Skip to main content
  • Review Article
  • Open access
  • Published:

HDAC Inhibition and Graft Versus Host Disease

Abstract

Histone deacetylase (HDAC) inhibitors are currently used clinically as anticancer drugs. Recent data have demonstrated that some of these drugs have potent antiinflammatory or immunomodulatory effects at noncytotoxic doses. The immunomodulatory effects have shown potential for therapeutic benefit after allogeneic bone marrow transplantation in several experimental models of graft versus host disease (GVHD). These effects, at least in part, result from the ability of HDAC inhibitors (HDACi) to suppress the function of host antigen presenting cells such as dendritic cells (DC). HDACi reduce the dendritic cell (DC) responses, in part, by enhancing the expression of indoleamine 2,3-dioxygenase (IDO) in a signal transducer and activator of transcription-3 (STAT-3) dependent manner. They also alter the function of other immune cells such as T regulatory cells and natural killer (NK) cells, which also play important roles in the biology of GVHD. Based on these observations, a clinical trial has been launched to evaluate the impact of HDAC inhibitors on clinical GVHD. The experimental, mechanistic studies along with the brief preliminary observations from the ongoing clinical trial are discussed in this review.

Introduction

Allogeneic hematopoietic cell transplantation (HCT) is an important therapeutic option for a variety of malignant and nonmalignant conditions. The therapeutic potential of allogeneic HCT relies on the graft-versus-leukemia (GVL) effect, which eradicates residual malignant cells by immunologic mechanisms (1). However, graft versus host disease (GVHD) remains the most frequent and serious complication following allogeneic HCT and limits the broader application of this important therapy. GVHD results from immunologically mediated injury to host tissues (2,3). Consequently, GVHD and GVL reactivity are tightly linked (4). As the number of allogeneic HCT continues to increase, a greater understanding of the pathogenesis of GVHD is being made that may lead to the development of more effective therapies and treatment strategies.

The pathophysiology of GVHD is known to involve donor T-cell interactions with host antigen presenting cells (APCs) and the subsequent induction of proinflammatory cytokines and cellular effectors that cause target organ damage (5). Because host APCs are critical for induction of GVHD by priming donor CD4+ and CD8+ T, targeting host APCs may be a promising strategy to prevent GVHD (6). Clinical observations also support the role of APCs in the development of GVHD and the attractiveness of an approach that targets the role APCs play (7,8).

Acetylation of histones represents one of several epigenetic modifications (9,10). Altering gene expression through chro-matin modifications induced by acetylation and deacetylation of histone tails has gained wide attention (11). Histone deacetylase inhibitors (HDACi) cause reversible inhibition of HDAC enzymes, remodel chromatin, regulate gene expression (12) and have shown efficacy in vitro and in vivo as antitumor agents (1316). Phase I/II clinical trials have demonstrated that HDAC inhibition is well tolerated and suberoylanilide hydroxamic acid (SAHA) or vorinostat is now a Food and Drug Administration (FDA) approved drug (15,16). The immunomodulatory effects of HDAC inhibitors, however, have been largely unrecognized until recently. Burgeoning evidence demonstrates that these agents have potent antiinflammatory effects at noncytotoxic doses and concentration (17,18).

In this review, we discuss the clinical features and pathophysiology of GVHD briefly and discuss the exciting and novel observations pertaining to the immunoregulatory effects of HDACi on GVHD. We summarize our current knowledge of the role of HDACs in the complex regulation of GVHD and GVL, and discuss several other studies offering potential molecular mechanisms of action for HDAC inhibition and prevention of alloresponses. Finally, we describe an ongoing Phase II clinical trial that attempts to translate the preclinical studies on HDAC inhibition and GVHD into a proof-of-concept clinical trial.

Clinical Features of GVHD

GVHD occurs when donor T cells respond to histoincompatible antigens on the host tissues and clinically presents in an acute or chronic form. Historically, acute and chronic forms were defined arbitrarily on the basis of the time frame after transplant. Classically, acute GVHD develops within the first 100 days of transplant or can occur beyond 100 days after transplant with persistent, recurrent or late-onset symptoms. The principle target organs include the skin, liver and GI tract. The signs and symptoms can be characterized by diffuse maculopapular rash (Figure 1), anorexia, profuse diarrhea, nausea, vomiting, ileus and cholestatic hepatitis (Table 1). Despite HLA identity between a patient and donor and the current immunoprophylaxis, about 40% of patients with acute GVHD require treatment with high-dose steroids (1). The incidence of acute GVHD is even higher in patients who received mismatched donor grafts. Chronic GVHD is a complex, multisystem disorder with myriad manifestations that can involve essentially any organ and, typically, is characterized by fibrosis (Table 2) (19). Chronic GVHD may emerge from acute disease (progressive type), develop following a period of resolution from acute disease (quiescent or interrupted type), or occur de novo. Some patients may experience overlap syndrome in which clinical features of acute and chronic GVHD appear together (20). The incidence of chronic GVHD is 60% to 70%, depending on the type of donor (19). Specific signs and symptoms, including erythematous rash, nausea, vomiting, diarrhea and liver dysfunction are shared between the two (Table 2).

Figure 1
figure 1

Acute GVHD of the skin. Photograph courtesy U of Michigan, BMT program.

Table 1 Acute GVHD symptoms.
Table 2 Chronic GVHD symptoms.

Pathophysiology of GVHD

The pathophysiology of GVHD is complex and can be considered as a normal immune response that has gone awry. GVHD also can be considered as a complex immune response that has gone awry and can be understood as a pathway that consists of triggers, sensors, mediators and effectors of GVHD.

Triggers for induction of GVHD

As with all immune responses, certain triggers are critical for induction of acute GVHD.

(a) Disparities between Histocompatibility Antigens. Antigen disparity can be at the level of major histocompatibility complex (MHC), that is, MHC mismatched or at the level of minor histocompatibility antigens (miHA) that is, MHC matched but miHA mismatched. In humans, the MHC gene is on chromosome 6 and encodes the human leukocyte antigens (HLA) (21). The severity of acute GVHD is directly related to the degree of MHC mismatch (22). In bone marrow transplants (BMT) that are MHC matched but miHA disparate, donor T cells still recognize MHC peptide derived from the products of recipient polymorphic genes, the miHAs (2325). The expression of miHAs is wide and variable. Some miHAs such as HA-1, HA-2, HB-1 and BCL2A1 are found primarily on hematopoietic cells, whereas some others such as the H-Y antigens, HA-3, HA-8, and UGT2B17 are ubiquitous (26).

(b) Damage induced by conditioning regimens and underlying diseases. Under most circumstances, the initiation of an adaptive immune response is triggered by the innate immune response. The innate immune system is triggered by certain exogenous and endogenous molecules. This is likely the case in the induction of acute GVHD. Pattern recognition receptors such as Toll-like receptors (TLRs) and nucleotide-binding oligomerization domain containing 2 (NOD2) (27) play an essential role in innate immunity by recognizing conserved damage or pathogen-associated molecular patterns (PAMPs) and initiating the cellular signaling pathways that activate cytokine secretion, such as NF-kB (28,29). The PAMPs such as lipopolysaccharide (LPS), CpG, and MDP2 which are recognized by TLR-4, TLR-9 and NOD2 respectively, are released during the chemotherapeutic and radiotherapeutic conditioning regimens performed before the infusion of BMT donor cells (3034). In this way, the conditioning regimens amplify the secretion of proinflammatory cytokines such as IL-1, TNFα; (31,35,36), IL-6 (37) and other interferon family members in a process described as a 201C;cytokine storm.” In addition to the exogenous microbial associated molecules, endogenous triggers as a consequence of damage, called damage-associated molecular patterns (DAMPs) might also play a critical role in GVHD (29). In fact the proinflammatory cytokines themselves might serve as DAMPs.

Sensors of GVHD

The triggers that initiate an immune response have to be sensed and presented. Antigen presenting cells (APCs) might be considered the sensors for acute GVHD. The APCs sense the DAMPs (i.e., present the MHC-disparate or miHA-disparate protein and provide the critical secondary (costimulatory) and tertiary (cytokine) signals for activation of the alloreactive T cells), the mediators of acute GVHD. APCs sense allodisparity through MHC and peptide complexes. Dendritic cells (DCs) are the most potent APCs and the primary sensors of allodisparity (38). Recipient DCs that have been primed by the conditioning regimen will process and present MHC and peptide complexes to donor T cells at the time of transplant (39). At later time points, donor DCs may take over this role (40,41). In the case of hematopoietic cell transplants (HCT), recipient DCs present the endogenous and the exogenous antigens to donor CD8+ and CD4+ T cells, respectively. There is no predilection for allopeptides to be recognized by either CD4+ or CD8+ mediated presentation. As noted earlier, DCs are important initiators of GVHD. The role of DC subsets in GVHD is just beginning to be understood (4244). However, the kinetics of the switch from recipient to donor APCs, the contributions of different APC subsets, the importance of direct alloantigen presentation, and the magnitude of indirect alloantigen presentation in GVHD remain to be determined.

APCs provide the critical costimulation signals for turning on the acute GVHD process. The interaction between the MHC/allopeptide complex on APCs and the TCR of donor T cells along with the signal via T-cell costimulatory molecules and their ligands on APCs is required to achieve T-cell activation, proliferation, differentiation, and survival (45,46) and the in vivo blockade of positive costimulatory molecules (such as CD28, ICOS, CD40, CD30 and so on) (4752), or inhibitory signals (such as PD-1 and CTLA-4) mitigate or exacerbate acute GVHD respectively (53).

As mentioned above, the inflammatory cytokines and DAMP ligands released during pretransplant conditioning regimens act as a third signal to enhance recipient APC and donor T-cell interactions. In addition, various modulations of APCs can influence GVHD development. Recent data show that exposure to granulocyte colony-stimulating factor (G-CSF) shortly after HCT, in combination with a TBI-conditioning regimen, significantly worsened GVHD in mice (54). Histone deacetylase inhibitors such as suberonylanilide hydroxamic acid (SAHA) and ITF 2357 have been shown to reduce development of GVHD in murine models by modulating host DC functions (5557) (discussed below).

Mediators of GVHD. These primarily include the donor T-cell subsets and the donor NK cells. Evidence suggests that alloreactive donor T cells consist of several subsets with different stimuli responsiveness, activation thresholds, and effector functions. The alloantigen composition of the host determines which donor T-cell subsets differentiate and proliferate. As mentioned previously, in the majority of HLA-matched HCT, acute GVHD may be induced by either or both CD4+ and CD8+ subset responses to MiHAs (58). The repertoire and immunodominance of the GVHD-associated peptides presented by MHC class I and class II molecules has not been defined (59). Donor naïve CD62L+ T cells are the primary alloreactive T cells that drive the GVHD reaction, while the donor effector memory CD62L T cells do not (60,61). Interestingly, donor Tregs expressing CD62L are also critical to the regulation of GVHD (62,63). We now know that it is possible to modulate the alloreactivity of naïve T cells by inducing anergy with costimulation blockade, deletion via cytokine modulation or mixed chimerism. Donor effector memory T cells that are nonalloreactive do not induce GVHD, yet are able to transfer functional memory (60) and mediate GVL (64). In addition, lymphopenia-induced proliferation gives rise to cells that are like memory T cells and enhance the graft-versustumor effect after donor leukocyte injection (DLI) (65). In contrast, memory T cells that are alloreactive can cause severe GVHD (6668).

GVHD is regulated negatively by regulatory T cells (Tregs). Distinct subsets of Tregs exist: the naturally occurring CD4+ CD25+ Tregs that express the Forkhead Box Protein P3 (FOXP3), CD4+ CD25IL10+ Tr cells, γδ T cells, double negative (DN) T cells, and NKT cells (6974). In mouse BMT models, naturally occurring donor-derived Tregs suppress the proliferation of conventional T cells, prevent GVHD and preserve GVL effects depending upon the ratio of effector T cells to Tregs (7580). Furthermore, viral immunity is preserved in the presence of Tregs after allogeneic HCT (81). Mechanisms that enhance Treg numbers and function might therefore be very effective in enhancing alloBMT. HDACi have been shown to have such salutary effects of natural Tregs (discussed further below). In addition, based on the dominant cytokines that are produced upon activation, T cells can be distinguished into various subsets such as Th1, Th2 and Th17 cells. The Th1 cytokines (IFN-γ, IL-2 and TNF-α;) have been implicated in the pathophysiology of acute GVHD (8284). IL-2 production by donor T cells remains the main target of many current clinical therapeutic and prophylactic approaches, such as cyclosporine, tacrolimus and monoclonal antibodies (mAbs) against the IL-2 and its receptor to control acute GVHD (85,86). But emerging data indicate an important role for IL-2 in the generation and maintenance of CD4+CD25+ Foxp3+ Tregs, suggesting that prolonged interference with IL-2 may have an unintended consequence in the prevention of the development of long-term tolerance after allogeneic HCT (8790). Furthermore the role of Th1/Th2 and Th17 cytokines is complex and might be model dependent (91103). Moreover these cells are required for the GVL effects.

Donor natural killer (NK) cells which are inhibited by recognition of class I alleles on target cells via their killer cell immunoglobulin-like receptors (KIR) are emerging as key effectors in the GVH process. They have been shown to specifically downregulate host APC-mediated activation of alloreactive T cells perhaps by directly killing APCs without losing the beneficial effects (104106).

Effectors and Amplifiers of GVHD

The effector phase that leads to GVHD target organ damage is a complex cascade that involves cytolytic cellular effectors such as CD8 CTLs, CD4 T cells, NK cells and inflammatory molecules such as IL-1b, TNFα, IFNγ and reactive oxygen species. The cellular effectors require cell-to-cell contact to kill the cells of the target tissues via activation of perforingranzyme, Fas-FasL (CD95-CD95L), or TNFR-TRAIL pathways. Other CTL-killing mechanisms such as TWEAK, and LT β/LIGHT pathways also have been implicated in GVHD (107114). It is important to note that CTL pathways are essential for GVL effects as well. Inflammatory pathways, by contrast, based on animal models, do not require cell-to-cell contact to kill target cells and are not particularly critical of GVL. GVHD damage by the cellular effectors is amplified by these inflammatory mediators including IFNγ produced by T cells, TNFα (115) and IL-1 (116) produced by T cells and monocytes/macrophages, and nitric oxide (NO) produced by monocytes/macrophages (117,118).

All of the above aspects of the biology of acute GVHD may be summarized in a cyclical three-step model: (step 1) conditioning regimen-related damage and the release of DAMPs such as LPS, (step 2) donor T-cell proliferation and (step 3) target organ damage by effectors (Figure 2). While this allows for accessing the biology of GVHD, it is important to note that GVHD is a complicated systemic process with many unknowns and is not a simplified, linear or cyclical process. Nonetheless, based on our current understanding, agents that reduce inflammatory cytokines such as TNF and IL-1, but spare T-cell CTL functions and enhance donor Tregs and NK cell functions may be ideal for reducing GVHD without compromising GVL significantly. Experimental data suggests that HDACi may be able to provide such an effect (see Figure 3 and discussion).

Figure 2
figure 2

Pathophysiology of GVHD. Three phases of GVHD pathophysiology. From: Reddy P, Ferrara JLM. (2009 Feb 28) Mouse models of graft-versus-host disease. In: StemBook [Internet]. Cambridge (MA): Harvard Stem Cell Institute; 2008-. Available at: https://doi.org/www.stembook.org/node/548

Figure 3
figure 3

The regulation of immune cells by histone deacetylase inhibitors (HDACi). HDACi have direct and indirect effects on various immune cellular subsets: HDACi play an important role in the negative regulation of APCs, reduce the secretion of inflammatory cytokines, increase the numbers and function of naturally occurring regulatory T cells (Tregs), and activate natural killer (NK) cell-mediated activity

Histone Deacetylase Inhibitors (HDACi)

Histones are major structural proteins that package DNA into chromatin and play an important role in gene regulation. DNA wraps around a histone octamer composed of histones H2A, H2B, H3 and H4 to form a nucleosome and the histone H1 links the octameric core into chromatin. Covalent modification on the amino terminal of the core histones through methylation, ADP-ribosylation, phosphorylation, ubiquitylation and acetylation (10,119,120) affect nuclear replication, chromatin assembly and transcription (121123), and thus provide insight into the epigenetic regulation of gene expression (10,124).

Histone acetylation is tightly regulated by the balance of histone acetyltransferases (HATs) and histone deacetylases (HDACs). HAT enzymes, which now include more than 20 members (125), act by acetylating specific lysine residues of the histone components of chromatin while HDACs deacetylate the lysine residues. HDACs comprise a family of 18 genes subdivided into four distinct classes: Class I (HDAC1, 2, 3, and 8), class II (HDAC5, 6, 7, 9, and 10), and class IV (HDAC 11) share sequence similarity and require Zn+ dependent enzymatic activity (126128). Class III is a structurally unrelated NAD+ dependent subfamily and belongs to the Sirtuin family (127). Much research on these enzymes has focused on their ability to modulate acetylation of histones and the regulation of chromatin (129,130). Emerging data demonstrate that HDACs can also target nonhistone cellular proteins (127). It is now becoming increasingly known that acetylation of several nonhistone proteins by the HATs and HDACs is an important posttranslational modification that regulates their function, stability, protein-protein/proteinDNA interactions, signaling and functions (131) and that disruption in the balance of acetylation and deacetylation affects a broad range of human disorders, including oncogenesis and immune dysfunction (132). However, not all HDACs are expressed in all cells; even in those that express them, their location, target proteins and functions might thus vary. Thus, the specificity of different HDACs and their nonhistone proteins, and more importantly, the consequences of targeting specific HDACs in modulating cellular growth, differentiation and immune responses are poorly understood.

However, HDAC inhibitors have emerged as an important class of anticancer agents (133135). HDACi are diverse and can be divided into six classes based on their chemical structure, which include hydroxamic acid derivatives, carboxylates, benzamides, electrophilic ketones, cyclic peptides and miscellaneous compounds (136,137). These agents inhibit the enzymatic activity of primarily class I and II HDACs with varying efficiency (126,138,139), thereby causing increased histone acetylation and gene transcription. Two of them, SAHA and ITF 2357, are hydroxamic-containing agents, and the former was approved by the FDA for treatment of cutaneous T-cell lymphoma (135,140,141). The HDACi, including SAHA and ITF 2357, have differential effects on various zinc-dependent HDAC enzymes, that is, class I and II HDACs (18,142). Thus, the specific HDACs that are critical for the various biological and clinical effects observed upon treatment with HDACi are not known (126). While a large range of different HDACi have been studied and developed for cancer therapy, we and others have demonstrated that HDACi at lower and noncytotoxic concentrations possess a novel and potent antiinflammatory and immunoregulatory effect (17,18). Emerging data from multiple laboratories demonstrate that HDACi can suppress several inflammatory and immune-mediated diseases such as lupus, sepsis, inflammatory bowel disease, rheumatoid arthritis, autoimmune diabetes, allograft tolerance and GVHD in preclinical models (17,18,55,56,143148).

Several of these are discussed in accompanying articles. Here we will focus on GVHD and discuss potential mechanisms of regulation by HDACi (Figure 3).

Impact of HDAC Inhibition on Experimental GVHD

Insights into the cellular and molecular pathogenesis of GVHD implicate proinflammatory cytokines and host APCs, such as DCs, as important targets for reducing GVHD (3,149). SAHA or ITF 2357 are such agents that are currently in clinical trials for treatment of cancers (150). Micromolar concentrations of SAHA are required for antitumor effects, whereas nanomolar concentrations of SAHA reduce the secretion of inflammatory cytokines such as TNF-α, IFN-γ, IL-1β and IL-12 (17,143). Given the antiinflammatory properties of these agents, and based on the central role of proinflammatory cytokines in the pathogenesis of acute GVHD, we investigated the role of SAHA in a well-characterized murine model of allogeneic HCT. SAHA or ITF 2357 were administered during the amplification of the proinflammatory cascade early in the time course of transplant without interrupting the initial donor T-cell interaction with host APCs (35,151,152). SAHA significantly reduced serum levels of TNF-α, IL-1, and IFN-γ after alloBMT (55). Furthermore, this reduction in the proinflammatory cytokines was associated with a reduction in the GVHD mortality and GVHD-specific target organ damage in multiple murine models (55). SAHA administration following allogeneic HCT did not affect donor T-cell responses to host antigens as determined by their proliferative and CTL responses (55). In addition, the inhibition of cytokines by SAHA temporally correlated with enhanced acetylation of histone H3 and was associated with the downregulation of TNF-α and IFN-γ mRNA after allogeneic HCT (55). Thus the reduction in GVHD likely was primarily due to the inhibition of the inflammatory cytokines and not due to a direct effect on donor T-cell responses in these models.

It is often challenging to separate the toxicity from GVHD with the beneficial GVL effects, a well-recognized and potent form of immunotherapy for malignancies (1). While inflammatory cytokines contribute to the toxicity of GVHD, they have a more limited role in the eradication of residual leukemia, which is primarily mediated by donor CTLs and NK cells (76,95,153155). Consistent with the preservation of donor T-cell functions, we have found that SAHA administration led to the disruption of inflammatory cytokine cascades, but maintained CTLs, thereby attenuating GVHD mortality and preserving GVL effects and improving leukemia-free survival (55). By contrast, the syngeneic animals that received SAHA did not eliminate the tumor completely, demonstrating the requirement of GVL for tumor eradication. These observations were confirmed in additional tumor and alloBMT murine models, thus ruling out any tumor- or model-specific artifacts. Thus, the maintenance of donor T-cell responses to host antigens after SAHA treatment also preserved the beneficial GVL effect in multiple mouse models of allogeneic HCT. Similar observations on GVHD reduction were made by other groups in other different models (148).

HDAC inhibition modulates the function of APCs. DCs serve as the sentinels of the immune response and function as the most potent APCs (156). They initiate innate immune responses primarily through PRRs, and shape adaptive immunity through the modulation of T-cell responses (157). Because host APCs are critical for the induction of alloresponses and are the major sources of proinflammatory cytokines, following our initial observation that SAHA administration suppressed proinflammatory cytokines and reduced GVHD (55), we investigated the effect of SAHA and ITF 2357 on the function of DCs (56). Bone marrow-derived DCs treated with SAHA or ITF 2357 and then stimulated with TLR agonists such as LPS or other TLR ligands (lipoteichoic acid, peptidoglycan, dsRNA poly[IC] and CpG DNA) (158,159), secreted significantly reduced amounts of proinflammatory cytokines such as IL-1b, TNF-α, IL-12, and IL-6 in a dose-dependent manner (56). DCs treated with SAHA and ITF 2357 also demonstrated reduced in vitro allostimulatory responses. This was due to decreased proliferation of the allogeneic T cells and not a consequence of enhanced apoptosis. More importantly, despite the reduction in proliferation of the T cells, their CTL functions were preserved against the allotargets. Consistent with the decreased in vitro alloproliferative responses and reduced amounts of cytokines, when the host type DCs were treated with SAHA and infused in the alloBMT recipients early after BMT, they reduced both CD4 and CD8 driven GVHD (56). The reduction in GVHD was once again associated with reduced levels of proinflammatory cytokines (56).

HDACi induce IDO and regulate DCs. Indoleamine 2,3-dioxygenase (IDO) is an intracellular enzyme that degrades tryptophan, an amino acid that is essential for T-cell activation (160). Treatment of DCs with SAHA increased IDO expression at the protein and mRNA levels (56). The increase in IDO expression was associated with histone (H4) acetylation in the IDO promoter region (56). Utilizing three complementary approaches, siRNA, pharmacologic inhibition by 1-MT, and genetically deficient IDO-/- mice, the importance of IDO induction in the DCs by HDACi was investigated. IDO-specific siRNA silenced the mRNA expression of IDO in the SAHA-treated DCs and significantly reversed the suppression of the proinflammatory cytokine, TNF-α, upon LPS stimulation. Likewise, LPS-stimulated DCs from IDO-/- animals or those treated with 1-MT also demonstrated the loss of suppression of proinflammatory cytokine secretion by HDACi. Consistent with our data from murine BM DCs, HDACi also reduced the innate and allostimulatory responses of DCs derived from healthy human volunteers (56). Furthermore, when bone marrow chimeras generated by utilizing IDO-deficient animals (IDO-/- → B6) (such that only host hematopoietic-derived APCs were incapable of generating IDO), were used as recipients in alloBMT, these animals were not protected by administration of SAHA or ITF 2357, demonstrating a loss of HDACi-induced reduction in GVHD. These data indicate a key role for IDO induction by host APCs in the HDACi induced GVHD protection.

STAT-3 is necessary for induction of IDO by HDACi. The critical pathways responsible for the induction of IDO following treatment with HDACi was further dissected. Signaling via Janus kinase (JAK)-signal transducer and activator of transcription (STAT) pathways positively and negatively regulate all cell types involved in immune responses (161,162). There are seven STAT transcription factors (161) and while each of the STAT proteins have distinct and overlapping functions, STAT-3 is critical for negative regulation of proinflammatory cytokine secretion by monocyte/DCs but for enhancing T-cell function (163). Data from lethal STAT-3 knock-out mice (164), tumor immunotherapy models (165167), and, more importantly, humans with loss of function mutations of STAT-3 (Hyper IgE syndrome patients) show enhanced inflammatory phenotype (168170), demonstrating an essential role for STAT-3 in suppressing immune responses. These findings indicate that STAT-3 plays a critical role in negative regulation of DCs (171). Posttranslational modification of STAT-3 either by phosphorylation and/or acetylation activates its functions (172178). We therefore, reasoned that HDACi might activate STAT-3 by acetylation and that may be critical for induction of IDO and regulation of DCs (174). STAT-3 was acetylated following SAHA-or ITF 2357-treatment of DCs. Although HDACi acetylated STAT-3 it did not alter its phosphorylation status (174). Furthermore, when the effects of SAHA and ITF 2357 were tested on the induction of IDO in cell lines expressing pcDNA3 empty vector (STAT-3 null), wild type STAT-3, and STAT-3 mutantK685R (that contains Lys685-to-Arg substitution and therefore cannot be acetylated-K685R), HDAC inhibition enhanced IDO expression in the WT STAT-3 transfected cells but not in the null control or the acetylation resistant STAT-3 mutantK685R. These data suggest a critical role for STAT-3 acetylation in the induction of IDO. The relevance of STAT-3 in altering DC function by HDACi was evaluated further by utilizing a drug that disrupts STAT-3 DNA complex formation, JSI-124 specifically (179). Murine DCs, when treated with JSI-124 and then conditioned with SAHA and ITF 2357, did not show suppression of LPS-induced secretion of proinflammatory cytokines such as TNF-α secretion or a reduction in allogeneic T-cell proliferation when compared with diluents treated DCs. Thus, the disruption of STAT-3 activity with JSI-124 mitigated the suppressive effects of HDAC inhibition on DCs. Other studies have demonstrated that STAT3 acetylation by the HAT CBP has been correlated with increased DNA-binding and transactivation activity (174,180,181,182). Conversely, deacetylation of STAT3 by the HDAC Sirtuin 1 correlates with decreased STAT3 tyrosine phosphorylation and activity (183). Nonetheless, collectively, these data demonstrate that acetylation of STAT-3 is necessary for its activation and for the regulation of DCs. However, whether acetylation alone is sufficient in the absence of phosphorylation remains to be investigated. These and other potential effects of HDACi on DCs are summarized in Figure 4.

Figure 4
figure 4

The regulation of DCs by histone deacetylase inhibitors (HDACi).

Specific HDAC enzymes in the regulation of DCs. HDACi mediated suppression of DCs demonstrate the overall impact on DCs from global suppression of class I/II HDACs. The role of specific HDAC enzymes in the regulation of DCs is being evaluated currently. To this end, recent studies by Sotomayor and colleagues have evaluated the impact of HDAC11 and HDAC6 on DCs (184,185). HDAC11 is a newly characterized member of the HDAC family (186). Villagra et al. (184), showed that HDAC11 regulates the expression of IL-10 negatively in mouse and human APCs (DCs and macrophages), primarily by interacting with the distal segment of the promoter of the gene encoding this cytokine. IL-10 is an antiinflammatory cytokine that is an important mediator in influencing the function of APCs at the site of antigen encounter, and, thus, serves a key role in tolerance induction and regulation of inflammation (187189). Overexpression of HDAC11 abrogated the expression of IL-10 mRNA in LPS-treated macrophages. When HDAC11 was “knocked down” by the transduction of primary mouse macrophages with short hairpin RNA, LPS stimulation resulted in higher expression of IL-10 mRNA. These findings were confirmed in additional experiments using two macrophage cell lines (derived from RAW264.7) lacking HDAC11 expression. Furthermore, in RAW264.7 cells transfected with an enzymatically inactive mutant HDAC11 with deletion of its deacetyltransferase domain, demonstrated increased expression of IL-10 mRNA, suggesting that intact deacetylase activity was required for HDAC11-mediated inhibition of IL-10 in APCs. APCs overexpressing or lacking HDAC11 altered CD4+ T-cell proliferation. Specifically, overexpression of HDAC11 in APCs activated naïve antigen-specific CD4+ T cells and restored the responsiveness of tolerant T cells, whereas APCs lacking HDAC11 functionally impaired CD4+ T-cell proliferation and they produced less IL-2 and IFN-γ. More recently, Dubovsky et al. (185) report that overexpression of HDAC6 induced transcriptional activation of IL-10 gene expression, the opposite effect seen with HDAC11. These data demonstrate the role of specific HDACs, HDAC11 and HDAC6, and the inflammatory response of APCs. They demonstrate that, in contrast to global inhibition of HDACs, the inhibition of specific HDACs might lead to a distinct and/or opposite effect on DC responses.

Nonetheless, they collectively demonstrate that HDACs could serve as potential therapeutic targets for influencing APC/DC-mediated immune responses.

HDAC inhibition and regulatory T-cell (Tregs) function. As noted above, donor Tregs reduce GVHD, but do not diminish GVL significantly after alloBMT. Recently, Tao et al. (147) reported that HDACi expands in vivo Treg cell population and also increased the activity of these cells. Several mouse models of alloimmunity and autoimmunity were studied for the in vivo analysis of HDAC tivating gene-2 (Rag2)-deficient mouse model and two adoptive transfer models. In each model, HDAC inhibition increased the absolute numbers and proportion of Treg cells, primarily in the CD4+ CD25+ Foxp3+ T-cell subset. When the effects of HDACi therapy were evaluated in vivo, there was increased expression and acetylation of FOXP3+ and also an increase in the Treg associated genes, such as CTLA4 and GITR, while IL-2 was repressed. Thus, the expression of multiple Treg-associated genes was increased with HDACi. HDAC inhibition promoted acetylation of histones in Treg cells and increased acetylation on several lysines in the forkhead domain of FOXP3+. When these lysines were mutated, FOXP3+ could not repress IL-2 expression and was less able to suppress conventional T-cell activity in vitro, demonstrating that the enhanced function of Tregs is in part due to direct targeting (acetylation) of nonhistone protein Foxp3. They also demonstrate that HDAC9, expressed in higher amounts in Treg cells than conventional T cells, was critical for modulating Tregs. They further demonstrated that the beneficial effects of HDACi on allograft rejection and IBD models. The direct impact of HDACi on donor Tregs after alloBMT is under active investigation currently by our group. Nonetheless, data extrapolated from Tao et al.’s observation suggest that HDACi might also have Treg enhancing effects after alloBMT and that this may be another potential GVHD protective effect of HDACi.

HDAC inhibition on NK-cell function and tumor immunogenicity. Donor NK cells reduce GVHD by eliminating host APCs while promoting GVL by direct elimination of the host tumors. Acetylation and deacetylation also may play an important role in NK-cell activity (186). HDAC inhibition with SAHA treatment has been reported to increase the functional expression of NK cell-mediated killing through NKG2, member D (NKG2D) ligands including MHC class I-related chain A and B (MICA/B) in Jurkat T-cell leukemia, thereby making them more sensitive to NK cell-mediated lyses (190). The effect of HDACi on NK-cell activity also was investigated by Armeanu et al. (191) who showed that treatment of human hepatocellular carcinoma cells with the HDACi sodium valproate (VPA) mediated the lyses of malignant cells via NKG2D expressed on cytotoxic lymphocytes. VPA induced the transcription of MICA/B in hepato-cellular carcinoma cells, which led to increased cell surface expression, followed by lyses of the cancer cells. These data support a role for HDACi in stimulating NK cell-mediated activity, which may contribute to antitumor immune responses while regulating GVHD. This, however, remains to be demonstrated directly in experimental GVHD models.

Skov et al. (190) demonstrated enhanced NK cell-mediated killing on multiple types of cancer cells following HDACi treatment, which included B-cell acute lymphoblastic leukemia, acute myelogenous leukemia, multiple myeloma, malignant non-Hodgkin lymphoma, T-cell acute lymphoblastic leukemia, mantle cell lymphoma, multiple myeloma, epithelial breast adenocarcinoma, epithelial cervix adenocarcinoma, and epithelial colorectal adenocarcinoma. This was consistent with other studies, which have shown a range of different cancer types characterized by constitutive expression of MICA/B (192194). Interestingly, Skov et al. (190) showed that two other cancer cell lines tested did not respond to HDACi treatment by increased MICA/B expression. Therefore, the molecular basis for the selective expression of MICA/B on different cancer cells by HDACi treatment remains unknown. In any event, another study demonstrated that HDACi enhanced the immune susceptibility of two specific forms of primary human acute myeloid leukemia (195). HDACi also induced apoptosis of leukemic blasts AML expressing the PML-RAR or AML1-ETO oncoproteins, independent of p53, through activation of a specific death receptor pathway (TRAIL and Fas signaling pathways) (196). Collectively, these results suggest that HDACi regulate GVHD by modulating proinflammatory cytokine secretion, host APC function and perhaps by enhancing Tregs. However, they still may preserve GVL by preservation of donor T-cell CTL functions, increasing NK reactivity and the immunogenicity of the tumor cells.

The impact of HDAC inhibition on other allograft models. The effects of HDACi on other allograft models, including rat and canine transplant models, also have been investigated (197,198). FR276457, a hydroxamic derivative HDAC inhibitor, was shown to prevent allograft rejection in a rat cardiac transplant model. When administered alone as monotherapy, the drug demonstrated strong efficacy and demonstrated dramatic allograft survival when used in combination with tacrolimus. Another HDAC inhibitor, FR235222, also prolonged graft survival in rat cardiac transplant model (199). HDACi therapy allograft survival of rapamycin in murine cardiac and islet cell transplant models (40). The data support the role for HDACi as potential therapeutic agents in mitigating alloresponses after BMT and solid organ allografting and, perhaps, can be used as useful adjuncts to current standard immunoprophylaxis drugs such as the CNI and mTOR inhibitors.

Ongoing Translation of HDAC Inhibition for GVHD Prevention

The literature reviewed herein suggests that HDACi have direct and indirect effects on various immune cellular subsets, depicted in Figure 3. HDACi play an important role in the negative regulation of APCs (56), reduce the secretion of inflammatory cytokines, such as TNF-α, IFN-γ, IL-1β and IL-12 (17,55,143), increase the numbers and function of natural CD4+ CD25+ FOXP3+ Tregs (147,200), and activate NK cell-mediated activity (190,191). HDAC inhibition reduced GVHD and preserved GVL in murine models (55) and regulated both murine and human APCs (57,147,200,201). The HDACi, SAHA and ITF 2357 thus appear to exhibit the desirable regulatory effects on multiple mediators of GVHD (that is, inhibit proinflammatory secretion, host APCs but promote donor Treg and NK-cell responses) (3),202, see Figure 3). In addition, the pharmacokinetics of oral ITF2357 recently has been studied (203) and both oral SAHA (vorinostat) and ITF2357 (givinostat) have good safety profiles in humans.

Therefore, given these preclinical observations and the good therapeutic index of the oral preparations of some of these agents, such as SAHA and ITF2357, a clinical trial has been launched at the University of Michigan and at Washington University to test the concept that deacetylase inhibition, when used as an adjunct with standard prophylaxis with CNI, will reduce the incidence and severity of acute GVHD after reduced intensity conditioning (RIC) in a matched related donor allogeneic HCT. This trial is based on the institutional experience of GVHD with RIC regimens. Prior experience with this approach showed a 42% incidence of grades II-IV GVHD, with a 50% two-year survival rate (204). The trial is now built on this experience to evaluate whether adding HDACi, SAHA (vorinostat) will reduce the incidence of grade II-IV GVHD to 25%. It is an open label, nonrandomized, Phase II clinical trial using the same RIC regimen (fludarabine and busulfan) that has been utilized for approximately 10 years at our institution. The GVHD prophylaxis backbone consists of tacrolimus (day 3 to day 56, followed by a taper over 4 months) and mycophenolate mofeitil (day 0 to day 28). Vorinostat currently is being administered orally from day 10 to day 100 at 100 mg twice daily. So far, 20 patients have been treated on this trial. Because SAHA has not been tested previously in this setting, it was started at 100 mg twice daily based on clinical data demonstrating that this dose of induced acetylation in circulating PBMCs. Because the 100 mg dose appeared safe in the first cohort of 10 patients, the dose was escalated to 200 mg twice daily in an attempt to enhance efficacy. However, even though no dose limiting toxicities were reached on the 200 mg twice daily dose, there appeared to be a greater incidence of thrombocytopenia on the increased dose. Therefore the study dose has been deescalated back to the 100 mg twice daily after 19 patients, and this dosing will be used for the remainder of the study. Thus far, all 20 patients on the trial have engrafted successfully. There have been no dose-limiting toxicities. No serious adverse events related to drugs, including myelosuppression, liver or kidney toxicities have been observed in this early cohort of patients. Only four patients have developed grade 2 GI GVHD, and in all four of these patients, GVHD resolved with systemic and topical therapy. To our knowledge, this is the first human study of HDAC inhibition in allogeneic BMT patients. If successful, it could lead to the development of an entirely new class of immunomodulatory therapy for GVHD, and perhaps for immune/inflammatory diseases. Alternatively, even if the primary endpoint of the clinical trial is not met, this study will generate data that likely will allow for new lines of laboratory investigation that may foster a better understanding of the biology and role of HDACs in GVHD and immune responses.

Disclosures

The authors declare that they have no competing interests as defined by Molecular Medicine, or other interests that might be perceived to influence the results and discussion reported in this paper.

References

  1. Appelbaum FR. (2001) Haematopoietic cell transplantation as immunotherapy. Nature. 411:385–9.

    Article  CAS  PubMed  Google Scholar 

  2. Blazar BR, Murphy WJ. (2005) Bone marrow transplantation and approaches to avoid graft-versus-host disease (GVHD). Philos. Trans. R. Soc. Lond. B. Biol. Sci. 360:1747–67.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Welniak LA, Blazar BR, Murphy WJ. (2007) Immunobiology of allogeneic hematopoietic stem cell transplantation. Annu. Rev. Immunol. 25:139–70.

    Article  CAS  PubMed  Google Scholar 

  4. Nash RA, et al. (2000) Phase III study comparing methotrexate and tacrolimus with methotrexate and cyclosporine for prophylaxis of acute graft-versus-host disease after marrow transplantation from unrelated donors. Blood. 96:2062–8.

    CAS  PubMed  Google Scholar 

  5. Antin JH, Ferrara JL. (1992) Cytokine dysregulation and acute graft-versus-host disease. Blood. 80:2964–8.

    CAS  PubMed  Google Scholar 

  6. Duffner UA, et al. (2004) Host dendritic cells alone are sufficient to initiate acute graft-versus-host disease. J. Immunol. 172:7393–8.

    Article  CAS  PubMed  Google Scholar 

  7. Mohty M. (2007) Dendritic cells and acute graft-versus-host disease after allogeneic stem cell transplantation. Leuk. Lymphoma. 48:1696–701.

    Article  CAS  PubMed  Google Scholar 

  8. Nachbaur D, Kircher B. (2005) Dendritic cells in allogeneic hematopoietic stem cell transplantation. Leuk. Lymphoma. 46:1387–96.

    Article  CAS  PubMed  Google Scholar 

  9. Esteller M. (2007) Cancer epigenomics: DNA methylomes and histone-modification maps. Nat. Rev. Genet. 8:286–98.

    Article  CAS  PubMed  Google Scholar 

  10. Jenuwein T, Allis CD. (2001) Translating the histone code. Science. 293:1074–80.

    Article  CAS  PubMed  Google Scholar 

  11. Villagra A, Sotomayor EM, Seto E. (2010) Histone deacetylases and the immunological network: implications in cancer and inflammation. Oncogene. 29:157–73.

    Article  CAS  PubMed  Google Scholar 

  12. Johnstone RW. (2002) Histone-deacetylase inhibitors: novel drugs for the treatment of cancer. Nat. Rev. Drug. Discov. 1:287–99.

    Article  CAS  PubMed  Google Scholar 

  13. Marks PA, Miller T, Richon VM. (2003) Histone deacetylases. Curr. Opin. Pharmacol. 3:344–51.

    Article  CAS  PubMed  Google Scholar 

  14. Richon VM, O’Brien JP. (2002) Histone deacetylase inhibitors: a new class of potential therapeutic agents for cancer treatment. Clin. Cancer Res. 8:662–4.

    PubMed  Google Scholar 

  15. Kelly WK, et al. (2003) Phase I clinical trial of histone deacetylase inhibitor: Suberoylanilide hydroxamic acid administered intravenously. Clin. Cancer Res. 9:3578–88.

    CAS  PubMed  Google Scholar 

  16. Kelly WK, et al. (2005) Phase I study of an oral histone deacetylase inhibitor, suberoylanilide hydroxamic acid, in patients with advanced cancer. J. Clin. Oncol. 23:3923–31.

    Article  CAS  PubMed  Google Scholar 

  17. Leoni F, et al. (2002) The antitumor histone deacetylase inhibitor suberoylanilide hydroxamic acid exhibits antiinflammatory properties via suppression of cytokines. Proc. Natl. Acad. Sci. U. S. A. 99:2995–3000.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Leoni F, et al. (2005) The histone deacetylase inhibitor ITF2357 reduces production of proinflammatory cytokines in vitro and systemic inflammation in vivo. Mol. Med. 11:1–15.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Pavletic SZ, et al. (2005) Prognostic factors of chronic graft-versus-host disease after allogeneic blood stem-cell transplantation. Am. J. Hematol. 78:265–74.

    Article  PubMed  Google Scholar 

  20. Filipovich AH, et al. (2005) National Institutes of Health consensus development project on criteria for clinical trials in chronic graft-versus-host disease: I. Diagnosis and staging working group report. Biol. Blood Marrow Transplant. 11:945–56.

    Article  PubMed  Google Scholar 

  21. Petersdorf EW, Malkki M. (2006) Genetics of risk factors for graft-versus-host disease. Semin. Hematol. 43:11–23.

    Article  CAS  PubMed  Google Scholar 

  22. Flomenberg N, et al. (2004) Impact of HLA class I and class II high-resolution matching on outcomes of unrelated donor bone marrow transplantation: HLA-C mismatching is associated with a strong adverse effect on transplantation outcome. Blood. 104:1923–30.

    Article  CAS  PubMed  Google Scholar 

  23. Den Haan JM, et al. (1995) Identification of a graft versus host disease-associated human minor histocompatibility antigen. Science. 268:1476–80.

    Article  Google Scholar 

  24. Goulmy E, et al. (1996) Mismatches of minor histocompatibility antigens between HLA-identical donors and recipients and the development of graft-versus-host disease after bone marrow transplantation. N. Engl. J. Med. 334:281–5.

    Article  CAS  PubMed  Google Scholar 

  25. Murata M, Warren EH, Riddell SR. (2003) A human minor histocompatibility antigen resulting from differential expression due to a gene deletion. J. Exp. Med. 197:1279–89.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Bleakley M, Riddell SR. (2004) Molecules and mechanisms of the graft-versus-leukaemia effect. Nat. Rev. Cancer. 4:371–80.

    Article  CAS  PubMed  Google Scholar 

  27. Inohara N, Nunez G. (2003) NODs: intracellular proteins involved in inflammation and apoptosis. Nat. Rev. Immunol. 3:371–82.

    Article  CAS  PubMed  Google Scholar 

  28. Medzhitov R. (2007) Recognition of microorganisms and activation of the immune response. Nature. 449:819–26.

    Article  CAS  PubMed  Google Scholar 

  29. Chen GY, Tang J, Zheng P, Liu Y. (2009) CD24 and Siglec-10 selectively repress tissue damage-induced immune responses. Science. 323:1722–5.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Cooke KR, et al. (1998) Tumor necrosis factor-alpha production to lipopolysaccharide stimulation by donor cells predicts the severity of experimental acute graft-versus-host disease. J. Clin. Invest. 102:1882–91.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Hill GR, Ferrara JL. (2000) The primacy of the gastrointestinal tract as a target organ of acute graft-versus-host disease: Rationale for the use of cytokine shields in allogeneic bone marrow transplantation. Blood. 95:2754–9.

    CAS  PubMed  Google Scholar 

  32. Taylor PA, et al. (2008) TLR agonists regulate alloresponses and uncover a critical role for donor APCs in allogeneic bone marrow rejection. Blood. 112:3508–16.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Holler E, et al. (2006) Prognostic significance of NOD2/CARD15 variants in HLA-identical sibling hematopoietic stem cell transplantation: effect on long-term outcome is confirmed in 2 independent cohorts and may be modulated by the type of gastrointestinal decontamination. Blood. 107:4189–93.

    Article  CAS  PubMed  Google Scholar 

  34. Holler E, et al. (2004) Both donor and recipient NOD2/CARD15 mutations associate with transplant-related mortality and GvHD following allogeneic stem cell transplantation. Blood. 104:889–94.

    Article  CAS  PubMed  Google Scholar 

  35. Hill GR, et al. (1997) Total body irradiation and acute graft-versus-host disease: The role of gastrointestinal damage and inflammatory cytokines. Blood. 90:3204–13.

    CAS  PubMed  Google Scholar 

  36. Xun CQ, Thompson JS, Jennings CD, Brown SA, Widmer MB. (1994) Effect of total body irradiation, busulfan-cyclophosphamide, or cyclophosphamide conditioning on inflammatory cytokine release and development of acute and chronic graft-versus-host disease in H-2-incompatible transplanted SCID mice. Blood. 83:2360–7.

    CAS  PubMed  Google Scholar 

  37. Chen X, et al. (2009) Blockade of interleukin-6 signaling augments regulatory T cell reconstitution and attenuates the severity of graft versus host disease. Blood. 114:891–900.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Banchereau J, Steinman RM. (1998) Dendritic cells and the control of immunity. Nature. 392:245–52.

    Article  CAS  PubMed  Google Scholar 

  39. Shlomchik WD, et al. (1999) Prevention of graft versus host disease by inactivation of host antigen-presenting cells. Science. 285:412–5.

    Article  CAS  PubMed  Google Scholar 

  40. Reddy P, et al. (2005) A crucial role for antigenpresenting cells and alloantigen expression in graft-versus-leukemia responses. Nat. Med. 11:1244–9.

    Article  CAS  PubMed  Google Scholar 

  41. Matte CC, et al. (2004) Donor APCs are required for maximal GVHD but not for GVL. Nat. Med. 10:987–92.

    Article  CAS  PubMed  Google Scholar 

  42. Hadeiba H, et al. (2008) CCR9 expression defines tolerogenic plasmacytoid dendritic cells able to suppress acute graft-versus-host disease. Nat. Immunol. 9:1253–60.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Koyama M, et al. (2009) Plasmacytoid dendritic cells prime alloreactive T cells to mediate graft-versus-host disease as antigen-presenting cells. Blood. 113:2088–95.

    Article  CAS  PubMed  Google Scholar 

  44. Banovic T, et al. (2009) Graft-versus-host disease prevents the maturation of plasmacytoid dendritic cells. J. Immunol. 182:912–20.

    Article  CAS  PubMed  Google Scholar 

  45. Sharpe AH, Freeman GJ. (2002) The B7-CD28 superfamily. Nat. Rev. Immunol. 2:116–26.

    Article  CAS  PubMed  Google Scholar 

  46. Li XC, Rothstein DM, Sayegh MH. (2009) Costimulatory pathways in transplantation: challenges and new developments. Immunol. Rev. 229:271–93.

    Article  CAS  PubMed  Google Scholar 

  47. Blazar BR, et al. (2001) Ligation of 4-1BB (CDw137) regulates graft-versus-host disease, graft-versus-leukemia, and graft rejection in allogeneic bone marrow transplant recipients. J. Immunol. 166:3174–83.

    Article  CAS  PubMed  Google Scholar 

  48. Blazar BR, et al. (2004) CD30/CD30 ligand (CD153) interaction regulates CD4+ T cellmediated graft-versus-host disease. J. Immunol. 173:2933–41.

    Article  CAS  PubMed  Google Scholar 

  49. Blazar BR, et al. (2003) Ligation of OX40 (CD134) regulates graft-versus-host disease (GVHD) and graft rejection in allogeneic bone marrow transplant recipients. Blood. 101:3741–8.

    Article  CAS  PubMed  Google Scholar 

  50. Blazar BR, Taylor PA, Linsley PS, Vallera DA. (1994) In vivo blockade of CD28/CTLA4: B7/BB1 interaction with CTLA4-Ig reduces lethal murine graft-versus-host disease across the major histocompatibility complex barrier in mice. Blood. 83:3815–25.

    CAS  PubMed  Google Scholar 

  51. Blazar BR, et al. (1997) Blockade of CD40 ligand-CD40 interaction impairs CD4+ T cell-mediated alloreactivity by inhibiting mature donor T cell expansion and function after bone marrow transplantation. J. Immunol. 158:29–39.

    CAS  PubMed  Google Scholar 

  52. Hubbard VM, et al. (2005) Absence of inducible costimulator on alloreactive T cells reduces graft versus host disease and induces Th2 deviation. Blood. 106:3285–92.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Blazar BR, et al. (2003) Blockade of programmed death-1 engagement accelerates graft-versus-host disease lethality by an IFN-gamma-dependent mechanism. J. Immunol. 171:1272–7.

    Article  CAS  PubMed  Google Scholar 

  54. Morris ES, et al. (2009) Induction of natural killer T cell-dependent alloreactivity by administration of granulocyte colony-stimulating factor after bone marrow transplantation. Nat. Med. 15:436–41.

    Article  CAS  PubMed  Google Scholar 

  55. Reddy P, et al. (2004) Histone deacetylase inhibitor suberoylanilide hydroxamic acid reduces acute graft-versus-host disease and preserves graft-versus-leukemia effect. Proc. Natl. Acad. Sci. U. S. A. 101:3921–6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Reddy P, et al. (2008) Histone deacetylase inhibition modulates indoleamine 2,3-dioxygenase-dependent DC functions and regulates experimental graft-versus-host disease in mice. J. Clin. Invest. 118:2562–73.

    CAS  PubMed  PubMed Central  Google Scholar 

  57. Sun Y, et al. (2009) Cutting edge: Negative regulation of dendritic cells through acetylation of the nonhistone protein STAT-3. J. Immunol. 182:5899–903.

    Article  CAS  PubMed  Google Scholar 

  58. Wu CJ, Ritz J. (2006) Induction of tumor immunity following allogeneic stem cell transplantation. Adv. Immunol. 90:133–73.

    Article  CAS  PubMed  Google Scholar 

  59. Spierings E, et al. (2006) A uniform genomic minor histocompatibility antigen typing methodology and database designed to facilitate clinical applications. PLoS ONE. 1:e42.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  60. Anderson BE, et al. (2003) Memory CD4+ T cells do not induce graft-versus-host disease. J. Clin. Invest. 112:101–108.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. Chen BJ, Cui X, Sempowski GD, Liu C, Chao NJ. (2004) Transfer of allogeneic CD62L- memory T cells without graft-versus-host disease. Blood. 103:1534–41.

    Article  CAS  PubMed  Google Scholar 

  62. Ermann J, et al. (2005) Only the CD62L+ subpopulation of CD4+CD25+ regulatory T cells protects from lethal acute GVHD. Blood. 105:2220–6.

    Article  CAS  PubMed  Google Scholar 

  63. Taylor PA, et al. (2004) L-Selectin(hi) but not the L-selectin(lo) CD4+25+ T-regulatory cells are potent inhibitors of GVHD and BM graft rejection. Blood. 104:3804–12.

    Article  CAS  PubMed  Google Scholar 

  64. Zheng H, et al. (2008) Effector memory CD4+ T cells mediate graft-versus-leukemia without inducing graft-versus-host disease. Blood. 111:2476–84.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  65. Miller JS, et al. (2007) Lymphodepletion followed by donor lymphocyte infusion (DLI) causes significantly more acute graft-versus-host disease than DLI alone. Blood. 110:2761–3.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  66. Zhang Y, Joe G, Hexner E, Zhu J, Emerson SG. (2005) Alloreactive memory T cells are responsible for the persistence of graft-versus-host disease. J. Immunol. 174:3051–8.

    Article  CAS  PubMed  Google Scholar 

  67. Zhang Y, Joe G, Hexner E, Zhu J, Emerson SG. (2005) Host-reactive CD8+ memory stem cells in graft-versus-host disease. Nat. Med. 11:1299.

    Article  CAS  PubMed  Google Scholar 

  68. Dutt S, et al. (2007) Naive and memory T cells induce different types of graft-versus-host disease. J. Immunol. 179:6547–54.

    Article  CAS  PubMed  Google Scholar 

  69. Blazar BR, Taylor PA. (2005) Regulatory T cells. Biol. Blood Marrow Transpl. 11:46–9.

    Article  Google Scholar 

  70. Cohen JL, Boyer O. (2006) The role of CD4+CD25hi regulatory T cells in the physiopathogeny of graft-versus-host disease. Curr. Opin. Immunol. 18:580–5.

    Article  CAS  PubMed  Google Scholar 

  71. Maeda Y, et al. (2005) Critical role of host gammadelta T cells in experimental acute graft-versus-host disease. Blood. 106:749–55.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  72. Roncarolo MG. (1997) The role of interleukin-10 in transplantation and GVHD. In: Graft-vs.-host disease. Ferrara JLM, Deeg HJ and Burakoff SJ (eds.) Marcel Dekker Inc., New York, pp 693–715.

    Google Scholar 

  73. Young KJ, DuTemple B, Phillips MJ, Zhang L. (2003) Inhibition of graft-versus-host disease by double-negative regulatory T cells. J. Immunol. 171:134–41.

    Article  CAS  PubMed  Google Scholar 

  74. Zeng D, et al. (1999) Bone marrow NK1.1(−) and NK1.1(+) T cells reciprocally regulate acute graft versus host disease. J. Exp. Med. 189:1073–81.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  75. Cohen JL, Trenado A, Vasey D, Klatzmann D, Salomon BL. (2002) CD4(+)CD25(+) immunoregulatory T Cells: new therapeutics for graft-versus-host disease. J. Exp. Med. 196:401–6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  76. Edinger M, et al. (2003) CD4+CD25+ regulatory T cells preserve graft-versus-tumor activity while inhibiting graft-versus-host disease after bone marrow transplantation. Nat. Med. 9:1144–50.

    Article  CAS  PubMed  Google Scholar 

  77. Hoffmann P, Ermann J, Edinger M, Fathman CG, Strober S. (2002) Donor-type CD4(+)CD25(+) regulatory T cells suppress lethal acute graft-versus-host disease after allogeneic bone marrow transplantation. J. Exp. Med. 196:389–99.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  78. Jones SC, Murphy GF, Korngold R. (2003) Post-hematopoietic cell transplantation control of graft-versus-host disease by donor CD425 T cells to allow an effective graft-versus-leukemia response. Biol. Blood Marrow Transpl. 9:243–56.

    Article  Google Scholar 

  79. Taylor PA, Lees CJ and Blazar BR. (2002) The infusion of ex vivo activated and expanded CD4(+)CD25(+) immune regulatory cells inhibits graft-versus-host disease lethality. Blood. 99:3493–9.

    Article  CAS  PubMed  Google Scholar 

  80. Coghill JM, Carlson MJ, Moran TP, Serody JS. (2008) The biology and therapeutic potential of natural regulatory T-cells in the bone marrow transplant setting. Leuk. Lymphoma 49:1860–9.

    Article  CAS  PubMed  Google Scholar 

  81. Nguyen VH, et al. (2008) The impact of regulatory T cells on T-cell immunity following hematopoietic cell transplantation. Blood. 111:945–53.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  82. Ferrara JL, Krenger W. (1998) Graft-versus-host disease: the influence of type 1 and type 2 T cell cytokines. Transf. Med. Rev. 12:1–17.

    Article  CAS  Google Scholar 

  83. Ferrara JLM. (1994) The cytokine storm of acute graft-versus host disease. Haematol. Rev. 8:27.

    Google Scholar 

  84. Reddy P. (2003) Pathophysiology of acute graft-versus-host disease. Hematol. Oncol. 21:149–61.

    Article  PubMed  Google Scholar 

  85. Ratanatharathorn V, et al. (1998) Phase III study comparing methotrexate and tacrolimus (prograf, FK506) with methotrexate and cyclosporine for graft-versus-host disease prophylaxis after HLA-identical sibling bone marrow transplantation. Blood. 92:2303–14.

    CAS  PubMed  Google Scholar 

  86. Liu EH, Siegel RM, Harlan DM, O’Shea JJ. (2007) T cell-directed therapies: lessons learned and future prospects. Nat. Immunol. 8:25–30.

    Article  CAS  PubMed  Google Scholar 

  87. Zeiser R, et al. (2006) Inhibition of CD4+CD25+ regulatory T-cell function by calcineurin-dependent interleukin-2 production. Blood. 108:390–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  88. Zhang H, et al. (2005) Lymphopenia and interleukin-2 therapy alter homeostasis of CD4+CD25+ regulatory T cells. Nat. Med. 11:1238–43.

    Article  CAS  PubMed  Google Scholar 

  89. Liston A, Rudensky AY. (2007) Thymic development and peripheral homeostasis of regulatory T cells. Curr. Opin. Immunol. 19:176–85.

    Article  CAS  PubMed  Google Scholar 

  90. Gavin MA, et al. (2007) Foxp3-dependent programme of regulatory T-cell differentiation. Nature. 445:771–5.

    Article  CAS  PubMed  Google Scholar 

  91. Fowler DH, Kurasawa K, Smith R, Eckhaus MA, Gress RE. (1994) Donor CD4-enriched cells of Th2 cytokine phenotype regulate graft-versus-host disease without impairing allogeneic engraftment in sublethally irradiated mice. Blood. 84:3540–9.

    CAS  PubMed  Google Scholar 

  92. Krenger W, Snyder KM, Byon JC, Falzarano G, Ferrara JL. (1995) Polarized type 2 alloreactive CD4+ and CD8+ donor T cells fail to induce experimental acute graft-versus-host disease. J. Immunol. 155:585–93.

    CAS  PubMed  Google Scholar 

  93. Pan L, Delmonte J, Jalonen C, Ferrara J. (1995) Pretreatment of donor mice with granulocyte colony-stimulating factor polarizes donor T-lymphocytes toward type-2 cytokine production and reduces severity of experimental graft-versus-host disease. Blood. 86:4422–9.

    CAS  PubMed  Google Scholar 

  94. Hill GR, et al. (1998) Interleukin-11 promotes T cell polarization and prevents acute graft-versus-host disease after allogeneic bone marrow transplantation. J. Clin. Invest. 102:115–23.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  95. Reddy P, et al. (2003) Pretreatment of donors with interleukin-18 attenuates acute graft-versus-host disease via STAT6 and preserves graft-versus-leukemia effects. Blood. 101:2877–85.

    Article  CAS  PubMed  Google Scholar 

  96. Foley JE, et al. (2005) Ex vivo rapamycin generates donor Th2 cells that potently inhibit graft-versus-host disease and graft-versus-tumor effects via an IL-4-dependent mechanism. J. Immunol. 175:5732–43.

    Article  CAS  PubMed  Google Scholar 

  97. Jung U, et al. (2006) Ex vivo rapamycin generates Th1/Tc1 or Th2/Tc2 effector T cells with enhanced in vivo function and differential sensitivity to post-transplant rapamycin therapy. Biol. Blood Marrow Transpl. 12:905–18.

    Article  CAS  Google Scholar 

  98. Fowler DH, Gress RE. (2000) Th2 and Tc2 cells in the regulation of GVHD, GVL, and graft rejection: considerations for the allogeneic transplantation therapy of leukemia and lymphoma. Leuk. Lymphoma 38:221–34.

    Article  CAS  PubMed  Google Scholar 

  99. Tawara I, et al. (2008) Combined Th2 cytokine deficiency in donor T cells aggravates experimental acute graft-vs-host disease. Exp. Hematol. 36:988–96.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  100. Nikolic B, Lee S, Bronson R, Grusby M, Sykes M. (2000) Th1 and Th2 mediate acute graft- versus-host disease, each with distinct endorgan targets. J. Clin. Invest. 105:1289–98.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  101. Yi T, et al. (2008) Absence of donor Th17 leads to augmented Th1 differentiation and exacerbated acute graft-versus-host disease. Blood. 112:2101–10.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  102. Kappel LW, et al. (2009) IL-17 contributes to CD4-mediated graft-versus-host disease. Blood. 113:945–52.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  103. Carlson MJ, et al. (2009) In vitro-differentiated TH17 cells mediate lethal acute graft-versus-host disease with severe cutaneous and pulmonary pathologic manifestations. Blood. 113:1365–74.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  104. Asai O, et al. (1998) Suppression of graft-versus-host disease and amplification of graft-versus-tumor effects by activated natural killer cells after allogeneic bone marrow transplantation. J. Clin. Invest. 101:1835–42.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  105. Baker J, Verneris MR, Ito M, Shizuru JA, Negrin RS. (2001) Expansion of cytolytic CD8(+) natural killer T cells with limited capacity for graft-versus-host disease induction due to interferon gamma production. Blood. 97:2923–31.

    Article  CAS  PubMed  Google Scholar 

  106. Nishimura R, et al. (2008) In vivo trafficking and survival of cytokine-induced killer cells resulting in minimal GVHD with retention of antitumor activity. Blood. 112:2563–74.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  107. Brown GR, Lee E, Thiele DL. (2002) TNF-TNFR2 interactions are critical for the development of intestinal graft-versus-host disease in MHC class II-disparate (C57BL/6J→C57BL/6J x bm12)F1 mice. J. Immunol. 168:3065–71.

    Article  CAS  PubMed  Google Scholar 

  108. Brown GR, Lee EL, El-Hayek J, Kintner K, Luck C. (2005) IL-12-independent LIGHT signaling enhances MHC class II disparate CD4+ T cell alloproliferation, IFN-gamma responses, and intestinal graft-versus-host disease. J. Immunol. 174:4688–95.

    Article  CAS  PubMed  Google Scholar 

  109. Kagi D, et al. (1994) Fas and perforin pathways as major mechanisms of T cell-mediated cytotoxicity. Science. 265:528–30.

    Article  CAS  PubMed  Google Scholar 

  110. Sato K, et al. (2005) TRAIL-transduced dendritic cells protect mice from acute graft-versus-host disease and leukemia relapse. J. Immunol. 174:4025–33.

    Article  CAS  PubMed  Google Scholar 

  111. Schmaltz C, et al. (2002) T cells require TRAIL for optimal graft-versus-tumor activity. Nat. Med. 8:1433–7.

    Article  CAS  PubMed  Google Scholar 

  112. van den Brink MR, Burakoff SJ. (2002) Cytolytic pathways in haematopoietic stem-cell transplantation. Nat. Rev. Immunol. 2:273–81.

    Article  PubMed  CAS  Google Scholar 

  113. Xu Y, et al. (2006) Selective targeting of the LIGHT-HVEM costimulatory system for the treatment of graft-versus-host disease. Blood. 109:4097–104.

    Article  PubMed  CAS  Google Scholar 

  114. Zimmerman Z, et al. (2005) Effector cells derived from host CD8 memory T cells mediate rapid resistance against minor histocompatibility antigen-mismatched allogeneic marrow grafts without participation of perforin, Fas lig-and, and the simultaneous inhibition of 3 tumor necrosis factor family effector pathways. Biol. Blood Marrow Transpl. 11:576–86.

    Article  CAS  Google Scholar 

  115. Piguet PF, Grau GE, Allet B, Vassalli P. (1987) Tumor necrosis factor/cachectin is an effector of skin and gut lesions of the acute phase of graft versus host disease. J. Exp. Med. 166:1280–9.

    Article  CAS  PubMed  Google Scholar 

  116. Abhyankar S, Gilliland DG, Ferrara JL. (1993)Interleukin-1 is a critical effector molecule during cytokine dysregulation in graft versus host disease to minor histocompatibility antigens. Transplantation. 56:1518–23.

    Article  CAS  PubMed  Google Scholar 

  117. Krenger W, et al. (1996) Interferon-gamma suppresses T-cell proliferation to mitogen via the nitric oxide pathway during experimental acute graft-versus-host disease. Blood. 88:1113–21.

    CAS  PubMed  Google Scholar 

  118. Nestel FP, Greene RN, Kichian K, Ponka P, Lapp WS. (2000) Activation of macrophage cytostatic effector mechanisms during acute graft-versus-host disease: release of intracellular iron and nitric oxide-mediated cytostasis. Blood. 96:1836–43.

    CAS  PubMed  Google Scholar 

  119. Sterner DE, Berger SL. (2000) Acetylation of histones and transcription-related factors. Microbiol. Mol. Biol. Rev. 64:435–59.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  120. Lachner M, O’Sullivan RJ, Jenuwein T. (2003) An epigenetic road map for histone lysine methylation. J. Cell Sci. 116:2117–24.

    Article  CAS  PubMed  Google Scholar 

  121. Grunstein M. (1997) Histone acetylation in chromatin structure and transcription. Nature. 389:349–52.

    Article  CAS  PubMed  Google Scholar 

  122. Thompson JS, Ling X, Grunstein M. (1994) Histone H3 amino terminus is required for telomeric and silent mating locus repression in yeast. Nature. 369:245–7.

    Article  CAS  PubMed  Google Scholar 

  123. Durrin LK, Mann RK, Kayne PS, Grunstein M. (1991) Yeast histone H4 N-terminal sequence is required for promoter activation in vivo. Cell. 65:1023–31.

    Article  CAS  PubMed  Google Scholar 

  124. Allfrey VG, Pogo BG, Littau VC, Gershey EL, Mirsky AE. (1968) Histone acetylation in insect chromosomes. Science. 159:314–6.

    Article  CAS  PubMed  Google Scholar 

  125. Marmorstein R, Roth SY. (2001) Histone acetyltransferases: function, structure, and catalysis. Curr Opin Genet Dev 11:155–61.

    Article  CAS  PubMed  Google Scholar 

  126. Bolden JE, Peart MJ, Johnstone RW. (2006) Anticancer activities of histone deacetylase inhibitors. Nat. Rev. Drug. Discov. 5:769–84.

    Article  CAS  PubMed  Google Scholar 

  127. Yang X-J, Seto E. (2008) The Rpd3/Hda1 family of lysine deacetylases: From bacteria and yeast to mice and men. Nat. Rev. Mol. Cell. Biol. 9:206–18.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  128. Yang X-J, Seto E. (2008) Lysine acetylation: Codified crosstalk with other posttranslational modifications. Mol. Cell 31:449–61.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  129. Narlikar GJ, Fan HY, Kingston RE. (2002) Cooperation between complexes that regulate chromatin structure and transcription. Cell. 108:475–87.

    Article  CAS  PubMed  Google Scholar 

  130. Kouzarides T. (2007) Chromatin modifications and their function. Cell. 128:693–705.

    Article  CAS  PubMed  Google Scholar 

  131. Kim SC, et al. (2006) Substrate and functional diversity of lysine acetylation revealed by a proteomics survey. Mol. Cell. 23:607–618.

    Article  CAS  PubMed  Google Scholar 

  132. Redner RL, Wang J, Liu JM. (1999) Chromatin remodeling and leukemia: new therapeutic paradigms. Blood. 94:417–28.

    CAS  PubMed  Google Scholar 

  133. Bhalla KN. (2005) Epigenetic and chromatin modifiers as targeted therapy of hematologic malignancies. J. Clin. Oncol. 23:3971–93.

    Article  CAS  PubMed  Google Scholar 

  134. Byrd JC, et al. (2005) A phase 1 and pharmacodynamic study of depsipeptide (FK228) in chronic lymphocytic leukemia and acute myeloid leukemia. Blood. 105:959–67.

    Article  CAS  PubMed  Google Scholar 

  135. Mann BS, Johnson JR, Cohen MH, Justice R, Pazdur R. (2007) FDA approval summary: Vorinostat for treatment of advanced primary cutaneous T-cell lymphoma. Oncologist. 12:1247–52.

    Article  CAS  PubMed  Google Scholar 

  136. Mai A, et al. (2005) Histone deacetylation in epigenetics: an attractive target for anticancer therapy. Med. Res. Rev. 25:261–309.

    Article  CAS  PubMed  Google Scholar 

  137. Miller TA, Witter DJ, Belvedere S. (2003) Histone deacetylase inhibitors. J. Med. Chem. 46:5097–16.

    Article  CAS  PubMed  Google Scholar 

  138. Kelly WK, Marks PA. (2005) Drug insight: Histone deacetylase inhibitors—development of the new targeted anticancer agent suberoylanilide hydroxamic acid. Nat. Clin. Pract. Oncol. 2:150–7.

    Article  CAS  PubMed  Google Scholar 

  139. Finnin MS, et al. (1999) Structures of a histone deacetylase homologue bound to the TSA and SAHA inhibitors. Nature. 401:188–93.

    Article  CAS  PubMed  Google Scholar 

  140. Duvic M, et al. (2007) Phase 2 trial of oral vorinostat (suberoylanilide hydroxamic acid, SAHA) for refractory cutaneous T-cell lymphoma (CTCL). Blood. 109:31–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  141. Mann BS, et al. (2007) Vorinostat for treatment of cutaneous manifestations of advanced primary cutaneous T-cell lymphoma. Clin Cancer Res. 13:2318–22.

    Article  CAS  PubMed  Google Scholar 

  142. Marks PA. (2007) Discovery and development of SAHA as an anticancer agent. Oncogene. 26:1351–6.

    Article  CAS  PubMed  Google Scholar 

  143. Mishra N, Reilly CM, Brown DR, Ruiz P, Gilkeson GS. (2003) Histone deacetylase inhibitors modulate renal disease in the MRL-lpr/lpr mouse. J. Clin. Invest. 111:539–52.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  144. Skov S, et al. (2003) Histone deacetylase inhibitors: a new class of immunosuppressors targeting a novel signal pathway essential for CD154 expression. Blood 101:1430–8.

    Article  CAS  PubMed  Google Scholar 

  145. Glauben R, et al. (2006) Histone hyperacetylation is associated with amelioration of experimental colitis in mice. J. Immunol. 176:5015–22.

    Article  CAS  PubMed  Google Scholar 

  146. Glauben R, et al. (2008) Histone deacetylases: Novel targets for prevention of colitis-associated cancer in mice. Gut. 57:613–22.

    Article  CAS  PubMed  Google Scholar 

  147. Tao R, et al. (2007) Deacetylase inhibition promotes the generation and function of regulatory T cells. Nat. Med. 13:1299–307.

    Article  CAS  PubMed  Google Scholar 

  148. Leng C, et al. (2006) 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. 34:776–87.

    Article  CAS  PubMed  Google Scholar 

  149. Shlomchik WD. (2007) Graft-versus-host disease. Nat. Rev. Immunol. 7:340–52.

    Article  CAS  PubMed  Google Scholar 

  150. Minucci S, Pelicci PG. (2006) Histone deacetylase inhibitors and the promise of epigenetic (and more) treatments for cancer. Nat. Rev. Cancer. 6:38–51.

    Article  CAS  PubMed  Google Scholar 

  151. Reddy P, et al. (2001) Interleukin-18 regulates acute graft-versus-host disease by enhancing Fas-mediated donor T cell apoptosis. J. Exp. Med. 194:1433–40.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  152. Zhang Y, Louboutin JP, Zhu J, Rivera AJ, Emerson SG. (2002) Preterminal host dendritic cells in irradiated mice prime CD8+ T cell-mediated acute graft-versus-host disease. J. Clin. Invest. 109:1335–44.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  153. Riddell SR, Murata M, Bryant S, Warren EH. (2002) Minor histocompatibility antigens—targets of graft versus leukemia responses. Int. J. Hematol. 76 Suppl 2:155–61.

    Article  PubMed  Google Scholar 

  154. Teshima T, et al. (1999) IL-11 separates graft-versus-leukemia effects from graft-versus-host disease after bone marrow transplantation. J. Clin. Invest. 104:317–25.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  155. Yang YG, Dey B, Sergio JJ, Sykes M. (1997) Interleukin-12 prevents severe acute graft-versus-host disease (GVHD) and GVHD-associated immune dysfunction in a fully major histocompatibility complex haplotype-mismatched murine bone marrow transplantation model. Transplantation. 64:1343–52.

    Article  CAS  PubMed  Google Scholar 

  156. Banchereau J, et al. (2000) Immunobiology of dendritic cells. Annu. Rev. Immunol. 18:767–811.

    Article  CAS  PubMed  Google Scholar 

  157. Medzhitov R, Janeway CA Jr. (2002) Decoding the patterns of self and nonself by the innate immune system. Science. 296:298–300.

    Article  CAS  PubMed  Google Scholar 

  158. Akira S. (2003) Mammalian Toll-like receptors. Curr. Opin. Immunol. 15:5–11.

    Article  CAS  PubMed  Google Scholar 

  159. Kobayashi K, et al. (2002) RICK/Rip2/CARDIAK mediates signalling for receptors of the innate and adaptive immune systems. Nature. 416:194–9.

    Article  CAS  PubMed  Google Scholar 

  160. Mellor AL, Munn DH. (2004) IDO expression by dendritic cells: Tolerance and tryptophan catabolism. Nat. Rev. Immunol. 4:762–74.

    Article  CAS  PubMed  Google Scholar 

  161. Murray PJ. (2007) The JAK-STAT signaling pathway: Input and output integration. J. Immunol. 178:2623–9.

    Article  CAS  PubMed  Google Scholar 

  162. Schindler C, Plumlee C. (2008) Inteferons pen the JAK-STAT pathway. Semin. Cell. Dev. Biol. 19:311–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  163. Stepkowski SM, Chen W, Ross JA, Nagy ZS, Kirken RA. (2008) STAT3: An important regulator of multiple cytokine functions. Transplantation. 85:1372–7.

    Article  CAS  PubMed  Google Scholar 

  164. Takeda K, et al. (1997) Targeted disruption of the mouse Stat3 gene leads to early embryonic lethality. Proc. Natl. Acad. Sci. U. S. A. 94:3801–4.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  165. Kortylewski M, et al. (2005) Inhibiting Stat3 signaling in the hematopoietic system elicits multicomponent antitumor immunity. Nat. Med. 11:1314–21.

    Article  CAS  PubMed  Google Scholar 

  166. Cheng F, et al. (2003) A critical role for Stat3 signaling in immune tolerance. Immunity. 19:425–36.

    Article  CAS  PubMed  Google Scholar 

  167. Yu H, Kortylewski M, Pardoll D. (2007) Crosstalk between cancer and immune cells: Role of STAT3 in the tumour microenvironment. Nat. Rev. Immunol. 7:41–51.

    Article  CAS  PubMed  Google Scholar 

  168. Milner JD, et al. (2008) Impaired T(H)17 cell differentiation in subjects with autosomal dominant hyper-IgE syndrome. Nature. 452:773–6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  169. Holland SM, et al. (2007) STAT3 mutations in the hyper-IgE syndrome. N. Engl. J. Med. 357:1608–19.

    Article  CAS  PubMed  Google Scholar 

  170. Minegishi Y, et al. (2007) Dominant-negative mutations in the DNA-binding domain of STAT3 cause hyper-IgE syndrome. Nature. 448:1058–62.

    Article  CAS  PubMed  Google Scholar 

  171. Barton BE. (2006) STAT3: A potential therapeutic target in dendritic cells for the induction of transplant tolerance. Expert Opin. Ther. Targets. 10:459–70.

    Article  CAS  PubMed  Google Scholar 

  172. Yang J, et al. (2005) Novel roles of unphosphorylated STAT3 in oncogenesis and transcriptional regulation. Cancer Res. 65:939–47.

    CAS  PubMed  Google Scholar 

  173. Nadiminty N, et al. (2006) Stat3 activation of NF-{kappa}B p100 processing involves CBP/p300-mediated acetylation. Proc. Natl. Acad. Sci. U. S. A. 103:7264–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  174. Yuan ZL, Guan YJ, Chatterjee D, Chin YE. (2005) Stat3 dimerization regulated by reversible acetylation of a single lysine residue. Science. 307:269–73.

    Article  CAS  PubMed  Google Scholar 

  175. Ray S, Boldogh I, Brasier AR. (2005) STAT3 NH2-terminal acetylation is activated by the hepatic acute-phase response and required for IL-6 induction of angiotensinogen. Gastroenterology. 129:1616–32.

    Article  CAS  PubMed  Google Scholar 

  176. Yang J, et al. (2007) Unphosphorylated STAT3 accumulates in response to IL-6 and activates transcription by binding to NFkappaB. Genes. Dev. 21:1396–408.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  177. Sehgal PB. (2008) Paradigm shifts in the cell biology of STAT signaling. Semin. Cell. Dev. Biol. 19:329–40.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  178. Hou T, Ray S, Lee C, Brasier AR. (2008) The STAT3 NH2-terminal domain stabilizes enhanceosome assembly by interacting with the p300 bromodomain. J. Biol. Chem. 283:30725–34.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  179. Blaskovich MA, et al. (2003) Discovery of JSI-124 (cucurbitacin I), a selective Janus kinase/signal transducer and activator of transcription 3 signaling pathway inhibitor with potent antitumor activity against human and murine cancer cells in mice. Cancer Res. 63:1270–9.

    CAS  PubMed  Google Scholar 

  180. Wang R, Cherukuri P, Luo J. (2005) Activation of Stat3 sequence-specific DNA binding and transcription by p300/CREB-binding proteinmediated acetylation. J. Biol. Chem. 280:11528–34.

    Article  CAS  PubMed  Google Scholar 

  181. Hu X, Ivashkiv LB. (2009) Cross-regulation of signaling pathways by interferon-gamma: implications for immune responses and autoimmune diseases. Immunity. 31:539–50.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  182. Melillo JA, et al. (2010) Dendritic cell (DC)-specific targeting reveals Stat3 as a negative regulator of DC function. J. Immunol. 184:2638–45.

    Article  CAS  PubMed  Google Scholar 

  183. Nie Y, et al. (2009) STAT3 inhibition of gluconeogenesis is downregulated by SirT1. Nat. Cell Biol. 11:492–500.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  184. Villagra A, et al. (2009) The histone deacetylase HDAC11 regulates the expression of interleukin 10 and immune tolerance. Nat. Immunol. 10:92–100.

    Article  CAS  PubMed  Google Scholar 

  185. Dubovsky JA, et al. (Circumventing immune tolerance through epigenetic modification. Curr. Pharm. Des. 16:268–76.

    Article  CAS  PubMed  Google Scholar 

  186. Gao L, Cueto MA, Asselbergs F, Atadja P. (2002) Cloning and functional characterization of HDAC11, a novel member of the human histone deacetylase family. J. Biol. Chem. 277:25748–55.

    Article  CAS  PubMed  Google Scholar 

  187. Moore KW, de Waal Malefyt R, Coffman RL, O’Garra A. (2001) Interleukin-10 and the interleukin-10 receptor. Annu. Rev. Immunol. 19:683–765.

    Article  CAS  PubMed  Google Scholar 

  188. Li MO, Flavell RA. (2008) Contextual regulation of inflammation: A duet by transforming growth factor-beta and interleukin-10. Immunity. 28:468–76.

    Article  PubMed  CAS  Google Scholar 

  189. Rubtsov YP, et al. (2008) Regulatory T cell-derived interleukin-10 limits inflammation at environmental interfaces. Immunity. 28:546–58.

    Article  CAS  PubMed  Google Scholar 

  190. Skov S, et al. (2005) Cancer cells become susceptible to natural killer cell killing after exposure to histone deacetylase inhibitors due to glycogen synthase kinase-3-dependent expression of MHC class I-related chain A and B. Cancer Res. 65:11136–45.

    Article  CAS  PubMed  Google Scholar 

  191. Armeanu S, et al. (2005) Natural killer cellmediated lysis of hepatoma cells via specific induction of NKG2D ligands by the histone deacetylase inhibitor sodium valproate. Cancer Res. 65:6321–9.

    Article  CAS  PubMed  Google Scholar 

  192. Groh V, et al. (1999) Broad tumor-associated expression and recognition by tumor-derived gamma delta T cells of MICA and MICB. Proc. Natl. Acad. Sci. U. S. A. 96:6879–84.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  193. Salih HR, et al. (2003) Functional expression and release of ligands for the activating immunoreceptor NKG2D in leukemia. Blood. 102:1389–96.

    Article  CAS  PubMed  Google Scholar 

  194. Pende D, et al. (2002) Major histocompatibility complex class I-related chain A and UL16-bind-ing protein expression on tumor cell lines of different histotypes: analysis of tumor susceptibility to NKG2D-dependent natural killer cell cytotoxicity. Cancer Res. 62:6178–86.

    CAS  PubMed  Google Scholar 

  195. Nebbioso A, et al. (2005) Tumor-selective action of HDAC inhibitors involves TRAIL induction in acute myeloid leukemia cells. Nat. Med. 11:77–84.

    Article  CAS  PubMed  Google Scholar 

  196. Insinga A, et al. (2005) Inhibitors of histone deacetylases induce tumor-selective apoptosis through activation of the death receptor pathway. Nat. Med. 11:71–6.

    Article  CAS  PubMed  Google Scholar 

  197. Kinugasa F, et al. (2008) Effect of a new immunosuppressant histon deacetylase (HDAC) inhibitor FR276457 in a rat cardiac transplant model. Biol. Pharm. Bull. 31:1723–6.

    Article  CAS  PubMed  Google Scholar 

  198. Kinugasa F, et al. (2009) Effect of the immunosuppressant histone deacetylase inhibitor FR276457 in a canine renal transplant model. Transpl. Immunol. 21:198–202.

    Article  CAS  PubMed  Google Scholar 

  199. Mori H, et al. (2003) FR235222, a fungal metabolite, is a novel immunosuppressant that inhibits mammalian histone deacetylase (HDAC) II. Biological activities in animal models. J. Antibiot. (Tokyo) 56:80–6.

    Article  CAS  Google Scholar 

  200. Reddy P, Zou W. (2007) Blocking HDACs boosts regulatory T cells. Nat. Med. 13:1282–4.

    Article  CAS  PubMed  Google Scholar 

  201. Bosisio D, et al. (2008) Blocking TH17-polarizing cytokines by histone deacetylase inhibitors in vitro and in vivo. J. Leukoc. Biol. 84:1540–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  202. Chen X, et al. (2007) Absence of regulatory T cell control of TH1 and TH17 cells is responsible for the autoimmune-mediated pathology in chronic graft versus host disease. Blood. 110:3804–13.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  203. Furlan A, et al. (2011) Pharmacokinetics, safety and inducible cytokine responses during a phase 1 trial of the oral histone deacetylase inhibitor ITF2357 (givinostat). Mol. Med. 17:353–362.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  204. Levine JE, et al. (2003) Lowered-intensity preparative regimen for allogeneic stem cell transplantation delays acute graft-versus-host disease but does not improve outcome for advanced hematologic malignancy. Biol. Blood Marrow Transpl. 9:189–97.

    Article  Google Scholar 

Download references

Acknowledgment

Supported by NIH grants: AI075284, HL090775 and CA143379 to PR.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Pavan Reddy.

Rights and permissions

Open Access This article is published under license to BioMed Central Ltd. This is an Open Access article is distributed under the terms of the Creative Commons Attribution License ( https://creativecommons.org/licenses/by/2.0 ), which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.

Reprints and permissions

About this article

Cite this article

Choi, S., Reddy, P. HDAC Inhibition and Graft Versus Host Disease. Mol Med 17, 404–416 (2011). https://doi.org/10.2119/molmed.2011.00007

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.2119/molmed.2011.00007

Keywords