Hematopoietic stem cell transplantation is the only curative treatment for many malignant hematologic diseases, with an often critical graft-versus-leukemia effect. Despite peritransplant prophylaxis, GVHD remains a significant cause of posthematopoietic stem cell transplantation morbidity and mortality. Traditional therapies have targeted T cells, yet immunostimulatory dendritic cells (DCs) are critical in the pathogenesis of GVHD. Furthermore, DCs also have tolerogenic properties. Monitoring of DC characteristics may be predictive of outcome, and therapies that target DCs are innovative and promising. DCs may be targeted in vivo or tolerogenic (tol) DCs may be generated in vitro and given in the peritransplant period. Other cellular therapies, notably regulatory T cells (Treg) and mesenchymal stem cells, mediate important effects through DCs and show promise for the prevention and treatment of GVHD in early human studies. Therapies are likely to be more effective if they have synergistic effects or target both DCs and T cells in vivo, such as tolDCs or Treg. Given the effectiveness of tolDCs in experimental models of GVHD and their safety in early human studies for type 1 diabetes, it is crucial that tolDCs be investigated in the prevention and treatment of human GVHD while ensuring conservation of graft-versus-leukemia effects.

Hematopoietic stem cell transplantation (HSCT) remains the only curative therapy for many high-risk malignant hematologic diseases, as well as numerous life-threatening genetic and hematologic disorders. However, despite peritransplant prophylaxis, HSCT is frequently complicated by GVHD, which leads to significant morbidity and mortality. The risk of GVHD limits the broader application of HSCT where it has the potential to cure autoimmune diseases, facilitate transplant tolerance, and correct immunologic deficiencies, including HIV infection.1  Conventional immunosuppressants remain the mainstay of treatment for GVHD, yet they frequently fail and carry a significant risk for infection.2,3  It is therefore of significant interest to identify new, effective, and safe prophylactic and therapeutic approaches, particularly those that maintain the critical graft-versus-leukemia (GVL) effect of HSCT. In this review, we consider advances that have been made in understanding the role of dendritic cells (DCs) in GVHD and address the challenge of monitoring, targeting, and exploiting these cells to improve therapeutic outcomes.

Our understanding of the pathogenesis of GVHD has advanced significantly over the past 45 years, since Billingham proposed that GVHD is the result of immunocompetent donor cells recognizing recipient antigens (Ags) in an immunocompromised host unable to reject donor cells.4  The principal immunocompetent donor effector cells are T cells, and the vigor of the immune response is driven by differences in MHC and minor histocompatibility antigens (miHA). Furthermore, the crucial role of Ag-presenting cells (APCs), in particular uniquely well-equipped donor and recipient DCs, has begun to be elucidated, not only in GVHD, but also in the GVL effect of HSCT.

DCs are rare, heterogeneous bone marrow (BM)–derived professional APCs, first characterized in mouse spleen by Steinman and Cohn,5  that are distributed ubiquitously in blood, lymphoid, and peripheral tissues, especially at portals of entry. They arise from hematopoietic stem cells through specialized progenitor subsets6  and are important in innate and adaptive immune function and in determining the balance between immunity and tolerance. In the normal steady state, DCs reside in “immature” or “semimature” states in the periphery where they constantly take up and process self-Ags and maintain self-tolerance.7  Immunostimulatory DCs have undergone maturation after recognition of exogenous and endogenous alarmins/danger signals by Toll-like receptors (TLRs).8  These signals include pathogen-associated molecular patterns in the form of microbial products and danger-associated molecular patterns, such as products of damaged or dying cells (eg, high-mobility group protein B1 or DNA). DCs are also matured by CD40 ligation and by proinflammatory cytokines that can induce DC maturation ex vivo, independent of CD40 ligation. Maturation is associated with up-regulation of cell surface MHC gene products, costimulatory molecules (CD40, CD80, and CD86, in addition to CD83 in humans), and appropriate chemokine receptors (in particular CCR7) that enhance the ability of DCs to home to secondary lymphoid tissue. Therein they present Ag to Ag-specific T cells and induce T-cell activation/proliferation. In turn, activated T cells drive DCs toward terminal maturation. These aspects of DC immunobiology have been reviewed in detail.9,,12 

DCs develop from HSCs in the BM and are derived from both myeloid and lymphoid progenitors (Figure 1).13,,16  This has been demonstrated in both mouse and human studies, in which all DC subsets can be generated from either a common lymphoid progenitor or common myeloid progenitor.6,17,19  The hematopoietic growth factor fms-like tyrosine kinase 3 ligand (Flt3L) plays a central role in steady-state DC development; this is evidenced by the majority of DC precursors being Flt3+ (CD135+) and culture with Flt3L resulting in all major DC subsets.6,17,20,21  GM-CSF is also important in DC hematopoiesis, as it gives rise to DCs from monocytes and early progenitors in the absence of intact Flt3L signaling and produces DCs under inflammatory conditions.6,17  Monocyte/macrophage M-CSF is also a DC poietin and can drive DC generation in mice independently of Flt3L.22 

Figure 1

DC hematopoiesis and subsets. (A) All identified DC subsets can be generated from either a common myeloid progenitor (CMP) or common lymphoid progenitor (CLP) depending on the cytokines and growth factors present. DCs can be broadly categorized as cDCs or precursor DCs. pDCs are understood to be a subset of precursor DCs that have plasma cell morphology, an immature phenotype, and secrete type I IFN after activation. Monocyte-derived DCs or “inflammatory DCs” are similar to cDCs in form and function and correlate with in vitro GM-CSF-generated DCs. cDCs can be categorized as lymphoid tissue resident and migratory DCs. DCs were categorized previously as lymphoid or myeloid (mDCs) based on the hypothesis that each had separate progenitors, a convention that has persisted in the experimental and clinical evaluation of DC subsets. Professional illustration by Alice Y. Chen. (B) In mice, pDCs are identified as CD11cloCD11b Siglec-H+PDCA-1+, whereas in humans, they are linMHC II+CD11cCD123(IL-3Rα)+BDCA2(CD303)+. Mouse mDCs are identified as CD11c+CD11b+B220 (CD45R) NK1.1, whereas human mDCs are lin MHCII+CD11c+CD123BDCA1(CD1b/c)+. Other phenotypic differences between mouse and human DC precursors are also listed in the table. HSC indicates hematopoietic stem cells; MPP, multipotent progenitor; CMP, common myeloid progenitor; CLP, common lymphoid progenitor; pDC, plasmacytoid DC; mDC, myeloid DC; LN, lymph node; and LP, lamina propria.

Figure 1

DC hematopoiesis and subsets. (A) All identified DC subsets can be generated from either a common myeloid progenitor (CMP) or common lymphoid progenitor (CLP) depending on the cytokines and growth factors present. DCs can be broadly categorized as cDCs or precursor DCs. pDCs are understood to be a subset of precursor DCs that have plasma cell morphology, an immature phenotype, and secrete type I IFN after activation. Monocyte-derived DCs or “inflammatory DCs” are similar to cDCs in form and function and correlate with in vitro GM-CSF-generated DCs. cDCs can be categorized as lymphoid tissue resident and migratory DCs. DCs were categorized previously as lymphoid or myeloid (mDCs) based on the hypothesis that each had separate progenitors, a convention that has persisted in the experimental and clinical evaluation of DC subsets. Professional illustration by Alice Y. Chen. (B) In mice, pDCs are identified as CD11cloCD11b Siglec-H+PDCA-1+, whereas in humans, they are linMHC II+CD11cCD123(IL-3Rα)+BDCA2(CD303)+. Mouse mDCs are identified as CD11c+CD11b+B220 (CD45R) NK1.1, whereas human mDCs are lin MHCII+CD11c+CD123BDCA1(CD1b/c)+. Other phenotypic differences between mouse and human DC precursors are also listed in the table. HSC indicates hematopoietic stem cells; MPP, multipotent progenitor; CMP, common myeloid progenitor; CLP, common lymphoid progenitor; pDC, plasmacytoid DC; mDC, myeloid DC; LN, lymph node; and LP, lamina propria.

Close modal

Overview of subsets

DCs can be broadly categorized as conventional DCs (cDCs) and precursor DCs (Figure 1).17  In the steady state, cDCs exhibit typical DC features (eg, cytoplasmic dendrites) and function (eg, Ag uptake, processing, and presentation).17  cDCs can be subdivided into migratory DCs, such as skin epidermal Langerhans cells (LCs) and dermal DCs, which present Ag in lymph nodes after its uptake in peripheral tissue, and resident DCs, which take up and present Ag within a lymphoid organ, such as splenic or thymic DCs (Table 1).6,17  Resident DCs can be further categorized in the mouse as CD8α, which is the predominant splenic population, and CD8α+, which is the major thymic population.6,17  CD8α+ DCs are involved in Ag cross-presentation and show high IL-12 secretion.6,17  Thymic cDCs primarily present self-Ag and are important in self-tolerance through T cell–negative selection and the production of naturally occurring regulatory T cells (Treg).23,25  Plasmacytoid DCs (pDCs) are a subset of precursor DCs which have an immature phenotype in the steady-state and plasma cell morphology (eg, lack dendrites).17  On activation, pDCs closely resemble cDCs in form and function.17  Monocyte-derived DCs, or “inflammatory DCs,” are similar to cDCs in form and function and correlate with in vitro GM-CSF-generated DCs.17 

Table 1

Phenotype and function of mouse conventional DC subsets

OrganPhenotypeLocationFunction and characteristics
Lymphoid-tissue resident DCs 
    Spleen CD8α+CD205+ T-cell area and marginal zone Uptake and cross-presentation of Ag from apoptotic cells on MHC class I 
 CD8α33D1+ Red pulp and bridging channels Predominant population; uptake and presentation of Ag on MHC class II 
    Thymus CD8α+ Medulla Predominant population; cross-presentation of Ag; self-tolerance 
 CD8α Cortex, medulla, corticomedullary junction Self-tolerance 
    LN CD8α+ Cortex Viral and self-Ag immunity; cross-presentation of Ag 
 CD8α Cortex Unknown 
Migratory DCs 
    Skin Langerin+ LC Epidermis Self-renew in situ, radiation resistant 
 CD103+CD11blo+ Langerin+ Dermis Viral and self-Ag immunity; cross-presentation of Ag 
 CD103CD11bhi− Langerin Dermis Unknown 
    Intestine CD103+CD11blo Peyer patches Unknown 
 CD103+CD11b+ Lamina propria Transfer pathogenic bacteria from gut to mesenteric LN 
 CD103CD11bhi Lamina propria Transport Ag to mesenteric LN from gut lumen 
    LN CD11cintCD40hiMHCIIhiCCR7+ Cortex Transport Ag to LN from periphery 
OrganPhenotypeLocationFunction and characteristics
Lymphoid-tissue resident DCs 
    Spleen CD8α+CD205+ T-cell area and marginal zone Uptake and cross-presentation of Ag from apoptotic cells on MHC class I 
 CD8α33D1+ Red pulp and bridging channels Predominant population; uptake and presentation of Ag on MHC class II 
    Thymus CD8α+ Medulla Predominant population; cross-presentation of Ag; self-tolerance 
 CD8α Cortex, medulla, corticomedullary junction Self-tolerance 
    LN CD8α+ Cortex Viral and self-Ag immunity; cross-presentation of Ag 
 CD8α Cortex Unknown 
Migratory DCs 
    Skin Langerin+ LC Epidermis Self-renew in situ, radiation resistant 
 CD103+CD11blo+ Langerin+ Dermis Viral and self-Ag immunity; cross-presentation of Ag 
 CD103CD11bhi− Langerin Dermis Unknown 
    Intestine CD103+CD11blo Peyer patches Unknown 
 CD103+CD11b+ Lamina propria Transfer pathogenic bacteria from gut to mesenteric LN 
 CD103CD11bhi Lamina propria Transport Ag to mesenteric LN from gut lumen 
    LN CD11cintCD40hiMHCIIhiCCR7+ Cortex Transport Ag to LN from periphery 

LN indicates lymph node.

Function of DC subsets

DC subsets differ in their immune functions, which has important implications for HSCT. Under steady-state conditions, human pDCs display lower levels of MHC and costimulatory molecules compared with conventional myeloid DCs (mDCs).26  In addition, because their Ag processing and loading ability are less efficient, pDCs stimulate T cells less effectively than mDCs.26  After their activation via TLR, pDCs secrete high levels of type 1 IFN and stimulate CD4+ and CD8+ T cells.11  This is in contrast to activated mDCs, which secrete IL-12 and promote T-helper type-1 (Th1) cell differentiation and CD8+ cytotoxic T lymphocyte (CTL) responses.26,27  pDCs have intrinsic tolerogenic properties; in the steady state, human thymic pDCs induce Treg, whereas liver and airway pDCs regulate oral and mucosal tolerance, respectively. pDCs have also been implicated in the regulation of disease activity in experimental models of autoimmunity and shown to exert disease-suppressing ability.28  This may be important after HSCT in terms of donor BM engraftment (tolerance), as well as for chronic GVHD (cGVHD), which has clinical features that overlap with autoimmune disease. Epidermal LCs may be immunostimulatory or tolerogenic, depending on their state of maturity, inciting immunogen, and the cytokine environment.29 

Mouse versus human DC subsets

DC subsets have been well characterized, especially in mice, and also in humans and other species (Figure 1).13,14,30,32  Most human work has been conducted in vitro; thus, knowledge of the in vivo function and development of human DC subsets is lacking. Direct comparisons between mouse and human DC subsets can be problematic because of phenotypic differences between the species (eg, multiple human DC subsets are capable of Ag cross-presentation and display high IL-12 secretion, and are thus comparable with the mouse CD8α+ DC subset).33  Likewise, flow cytometric characterization of DC subsets has been refined over time, and the evolution of “standard” DC markers makes comparisons between present and past studies difficult. Currently, in mice, pDCs are identified as CD11cloCD11b sialic acid binding immunoglobulin-like lectin H (Siglec-H)+PDCA-1+, whereas in humans, they are linMHC II+CD11cCD123(IL-3Rα)+BDCA2(CD303)+. Mouse mDCs are identified as CD11c+CD11b+B220 (CD45R) NK1.1, whereas human mDCs are linMHCII+CD11c+CD123BDCA1 (CD1b/c)+.

Although characterization of human DC subsets has been more difficult because of both their rarity and difficulty of access, DC subsets in the skin have been well characterized, although recent studies have begun to elucidate subsets in the blood and other organs. The epidermis contains Langerin+ LCs, whereas the dermis contains CD1a+ and CD14+ DCs, the former of which has an unknown function.34  CD14+ DCs are involved in B-cell differentiation both by activation of naive B cells to plasma cells35,36  and induction of naive CD4+ T cells to T follicular helper cells.37  Compared with other human skin DC subsets, LCs are potent activators of Ag-specific CD8+ T cells, which may be explained in part by their production of IL-15.34,35  Conventional DC subsets appear to be comparable in human blood and spleen in the steady state, with 3 major populations described, all of which are linCD11c+HLA-DR+, and unlike mouse subsets, cannot be further distinguished by CD8α.33  BDCA3(CD141)+ DCs are thought to be the human equivalent of mouse CD8α+ DCs, based on cell adhesion molecule 1 expression and their ability to cross-present Ag and secrete high IL-12, although more recent studies indicate that other human cDC subsets share these abilities.33,38  BDCA1(CD1b/c)+ DCs may be comparable with mouse CD8α DCs, whereas CD16+ DCs have been termed inflammatory monocytes, based on their high TNF-α and low IL-10 expression.33 

In addition to their capacity to stimulate innate and adaptive immunity, DCs can induce and maintain tolerance.28,39,40  Tolerogenic (tol) DCs present Ag to T cells but lack adequate costimulatory ability, deliver inhibitory signals (eg, via the programmed death [PD] pathway), and produce tolerance-promoting cytokines (IL-10).39  TolDCs do not support Ag-specific T-cell activation and proliferation but instead facilitate T-cell anergy/apoptosis and/or the generation or expansion of Treg.39  Importantly, bidirectional feedback between tolDCs and Treg has been demonstrated in humans and mice, whereby tolDCs promote the generation of Treg from naive T cells and Treg generate tolDCs from DC progenitors.41  Regulation of immunosuppressive tryptophan catabolism in DCs via activation of indoleamine 2,3-dioxygenase (IDO) may be an important mechanism of action of Treg42  and may underlie transplant tolerance in vivo. The close relationship between Treg and DCs is illustrated by the observation that increases in DCs lead to increases in Treg, whereas constitutive absence of DCs leads to fatal autoimmunity.43,45 

Mouse studies have demonstrated that CD4+ T cell–dependent (MHC-mismatched) acute GVHD (aGHVD) can be induced by either host or donor APCs, whereas host APCs are required for the initiation of CD8+ T cell–dependent (MHC-matched, multiple miHA-mismatched) aGVHD and donor APC amplify the process.46,48  Additional studies have tried to further characterize the contribution of different APC populations to the development of aGVHD. Whereas earlier mouse studies implicated host LCs in the pathogenesis of skin GVHD, more recent experiments using mice deficient in LCs question the relevance of LCs in the development of aGVHD.49,50 

Less is known about the role of DCs in cGVHD because of variability in clinical presentation (de novo cGVHD vs cGVHD evolving from aGVHD) and the lack of relevant mouse models. Both host and donor APCs have been implicated, but with target organ specificity; skin cGVHD can be induced by either donor or host APCs, whereas donor APCs are dominant in intestinal cGVHD.51,52  Thymic independent and dependent pathways likely contribute to cGVHD. Autoreactive CD4+ T cells have been implicated in cGVHD, and cGVHD occurs in patients with little thymic function or in those with intact thymic negative selection.52,53  Mouse studies have implicated engrafted donor anti–host CD4+ T cells in the evolution of aGVHD to cGVHD, whereby donor CD4+ T cells are generated in the milieu of CD8+ T cell–mediated aGVHD thymic damage, likely due to failure of thymic DCs to delete autoreactive CD4+ T cells.54  This has important implications, both in the pathogenesis of cGVHD, but also in its prevention, as keratinocyte growth factor prevents cGVHD likely because of thymic protection.54 

The development of GVHD, particularly aGVHD, has been divided traditionally into 3 phases (Figure 2). Phase 1 involves activation of APCs, particularly DCs, by cytokines released after recipient tissue damage. These DCs present acquired and processed Ag to T cells, which, in combination with simultaneous costimulation, leads to the second phase, donor T-cell activation. Mouse studies suggest that donor T-cell activation in GVHD requires costimulation via B7 family molecules (CD80/86)/CD28 and B7H/inducible costimulator and is inhibited by B7/CTL Ag (CTLA)–4 and PD-L1/PD interactions.55,56  After HSCT, DCs can present host Ag to donor T cells, either directly or indirectly. In the direct pathway, donor T cells are stimulated by allogeneic MHC or miHA molecules (in the more common MHC-matched setting) present on host APCs, whereas the indirect pathway involves presentation of acquired host Ags by engrafted donor APCs, particularly CD11c+ DCs.57  Mouse models have shown that the indirect pathway or “cross-presentation” of host Ag does not initiate aGVHD but that direct presentation by host DCs resistant to conditioning is required.46,48  Wang et al expanded knowledge of cross-presentation in an experimental model, demonstrating that donor APCs are activated by donor CD4+ T cells (initially activated by host APCs) dependent on CD40L and type I IFN, then cross-present acquired host hematopoietic and nonhematopoietic transmembrane proteins to donor CD8+ T cells.57  Using mAbs and/or transgenic/knockout donor mice, Markey et al examined the role of donor APC subsets and showed that donor cDCs are critical for cross-presentation of alloAg immediately after HSCT.58  Donor T-cell activation leads to the third phase, in which cytokines and cellular effectors, particularly CTLs, NK cells, and macrophages, mediate target cell injury and apoptosis.57 

Figure 2

Role of DCs in the pathogenesis of GVHD. (A) Recipient pretransplant conditioning results in target organ tissue damage, leading to the so-called “cytokine storm,” a progressive amplification of proinflammatory cytokine production and immune activation as inflammatory cytokines feed forward unabated. IL-1β, IL-6, and TNF-α are particularly implicated in this process. In addition to proinflammatory cytokines, conditioning-released damage-associated molecular patterns (DAMPS) and translocation of lipopolysaccharide in the intestine also lead to the activation of host and subsequently donor DCs, including epidermal LCs and dermal DCs in the skin. Mature DCs up-regulate MHC, costimulatory, and intercellular adhesion molecule expression. (B) DCs present host allo-Ag to donor T cells. Host DCs resistant to conditioning present alloAg via the direct pathway, whereas transplanted donor DCs present processed alloAg peptides on MHC syngeneic with donor T cells via the indirect pathway. Donor T-cell activation requires Ag presentation via MHC molecules to the T-cell Ag receptor (TCR), as well as stimulation via various costimulatory molecules. This interaction results in T-cell activation, proliferation, differentiation (Th1, Th2), migration to GVHD target organs, and secretion of various chemokines and cytokines, importantly IFN-γ and IL-2. (C) Cellular and inflammatory effectors lead to target organ tissue damage. CTLs mediate target cell apoptosis via interactions between TNF and TNF receptors, TRAIL (TNF-related apoptosis-inducing ligand)/TRAIL-R and Fas (CD95)/FasL interactions and release of cytotoxic mediators (perforin and granzyme). Recruited macrophages release TNF-α, IL-1, and NO, which also damage target cells. RT indicates radiation therapy; ICAM-1, intercellular adhesion molecule-1; VCAM-1, vascular cell adhesion molecule-1; ICOS, inducible costimulator; and NO, nitric oxide. (A-C) Professional illustrations by Alice Y. Chen.

Figure 2

Role of DCs in the pathogenesis of GVHD. (A) Recipient pretransplant conditioning results in target organ tissue damage, leading to the so-called “cytokine storm,” a progressive amplification of proinflammatory cytokine production and immune activation as inflammatory cytokines feed forward unabated. IL-1β, IL-6, and TNF-α are particularly implicated in this process. In addition to proinflammatory cytokines, conditioning-released damage-associated molecular patterns (DAMPS) and translocation of lipopolysaccharide in the intestine also lead to the activation of host and subsequently donor DCs, including epidermal LCs and dermal DCs in the skin. Mature DCs up-regulate MHC, costimulatory, and intercellular adhesion molecule expression. (B) DCs present host allo-Ag to donor T cells. Host DCs resistant to conditioning present alloAg via the direct pathway, whereas transplanted donor DCs present processed alloAg peptides on MHC syngeneic with donor T cells via the indirect pathway. Donor T-cell activation requires Ag presentation via MHC molecules to the T-cell Ag receptor (TCR), as well as stimulation via various costimulatory molecules. This interaction results in T-cell activation, proliferation, differentiation (Th1, Th2), migration to GVHD target organs, and secretion of various chemokines and cytokines, importantly IFN-γ and IL-2. (C) Cellular and inflammatory effectors lead to target organ tissue damage. CTLs mediate target cell apoptosis via interactions between TNF and TNF receptors, TRAIL (TNF-related apoptosis-inducing ligand)/TRAIL-R and Fas (CD95)/FasL interactions and release of cytotoxic mediators (perforin and granzyme). Recruited macrophages release TNF-α, IL-1, and NO, which also damage target cells. RT indicates radiation therapy; ICAM-1, intercellular adhesion molecule-1; VCAM-1, vascular cell adhesion molecule-1; ICOS, inducible costimulator; and NO, nitric oxide. (A-C) Professional illustrations by Alice Y. Chen.

Close modal

Host DCs are required for full GVL effects, although donor APCs can initiate GVL activity when low levels of tumor are present.48,59,60  Li and Waller found that depletion of donor BM CD11b+ myeloid cells in an experimental model enhanced survival of recipients with tumor61 ; more recently, they reported that, conversely, addition of CD11b cells, which were primarily precursor pDCs, augmented GVL without concomitant increase in GVHD.62  Clinical studies have demonstrated that increased graft pDC content is associated with relapse and decreased patient overall survival (OS).63  Low CD11c+ DCs, but not CD123+ DCs, in peripheral blood (PB) at the time of engraftment have also been associated with death and relapse.64  New insights concerning the role of both donor and recipient DC subsets in GVL, including graft precursor pDC content, and the impact of pretransplant manipulation of these subsets are clearly needed.

The role of DCs in GVL after donor leukocyte infusion has also been examined. In murine and clinical studies, GVL effects can be seen after donor leukocyte infusion without GVHD.65  The presence of host APCs and allo-MHC class I has been shown to be critical for GVL effects in mixed chimeras created in the fully MHC-mismatched setting, although results are not as clear in the MHC-matched minor Ag mismatched and clinical settings.66,68  Furthermore, there is evidence that donor leukocyte infusion-induced GVL effects in mice are dependent on MHC alloAg, but not miHC or tumor-associated Ags, in a CD4+ and CD8+ T cell–dependent manner, and that MHC class II–expressing host APCs are required for maximal GVL activity.60  Unfortunately, although donor leukocyte infusion enhances the GVL effect, it is often complicated by GVHD.69,70 

Given the role of host tissue damage in the pathogenesis of GVHD, it was thought that reduced intensity conditioning (RIC) would lead to less GVHD. However, whereas RIC has reduced transplant-related mortality, the incidence of aGVHD, although delayed, remains unchanged. In mouse HSCT after RIC, Turner et al demonstrated that, although the onset of GVHD was delayed, it was equally severe.71  The authors hypothesized that delays in GVHD were the result of maintenance of absolute numbers of host DCs and decreased TNF-α production, promoting Treg responses.71  As donor chimerism increased, donor-activated DCs increased and elevated TNF-α led to decreased Treg and onset of delayed, yet equally severe, GVHD.71 

Conditioning regimens can differ in host irradiation; however, some human and mouse DCs are resistant to radiation, particularly dermal DCs and epidermal LCs.72,74  In an experimental model, total body irradiation led to complete depletion of splenic and BM pDCs after 24 hours, whereas mDCs were maintained, but in decreased numbers.75  In addition, total body irradiation is important in DC activation; studies in mice have shown that inflammation from irradiation is critical for pDC but not mDC activation.26 

HSCs may be obtained either directly from BM or from the PB after expansion with G-CSF. As reviewed by Korbling and Anderlini,76  despite a significant increase in donor T cells in PB stem cell grafts, there is surprisingly no increase in aGVHD, although there seems to be an increase in cGVHD.76  Numerous studies have documented increased graft pDCs with G-CSF mobilization, with potential implications for outcome of HSCT, including that these cells may favor Treg function.77,79  G-CSF treatment has also been associated with decreased proinflammatory IL-12 production. In a mouse model of PB stem cell transplantation, G-CSF treatment of donors rather than recipients significantly reduced levels of TNF-α, probably via decreased donor DC TNF-α and IL-12 production.80  mDC IL-12 production was also significantly decreased in pediatric HSCT recipients who received G-CSF after transplantation.81  These differences must be taken into account when interpreting clinical studies.

DC engraftment

Clinical studies have revealed an association between low total DC numbers at the time of engraftment and decreased patient OS, increased relapse, and increased aGVHD (summarized in Table 2).64  Neither host DC count pretransplant nor graft DC count was associated with death or relapse.64  Although neither was independently significant, lower mDC count at engraftment was associated with decreased survival, increased relapse, and increased incidence of aGVHD.64  Lower circulating pDC count correlated only with increases in aGVHD.64  Skin GVHD has been associated with decreased human LC engraftment.49,82  However, this is thought to be a secondary effect related to steroid treatment and GVHD effector cells, as experimental studies have shown that donor T cells promote donor LC engraftment.49,82  Prospective studies are indicated to determine whether low DC count at the time of engraftment can be used as a predictive tool for GVHD.

Table 2

HSCT outcome in relation to DC analyses

Patient populationDC measurementOutcomeReference
Allo-HSCT (n = 49) Total PB DC count at engraftment < 4.97 cells/μL Survival ↓ 64  
92% PBSCT; 73% MAC  Relapse and aGVHD ↑  
 Low PB mDCs (CD11c+) at engraftment Survival ↓  
  Relapse and aGVHD ↑  
 Low PB pDCs (CD123+) at engraftment aGVHD ↑  
 Graft DC or DC count before transplant No association with death/relapse  

 
Allo-HSCT (n = 30) Higher PB pDCs (BDCA-2+) > day 100 (donor) cGVHD ↑ 83  
63% PBSCT; 83% MAC    

 
Allo-HSCT (n = 24) Higher total PB host DCs day 100 Survival ↓ 84  
100% BM; 87% RIC  aGVHD and cGVHD (grade II-IV)↑  

 
Allo-HSCT (n = 40) Low total PB DC, mDC (CD11c+), and pDC (CD123hi) counts GVHD severity ↑ 85  
90% PBSCT; 52% RIC > 7.9% CMRF-44+ CD11c+ DCs aGVHD ↑ (sensitivity, 87.5%; specificity, 79.2%)  
 CD83+/CD86+ CD11c+ DCs No association with aGVHD  

 
Allo-HSCT (n = 69) Graft pDC (CD123+) > 2.3 × 106/kg Relapse ↑ 63  
100% PBSCT; 54% MAC  OS and EFS ↓  

 
Allo-HSCT (n = 54) Lower PB pDCs (lin/CD11c/ILT3+) 3 mo after HSCT (median 92 days) aGVHD (grade II-IV) ↑ 88  
93% PBSCT; 100% RIC  OS and NRM ↓  
  Late infections ↑  
  Death ↑  
Patient populationDC measurementOutcomeReference
Allo-HSCT (n = 49) Total PB DC count at engraftment < 4.97 cells/μL Survival ↓ 64  
92% PBSCT; 73% MAC  Relapse and aGVHD ↑  
 Low PB mDCs (CD11c+) at engraftment Survival ↓  
  Relapse and aGVHD ↑  
 Low PB pDCs (CD123+) at engraftment aGVHD ↑  
 Graft DC or DC count before transplant No association with death/relapse  

 
Allo-HSCT (n = 30) Higher PB pDCs (BDCA-2+) > day 100 (donor) cGVHD ↑ 83  
63% PBSCT; 83% MAC    

 
Allo-HSCT (n = 24) Higher total PB host DCs day 100 Survival ↓ 84  
100% BM; 87% RIC  aGVHD and cGVHD (grade II-IV)↑  

 
Allo-HSCT (n = 40) Low total PB DC, mDC (CD11c+), and pDC (CD123hi) counts GVHD severity ↑ 85  
90% PBSCT; 52% RIC > 7.9% CMRF-44+ CD11c+ DCs aGVHD ↑ (sensitivity, 87.5%; specificity, 79.2%)  
 CD83+/CD86+ CD11c+ DCs No association with aGVHD  

 
Allo-HSCT (n = 69) Graft pDC (CD123+) > 2.3 × 106/kg Relapse ↑ 63  
100% PBSCT; 54% MAC  OS and EFS ↓  

 
Allo-HSCT (n = 54) Lower PB pDCs (lin/CD11c/ILT3+) 3 mo after HSCT (median 92 days) aGVHD (grade II-IV) ↑ 88  
93% PBSCT; 100% RIC  OS and NRM ↓  
  Late infections ↑  
  Death ↑  

PBSCT indicates peripheral blood stem cell transplant; EFS, event-free survival; ILT3, immunoglobulin-like transcript 3; and NRM, nonrelapse mortality.

DC chimerism

There are conflicting data on DC chimerism after HSCT. Early human studies demonstrated an association between full donor chimerism and cGVHD compared with mixed chimerism in some control patients without cGVHD.83  Chan et al assessed DC chimerism 100 days after transplantation and found that host DC persistence correlated with severe aGVHD and cGVHD.84  There were significant differences between the 2 studies, however, with the latter involving primarily RIC regimens and samples differentiated in vitro and analyzed by DNA PCR banding, rather than by conventional flow cytometry.84  Given the differential effects of myeloablative conditioning (MAC) versus RIC, further studies on DC chimerism in both populations are warranted to resolve the impact of DC chimerism on development of aGVHD and cGVHD.

DC activation status

The activation status of DCs is likely important in and potentially predictive of GVHD. Lau et al85  examined expression of CMRF-44, a cell surface marker that is expressed early during the activation and maturation of human mDCs, but not on freshly isolated DCs from healthy controls. The incidence of circulating CMRF-44+CD11c+ DCs correlated with onset and severity of aGVHD and was found to be predictive when used as a screening test before the onset of GVHD.85  In the same study, cell surface expression of CD83 and CD86, both of which are increased on human DC activation and are important in T-cell costimulation, were not predictive of GVHD.85  Larger studies investigating the predictive role of these DC activation/maturation markers and their anti-inflammatory versus proinflammatory cytokine production, such as IL-10 or IL-12, respectively, should be performed. Analysis of donor versus host origin DC expression of these immunoregulatory molecules could enhance the insights from these further evaluations.

DC subsets

Before current immunophenotyping of DC subsets, Waller et al demonstrated an association between high BM graft presumptive pDC progenitors and decreased cGVHD,86  although the incidence of leukemic relapse was increased. A more recent clinical study did not find an association between G-CSF–mobilized PB graft pDC content and GVHD; however, it confirmed the increased incidence of relapse, as well as decreased OS and event-free survival.86  Because these studies used different stem cell sources, the difference in the incidence of GVHD may be accounted for, in part, by differences in cytokine release and/or DC activation status after G-CSF administration.86  The association between relapse and graft pDCs highlights the importance of preserving the GVL effect with any intervention to decrease GVHD.

Human DC subsets have also been examined in the posttransplant period. Reddy et al documented a dependent association between low CD11c+ DCs in PB at the time of engraftment and death, relapse and aGVHD64 ; low CD123+ DC count was associated with aGVHD only. More recent studies in patients after MAC and RIC found a significant correlation between pDC count and increased GVHD, as well as pDC and mDC count and increased GVHD severity.63,85,87  Low pDC count 3 months after RIC transplant was also associated with severe aGVHD, decreased OS, and increased nonrelapse mortality, notably from GVHD and late infections.88  As with other analyses, there have been conflicting reports, as an earlier study associated high pDC count with cGVHD, although this was at a median of 14.5 months after transplantation.83  Larger studies comparing DC subsets after various conditioning regimens may help elucidate the differences between studies.

DC subsets have been examined extensively in experimental GVHD. By adoptively transferring DCs into MHC class II–deficient recipient mice, both pDCs and cDCs were found to be sufficient to induce comparable donor CD4+ T cell–dependent GVHD, although pDCs required an inflammatory environment created by host irradiation for activation and donor T-cell priming.26  Thus, pDCs expressing alloAg were sufficient to prime alloreactive T cells and induce GVHD. Similar to human studies, low pDCs (depleted by 120G8 Ab to BM stromal cell Ag-2) in the BM graft led to increased aGVHD, whereas there was no association between GVHD and pDC count in G-CSF–mobilized grafts.75  The authors indicated that these latter pDCs were mature, which may account for the difference in incidence of GVHD. In addition, although cDC reconstitution did not differ between control and GVHD mice, pDC maturation was abrogated in GVHD.75  Interestingly, GVHD led to a suppressive precursor DC population that may contribute to immune paralysis after transplantation.75  These findings concerning the role of DC subsets provide important insight into potential strategies for tolerance induction in HSCT.

Many current therapies significantly affect DC phenotype and function.89  More precisely, calcineurin inhibitors (CNI; cyclosporine or tacrolimus) suppress Ag presentation, whereas glucocorticosteroids inhibit DC maturation, activation, and production of TNF-α, IL-1β, and IL-12 after stimulation.90,92  DCs generated in the presence of CNI or rapamycin (sirolimus; the serine-threonine kinase inhibitor of the mammalian target of rapamycin) have decreased costimulatory molecule expression and T-cell allostimulatory capacity.90,93  In addition, epidermal LCs exposed to glucocorticosteroids are phenotypically immature and expand Treg via TGF-β production.94 

Ab therapy directed against immune cells is used both in the prevention and treatment of GVHD. Polyclonal anti-thymocyte globulin (ATG) Ab has been used before transplantation for T-cell depletion for decades.95  However, as reviewed by Mohty,96  ATG has diverse immunologic effects, including its impact on DCs. ATG inhibits experimental DC Ag uptake and maturation, induces complement-mediated lysis of DCs, and decreases the capacity of DCs to stimulate allogeneic T cells.96,97  In humans, ATG decreases DC Ag uptake, PB mDCs and pDCs, and mDC IL-12 production and allogeneic T-cell proliferation.66,67 

Alemtuzumab (Campath-1H), a lymphocyte-depleting humanized anti-CD52 mAb, has been used for both GVHD prevention and treatment. As well as depleting donor T cells, alemtuzumab may also target host DCs.95  Although its effects on DCs are not well studied, alemtuzumab depletes human PB DCs in vivo but has few significant effects on LCs or dermal DCs, which only weakly express the epitope.98,99  Multiple mAbs against the IL-2 receptor (CD25) have shown efficacy in second-line treatment of GVHD.95  Although much of these effects have been attributed to direct binding to T cells, recent work using daclizumab (humanized anti-CD25 mAb) has shown that it potently inhibits Ag-specific T-cell activation by mature DCs.100,101 

Historically, T cells have been the primary target in GVHD, but given the important role of DCs in its pathogenesis, APCs also represent an important target. DCs may be manipulated using multiple approaches in vivo or in vitro, in the latter case for the production of tolDC vaccines with the ability to regulate immunity and suppress GVHD. Approaches being evaluated include the following.

Pharmacologic interventions

Histone deacetylase (HDAC) inhibitors, used clinically as anticancer drugs, reduce DC TLR-induced costimulatory molecule expression, proinflammatory cytokine release, and T-cell allostimulatory activity (summarized in Table 3). Further, they increase Treg number and function via increased IDO expression in a signal transducer and activator of T cells (STAT3)–dependent manner.102,103  HDAC inhibition decreases GVHD in several experimental models while preserving GVL.103,105  Clinical testing of HDAC inhibition using agents, such as suberoylanilide hydroxamic acid (SAHA; vorinostat), in conjunction with CNI for the prophylaxis of GVHD after RIC allogeneic HSCT is in progress.103,104 

Table 3

Impact of interventional strategies on DCs and outcome in experimental HSCT

MechanismTherapyGVHD model; treatmentEffect on DCs*Effect on GVHDReference
HDAC inhibition SAHA or ITF2357 MHC-mismatched BALB/c → B6; TNF-α, IL-12, and IL-6 secretion ↓ Survival ↑ 104  
  Host type DCs pretreated with HDAC inhibitor infused days −1, 0, and 2 CD40/CD80/CD86 expression ↓ Clinical score ↓  
   Allogeneic T-cell proliferation ↓ (in vitro/in vivo) Serum TNF-α ↓  
    Preserved GVL  

 
Proteasome inhibition Bortezomib MHC-mismatched BALB/c → B6; Response to maturation signals ↓ Survival ↑ with early treatment 108  
  Early (days 0-2) versus late (days 6-8) treatment MHC class II, CD40/CD80/CD83/CD86 expression ↓ Survival ↓ with late treatment  
   Apoptosis ↑ Preserved GVL  
   Allogeneic T-cell proliferation ↓   

 
NF-κB inhibition RelB−/− BM chimera recipients MHC-mismatched BALB/c → B6 CD11chi DCs (cDCs) ↓ Survival ↑ 109  
   CD40/CD80/CD86 expression unchanged Clinical score ↓  
   IL-12, IL-6, TNF-α secretion ↓ (after CD40L)   
   CD4+ T-cell proliferation (in vitro/vivo); cytokine secretion ↓ (in vitro)   
   No difference PDCA-1+ DCs (pDCs), Treg (in vivo)   
 RelB−/− BM chimera donor MHC-mismatched B6 → B6D2F1  Late (> 3 wks) survival ↑ and clinical score ↓  

 
Anti-CD83 Polyclonal Ab Hu SCID; day 0  Survival ↑ 110  
    Preserved engraftment and GVL  
MechanismTherapyGVHD model; treatmentEffect on DCs*Effect on GVHDReference
HDAC inhibition SAHA or ITF2357 MHC-mismatched BALB/c → B6; TNF-α, IL-12, and IL-6 secretion ↓ Survival ↑ 104  
  Host type DCs pretreated with HDAC inhibitor infused days −1, 0, and 2 CD40/CD80/CD86 expression ↓ Clinical score ↓  
   Allogeneic T-cell proliferation ↓ (in vitro/in vivo) Serum TNF-α ↓  
    Preserved GVL  

 
Proteasome inhibition Bortezomib MHC-mismatched BALB/c → B6; Response to maturation signals ↓ Survival ↑ with early treatment 108  
  Early (days 0-2) versus late (days 6-8) treatment MHC class II, CD40/CD80/CD83/CD86 expression ↓ Survival ↓ with late treatment  
   Apoptosis ↑ Preserved GVL  
   Allogeneic T-cell proliferation ↓   

 
NF-κB inhibition RelB−/− BM chimera recipients MHC-mismatched BALB/c → B6 CD11chi DCs (cDCs) ↓ Survival ↑ 109  
   CD40/CD80/CD86 expression unchanged Clinical score ↓  
   IL-12, IL-6, TNF-α secretion ↓ (after CD40L)   
   CD4+ T-cell proliferation (in vitro/vivo); cytokine secretion ↓ (in vitro)   
   No difference PDCA-1+ DCs (pDCs), Treg (in vivo)   
 RelB−/− BM chimera donor MHC-mismatched B6 → B6D2F1  Late (> 3 wks) survival ↑ and clinical score ↓  

 
Anti-CD83 Polyclonal Ab Hu SCID; day 0  Survival ↑ 110  
    Preserved engraftment and GVL  

Hu SCID indicates human severe-combined immunodeficiency.

*

In vitro unless otherwise indicated.

Proteasome inhibitors have been studied in cancer and autoimmunity and are thought to induce apoptosis by blocking the degradation of proapoptotic proteins. Bortezomib, approved for the treatment of multiple myeloma, is thought to block the activation and nuclear translocation of NF-κB, a transcription factor central to DC maturation and inflammatory responses. Thus, inhibition of DC NF-κB activation with bortezomib or other inhibitors is an attractive strategy for GVHD prevention.106  Immature DCs treated with bortezomib fail to up-regulate MHC class II and costimulatory molecules in response to maturation signals, have decreased T-cell allostimulatory capacity, and are more susceptible to apoptosis.106,107  In experimental HSCT, bortezomib attenuates aGVHD yet preserves GVL.106,107  Whereas early treatment after HSCT prevents mild aGVHD in mice, later treatment increases mortality significantly,108  which may reflect loss of early effects on immature DCs. Notably, histopathologic observations in later bortezomib treatment have implicated severe colonic damage in increased GVHD-dependent mortality.106 

RelB, an NF-κB family subunit, has been shown to be critical within both host and donor APCs for the induction and maintenance of experimental GVHD.109  RelB in APCs is required for differentiation of Th1 effectors, but not for expansion or function of donor Treg.109  Inhibition of nuclear RelB translocation, with RelB inhibitors targeted to DCs using Ab, thus appears to be an attractive strategy for therapy of GVHD.109  Although these studies confirm NF-κB in DCs as an important therapeutic target, they also urge caution when considering bortezomib for the treatment of established GVHD given that late (vs early) treatment in an experimental model significantly increased mortality.

Biologic interventions

Activated DCs may be targeted by mAbs against cell surface molecules, including CD83, which is up-regulated on DC maturation. There is recent evidence that anti-CD83 (polyclonal Ab) decreases T-cell proliferation induced by DCs while maintaining antiviral T cell memory.110  In an experimental model, anti-CD83 therapy prevented GVHD while preserving HSC engraftment and GVL.110  Costimulatory signal blockade also prevented experimental GVHD, with the most significant effect achieved by blocking inducible costimulator (using mAb) and CD28 (CD28−/− donor T cells) with intact CTLA-4 signaling.55  Further mechanistic and therapeutic studies of mAbs directed against activated DCs are clearly justified.

DCs can be manipulated in vitro to produce tolDCs or “negative DC vaccines” for control of alloimmunity or allograft rejection (summarized in Table 4). TolDCs may be produced under specific culture conditions, by pharmacologic modification, or by cell sorting. Early studies showed that immature DCs generated from BM cells in GM-CSF, and with weak allostimulatory T-cell capacity, could prolong organ allograft survival.111,112  Subsequent reports have verified and extended these findings to show that immature or maturation-resistant tolDCs can promote tolerance in experimental organ and HSCT39,113,114  while still protecting against leukemia relapse.114 

Table 4

Use of tolDC therapy to inhibit GVHD in mice

TolDCGVHD model; treatmentEffect on DCs (in vitro)Effect on GVHDReference
DC conditioning     
    Rapamycin MHC-mismatched (B6 → BALB/c) MHC class II, CD80/CD86 expression ↓ Survival ↑ 117  
 Host-type DCs day 0 Intact in vivo trafficking Histopathology ↓  
  Intact CCR5, CCR7, and CD62L   

 
    VIP MHC (B6 → BALB/c, BALB/c → B6) and miHA-mismatched (B6 → F1) Host-type DCs day 2 and/or day 5 Donor CD4+ T-cell Ag-specific response ↓ Induce Treg Survival ↑ (miHA > MHA; early > late for MHA) 118,
146  

 
    SAHA (HDAC inhibitor) MHC-mismatched (BALB/c → B6) CD40/CD80 expression ↓ Survival ↑ 104  
 Host-type DCs days −1, 0, and 2 TNF-α ↓   
  T-cell proliferation ↓   

 
    TGF-β, IL-10 + LPS (DCreg)* MHC-matched, miHA-mismatched cGVHD (B10.D2 → BALB/c) Induce anergy of Ag-specific T cells Cutaneous GVHD ↓ 129  
 Host-type DCs days 2, 9, and 16 or days 18, 25, and 32 vs short-course rapamycin TNF-α, IL-12p70, and IFN-γ ↓   
  Induce Treg   

 
Subset (no DC conditioning)     
    CD8α+ DCs MHC (BALB/c → B6) and miHA (C3H.SW → B6) mismatched T-cell proliferation ↓ Survival ↑ 124  
 Host-type DCs days −8, −5 to −3, and −1 IFN-γ and TNF-α ↓ Histopathology ↓  
  IL-10 ↑   

 
    CCR9+ pDCs MHC-mismatched (BALB/c → B6) CD40/CD80/CD86 expression ↓ Survival ↑ 128  
 Host-type DCs day 0 (mobilized with Flt3L) Intermediate expression of MHC class II   
  Induce Treg   
  IL-12–producing effector T cells ↓   

 
    CD49+CD200R3+ DCs (naturally occurring DCregMHC-matched, miHA-mismatched cGVHD (B10.D2 → BALB/c) Anergy of Ag-specific T cells Cutaneous GVHD ↓ 130  
 Host-type DCs days 2, 9, and 16 TNF-α, IL-12p70, and IFN-γ ↓   
  Induce Treg   
TolDCGVHD model; treatmentEffect on DCs (in vitro)Effect on GVHDReference
DC conditioning     
    Rapamycin MHC-mismatched (B6 → BALB/c) MHC class II, CD80/CD86 expression ↓ Survival ↑ 117  
 Host-type DCs day 0 Intact in vivo trafficking Histopathology ↓  
  Intact CCR5, CCR7, and CD62L   

 
    VIP MHC (B6 → BALB/c, BALB/c → B6) and miHA-mismatched (B6 → F1) Host-type DCs day 2 and/or day 5 Donor CD4+ T-cell Ag-specific response ↓ Induce Treg Survival ↑ (miHA > MHA; early > late for MHA) 118,
146  

 
    SAHA (HDAC inhibitor) MHC-mismatched (BALB/c → B6) CD40/CD80 expression ↓ Survival ↑ 104  
 Host-type DCs days −1, 0, and 2 TNF-α ↓   
  T-cell proliferation ↓   

 
    TGF-β, IL-10 + LPS (DCreg)* MHC-matched, miHA-mismatched cGVHD (B10.D2 → BALB/c) Induce anergy of Ag-specific T cells Cutaneous GVHD ↓ 129  
 Host-type DCs days 2, 9, and 16 or days 18, 25, and 32 vs short-course rapamycin TNF-α, IL-12p70, and IFN-γ ↓   
  Induce Treg   

 
Subset (no DC conditioning)     
    CD8α+ DCs MHC (BALB/c → B6) and miHA (C3H.SW → B6) mismatched T-cell proliferation ↓ Survival ↑ 124  
 Host-type DCs days −8, −5 to −3, and −1 IFN-γ and TNF-α ↓ Histopathology ↓  
  IL-10 ↑   

 
    CCR9+ pDCs MHC-mismatched (BALB/c → B6) CD40/CD80/CD86 expression ↓ Survival ↑ 128  
 Host-type DCs day 0 (mobilized with Flt3L) Intermediate expression of MHC class II   
  Induce Treg   
  IL-12–producing effector T cells ↓   

 
    CD49+CD200R3+ DCs (naturally occurring DCregMHC-matched, miHA-mismatched cGVHD (B10.D2 → BALB/c) Anergy of Ag-specific T cells Cutaneous GVHD ↓ 130  
 Host-type DCs days 2, 9, and 16 TNF-α, IL-12p70, and IFN-γ ↓   
  Induce Treg   

VIP indicates vasoactive intestinal peptide; and LPS, lipopolysaccharide.

*

Depleted of CD40+CD80+CD86+ cells.

Pharmacologic manipulation of DCs (eg, using dexamethasone, rapamycin, or IL-10) renders DCs maturation-resistant and enhances their tolerogenic potential for inhibition of allograft rejection and GVHD. As an example, rapamycin-treated DCs (RAPA-DCs) resist maturation and have impaired capacity to stimulate allogeneic effector T cells yet promote Treg.93  When adoptively transferred to organ graft recipients, RAPA-DCs promote transplant survival and, in conjunction with a short course of host immunosuppression, can induce indefinite graft survival.93,115,116  When administered systemically in experimental GVHD, host-derived RAPA-DCs traffic to secondary lymphoid tissue and improve both survival and histopathologic grade of GVHD.117 

Similarly, vasoactive intestinal peptide is an immunosuppressive neuropeptide that has been used to generate host-derived tolDCs that increase Treg and abrogate aGVHD while maintaining GVL.118  Interestingly, early administration (by day 5) of these tolDCs is critical in the MHC-mismatched model. They were more effective in the miHA-mismatched model regardless of timing.118 

IDO is an important enzyme in tryptophan catabolism that is thought to be critical for control of Teff responses.119  After experimental GVHD, IDO expression in host APCs is increased via IFN-γ release by donor T cells. IDO−/− recipients have accelerated colonic GVHD and mortality, with enhanced T-cell proliferation and decreased apoptosis.120,121  Specific culture conditions (eg, low tryptophan or lipopolysaccharide and IFN-γ) can be used to generate tolDCs with increased IDO expression.122,123  Although these tolDCs have not been studied directly in experimental GVHD, DCs treated with the HDAC inhibitor SAHA display enhanced IDO expression and suppress experimental GVHD in an IDO-dependent manner.104  In addition, increasing colon IDO expression via the injection of kynurenine (tryptophan breakdown product) or a TLR7/TLR8 agonist (3M-011) abrogates experimental GVHD mortality.121 

Cell sorting can be used to isolate/purify tolDCs. Murine CD8α+ DCs are the principal DC subset involved in cross-presentation (Table 1) and have tolerogenic properties.124  In both MHC- and miHA-mismatched models of aGVHD, immunization of recipients with ex vivo–generated and FACS-sorted autologous CD8α+ DC pretransplant reduces GVHD in an IL-10–dependent, Ag-specific manner.124  These results confirm the therapeutic ability of CD8α+ DCs to modify aGVHD, as shown in earlier studies in which Flt3L administration expanded CD8α+ DCs in vivo and reduced aGVHD.125  Ildstadt and colleagues have also described how CD8α+/TCR “facilitating cells,” with a critical component of plasmacytoid precursor-like CD11c+/B220+/CD11b cells, enhance HSC engraftment in mice without increased GVHD.126,127  This effect was attributed to the induction of Ag-specific chimeric Treg that suppress effector T cells. Murine CCR9+ pDCs, obtained via Flt3L-induced mobilization and cell sorting, display an immature phenotype and prevent experimental aGVHD via induction of Treg and suppression of IL-17–producing effector T cells while maintaining IFN-γ–producing effector T cells.128  Overall, distinct subsets of ex vivo–fashioned tolDCs or endogenous DCs have potential for therapy of GVHD, and an important question is which subset is best suited for therapeutic application.

So-called “regulatory DCs” (DCreg), generated by culturing BM in GM-CSF, IL-10, and TGF-β, are proposed to have greater therapeutic efficacy than conventional tolDCs.129  There is evidence that DCreg exclusively express CD200R3 and that naturally occurring mouse CD49+CD200R3+ DCs are identical phenotypically and functionally.130  Both BM-derived and naturally occurring recipient-type DCreg protect against cutaneous cGVHD in a multiple miHA- or MHA- mismatched model via the generation of donor inducible Treg and anergic, Ag-specific CD4+ T cells.130  Moreover, depletion of CD49+CD200R3+ cells before alloHSCT enhanced the progression of cGVHD.130 

MSCs

Mesenchymal stem cells (MSCs) are rare, heterogeneous, pluripotent nonhematopoietic progenitors present in normal BM and adipose tissue that induce immune tolerance via effects on multiple immune cells, in particular DCs. Human MSCs impair DC maturation and induce T-cell hyporesponsiveness in a dose- and contact-dependent manner. The effect can be partly reversed by DC maturation and by blocking IL-10 or IL-6.131  TolDCs generated by coculture of DCs with human MSCs (MSC-DCs) induce Ag-specific Treg via activation of the Notch pathway, but they have not been studied in vivo.132,133 

MSCs have shown promise in the prevention and treatment of GVHD. As reviewed by Baron and Storb,134  while various mouse models have generated conflicting results, they suggest the importance of MSC dose, timing, and activation status. Phase 1 and 2 human studies have demonstrated safety and possible efficacy, and multicenter randomized blinded trials are currently underway.134  Interestingly, the combination of rapamycin and MSCs after experimental cardiac transplantation led to long-term graft survival with significantly increased splenic Treg and tolDCs.135  This also highlights the capacity of synergistic therapies in the promotion of tolerance.135 

MDSCs

Myeloid-derived suppressor cells (MDSCs) are heterogeneous hematopoietic precursor cells with immunosuppressive properties, first noted to aid tumor evasion in mice and humans.136  As reviewed by Lees et al,137  MDSCs modulate both innate and adaptive immunity. Although many of their functions are attributed to direct effects on T cells, MDSCs additionally inhibit the differentiation and maturation of DCs. In mice, MDSCs generated from BM cells in G-CSF, GM-CSF, and IL-13 (MDSC–IL-13) were more potent inhibitors of MHC-mismatched GVHD than conventional MDSCs.136  This inhibition was dependent on the L-arginine-depleting enzyme arginase.136  Importantly, MDSC–IL-13 do not impair the GVL effect in vivo.136 

Treg

As a bidirectional tolerogenic feedback loop exists between Treg and tolDCs, Treg therapy supports tolerance through effects on DCs.41  DCs also control the number and function of Treg.44  Host APC alloAg expression is necessary and sufficient for Treg function in both miHA- and MHC-mismatched mouse models of GVHD, independent of APC IL-10 or IDO expression.138  In addition, human Treg (generated via CD127 [IL-7Rα] negative selection) and Treg-conditioned DCs can abrogate xenogeneic GVHD via induction of immunosuppressive PD-L1 expression on conditioned DCs and on effector T cells in vivo.56  Furthermore, Ag-specific Treg can be induced and expanded by DCs, as demonstrated by human monocyte-derived DCs in an IDO-dependent manner.139 

Adoptive transfer of Treg is highly effective in the prevention of experimental GVHD; thus, phase 1 trials are underway with initial studies demonstrating safety and some efficacy.140,141  A major impediment to Treg therapy has been the generation of sufficient cell numbers, particularly for natural Treg.141  Interestingly, the addition of rapamycin (for restimulation of natural Treg or for the generation of iTreg with TGF-β) increases Treg yields, which may allow completion of dose escalation trials.141 

There are numerous open clinical trials for the prevention or treatment of GVHD currently studying pharmacologic or biologic interventions and cellular therapies that target or impact DCs (Table 5). Although not listed in Table 5, there are also many ongoing trials assessing the impact of conventional GVHD therapies (eg, corticosteroids, CNI, rapamycin) used in new combinations and via different routes (eg, topical, intrahepatic). Cellular therapy remains particularly intriguing, with the majority of active studies using MSCs. A single trial has been underway testing autologous DCs in the setting of relapsed hematologic malignancy; although the DCs are not being used for the prevention or treatment of GVHD, GVHD is a primary outcome measure of the study and the trial will hopefully demonstrate safety and feasibility of DC therapy in the SCT setting.

Table 5

Active clinical trials using interventions that target/impact DCs

IDConditionInterventionPhaseStudy typeSponsor
HDAC inhibition 
    NCT00810602 aGVHD Vorinostat plus tacrolimus and mycophenolate after RIC related donor allogeneic transplant; prevention II Single-agent, open-label, non-randomized safety/efficacy University of Michigan Cancer Center 
    NCT01111526 aGVHD Panobinostat (LBH589) plus corticosteroids; initial treatment I/II Non-randomized, open-label, safety/efficacy H. Lee Moffitt Cancer Center and Research Institute 
Proteasome inhibition 
    NCT01158105 Steroid-refractory cGVHD Bortezomib; treatment II Single-agent, open-label, safety/efficacy Baylor Research Institute 
    NCT00670423 GVHD Bortezomib plus tacrolimus and sirolimus after allogeneic PBSC transplant; prevention Single-agent, open-label, safety Indiana University School of Medicine 
    NCT01323920 aGVHD Bortezomib plus tacrolimus and methotrexate after myeloablative allogeneic SCT without HLA-matched related donor; prevention II Single-agent, open-label, safety/efficacy Dana-Farber Cancer Institute 
    NCT01163786 Bronchiolitis obliterans (cGVHD) Bortezomib; treatment II Single-agent, open-label, safety/efficacy Northwestern University 
Antibody therapy 
    NCT01012492 aGVHD Abatacept (CTLA4-Ig) plus cyclosporine and methotrexate after unrelated donor HSCT; prevention II Single group, open-label, safety Emory University 
Cellular therapy 
    NCT00603330 Grade II to IV steroid-refractory aGVHD MSCs; treatment II Single-agent, open-label, efficacy University Hospital of Liege 
    NCT00827398 Grade II to IV steroid-refractory aGVHD MSCs; treatment I/II Single-agent, open-label, safety/efficacy UMC Utrecht 
    NCT00759018 Grade II to IV steroid-refractory aGVHD MSCs; treatment NA Expanded access (pediatrics) Osiris Therapeutics 
    NCT00826046 Grade II to IV steroid-refractory aGVHD MSCs; treatment NA Expanded access (adults) Osiris Therapeutics 
    NCT01522716 Steroid-refractory cGVHD MSCs; treatment Single-agent, open-label, safety/efficacy Karolinska Institute 
    NCT01045382 GVHD MSCs versus placebo in HLA-mismatched allogeneic transplant after non-myeloablative conditioning; prevention II Randomized, double-blind, safety/efficacy University Hospital of Liege 
    NCT01222039 cGVHD Conventional therapy versus conventional therapy plus MSCs derived from adipose tissue; treatment I/II Multicenter, randomized, safety/efficacy Fundacion Progreso y Salud, Spain 
    NCT00957931 GVHD Haploidentical MSCs in MUD HCT in patients with high-risk non-malignant RBC disorders after RIC; prevention II Non-randomized, open-label, efficacy Stanford University 
    NCT01050764 GVHD Allogeneic Treg plus allogeneic conventional T cells after allogeneic MAC HCT with haploidentical related donor for patients with hematologic malignancies; prevention II Non-randomized, open-label, safety/efficacy Stanford University 
    NCT00935597 GVHD Host DC infusion after allogeneic SCT for prevention or treatment of relapsed disease in patients with advanced hematologic malignancies Non-randomized, open-label, safety/efficacy Mt Sinai School of Medicine 
IDConditionInterventionPhaseStudy typeSponsor
HDAC inhibition 
    NCT00810602 aGVHD Vorinostat plus tacrolimus and mycophenolate after RIC related donor allogeneic transplant; prevention II Single-agent, open-label, non-randomized safety/efficacy University of Michigan Cancer Center 
    NCT01111526 aGVHD Panobinostat (LBH589) plus corticosteroids; initial treatment I/II Non-randomized, open-label, safety/efficacy H. Lee Moffitt Cancer Center and Research Institute 
Proteasome inhibition 
    NCT01158105 Steroid-refractory cGVHD Bortezomib; treatment II Single-agent, open-label, safety/efficacy Baylor Research Institute 
    NCT00670423 GVHD Bortezomib plus tacrolimus and sirolimus after allogeneic PBSC transplant; prevention Single-agent, open-label, safety Indiana University School of Medicine 
    NCT01323920 aGVHD Bortezomib plus tacrolimus and methotrexate after myeloablative allogeneic SCT without HLA-matched related donor; prevention II Single-agent, open-label, safety/efficacy Dana-Farber Cancer Institute 
    NCT01163786 Bronchiolitis obliterans (cGVHD) Bortezomib; treatment II Single-agent, open-label, safety/efficacy Northwestern University 
Antibody therapy 
    NCT01012492 aGVHD Abatacept (CTLA4-Ig) plus cyclosporine and methotrexate after unrelated donor HSCT; prevention II Single group, open-label, safety Emory University 
Cellular therapy 
    NCT00603330 Grade II to IV steroid-refractory aGVHD MSCs; treatment II Single-agent, open-label, efficacy University Hospital of Liege 
    NCT00827398 Grade II to IV steroid-refractory aGVHD MSCs; treatment I/II Single-agent, open-label, safety/efficacy UMC Utrecht 
    NCT00759018 Grade II to IV steroid-refractory aGVHD MSCs; treatment NA Expanded access (pediatrics) Osiris Therapeutics 
    NCT00826046 Grade II to IV steroid-refractory aGVHD MSCs; treatment NA Expanded access (adults) Osiris Therapeutics 
    NCT01522716 Steroid-refractory cGVHD MSCs; treatment Single-agent, open-label, safety/efficacy Karolinska Institute 
    NCT01045382 GVHD MSCs versus placebo in HLA-mismatched allogeneic transplant after non-myeloablative conditioning; prevention II Randomized, double-blind, safety/efficacy University Hospital of Liege 
    NCT01222039 cGVHD Conventional therapy versus conventional therapy plus MSCs derived from adipose tissue; treatment I/II Multicenter, randomized, safety/efficacy Fundacion Progreso y Salud, Spain 
    NCT00957931 GVHD Haploidentical MSCs in MUD HCT in patients with high-risk non-malignant RBC disorders after RIC; prevention II Non-randomized, open-label, efficacy Stanford University 
    NCT01050764 GVHD Allogeneic Treg plus allogeneic conventional T cells after allogeneic MAC HCT with haploidentical related donor for patients with hematologic malignancies; prevention II Non-randomized, open-label, safety/efficacy Stanford University 
    NCT00935597 GVHD Host DC infusion after allogeneic SCT for prevention or treatment of relapsed disease in patients with advanced hematologic malignancies Non-randomized, open-label, safety/efficacy Mt Sinai School of Medicine 

PBSC indicates peripheral blood stem cell; MUD, matched-unrelated donor; RBC, red blood cells; and NA, not applicable.

Despite therapies that broadly target effector T cells or globally suppress immunity, GVHD remains a significant cause of post-HSCT morbidity and mortality. Given the tolerogenic potential of some DC subsets and the critical role of others in the pathogenesis of GVHD, differences in DC characteristics may be used to predict outcome, whereas targeting DCs is an innovative treatment approach. Likewise, DCs may be targeted directly in vivo through molecular pathways or cell surface expression of maturation markers or costimulatory molecules, or tolDCs may be generated in vitro and given in the peritransplant period (summarized in Figure 3). Other cellular therapies, including Treg, mediate dominant immunosuppressive effects by restraining DC stimulatory functions. Given the importance of the GVL effect, any therapy targeting or using DCs must conserve this process.

Figure 3

Potential DC-based therapies for GVHD. TolDCs can be used as a negative cellular vaccine after in vitro generation via their pharmacologic manipulation, cell sorting (subsets), or expansion after interaction with other immune regulatory cell populations. In vivo, DCs can be targeted by the inhibition of molecular pathways (HDAC; NF-κB) or the expression of maturation markers or costimulatory molecules (eg, CD80/CD86; CD83). Other cellular therapies, such as MSCs, MDSCs, and Treg, mediate immunosuppressive effects through DCs. VIP indicates vasoactive intestinal peptide. Professional illustration by Alice Y. Chen.

Figure 3

Potential DC-based therapies for GVHD. TolDCs can be used as a negative cellular vaccine after in vitro generation via their pharmacologic manipulation, cell sorting (subsets), or expansion after interaction with other immune regulatory cell populations. In vivo, DCs can be targeted by the inhibition of molecular pathways (HDAC; NF-κB) or the expression of maturation markers or costimulatory molecules (eg, CD80/CD86; CD83). Other cellular therapies, such as MSCs, MDSCs, and Treg, mediate immunosuppressive effects through DCs. VIP indicates vasoactive intestinal peptide. Professional illustration by Alice Y. Chen.

Close modal

Further understanding of the precise immunoregulatory properties of DCs and the development of DC-based therapies for GVHD will expand HSCT use beyond treatment of malignant disease and allow its use in patients lacking MHC-matched donors. Early work by Shlomchik et al elucidated the critical role of miHA expression by host hematopoietic APCs for CD8+ T cell–driven GVHD2,46 ; thus, therapies that orchestrate the successful and timely suppression and/or ablation of host DCs are expected to be particularly beneficial to patients after MHC-matched HSCT. Very recent findings suggest that recipient nonhematopoietic APCs in target organs may be central to promoting indirect CD4+ T cell-mediated aGVHD.142  In these studies, host CD11c+ DCs suppressed GVHD development. These and other data discussed herein underscore the importance of developing a clear understanding of DC involvement in the complex immunopathology of GVHD. Likewise, infused recipient Ag-pulsed donor tolDCs or recipient tolDCs presenting alloAg and miHA have the potential to prevent the pathologic alloresponses of donor T cells and benefit HSCT patients given either MHC-matched or mismatched transplants.

Finally, given the role of both DCs and T cells in the pathogenesis of GVHD, synergistic therapies or those that target both cell types in vivo may be more effective. Cellular therapies, specifically tolDCs and Treg, are intriguing in their ability to modulate one another in vivo. Importantly, cellular therapies have begun in humans. Human tolDCs have been generated and characterized in vitro using clinical-grade reagents.143  Recently, a DC-based vaccine for the treatment of prostate cancer was approved by the FDA,144  and the first report has appeared of a phase 1 safety study of tolDCs in patients with type 1 (autoimmune) diabetes.145  As other forms of innovative cell therapy, including testing of Treg, are underway for the prevention of GVHD,140,141  there would appear to be adequate justification for phase 1 studies of tolDCs alone and in combination with Treg in HSCT.

The publication costs of this article were defrayed in part by page charge payment. Therefore, and solely to indicate this fact, this article is hereby marked “advertisement” in accordance with 18 USC section 1734.

The authors thank Brian Rosborough for critical reading of the manuscript.

This work was supported by the National Institutes of Health (grants R01AI67541 and R01AI60994, A.W.T.; grants R01HL93716 and RO1GM63569, M.Y.M.; and grant K99/R00 HL97155, H.R.T.) and the Pittsburgh Foundation (M.Y.M.).

National Institutes of Health

Contribution: E.O.S. and A.W.T. wrote the manuscript; H.R.T. assisted in writing the manuscript and generation of the figures; and M.Y.M. assisted in editing the manuscript.

Conflict-of-interest disclosure: The authors declare no competing financial interests.

Correspondence: Angus W. Thomson, Starzl Transplantation Institute, University of Pittsburgh School of Medicine, 200 Lothrop St, BST W1540, Pittsburgh, PA 15261; e-mail: thomsonaw@upmc.edu.

1
Hutter
G
Nowak
D
Mossner
M
et al
Long-term control of HIV by CCR5 Delta32/Delta32 stem-cell transplantation.
N Engl J Med
2009
360
7
692
698
2
Shlomchik
WD
Graft-versus-host disease.
Nat Rev Immunol
2007
7
5
340
352
3
Ferrara
JL
Levine
JE
Reddy
P
Holler
E
Graft-versus-host disease.
Lancet
2009
373
9674
1550
1561
4
Billingham
RE
The biology of graft-versus-host reactions.
Harvey Lect
1966
62
21
78
5
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
6
Watowich
SS
Liu
YJ
Mechanisms regulating dendritic cell specification and development.
Immunol Rev
2010
238
1
76
92
7
Steinman
RM
Hawiger
D
Nussenzweig
MC
Tolerogenic dendritic cells.
Annu Rev Immunol
2003
21
685
711
8
Iwasaki
A
Medzhitov
R
Toll-like receptor control of the adaptive immune responses.
Nat Immunol
2004
5
10
987
995
9
Banchereau
J
Steinman
RM
Dendritic cells and the control of immunity.
Nature
1998
392
6673
245
252
10
Banchereau
J
Briere
F
Caux
C
et al
Immunobiology of dendritic cells.
Annu Rev Immunol
2000
18
767
811
11
Gilliet
M
Cao
W
Liu
YJ
Plasmacytoid dendritic cells: sensing nucleic acids in viral infection and autoimmune diseases.
Nat Rev Immunol
2008
8
8
594
606
12
Steinman
RM
Hemmi
H
Dendritic cells: translating innate to adaptive immunity.
Curr Top Microbiol Immunol
2006
311
17
58
13
Schmid
MA
Kingston
D
Boddupalli
S
Manz
MG
Instructive cytokine signals in dendritic cell lineage commitment.
Immunol Rev
2010
234
1
32
44
14
Liu
K
Nussenzweig
MC
Origin and development of dendritic cells.
Immunol Rev
2010
234
1
45
54
15
Buza-Vidas
N
Woll
P
Hultquist
A
et al
FLT3 expression initiates in fully multipotent mouse hematopoietic progenitor cells.
Blood
2011
118
6
1544
1548
16
Akashi
K
Traver
D
Miyamoto
T
Weissman
IL
A clonogenic common myeloid progenitor that gives rise to all myeloid lineages.
Nature
2000
404
6774
193
197
17
Shortman
K
Naik
SH
Steady-state and inflammatory dendritic-cell development.
Nat Rev Immunol
2007
7
1
19
30
18
Manz
MG
Traver
D
Miyamoto
T
Weissman
IL
Akashi
K
Dendritic cell potentials of early lymphoid and myeloid progenitors.
Blood
2001
97
11
3333
3341
19
Chicha
L
Jarrossay
D
Manz
MG
Clonal type I interferon-producing and dendritic cell precursors are contained in both human lymphoid and myeloid progenitor populations.
J Exp Med
2004
200
11
1519
1524
20
D'Amico
A
Wu
L
The early progenitors of mouse dendritic cells and plasmacytoid predendritic cells are within the bone marrow hemopoietic precursors expressing Flt3.
J Exp Med
2003
198
2
293
303
21
Brasel
K
De Smedt
T
Smith
JL
Maliszewski
CR
Generation of murine dendritic cells from flt3-ligand-supplemented bone marrow cultures.
Blood
2000
96
9
3029
3039
22
Fancke
B
Suter
M
Hochrein
H
O'Keeffe
M
M-CSF: a novel plasmacytoid and conventional dendritic cell poietin.
Blood
2008
111
1
150
159
23
Wu
L
Liu
YJ
Development of dendritic-cell lineages.
Immunity
2007
26
6
741
750
24
Wu
L
Shortman
K
Heterogeneity of thymic dendritic cells.
Semin Immunol
2005
17
4
304
312
25
Atibalentja
DF
Murphy
KM
Unanue
ER
Functional redundancy between thymic CD8alpha+ and Sirpalpha+ conventional dendritic cells in presentation of blood-derived lysozyme by MHC class II proteins.
J Immunol
2011
186
3
1421
1431
26
Koyama
M
Hashimoto
D
Aoyama
K
et al
Plasmacytoid dendritic cells prime alloreactive T cells to mediate graft-versus-host disease as antigen-presenting cells.
Blood
2009
113
9
2088
2095
27
Young
JW
Merad
M
Hart
DN
Dendritic cells in transplantation and immune-based therapies.
Biol Blood Marrow Transplant
2007
13
1 suppl 1
23
32
28
Matta
BM
Castellaneta
A
Thomson
AW
Tolerogenic plasmacytoid DCs.
Eur J Immunol
2010
40
10
2667
2676
29
Romani
N
Clausen
BE
Stoitzner
P
Langerhans cells and more: Langerin-expressing dendritic cell subsets in the skin.
Immunol Rev
2010
234
1
120
141
30
Coquerelle
C
Moser
M
DC subsets in positive and negative regulation of immunity.
Immunol Rev
2010
234
1
317
334
31
Heath
WR
Carbone
FR
Dendritic cell subsets in primary and secondary T cell responses at body surfaces.
Nat Immunol
2009
10
12
1237
1244
32
Doulatov
S
Notta
F
Eppert
K
Nguyen
LT
Ohashi
PS
Dick
JE
Revised map of the human progenitor hierarchy shows the origin of macrophages and dendritic cells in early lymphoid development.
Nat Immunol
2010
11
7
585
593
33
Mittag
D
Proietto
AI
Loudovaris
T
et al
Human dendritic cell subsets from spleen and blood are similar in phenotype and function but modified by donor health status.
J Immunol
2011
186
6207
6217
34
Ueno
H
Schmitt
N
Klechevsky
E
et al
Harnessing human dendritic cell subsets for medicine.
Immunol Rev
2010
234
1
199
212
35
Klechevsky
E
Morita
R
Liu
M
et al
Functional specializations of human epidermal Langerhans cells and CD14+ dermal dendritic cells.
Immunity
2008
29
3
497
510
36
Caux
C
Massacrier
C
Vanbervliet
B
et al
CD34+ hematopoietic progenitors from human cord blood differentiate along two independent dendritic cell pathways in response to granulocyte-macrophage colony-stimulating factor plus tumor necrosis factor alpha: II. Functional analysis.
Blood
1997
90
4
1458
1470
37
Schmitt
N
Morita
R
Bourdery
L
et al
Human dendritic cells induce the differentiation of interleukin-21-producing T follicular helper-like cells through interleukin-12.
Immunity
2009
31
1
158
169
38
Villadangos
JA
Shortman
K
Found in translation: the human equivalent of mouse CD8+ dendritic cells.
J Exp Med
2010
207
6
1131
1134
39
Morelli
AE
Thomson
AW
Tolerogenic dendritic cells and the quest for transplant tolerance.
Nat Rev Immunol
2007
7
8
610
621
40
Maldonado
RA
von Andrian
UH
How tolerogenic dendritic cells induce regulatory T cells.
Adv Immunol
2010
108
111
165
41
Min
WP
Zhou
D
Ichim
TE
et al
Inhibitory feedback loop between tolerogenic dendritic cells and regulatory T cells in transplant tolerance.
J Immunol
2003
170
3
1304
1312
42
Fallarino
F
Grohmann
U
Hwang
KW
et al
Modulation of tryptophan catabolism by regulatory T cells.
Nat Immunol
2003
4
12
1206
1212
43
Darrasse-Jeze
G
Deroubaix
S
Mouquet
H
et al
Feedback control of regulatory T cell homeostasis by dendritic cells in vivo.
J Exp Med
2009
206
9
1853
1862
44
Swee
LK
Bosco
N
Malissen
B
Ceredig
R
Rolink
A
Expansion of peripheral naturally occurring T regulatory cells by Fms-like tyrosine kinase 3 ligand treatment.
Blood
2009
113
25
6277
6287
45
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
46
Shlomchik
WD
Couzens
MS
Tang
CB
et al
Prevention of graft versus host disease by inactivation of host antigen-presenting cells.
Science
1999
285
5426
412
415
47
Duffner
UA
Maeda
Y
Cooke
KR
et al
Host dendritic cells alone are sufficient to initiate acute graft-versus-host disease.
J Immunol
2004
172
12
7393
7398
48
Matte
CC
Liu
J
Cormier
J
et al
Donor APCs are required for maximal GVHD but not for GVL.
Nat Med
2004
10
9
987
992
49
Merad
M
Hoffmann
P
Ranheim
E
et al
Depletion of host Langerhans cells before transplantation of donor alloreactive T cells prevents skin graft-versus-host disease.
Nat Med
2004
10
5
510
517
50
Li
H
Kaplan
DH
Matte-Martone
C
et al
Langerhans cells are not required for graft-versus-host disease.
Blood
2011
117
2
697
707
51
Anderson
BE
McNiff
JM
Jain
D
Blazar
BR
Shlomchik
WD
Shlomchik
MJ
Distinct roles for donor- and host-derived antigen-presenting cells and costimulatory molecules in murine chronic graft-versus-host disease: requirements depend on target organ.
Blood
2005
105
5
2227
2234
52
Zhao
D
Young
JS
Chen
YH
et al
Alloimmune response results in expansion of autoreactive donor CD4+ T cells in transplants that can mediate chronic graft-versus-host disease.
J Immunol
2011
186
2
856
868
53
Sakoda
Y
Hashimoto
D
Asakura
S
et al
Donor-derived thymic-dependent T cells cause chronic graft-versus-host disease.
Blood
2007
109
4
1756
1764
54
Zhang
Y
Hexner
E
Frank
D
Emerson
SG
CD4+ T cells generated de novo from donor hemopoietic stem cells mediate the evolution from acute to chronic graft-versus-host disease.
J Immunol
2007
179
5
3305
3314
55
Li
J
Semple
K
Suh
WK
et al
Roles of CD28, CTLA4, and inducible costimulator in acute graft-versus-host disease in mice.
Biol Blood Marrow Transplant
2011
17
7
962
969
56
Amarnath
S
Costanzo
CM
Mariotti
J
et al
Regulatory T cells and human myeloid dendritic cells promote tolerance via programmed death ligand-1.
PLoS Biol
2010
8
2
e1000302
57
Wang
X
Li
H
Matte-Martone
C
et al
Mechanisms of antigen presentation to T cells in murine graft-vs-host disease: cross-presentation and the appearance of cross-presentation.
Blood
2011
118
24
6426
6437
58
Markey
KA
Banovic
T
Kuns
RD
et al
Conventional dendritic cells are the critical donor APC presenting alloantigen after experimental bone marrow transplantation.
Blood
2009
113
22
5644
5649
59
Reddy
P
Maeda
Y
Liu
C
Krijanovski
OI
Korngold
R
Ferrara
JL
A crucial role for antigen-presenting cells and alloantigen expression in graft-versus-leukemia responses.
Nat Med
2005
11
11
1244
1249
60
Chakraverty
R
Eom
HS
Sachs
J
et al
Host MHC class II+ antigen-presenting cells and CD4 cells are required for CD8-mediated graft-versus-leukemia responses following delayed donor leukocyte infusions.
Blood
2006
108
6
2106
2113
61
Li
JM
Waller
EK
Donor antigen-presenting cells regulate T-cell expansion and antitumor activity after allogeneic bone marrow transplantation.
Biol Blood Marrow Transplant
2004
10
8
540
551
62
Li
JM
Southerland
LT
Lu
Y
et al
Activation, immune polarization, and graft-versus-leukemia activity of donor T cells are regulated by specific subsets of donor bone marrow antigen-presenting cells in allogeneic hemopoietic stem cell transplantation.
J Immunol
2009
183
12
7799
7809
63
Rajasekar
R
Lakshmi
KM
George
B
et al
Dendritic cell count in the graft predicts relapse in patients with hematologic malignancies undergoing an HLA-matched related allogeneic peripheral blood stem cell transplant.
Biol Blood Marrow Transplant
2010
16
6
854
860
64
Reddy
V
Iturraspe
JA
Tzolas
AC
Meier-Kriesche
HU
Schold
J
Wingard
JR
Low dendritic cell count after allogeneic hematopoietic stem cell transplantation predicts relapse, death, and acute graft-versus-host disease.
Blood
2004
103
11
4330
4335
65
Mackinnon
S
Papadopoulos
EB
Carabasi
MH
et al
Adoptive immunotherapy evaluating escalating doses of donor leukocytes for relapse of chronic myeloid leukemia after bone marrow transplantation: separation of graft-versus-leukemia responses from graft-versus-host disease.
Blood
1995
86
4
1261
1268
66
Mapara
MY
Kim
YM
Wang
SP
Bronson
R
Sachs
DH
Sykes
M
Donor lymphocyte infusions mediate superior graft-versus-leukemia effects in mixed compared to fully allogeneic chimeras: a critical role for host antigen-presenting cells.
Blood
2002
100
5
1903
1909
67
Durakovic
N
Radojcic
V
Skarica
M
et al
Factors governing the activation of adoptively transferred donor T cells infused after allogeneic bone marrow transplantation in the mouse.
Blood
2007
109
10
4564
4574
68
Peggs
KS
Thomson
K
Hart
DP
et al
Dose-escalated donor lymphocyte infusions following reduced intensity transplantation: toxicity, chimerism, and disease responses.
Blood
2004
103
4
1548
1556
69
Schmid
C
Labopin
M
Nagler
A
et al
Donor lymphocyte infusion in the treatment of first hematological relapse after allogeneic stem-cell transplantation in adults with acute myeloid leukemia: a retrospective risk factors analysis and comparison with other strategies by the EBMT Acute Leukemia Working Party.
J Clin Oncol
2007
25
31
4938
4945
70
Kolb
HJ
Schattenberg
A
Goldman
JM
et al
Graft-versus-leukemia effect of donor lymphocyte transfusions in marrow grafted patients.
Blood
1995
86
5
2041
2050
71
Turner
BE
Kambouris
ME
Sinfield
L
et al
Reduced intensity conditioning for allogeneic hematopoietic stem-cell transplant determines the kinetics of acute graft-versus-host disease.
Transplantation
2008
86
7
968
976
72
Durakovic
N
Bezak
KB
Skarica
M
et al
Host-derived Langerhans cells persist after MHC-matched allografting independent of donor T cells and critically influence the alloresponses mediated by donor lymphocyte infusions.
J Immunol
2006
177
7
4414
4425
73
Auffermann-Gretzinger
S
Eger
L
Bornhauser
M
et al
Fast appearance of donor dendritic cells in human skin: dynamics of skin and blood dendritic cells after allogeneic hematopoietic cell transplantation.
Transplantation
2006
81
6
866
873
74
Bogunovic
M
Ginhoux
F
Wagers
A
et al
Identification of a radio-resistant and cycling dermal dendritic cell population in mice and men.
J Exp Med
2006
203
12
2627
2638
75
Banovic
T
Markey
KA
Kuns
RD
et al
Graft-versus-host disease prevents the maturation of plasmacytoid dendritic cells.
J Immunol
2009
182
2
912
920
76
Korbling
M
Anderlini
P
Peripheral blood stem cell versus bone marrow allotransplantation: does the source of hematopoietic stem cells matter?
Blood
2001
98
10
2900
2908
77
Arpinati
M
Chirumbolo
G
Urbini
B
Perrone
G
Rondelli
D
Anasetti
C
Role of plasmacytoid dendritic cells in immunity and tolerance after allogeneic hematopoietic stem cell transplantation.
Transpl Immunol
2003
11
3
345
356
78
Morris
ES
MacDonald
KP
Hill
GR
Stem cell mobilization with G-CSF analogs: a rational approach to separate GVHD and GVL?
Blood
2006
107
9
3430
3435
79
Chakraverty
R
Sykes
M
The role of antigen-presenting cells in triggering graft-versus-host disease and graft-versus-leukemia.
Blood
2007
110
1
9
17
80
Reddy
V
Hill
GR
Pan
L
et al
G-CSF modulates cytokine profile of dendritic cells and decreases acute graft-versus-host disease through effects on the donor rather than the recipient.
Transplantation
2000
69
4
691
693
81
Fagnoni
FF
Oliviero
B
Giorgiani
G
et al
Reconstitution dynamics of plasmacytoid and myeloid dendritic cell precursors after allogeneic myeloablative hematopoietic stem cell transplantation.
Blood
2004
104
1
281
289
82
Collin
MP
Hart
DN
Jackson
GH
et al
The fate of human Langerhans cells in hematopoietic stem cell transplantation.
J Exp Med
2006
203
1
27
33
83
Clark
FJ
Freeman
L
Dzionek
A
et al
Origin and subset distribution of peripheral blood dendritic cells in patients with chronic graft-versus-host disease.
Transplantation
2003
75
2
221
225
84
Chan
GW
Gorgun
G
Miller
KB
Foss
FM
Persistence of host dendritic cells after transplantation is associated with graft-versus-host disease.
Biol Blood Marrow Transplant
2003
9
3
170
176
85
Lau
J
Sartor
M
Bradstock
KF
Vuckovic
S
Munster
DJ
Hart
DN
Activated circulating dendritic cells after hematopoietic stem cell transplantation predict acute graft-versus-host disease.
Transplantation
2007
83
7
839
846
86
Waller
EK
Rosenthal
H
Jones
TW
et al
Larger numbers of CD4(bright) dendritic cells in donor bone marrow are associated with increased relapse after allogeneic bone marrow transplantation.
Blood
2001
97
10
2948
2956
87
Rajasekar
R
Mathews
V
Lakshmi
KM
et al
Plasmacytoid dendritic cell count on day 28 in HLA-matched related allogeneic peripheral blood stem cell transplant predicts the incidence of acute and chronic GVHD.
Biol Blood Marrow Transplant
2008
14
3
344
350
88
Mohty
M
Blaise
D
Faucher
C
et al
Impact of plasmacytoid dendritic cells on outcome after reduced-intensity conditioning allogeneic stem cell transplantation.
Leukemia
2005
19
1
1
6
89
Hackstein
H
Thomson
AW
Dendritic cells: emerging pharmacological targets of immunosuppressive drugs.
Nat Rev Immunol
2004
4
1
24
35
90
Lee
YR
Yang
IH
Lee
YH
et al
Cyclosporin A and tacrolimus, but not rapamycin, inhibit MHC-restricted antigen presentation pathways in dendritic cells.
Blood
2005
105
10
3951
3955
91
Lee
YH
Lee
YR
Im
SA
et al
Calcineurin inhibitors block MHC-restricted antigen presentation in vivo.
J Immunol
2007
179
9
5711
5716
92
Piemonti
L
Monti
P
Allavena
P
et al
Glucocorticoids affect human dendritic cell differentiation and maturation.
J Immunol
1999
162
11
6473
6481
93
Turnquist
H
Raimondi
G
Zahorchak
AF
Fischer
RT
Wang
Z
Thomson
AW
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
7018
7031
94
Stary
G
Klein
I
Bauer
W
et al
Glucocorticosteroids modify Langerhans cells to produce TGF-beta and expand regulatory T cells.
J Immunol
2011
186
1
103
112
95
Paczesny
S
Choi
SW
Ferrara
JL
Acute graft-versus-host disease: new treatment strategies.
Curr Opin Hematol
2009
16
6
427
436
96
Mohty
M
Mechanisms of action of antithymocyte globulin: T-cell depletion and beyond.
Leukemia
2007
21
7
1387
1394
97
Naujokat
C
Berges
C
Fuchs
D
Sadeghi
M
Opelz
G
Daniel
V
Antithymocyte globulins suppress dendritic cell function by multiple mechanisms.
Transplantation
2007
83
4
485
497
98
Auffermann-Gretzinger
S
Eger
L
Schetelig
J
Bornhauser
M
Heidenreich
F
Ehninger
G
Alemtuzumab depletes dendritic cells more effectively in blood than in skin: a pilot study in patients with chronic lymphocytic leukemia.
Transplantation
2007
83
9
1268
1272
99
Ratzinger
G
Reagan
JL
Heller
G
Busam
KJ
Young
JW
Differential CD52 expression by distinct myeloid dendritic cell subsets: implications for alemtuzumab activity at the level of antigen presentation in allogeneic graft-host interactions in transplantation.
Blood
2003
101
4
1422
1429
100
Wuest
SC
Edwan
JH
Martin
JF
et al
A role for interleukin-2 trans-presentation in dendritic cell-mediated T-cell activation in humans, as revealed by daclizumab therapy.
Nat Med
2011
17
5
604
609
101
Schluns
KS
Window of opportunity for daclizumab.
Nat Med
2011
17
5
545
547
102
Sun
Y
Chin
YE
Weisiger
E
et al
Cutting edge: negative regulation of dendritic cells through acetylation of the nonhistone protein STAT-3.
J Immunol
2009
182
10
5899
5903
103
Choi
S
Reddy
P
HDAC inhibition and graft versus host disease.
Mol Med
2011
17
5
404
416
104
Reddy
P
Sun
Y
Toubai
T
et al
Histone deacetylase inhibition modulates indoleamine 2,3-dioxygenase-dependent DCs functions and regulates experimental graft-versus-host disease in mice.
J Clin Invest
2008
118
7
2562
2573
105
Leng
C
Gries
M
Ziegler
J
et al
Reduction of graft-versus-host disease by histone deacetylase inhibitor suberonylanilide hydroxamic acid is associated with modulation of inflammatory cytokine milieu and involves inhibition of STAT1.
Exp Hematol
2006
34
6
776
787
106
Vodanovic-Jankovic
S
Hari
P
Jacobs
P
Komorowski
R
Drobyski
WR
NF-kappaB as a target for the prevention of graft-versus-host disease: comparative efficacy of bortezomib and PS-1145.
Blood
2006
107
2
827
834
107
Sun
K
Welniak
LA
Panoskaltsis-Mortari
A
et al
Inhibition of acute graft-versus-host disease with retention of graft-versus-tumor effects by the proteasome inhibitor bortezomib.
Proc Natl Acad Sci U S A
2004
101
21
8120
8125
108
Tao
Y
Zhang
W
Fang
Y
et al
Bortezomib attenuates acute graft-vs.-host disease through interfering with host immature dendritic cells.
Exp Hematol
2011
39
6
710
720
109
MacDonald
KP
Kuns
RD
Rowe
V
et al
Effector and regulatory T-cell function is differentially regulated by RelB within antigen-presenting cells during GVHD.
Blood
2007
109
11
5049
5057
110
Wilson
J
Cullup
H
Lourie
R
et al
Antibody to the dendritic cell surface activation antigen CD83 prevents acute graft-versus-host disease.
J Exp Med
2009
206
2
387
398
111
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
112
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
113
Morelli
AE
Thomson
AW
Dendritic cells: regulators of alloimmunity and opportunities for tolerance induction.
Immunol Rev
2003
196
125
146
114
Sato
K
Yamashita
N
Yamashita
N
Baba
M
Matsuyama
T
Regulatory dendritic cells protect mice from murine acute graft-versus-host disease and leukemia relapse.
Immunity
2003
18
3
367
379
115
Taner
T
Hackstein
H
Wang
Z
Morelli
AE
Thomson
AW
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
116
Thomson
AW
Turnquist
HR
Raimondi
G
Immunoregulatory functions of mTOR inhibition.
Nat Rev Immunol
2009
9
5
324
337
117
Reichardt
W
Durr
C
von Elverfeldt
D
et al
Impact of mammalian target of rapamycin inhibition on lymphoid homing and tolerogenic function of nanoparticle-labeled dendritic cells following allogeneic hematopoietic cell transplantation.
J Immunol
2008
181
7
4770
4779
118
Chorny
A
Gonzalez-Rey
E
Fernandez-Martin
A
Ganea
D
Delgado
M
Vasoactive intestinal peptide induces regulatory dendritic cells that prevent acute graft-versus-host disease while maintaining the graft-versus-tumor response.
Blood
2006
107
9
3787
3794
119
Curti
A
Trabanelli
S
Salvestrini
V
Baccarani
M
Lemoli
RM
The role of indoleamine 2,3-dioxygenase in the induction of immune tolerance: focus on hematology.
Blood
2009
113
11
2394
2401
120
Jasperson
LK
Bucher
C
Panoskaltsis-Mortari
A
et al
Indoleamine 2,3-dioxygenase is a critical regulator of acute graft-versus-host disease lethality.
Blood
2008
111
6
3257
3265
121
Jasperson
LK
Bucher
C
Panoskaltsis-Mortari
A
Mellor
AL
Munn
DH
Blazar
BR
Inducing the tryptophan catabolic pathway, indoleamine 2,3-dioxygenase (IDO), for suppression of graft-versus-host disease (GVHD) lethality.
Blood
2009
114
24
5062
5070
122
Jurgens
B
Hainz
U
Fuchs
D
Felzmann
T
Heitger
A
Interferon-gamma-triggered indoleamine 2,3-dioxygenase competence in human monocyte-derived dendritic cells induces regulatory activity in allogeneic T cells.
Blood
2009
114
15
3235
3243
123
Brenk
M
Scheler
M
Koch
S
et al
Tryptophan deprivation induces inhibitory receptors ILT3 and ILT4 on dendritic cells favoring the induction of human CD4+CD25+ Foxp3+ T regulatory cells.
J Immunol
2009
183
1
145
154
124
Toubai
T
Malter
C
Tawara
I
et al
Immunization with host-type CD8alpha+ dendritic cells reduces experimental acute GVHD in an IL-10-dependent manner.
Blood
2010
115
3
724
735
125
Teshima
T
Reddy
P
Lowler
KP
et al
Flt3 ligand therapy for recipients of allogeneic bone marrow transplants expands host CD8 alpha(+) dendritic cells and reduces experimental acute graft-versus-host disease.
Blood
2002
99
5
1825
1832
126
Fugier-Vivier
IJ
Rezzoug
F
Huang
Y
et al
Plasmacytoid precursor dendritic cells facilitate allogeneic hematopoietic stem cell engraftment.
J Exp Med
2005
201
3
373
383
127
Huang
Y
Bozulic
LD
Miller
T
Xu
H
Hussain
LR
Ildstad
ST
CD8alpha+ plasmacytoid precursor DCs induce antigen-specific regulatory T cells that enhance HSC engraftment in vivo.
Blood
2011
117
8
2494
2505
128
Hadeiba
H
Sato
T
Habtezion
A
Oderup
C
Pan
J
Butcher
EC
CCR9 expression defines tolerogenic plasmacytoid dendritic cells able to suppress acute graft-versus-host disease.
Nat Immunol
2008
9
11
1253
1260
129
Fujita
S
Sato
Y
Sato
K
et al
Regulatory dendritic cells protect against cutaneous chronic graft-versus-host disease mediated through CD4+CD25+Foxp3+ regulatory T cells.
Blood
2007
110
10
3793
3803
130
Sato
K
Eizumi
K
Fukaya
T
et al
Naturally occurring regulatory dendritic cells regulate murine cutaneous chronic graft-versus-host disease.
Blood
2009
113
19
4780
4789
131
Beyth
S
Borovsky
Z
Mevorach
D
et al
Human mesenchymal stem cells alter antigen-presenting cell maturation and induce T-cell unresponsiveness.
Blood
2005
105
5
2214
2219
132
Aldinucci
A
Rizzetto
L
Pieri
L
et al
Inhibition of immune synapse by altered dendritic cell actin distribution: a new pathway of mesenchymal stem cell immune regulation.
J Immunol
2010
185
9
5102
5110
133
Li
YP
Paczesny
S
Lauret
E
et al
Human mesenchymal stem cells license adult CD34+ hemopoietic progenitor cells to differentiate into regulatory dendritic cells through activation of the Notch pathway.
J Immunol
2008
180
3
1598
1608
134
Baron
F
Storb
R
Mensenchymal stromal cells: a new tool against graft-versus-host disease? [published online ahead of print September 29, 2011].
Biol Blood Marrow Transplant
135
Ge
W
Jiang
J
Baroja
ML
et al
Infusion of mesenchymal stem cells and rapamycin synergize to attenuate alloimmune responses and promote cardiac allograft tolerance.
Am J Transplant
2009
9
8
1760
1772
136
Highfill
SL
Rodriguez
PC
Zhou
Q
et al
Bone marrow myeloid-derived suppressor cells (MDSCs) inhibit graft-versus-host disease (GVHD) via an arginase-1-dependent mechanism that is up-regulated by interleukin-13.
Blood
2010
116
25
5738
5747
137
Lees
JR
Azimzadeh
AM
Bromberg
JS
Myeloid derived suppressor cells in transplantation.
Curr Opin Immunol
2011
23
5
692
697
138
Tawara
I
Shlomchik
WD
Jones
A
et al
A crucial role for host APCs in the induction of donor CD4+CD25+ regulatory T cell-mediated suppression of experimental graft-versus-host disease.
J Immunol
2010
185
7
3866
3872
139
Chung
DJ
Rossi
M
Romano
E
et al
Indoleamine 2,3-dioxygenase-expressing mature human monocyte-derived dendritic cells expand potent autologous regulatory T cells.
Blood
2009
114
3
555
563
140
Brunstein
CG
Miller
JS
Cao
Q
et al
Infusion of ex vivo expanded T regulatory cells in adults transplanted with umbilical cord blood: safety profile and detection kinetics.
Blood
2011
117
3
1061
1070
141
Hippen
KL
Riley
JL
June
CH
Blazar
BR
Clinical perspectives for regulatory T cells in transplantation tolerance.
Semin Immunol
2011
23
6
462
468
142
Koyama
M
Kuns
RD
Olver
SD
et al
Recipient nonhematopoietic antigen-presenting cells are sufficient to induce lethal acute graft-versus-host disease.
Nat Med
2012
18
1
135
142
143
Naranjo-Gomez
M
Raich-Regue
D
Onate
C
et al
Comparative study of clinical grade human tolerogenic dendritic cells.
J Transl Med
2011
9
1
89
144
Thara
E
Dorff
TB
Pinski
JK
Quinn
DI
Vaccine therapy with sipuleucel-T (Provenge) for prostate cancer.
Maturitas
2011
69
4
296
303
145
Giannoukakis
N
Phillips
B
Finegold
D
Harnaha
J
Trucco
M
Phase I (safety) study of autologous tolerogenic dendritic cells in type 1 diabetic patients.
Diabetes Care
2011
34
9
2026
2032
146
Gonzalez-Rey
E
Chorny
A
Fernandez-Martin
A
Ganea
D
Delgado
M
Vasoactive intestinal peptide generates human tolerogenic dendritic cells that induce CD4 and CD8 regulatory T cells.
Blood
2006
107
9
3632
3638
Sign in via your Institution