Acute and chronic graft-versus-host disease (GVHD) are potentially lethal complications after stem cell transplantation (SCT). Steroids are the appropriate first-line treatment for both. However, if patients do not adequately benefit from steroid therapy, mortality is high and standardized treatment algorithms are lacking. This is mainly because of limited data from prospective, randomized clinical trials. In addition, most of the available treatment options only induce clinical benefits in a limited proportion of patients. Thus, there is an urgent clinical need to develop more potent immunosuppressive treatment strategies for patients suffering from acute or chronic steroid-refractory GVHD while maintaining the graft versus tumor effect to avoid a potential rise in relapse-related mortality. The increasing knowledge about host- as well as donor-derived variables favoring GVHD development and the increasing armamentarium of immune-modulatory agents entering preclinical and clinical research will probably allow more effective treatment of GVHD in the future. This review describes novel developments in the treatment of steroid-refractory GVHD, with a special focus on the rationale behind promising pharmacologic compounds or up-coming cellular therapies.

In 1957, E. D. Thomas and colleagues first described the infusion of bone marrow cells into patients after prior radio- or chemotherapy in their seminal New England Journal of Medicine paper.1  This work initiated 5 decades of basic and clinical research in the field of stem cell transplantation (SCT) and nowadays SCT is the treatment of choice for many malignant and benign hematopoietic diseases. It was known before 1957 from preclinical animals studies that tranplantation of splenocytes from noncongenic donor strains, while facilitating hematopoietic recovery, induced a severe illness, characterized by progressive weight loss, hunched posture, and diarrhea.2  In the following years it has become clear that this was not primarily because of the conditioning therapy, but that it was an immune-mediated syndrome, which is now referred to as graft versus host disease (GVHD). GVHD nowadays remains not only a major cause of non-relapse mortality, but also induces substantial morbidity, which can severely affect quality of life. GVHD is a very complex immunologic disorder characterized by a plethora of clinical presentations.3  Importantly, the predominant use of peripheral blood stem cells (PBSC) rather than bone marrow (BM) and the increasing proportion of reduced intensity conditioning (RIC) regimens has made multifacetted chronic GVHD (cGVHD) more and more clinically relevant.4,5  The incidence of GVHD thus depends on several variables, including the donor type (related versus unrelated, matched versus mismatched or haploidentical), the type of conditioning (total boby irradiation [TBI] versus nonTBI), the donor's sex (female or male, versus all other sex combinations) and the stem cell source (PBSC versus BM).6  Despite the increasing importance of cGVHD in clinical practice, preclinical animal cGVHD models are scarce. This is in contrast to acute GVHD (aGVHD), which can be adaquately described by several animal models7  enabling researchers to study its immune pathogenesis and test novel treatment approaches.

Steroids are still the first-line GVHD treatment8  and patients not responding to steroid therapy are at high risk of dying from GVHD or its related complications.9  Currently, there is no common standard treatment strategy for steroid-refractory GVHD patients. This review will focus on novel developments in the prevention of GVHD development and the treatment of steroid-refractory GVHD, with a special focus on promising innovative compounds as well as on up-coming cellular therapies.

Modification of conditioning-induced tissue damage and danger signals

Conditioning of the recipient before the donor's stem cells are transferred is usually done by chemo- or radio-chemotherapy, both of which induce significant tissue damage. This results in the accumulation of “danger signals,” which are signals released from injured, stressed, or dying cells and which in turn activate antigen-presenting cells.10  The gastrointestinal (GI) tract is a predisposed organ for the development of aGVHD, because tissue damage induces partial disruption of the barrier function between the sterile submucosal area and the colonized intraluminal space allowing the transit of bacteria. In addition, local release of danger signals from activated T cells11  may activate innate and adaptive immune cells. Both, bacterial components and extracellular danger signals transmit their signals via toll-like receptors (TLRs) or via NOD-like receptors expressed on innate immune cells.12  Zeiser and colleagues elegantly demonstrated in a murine aGVHD model that high levels of adenosine triphosphate (ATP), an important extracellular danger signal released by dying cells, can be detected in the GI tract of animals receiving TBI and in the peritoneal fluid of patients receiving TBI before SCT.13  ATP induces higher expression of the costimulatory molecules CD80 and CD86 on antigen-presenting cells (APC) which leads to a more efficient priming of allo-specific T cells. The receptor for ATP on recipient's APC is P2X7R. This receptor has been described to activate the inflammasome, thereby also allowing more potent allo-stimulatory T-cell priming. Accordingly, pharmacologic blockade of P2X7R or reconstitution of animals with P2X7R-deficient APC before SCT substantially reduced the incidence of GVHD, increased the number of regulatory T cells (Treg) and decreased IFN-γ producing effector T cells.13  These observations underscore the importance of ATP-mediated danger signaling in tipping the balance towards a more allo-reactive and pro-inflammatory milieu. SNP data from 152 patients who underwent matched sibling allogeneic SCT support the clinical relevance of these findings. Here, overall survival was significantly shorter in recipients carrying the CC genotype in the P2X7R gene and in patients receiving stem cells from donors carrying the CC genotype. The CC genotype was also identified as an independent prognostic factor for overall survival.14  Thus, inhibition of P2X7R signaling by targeted drugs (such as A-438079) might represent a promising strategy for preventing aGVHD caused by tissue damage during conditioning (Figure 1).

Figure 1

Targeting danger signaling and APCs. The left panel highlights the central role of danger signals and APC activation during GVHD. Tissue damage induced by the conditioning regimen induces the release of ATP and transit of bacteria/bacterial components, which in turn induces activation of DC (aDC) leading to priming of allo-reactive T cells. The right panel illustrates potential strategies interfering with these processes of APC activation to inhibit or prevent GVHD development. Promising strategies include TLR-inhibition by MyD88-inhibitors (ST825), APC modification by HDACi or TKI or blockade of ATP-signaling by P2X7-inhibitors (A-438079). Moreover, stimulation of APC by MDP might induce a tolerogenic APC-phenotype. Compounds in clinical testing are shown in brown, promising strategies to be tested in the future in humans are given in red. TC indicates T-cell; DC, dendritic cell; aDC, activated DC; Treg, regulatory T-cell; PRR, pattern recognition receptor; TLR, toll-like receptor; HDACi, histone deacetylase inhibitor; TKI, tyrosine kinase inhibitor; NOD/CARD, nucleotidebinding oligomerization domain containing/caspase recruitment domain; tDC, tolerogenic DC; and MDP, muramyl-dipeptide.

Figure 1

Targeting danger signaling and APCs. The left panel highlights the central role of danger signals and APC activation during GVHD. Tissue damage induced by the conditioning regimen induces the release of ATP and transit of bacteria/bacterial components, which in turn induces activation of DC (aDC) leading to priming of allo-reactive T cells. The right panel illustrates potential strategies interfering with these processes of APC activation to inhibit or prevent GVHD development. Promising strategies include TLR-inhibition by MyD88-inhibitors (ST825), APC modification by HDACi or TKI or blockade of ATP-signaling by P2X7-inhibitors (A-438079). Moreover, stimulation of APC by MDP might induce a tolerogenic APC-phenotype. Compounds in clinical testing are shown in brown, promising strategies to be tested in the future in humans are given in red. TC indicates T-cell; DC, dendritic cell; aDC, activated DC; Treg, regulatory T-cell; PRR, pattern recognition receptor; TLR, toll-like receptor; HDACi, histone deacetylase inhibitor; TKI, tyrosine kinase inhibitor; NOD/CARD, nucleotidebinding oligomerization domain containing/caspase recruitment domain; tDC, tolerogenic DC; and MDP, muramyl-dipeptide.

Close modal

During this early process of danger signaling other nonpurinergic signaling cascades might be as important as free ATP. Among these, pattern recognition receptors (PRRs) sensing microbial components such as lipopolysaccharide (LPS) or flagellin15  and extracellular matrix components16  are important as well. Genetic association studies linking TLR4 polymorphism (Thr399Ile) in patients and donors with an increased the risk for severe aGVHD17  suggest an important role for TLRs in allo-SCT. Moreover, cotreatment of animals with TLR9 agonistic CPG oligonucleotides aggravated GVHD lethality because it increased host-derived IFN-γ production.18  Functional data using mice deficient in the TLR signaling components MyD88 and TRIF or in TLR9 support the role of TLRs in GVHD development.19  Deficiency in TLR9 may even led to increased survival.19  Thus, preclinical testing of novel MyD88-inhibitors such as ST282520  might be a rational approach to modify microorganism-mediated TLR activation during GVHD (Figure 1).

In addition to TLRs, NOD/CARD15 also regulates GVHD pathogenesis by sensing bacterial products. Polymorphisms of NOD/CARD15 are linked to GVHD pathogenesis.21  Single nucleotide polymorphisms (SNPs) resulting in reduced function of NOD/CARD15 induce a hyperactive phenotype of APC,22  which might contribute to hyperactivation of allo-reactive T cells during GVHD. In line with this idea, NOD2-deficiency in mice induces an increased allo-priming of T cells.23  Thus, inter-individual differences in the sensitivity toward pathogen-associated microbial patterns (PAMPs) as a consequence of SNPs probably play a role in the deregulation of the immune system during allo-reaction/GVHD. As a consequence, actual concepts transferred from models for inflammatory bowel disease24  are testing the value of continuous NOD2 stimulation via its cognate ligand muramyl-dipeptide to prevent GVHD (Figure 1).

Targeting recipient and donor type APC

The previous topic of danger signal-mediated regulation of inflammatory responses already highlighted the important role of APC as central regulators of allogeneic T-cell priming. Host APC survive for a considerable time within the recipient and are very potent allogeneic T-cell activators because they express minor histocompatibility antigens (miHA) that induce allo-miHA-specific T-cell activation. Even after sublethal irradiation, 30% of host dendritic cells (DC) are still present in the spleen 24 hours after challenge,25  allowing rapid activation of donor T cells. Because of their high degree of radioresistance, tissue-specific DCs including Langerhans cells (LCs) in the skin undergo a markedly less dynamic turnover and can still be found months after SCT.26  LCs are predominatly replaced in case of an ongoing skin GVHD.27  After replacement of the DC compartment by donor-derived DCs, CD8+ T cells are primarily activated via crosspresentation of miHA by CD103+ or CD8+ DCs. There are various approaches to target DCs or DC function to modify the course of GVHD. Notably, currently used immunosuppressive agents including cyclosporin, mycophenolate mofetil and steroids also modify DC function.28-30  However, more innovative strategies have been proposed in more recent reports. An interesting and clinically feasible strategy is the in vivo tailoring of DCs into a tolerogenic phenotype. Targeting the process of histone acteylation for example appears to be a very interesting approach not only for direct elimination of malignant T cells, but also for immune-modulation. We could recently demonstrate that histone deacetylase inhibitors (HDACi) skew DC differentiation by preventing the acquisition of the DC hallmark CD1a as well as by affecting the expression of costimulatory and adhesion molecules.31  The observed defects in DC function on exposure to HDACi obviously reflect impaired signaling through nuclear factor-kappa B, IRF-3 and IRF-8.31  In addition, HDACi exert substantial effects on the APC-induced cytokine storm during GVHD (that is, reduced TNF-α, IFN-γ, IL-1β, and IL-12 levels) and they impair TLR-induced expression of CD40 and CD80 as well as the APC's allo-stimulatory capacity.32  In line with these in vitro effects, injection of in vitro HDACi-treated DCs reduced experimental GVHD in murine SCT models, mainly because of induction of IDO, a well known suppressor of DC and T-cell function.32 

Moreover, HDACi promote Treg expansion, as FoxP3 interacts with HDAC 7 and 9, which maintain the Treg phenotype, and with the histone acetyltransferase TIP60.33  Accordingly, mice exposed to the HDACi trichostatin A have increased numbers and functionally improved Treg, increasing donor-specific allograft tolerance.34  Thus, the impact of HDACi on DCs as well as on the Treg compartment might at least in part explain the promising effects of suberoylanilide hydroxamic acid and ITF 2357 in murine GVHD models.32,33  Together, the available preclinical data suggest that HDACi might have a huge potential for prevention and treatment of GVHD and currently various clinical phase 1/2 trials testing the efficacy of single agent panobinostat or its combination with steroids in the treatment or prevention of aGvHD are ongoing (Figure 1).

Finally, tyrosine kinase inhibitors (TKI) also exert beneficial effects in the treatment of GVHD. The most frequently used TKI is imatinib, which primarily induces responses in sclerodermiformal skin and pulmonary cGVHD.35  The obvious rational for TKIs in cGVHD is based on their inhition of platelet-derived growth factor-receptor (PDGF-R) signaling, which is assumed to regulate tissue scarring triggered by chronic inflammation,36  because patients with cGVHD have been described to exhibit elevated levels of agonistic antibodies binding to the PDGF-R.37  As a result of this finding, imatinib, an inhibitor of PDGF intracellular signaling, has been studied in various patient populations with fibrotic cGVHD. One hundred mg/d imatinib induced a response rate of 79% in a prospective study including 19 patients with refractory cGVHD involving the skin, lung and bowel. Seven of 19 patients achieved a complete remission (CR).38  Toxicity included fluid retention and myelosuppression and only 3 patients discontinued because of toxicity. Another group found a somewhat lower response rate (50%) in a retrospective study including 14 patients with extensive cGVHD of the skin and joints. Here, the median dose was 400 mg/d leading to a higher rate of toxicity and subsequent treatment discontinuation.39 

In addition to its well known effects on PDGF-R signaling, we and others have demonstrated that imatinib inhibits DCs and monocyte/macrophage function,40,41  and potentially affects T cells.42  In more detail, imatinib reduces human monocyte-derived DCs differentiation by inhibiting the phosphatidylinositol 3-kinase/Akt and the NF kappa B pathway, which is paralleled by a reduced capacity of TKI-exposed DCs to activate T cells.40  We further extended these finding to monocytes and macrophages, which are similarly characterized by a markedly impaired cytokine response to TLR-agonists in the presence of imatinib.41  Others have provided evidence that imatinib and the second generation TKI nilotinib also inhibit T-cell responses.43  So far, no correlative immunologic data from GVHD patients receiving imatinib are available. However, the immunomodulatory function of TKI might at least in part explain their beneficial effects seen in cGVHD and a more systematic testing of these compounds either as monotherapy or in combination with other treatment modalities (ie, rituximab) in cGVHD is currently ongoing (Figure 1).

Targeting the T-cell compartment

Various CD4+ T-cell subpopulations are involved in the pathogenesis of GVHD. Each individual GVHD manifestation appears to have a distinct T-cell milieu, which is determined not only by the target structures but also by the respective inflammatory environment. The understanding of the role of T-cell subsets in humans is derived from cytokine quantifications in peripheral blood, lesional tissue biopsies or from SNPs in cytokine genes and their association with GVHD.44-46  Murine models allow a more concise description of the functional involvement of CD4+ T-cell supopulations in aGVHD. Donor TH1 cells predominantly mediate GI and hepatic aGVHD.47  When IFN-γ deficient CD4+ T cells are transferred, T cells differentiate into TH2 and TH17 cells which aggravate pulmonary and skin aGVHD. In addition, absence of IL-4 and IFN-γ augments TH17 differentiation inducing skin aGVHD.47  Finally, if cells deficient in IFN-γ and IL-17 are transferred, augmented TH2 differentiation induced idiopathic pneumonia.47  This data supports the model that tissue-specific migration patterns of TH1, TH2, and TH17 cells occur, which mediate organ-specific GVHD. In humans, the picture might be more complex: the TH17/Treg ratio detected in lesional biopsies from the GI tract and skin is positively correlated with severity of aGVHD.48  In addition, sole quantification of TH17 did not show a difference between affected and healthy skin. In contrast, significantly more IFN-γ producing T cells were detected in biopsies from affected areas.48  This data suggest that detection of individual T-cell subpopulations might not be sufficient to reflect the complex immunologic processes in GVHD.

However, it is tempting to speculate that fine-tuning of CD4+ T-cell diversification might influence the course of GVHD. Recent reports emphasized the importance of IL-21 derived from donor T cells for GVHD development.49,50  Disruption of the IL-21 pathway in donor CD4+ T cells markedly impairs development of GI aGVHD while preserving the graft versus tumor effect.50  Surprisingly, disruption of IL-21 signaling had no impact on IL-17 production, impaired IFN-γ secretion in colon-infiltrating T cells and increased TH2 and induced Treg (iTreg).49  Recent reports also emphasized signaling molecules as interesting targets for fine-tuning of T-cell differentiation within the recipient. STAT1 for example interferes with TH1 differentiation, as reflected by an impaired in vivo activation of the cells with subsequent reduction of IFN-γ production.51  Moreover, STAT1 is a natural inhibitor of FoxP3 and its inhibition boosts in vivo iTreg conversion and naturally occurring Treg (nTreg) expansion. Future development of more specific STAT1 inhibitors will allow pre-clinical testing of this concept in murine GVHD models (Figure 2).

Figure 2

Targeting the T-cell compartment. The left panel highlights the central role of T-cell activation within SLO for GVHD initiation. APC activate and expand allo-reactive T cells, which then migrate to target organs. Secretion of pro-inflammatory cytokines, such as IFN-γ and IL-21 are hallmarks of a sustained alloreactive T-cell response. In this GVHD-favouring scenario, the balance within the T-cell compartment is on the effector T-cell side. The right panel depicts potential strategies inhibiting T-cell activation or SLO occupancy to inhibit or prevent GVHD development. Antibodies inhibiting SLO entry might help to prevent T-cell priming, whereas blockade of T-cell exit by FTY720 inhibits the migration of GVHD-mediating T cells to GVHD target organs. Other drugs help to tip the balance toward the Treg compartment, such as mTOR inhibitors, STAT1 inhibitors or antibodies blocking IL-21. The activation process of T cells can also be directly inhibited by PKC-inhibitors (eg AEB071). Compounds in clinical testing are shown in brown, promising T-cell targeting strategies to be tested in future clinical trials are given in red. TC indicates T-cell; Treg, regulatory T-cell; HEV, high endothelial venules; mTOR, mammalian target of rapamycin; mAb, monoclonal antibody; MadCAM, mucosal addressin cell adhesion molecule; and SLO, secondary lymphoid organ.

Figure 2

Targeting the T-cell compartment. The left panel highlights the central role of T-cell activation within SLO for GVHD initiation. APC activate and expand allo-reactive T cells, which then migrate to target organs. Secretion of pro-inflammatory cytokines, such as IFN-γ and IL-21 are hallmarks of a sustained alloreactive T-cell response. In this GVHD-favouring scenario, the balance within the T-cell compartment is on the effector T-cell side. The right panel depicts potential strategies inhibiting T-cell activation or SLO occupancy to inhibit or prevent GVHD development. Antibodies inhibiting SLO entry might help to prevent T-cell priming, whereas blockade of T-cell exit by FTY720 inhibits the migration of GVHD-mediating T cells to GVHD target organs. Other drugs help to tip the balance toward the Treg compartment, such as mTOR inhibitors, STAT1 inhibitors or antibodies blocking IL-21. The activation process of T cells can also be directly inhibited by PKC-inhibitors (eg AEB071). Compounds in clinical testing are shown in brown, promising T-cell targeting strategies to be tested in future clinical trials are given in red. TC indicates T-cell; Treg, regulatory T-cell; HEV, high endothelial venules; mTOR, mammalian target of rapamycin; mAb, monoclonal antibody; MadCAM, mucosal addressin cell adhesion molecule; and SLO, secondary lymphoid organ.

Close modal

A more useful, already druggable target is protein kinase C (PKC)θ. PKCθ is critical for T-cell survival and regulates the T-cell activation threshold.52  Genetic mouse models enabled validation of PKCθ as promising therapeutic target for GVHD prevention.53  When focusing on clinical translation of these findings, AEB071 represents a potent and selective inhibitor of novel and classic PKCs, decreasing IL-2 and IFN-γ production in T cells.54  In preclinical monkey studies, oral AEB071 mono- or combination-therapy prolongs survival of rat heart and kidney allografts.55  Most importantly, AEB071 blocks T-cell activation through mechanisms distinct from calcineurin inhibitors (CNI), the current standard therapy in GVHD prophylaxis and may therefore lack toxicities associated with CNIs. Consequently, AEB071 is currently tested in combination with tacrolimus versus mycophenolate mofetil plus tacrolimus in solid organ transplantion. AEB071 might also be a promising drug for GVHD treatment or prevention after allogeneic SCT in humans (Figure 2).

Another interesting group of novel agents potentially limiting alloreactivity of T cells are compunds interfering with T-cell localization, as T cells enter secondary lymphoid organs (SLO) for T-cell priming and subsequently GVHD target organs to induce tissue damage.56  As an example, FTY720, a sphingosine analog has been shown to prevent GVHD57  by blocking sphingosine 1-phosphate receptor-mediated T-cell egress from SLO normally induced by a sphingosine 1-phosphate gradient from blood and lymph fluid to tissue.58  As an alternative approach, Beilhack and coworkers extensively tested strategies of blocking T-cell access to SLO.59  It is generally assumed that primed T cells are instructed to migrate to the respective target organs.60  In GVHD, lack of specific priming sites did not affect disease severity in the respective target organ and a single priming site is sufficient for aGVHD induction.59  Intriguingly, blockade of SLO access by application of blocking mAbs to MAdCAM-1 and CD62L to splenectomized mice almost completely prevented aGVHD lethality, supporting the importance of SLO occupancy by donor-derived T cells for their priming.59  This data shows that blockade of SLO access might be a rational approach to prevent the development of aGVHD (Figure 2).

Finally, proteasome inhibitors are also a novel group of agents potentially reversing alloreactivity of T cells toward tolerance. Bortezomib, the first in kind proteasome inhibitor widely applied for treatment of multiple myeloma, has been shown in vitro to selectively induce apoptosis in activated alloreactive T cells via caspase activation and cleavage of the antiapoptotic bcl-2 protein.61  Notably, there was no inhibition of the anti-tumor effect in the mouse model,62  although a slight concern regarding possible aggravation of GI symptoms was raised.63  Thus, soon clinical case reports on myeloma patients with progressive disease after allo-SCT followed: Mateos-Mazon et al reported on 5 patients with relapsed myeloma and GVHD treated with bortezomib.64  Four of these patients showed an improvement of their GVHD. Based on these promising results, first clinical studies are being performed including bortezomib as part of the GVHD prophylaxis regimen demonstrating its feasibility and minimal toxicity.65  Randomized trials have yet to be published.

Targeting GVHD-associated neovascularization

Infused allogeneic T cells primarily interact with circulating cells within the blood stream and with endothelial cells lining the inner face of the vessel. The endothelial cell (EC) compartment can be damaged by conditioning regiments, especially by TBI. Because of endothelial damage, host ECs are at least in part replaced by donor-derived endothelium by vasculogenesis.66  Interestingly, some types of ECs (that is, sinusoidal endothelial cells) have been demonstrated to excert immuno-modulatory effects.67  Besides, the vessel endothelium can directly activate alloreactive CD8+ T cells in vitro and in vivo in the absence of classic APC.68  It is conceivable that this first step of allo-activation within the host is an important event for priming and expansion of allo-reactive T cells, which subsequently invade the interstitial space and target epithelial structures. Thus, conditioning-mediated endothelial damage and replacement of EC because of recruitment of endothelial progenitor cells represents an interesting therapeutic target in GVHD. Of note, in cGVHD of the skin the vascularization process is rather insufficient, causing rarification of vessels within the involved areas.69  In aGVHD models, treatment of recipient animals with the anti-vascular endothelial-cadherin antibody E4G10 inhibited neovascularization by donor-derived cells and inhibited GVHD and tumor growth with a subsequent increase of survival rates.70  This study provides a strong rationale for further testing of anti-angiogenic strategies in GVHD.

In addition, vascular endothelial growth factor (VEGF) expression in GVHD is negatively associated with GVHD severity and non-relapse mortality.71  Recent data further demonstrated that angiopoetin-2 and thrombomodulin are increased in steroid-refractory GVHD.72  The correlative data of a negative impact of increased VEGF levels for GVHD are somehow contradicted by pre-clinical results showing that VEGF-R1 or R2 blockade not only aggravated GVHD severity, but also reduces engraftment and increased mortality.70  Thus, blocking the VEGF/VEGF-R appears not to be the appropriate approach for aGVHD treatment, but obviously more pre-clinical and translational clinical data are needed to explore the potential of angiogenesis/vasculogenesis targeting in the management of GVHD, especially because recent data show that various anti-angiogenic compounds targeting the VEGF or PDGF pathways seem to be capable of modulating immune responses.73 

Similarly, the anti-angiogenic agent thalidomide has yielded discrepant results: while early studies in cGVHD have been very promising,74  later publications only confirmed an overall response rate around 40% in cGVHD,75  no efficacy in acute GVHD76  and a possibly detrimental effect in GVHD prophylaxis.77  In this context it might not be surprising, that the thalidomide-derivative lenalidomide given within 6 months after allo-SCT, rather than showing protective properties, led to an almost 50% discontinuation rate because of GVHD and was therefore not considered feasible by the authors.78 

Donor Treg infusion (DTI)

Natural occurring CD4+CD25+ Treg tip the balance between auto- and tumor-immunity. Treg also regulate allo-reactivity in vitro and in vivo.79  In murine models adoptively transferred Treg prevent the development of aGVHD without affecting the graft versus tumor response.80  Accordingly, Treg content in the graft or the donor's blood correlates with aGVHD incidence after HLA-identical sibling SCT.81,82  Of note, relapse-related mortality or infections were not increased in patients receiving grafts with high Treg numbers. As a consequence, recently the first clinical trial of adoptive Treg transfer for prevention of aGVHD was reported.83  Twenty-eight patients undergoing haploidentical stem cell transplantation received freshly isolated donor Treg on day −4, which was then followed by transfer of highly purified CD34+ stem cells together with conventional T cells. Patients did not receive any prophylactic immunosuppression. For safety reasons of this pilot trial, patients received only 25% conventional T cells compared with Treg. None of the first 4 patients developed aGVHD, and therefore, conventional T cells were escalated to 50% of the Treg cell dose. Rapid and stable engraftment was seen and only 2 patients developed grade II aGVHD. So far, none of the patients has developed cGVHD. When compared with a dataset of 152 patients receiving haploidentical SCT without Treg transfer, the approach rather promoted lymphoid reconstitution and improved immunity to opportunistic pathogens. Fourteen patients died during the study and only 1 patient in this high risk patient population died from relapse. The study is small, the median follow-up is short and thus the data too premature to draw definite conclusions. However, DTI appears to be feasible and safe in terms of GVHD prevention. Randomized studies are now needed to test the preventive as well as the therapeutic potential of DTI and to evaluate optimized application schedules (Figure 3).

Figure 3

Cellular therapies including MSC, Treg, NKT-, and B cell targeting. The left panel highlights the central role of various cell types including B and NKT cells for effector T-cell activation during GVHD. In this GVHD favoring scenario NKT-cell–derived IFN-γ and B cells function as T-cell activators, while Treg expansion is suppressed. The right panel depicts potential strategies how this processes can be manipulated to favor a more tolerogenic environment. Adoptive transfer of in vitro generated or donor-derived Treg, MSC or NKT cells either directly inhibits effector T-cell activation and expansion, or indirectly tips the balance toward a tolerogenic milieu by inducing Treg expansion. Moreover, cellular compartments known to be involved in the activation of alloreactive T cells, such as B cells can be depleted by monoclonal antibodies directly targeting B cells (ie rituximab). Therapeutic strategies currently in clincial testing are shown in brown, preclinical concepts in red. MSC indicates mesenchymal stem cell; Treg, regulatory T-cell; NKT, natural killer T-cell; BC, B cell; DC, dendritic cell; TC, T-cell; and SLO, secondary lymphoid organ.

Figure 3

Cellular therapies including MSC, Treg, NKT-, and B cell targeting. The left panel highlights the central role of various cell types including B and NKT cells for effector T-cell activation during GVHD. In this GVHD favoring scenario NKT-cell–derived IFN-γ and B cells function as T-cell activators, while Treg expansion is suppressed. The right panel depicts potential strategies how this processes can be manipulated to favor a more tolerogenic environment. Adoptive transfer of in vitro generated or donor-derived Treg, MSC or NKT cells either directly inhibits effector T-cell activation and expansion, or indirectly tips the balance toward a tolerogenic milieu by inducing Treg expansion. Moreover, cellular compartments known to be involved in the activation of alloreactive T cells, such as B cells can be depleted by monoclonal antibodies directly targeting B cells (ie rituximab). Therapeutic strategies currently in clincial testing are shown in brown, preclinical concepts in red. MSC indicates mesenchymal stem cell; Treg, regulatory T-cell; NKT, natural killer T-cell; BC, B cell; DC, dendritic cell; TC, T-cell; and SLO, secondary lymphoid organ.

Close modal

In vivo Treg expansion—GVHD treatment and prevention by mTOR inhibition

In addition to adoptive Treg therapies, the Treg compartment can also be modified by pharmacologic intervention. We recently demonstrated that expansion of adoptively transferred Treg in vivo is critical for their GVHD suppressive activity.84  The mammalian target of rapamycin (mTOR) functions as rheostat regulating this process. mTOR regulation is a dynamic and oscillatory process characterized by an early down-regulation of the leptin-mTOR pathway followed by a rapid increase, which is a prerequisite for Treg expandability.85  Rapamycin induces in vitro expansion of nTreg,86  which has also recently been used to establish GMP-conform expansion protocols for human CD4+CD25+ T cells.87  Data from murine in vivo SCT models support the observation of Treg-supportive effects of rapamycin88  including increased generation of thymic Treg89  and infiltration into GVHD target organs.90  Of note, Treg expansion by rapamycin preserves the graft-versus- leukemia effect. Very recent data support the importance of iTreg in addition to nTreg to allow complete coverage of autoreactive T-cell clones, thus enabling rescue of foxp3-deficient animals from auto-immunity.91  Interestingly, rapamycin together with IL-2 enables expansion of donor-derived nTreg and conversion of CD25 T cells to iTreg by IL-2, which potently inhibits development of GVHD in vivo.92  Thus, combination of adoptive Treg transfers with in vivo application of rapamycin, or the in vivo application of rapamycin in combination with IL-2 to get higher yields of nTreg and iTreg represents a promising strategy of pharmacologic Treg modification to treat or prevent GVHD (Figure 3).

The clinical potential of mTOR inhibition is emphasized by various data from small clinical trials. As an example, when combined with mycophenolate mofetil and anti-thymocyte globulin (ATG), CNIs can be replaced by sirolimus within the FLAMSA-RIC protocol.93  Using this approach, 21% of patients developed grade II-IV aGVHD, 30% cGVHD and nonrelapse mortality rate was 14%. Of note, in this setting sirolimus did not induce transplant-associated thrombotic microangiopathy (TA-TMA). Seventy-five percent of the patients were alive after a median follow-up of 10 months.93  In contrast, when combined with CNIs, various groups recently reported that sirolimus-based GVHD prophylaxis in patients receiving myeloablative or RIC-SCT induces severe toxicities, such as transplantation associated microangiopathy or sinusoidal obstructive syndrome.94,95 

When used for treatment but not prophylaxis of GVHD, sirolimus appears to be a promising agent even in steroid-refractory aGVHD.96  Overall response rate of a recently reported study including 34 patients was 76% with 42% CR in steroid-refractory and 67% of steroid-intolerant patients leading to a 1-year survival of 44%. Sirolimus might even be used as front-line therapy of aGVHD instead of steroids, inducing CR in up to 50% of patients.97  With respect to treatment of cGVHD, a retrospective analysis including 34 patients with severe sclerodermatous cGVHD treated with sirolimus or everolimus (including 12 receiving only mTOR inhibitors) also suggest substantial clinical efficacy in cGVHD.98  The overall response rate was again 76%, including CR and PR in 6 and 20 patients, respectively. Steroids could be tapered and stopped in a significant number of patients. Relevant side effects included hyperlipidemia, impaired wound healing and TA-TMA in 2 patients. After a median follow-up of 723 days, 26 of the 34 patients were still alive. Overall survival at 3 years since initiation of mTOR-inhibitor therapy is 72%. Unfortunately, data on immunologic surrogate providing biomarkers for clinical responses to mTOR inhibition are not available so far. Future studies will have to clarify the role of sirolimus in GVHD treatment and prophylaxis, but combination with CNI should be used with caution, because of increased toxicity and because CNIs antagonize the effects of mTOR inhibitors on the generation of Tregs by inhibiting IL-2 production.

NKT cells

Natural Killer T (NKT) cells express an invariant Vα24 TCR-chain (Vα14 in mice) and secrete large amounts of IL-4 and/or IFN-γ after activation via the T-cell receptor, which recognizes glycolipids presented by the nonpolymorphic CD1 days antigen-presenting molecule.99-101  Various reports provide evidence that both, host- and donor-derived NKT cells protect from GVHD.102-104  This effect primarily depends on NKT-cell secreted IL-4, which is known to drive TH2-polarization of conventional donor-derived T cells,105,106  attenuating their capacity to mediate GVHD.105,107,108  Thus, NKT cells expressing IL-4 can be referred to as another regulatory lymphocyte population. Activation of host NKT cells appears to be crucial for GVHD prevention, as transplantation of CD1 days- and Jα-18–deficient host mice with wild-type transplants allowed graft versus leukemia reaction but failed to prevent GVHD.103  In a murine SCT setting modeling human aGVHD-protective RIC protocols using total lymphoid irradiation and anti-thymocyte globulin (ATG) as conditioning regimen, host NKT cells cooperated with donor-Treg to induce IL-4–dependent in vivo Treg expansion and prevented lethal GVHD.109  Moreover, in mice highly purified NKT-cell infusion protected from GVHD development by limiting T cell–mediated secretion of pro-inflammatory cytokines, such as IFN-γ and TNF-α, while the graft- versus-leukemia effect was preserved.102  This observation underscores the therapeutic potential of donor NKT cell for immune-modulation within the recipient and their potential as cellular immuno-suppressant, possibly in combination with DTI. However, data from transplant models showing reduced GVHD severity in recipients of NKT-deficient grafts,110  demonstrate that donor NKT cells might under certain conditions also contribute to the pathogenesis of GVHD. Thus, further research is needed to determine how NKT cells are optimally modulated to skew the T-cell response toward TH2 cells, as they might also boost GVHD.

Mesenchymal stem cells

The bone marrow contains an adherent bone marrow–derived fibroblast-like cell, which as a consequence of its multipotency (ie, differentiation into adipocytes, osteoblasts and chondrocytes), has been referred to as mesenchymal stem/stromal cells (MSC).111  MSC are potent immune-regulators. This information set the stage for their clinical testing as cellular immunosuppressants in GVHD.112  MSC primarily affect T-cell biology; however, the exact mechanisms of immunregulation are not well defined. MSC-mediated inhibition of T-cell proliferation is assumed to be mediated by several mechanisms including secretion of TGF-ß, HGF, PGE2, galectin, and shaping of DC toward a more tolerogenic phenotype.113  Moreover, MSC express IDO, which in turn depletes the essential amino acid tryptophan leading to an accumulation of the T-cell toxic metabolite kynurenin.114  Recent data demonstrated that MSC also induce Treg.115  Interestingly, it is the possible to transplant third-party MSC without any need for HLA-identity, making this treatment quite convenient. The reason for this phenomenon might be the lack of MHC class I and II and low costimulatory molecule expression.116  Importantly, not all reports confirmed the effects of MSC in treatment of refractory aGVHD,117  but the largest phase 2 trial available so far demonstrated high efficacy of this approach. This trial included 30 of 55 patients achieving complete resolution of steroid-refractory aGVHD.118  Of note, the median dose in this trial was 50% higher than in the negative trial. None of the patients experienced relevant side-effects, especially no increase in infection or relapse rates. This is in line with previous observations that MSC predominantly inhibit allogeneic but not anti-viral T-cell responses in vivo.119  Moreover, complete response was associated with increased overall survival. The currently ongoing phase 3 trial is not yet fully published but will help to define the value of MSC in the treatment of steroid-refractory aGVHD more accurately (Figure 3). Finally, early clinical data also suggest a clincial potential of MSC in the treatment of cGVHD.120 

B-cell targeting

B cells have mostly been appreciated for their ability to produce highly pathogen-specific antibodies. However, in recent years physiologic antibody-independent immune functions such as antigen presentation to T cells, immune regulation and cytokine secretion have been investigated.121  Correspondingly, in several autoimmune diseases antibody-independent B-cell functions have been described to play an important role.122  In patients with autoimmune diseases antigen presentation of auto-reactive B to T cells was found.123  Similarly, a dysfunctional B-cell compartment has been detected in cGVHD: Patients with cGvHD have a high number of activated memory (CD27+) B-cells, higher levels of B cell–activating factor of the tumor necrosis family and donor-derived allo-antibodies. Besides, they show a faster B-cell reconstitution.124,125 

Although the involvement of B cells in the pathogenesis of aGVHD is less well studied, it is correlated with the number of B cells in the transplant and depletion of B cells resulted in a reduction of aGVHD.126-128  Still, as GVHD is predominantly T cell–mediated, it remains unclear whether occurrence of allo-antibodies represents a bystander effect or is a relevant cofactor.

In the light of the sometimes rapid response of GVHD to B-cell depletion and the serum half-life of immunoglobulins a predominantly antibody-mediated mechanism of GVHD seems unlikely, rendering a direct effect on B cells possible. This is supported by the fact, that other treatment modalities like mycophenolate mofetil, steroids, extracorporal photophoresis and ATG also have direct effects on B cells. ATG, one of the oldest immunosuppressive agents, not only targets T cells but also an array of epitopes expressed by B cells including CD19, CD20 and many others.129  Interestingly, HMG-CoA-Reductase inhibitors have also been demonstrated to inhibit aGVHD130  potentially by targeting B cells among other cellular compartments.131  Rituximab as a B cell–specific agent has shown considerable clinical benefit in patients with auto-immune disease, probably by preventing B cell–mediated T-cell activation in addition to B-cell depletion.132  Correspondingly, several small trials showed clinical effectiveness of rituximab in cGVHD. GVHD of the skin responded better than cGVHD of the visceral organs,133  but successful treatment of cGVHD of inner organs has been reported anecdotally.134  As CD20 is a promising target not only in malignant disease several alternative, humanized or human antibodies such as ocrelizumab, GA101 and ofatumumab are currently being tested and should also be investigated in GVHD. Thus, B cells represent exciting targets for the prevention and treatment of GVHD. The development of B-cell–specific agents in other fields will rapidly offer an arsenal of therapeutics that need to be investigated in the context of GVHD.

Acute and chronic GVHD are a potentially lethal complications and still limit the success of SCT in a considerable proportion of patients. Steroids are the current treatment standard for GVHD. However, if it does not resolve and the patient is steroid-refractory, the mortality rate is high. We here reviewed pre-clinical and early clinical approaches representing potential future strategies for treating GVHD. This includes novel pharmacologic agents (eg, HDAC inhibitors, proteasome inhibitors, antibodies targeting IL-21 or adhesion molecules) or up-coming cellular therapeutics such as Treg, NKT, MSC, or B-cell targeting. The recent presentation of promising first clinical data from Treg or MSC therapy support the feasibility of this concept. It remains critical that well designed clinical trials will be performed to define the value of the respective novel treatment strategy in steroid-refractory GVHD.

D.W. is supported by the Austrian Science Fund FWF, the OEGHO, and the TILAK. P.B. is supported by DFG and Deutsche Krebshilfe.

Contribution: D.W., M.v.L.T., and P.B. wrote the first draft of the paper; A.M.W. wrote the Treg part; M.S. the mTOR section, M.v.B.B. the B-cell part; and S.A.E.H. the APC section; and all authors wrote the final version of the article.

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

Correspondence: Peter Brossart, MD, or Dominik Wolf, MD, Department of Hematology and Oncology, University Hospital Bonn, Wilhelmstraße 35, 53111 Bonn, Germany; e-mail: peter.brossart@ukb.uni-bonn.de or dominik.wolf@i-med.ac.at.

1
Thomas
 
ED
Lochte
 
HLJ
LU
 
WC
FerreBee
 
JW
Intravenous infusion of bone marrow in patients receiving radiation and chemotherapy.
N Engl J Med
1957
, vol. 
257
 
11
(pg. 
491
-
496
)
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
Ferrara
 
JL
Levine
 
JE
Reddy
 
P
Holler
 
E
Graft-versus-host disease.
Lancet
2009
, vol. 
373
 
9674
(pg. 
1550
-
1561
)
4
Landfried
 
K
Wolff
 
D
Holler
 
E
Pathophysiology and management of graft-versus-host disease in the era of reduced-intensity conditioning.
Curr Opin Oncol
2009
, vol. 
21
 
Suppl 1
(pg. 
S39
-
41
)
5
Storek
 
J
Gooley
 
T
Siadak
 
M
, et al. 
Allogeneic peripheral blood stem cell transplantation may be associated with a high risk of chronic graft-versus-host disease.
Blood
1997
, vol. 
90
 
12
(pg. 
4705
-
4709
)
6
Flowers
 
ME
Inamoto
 
Y
Carpenter
 
PA
, et al. 
Comparative analysis of risk factors for acute graft-versus-host disease and for chronic graft-versus-host disease according to National Institutes of Health consensus criteria.
Blood
2011
, vol. 
117
 
11
(pg. 
3214
-
3219
)
7
Strober
 
S
Spitzer
 
TR
Lowsky
 
R
Sykes
 
M
Translational studies in hematopoietic cell transplantation: Treatment of hematologic malignancies as a stepping stone to tolerance induction. [published online ahead of print June 24, 2011]
Semin Immunol
 
8
Salmasian
 
H
Rohanizadegan
 
M
Banihosseini
 
S
, et al. 
Corticosteroid regimens for treatment of acute and chronic graft versus host disease (GvHD) after allogenic stem cell transplantation.
Cochrane Database Syst Rev
2010
1
pg. 
CD005565
 
9
Deeg
 
HJ
How I treat refractory acute GVHD.
Blood
2007
, vol. 
109
 
10
(pg. 
4119
-
4126
)
10
Matzinger
 
P
Tolerance, danger, and the extended family.
Annu Rev Immunol
1994
, vol. 
12
 (pg. 
991
-
1045
)
11
Schenk
 
U
Westendorf
 
AM
Radaelli
 
E
, et al. 
Purinergic control of T cell activation by ATP released through pannexin-1 hemichannels.
Sci Signal
2008
, vol. 
1
 
39
pg. 
ra6
 
12
Zeiser
 
R
Penack
 
O
Holler
 
E
Idzko
 
M
Danger signals activating innate immunity in graft-versus-host disease.
J Mol Med (Berl)
2011
, vol. 
89
 
9
(pg. 
833
-
845
)
13
Wilhelm
 
K
Ganesan
 
J
Muller
 
T
, et al. 
Graft-versus-host disease is enhanced by extracellular ATP activating P2X7R.
Nat Med
2010
, vol. 
16
 
12
(pg. 
1434
-
1438
)
14
Lee
 
KH
Park
 
SS
Kim
 
I
, et al. 
P2X7 receptor polymorphism and clinical outcomes in HLA-matched sibling allogeneic hematopoietic stem cell transplantation.
Haematologica
2007
, vol. 
92
 
5
(pg. 
651
-
657
)
15
Casanova
 
JL
Abel
 
L
Quintana-Murci
 
L
Human TLRs and IL-1Rs in host defense: natural insights from evolutionary, epidemiological, and clinical genetics.
Annu Rev Immunol
2011
, vol. 
29
 (pg. 
447
-
491
)
16
Yu
 
L
Wang
 
L
Chen
 
S
Endogenous toll-like receptor ligands and their biological significance.
J Cell Mol Med
2010
, vol. 
14
 
11
(pg. 
2592
-
2603
)
17
Elmaagacli
 
AH
Koldehoff
 
M
Hindahl
 
H
, et al. 
Mutations in innate immune system NOD2/CARD 15 and TLR-4 (Thr399Ile) genes influence the risk for severe acute graft-versus-host disease in patients who underwent an allogeneic transplantation.
Transplantation
2006
, vol. 
81
 
2
(pg. 
247
-
254
)
18
Taylor
 
PA
Ehrhardt
 
MJ
Lees
 
CJ
, et al. 
TLR agonists regulate alloresponses and uncover a critical role for donor APCs in allogeneic bone marrow rejection.
Blood
2008
, vol. 
112
 
8
(pg. 
3508
-
3516
)
19
Heimesaat
 
MM
Nogai
 
A
Bereswill
 
S
, et al. 
MyD88/TLR9 mediated immunopathology and gut microbiota dynamics in a novel murine model of intestinal graft-versus-host disease.
Gut
2010
, vol. 
59
 
8
(pg. 
1079
-
1087
)
20
Loiarro
 
M
Capolunghi
 
F
Fanto
 
N
, et al. 
Pivotal Advance: Inhibition of MyD88 dimerization and recruitment of IRAK1 and IRAK4 by a novel peptidomimetic compound.
J Leukoc Biol
2007
, vol. 
82
 
4
(pg. 
801
-
810
)
21
Holler
 
E
Rogler
 
G
Brenmoehl
 
J
, et al. 
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
2006
, vol. 
107
 
10
(pg. 
4189
-
4193
)
22
Maeda
 
S
Hsu
 
LC
Liu
 
H
, et al. 
Nod2 mutation in Crohn's disease potentiates NF-kappaB activity and IL-1beta processing.
Science
2005
, vol. 
307
 
5710
(pg. 
734
-
738
)
23
Penack
 
O
Smith
 
OM
Cunningham-Bussel
 
A
, et al. 
NOD2 regulates hematopoietic cell function during graft-versus-host disease.
J Exp Med
2009
, vol. 
206
 
10
(pg. 
2101
-
2110
)
24
Watanabe
 
T
Asano
 
N
Murray
 
PJ
, et al. 
Muramyl dipeptide activation of nucleotide-binding oligomerization domain 2 protects mice from experimental colitis.
J Clin Invest
2008
, vol. 
118
 
2
(pg. 
545
-
559
)
25
Zhang
 
Y
Louboutin
 
JP
Zhu
 
J
Rivera
 
AJ
Emerson
 
SG
Preterminal host dendritic cells in irradiated mice prime CD8+ T cell-mediated acute graft-versus-host disease.
J Clin Invest
2002
, vol. 
109
 
10
(pg. 
1335
-
1344
)
26
Merad
 
M
Manz
 
MG
Karsunky
 
H
, et al. 
Langerhans cells renew in the skin throughout life under steady-state conditions.
Nat Immunol
2002
, vol. 
3
 
12
(pg. 
1135
-
1141
)
27
Merad
 
M
Hoffmann
 
P
Ranheim
 
E
, et al. 
Depletion of host Langerhans cells before transplantation of donor alloreactive T cells prevents skin graft-versus-host disease.
Nat Med
2004
, vol. 
10
 
5
(pg. 
510
-
517
)
28
Lee
 
YR
Yang
 
IH
Lee
 
YH
, et al. 
Cyclosporin A and tacrolimus, but not rapamycin, inhibit MHC-restricted antigen presentation pathways in dendritic cells.
Blood
2005
, vol. 
105
 
10
(pg. 
3951
-
3955
)
29
Mehling
 
A
Grabbe
 
S
Voskort
 
M
Schwarz
 
T
Luger
 
TA
Beissert
 
S
Mycophenolate mofetil impairs the maturation and function of murine dendritic cells.
J Immunol
2000
, vol. 
165
 
5
(pg. 
2374
-
2381
)
30
Piemonti
 
L
Monti
 
P
Allavena
 
P
, et al. 
Glucocorticoids affect human dendritic cell differentiation and maturation.
J Immunol
1999
, vol. 
162
 
11
(pg. 
6473
-
6481
)
31
Nencioni
 
A
Beck
 
J
Werth
 
D
, et al. 
Histone deacetylase inhibitors affect dendritic cell differentiation and immunogenicity.
Clin Cancer Res
2007
, vol. 
13
 
13
(pg. 
3933
-
3941
)
32
Reddy
 
P
Sun
 
Y
Toubai
 
T
, et al. 
Histone deacetylase inhibition modulates indoleamine 2,3-dioxygenase-dependent DC functions and regulates experimental graft-versus-host disease in mice.
J Clin Invest
2008
, vol. 
118
 
7
(pg. 
2562
-
2573
)
33
Li
 
N
Zhao
 
D
Kirschbaum
 
M
, et al. 
HDAC inhibitor reduces cytokine storm and facilitates induction of chimerism that reverses lupus in anti-CD3 conditioning regimen.
Proc Natl Acad Sci U S A
2008
, vol. 
105
 
12
(pg. 
4796
-
4801
)
34
Tao
 
R
de Zoeten
 
EF
Ozkaynak
 
E
, et al. 
Deacetylase inhibition promotes the generation and function of regulatory T cells.
Nat Med
2007
, vol. 
13
 
11
(pg. 
1299
-
1307
)
35
Magro
 
L
Catteau
 
B
Coiteux
 
V
Bruno
 
B
Jouet
 
JP
Yakoub-Agha
 
I
Efficacy of imatinib mesylate in the treatment of refractory sclerodermatous chronic GVHD.
Bone Marrow Transplant
2008
, vol. 
42
 
11
(pg. 
757
-
760
)
36
Johnson
 
RJ
Raines
 
EW
Floege
 
J
, et al. 
Inhibition of mesangial cell proliferation and matrix expansion in glomerulonephritis in the rat by antibody to platelet-derived growth factor.
J Exp Med
1992
, vol. 
175
 
5
(pg. 
1413
-
1416
)
37
Svegliati
 
S
Olivieri
 
A
Campelli
 
N
, et al. 
Stimulatory autoantibodies to PDGF receptor in patients with extensive chronic graft-versus-host disease.
Blood
2007
, vol. 
110
 
1
(pg. 
237
-
241
)
38
Olivieri
 
A
Locatelli
 
F
Zecca
 
M
, et al. 
Imatinib for refractory chronic graft-versus-host disease with fibrotic features.
Blood
2009
, vol. 
114
 
3
(pg. 
709
-
718
)
39
Magro
 
L
Mohty
 
M
Catteau
 
B
, et al. 
Imatinib mesylate as salvage therapy for refractory sclerotic chronic graft-versus-host disease.
Blood
2009
, vol. 
114
 
3
(pg. 
719
-
722
)
40
Appel
 
S
Rupf
 
A
Weck
 
MM
, et al. 
Effects of imatinib on monocyte-derived dendritic cells are mediated by inhibition of nuclear factor-kappaB and Akt signaling pathways.
Clin Cancer Res
2005
, vol. 
11
 
5
(pg. 
1928
-
1940
)
41
Wolf
 
AM
Wolf
 
D
Rumpold
 
H
, et al. 
The kinase inhibitor imatinib mesylate inhibits TNF-{alpha} production in vitro and prevents TNF-dependent acute hepatic inflammation.
Proc Natl Acad Sci U S A
2005
, vol. 
102
 
38
(pg. 
13622
-
13627
)
42
Mumprecht
 
S
Matter
 
M
Pavelic
 
V
Ochsenbein
 
AF
Imatinib mesylate selectively impairs expansion of memory cytotoxic T cells without affecting the control of primary viral infections.
Blood
2006
, vol. 
108
 
10
(pg. 
3406
-
3413
)
43
Chen
 
J
Schmitt
 
A
Chen
 
B
, et al. 
Nilotinib hampers the proliferation and function of CD8+ T lymphocytes through inhibition of T cell receptor signalling.
J Cell Mol Med
2008
, vol. 
12
 
5B
(pg. 
2107
-
2118
)
44
Cavet
 
J
Dickinson
 
AM
Norden
 
J
Taylor
 
PR
Jackson
 
GH
Middleton
 
PG
Interferon-gamma and interleukin-6 gene polymorphisms associate with graft-versus-host disease in HLA-matched sibling bone marrow transplantation.
Blood
2001
, vol. 
98
 
5
(pg. 
1594
-
1600
)
45
Ritchie
 
D
Seconi
 
J
Wood
 
C
Walton
 
J
Watt
 
V
Prospective monitoring of tumor necrosis factor alpha and interferon gamma to predict the onset of acute and chronic graft-versus-host disease after allogeneic stem cell transplantation.
Biol Blood Marrow Transplant
2005
, vol. 
11
 
9
(pg. 
706
-
712
)
46
Roy
 
J
Blazar
 
BR
Ochs
 
L
Weisdorf
 
DJ
The tissue expression of cytokines in human acute cutaneous graft-versus-host disease.
Transplantation
1995
, vol. 
60
 
4
(pg. 
343
-
348
)
47
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
)
48
Ratajczak
 
P
Janin
 
A
Peffault de Latour
 
R
, et al. 
Th17/Treg ratio in human graft-versus-host disease.
Blood
2010
, vol. 
116
 
7
(pg. 
1165
-
1171
)
49
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
)
50
Hanash
 
AM
Kappel
 
LW
Yim
 
NL
, et al. 
Abrogation of donor T cell IL-21 signaling leads to tissue-specific modulation of immunity and separation of GVHD from GVL.
Blood
2011
, vol. 
118
 
2
(pg. 
446
-
455
)
51
Ma
 
H
Lu
 
C
Ziegler
 
J
, et al. 
Absence of Stat1 in donor CD4+ T cells promotes the expansion of Tregs and reduces graft-versus-host disease in mice.
J Clin Invest
2011
, vol. 
121
 
7
(pg. 
2554
-
2569
)
52
Baier
 
G
Wagner
 
J
PKC inhibitors: potential in T cell-dependent immune diseases.
Curr Opin Cell Biol
2009
, vol. 
21
 
2
(pg. 
262
-
267
)
53
Valenzuela
 
JO
Iclozan
 
C
Hossain
 
MS
, et al. 
PKCtheta is required for alloreactivity and GVHD but not for immune responses toward leukemia and infection in mice.
J Clin Invest
2009
, vol. 
119
 
12
(pg. 
3774
-
3786
)
54
Skvara
 
H
Dawid
 
M
Kleyn
 
E
, et al. 
The PKC inhibitor AEB071 may be a therapeutic option for psoriasis.
J Clin Invest
2008
, vol. 
118
 
9
(pg. 
3151
-
3159
)
55
Vincenti
 
F
Kirk
 
AD
What's next in the pipeline.
Am J Transplant
2008
, vol. 
8
 
10
(pg. 
1972
-
1981
)
56
Beilhack
 
A
Schulz
 
S
Baker
 
J
, et al. 
In vivo analyses of early events in acute graft-versus-host disease reveal sequential infiltration of T-cell subsets.
Blood
2005
, vol. 
106
 
3
(pg. 
1113
-
1122
)
57
Kim
 
YM
Sachs
 
T
Asavaroengchai
 
W
Bronson
 
R
Sykes
 
M
Graft-versus-host disease can be separated from graft-versus-lymphoma effects by control of lymphocyte trafficking with FTY720.
J Clin Invest
2003
, vol. 
111
 
5
(pg. 
659
-
669
)
58
Schwab
 
SR
Cyster
 
JG
Finding a way out: lymphocyte egress from lymphoid organs.
Nat Immunol
2007
, vol. 
8
 
12
(pg. 
1295
-
1301
)
59
Beilhack
 
A
Schulz
 
S
Baker
 
J
, et al. 
Prevention of acute graft-versus-host disease by blocking T-cell entry to secondary lymphoid organs.
Blood
2008
, vol. 
111
 
5
(pg. 
2919
-
2928
)
60
Johansson-Lindbom
 
B
Svensson
 
M
Wurbel
 
MA
Malissen
 
B
Marquez
 
G
Agace
 
W
Selective generation of gut tropic T cells in gut-associated lymphoid tissue (GALT): requirement for GALT dendritic cells and adjuvant.
J Exp Med
2003
, vol. 
198
 
6
(pg. 
963
-
969
)
61
Blanco
 
B
Perez-Simon
 
JA
Sanchez-Abarca
 
LI
, et al. 
Bortezomib induces selective depletion of alloreactive T lymphocytes and decreases the production of Th1 cytokines.
Blood
2006
, vol. 
107
 
9
(pg. 
3575
-
3583
)
62
Sun
 
K
Welniak
 
LA
Panoskaltsis-Mortari
 
A
, et al. 
Inhibition of acute graft-versus-host disease with retention of graft-versus-tumor effects by the proteasome inhibitor bortezomib.
Proc Natl Acad Sci U S A
2004
, vol. 
101
 
21
(pg. 
8120
-
8125
)
63
Sun
 
K
Wilkins
 
DE
Anver
 
MR
, et al. 
Differential effects of proteasome inhibition by bortezomib on murine acute graft-versus-host disease (GVHD): delayed administration of bortezomib results in increased GVHD-dependent gastrointestinal toxicity.
Blood
2005
, vol. 
106
 
9
(pg. 
3293
-
3299
)
64
Mateos-Mazon
 
J
Perez-Simon
 
JA
Lopez
 
O
Hernandez
 
E
Etxebarria
 
J
San Miguel
 
JF
Use of bortezomib in the management of chronic graft-versus-host disease among multiple myeloma patients relapsing after allogeneic transplantation.
Haematologica
2007
, vol. 
92
 
9
(pg. 
1295
-
1296
)
65
Koreth
 
J
Stevenson
 
KE
Kim
 
HT
, et al. 
Bortezomib, tacrolimus, and methotrexate for prophylaxis of graft-versus-host disease after reduced-intensity conditioning allogeneic stem cell transplantation from HLA-mismatched unrelated donors.
Blood
2009
, vol. 
114
 
18
(pg. 
3956
-
3959
)
66
Jiang
 
S
Walker
 
L
Afentoulis
 
M
, et al. 
Transplanted human bone marrow contributes to vascular endothelium.
Proc Natl Acad Sci U S A
2004
, vol. 
101
 
48
(pg. 
16891
-
16896
)
67
Limmer
 
A
Ohl
 
J
Kurts
 
C
, et al. 
Efficient presentation of exogenous antigen by liver endothelial cells to CD8+ T cells results in antigen-specific T-cell tolerance.
Nat Med
2000
, vol. 
6
 
12
(pg. 
1348
-
1354
)
68
Kreisel
 
D
Krupnick
 
AS
Gelman
 
AE
, et al. 
Non-hematopoietic allograft cells directly activate CD8+ T cells and trigger acute rejection: an alternative mechanism of allorecognition.
Nat Med
2002
, vol. 
8
 
3
(pg. 
233
-
239
)
69
Biedermann
 
BC
Sahner
 
S
Gregor
 
M
, et al. 
Endothelial injury mediated by cytotoxic T lymphocytes and loss of microvessels in chronic graft versus host disease.
Lancet
2002
, vol. 
359
 
9323
(pg. 
2078
-
2083
)
70
Penack
 
O
Henke
 
E
Suh
 
D
, et al. 
Inhibition of neovascularization to simultaneously ameliorate graft-vs-host disease and decrease tumor growth.
J Natl Cancer Inst
2010
, vol. 
102
 
12
(pg. 
894
-
908
)
71
Nachbaur
 
D
Schumacher
 
P
Auberger
 
J
Clausen
 
J
Kircher
 
B
Vascular endothelial growth factor and activin-a serum levels following allogeneic hematopoietic stem cell transplantation.
Biol Blood Marrow Transplant
2007
, vol. 
13
 
8
(pg. 
942
-
947
)
72
Luft
 
T
Dietrich
 
S
Falk
 
C
, et al. 
Steroid-refractory GVHD: T-cell attack within a vulnerable endothelial system.
Blood
2011
, vol. 
118
 
6
(pg. 
1685
-
1692
)
73
Heine
 
A
Held
 
SA
Bringmann
 
A
Holderried
 
TA
Brossart
 
P
Immunomodulatory effects of anti-angiogenic drugs.
Leukemia
2011
, vol. 
25
 
6
(pg. 
899
-
905
)
74
Vogelsang
 
GB
Farmer
 
ER
Hess
 
AD
, et al. 
Thalidomide for the treatment of chronic graft-versus-host disease.
N Engl J Med
1992
, vol. 
326
 
16
(pg. 
1055
-
1058
)
75
Browne
 
PV
Weisdorf
 
DJ
DeFor
 
T
, et al. 
Response to thalidomide therapy in refractory chronic graft-versus-host disease.
Bone Marrow Transplant
2000
, vol. 
26
 
8
(pg. 
865
-
869
)
76
Kulkarni
 
S
Powles
 
R
Sirohi
 
B
, et al. 
Thalidomide after allogeneic haematopoietic stem cell transplantation: activity in chronic but not in acute graft-versus-host disease.
Bone Marrow Transplant
2003
, vol. 
32
 
2
(pg. 
165
-
170
)
77
Chao
 
NJ
Parker
 
PM
Niland
 
JC
, et al. 
Paradoxical effect of thalidomide prophylaxis on chronic graft-vs.-host disease.
Biol Blood Marrow Transplant
1996
, vol. 
2
 
2
(pg. 
86
-
92
)
78
Kneppers
 
E
van der Holt
 
B
Kersten
 
MJ
, et al. 
Lenalidomide maintenance following non-myeloablative allogeneic stem cell transplantation in multiple myeloma is not feasible: results of the HOVON 76 trial.
Blood
2011
, vol. 
118
 
9
(pg. 
2413
-
2419
)
79
Hoffmann
 
P
Ermann
 
J
Edinger
 
M
Fathman
 
CG
Strober
 
S
Donor-type CD4(+)CD25(+) regulatory T cells suppress lethal acute graft-versus-host disease after allogeneic bone marrow transplantation.
J Exp Med
2002
, vol. 
196
 
3
(pg. 
389
-
399
)
80
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
)
81
Rezvani
 
K
Mielke
 
S
Ahmadzadeh
 
M
, et al. 
High donor FOXP3-positive regulatory T-cell (Treg) content is associated with a low risk of GVHD following HLA-matched allogeneic SCT.
Blood
2006
, vol. 
108
 
4
(pg. 
1291
-
1297
)
82
Wolf
 
D
Wolf
 
AM
Fong
 
D
, et al. 
Regulatory T cells in the graft and the risk of acute graft-versus-host disease after allogeneic stem cell transplantation.
Transplantation
2007
, vol. 
83
 
8
(pg. 
1107
-
1113
)
83
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
)
84
Wolf
 
AM
Eller
 
K
Zeiser
 
R
, et al. 
The sphingosine 1-phosphate receptor agonist FTY720 potently inhibits regulatory T cell proliferation in vitro and in vivo.
J Immunol
2009
, vol. 
183
 
6
(pg. 
3751
-
3760
)
85
Procaccini
 
C
De Rosa
 
V
Galgani
 
M
, et al. 
An oscillatory switch in mTOR kinase activity sets regulatory T cell responsiveness.
Immunity
2010
, vol. 
33
 
6
(pg. 
929
-
941
)
86
Battaglia
 
M
Stabilini
 
A
Roncarolo
 
MG
Rapamycin selectively expands CD4+CD25+FoxP3+ regulatory T cells.
Blood
2005
, vol. 
105
 
12
(pg. 
4743
-
4748
)
87
Tresoldi
 
E
Dell'albani
 
I
Stabilini
 
A
, et al. 
Stability of human rapamycin-expanded CD4+CD25+ T regulatory cells.
Haematologica
2011
, vol. 
96
 
9
(pg. 
1357
-
1365
)
88
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
)
89
Coenen
 
JJ
Koenen
 
HJ
van Rijssen
 
E
, et al. 
Rapamycin, not cyclosporine, permits thymic generation and peripheral preservation of CD4+ CD25+ FoxP3+ T cells.
Bone Marrow Transplant
2007
, vol. 
39
 
9
(pg. 
537
-
545
)
90
Palmer
 
JM
Chen
 
BJ
DeOliveira
 
D
Le
 
ND
Chao
 
NJ
Novel mechanism of rapamycin in GVHD: increase in interstitial regulatory T cells.
Bone Marrow Transplant
2010
, vol. 
45
 
2
(pg. 
379
-
384
)
91
Haribhai
 
D
Williams
 
JB
Jia
 
S
, et al. 
A Requisite Role for Induced Regulatory T Cells in Tolerance Based on Expanding Antigen Receptor Diversity.
Immunity
2011
, vol. 
35
 
1
(pg. 
109
-
122
)
92
Shin
 
HJ
Baker
 
J
Leveson-Gower
 
DB
Smith
 
AT
Sega
 
EI
Negrin
 
RS
Rapamycin and IL-2 reduce lethal acute graft-versus-host disease associated with increased expansion of donor type CD4+CD25+Foxp3+ regulatory T cells.
Blood
2011
, vol. 
118
 
8
(pg. 
2342
-
2350
)
93
Schleuning
 
M
Judith
 
D
Jedlickova
 
Z
, et al. 
Calcineurin inhibitor-free GVHD prophylaxis with sirolimus, mycophenolate mofetil and ATG in Allo-SCT for leukemia patients with high relapse risk: an observational cohort study.
Bone Marrow Transplant
2009
, vol. 
43
 
9
(pg. 
717
-
723
)
94
Cutler
 
C
Stevenson
 
K
Kim
 
HT
, et al. 
Sirolimus is associated with veno-occlusive disease of the liver after myeloablative allogeneic stem cell transplantation.
Blood
2008
, vol. 
112
 
12
(pg. 
4425
-
4431
)
95
Platzbecker
 
U
von Bonin
 
M
Goekkurt
 
E
, et al. 
Graft-versus-host disease prophylaxis with everolimus and tacrolimus is associated with a high incidence of sinusoidal obstruction syndrome and microangiopathy: results of the EVTAC trial.
Biol Blood Marrow Transplant
2009
, vol. 
15
 
1
(pg. 
101
-
108
)
96
Hoda
 
D
Pidala
 
J
Salgado-Vila
 
N
, et al. 
Sirolimus for treatment of steroid-refractory acute graft-versus-host disease.
Bone Marrow Transplant
2010
, vol. 
45
 
8
(pg. 
1347
-
1351
)
97
Pidala
 
J
Kim
 
J
Anasetti
 
C
Sirolimus as primary treatment of acute graft-versus-host disease following allogeneic hematopoietic cell transplantation.
Biol Blood Marrow Transplant
2009
, vol. 
15
 
7
(pg. 
881
-
885
)
98
Jedlickova
 
Z
Burlakova
 
I
Bug
 
G
Baurmann
 
H
Schwerdtfeger
 
R
Schleuning
 
M
Therapy of sclerodermatous chronic graft-versus-host disease with mammalian target of rapamycin inhibitors.
Biol Blood Marrow Transplant
2011
, vol. 
17
 
5
(pg. 
657
-
663
)
99
Brigl
 
M
Brenner
 
MB
CD1: antigen presentation and T cell function.
Annu Rev Immunol
2004
, vol. 
22
 (pg. 
817
-
890
)
100
Kronenberg
 
M
Toward an understanding of NKT cell biology: progress and paradoxes.
Annu Rev Immunol
2005
, vol. 
23
 (pg. 
877
-
900
)
101
Taniguchi
 
M
Harada
 
M
Kojo
 
S
Nakayama
 
T
Wakao
 
H
The regulatory role of Valpha14 NKT cells in innate and acquired immune response.
Annu Rev Immunol
2003
, vol. 
21
 (pg. 
483
-
513
)
102
Leveson-Gower
 
DB
Olson
 
JA
Sega
 
EI
, et al. 
Low doses of natural killer T cells provide protection from acute graft-versus-host disease via an IL-4-dependent mechanism.
Blood
2011
, vol. 
117
 
11
(pg. 
3220
-
3229
)
103
Pillai
 
AB
George
 
TI
Dutt
 
S
Teo
 
P
Strober
 
S
Host NKT cells can prevent graft-versus-host disease and permit graft antitumor activity after bone marrow transplantation.
J Immunol
2007
, vol. 
178
 
10
(pg. 
6242
-
6251
)
104
Zeng
 
D
Lewis
 
D
Dejbakhsh-Jones
 
S
, et al. 
Bone marrow NK1.1(-) and NK1.1(+) T cells reciprocally regulate acute graft versus host disease.
J Exp Med
1999
, vol. 
189
 
7
(pg. 
1073
-
1081
)
105
Hashimoto
 
D
Asakura
 
S
Miyake
 
S
, et al. 
Stimulation of host NKT cells by synthetic glycolipid regulates acute graft-versus-host disease by inducing Th2 polarization of donor T cells.
J Immunol
2005
, vol. 
174
 
1
(pg. 
551
-
556
)
106
Singh
 
N
Hong
 
S
Scherer
 
DC
, et al. 
Cutting edge: activation of NK T cells by CD1d and alpha-galactosylceramide directs conventional T cells to the acquisition of a Th2 phenotype.
J Immunol
1999
, vol. 
163
 
5
(pg. 
2373
-
2377
)
107
Lan
 
F
Zeng
 
D
Higuchi
 
M
Huie
 
P
Higgins
 
JP
Strober
 
S
Predominance of NK1.1+TCR alpha beta+ or DX5+TCR alpha beta+ T cells in mice conditioned with fractionated lymphoid irradiation protects against graft-versus-host disease: “natural suppressor” cells.
J Immunol
2001
, vol. 
167
 
4
(pg. 
2087
-
2096
)
108
Rigby
 
SM
Rouse
 
T
Field
 
EH
Total lymphoid irradiation nonmyeloablative preconditioning enriches for IL-4-producing CD4+-TNK cells and skews differentiation of immunocompetent donor CD4+ cells.
Blood
2003
, vol. 
101
 
5
(pg. 
2024
-
2032
)
109
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
)
110
Haraguchi
 
K
Takahashi
 
T
Matsumoto
 
A
, et al. 
Host-residual invariant NK T cells attenuate graft-versus-host immunity.
J Immunol
2005
, vol. 
175
 
2
(pg. 
1320
-
1328
)
111
Prockop
 
DJ
Marrow stromal cells as stem cells for nonhematopoietic tissues.
Science
1997
, vol. 
276
 
5309
(pg. 
71
-
74
)
112
Le Blanc
 
K
Rasmusson
 
I
Sundberg
 
B
, et al. 
Treatment of severe acute graft-versus-host disease with third party haploidentical mesenchymal stem cells.
Lancet
2004
, vol. 
363
 
9419
(pg. 
1439
-
1441
)
113
Tolar
 
J
Le Blanc
 
K
Keating
 
A
Blazar
 
BR
Concise review: hitting the right spot with mesenchymal stromal cells.
Stem Cells
2010
, vol. 
28
 
8
(pg. 
1446
-
1455
)
114
Meisel
 
R
Zibert
 
A
Laryea
 
M
Gobel
 
U
Daubener
 
W
Dilloo
 
D
Human bone marrow stromal cells inhibit allogeneic T-cell responses by indoleamine 2,3-dioxygenase-mediated tryptophan degradation.
Blood
2004
, vol. 
103
 
12
(pg. 
4619
-
4621
)
115
Selmani
 
Z
Naji
 
A
Zidi
 
I
, et al. 
Human leukocyte antigen-G5 secretion by human mesenchymal stem cells is required to suppress T lymphocyte and natural killer function and to induce CD4+CD25highFOXP3+ regulatory T cells.
Stem Cells
2008
, vol. 
26
 
1
(pg. 
212
-
222
)
116
Le Blanc
 
K
Tammik
 
C
Rosendahl
 
K
Zetterberg
 
E
Ringden
 
O
HLA expression and immunologic properties of differentiated and undifferentiated mesenchymal stem cells.
Exp Hematol
2003
, vol. 
31
 
10
(pg. 
890
-
896
)
117
von Bonin
 
M
Stolzel
 
F
Goedecke
 
A
, et al. 
Treatment of refractory acute GVHD with third-party MSC expanded in platelet lysate-containing medium.
Bone Marrow Transplant
2009
, vol. 
43
 
3
(pg. 
245
-
251
)
118
Le Blanc
 
K
Frassoni
 
F
Ball
 
L
, et al. 
Mesenchymal stem cells for treatment of steroid-resistant, severe, acute graft-versus-host disease: a phase II study.
Lancet
2008
, vol. 
371
 
9624
(pg. 
1579
-
1586
)
119
Karlsson
 
H
Samarasinghe
 
S
Ball
 
LM
, et al. 
Mesenchymal stem cells exert differential effects on alloantigen and virus-specific T-cell responses.
Blood
2008
, vol. 
112
 
3
(pg. 
532
-
541
)
120
Weng
 
JY
Du
 
X
Geng
 
SX
, et al. 
Mesenchymal stem cell as salvage treatment for refractory chronic GVHD.
Bone Marrow Transplant
2010
, vol. 
45
 
12
(pg. 
1732
-
1740
)
121
Chan
 
OT
Hannum
 
LG
Haberman
 
AM
Madaio
 
MP
Shlomchik
 
MJ
A novel mouse with B cells but lacking serum antibody reveals an antibody-independent role for B cells in murine lupus.
J Exp Med
1999
, vol. 
189
 
10
(pg. 
1639
-
1648
)
122
Chan
 
O
Shlomchik
 
MJ
A new role for B cells in systemic autoimmunity: B cells promote spontaneous T cell activation in MRL-lpr/lpr mice.
J Immunol
1998
, vol. 
160
 
1
(pg. 
51
-
59
)
123
Rahman
 
A
Isenberg
 
DA
Systemic lupus erythematosus.
N Engl J Med
2008
, vol. 
358
 
9
(pg. 
929
-
939
)
124
Greinix
 
HT
Pohlreich
 
D
Kouba
 
M
, et al. 
Elevated numbers of immature/transitional CD21- B lymphocytes and deficiency of memory CD27+ B cells identify patients with active chronic graft-versus-host disease.
Biol Blood Marrow Transplant
2008
, vol. 
14
 
2
(pg. 
208
-
219
)
125
Sarantopoulos
 
S
Stevenson
 
KE
Kim
 
HT
, et al. 
Altered B cell homeostasis and excess BAFF in human chronic graft-versus-host disease.
Blood
2009
, vol. 
113
 
16
(pg. 
3865
-
3874
)
126
Christopeit
 
M
Schutte
 
V
Theurich
 
S
Weber
 
T
Grothe
 
W
Behre
 
G
Rituximab reduces the incidence of acute graft-versus-host disease.
Blood
2009
, vol. 
113
 
13
(pg. 
3130
-
3131
)
127
Crocchiolo
 
R
Castagna
 
L
El-Cheikh
 
J
, et al. 
Prior rituximab administration is associated with reduced rate of acute GVHD after in vivo T-cell depleted transplantation in lymphoma patients.
Exp Hematol
2011
, vol. 
39
 
9
(pg. 
892
-
896
)
128
Dominietto
 
A
Tedone
 
E
Soracco
 
M
, et al. 
In vivo B cell depletion with rituximab for alternative donor hemopoietic SCT. [published online ahead of print Apr 4, 2011]
Bone Marrow Transplant
 
129
Mohty
 
M
Mechanisms of action of antithymocyte globulin: T-cell depletion and beyond.
Leukemia
2007
, vol. 
21
 
7
(pg. 
1387
-
1394
)
130
Hamadani
 
M
Awan
 
FT
Devine
 
SM
The impact of HMG-CoA reductase inhibition on the incidence and severity of graft-versus-host disease in patients with acute leukemia undergoing allogeneic transplantation.
Blood
2008
, vol. 
111
 
7
(pg. 
3901
-
3902
)
131
Shimabukuro-Vornhagen
 
A
Liebig
 
T
von Bergwelt-Baildon
 
M
Statins inhibit human APC function: implications for the treatment of GVHD.
Blood
2008
, vol. 
112
 
4
(pg. 
1544
-
1545
)
132
Tokunaga
 
M
Fujii
 
K
Saito
 
K
, et al. 
Down-regulation of CD40 and CD80 on B cells in patients with life-threatening systemic lupus erythematosus after successful treatment with rituximab.
Rheumatology (Oxford)
2005
, vol. 
44
 
2
(pg. 
176
-
182
)
133
van Dorp
 
S
Resemann
 
H
Te Boome
 
L
, et al. 
The immunological phenotype of rituximab-sensitive chronic GVHD: a phase II study.
Haematologica
2011
, vol. 
96
 
9
(pg. 
1380
-
1384
)
134
Zaja
 
F
Bacigalupo
 
A
Patriarca
 
F
, et al. 
Treatment of refractory chronic GVHD with rituximab: a GITMO study.
Bone Marrow Transplant
2007
, vol. 
40
 
3
(pg. 
273
-
277
)
Sign in via your Institution