spontaneous fatal autoimmunity. J Exp Med. 2009;206(3):549–559. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Thomson AW, Metes DM, Ezzelarab MB, et al. Regulatory dendritic cells for human organ transplantation. Transplant Rev (Orlando). 2019;33(3):130–136. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Suuring M, Moreau A. Regulatory macrophages and tolerogenic dendritic cells in myeloid regulatory cell-based therapies. Int J Mol Sci. 2021;22(15). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Huang H, Dawicki W, Zhang X, et al. Tolerogenic dendritic cells induce CD4+CD25hiFoxp3+ regulatory T cell differentiation from CD4+CD25-/loFoxp3- effector T cells. J Immunol. 2010;185(9):5003–5010. [DOI] [PubMed] [Google Scholar]
  • 20.Riquelme P, Tomiuk S, Kammler A, et al. IFN-γ-induced iNOS expression in mouse regulatory macrophages prolongs allograft survival in fully immunocompetent recipients. Mol Ther. 2013;21(2):409–422. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Riquelme P, Hutchinson JA. Novel molecules mediate specialized functions of human regulatory macrophages. Curr Opin Organ Transplant. 2018;23(5):533–537. [DOI] [PubMed] [Google Scholar]
  • 22.Rosborough BR, Raïch-Regué D, Turnquist HR, et al. Regulatory myeloid cells in transplantation. Transplantation. 2014;97(4):367–379. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Zahorchak AF, DeRiggi ML, Muzzio JL, et al. Manufacturing and validation of Good Manufacturing Practice-compliant regulatory dendritic cells for infusion into organ transplant recipients. Cytotherapy. 2023;25(4):432–441. [DOI] [PubMed] [Google Scholar]
  • 24.Moreau A, Kervella D, Bouchet-Delbos L, et al. A Phase I/IIa study of autologous tolerogenic dendritic cells immunotherapy in kidney transplant recipients. Kidney Int. 2023;103(3):627–637. [DOI] [PubMed] [Google Scholar]
  • 25.Zahorchak AF, Macedo C, Hamm DE, et al. High PD-L1/CD86 MFI ratio and IL-10 secretion characterize human regulatory dendritic cells generated for clinical testing in organ transplantation. Cell Immunol. 2018;323:9–18. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Hutchinson JA, Ahrens N, Geissler EK. MITAP-compliant characterization of human regulatory macrophages. Transpl Int. 2017;30(8):765–775. [DOI] [PubMed] [Google Scholar]
  • 27.Hutchinson JA, Riquelme P, Sawitzki B, et al. Cutting Edge: Immunological consequences and trafficking of human regulatory macrophages administered to renal transplant recipients. J Immunol. 2011;187(5):2072–2078. [DOI] [PubMed] [Google Scholar]
  • 28.Sawitzki B, Harden PN, Reinke P, et al. Regulatory cell therapy in kidney transplantation (The ONE Study): a harmonised design and analysis of seven non-randomised, single-arm, phase 1/2A trials. Lancet. 2020;395(10237):1627–1639. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Macedo C, Tran LM, Zahorchak AF, et al. Donor-derived regulatory dendritic cell infusion results in host cell cross-dressing and T cell subset changes in prospective living donor liver transplant recipients. Am J Transplant. 2021;21(7):2372–2386. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Divito SJ, Wang Z, Shufesky WJ, et al. Endogenous dendritic cells mediate the effects of intravenously injected therapeutic immunosuppressive dendritic cells in transplantation. Blood. 2010;116(15):2694–2705. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Morelli AE, Thomson AW. Orchestration of transplantation tolerance by regulatory dendritic cell therapy or in-situ targeting of dendritic cells. Curr Opin Organ Transplant. 2014;19(4):348–356. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Ezzelarab MB, Lu L, Guo H, et al. Eomesodermin(lo) CTLA4(hi) alloreactive CD8+ memory T cells are associated with prolonged renal transplant survival induced by regulatory dendritic cell infusion in CTLA4 immunoglobulin-treated nonhuman primates. Transplantation. 2016;100(1):91–102. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Ezzelarab MB, Lu L, Shufesky WF, et al. Donor-derived regulatory dendritic cell infusion maintains donor-reactive CD4(+)CTLA4(hi) T cells in non-human primate renal allograft recipients treated with CD28 co-stimulation blockade. Front Immunol. 2018;9:250. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Ezzelarab MB, Raich-Regue D, Lu L, et al. Renal allograft survival in nonhuman primates infused with donor antigen-pulsed autologous regulatory dendritic cells. Am J Transplant. 2017;17(6):1476–1489. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Ezzelarab MB, Zahorchak AF, Lu L, et al. Regulatory dendritic cell infusion prolongs kidney allograft survival in nonhuman primates. Am J Transplant. 2013;13(8):1989–2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Tran LM, Macedo C, Zahorchak AF, et al. Donor-derived regulatory dendritic cell infusion modulates CD8+ and NK cell responses after live donor liver transplantation. Sci Transl Med. 2023; 15(717):eadf4287. [DOI] [PubMed] [Google Scholar]
  • 37.Bériou G, Pêche H, Guillonneau C, et al. Donor-specific allograft tolerance by administration of recipient-derived immature dendritic cells and suboptimal immunosuppression. Transplantation. 2005;79(8):969–972. [DOI] [PubMed] [Google Scholar]
  • 38.Rastellini C, Lu L, Ricordi C, et al. Granulocyte/macrophage colony-stimulating factor-stimulated hepatic dendritic cell progenitors prolong pancreatic islet allograft survival. Transplantation. 1995;60(11):1366–1370. [PMC free article] [PubMed] [Google Scholar]
  • 39.Segovia M, Louvet C, Charnet P, et al. Autologous dendritic cells prolong allograft survival through Tmem176b-dependent antigen cross-presentation. Am J Transplant. 2014;14(5):1021–1031. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Taner T, Hackstein H, Wang Z, et al. Rapamycin-treated, alloantigen-pulsed host dendritic cells induce ag-specific T cell regulation and prolong graft survival. Am J Transplant. 2005;5(2):228–236. [DOI] [PubMed] [Google Scholar]
  • 41.Fu F, Li Y, Qian S, et al. Costimulatory molecule-deficient dendritic cell progenitors (MHC class II+, CD80dim, CD86-) prolong cardiac allograft survival in nonimmunosuppressed recipients. Transplantation. 1996;62(5):659–665. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Lutz MB, Suri RM, Niimi M, et al. Immature dendritic cells generated with low doses of GM-CSF in the absence of IL-4 are maturation resistant and prolong allograft survival in vivo. Eur J Immunol. 2000;30(7):1813–1822. [DOI] [PubMed] [Google Scholar]
  • 43.Cai S, Hou J, Fujino M, et al. iPSC-derived regulatory dendritic cells inhibit allograft rejection by generating alloantigen-specific regulatory T cells. Stem Cell Reports. 2017;8(5):1174–1189. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Morelli AE, Thomson AW. Tolerogenic dendritic cells and the quest for transplant tolerance. Nat Rev Immunol. 2007;7(8):610–621. [DOI] [PubMed] [Google Scholar]
  • 45.Ochando J, Ordikhani F, Jordan S, et al. Tolerogenic dendritic cells in organ transplantation. Transpl Int. 2020;33(2):113–127. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Voll RE, Herrmann M, Roth EA, et al. Immunosuppressive effects of apoptotic cells. Nature. 1997;390(6658):350–351. [DOI] [PubMed] [Google Scholar]
  • 47.Wang Z, Larregina AT, Shufesky WJ, et al. Use of the inhibitory effect of apoptotic cells on dendritic cells for graft survival via T-cell deletion and regulatory T cells. Am J Transplant. 2006;6(6):1297–1311. [DOI] [PubMed] [Google Scholar]
  • 48.Szondy Z, Sarang Z, Kiss B, et al. Anti-inflammatory mechanisms triggered by apoptotic cells during their clearance. Front Immunol. 2017;8:909. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.McCarthy DP, Bryant J, Galvin JP, et al. Tempering allorecognition to induce transplant tolerance with chemically modified apoptotic donor cells. Am J Transplant. 2015;15(6):1475–1483. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Getts DR, Turley DM, Smith CE, et al. Tolerance induced by apoptotic antigen-coupled leukocytes is induced by PD-L1+ and IL-10-producing splenic macrophages and maintained by T regulatory cells. J Immunol. 2011;187(5):2405–2417. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Zhang L, DeBerge M, Wang J, et al. Receptor tyrosine kinase MerTK suppresses an allogenic type I IFN response to promote transplant tolerance. Am J Transplant. 2019;19(3):674–685. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Kheradmand T, Wang S, Bryant J, et al. Ethylenecarbodiimide-fixed donor splenocyte infusions differentially target direct and indirect pathways of allorecognition for induction of transplant tolerance. J Immunol. 2012;189(2):804–812. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Zhou B, Zhang Y, Zhang D, et al. ECDI-fixed donor splenocytes prolong skin allograft survival by promoting M2 macrophage polarization and inducing regulatory T cells. FASEB Bioadv. 2019;1(11):706–718. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Lai X, Qiu L, Zhao Y, et al. Ethylene carbodiimide-fixed donor splenocytes combined with α−1 antitrypsin induce indefinite donor-specific protection to mice cardiac allografts. Transpl Int. 2017;30(3):305–317. [DOI] [PubMed] [Google Scholar]
  • 55.Ding J, Liu S, Zhang D, et al. Transfusion of ethylene carbodiimide-fixed donor splenocytes prolongs survival of vascularized skin allografts. J Surg Res. 2018;221:343–352. [DOI] [PubMed] [Google Scholar]
  • 56.Kheradmand T, Wang S, Gibly RF, et al. Permanent protection of PLG scaffold transplanted allogeneic islet grafts in diabetic mice treated with ECDI-fixed donor splenocyte infusions. Biomaterials. 2011;32(20):4517–4524. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Kaneko K, Morelli AE, Wang Z, et al. Alloantigen presentation by ethylcarbodiimide-treated dendritic cells induces T cell hyporesponsiveness, and prolongs organ graft survival. Clin Immunol. 2003;108(3):190–198. [DOI] [PubMed] [Google Scholar]
  • 58.Luo X, Pothoven KL, McCarthy D, et al. ECDI-fixed allogeneic splenocytes induce donor-specific tolerance for long-term survival of islet transplants via two distinct mechanisms. Proc Natl Acad Sci U S A. 2008;105(38):14527–14532. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Martin AJ, McCarthy D, Waltenbaugh C, et al. Ethylenecarbodiimide-treated splenocytes carrying male CD4 epitopes confer histocompatibility Y chromosome antigen transplant protection by inhibiting CD154 upregulation. J Immunol. 2010;185(6):3326–3336. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Albert MH, Yu XZ, Magg T. Ethylenecarbodiimide-coupled allogeneic antigen presenting cells induce human CD4+ regulatory T cells. Clin Immunol. 2008;129(3):381–393. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Singh A, Ramachandran S, Graham ML, et al. Long-term tolerance of islet allografts in nonhuman primates induced by apoptotic donor leukocytes. Nat Commun. 2019;10(1):3495. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Chen G, Kheradmand T, Bryant J, et al. Intragraft CD11b(+) IDO(+) cells mediate cardiac allograft tolerance by ECDI-fixed donor splenocyte infusions. Am J Transplant. 2012;12(11):2920–2929. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Bryant J, Lerret NM, Wang JJ, et al. Preemptive donor apoptotic cell infusions induce IFN-γ-producing myeloid-derived suppressor cells for cardiac allograft protection. J Immunol. 2014;192(12):6092–6101. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Cao P, Sun Z, Zhang F, et al. TGF-β enhances immunosuppression of myeloid-derived suppressor cells to induce transplant immune tolerance through affecting Arg-1 expression. Front Immunol. 2022;13:919674. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Fujimoto K, Uchida K, Yin E, et al. Analysis of therapeutic potential of monocytic myeloid-derived suppressor cells in cardiac allotransplantation. Transpl Immunol. 2021;67:101405. [DOI] [PubMed] [Google Scholar]
  • 66.Kushwah R, Wu J, Oliver JR, et al. Uptake of apoptotic DC converts immature DC into tolerogenic DC that induce differentiation of Foxp3+ Treg. Eur J Immunol. 2010;40(4):1022–1035. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Sheehan J, Cruickshank P, Boshart G. Notes- A convenient synthesis of water-soluble carbodiimides. J Org Chem. 1961;26(7):2525–2528. [Google Scholar]
  • 68.Miller SD, Wetzig RP, Claman HN. The induction of cell-mediated immunity and tolerance with protein antigens coupled to syngeneic lymphoid cells. J Exp Med. 1979;149(3):758–773. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Jenkins MK, Schwartz RH. Antigen presentation by chemically modified splenocytes induces antigen-specific T cell unresponsiveness in vitro and in vivo. J Exp Med. 1987;165(2):302–319. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Madsen JC, Superina RA, Wood KJ, et al. Immunological unresponsiveness induced by recipient cells transfected with donor MHC genes. Nature. 1988;332(6160):161–164. [DOI] [PubMed] [Google Scholar]
  • 71.Wang S, Zhang X, Zhang L, et al. Preemptive tolerogenic delivery of donor antigens for permanent allogeneic islet graft protection. Cell Transplant. 2015;24(6):1155–1165. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Lai X, Yao Z, Ning F, et al. Blockade of OX40/OX40L pathway combined with ethylene-carbodiimide-fixed donor splenocytes induces donor-specific allograft tolerance in presensitized recipients. Ann Transl Med. 2020;8(4):84. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Wang S, Tasch J, Kheradmand T, et al. Transient B-cell depletion combined with apoptotic donor splenocytes induces xeno-specific T- and B-cell tolerance to islet xenografts. Diabetes. 2013;62(9):3143–3150. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Schneiderman J, Qiu L, Yeap XY, et al. Pre-transplant infusion of donor leukocytes treated with extracorporeal photochemotherapy induces immune hypo-responsiveness and long-term allograft survival in murine models. Sci Rep. 2022;12(1):7298. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Zheng DH, Dou LP, Wei YX, et al. Uptake of donor lymphocytes treated with 8-methoxypsoralen and ultraviolet A light by recipient dendritic cells induces CD4+CD25+Foxp3+ regulatory T cells and down-regulates cardiac allograft rejection. Biochem Biophys Res Commun. 2010;395(4):540–546. [DOI] [PubMed] [Google Scholar]
  • 76.Yu S, Dangi A, Burnette M, et al. Acute murine cytomegalovirus disrupts established transplantation tolerance and causes recipient allo-sensitization. Am J Transplant. 2021;21(2):515–524. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Dangi A, Husain I, Jordan CZ, et al. Conversion of CD73hiFR4hi anergic T cells to IFN-γ-producing effector cells disrupts established immune tolerance. J Clin Invest. 2023;133(5). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Chen L, Wang T, Zhou P, et al. TLR engagement prevents transplantation tolerance. Am J Transplant. 2006;6(10):2282–2291. [DOI] [PubMed] [Google Scholar]
  • 79.Ezzelarab MB, Cooper DK, Thomson AW. Cell-based immunosuppression in kidney transplantation: the value of non-human primate studies. Kidney Int. 2015;88(5):1196–1197. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Kirk AD. Transplantation tolerance: a look at the nonhuman primate literature in the light of modern tolerance theories. Crit Rev Immunol. 1999;19(5–6):349–388. [PubMed] [Google Scholar]
  • 81.Lei J, Kim JI, Shi S, et al. Pilot study evaluating regulatory T cell-promoting immunosuppression and nonimmunogenic donor antigen delivery in a nonhuman primate islet allotransplantation model. Am J Transplant. 2015;15(10):2739–2749. [DOI] [PubMed] [Google Scholar]
  • 82.Thomson AW, Turnquist HR, Raimondi G. Immunoregulatory functions of mTOR inhibition. Nat Rev Immunol. 2009;9(5):324–337. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Bestard O, Cruzado JM, Grinyó JM. Inhibitors of the mammalian target of rapamycin and transplant tolerance. Transplantation. 2009;87(8 Suppl):S27–29. [DOI] [PubMed] [Google Scholar]
  • 84.Turnquist HR, Raimondi G, Zahorchak AF, et al. Rapamycin-conditioned dendritic cells are poor stimulators of allogeneic CD4+ T cells, but enrich for antigen-specific Foxp3+ T regulatory cells and promote organ transplant tolerance. J Immunol. 2007;178(11):7018–7031. [DOI] [PubMed] [Google Scholar]
  • 85.Dangi A, Yu S, Lee FT, et al. Donor apoptotic cell-based therapy for effective inhibition of donor-specific memory T and B cells to promote long-term allograft survival in allosensitized recipients. Am J Transplant. 2020;20(10):2728–2739. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Goldstein S, Reddy P. Tolerance without toxicity? α1-antitrypsin as a novel alternative to immunosuppression. Expert Rev Clin Immunol. 2012;8(5):397–399. [DOI] [PubMed] [Google Scholar]
  • 87.Chen G, Li J, Chen L, et al. ECDI-fixed allogeneic splenocytes combined with α1-antitrypsin prolong survival of rat renal allografts. Int Immunopharmacol. 2015;26(1):43–49. [DOI] [PubMed] [Google Scholar]
  • 88.Xingqiang L, Fen N, Zhongpeng Y, et al. Ethylene carbodiimide-fixed donor splenocytes combined with cordycepin induce long-term protection to mice cardiac allografts. Transpl Immunol. 2019;56:101196. [DOI] [PubMed] [Google Scholar]
  • 89.Gao W, Lu Y, El Essawy B, et al. Contrasting effects of cyclosporine and rapamycin in de novo generation of alloantigen-specific regulatory T cells. Am J Transplant. 2007;7(7):1722–1732. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Pêche H, Renaudin K, Beriou G, et al. Induction of tolerance by exosomes and short-term immunosuppression in a fully MHC-mismatched rat cardiac allograft model. Am J Transplant. 2006;6(7):1541–1550. [DOI] [PubMed] [Google Scholar]
  • 91.Ma B, Yang JY, Song WJ, et al. Combining exosomes derived from immature DCs with donor antigen-specific Treg cells induces tolerance in a rat liver allograft model. Sci Rep. 2016;6:32971. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Shah S, Daneshmandi S, Hughes KR, et al. Optimizing PLG nanoparticle-peptide delivery platforms for transplantation tolerance using an allogeneic skin transplant model. Biomaterials. 2019;210:70–82. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Maldonado RA, LaMothe RA, Ferrari JD, et al. Polymeric synthetic nanoparticles for the induction of antigen-specific immunological tolerance. Proc Natl Acad Sci U S A. 2015;112(2):E156–165. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Lau CYJ, Benne N, Lou B, et al. Tuning surface charges of peptide nanofibers for induction of antigen-specific immune tolerance: an introductory study. J Pharm Sci. 2022;111(4):1004–1011. [DOI] [PubMed] [Google Scholar]
  • 95.Stead SO, Kireta S, McInnes SJP, et al. Murine and non-human primate dendritic cell targeting nanoparticles for in vivo generation of regulatory T-cells. ACS Nano. 2018;12(7):6637–6647. [DOI] [PubMed] [Google Scholar]
  • 96.Stead SO, McInnes SJP, Kireta S, et al. Manipulating human dendritic cell phenotype and function with targeted porous silicon nanoparticles. Biomaterials. 2018;155:92–102. [DOI] [PubMed] [Google Scholar]
  • 97.Braza MS, van Leent MMT, Lameijer M, et al. Inhibiting inflammation with myeloid cell-specific nanobiologics promotes organ transplant acceptance. Immunity. 2018;49(5):819–828.e816. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.van Leent MMT, Meerwaldt AE, Berchouchi A, et al. A modular approach toward producing nanotherapeutics targeting the innate immune system. Sci Adv. 2021;7(10). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Figdor CG, van Kooyk Y, Adema GJ. C-type lectin receptors on dendritic cells and Langerhans cells. Nat Rev Immunol. 2002;2(2):77–84. [DOI] [PubMed] [Google Scholar]
  • 100.Idoyaga J, Fiorese C, Zbytnuik L, et al. Specialized role of migratory dendritic cells in peripheral tolerance induction. J Clin Invest. 2013;123(2):844–854. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.Loschko J, Heink S, Hackl D, et al. Antigen targeting to plasmacytoid dendritic cells via Siglec-H inhibits Th cell-dependent autoimmunity. J Immunol. 2011;187(12):6346–6356. [DOI] [PubMed] [Google Scholar]
  • 102.Mathiesen CBK, Carlsson MC, Brand S, et al. Genetically engineered cell factories produce glycoengineered vaccines that target antigen-presenting cells and reduce antigen-specific T-cell reactivity. J Allergy Clin Immunol. 2018;142(6):1983–1987. [DOI] [PubMed] [Google Scholar]
  • 103.Sykes M, Chandran S, Kawai T, et al. Meeting Report: The Fifth International Samuel Strober Workshop on Clinical Immune Tolerance. Transplantation. 2023;107(3):564–569. [DOI] [PubMed] [Google Scholar]
  • 104.Hutchinson JA, Brem-Exner BG, Riquelme P, et al. A cell-based approach to the minimization of immunosuppression in renal transplantation. Transpl Int. 2008;21(8):742–754. [DOI] [PubMed] [Google Scholar]
  • 105.Hutchinson JA, Riquelme P, Brem-Exner BG, et al. Transplant acceptance-inducing cells as an immune-conditioning therapy in renal transplantation. Transpl Int. 2008;21(8):728–741. [DOI] [PubMed] [Google Scholar]
  • 106.Mevorach D, Zuckerman T, Reiner I, et al. Single infusion of donor mononuclear early apoptotic cells as prophylaxis for graft-versus-host disease in myeloablative HLA-matched allogeneic bone marrow transplantation: a phase I/IIa clinical trial. Biol Blood Marrow Transplant. 2014;20(1):58–65. [DOI] [PubMed] [Google Scholar]
  • 107.Li M, Zhang X, Zheng X, et al. Tolerogenic dendritic cells transferring hyporesponsiveness and synergizing T regulatory cells in transplant tolerance. Int Immunol. 2008;20(2):285–293. [DOI] [PubMed] [Google Scholar]
  • 108.Pêche H, Trinité B, Martinet B, et al. Prolongation of heart allograft survival by immature dendritic cells generated from recipient type bone marrow progenitors. Am J Transplant. 2005;5(2):255–267. [DOI] [PubMed] [Google Scholar]
  • 109.Bryant J, Hlavaty KA, Zhang X, et al. Nanoparticle delivery of donor antigens for transplant tolerance in allogeneic islet transplantation. Biomaterials. 2014;35(31):8887–8894. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110.Luo X, Du G, Long Y, et al. Programmed death ligand-1-overexpressing donor exosomes mediate donor-specific immunosuppression by delivering co-inhibitory signals to donor-specific T cells. Adv Healthc Mater. 2023:12(25):e2300670. [DOI] [PubMed] [Google Scholar]
  • 111.Pêche H, Heslan M, Usal C, et al. Presentation of donor major histocompatibility complex antigens by bone marrow dendritic cell-derived exosomes modulates allograft rejection. Transplantation. 2003;76(10):1503–1510. [DOI] [PubMed] [Google Scholar]

RESOURCES