The last 6 decades have seen major advances in the understanding of immunologic diseases, driven by preclinical animal models. Indeed, bone marrow transplantation (BMT) has its genesis in rodent models dating back to the 1950s. Allogeneic BMT and its major complication, graft-versus-host disease (GVHD), represent a paradigm for the translation of preclinical concepts into clinical practice. The appreciation that GVHD can be thought of as a stepwise escalation in immune activation characterized by eventual massive target tissue apoptosis has allowed the design of rational approaches to better manage patients. Here, we describe the pathophysiology of GVHD as defined in preclinical models, focusing on the successes and failures of this research to instruct and translate clinical practice. We also provide a commentary on the limitations of these models so that they may be better appreciated and addressed in future studies. Notable preclinical successes include the definition of modern immune suppression, reductions in conditioning intensity, posttransplant cyclophosphamide, and the promotion of regulatory T-cell reconstitution. New strategies including naïve T-cell depletion, focused cytokine and chemokine inhibition, and the blockade of costimulation now also appear highly promising and very likely to translate into patients in the near future.

Allogeneic peripheral blood stem cell or bone marrow transplantation (hereafter referred to as BMT) continues to expand in use as curative therapy for hematologic malignancies, predominantly as a vehicle for generating potent immunologic graft-versus-leukemia (GVL) effects. Modern BMT has its genesis in the early 1950s from the original rodent studies demonstrating the ability of bone marrow to prevent radiation lethality.1,2  As early as 1956, a GVL effect was mooted,3  but it was not until 1968 that the first successful allogeneic BMT was performed.4  Improvements in donor and recipient selection, conditioning regimens, graft-versus-host disease (GVHD) prophylaxis and treatment, and attention to infectious complications mean that, in 2014, the majority of transplant-related mortality is due to relapse of primary malignancy. However, GVHD remains a major cause of treatment failure, resulting in mortality in 20% of recipients.5  A recent study reviewed transplant outcome between 1993 and 1997, and compared these to more recent outcomes in the period 2003 through 2007.6  Considering all indications for transplant, 70% of patients developed grade II-IV acute GVHD (aGVHD) in the latter cohort, a slight decrease compared with 77% in the earlier group. This small improvement reflects the reduction in incidence of severe (grade III/IV) GVHD which decreased from 30% to 14% in the 2003 to 2007 cohort. Data pertaining to chronic GVHD are more elusive, but it is estimated that 60% to 80% of long-term survivors of BMT experience some degree of chronic GVHD.7  More recent studies reintroducing T-cell–depleting antibodies in the peritransplant period have demonstrated further reductions in the incidence of significant GVHD; however, these reductions have not been associated with improvements in survival.8  Thus, at a time of intense activity in experimental modeling of GVHD and GVL, only modest improvements in clinical outcome are being realized.

So what insights have been gained from these preclinical studies? Is it too early yet to see the effects of this work translated, are there scientific limitations of these studies to date that limit their clinical relevance, or is the translation to a relatively small discipline too difficult? Here, we propose that, although all of the above apply to some extent, experimental studies have made major contributions to clinical practice over the last 3 decades that are not broadly appreciated.

This review will focus on the pathophysiology of GVHD and, in particular, the contributions made by experimental models to the understanding of GVHD and the clinical practice of transplantation. In addition, we will highlight the limitations of experimental systems in the hope that some of these issues can be better appreciated and addressed scientifically as the field moves forward. Finally, we will outline recently defined pathways of GVHD that hold particular promise for clinical translation.

The majority of animal models currently used to study GVHD use inbred mice. The advantages of murine systems include the ability to: (1) control environmental conditions (including microbiota and conditioning regimens), (2) control major histocompatibility complex (MHC) and minor histocompatibility antigen disparities, (3) transplant large numbers of recipients simultaneously, (4) genetically mutate key molecules in donor and/or recipients and within specific cell populations, (5) image and define immune reactions in real time and at multiple time points, (6) test lead therapeutics in a relevant disease model, and (7) the utilization of a relatively cost-effective model. The GVHD that develops in these systems is in response to alloantigen, as in patients, and on the whole faithfully mirrors the GVHD seen in the clinic. Furthermore, the ability to strictly control multiple variables enhances scientific rigor and facilitates the dissection of mechanisms of disease. However, humans are not inbred; they come to transplant as genetically diverse individuals, at varying ages, with varying diseases in varying states of remission, and with varying exposure to many, often toxic, previous therapies. Although it is easy to blame these differences for the failure of many preclinical mouse concepts to be emulated in patients, appreciating the potential limitations of the initial preclinical and/or the subsequent clinical study is more often informative. In particular, it should be noted that most murine models of GVHD (regardless of whether they are MHC matched or not) induce early CD4-dependent GVHD that is characterized by inflammatory cytokine dysregulation and T helper 1 (Th1) differentiation.9  This degree of CD4 dependence is unlikely to be as dominant in patients receiving HLA-matched grafts and immune suppression with calcineurin inhibitors. Indeed, the absence of immune suppression in murine models has the capacity to identify pathways of GVHD in mice that may well not even be operative clinically and this is a fundamental limitation of these systems. An additional weakness is the almost exclusive use of total body irradiation (TBI) in animal systems, which is increasingly disparate to clinical practice. Preclinical studies should thus ideally analyze both CD4- and CD8-dependent GVHD, and this rather than the use of MHC disparate or “clinically relevant” MHC matched models is paramount. It is no surprise then that seminal studies leading to the generation of our current immune suppression regimens come from studies in canines in the 1980s.10  These canine studies continue to this day and provide a robust preclinical model to analyze the therapeutic efficacy of therapeutics, allowing their prioritization for subsequent clinical trials. In particular, canine models overcome the issue of inbred backgrounds and better permit the analysis of therapies concurrent with clinically relevant immune suppression, antibiotics, etc. Their weakness is their cost and lack of genetic manipulation, meaning that murine and canine systems are likely to continue to perform complementary functions in the modeling of GVHD.

The ultimate goal of allogeneic BMT is to separate GVHD and GVL, which may ultimately require genetic manipulation of the T-cell receptor (TCR) to ensure specificity for recipient leukemia or at least hematopoiesis, an approach now feasible.11  Nevertheless, even small shifts in favor of GVL are advantageous, as demonstrated by improved outcome in studies using peripheral blood stem cell grafts as opposed to bone marrow.12  This may be possible by targeting potential inflammatory molecules that mediate GVHD13  but are dispensable for the cognate interaction and cytotoxicity between cytotoxic T lymphocyte and leukemia that appears to be a prerequisite for GVL responses.14  The experimental GVHD literature demonstrates numerous therapeutic maneuvers that appear to achieve this aim. However, there remain a number of caveats to these studies that relate to limitations in the experimental GVHD systems and leukemia targets used. As noted, many of the therapeutic interventions published have demonstrated efficacy in CD4-dependent GVHD models (inflammatory cytokine inhibition as an example) but the GVL systems used experimentally are largely a measure of GVL that is CD8-dependent.15,16  Thus, the separation of the 2 processes may be overestimated due to the differential pathways used by GVHD and GVL in these systems. This is, of course, only an issue if the same does not hold true clinically. Although we do not yet have a complete understanding of the relative importance of class I– vs class II–dependent GVHD in the clinic when both pathways are operative, it appears that experimental models overestimate CD4-dependent pathways relative to those in the clinic.9 

The second issue relates to the leukemia targets used in experimental studies. The vast majority of animal systems have used cell lines on the recipient strain background to study GVL. Some of these cell lines (particularly B-cell lymphoma lines) grow variably in vivo and are exquisitely sensitive to a GVL effect such that they rarely survive in the presence of any alloreactive T-cell response. Thus, many of these studies to date demonstrate that a GVL effect remains following a given therapeutic intervention, but it is not possible to ascertain the quality of that response. Second, it is clear that different cell lines are sensitive to different killing pathways17  (likely the case in human leukemia also), meaning that multiple targets sensitive to killing by the pathway in question should ideally be tested in vivo. Third, it is becoming clear that primary leukemia (generated by retroviral insertion of appropriate oncogenes) may model GVL in a more realistic and clinically relevant fashion than cell lines.14,18  Strikingly different results in experimental studies of regulatory T cells (Tregs) are a case in point.19,20  For all these reasons, it seems clear that the use of cell lines in mouse models of GVHD to date has generated data that probably underestimate the effect of therapeutic interventions on the quality of GVL responses and this is likely an inherent weakness in murine systems. However, studying GVHD and GVL that require similar CD4- or CD8-dependent pathways while using primary leukemia rather than cell lines will help to generate data that faithfully model clinical GVL.

The characteristic pathologic feature of aGVHD is target tissue (skin, liver, gastrointestinal [GI] tract) apoptosis. Although idiopathic pneumonia syndrome is increasingly viewed as a manifestation of aGVHD in the lung, apoptosis is generally not a feature. aGVHD was initially described as a “cytokine storm” involving a three-step disease process by Ferrara and Deeg.21  These steps involve: (1) transplant conditioning and associated inflammation, (2) donor T-cell priming and differentiation, and (3) an effector phase of tissue apoptosis mediated by inflammatory cytokines and cellular (T and NK cell) effectors. It still remains useful to break GVHD down into these phases, particularly when considering where advances in the field of clinical transplantation originate, and where the limitations in this paradigm arise.

(1) Transplant conditioning and inflammation

Clinical allogeneic stem cell transplantation has traditionally involved the use of myeloablative chemotherapy, with or without TBI as a cytoreductive and immunosuppressive preparative regimen. For technical reasons largely relating to access and reproducibility, almost all preclinical animal studies use TBI alone as conditioning. In the mid 1990s, it became clear that the intensity of conditioning was an important factor in determining the tempo and nature of subsequent GVHD. Mouse models were instrumental in demonstrating the mechanism by which pretransplant conditioning contributes to GVHD pathophysiology (Figure 1).22-24  Many groups demonstrated that intensity of TBI and chemotherapy used during conditioning dictated the integrity of the GI mucosa and the subsequent transfer of bacterial lipopolysaccharide22-25  and other “danger/pathogen-associated molecular patterns” (DAMPS/PAMPS) into the systemic circulation. Indeed, the role of the microbiota in promoting GVHD has been a central theme in the last 4 decades of BMT research and more recent data has demonstrated profound effects of GVHD on the modification of microbiota diversity that promotes the GVHD response.26,27  It is thus now clear that lipopolysaccharide is but one of many microbiome-derived products that drive GVHD.26,28  The characteristic proinflammatory cytokines released by recipient cells at this early stage of GVHD pathogenesis are tumor necrosis factor (TNF) and interleukins 1 and 6 (IL-1 and IL-6).22,23,29,30  This inflammation has 2 potential effects. First, it activates recipient antigen-presenting cells (APCs), enhancing the ability of professional APCs to prime T cells and also likely promoting antigen presentation by nonprofessional APCs within tissue.31  Second, these cytokines may provide complimentary costimulatory signals to donor T cells (eg, TNF, IL-6).13,32  Finally, they generate tissue inflammation, allowing the access of T cells to GVHD target tissue.33 

Figure 1

GVHD pathophysiology phase 1: transplant conditioning and inflammation. Following conditioning (radiation and/or chemotherapy), the integrity of the GI mucosa becomes compromised allowing the release of DAMPS and PAMPS, which in turn promote the production of proinflammatory cytokines from recipient cells. These cytokines contribute to host APC (hematopoietic and nonhematopoietic) activation in the gut and lymphoid tissue. GVHD impacts on the gut microbiota, reducing its diversity with a loss of enteric commensal organisms and an outgrowth of pathogenic microbes that further exacerbates the pathological DAMP/PAMP cascade.

Figure 1

GVHD pathophysiology phase 1: transplant conditioning and inflammation. Following conditioning (radiation and/or chemotherapy), the integrity of the GI mucosa becomes compromised allowing the release of DAMPS and PAMPS, which in turn promote the production of proinflammatory cytokines from recipient cells. These cytokines contribute to host APC (hematopoietic and nonhematopoietic) activation in the gut and lymphoid tissue. GVHD impacts on the gut microbiota, reducing its diversity with a loss of enteric commensal organisms and an outgrowth of pathogenic microbes that further exacerbates the pathological DAMP/PAMP cascade.

Close modal

Thus, strategies to reduce GVHD at the conditioning phase have focused on a: (1) reduced-intensity conditioning (RIC), (2) manipulation of the microbiota, and (3) the blockade of inflammatory cytokines that promote the downstream priming T cells and their entry to tissue (discussed in section 3).

Canine models of allogeneic BMT were first used to address concerns that decreasing the intensity of the conditioning regimen would result in graft rejection (enhanced host-versus-graft effects) and loss of GVL effects. In the 1990s, these models confirmed that GVHD was reduced with so-called RIC protocols,34  and that poor engraftment could be overcome by the addition of peritransplant immune suppression, defining the combination of cyclosporine and mycophenolate that is used today.34-36  Murine data at this time confirmed the ability and mechanism by which conditioning intensity dictated the severity of GVHD.23  Because the toxicity of myeloablative regimens generally prevented elderly patients from receiving transplants, the clinical introduction of RIC was rapid and the first reports were published in 1997 and 1998.37,38  Although numerous conditioning regimens are now in clinical practice, the use of total lymphoid irradiation and anti-thymocyte globulin deserves special mention. This protocol was pioneered in animal models and shown to prevent GVHD but not GVL by enrichment of an IL-4–secreting recipient natural killer (NK) T-cell subset.39  This protocol has been successfully translated to the clinic with outcomes as predicted by animal models.40  Today, approximately one-third of patients receive transplants after RIC, a procedure that had its genesis in animal models. Attempts to improve GI tract integrity after myeloablative conditioning with cytokines such as IL-11 and KGF were promising approaches based on animal models24  but found limited therapeutic success due to either toxicity or incomplete efficacy in humans.41,42 

The relationship between the GI tract microbiota and GVHD was first reported in 197428  and the clinical importance and translation has been exemplified by the ability of gut decontamination to attenuate GVHD clinically.43  The current era of bacterial multidrug resistance severely limits this approach but it is likely that manipulation of the microbiota to a more tolerogeneic phenotype will be possible in the near future, as demonstrated in recent animal models.44  Clearly, the increased understanding of the influence of conditioning on GVHD gleaned from mouse models has had an enormous influence on clinical BMT in the modern era.

(2) Donor T-cell priming and differentiation

Priming of donor T cells

The priming and activation of donor T cells is the hallmark of the second phase of GVHD pathogenesis. The process requires the interaction of the donor T cell with an APC-presenting alloantigen (usually a minor histocompatibility antigen) which drives T-cell activation and differentiation (Figure 2). The site and type of APC involved in the initiation of GVHD has been the focus of a great deal of investigation in murine systems over the last 15 years. Current data support the notion that donor CD8 T cells are predominantly activated by recipient hematopoietic APCs whereas donor CD4 T cells can also be activated by recipient nonhematopoietic APC within the GI tract.31,45,46  Donor APCs also contribute to GVHD, once donor T cells are primed by recipient APCs.47  Studies using stringent conditional ablation of individual APC subsets have demonstrated that no single-recipient APC subset is mandatory in this process. In particular, despite the long-held and frequently quoted paradigm, recipient dendritic cells (DCs) are not required to initiate GVHD and may even regulate subsequent donor T-cell expansion.31,48  Although recipient DCs appear important in the generation of Treg responses,49  we have demonstrated that DCs can attenuate GVHD induced by alloantigen-specific TCR-transgenic effector T cells via a process of deletion.31  In this scenario, Tregs do not expand and so the process can also occur in the absence of effects on Tregs. In addition, both recipient B cells50  and macrophages51,52  appear capable of regulating T-cell expansion and aGVHD. It should, however, be noted that donor DCs would appear the critical donor APCs in regard to alloantigen presentation and the maintenance of GVHD.53-55  It is now clear that in the presence of GVHD, donor DCs have an impaired ability to present exogenous antigen, particularly via MHC class II, and that this contributes to poor immune function in patients with GVHD.56  The level of immune incompetence during GVHD had been historically attributed to defects within the T-cell compartment alone, and it will now be important to consider both the donor DC and T cell in immune-restorative therapy during GVHD.56 

Figure 2

GVHD pathophysiology phase 2: donor T-cell priming and differentiation. Donor CD4 T cells contained within the graft are activated by the inflammatory milieu early after conditioning, facilitating their rapid access to the gut and lymphoid tissue. Once in the gut, MHC class II–expressing recipient nonhematopoietic APCs can initiate priming to host antigens while recipient hematopoietic APC initiate priming in lymphoid tissue. Recipient hematopoietic APCs appear to be the dominant APCs for CD8 T-cell priming. Donor APCs can further contribute to this priming process. Activation in the presence of various cytokines (particularly IL-4, IL-6, and IL-12) instructs T-cell differentiation along specific lineage pathways (type 2, type 17, and type 1, respectively). The transcription factors GATA-3, RORγt, and T-bet are critical for these Th2, Th17, and Th1 differentiation pathways. Tregs are differentiated in the presence of IL-2 and TGFβ (in the absence of IL-6) and abrogate the differentiation of effector T cells via effects on DCs and effector T cells themselves.

Figure 2

GVHD pathophysiology phase 2: donor T-cell priming and differentiation. Donor CD4 T cells contained within the graft are activated by the inflammatory milieu early after conditioning, facilitating their rapid access to the gut and lymphoid tissue. Once in the gut, MHC class II–expressing recipient nonhematopoietic APCs can initiate priming to host antigens while recipient hematopoietic APC initiate priming in lymphoid tissue. Recipient hematopoietic APCs appear to be the dominant APCs for CD8 T-cell priming. Donor APCs can further contribute to this priming process. Activation in the presence of various cytokines (particularly IL-4, IL-6, and IL-12) instructs T-cell differentiation along specific lineage pathways (type 2, type 17, and type 1, respectively). The transcription factors GATA-3, RORγt, and T-bet are critical for these Th2, Th17, and Th1 differentiation pathways. Tregs are differentiated in the presence of IL-2 and TGFβ (in the absence of IL-6) and abrogate the differentiation of effector T cells via effects on DCs and effector T cells themselves.

Close modal

The costimulatory signals between the APC and T cell have been the subject of intense preclinical study over the last 25 years. In particular, blockade of the CD40-CD154 and CD80/CD86-CD28/CTLA4 axes have been explored in an attempt to induce alloantigen-specific T-cell anergy and tolerance (reviewed in Kinnear et al57 ). Clinical reagents that inhibit the CD80/CD86-CD28/CTLA4 axis are now available (belatacept, abatacept) and have proven to be effective, nontoxic alternatives to calcineurin inhibitors in renal transplantation, although in humans these agents do not appear to induce the tolerance seen in rodents.57  These agents are just beginning to be explored in clinical BMT and may be of particular value in patients intolerant of calcineurin inhibitors.

Donor T-cell expansion and differentiation

Once primed by alloantigen, donor T cells undergo proliferation and differentiation such that they secrete a predictable suite of cytokines and/or gain cytolytic function. This process of generating terminally differentiated effector T cells is the principal determinant of GVHD. Mouse studies have clearly demonstrated that it is the priming and differentiation of naïve (rather than memory) T cells that results in GVHD58,59  and early human data support this paradigm.60,61  This raises the possibility that depletion of naïve T cells may prevent GVHD while allowing the transfer of donor memory T cells that may provide pathogen-specific immunity. This concept is currently being tested clinically in the United States. The proliferation and expansion of donor T cells requires IL-2 and its autocrine binding via the IL-2 receptor complex (CD25 and CD122). The mainstay of both GVHD prophylaxis and treatment is calcineurin inhibitors and steroids, respectively, both of which decrease the IL-262  available to drive effector T-cell proliferation and survival. aGVHD, particularly that targeting the GI tract, has been defined as a Th1-mediated disease and the classical Th1-derived cytokines (eg, interferon γ [IFNγ] and TNF) are systemically elevated and clearly mediate target organ pathology in preclinical models.23,24,63,64  Donor Th1 differentiation, characterized by expression of the transcription factor t-bet, is driven in these systems by IL-12 secretion from APCs and this cytokine (together with IL-23) is now a target for neutralization in clinical BMT given the availability of clinical-grade inhibitors. It is becoming clear that GVHD within individual organs may be generated by different T-cell differentiation programs. In particular, it has been noted that Th17 differentiation tends to result in GVHD within the skin and lung.65-67  Th17 differentiation, characterized by expression of the transcription factor RORγt, is induced by IL-6 (in the presence of transforming growth factor β [TGFβ], IL-21, and IL-23) which is secreted by numerous cell types including APCs. IL-6 inhibition is now being tested in phase 2-3 trials in clinical BMT and efficacy of this approach should be available shortly. Donor Th17 cells secrete IL-17A, IL-17F, IL-21, and IL-22, most of which have been shown to be pathogenic in preclinical systems.66,68-70  Th2 differentiation, characterized by secretion of IL-4 and expression of the transcription factor GATA3, may also be involved in GVHD, preferentially within liver and skin.67,71  In general, CD8 T cells can also differentiate along similar pathways although to date, research has focused only on T cytotoxic (Tc)-1 and Tc2 differentiation (although there is growing interest in the Tc17 lineage) and these cells appear to have differential ability to induce aGVHD and GVL.66,72  In particular, Tc2 cells appear to offer a favorable profile with regard to their ability to mediate GVL rather than GVHD.72  A more recently described follicular helper T-cell (TFH) subset is characterized by expression of the transcription factor bcl6 and secretion of IL-21. TFH cells appear to be important in generating germinal center (B-cell) reactions and alloantibody that is involved in the manifestations of chronic GVHD, particularly within the lung.73  IL-21 derived from TFH and Th17 cells thus appears an attractive target to prevent both acute and chronic GVHD.66,70 

The identification of Tregs, characterized by expression of the transcription factor FoxP3 and induced by IL-2 and TGFβ, has led to an explosion in interest in this cell subset for the induction of tolerance. It is clear from preclinical and more recent clinical studies that both acute and particularly chronic forms of GVHD are characterized by major quantitative and qualitative defects in Tregs.74,75  Therapeutic approaches to harness the regulatory potential of these cells have revolved around strategies to enhance their expansion in vivo, or to select large numbers (with or without in vitro expansion), for adoptive transfer. It became clear very early that clinical calcineurin-based immune suppression prevented Treg differentiation and expansion, a phenomenon not seen with the mammalian target of rapamycin inhibitor, rapamycin.76  This has led to trials of rapamycin both in clinical GVHD prophylaxis and treatment protocols and for the expansion of Tregs in vitro.77  Approaches to expand Tregs in vivo have focused on their preferential requirement for IL-2 and indeed exogenous low-dose IL-2 results in a marked expansion of Tregs and improvement in chronic GVHD in a proportion of patients.78  In addition, it is now possible to purify small numbers of Tregs based on CD25 selection that are nevertheless sufficient to allow profound modulation of effector T cells when transferred after haplomismatched BMT,79  an approach translated from the preclinical systems pioneered by the Negrin laboratory.76  Alternatively, large numbers of Treg can be expanded in vitro based on culture systems using IL-2 and TGF (± rapamycin)80  and the ability of these cells to modulate GVHD is the subject of intense clinical investigation. Additional novel approaches to expand Tregs using histone deacetylase (HDAC) inhibitors have also recently translated from animal models into clinical practice.81  The administration of demethylating agents after experimental BMT also has promising effects on the induction of Tregs.82  It is worth noting that IL-10 and TGFβ are major immunoregulatory cytokines produced by Treg and T-regulatory (Tr)-1 (FoxP3neg IL-10 producing) regulatory cells and IL-10 polymorphisms were among the first genetic polymorphisms to be correlated with disease outcome in GVHD.83  Approaches to enhance Treg reconstitution after BMT now represent the most promising strategy to prevent and treat GVHD and demonstrate the ability of preclinical research to guide clinical therapeutic intervention.

Cytokine administration to enhance thymopoiesis and improve immune reconstitution after T-cell–depleted transplantation is an attractive proposition supported by numerous preclinical studies (reviewed in Dudakov and van den Brink84 ). The cytokines KGF, IL-7, and IL-22 in particular appear promising and some have entered clinical testing.84,85  The use of these agents in T-cell–replete transplantation, however, remains more problematic because IL-7 and IL-22 at least may also promote GVHD.68,69 

Although the use of standard immune suppression potently inhibits donor T-cell expansion and differentiation, the fact that at least half of patients still develop GVHD demonstrates that standard therapy is less than perfect. Perhaps one of the most interesting therapeutic approaches to prevent alloreactive donor T-cell expansion is the administration of high doses of cyclophosphamide early after haplomismatched BMT to recipients otherwise receiving no immune suppression. This approach has been developed in animal models for over 4 decades (reviewed in Luznik et al86 ) and results in efficient deletion of alloreactive effector T cells while sparing Tregs.87  The translation of this approach from mice to patients has been strikingly successful88  and has permitted increased BMT activity over the last 5 years in patient groups previously untreatable. The administration of bortezomib in the peritransplant period also promotes alloreactive T-cell depletion in vivo and has also generated promising results in phase 2 studies89  that have been translated from earlier animal studies.

(3) The effector phase

aGVHD

The cumulative effect of transplant conditioning and inflammation, the subsequent promotion of alloantigen presentation, and T-cell priming and differentiation leads to an alarming immune response in which inflammatory cytokines and cytolytic T and NK cells mediate antigen-independent and -dependent killing of target tissues (Figure 3). The movement of T cells into target organs is mediated by chemokines and the recent clinical studies blocking CCR5 have demonstrated promising results that are consistent with the earlier animal studies.90  It has become clear over the last decade that inflammatory cytokines (particularly TNF and IL-1), largely the result of CD4-dependent GVHD, can induce target tissue apoptosis independent of any cognate (ie, TCR-MHC II) interaction.30  The inflammatory cytokines so characteristic of aGVHD are derived from both monocytes/macrophages and T cells themselves and cytokines from the latter clearly amplify cytokine dysregulation in the former.24  In contrast, target tissue injury in CD8-dependent GVHD does appear to primarily require cognate interaction between T cells and the target tissue (ie, TCR-MHC II).14  In this way, the cytolytic machinery within the T cell may invoke apoptosis in adjacent cells via perforin and granzyme molecules,91  TNF, and the Fas/FasL signaling pathway.92-94  Importantly, these same T cells use perforin,95  TNF-related apoptosis-inducing ligand,96  and TNF16  mediated cytolytic pathways for their GVL effects (where both CD4 and CD8 T cells require cognate TCR-MHC interactions with target leukemia)14,97  and so targeting these pathways has generally been deleterious on GVL effects. Nevertheless, these pathways may be viable targets for the treatment of severe GVHD, where GVL effects may have already effectively eradicated residual leukemia.

Figure 3

GVHD pathophysiology phase 3: the effector phase. During the effector phase of GVHD, inflammatory cytokines (IFNγ, TNF, lymphotoxin, and IL-6) derived from macrophages and T cells mediate apoptosis in target tissues, particularly within the gut. Donor Th1/Tc1, Th2/Tc2, and Th17/Tc17 cells elicit GVHD with relatively tissue-specific patterns mediated in part by their respective chemokine profiles and the relative sensitivity of the target to effector cytokines generated by each lineage. Cytolytic T and NK cells mediate antigen-dependent killing of target tissues via the perforin/granzyme and TNF member pathways.

Figure 3

GVHD pathophysiology phase 3: the effector phase. During the effector phase of GVHD, inflammatory cytokines (IFNγ, TNF, lymphotoxin, and IL-6) derived from macrophages and T cells mediate apoptosis in target tissues, particularly within the gut. Donor Th1/Tc1, Th2/Tc2, and Th17/Tc17 cells elicit GVHD with relatively tissue-specific patterns mediated in part by their respective chemokine profiles and the relative sensitivity of the target to effector cytokines generated by each lineage. Cytolytic T and NK cells mediate antigen-dependent killing of target tissues via the perforin/granzyme and TNF member pathways.

Close modal

Perhaps the dominant effector of GVHD identified in preclinical models is that of the TNF family (eg, TNF and lymphotoxin), especially in GVHD of the GI tract.16,17,32,98,99  Clinical evidence for the importance of TNF in GVHD was initially obtained in studies of donor and recipient genetic polymorphisms, which established that high donor TNF production and increased recipient responses to TNF via receptor signaling contributed to GVHD risk.100  The importance of the TNF signaling pathway in clinical BMT has been suggested by the apparent efficacy of TNF blocking agents in the prevention and treatment of GVHD.101-104  However, TNF is not present in serum of patients receiving immune suppression, in contrast to mice that do not, leading groups to use the soluble TNF receptor-1 as a surrogate.105  Furthermore, a recent study by the Blood and Marrow Transplant Clinical Trials Network suggested that TNF inhibition had limited efficacy as an adjunct to steroids (and calcineurin inhibition) in the treatment of aGVHD.106  This again highlights the fact that although significant efficacy may be seen following single-agent therapeutic intervention in animal models, concurrent standard clinical therapies may have already modified this pathway.

IL-1 is a potent proinflammatory cytokine that is important in animal models of GVHD and a large number of other acute and chronic inflammatory diseases.107,108  Early studies in the mouse demonstrated that IL-1 is an important mediator of GVHD and preclinical studies suggested that this molecule would be a target for clinical therapy.107,109  However, a phase 3 trial involving a large number of patients indicated that IL-1 blockade was insufficient to prevent GVHD, nor could it impact toxicity or overall survival.110  Given this data and the paradoxical effects of IL-18 in GVHD,111  although the inflammasome may be involved in GVHD in animal models,112  it would appear an unattractive target in clinical BMT. Likewise, it is clear that IFNγ plays complex, organ-specific, pathogenic and protective effects in GVHD63  and is inhibited by calcineurin inhibition113  such that it appears a complex therapeutic target in isolation.

Animal models clearly demonstrate IL-6 as a major mediator of GVHD with both direct effects on target tissue and important effects on Th17 differentiation.13,114  Importantly, clinical data does suggest that IL-6 is dysregulated in BMT patients,115  making it a more attractive target, currently under investigation in clinical trials.

Chronic GVHD

Our understanding of the mechanisms of chronic GVHD is very limited. Because chronic GVHD is characterized by end-organ fibrosis, it is of no surprise that the effector pathways differ from those mediating aGVHD at both a cellular and molecular level. However, like aGVHD, it also appears that differing pathways of disease operate within individual target organs. Certainly fibrosis in the skin appears to be an IL-17–dependent process66  whereas aberrant B-cell function and alloantibody deposition appear important in lung and liver.73  Whether the latter process also involves Th17 and/or TFH differentiation remains to be determined. Certainly preclinical studies have shown TGFβ, derived from tissue macrophages, as a major mediator of chronic GVHD.116  However, the critical role for TGFβ in regulatory responses make therapeutic inhibition a double-edged sword. Strategies for the prevention of chronic GVHD will thus likely need to focus on an aberrant pathway of T-cell differentiation or the final common initiating cell, putatively macrophages and/or B and plasma cells.

Over the last 60 years, animal models have played a critical role in shaping our understanding of GVHD and GVL responses and this is perhaps their greatest contribution to the field. Nonetheless, they have also generated major new paradigms that have instructed our clinical practice and examples of these contributions are shown in Table 1. As we move forward, it is useful to consider where the next advances will come from and how we should assess the potential of a therapeutic intervention defined in a preclinical study to translate into clinical practice. There are some obvious criteria and these are shown in Table 2. Based on these criteria, we suggest that therapeutics aimed at Treg reconstitution, alloreactive and naive T-cell depletion, and IL-6, IL-12p40, and IL-21 inhibition are the most immediately promising candidates and should be a high priority for clinical trials.

Table 1

Examples of the concepts generated in animal models

Concepts generated
Established clinical procedures 
 BMT 
 GVL 
 T-cell depletion 
 Peritransplant immune suppression 
 Donor lymphocyte infusions 
 RIC 
 TLI-based conditioning 
 Posttransplant cyclophosphamide 
Promising concepts under investigation 
 Treg expansion (eg, adoptive transfer, IL-2, HDAC inhibition, demethylating agents) 
 Naïve T-cell depletion 
 Alternative alloreactive T-cell depletion strategies (eg, bortezomib) 
 Cytokine inhibition (eg, IL-6, IL-12, IL-21) 
 Chemokine blockade (CCR5) 
 CAR-expressing donor lymphocyte infusions 
 Costimulation blockade 
 Manipulation of microbiota 
Concepts generated
Established clinical procedures 
 BMT 
 GVL 
 T-cell depletion 
 Peritransplant immune suppression 
 Donor lymphocyte infusions 
 RIC 
 TLI-based conditioning 
 Posttransplant cyclophosphamide 
Promising concepts under investigation 
 Treg expansion (eg, adoptive transfer, IL-2, HDAC inhibition, demethylating agents) 
 Naïve T-cell depletion 
 Alternative alloreactive T-cell depletion strategies (eg, bortezomib) 
 Cytokine inhibition (eg, IL-6, IL-12, IL-21) 
 Chemokine blockade (CCR5) 
 CAR-expressing donor lymphocyte infusions 
 Costimulation blockade 
 Manipulation of microbiota 

CAR, chimeric antigen receptor; TLI, total lymphoid irradiation.

Table 2

Predicting the translation of preclinical therapeutic strategies

Will a new therapeutic strategy translate into the clinic?
1. Is the pathway in question still active in patients (or animals) receiving immune suppression? If not, does preclinical evidence suggest the inhibition of the pathway in isolation will be feasible and more efficacious than current immune suppression? 
2. Is the effect large (complete) rather than incremental (partial)? 
3. Does the intervention inhibit GVHD at multiple stages of its pathophysiology, and preferably early? 
4. Is the pathway active in both class I– and class II–dependent GVHD? If not, it should at least not have opposing effects in CD8 and CD4 T-cell–dependent GVHD. 
5. Are there potential toxicities in humans that might reasonably not be evident in the animal model? 
6. Are there detrimental effects of the intervention on GVL and pathogenic-specific immunity and if so is this manageable and outweighed by the potential benefits in treating GVHD? 
7. Bearing in mind that pharmaceutical companies have minimal interest in BMT as a sole market, is the pathway operative in other disease settings such that the generation and development of reagents to the clinic is likely to be commercially viable? 
Will a new therapeutic strategy translate into the clinic?
1. Is the pathway in question still active in patients (or animals) receiving immune suppression? If not, does preclinical evidence suggest the inhibition of the pathway in isolation will be feasible and more efficacious than current immune suppression? 
2. Is the effect large (complete) rather than incremental (partial)? 
3. Does the intervention inhibit GVHD at multiple stages of its pathophysiology, and preferably early? 
4. Is the pathway active in both class I– and class II–dependent GVHD? If not, it should at least not have opposing effects in CD8 and CD4 T-cell–dependent GVHD. 
5. Are there potential toxicities in humans that might reasonably not be evident in the animal model? 
6. Are there detrimental effects of the intervention on GVL and pathogenic-specific immunity and if so is this manageable and outweighed by the potential benefits in treating GVHD? 
7. Bearing in mind that pharmaceutical companies have minimal interest in BMT as a sole market, is the pathway operative in other disease settings such that the generation and development of reagents to the clinic is likely to be commercially viable? 

The authors acknowledge Madeleine Flynn, Queensland Institute of Medical Research Berghofer Medical Research Institute, for assistance with the illustrations. K.A.M. is a National Health and Medical Research Council Clinical Training Fellow, K.P.A.M. is a Cancer Council Queensland Senior Research Fellow, and G.R.H. is a National Health and Medical Research Council Australia Fellow and a Queensland Health Senior Clinical Research Fellow.

Contribution: K.A.M., K.P.A.M., and G.R.H. all contributed to the writing of the manuscript.

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

Correspondence: Geoffrey R. Hill, QIMR Berghofer Medical Research Institute, 300 Herston Rd, Herston, QLD, Australia, 4006; e-mail: geoff.hill@qimrberghofer.edu.au.

1
Jacobson
 
LO
Simmons
 
EL
Marks
 
EK
Robson
 
MJ
Bethard
 
WF
Gaston
 
EO
The role of the spleen in radiation injury and recovery.
J Lab Clin Med
1950
, vol. 
35
 
5
(pg. 
746
-
770
)
2
Lorenz
 
E
Uphoff
 
D
Reid
 
TR
Shelton
 
E
Modification of irradiation injury in mice and guinea pigs by bone marrow injections.
J Natl Cancer Inst
1951
, vol. 
12
 
1
(pg. 
197
-
201
)
3
Barnes
 
DW
Corp
 
MJ
Loutit
 
JF
Neal
 
FE
Treatment of murine leukaemia with X rays and homologous bone marrow; preliminary communication.
BMJ
1956
, vol. 
2
 
4993
(pg. 
626
-
627
)
4
Gatti
 
RA
Meuwissen
 
HJ
Allen
 
HD
Hong
 
R
Good
 
RA
Immunological reconstitution of sex-linked lymphopenic immunological deficiency.
Lancet
1968
, vol. 
2
 
7583
(pg. 
1366
-
1369
)
5
Center for International Blood and Marrow Transplant Research
Current uses and outcomes of hematopoietic stem cell transplantation 2012: summary slides.
 
CIBMTR Web site. http://www.cibmtr.org. Accessed May 1, 2014
6
Gooley
 
TA
Chien
 
JW
Pergam
 
SA
, et al. 
Reduced mortality after allogeneic hematopoietic-cell transplantation.
N Engl J Med
2010
, vol. 
363
 
22
(pg. 
2091
-
2101
)
7
Ratanatharathorn
 
V
Nash
 
RA
Przepiorka
 
D
, et al. 
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. 1998;92(7):2303-2314
8
Theurich
 
S
Fischmann
 
H
Shimabukuro-Vornhagen
 
A
, et al. 
Polyclonal anti-thymocyte globulins for the prophylaxis of graft-versus-host disease after allogeneic stem cell or bone marrow transplantation in adults.
Cochrane Database Syst Rev
2012
, vol. 
9
 pg. 
CD009159
 
9
Reddy
 
P
Negrin
 
R
Hill
 
GR
Mouse models of bone marrow transplantation.
Biol Blood Marrow Transplant
2008
, vol. 
14
 
1 suppl 1
(pg. 
129
-
135
)
10
Storb
 
R
Thomas
 
ED
Graft-versus-host disease in dog and man: the Seattle experience.
Immunol Rev
1985
, vol. 
88
 (pg. 
215
-
238
)
11
Kochenderfer
 
JN
Rosenberg
 
SA
Treating B-cell cancer with T cells expressing anti-CD19 chimeric antigen receptors.
Nat Rev Clin Oncol
2013
, vol. 
10
 
5
(pg. 
267
-
276
)
12
Bensinger
 
WI
Martin
 
PJ
Storer
 
B
, et al. 
Transplantation of bone marrow as compared with peripheral-blood cells from HLA-identical relatives in patients with hematologic cancers.
N Engl J Med
2001
, vol. 
344
 
3
(pg. 
175
-
181
)
13
Tawara
 
I
Koyama
 
M
Liu
 
C
, et al. 
Interleukin-6 modulates graft-versus-host responses after experimental allogeneic bone marrow transplantation.
Clin Cancer Res
2011
, vol. 
17
 
1
(pg. 
77
-
88
)
14
Matte-Martone
 
C
Liu
 
J
Jain
 
D
McNiff
 
J
Shlomchik
 
WD
CD8+ but not CD4+ T cells require cognate interactions with target tissues to mediate GVHD across only minor H antigens, whereas both CD4+ and CD8+ T cells require direct leukemic contact to mediate GVL.
Blood
2008
, vol. 
111
 
7
(pg. 
3884
-
3892
)
15
Hill
 
GR
Cooke
 
KR
Teshima
 
T
, et al. 
Interleukin-11 promotes T cell polarization and prevents acute graft-versus-host disease after allogeneic bone marrow transplantation.
J Clin Invest
1998
, vol. 
102
 
1
(pg. 
115
-
123
)
16
Hill
 
GR
Teshima
 
T
Gerbitz
 
A
, et al. 
Differential roles of IL-1 and TNF-alpha on graft-versus-host disease and graft versus leukemia.
J Clin Invest
1999
, vol. 
104
 
4
(pg. 
459
-
467
)
17
Markey
 
KA
Burman
 
AC
Banovic
 
T
, et al. 
Soluble lymphotoxin is an important effector molecule in GVHD and GVL.
Blood
2010
, vol. 
115
 
1
(pg. 
122
-
132
)
18
Matte
 
CC
Cormier
 
J
Anderson
 
BE
, et al. 
Graft-versus-leukemia in a retrovirally induced murine CML model: mechanisms of T-cell killing.
Blood
2004
, vol. 
103
 
11
(pg. 
4353
-
4361
)
19
Edinger
 
M
Hoffmann
 
P
Ermann
 
J
, et al. 
CD4+CD25+ regulatory T cells preserve graft-versus-tumor activity while inhibiting graft-versus-host disease after bone marrow transplantation.
Nat Med
2003
, vol. 
9
 
9
(pg. 
1144
-
1150
)
20
Zhang
 
P
Tey
 
SK
Koyama
 
M
, et al. 
Induced regulatory T cells promote tolerance when stabilized by rapamycin and IL-2 in vivo.
J Immunol
2013
, vol. 
191
 
10
(pg. 
5291
-
5303
)
21
Ferrara
 
JL
Deeg
 
HJ
Graft-versus-host disease.
N Engl J Med
1991
, vol. 
324
 
10
(pg. 
667
-
674
)
22
Nestel
 
FP
Price
 
KS
Seemayer
 
TA
Lapp
 
WS
Macrophage priming and lipopolysaccharide-triggered release of tumor necrosis factor alpha during graft-versus-host disease.
J Exp Med
1992
, vol. 
175
 
2
(pg. 
405
-
413
)
23
Hill
 
GR
Crawford
 
JM
Cooke
 
KR
Brinson
 
YS
Pan
 
L
Ferrara
 
JL
Total body irradiation and acute graft-versus-host disease: the role of gastrointestinal damage and inflammatory cytokines.
Blood
1997
, vol. 
90
 
8
(pg. 
3204
-
3213
)
24
Hill
 
GR
Ferrara
 
JL
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
2000
, vol. 
95
 
9
(pg. 
2754
-
2759
)
25
Cooke
 
KR
Gerbitz
 
A
Crawford
 
JM
, et al. 
LPS antagonism reduces graft-versus-host disease and preserves graft-versus-leukemia activity after experimental bone marrow transplantation.
J Clin Invest
2001
, vol. 
107
 
12
(pg. 
1581
-
1589
)
26
Jenq
 
RR
Ubeda
 
C
Taur
 
Y
, et al. 
Regulation of intestinal inflammation by microbiota following allogeneic bone marrow transplantation.
J Exp Med
2012
, vol. 
209
 
5
(pg. 
903
-
911
)
27
Eriguchi
 
Y
Takashima
 
S
Oka
 
H
, et al. 
Graft-versus-host disease disrupts intestinal microbial ecology by inhibiting Paneth cell production of α-defensins.
Blood
2012
, vol. 
120
 
1
(pg. 
223
-
231
)
28
van Bekkum
 
DW
Roodenburg
 
J
Heidt
 
PJ
van der Waaij
 
D
Mitigation of secondary disease of allogeneic mouse radiation chimeras by modification of the intestinal microflora.
J Natl Cancer Inst
1974
, vol. 
52
 
2
(pg. 
401
-
404
)
29
Xun
 
CQ
Thompson
 
JS
Jennings
 
CD
Brown
 
SA
Widmer
 
MB
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
1994
, vol. 
83
 
8
(pg. 
2360
-
2367
)
30
Teshima
 
T
Ordemann
 
R
Reddy
 
P
, et al. 
Acute graft-versus-host disease does not require alloantigen expression on host epithelium.
Nat Med
2002
, vol. 
8
 
6
(pg. 
575
-
581
)
31
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
, vol. 
18
 
1
(pg. 
135
-
142
)
32
Hill
 
GR
Teshima
 
T
Rebel
 
VI
, et al. 
The p55 TNF-alpha receptor plays a critical role in T cell alloreactivity.
J Immunol
2000
, vol. 
164
 
2
(pg. 
656
-
663
)
33
Chakraverty
 
R
Côté
 
D
Buchli
 
J
, et al. 
An inflammatory checkpoint regulates recruitment of graft-versus-host reactive T cells to peripheral tissues.
J Exp Med
2006
, vol. 
203
 
8
(pg. 
2021
-
2031
)
34
Yu
 
C
Storb
 
R
Mathey
 
B
, et al. 
DLA-identical bone marrow grafts after low-dose total body irradiation: effects of high-dose corticosteroids and cyclosporine on engraftment.
Blood
1995
, vol. 
86
 
11
(pg. 
4376
-
4381
)
35
Storb
 
R
Yu
 
C
Wagner
 
JL
, et al. 
Stable mixed hematopoietic chimerism in DLA-identical littermate dogs given sublethal total body irradiation before and pharmacological immunosuppression after marrow transplantation.
Blood
1997
, vol. 
89
 
8
(pg. 
3048
-
3054
)
36
Yu
 
C
Seidel
 
K
Nash
 
RA
, et al. 
Synergism between mycophenolate mofetil and cyclosporine in preventing graft-versus-host disease among lethally irradiated dogs given DLA-nonidentical unrelated marrow grafts.
Blood
1998
, vol. 
91
 
7
(pg. 
2581
-
2587
)
37
Giralt
 
S
Estey
 
E
Albitar
 
M
, et al. 
Engraftment of allogeneic hematopoietic progenitor cells with purine analog-containing chemotherapy: harnessing graft-versus-leukemia without myeloablative therapy.
Blood
1997
, vol. 
89
 
12
(pg. 
4531
-
4536
)
38
Khouri
 
IF
Keating
 
M
Körbling
 
M
, et al. 
Transplant-lite: induction of graft-versus-malignancy using fludarabine-based nonablative chemotherapy and allogeneic blood progenitor-cell transplantation as treatment for lymphoid malignancies.
J Clin Oncol
1998
, vol. 
16
 
8
(pg. 
2817
-
2824
)
39
Pillai
 
AB
George
 
TI
Dutt
 
S
Strober
 
S
Host natural killer T cells induce an interleukin-4-dependent expansion of donor CD4+CD25+Foxp3+ T regulatory cells that protects against graft-versus-host disease.
Blood
2009
, vol. 
113
 
18
(pg. 
4458
-
4467
)
40
Kohrt
 
HE
Turnbull
 
BB
Heydari
 
K
, et al. 
TLI and ATG conditioning with low risk of graft-versus-host disease retains antitumor reactions after allogeneic hematopoietic cell transplantation from related and unrelated donors.
Blood
2009
, vol. 
114
 
5
(pg. 
1099
-
1109
)
41
Antin
 
JH
Lee
 
SJ
Neuberg
 
D
, et al. 
A phase I/II double-blind, placebo-controlled study of recombinant human interleukin-11 for mucositis and acute GVHD prevention in allogeneic stem cell transplantation.
Bone Marrow Transplant
2002
, vol. 
29
 
5
(pg. 
373
-
377
)
42
Levine
 
JE
Blazar
 
BR
DeFor
 
T
Ferrara
 
JL
Weisdorf
 
DJ
Long-term follow-up of a phase I/II randomized, placebo-controlled trial of palifermin to prevent graft-versus-host disease (GVHD) after related donor allogeneic hematopoietic cell transplantation (HCT).
Biol Blood Marrow Transplant
2008
, vol. 
14
 
9
(pg. 
1017
-
1021
)
43
Beelen
 
DW
Elmaagacli
 
A
Müller
 
KD
Hirche
 
H
Schaefer
 
UW
Influence of intestinal bacterial decontamination using metronidazole and ciprofloxacin or ciprofloxacin alone on the development of acute graft-versus-host disease after marrow transplantation in patients with hematologic malignancies: final results and long-term follow-up of an open-label prospective randomized trial.
Blood
1999
, vol. 
93
 
10
(pg. 
3267
-
3275
)
44
Atarashi
 
K
Tanoue
 
T
Oshima
 
K
, et al. 
Treg induction by a rationally selected mixture of Clostridia strains from the human microbiota.
Nature
2013
, vol. 
500
 
7461
(pg. 
232
-
236
)
45
Toubai
 
T
Tawara
 
I
Sun
 
Y
, et al. 
Induction of acute GVHD by sex-mismatched H-Y antigens in the absence of functional radiosensitive host hematopoietic-derived antigen presenting cells.
Blood
2012
, vol. 
119
 
16
(pg. 
3844
-
3853
)
46
Shlomchik
 
WD
Couzens
 
MS
Tang
 
CB
, et al. 
Prevention of graft versus host disease by inactivation of host antigen-presenting cells.
Science
1999
, vol. 
285
 
5426
(pg. 
412
-
415
)
47
Matte
 
CC
Liu
 
J
Cormier
 
J
, et al. 
Donor APCs are required for maximal GVHD but not for GVL.
Nat Med
2004
, vol. 
10
 
9
(pg. 
987
-
992
)
48
Li
 
H
Demetris
 
AJ
McNiff
 
J
, et al. 
Profound depletion of host conventional dendritic cells, plasmacytoid dendritic cells, and B cells does not prevent graft-versus-host disease induction.
J Immunol
2012
, vol. 
188
 
8
(pg. 
3804
-
3811
)
49
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
, vol. 
185
 
7
(pg. 
3866
-
3872
)
50
Rowe
 
V
Banovic
 
T
MacDonald
 
KP
, et al. 
Host B cells produce IL-10 following TBI and attenuate acute GVHD after allogeneic bone marrow transplantation.
Blood
2006
, vol. 
108
 
7
(pg. 
2485
-
2492
)
51
MacDonald
 
KP
Palmer
 
JS
Cronau
 
S
, et al. 
An antibody against the colony-stimulating factor 1 receptor depletes the resident subset of monocytes and tissue- and tumor-associated macrophages but does not inhibit inflammation.
Blood
2010
, vol. 
116
 
19
(pg. 
3955
-
3963
)
52
Hashimoto
 
D
Chow
 
A
Greter
 
M
, et al. 
Pretransplant CSF-1 therapy expands recipient macrophages and ameliorates GVHD after allogeneic hematopoietic cell transplantation.
J Exp Med
2011
, vol. 
208
 
5
(pg. 
1069
-
1082
)
53
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
, vol. 
109
 
11
(pg. 
5049
-
5057
)
54
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
, vol. 
113
 
22
(pg. 
5644
-
5649
)
55
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
, vol. 
118
 
24
(pg. 
6426
-
6437
)
56
Markey
 
KA
Koyama
 
M
Kuns
 
RD
, et al. 
Immune insufficiency during GVHD is due to defective antigen presentation within dendritic cell subsets.
Blood
2012
, vol. 
119
 
24
(pg. 
5918
-
5930
)
57
Kinnear
 
G
Jones
 
ND
Wood
 
KJ
Costimulation blockade: current perspectives and implications for therapy.
Transplantation
2013
, vol. 
95
 
4
(pg. 
527
-
535
)
58
Anderson
 
BE
McNiff
 
J
Yan
 
J
, et al. 
Memory CD4+ T cells do not induce graft-versus-host disease.
J Clin Invest
2003
, vol. 
112
 
1
(pg. 
101
-
108
)
59
Chen
 
BJ
Cui
 
X
Sempowski
 
GD
Liu
 
C
Chao
 
NJ
Transfer of allogeneic CD62L- memory T cells without graft-versus-host disease.
Blood
2004
, vol. 
103
 
4
(pg. 
1534
-
1541
)
60
Foster
 
AE
Marangolo
 
M
Sartor
 
MM
, et al. 
Human CD62L- memory T cells are less responsive to alloantigen stimulation than CD62L+ naive T cells: potential for adoptive immunotherapy and allodepletion.
Blood
2004
, vol. 
104
 
8
(pg. 
2403
-
2409
)
61
Bleakley
 
M
Otterud
 
BE
Richardt
 
JL
, et al. 
Leukemia-associated minor histocompatibility antigen discovery using T-cell clones isolated by in vitro stimulation of naive CD8+ T cells.
Blood
2010
, vol. 
115
 
23
(pg. 
4923
-
4933
)
62
Thomson
 
AW
Bonham
 
CA
Zeevi
 
A
Mode of action of tacrolimus (FK506): molecular and cellular mechanisms.
Ther Drug Monit
1995
, vol. 
17
 
6
(pg. 
584
-
591
)
63
Burman
 
AC
Banovic
 
T
Kuns
 
RD
, et al. 
IFNgamma differentially controls the development of idiopathic pneumonia syndrome and GVHD of the gastrointestinal tract.
Blood
2007
, vol. 
110
 
3
(pg. 
1064
-
1072
)
64
Robb
 
RJ
Hill
 
GR
The interferon-dependent orchestration of innate and adaptive immunity after transplantation.
Blood
2012
, vol. 
119
 
23
(pg. 
5351
-
5358
)
65
Carlson
 
MJ
West
 
ML
Coghill
 
JM
Panoskaltsis-Mortari
 
A
Blazar
 
BR
Serody
 
JS
In vitro-differentiated TH17 cells mediate lethal acute graft-versus-host disease with severe cutaneous and pulmonary pathologic manifestations.
Blood
2009
, vol. 
113
 
6
(pg. 
1365
-
1374
)
66
Hill
 
GR
Olver
 
SD
Kuns
 
RD
, et al. 
Stem cell mobilization with G-CSF induces type 17 differentiation and promotes scleroderma.
Blood
2010
, vol. 
116
 
5
(pg. 
819
-
828
)
67
Yi
 
T
Chen
 
Y
Wang
 
L
, et al. 
Reciprocal differentiation and tissue-specific pathogenesis of Th1, Th2, and Th17 cells in graft-versus-host disease.
Blood
2009
, vol. 
114
 
14
(pg. 
3101
-
3112
)
68
Serody
 
JS
Hill
 
GR
The IL-17 differentiation pathway and its role in transplant outcome.
Biol Blood Marrow Transplant
2012
, vol. 
18
 
1 suppl
(pg. 
S56
-
S61
)
69
Couturier
 
M
Lamarthée
 
B
Arbez
 
J
, et al. 
IL-22 deficiency in donor T cells attenuates murine acute graft-versus-host disease mortality while sparing the graft-versus-leukemia effect.
Leukemia
2013
, vol. 
27
 
7
(pg. 
1527
-
1537
)
70
Bucher
 
C
Koch
 
L
Vogtenhuber
 
C
, et al. 
IL-21 blockade reduces graft-versus-host disease mortality by supporting inducible T regulatory cell generation.
Blood
2009
, vol. 
114
 
26
(pg. 
5375
-
5384
)
71
Nikolic
 
B
Lee
 
S
Bronson
 
RT
Grusby
 
MJ
Sykes
 
M
Th1 and Th2 mediate acute graft-versus-host disease, each with distinct end-organ targets.
J Clin Invest
2000
, vol. 
105
 
9
(pg. 
1289
-
1298
)
72
Fowler
 
DH
Breglio
 
J
Nagel
 
G
Eckhaus
 
MA
Gress
 
RE
Allospecific CD8+ Tc1 and Tc2 populations in graft-versus-leukemia effect and graft-versus-host disease.
J Immunol
1996
, vol. 
157
 
11
(pg. 
4811
-
4821
)
73
Srinivasan
 
M
Flynn
 
R
Price
 
A
, et al. 
Donor B-cell alloantibody deposition and germinal center formation are required for the development of murine chronic GVHD and bronchiolitis obliterans.
Blood
2012
, vol. 
119
 
6
(pg. 
1570
-
1580
)
74
Cohen
 
JL
Trenado
 
A
Vasey
 
D
Klatzmann
 
D
Salomon
 
BL
CD4(+)CD25(+) immunoregulatory T cells: new therapeutics for graft-versus-host disease.
J Exp Med
2002
, vol. 
196
 
3
(pg. 
401
-
406
)
75
Matsuoka
 
K
Kim
 
HT
McDonough
 
S
, et al. 
Altered regulatory T cell homeostasis in patients with CD4+ lymphopenia following allogeneic hematopoietic stem cell transplantation.
J Clin Invest
2010
, vol. 
120
 
5
(pg. 
1479
-
1493
)
76
Zeiser
 
R
Nguyen
 
VH
Beilhack
 
A
, et al. 
Inhibition of CD4+CD25+ regulatory T-cell function by calcineurin-dependent interleukin-2 production.
Blood
2006
, vol. 
108
 
1
(pg. 
390
-
399
)
77
Abouelnasr
 
A
Roy
 
J
Cohen
 
S
Kiss
 
T
Lachance
 
S
Defining the role of sirolimus in the management of graft-versus-host disease: from prophylaxis to treatment.
Biol Blood Marrow Transplant
2013
, vol. 
19
 
1
(pg. 
12
-
21
)
78
Koreth
 
J
Matsuoka
 
K
Kim
 
HT
, et al. 
Interleukin-2 and regulatory T cells in graft-versus-host disease.
N Engl J Med
2011
, vol. 
365
 
22
(pg. 
2055
-
2066
)
79
Di Ianni
 
M
Falzetti
 
F
Carotti
 
A
, et al. 
Tregs prevent GVHD and promote immune reconstitution in HLA-haploidentical transplantation.
Blood
2011
, vol. 
117
 
14
(pg. 
3921
-
3928
)
80
Hippen
 
KL
Merkel
 
SC
Schirm
 
DK
, et al. 
Massive ex vivo expansion of human natural regulatory T cells (T(regs)) with minimal loss of in vivo functional activity.
Sci Transl Med
2011
, vol. 
3
 
83
pg. 
83ra41
 
81
Choi
 
SW
Braun
 
T
Chang
 
L
, et al. 
Vorinostat plus tacrolimus and mycophenolate to prevent graft-versus-host disease after related-donor reduced-intensity conditioning allogeneic haemopoietic stem-cell transplantation: a phase 1/2 trial.
Lancet Oncol
2014
, vol. 
15
 
1
(pg. 
87
-
95
)
82
Choi
 
J
Ritchey
 
J
Prior
 
JL
, et al. 
In vivo administration of hypomethylating agents mitigate graft-versus-host disease without sacrificing graft-versus-leukemia.
Blood
2010
, vol. 
116
 
1
(pg. 
129
-
139
)
83
Lin
 
MT
Storer
 
B
Martin
 
PJ
, et al. 
Relation of an interleukin-10 promoter polymorphism to graft-versus-host disease and survival after hematopoietic-cell transplantation.
N Engl J Med
2003
, vol. 
349
 
23
(pg. 
2201
-
2210
)
84
Dudakov
 
JA
van den Brink
 
MR
Greater than the sum of their parts: combination strategies for immune regeneration following allogeneic hematopoietic stem cell transplantation.
Best Pract Res Clin Haematol
2011
, vol. 
24
 
3
(pg. 
467
-
476
)
85
Dudakov
 
JA
Hanash
 
AM
Jenq
 
RR
, et al. 
Interleukin-22 drives endogenous thymic regeneration in mice.
Science
2012
, vol. 
336
 
6077
(pg. 
91
-
95
)
86
Luznik
 
L
O’Donnell
 
PV
Fuchs
 
EJ
Post-transplantation cyclophosphamide for tolerance induction in HLA-haploidentical bone marrow transplantation.
Semin Oncol
2012
, vol. 
39
 
6
(pg. 
683
-
693
)
87
Kanakry
 
CG
Ganguly
 
S
Zahurak
 
M
, et al. 
Aldehyde dehydrogenase expression drives human regulatory T cell resistance to posttransplantation cyclophosphamide.
Sci Transl Med
2013
 
5(211):211ra157
88
Bashey
 
A
Zhang
 
X
Sizemore
 
CA
, et al. 
T-cell-replete HLA-haploidentical hematopoietic transplantation for hematologic malignancies using post-transplantation cyclophosphamide results in outcomes equivalent to those of contemporaneous HLA-matched related and unrelated donor transplantation.
J Clin Oncol
2013
, vol. 
31
 
10
(pg. 
1310
-
1316
)
89
Koreth
 
J
Stevenson
 
KE
Kim
 
HT
, et al. 
Bortezomib-based graft-versus-host disease prophylaxis in HLA-mismatched unrelated donor transplantation.
J Clin Oncol
2012
, vol. 
30
 
26
(pg. 
3202
-
3208
)
90
Reshef
 
R
Luger
 
SM
Hexner
 
EO
, et al. 
Blockade of lymphocyte chemotaxis in visceral graft-versus-host disease.
N Engl J Med
2012
, vol. 
367
 
2
(pg. 
135
-
145
)
91
Graubert
 
TA
DiPersio
 
JF
Russell
 
JH
Ley
 
TJ
Perforin/granzyme-dependent and independent mechanisms are both important for the development of graft-versus-host disease after murine bone marrow transplantation.
J Clin Invest
1997
, vol. 
100
 
4
(pg. 
904
-
911
)
92
Braun
 
MY
Lowin
 
B
French
 
L
Acha-Orbea
 
H
Tschopp
 
J
Cytotoxic T cells deficient in both functional fas ligand and perforin show residual cytolytic activity yet lose their capacity to induce lethal acute graft-versus-host disease.
J Exp Med
1996
, vol. 
183
 
2
(pg. 
657
-
661
)
93
Hattori
 
K
Hirano
 
T
Miyajima
 
H
, et al. 
Differential effects of anti-Fas ligand and anti-tumor necrosis factor alpha antibodies on acute graft-versus-host disease pathologies.
Blood
1998
, vol. 
91
 
11
(pg. 
4051
-
4055
)
94
Maeda
 
Y
Levy
 
RB
Reddy
 
P
, et al. 
Both perforin and Fas ligand are required for the regulation of alloreactive CD8+ T cells during acute graft-versus-host disease.
Blood
2005
, vol. 
105
 
5
(pg. 
2023
-
2027
)
95
Pan
 
L
Teshima
 
T
Hill
 
GR
, et al. 
Granulocyte colony-stimulating factor-mobilized allogeneic stem cell transplantation maintains graft-versus-leukemia effects through a perforin-dependent pathway while preventing graft-versus-host disease.
Blood
1999
, vol. 
93
 
12
(pg. 
4071
-
4078
)
96
Schmaltz
 
C
Alpdogan
 
O
Kappel
 
BJ
, et al. 
T cells require TRAIL for optimal graft-versus-tumor activity.
Nat Med
2002
, vol. 
8
 
12
(pg. 
1433
-
1437
)
97
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
, vol. 
11
 
11
(pg. 
1244
-
1249
)
98
Schmaltz
 
C
Alpdogan
 
O
Muriglan
 
SJ
, et al. 
Donor T cell-derived TNF is required for graft-versus-host disease and graft-versus-tumor activity after bone marrow transplantation.
Blood
2003
, vol. 
101
 
6
(pg. 
2440
-
2445
)
99
Speiser
 
DE
Bachmann
 
MF
Frick
 
TW
, et al. 
TNF receptor p55 controls early acute graft-versus-host disease.
J Immunol
1997
, vol. 
158
 
11
(pg. 
5185
-
5190
)
100
Wilson
 
AG
Symons
 
JA
McDowell
 
TL
McDevitt
 
HO
Duff
 
GW
Effects of a polymorphism in the human tumor necrosis factor alpha promoter on transcriptional activation.
Proc Natl Acad Sci U S A
1997
, vol. 
94
 
7
(pg. 
3195
-
3199
)
101
Levine
 
JE
Paczesny
 
S
Mineishi
 
S
, et al. 
Etanercept plus methylprednisolone as initial therapy for acute graft-versus-host disease.
Blood
2008
, vol. 
111
 
4
(pg. 
2470
-
2475
)
102
Kennedy
 
GA
Butler
 
J
Western
 
R
Morton
 
J
Durrant
 
S
Hill
 
GR
Combination antithymocyte globulin and soluble TNFalpha inhibitor (etanercept) +/- mycophenolate mofetil for treatment of steroid refractory acute graft-versus-host disease.
Bone Marrow Transplant
2006
, vol. 
37
 
12
(pg. 
1143
-
1147
)
103
Hervé
 
P
Flesch
 
M
Tiberghien
 
P
, et al. 
Phase I-II trial of a monoclonal anti-tumor necrosis factor alpha antibody for the treatment of refractory severe acute graft-versus-host disease.
Blood
1992
, vol. 
79
 
12
(pg. 
3362
-
3368
)
104
Holler
 
E
Kolb
 
HJ
Wilmanns
 
W
Treatment of GVHD—TNF-antibodies and related antagonists.
Bone Marrow Transplant
1993
, vol. 
12
 
suppl 3
(pg. 
S29
-
S31
)
105
Choi
 
SW
Kitko
 
CL
Braun
 
T
, et al. 
Change in plasma tumor necrosis factor receptor 1 levels in the first week after myeloablative allogeneic transplantation correlates with severity and incidence of GVHD and survival.
Blood
2008
, vol. 
112
 
4
(pg. 
1539
-
1542
)
106
Alousi
 
AM
Weisdorf
 
DJ
Logan
 
BR
, et al. 
Blood and Marrow Transplant Clinical Trials Network
Etanercept, mycophenolate, denileukin, or pentostatin plus corticosteroids for acute graft-versus-host disease: a randomized phase 2 trial from the Blood and Marrow Transplant Clinical Trials Network.
Blood
2009
, vol. 
114
 
3
(pg. 
511
-
517
)
107
Abhyankar
 
S
Gilliland
 
DG
Ferrara
 
JL
Interleukin-1 is a critical effector molecule during cytokine dysregulation in graft versus host disease to minor histocompatibility antigens.
Transplantation
1993
, vol. 
56
 
6
(pg. 
1518
-
1523
)
108
Dinarello
 
CA
Interleukin-1 in the pathogenesis and treatment of inflammatory diseases.
Blood
2011
, vol. 
117
 
14
(pg. 
3720
-
3732
)
109
McCarthy
 
PL
Abhyankar
 
S
Neben
 
S
, et al. 
Inhibition of interleukin-1 by an interleukin-1 receptor antagonist prevents graft-versus-host disease.
Blood
1991
, vol. 
78
 
8
(pg. 
1915
-
1918
)
110
Antin
 
JH
Weisdorf
 
D
Neuberg
 
D
, et al. 
Interleukin-1 blockade does not prevent acute graft-versus-host disease: results of a randomized, double-blind, placebo-controlled trial of interleukin-1 receptor antagonist in allogeneic bone marrow transplantation.
Blood
2002
, vol. 
100
 
10
(pg. 
3479
-
3482
)
111
Min
 
CK
Maeda
 
Y
Lowler
 
K
, et al. 
Paradoxical effects of interleukin-18 on the severity of acute graft-versus-host disease mediated by CD4+ and CD8+ T-cell subsets after experimental allogeneic bone marrow transplantation.
Blood
2004
, vol. 
104
 
10
(pg. 
3393
-
3399
)
112
Jankovic
 
D
Ganesan
 
J
Bscheider
 
M
, et al. 
The Nlrp3 inflammasome regulates acute graft-versus-host disease.
J Exp Med
2013
, vol. 
210
 
10
(pg. 
1899
-
1910
)
113
Lemaître
 
PH
Vokaer
 
B
Charbonnier
 
LM
, et al. 
Cyclosporine A drives a Th17- and Th2-mediated posttransplant obliterative airway disease.
Am J Transplant
2013
, vol. 
13
 
3
(pg. 
611
-
620
)
114
Chen
 
X
Das
 
R
Komorowski
 
R
, et al. 
Blockade of interleukin-6 signaling augments regulatory T-cell reconstitution and attenuates the severity of graft-versus-host disease.
Blood
2009
, vol. 
114
 
4
(pg. 
891
-
900
)
115
Symington
 
FW
Symington
 
BE
Liu
 
PY
Viguet
 
H
Santhanam
 
U
Sehgal
 
PB
The relationship of serum IL-6 levels to acute graft-versus-host disease and hepatorenal disease after human bone marrow transplantation.
Transplantation
1992
, vol. 
54
 
3
(pg. 
457
-
462
)
116
Banovic
 
T
MacDonald
 
KP
Morris
 
ES
, et al. 
TGF-beta in allogeneic stem cell transplantation: friend or foe?
Blood
2005
, vol. 
106
 
6
(pg. 
2206
-
2214
)
Sign in via your Institution