Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2014 Mar 5.
Published in final edited form as: Curr Opin Hematol. 2009 Nov;16(6):427–436. doi: 10.1097/MOH.0b013e3283319a6f

Acute Graft versus Host Disease: New Treatment Strategies

Sophie Paczesny a, Sung W Choi a, James LM Ferrara a
PMCID: PMC3943343  NIHMSID: NIHMS378068  PMID: 19812490

Abstract

Purpose of the review

Graft-versus host disease (GVHD) remains a major cause of morbidity and mortality after allogeneic hematopoietic cell transplantation (HCT), despite improvements in our understanding of its pathophysiology as well as the generation of new monoclonal antibodies, immunomodulatory chemotherapy, cellular therapeutics and supportive care. Herein, we review therapies that have proven effective as well as newer agents that have recently improved GVHD response rates and survival following HCT.

Recent findings

Novel approaches to prevent or treat GVHD are often based on evidence from experimental models. Our understanding of the pathophysiology of GVHD may lead to the development of innovative strategies that target both soluble and cellular effectors. Among such agents are sirolimus, anti-TNF antibodies, anti-LFA-3–IgG fusion protein, extracorporeal photopheresis, mesenchymal stem cells and regulatory T cells.

Summary

Obstacles to the improvement of HCT include the tight linkage between GVHD toxicity and the beneficial graft versus leukemia effect (GVL), as well as the impairment of immune reconstitution by immunomodulatory drugs leading to life-threatening infections. The design of newer phase I/II clinical trials are underway. Future therapies are likely to include modulation of cell types that play key roles in the GVH process, including regulatory T cells, dendritic cells, NKT cells and B cells.

Keywords: Allogeneic hematopoietic cell transplantation (HCT), graft versus host disease (GVHD), immunomodulatory drugs

Introduction

Graft versus Host Disease (GVHD) is the principal complication of allogeneic HCT that limits the wider application of this therapeutic approach to patients with high-risk hematologic malignancies. The pathophysiology of acute GVHD is complex and can be considered in a framework of three sequential phases. In Phase I, the recipient conditioning regimen damages host tissues and causes release of pro-inflammatory cytokines. As a consequence, host antigen presenting cells (APCs) mature, acquiring adhesion and co-stimulatory molecules. In Phase II, host APCs activate mature donor T cells which subsequently proliferate and produce additional cytokines. Phase III involves inflammatory and cellular effectors that trigger additional inflammatory responses and together mediate target tissue damage [1**,2*]. Novel agents can act at different points of these three phases, and most current therapies are not specific to any single phase.

Prevention of GVHD

The most widely used GVHD prophylaxis following full intensity conditioning includes a combination of a calcineurin inhibitor (e.g. cyclosporine, tacrolimus, sirolimus) with “short course” methotrexate (MTX). This standard regimen was first described in 1986 by Storb et al. [3] and several clinical trials have shown superiority in reducing the incidence of GVHD and improving survival using this combination compared to either agent alone [4-6]. A recent meta-analysis of prophylaxis regimens for GVHD further supports the use of cyclosporine-MTX or tacrolimus-MTX over cyclosporine alone [7*]. Tacrolimus and cyclosporine both interrupt the T-lymphocyte signaling pathway via inhibition of calcineurin, an activator of Nuclear Factor of Activated T cell (NFATc). In many centers tacrolimus has replaced cyclosporine; several studies have shown that tacrolimus-MTX is superior to cyclosporine-MTX in reducing acute GVHD although long-term survival is not affected [5,8].

Several other immunosuppressive agents are also used as GVHD prophylaxis. Sirolimus, mTOR (mammalian Target of Rapamycin), an inhibitor of activated T cells via coupling to FK binding protein 12 (FKBP12), may also expand and maintain of CD4+CD25hiFOXP3+ regulatory T cells (Tregs) [9,10]. Furthermore, sirolimus may inhibit functions of dendritic cells, which are important in the initiation of GVHD [11-14].

The combination of sirolimus and tacrolimus has resulted in rapid engraftment, a low incidence of acute GVHD, reduced transplant-related toxicity, and improved survival in phase II trials [15,16]. The Bone Marrow Transplant Clinical Trials Network (BMT-CTN) is currently conducting a prospective phase III trial of sirolimus-tacrolimus versus tacrolimus-MTX following HLA-matched, related peripheral blood stem cell transplantation. Recent reports of sinusoidal obstruction syndrome/veno-occlusive disease have been associated with sirolimus [16,17].

Mycophenolate mofetil (MMF) is the prodrug of mycophenolic acid which is a selective inhibitor of inosine monophosphate dehydrogenase, an enzyme critical to the de novo synthesis of guanosine nucleotide. MMF inhibits T cell proliferation, and is now commonly used in combination with a calcineurin inhibitor for GVHD prophylaxis, although the optimal prophylaxis regimen following reduced-intensity HCT is not well established [18-22].

Multiple factors influence the strategies to prevent GVHD in individual patients, including risk of relapse, organ dysfunction, patient performance status, and risk of infections. A recent study of international HCT registry data from 1995 to 2002 reported risk factors for grade II-IV acute GVHD in 1,960 adults after HLA-identical sibling myeloablative transplant for leukemia [23*]. The cumulative incidence of grade II to IV acute GVHD was 35% (95% CI, 33% to 37%). In multivariable analyses, factors significantly associated with grade II to IV acute GVHD were total-body irradiation versus busulfan peripheral blood versus bone marrow, recipient age 40 and older, CML versus AML/ALL, white/Black versus Asian/Hispanic race, Karnofsky performance score less than 90, and recipient/donor cytomegalovirus-seronegative versus either positive.

For recipients of HLA-mismatched donor grafts, many centers have previously attempted to decrease the risk of GVHD by ex-vivo T-cell depletion. This approach has been limited, however, by an increased incidence of relapse as well as life-threatening infections [24]. Anti-thymocyte globulin (ATG) or alemtuzumab have been used to deplete T cells in vivo. ATG is a polyclonal antibodies mixture either of horse or rabbit origin that are directed against multiple epitopes of human T cells [25-29]. Alemtuzumab is a monoclonal antibody specific for CD52 antigen, which is present on all human leukocytes [30-34]. These approaches can reduce acute GVHD but they increase the same complications observed with ex-vivo T-cell depletion [35-37]. With respect to long-term overall survival, the benefit of these prophylactic regimens compared to a calcineurin inhibitor + MTX remains uncertain.

Treatment of acute GVHD

Acute GVHD can include the skin, liver and gastrointestinal (GI) tract and the current grading system (grades I-IV) for acute GVHD is based upon the extent of target organ involvement [38]. Significant (grade II or greater) GVHD is usually treated with high dose methylprednisone, typically starting at 2 mg/kg/day. Durable responses occur in less than half of patients with grade II to IV GVHD [39-42], and the most important predictor of long-term survival in patient with acute GVHD is the primary response to the first line of treatment [42]. Higher doses (> 2 mg/kg/day) have failed to show any advantage [43]. Because steroid-resistant GVHD has such a poor outcome, many clinicians prefer aggressive primary treatment of GVHD, even though protracted therapies with steroids incur multiple complications. Treatment with high dose steroids often continues for 7 days or more, with a progressive taper over 8 weeks or longer depending on the clinical response. In this light, a recent retrospective study from Seattle is particularly interesting. Low-dose prednisone (1 mg/kg/day) was compared to standard dose (2 mg/kg/day) for initial treatment of acute GVHD [44*]. The non-relapse mortality and overall survival were similar between regimens, with a reduced risk of invasive fungal infections and shorter hospitalizations in the low-dose prednisone group.

Local steroid therapy is also often commonly used in low grade GVHD, specifically of the skin and GI tract. For grade I skin GVHD, many centers use triamcinolone 0.1% ointment or cream to affected areas, and hydrocortisone 0.1% ointment or cream to affected facial lesions. For GI GVHD, oral non-absorbable steroids such as budesonide or beclamethasone dipropionate (BDP) have been used [45*]. A small single-center study of oral budesonide in addition to systemic corticosteroids in patients with acute GI tract GVHD showed complete responses in 77% of patients compared to 32% in the historical controls [46]. A randomized, placebo-controlled trial of oral BDP for GI tract GVHD showed that oral BDP reduced flares of GVHD following a prednisone taper and resulted in superior survival at 1 year post transplant [47]. Other attempts to deliver the steroids to the target organ include intra-arterial administration of steroids for GI and hepatic GVHD [48,49]. A recent study of 17 patients with systemic, steroid-resistant GVHD reported 35% complete responses and 29% partial responses for this approach [50].

ATG is commonly used to treat steroid-refractory GVHD, but its use has been primarily limited by severe life-threatening opportunistic infections [51]. Response rates following ATG are highest in patients with skin involvement and lowest in those with liver involvement [51,52]. A prospective randomized trial comparing ATG and steroids at 5 mg/kg to steroids at 5 mg/kg alone as second-line therapy showed no significant difference between groups [53].

Various monoclonal antibodies to cell surface antigens on GVHD effector cells have also been used as treatment for the disease. There are case reports of resolution of acute GVHD with alemtuzumab [54,55]. Delayed immune reconstitution in these patients results in serious infections, particularly CMV infections [36,37,56]. Visilizumab, a humanized monoclonal anti-CD3 antibody, induces selective apoptosis of activated T cells after binding to CD3 [57,58], as does OKT3 [59]. Both agents induce 30- 50% overall response rates but life-threatening opportunistic infections remain a significant toxicity and no overall survival benefit has been reported.

A monoclonal antibody directed against CD147, a neurothelin expressed on activated T and B lymphocytes, monocytes and dendritic cells, has been tested in steroid-refractory GVHD. A pilot study showed a 41% overall response rate [60] but a phase II/III trial comparing anti-CD147 to ATG in 92 patients with steroid-refractory GVHD did not show any differences in either response or survival [61].

Several anti-IL-2-receptor (CD25) antibodies have been developed and studied extensively: inolimomab, a murine antibody specific for IL-2α receptor [62,63]; basiliximab, a chimeric murine/human monoclonal antibody specific for IL-2Rα [64-66] daclizumab, a humanized monoclonal antibody to the alpha subunit of the IL-2R [67]; and denileukin diftitox, a fusion protein combining sequences from CD25 with sequences from the diphtheria toxin leading to the toxic lysis of activated lymphocytes [68]. These agents have shown efficacy as second-line treatment, but infections remain the most significant problem associated with these drugs [62-67]. Denileukin diftitox, the only agent that is still currently under investigation, produced a 71% response rate for steroid-refractory acute GVHD in a phase I trial [68] but only a 27% response rate at day 100, in a phase II trial [69].

Alefacept is a fusion protein of the first extracellular domains of human LFA-3 and the Fc portion of IgG1 that blocks CD2–LFA-3 costimulation by selective binding to CD2 on memory effector T cells. Alefacept has been shown to be active in T cell mediated autoimmune disease, particularly in psoriasis [70]. Three small clinical studies have demonstrated response rates of more than 70% for steroid refractory GVHD [71-73].

A different strategy to treat GVHD is blockade of the inflammatory cytokine TNFα. Two anti-TNFα monoclonal antibodies have been used: infliximab, a chimeric monoclonal antibody that binds to TNFα and that lyses cells producing TNFα; and etanercept, a recombinant DNA protein composed of TNF receptor II linked to the Fc portion of human IgG1. Infliximab resulted in a 19% complete response rate in 32 patients treated for grade II-IV steroid-refractory acute GVHD in a multicenter, retrospective analysis. These patients experienced high rates of infectious complications as would be expected with exposure to high dose steroids [74]. The combination of etanercept and steroids was recently tested as first-line therapy for patients with grade II-IV acute GVHD in a single-center trial. Complete response rates of approximately 70% were observed four weeks following treatment, although this finding has not yet been reproduced [75*].

Two new anti-TNF antibodies have been approved by the Food and Drug Administration: adalimumab, a fully human monoclonal anti-TNF antibody that has been recently tested in both Crohn's disease [76,77] and juvenile rheumatoid arthritis [78]; and certolizumab pegol, a pegylated humanized Fab' fragment of an anti–TNF-α monoclonal antibody that has also used to treat Crohn's disease [79] and rheumatoid arthritis [80]. The cytotoxic effects of the newer anti-TNF agents are mechanistically different, e.g. infliximab and adalimumab exert complement-dependent cytotoxicity and induce apoptosis in Jurkat T cells that express transmembrane TNFα, while etanercept showed considerably lower levels of both activities [81,82].

Pentostatin is a purine analog that reduces DNA synthesis by inhibiting adenosine deaminase. In a phase I trial, pentostatin led to a 78% overall response rate in steroid-refractory acute GVHD, although infectious complications were frequent [83].

Sirolimus has shown some efficacy in the treatment of both acute and chronic GVHD [84,85] with a twenty-four percent complete response rate observed in patients with steroid-refractory acute GVHD [84].

MMF produced a 65% response rate when it was used with a calcineurin inhibitor and prednisolone to treat steroid-refractory acute GVHD [86]. Several small studies have confirmed these promising results [87-89]. Interests in MMF increased further following the results of a phase II, randomized, four-arm trial of 180 patients with newly diagnosed acute GVHD. Patients were treated with standard corticosteroids plus one of the four new agents: MMF, etanercept, denileukin diftitox and pentostatin. MMF produced the best results in terms of toxicity profile, response, survival, incidence of chronic GVHD, and infections [90**]. A randomized phase III study to validate these finding is currently being planned. Should be noted that in a multicenter randomized trial, the addition of MMF to the initial systemic treatment regimen for chronic GVHD led to no difference in the cumulative incidence of chronic GVHD and resulted in a hazard ratio of death of 1.99 (95% confidence interval, 0.9 - 4.3) compared to the control arm [91].

Extracorporeal photopheresis (ECP) is increasingly used in chronic GVHD and is also used in some centers as an adjunct therapy for acute GVHD in an effort to minimize corticosteroid exposure. This technique exposes peripheral blood mononuclear cells to photo-activated 8 methoxypsoralen (8-MOP) and ultraviolet A radiation, which covalently binds and cross-links DNA, initiating apoptosis [92,93]. Experimental studies suggest ECP increases number of regulatory T cells in both humans and mice [94*,95]. The mouse model is of interest as it showed that the transfer of cells treated with ECP reverses established GVHD by increasing donor regulatory T cells and reduces the number of donor effector lymphocytes even though they had never been directly exposed to 8-MOP and ultraviolet A radiation [94*]. A recent phase II study of 59 patients receiving ECP for treatment of steroid-refractory GVHD demonstrated complete response rates of 60%, with a 59% overall survival at 4 years [96]. Other small studies showed complete response rate of 52% to 83% [97-99]. Response rates may depend on the length of time from diagnosis and the grade of GVHD at the time of treatment [98,99].

The infusions of mesenchymal stem cells (MSC) represent yet another approach for the treatment of acute GVHD. In a phase II study, thirty of 55 patients (55%) with steroids-resistant GVHD showed complete responses to MSC infusions [100*]. These positive results were not reproduced, however, in a second study where only 2 out of 13 patients (15%) with steroid-refractory GVHD responded [101]. Clinical studies have not shown an increased risk of severe infections, and a randomized phase III trial is currently ongoing. MSC have been almost impossible to detect after infusion when administered in vivo, and thus little is known regarding their migration, their mechanism of action, or their persistence. In vitro, MSC suppress the responses of numbers of cell types including T cells, antigen-presenting cells, natural-killer cells, and B cells [102]. Experimental models have suggested a potential risk of malignant transformation [103,104].

The results of selected recent studies of therapies for GVHD are summarized in Table 1. There are currently no criteria to identify patients who are likely to benefit from these second line agents. The diagnosis of acute GVHD is still based on clinical criteria that may be confirmed by biopsy of one of the three target organs (skin, gastrointestinal tract, or liver). The severity of acute GVHD is graded from I to IV using a standardized system that evaluates the involvement of three principal target organs. Thus, new blood tests with predictive value for long-term survival such as the four biomarker panel [105,106*]) may be useful to further identify high-risk groups and their outcomes (Figure 1). Such blood tests could facilitate the decisions to introduce more intensive therapy at an earlier time for patients presenting with a high-risk biomarker panel. It may also guide the speed of steroid taper in patients with low-risk panels. Such biomarkers may also help identify patients who are not likely to respond to traditional treatment and who are at very high risk for subsequent morbidity and mortality. If validated, biomarkers could thus result in immunosuppressive treatment plans tailored to patients in several risk strata.

Table 1. Selected studies of novel agents as secondary or first line therapy in acute GVHD.

Novel Therapy Type of trial Type of treatment Sample size Overall Response at 4 weeks Survival at 3-6 months Study Reference
Denileukin diftitox Phase I steroid refractory 30 71% 33% Ho et al. 68
Denileukin diftitox Phase II steroid refractory 22 41% ND Shaughnessy et al. 69
Denileukin diftitox Phase II randomized Initial therapy 60 60% 49% Alousi et al. 90
Etanercept Phase II randomized Initial therapy 60 48% 56% Alousi et al. 90
Pentostatin Phase I steroid refractory 23 78% ND Bolanos-Meade et al. 83
Pentostatin Phase II randomized Initial therapy 60 62% 52% Alousi et al. 90
MMF Phase I/II steroid refractory 48 72% 78% Basara et al. 87
MMF Phase I/II steroid refractory 10 60% 70% Krejci et al. 88
MMF Phase I/II steroid refractory 6 67% 64% Takami et al. 89
MMF Phase II randomized Initial therapy 60 78% 64% Alousi et al. 90
Sirolimus Phase I/II steroid refractory 21 57% 28% Benito et al. 84
BDP Phase II randomized Initial therapy 129 71% 87% Hockenbery et al. 46
ECP Phase II steroid refractory 59 68% ND Greinix et al. 96
ECP Phase II steroid refractory 23 52% 48% Perfetti et al. 97
ECP Phase II steroid refractory 41 (children) 73% 100% Calore et al. 98
ECP Phase II steroid refractory 15 (children) 0% for grade IV GVHD ND Berger et al. 99
MSC Phase II steroid refractory 55 71% ND Le Blanc et al. 100
MSC Phase II steroid refractory 13 15% 31% von Bonin et al. 101
Alefacept Phase I steroid refractory 16 75% ND Shapira et al. 72

GVHD indicates graft versus host disease, MMF, mycophenolate mofetil, BDP, beclomethasone diproprionate, ECP, extracorporeal photophoresis, MSC, mesenchymal stem cell

Figure 1. From discovery to validation of plasma biomarkers of acute graft versus host disease.

Figure 1

Panel A shows the heatmap of proteins levels measured sequential ELISA in the discovery set samples. Samples from 21 GVHD– patients (left) and 21 GVHD+ patients (right) are represented. Eleven proteins gave a P values for differences between GVHD+ and GVHD– patient plasma< 0.05. Panel B shows the receiver operating characteristic (ROC) curves of four individual discriminator proteins and the composite panel in the training set. Individual ROC curves for IL-2Ra, TNFR1, HGF, and IL-8 and the composite panel (Reproduced with permission [106].)

Future strategies

Novel approaches to prevent or treat GVHD are often based on evidence from experimental models. Recently, several groups have explored the potential of regulatory T cells (Tregs) in such models. In mice, naturally occuring Tregs develop in the thymus and express the Forkhead Box Protein P3 (FOXP3) gene as well as CD4 and CD25 surface markers. These cells have a vital role in preventing autoimmunity and pathology that is inflicted by uncontrolled immune responses [107]. In mouse BMT models, donor derived Tregs can suppress the proliferation of conventional T cells, prevent GVHD and preserve GVL effects [108-113]. Furthermore, viral immunity is preserved in the presence of Tregs after allogeneic HCT [114]. Clinical trials of this promising approach are ongoing, but isolation and expansion of human T regs is challenging and labor-intensive [115-117]. A recent study demonstrated that combined CD4+ donor lymphocyte infusion and low-dose recombinant IL-2 expands Tregs in vivo following allogeneic HCT [118].

The role of IL-17-producing CD4 T cells (Th17) in GVHD is controversial. Different groups have shown that Th17 cells reduce GVHD [119,120], but their presence does not appear to impact overall survival [120]. In addition, the differentiation of Th17 cells in vitro appears to cause lethal acute GVHD with severe cutaneous and pulmonary damage [121]. These differences may be due to variations among the models, and further analyses are required before Th17 cells are translated from the bench to the bedside.

T cells in murine models can be divided into naïve (CD62L+ CD44-), central memory (CD62L+ CD44+) and terminally differentiated effector/effector memory (CD62L- CD44-). Donor effector memory T cells (TEMs) do not cause GVHD [122-124] but they transfer functional memory [122] and mediate GVL [125] in murine studies. For human T lymphocytes, the markers that best distinguish these subsets are CD45RA and CCR7. In both CD4+ and CD8+ T cell subsets, a progression from naïve (CD45RA+ CCR7+) to central memory (CD45RA-CCR7+) to effector/memory (CD45RA- CCR7-) has been established [126,127]. Although the loss of CD62L expression also can divide memory T cell subsets, the loss of CCR7 appears to be more complete, particularly in the CD4+ subset [126]. Such T cell subpopulations may have relevance to clinical GVHD. High proportions of CD4+ and CCR7+ cells in the donor graft correlate with the incidence and severity of GVHD [128] and reference values for the CD4+ and CD8+ naïve and memory subsets within allografts have been established [129]. If validated, the selection and infusion of human TEMs could become a new approach to modulate GVHD.

Modulation of dendritic cells (DCs) may influence GVHD. A recent murine study demonstrated that exposure to granulocyte colony stimulating factor (G-CSF) shortly after HCT, in addition to a conditioning regimen with total body irradiation (TBI), significantly worsened GVHD. The mechanism underlining this effect is the stimulation of host dendritic cells (DCs) that subsequently activate donor natural killer T cells [130]. These data might explain the increased incidence of GVHD found in recipients receiving prophylactic G-CSF in the European BMT registry [131].

The properties of myeloid DCs can be modulated by several agents in order to render them tolerogenic. Histone deacetylase inhibitors such as suberonylanilide hydroxamic acid (SAHA) have been shown to reduce development of GVHD in murine models by modulating indoleamine 2,3-dioxygenase-dependent DCs functions [132*-134]. SAHA is currently in clinical trials to prevent GVHD. Another therapeutic approach to modulate DCs could be an antibody to the DC surface maturation antigen CD83; this antibody has been shown to prevent acute GVHD in a humanized mouse model with severe congenital immunodeficiency [135].

As noted earlier, DCs are important initiators of GVHD. Plasmacytoid dendritic cells (pDCs) have gained recent interest because of their potential tolerogenic effect in an immature state. In one study a subset of host pDCs that express CCR9 receptor has been shown to suppress GVHD [136]. However, in another study, host pDCs have been shown to induce GVHD in manner independent of Toll like receptors [137]. Thus the role of pDCs in acute GVHD is still controversial and confirmatory studies about their functions will be required.

The role of B cells in acute GVHD is currently under investigation, particularly regarding their possible role in attenuating the disease. In a study of 254 HCT recipients from sibling donors, the number of donor B cells in the graft inversely correlated with the cumulative incidence of grade II-IV acute GVHD [138]. A retrospective study of several hundred B-cell lymphoma patients showed that those receiving rituximab, a chimeric monoclonal CD20 antibody, within the 6 months prior to allogeneic HCT had significantly lower TRM (relative risk (RR) = 0.68) and lower incidence of grade II-IV acute GVHD (RR = 0.72) compared to controls [139]. The authors speculated that recipient B cells might function as semi-professional APCs to stimulate donor T cells. Mouse studies show that host B cells produce IL-10 following TBI and attenuate acute GVHD after allogeneic BMT [140].

Conclusions

The number of patients at high risk for GVHD is increasing, particularly as more allogeneic HCT are performed from unrelated donors and older patients. Our understanding of the complex pathophysiology of this disorder continues to increase, but few of these insights have thus far led to major therapeutic advances. Deleterious side effects of infectious complications and relapse of underlying malignancy remain barriers to successful new approaches. Several lines of investigation with cellular therapeutics and immunomodulating agents are currently in Phase II clinical trials. These promising therapies are most likely to achieve success when they are used early in the disease process, either as primary treatment or as prophylaxis.

References and recommended reading

Papers of particular interest, published within the annual period of review, have been highlighted as:

* of special interest

** of outstanding interest

  • 1**.Ferrara JL, Levine JE, Reddy P, Holler E. Graft-versus-host disease. Lancet. 2009 doi: 10.1016/S0140-6736(09)60237-3. A recent comprehensive review of acute and chronic GVHD pathophysiology, clinical features and treatments options. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2*.Shlomchik WD. Graft-versus-host disease. Nat Rev Immunol. 2007;7:340–352. doi: 10.1038/nri2000. A comprehensive review of the immune response underlying acute GVHD. [DOI] [PubMed] [Google Scholar]
  • 3.Storb R, Deeg HJ, Whitehead J, Appelbaum F, Beatty P, Bensinger W, Buckner CD, Clift R, Doney K, Farewell V, et al. Methotrexate and cyclosporine compared with cyclosporine alone for prophylaxis of acute graft versus host disease after marrow transplantation for leukemia. N Engl J Med. 1986;314:729–735. doi: 10.1056/NEJM198603203141201. [DOI] [PubMed] [Google Scholar]
  • 4.Hiraoka A, Ohashi Y, Okamoto S, Moriyama Y, Nagao T, Kodera Y, Kanamaru A, Dohy H, Masaoka T. Phase III study comparing tacrolimus (FK506) with cyclosporine for graft-versus-host disease prophylaxis after allogeneic bone marrow transplantation. Bone Marrow Transplant. 2001;28:181–185. doi: 10.1038/sj.bmt.1703097. [DOI] [PubMed] [Google Scholar]
  • 5.Horowitz MM, Przepiorka D, Bartels P, Buell DN, Zhang MJ, Fitzsimmons WE, Erdman J, Huang C, Hodosh E, Maher R, et al. Tacrolimus vs. cyclosporine immunosuppression: results in advanced-stage disease compared with historical controls treated exclusively with cyclosporine. Biol Blood Marrow Transplant. 1999;5:180–186. doi: 10.1053/bbmt.1999.v5.pm10392964. [DOI] [PubMed] [Google Scholar]
  • 6.Ratanatharathorn V, Nash RA, Przepiorka D, Devine SM, Klein JL, Weisdorf D, Fay JW, Nademanee A, Antin JH, Christiansen NP, 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;92:2303–2314. [PubMed] [Google Scholar]
  • 7*.Ram R, Gafter-Gvili A, Yeshurun M, Paul M, Raanani P, Shpilberg O. Prophylaxis regimens for GVHD: systematic review and meta-analysis. Bone Marrow Transplant. 2008 doi: 10.1038/bmt.2008.373. A meta-analysis of prophylaxis regimens for GVHD showing that both MTX- cyclosporine and MTX-tacrolimus are good acute GVHD prophylaxis treatment, although MTX-tacrolimus may be superior in terms of acute GVHD reduction. [DOI] [PubMed] [Google Scholar]
  • 8.Nash RA, Antin JH, Karanes C, Fay JW, Avalos BR, Yeager AM, Przepiorka D, Davies S, Petersen FB, Bartels P, et al. Phase 3 study comparing methotrexate and tacrolimus with methotrexate and cyclosporine for prophylaxis of acute graft-versus-host disease after marrow transplantation from unrelated donors. Blood. 2000;96:2062–2068. [PubMed] [Google Scholar]
  • 9.Baan CC, van der Mast BJ, Klepper M, Mol WM, Peeters AM, Korevaar SS, Balk AH, Weimar W. Differential effect of calcineurin inhibitors, anti-CD25 antibodies and rapamycin on the induction of FOXP3 in human T cells. Transplantation. 2005;80:110–117. doi: 10.1097/01.tp.0000164142.98167.4b. [DOI] [PubMed] [Google Scholar]
  • 10.Battaglia M, Stabilini A, Roncarolo MG. Rapamycin selectively expands CD4+CD25+FoxP3+ regulatory T cells. Blood. 2005;105:4743–4748. doi: 10.1182/blood-2004-10-3932. [DOI] [PubMed] [Google Scholar]
  • 11.Hackstein H, Taner T, Logar AJ, Thomson AW. Rapamycin inhibits macropinocytosis and mannose receptor-mediated endocytosis by bone marrow-derived dendritic cells. Blood. 2002;100:1084–1087. doi: 10.1182/blood.v100.3.1084. [DOI] [PubMed] [Google Scholar]
  • 12.Monti P, Mercalli A, Leone BE, Valerio DC, Allavena P, Piemonti L. Rapamycin impairs antigen uptake of human dendritic cells. Transplantation. 2003;75:137–145. doi: 10.1097/00007890-200301150-00025. [DOI] [PubMed] [Google Scholar]
  • 13.Hackstein H, Taner T, Zahorchak AF, Morelli AE, Logar AJ, Gessner A, Thomson AW. Rapamycin inhibits IL-4--induced dendritic cell maturation in vitro and dendritic cell mobilization and function in vivo. Blood. 2003;101:4457–4463. doi: 10.1182/blood-2002-11-3370. [DOI] [PubMed] [Google Scholar]
  • 14.Chiang PH, Wang L, Liang Y, Liang X, Qian S, Fung JJ, Bonham CA, Lu L. Inhibition of IL-12 signaling Stat4/IFN-gamma pathway by rapamycin is associated with impaired dendritic [correction of dendritc] cell function. Transplant Proc. 2002;34:1394–1395. doi: 10.1016/s0041-1345(02)02900-7. [DOI] [PubMed] [Google Scholar]
  • 15.Antin JH, Kim HT, Cutler C, Ho VT, Lee SJ, Miklos DB, Hochberg EP, Wu CJ, Alyea EP, Soiffer RJ. Sirolimus, tacrolimus, and low-dose methotrexate for graft-versus-host disease prophylaxis in mismatched related donor or unrelated donor transplantation. Blood. 2003;102:1601–1605. doi: 10.1182/blood-2003-02-0489. [DOI] [PubMed] [Google Scholar]
  • 16.Cutler C, Li S, Ho VT, Koreth J, Alyea E, Soiffer RJ, Antin JH. Extended follow-up of methotrexate-free immunosuppression using sirolimus and tacrolimus in related and unrelated donor peripheral blood stem cell transplantation. Blood. 2007;109:3108–3114. doi: 10.1182/blood-2006-09-046219. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Platzbecker U, von Bonin M, Goekkurt E, Radke J, Binder M, Kiani A, Stoehlmacher J, Schetelig J, Thiede C, Ehninger G, 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;15:101–108. doi: 10.1016/j.bbmt.2008.11.004. [DOI] [PubMed] [Google Scholar]
  • 18.Bornhauser M, Schuler U, Porksen G, Naumann R, Geissler G, Thiede C, Schwerdtfeger R, Ehninger G, Thiede HM. Mycophenolate mofetil and cyclosporine as graft-versus-host disease prophylaxis after allogeneic blood stem cell transplantation. Transplantation. 1999;67:499–504. doi: 10.1097/00007890-199902270-00001. [DOI] [PubMed] [Google Scholar]
  • 19.Kasper C, Sayer HG, Mugge LO, Schilling K, Scholl S, Issa MC, Hoffken K. Combined standard graft-versus-host disease (GvHD) prophylaxis with mycophenolate mofetil (MMF) in allogeneic peripheral blood stem cell transplantation from unrelated donors. Bone Marrow Transplant. 2004;33:65–69. doi: 10.1038/sj.bmt.1704299. [DOI] [PubMed] [Google Scholar]
  • 20.Mohty M, de Lavallade H, Faucher C, Bilger K, Vey N, Stoppa AM, Gravis G, Coso D, Viens P, Gastaut JA, et al. Mycophenolate mofetil and cyclosporine for graft-versus-host disease prophylaxis following reduced intensity conditioning allogeneic stem cell transplantation. Bone Marrow Transplant. 2004;34:527–530. doi: 10.1038/sj.bmt.1704640. [DOI] [PubMed] [Google Scholar]
  • 21.Vogelsang GB, Arai S. Mycophenolate mofetil for the prevention and treatment of graft-versus-host disease following stem cell transplantation: preliminary findings. Bone Marrow Transplant. 2001;27:1255–1262. doi: 10.1038/sj.bmt.1703076. [DOI] [PubMed] [Google Scholar]
  • 22.Niederwieser D, Maris M, Shizuru JA, Petersdorf E, Hegenbart U, Sandmaier BM, Maloney DG, Storer B, Lange T, Chauncey T, et al. Low-dose total body irradiation (TBI) and fludarabine followed by hematopoietic cell transplantation (HCT) from HLA-matched or mismatched unrelated donors and postgrafting immunosuppression with cyclosporine and mycophenolate mofetil (MMF) can induce durable complete chimerism and sustained remissions in patients with hematological diseases. Blood. 2003;101:1620–1629. doi: 10.1182/blood-2002-05-1340. [DOI] [PubMed] [Google Scholar]
  • 23*.Hahn T, McCarthy PL, Jr, Zhang MJ, Wang D, Arora M, Frangoul H, Gale RP, Hale GA, Horan J, Isola L, et al. Risk factors for acute graft-versus-host disease after human leukocyte antigen-identical sibling transplants for adults with leukemia. J Clin Oncol. 2008;26:5728–5734. doi: 10.1200/JCO.2008.17.6545. A study of worldwide HCT registry from 1995 to 2002 reported risk factors for grade II-IV acute GVHD in 1,960 adults after HLA-identical sibling myeloablative transplant for acute myeloid leukemia (AML), acute lymphoid leukemia (ALL) and chronic myeloid leukemia (CML) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.O'Reilly RJ. T-cell depletion and allogeneic bone marrow transplantation. Semin Hematol. 1992;29:20–26. [PubMed] [Google Scholar]
  • 25.Bacigalupo A, Lamparelli T, Bruzzi P, Guidi S, Alessandrino PE, di Bartolomeo P, Oneto R, Bruno B, Barbanti M, Sacchi N, et al. Antithymocyte globulin for graft-versus-host disease prophylaxis in transplants from unrelated donors: 2 randomized studies from Gruppo Italiano Trapianti Midollo Osseo (GITMO) Blood. 2001;98:2942–2947. doi: 10.1182/blood.v98.10.2942. [DOI] [PubMed] [Google Scholar]
  • 26.Byrne JL, Stainer C, Cull G, Haynes AP, Bessell EM, Hale G, Waldmann H, Russell NH. The effect of the serotherapy regimen used and the marrow cell dose received on rejection, graft-versus-host disease and outcome following unrelated donor bone marrow transplantation for leukaemia. Bone Marrow Transplant. 2000;25:411–417. doi: 10.1038/sj.bmt.1702165. [DOI] [PubMed] [Google Scholar]
  • 27.Finke J, Bertz H, Schmoor C, Veelken H, Behringer D, Wasch R, Kunzmann R, Heidecker L, Lang H, Meyer-Konig U, et al. Allogeneic bone marrow transplantation from unrelated donors using in vivo anti-T-cell globulin. Br J Haematol. 2000;111:303–313. doi: 10.1046/j.1365-2141.2000.02305.x. [DOI] [PubMed] [Google Scholar]
  • 28.Hansen JA, Gooley TA, Martin PJ, Appelbaum F, Chauncey TR, Clift RA, Petersdorf EW, Radich J, Sanders JE, Storb RF, et al. Bone marrow transplants from unrelated donors for patients with chronic myeloid leukemia. N Engl J Med. 1998;338:962–968. doi: 10.1056/NEJM199804023381405. [DOI] [PubMed] [Google Scholar]
  • 29.Hows J, Bradley BA, Gore S, Downie T, Howard M, Gluckman E. Prospective evaluation of unrelated donor bone marrow transplantation. The International Marrow Unrelated Search and Transplant (IMUST) Study. Bone Marrow Transplant. 1993;12:371–380. [PubMed] [Google Scholar]
  • 30.Barge RM, Starrenburg CW, Falkenburg JH, Fibbe WE, Marijt EW, Willemze R. Long-term follow-up of myeloablative allogeneic stem cell transplantation using Campath “in the bag” as T-cell depletion: the Leiden experience. Bone Marrow Transplant. 2006;37:1129–1134. doi: 10.1038/sj.bmt.1705385. [DOI] [PubMed] [Google Scholar]
  • 31.Delgado J, Pillai S, Benjamin R, Caballero D, Martino R, Nathwani A, Lovell R, Thomson K, Perez-Simon JA, Sureda A, et al. The effect of in vivo T cell depletion with alemtuzumab on reduced-intensity allogeneic hematopoietic cell transplantation for chronic lymphocytic leukemia. Biol Blood Marrow Transplant. 2008;14:1288–1297. doi: 10.1016/j.bbmt.2008.09.001. [DOI] [PubMed] [Google Scholar]
  • 32.Kennedy-Nasser AA, Bollard CM, Myers GD, Leung KS, Gottschalk S, Zhang Y, Liu H, Heslop HE, Brenner MK, Krance RA. Comparable outcome of alternative donor and matched sibling donor hematopoietic stem cell transplant for children with acute lymphoblastic leukemia in first or second remission using alemtuzumab in a myeloablative conditioning regimen. Biol Blood Marrow Transplant. 2008;14:1245–1252. doi: 10.1016/j.bbmt.2008.08.010. [DOI] [PubMed] [Google Scholar]
  • 33.Shore T, Harpel J, Schuster MW, Roboz GJ, Leonard JP, Coleman M, Feldman EJ, Silver RT. A study of a reduced-intensity conditioning regimen followed by allogeneic stem cell transplantation for patients with hematologic malignancies using Campath-1H as part of a graft-versus-host disease strategy. Biol Blood Marrow Transplant. 2006;12:868–875. doi: 10.1016/j.bbmt.2006.05.006. [DOI] [PubMed] [Google Scholar]
  • 34.Martinez C, Solano C, Ferra C, Sampol A, Valcarcel D, Perez-Simon JA. Alemtuzumab as treatment of steroid-refractory acute graft-versus-host disease: results of a phase II study. Biol Blood Marrow Transplant. 2009;15:639–642. doi: 10.1016/j.bbmt.2009.01.014. [DOI] [PubMed] [Google Scholar]
  • 35.Kroger N, Shaw B, Iacobelli S, Zabelina T, Peggs K, Shimoni A, Nagler A, Binder T, Eiermann T, Madrigal A, et al. Comparison between antithymocyte globulin and alemtuzumab and the possible impact of KIR-ligand mismatch after dose-reduced conditioning and unrelated stem cell transplantation in patients with multiple myeloma. Br J Haematol. 2005;129:631–643. doi: 10.1111/j.1365-2141.2005.05513.x. [DOI] [PubMed] [Google Scholar]
  • 36.Chakrabarti S, Mackinnon S, Chopra R, Kottaridis PD, Peggs K, O'Gorman P, Chakraverty R, Marshall T, Osman H, Mahendra P, et al. High incidence of cytomegalovirus infection after nonmyeloablative stem cell transplantation: potential role of Campath-1H in delaying immune reconstitution. Blood. 2002;99:4357–4363. doi: 10.1182/blood.v99.12.4357. [DOI] [PubMed] [Google Scholar]
  • 37.Chakrabarti S, Mautner V, Osman H, Collingham KE, Fegan CD, Klapper PE, Moss PA, Milligan DW. Adenovirus infections following allogeneic stem cell transplantation: incidence and outcome in relation to graft manipulation, immunosuppression, and immune recovery. Blood. 2002;100:1619–1627. doi: 10.1182/blood-2002-02-0377. [DOI] [PubMed] [Google Scholar]
  • 38.Przepiorka D, Weisdorf D, Martin P, Klingemann HG, Beatty P, Hows J, Thomas ED. 1994 Consensus Conference on Acute GVHD Grading. Bone Marrow Transplant. 1995;15:825–828. [PubMed] [Google Scholar]
  • 39.Chao NJ, Chen BJ. Prophylaxis and treatment of acute graft-versus-host disease. Semin Hematol. 2006;43:32–41. doi: 10.1053/j.seminhematol.2005.09.007. [DOI] [PubMed] [Google Scholar]
  • 40.MacMillan ML, Weisdorf DJ, Wagner JE, DeFor TE, Burns LJ, Ramsay NK, Davies SM, Blazar BR. Response of 443 patients to steroids as primary therapy for acute graft-versus-host disease: comparison of grading systems. Biol Blood Marrow Transplant. 2002;8:387–394. doi: 10.1053/bbmt.2002.v8.pm12171485. [DOI] [PubMed] [Google Scholar]
  • 41.Martin PJ, Schoch G, Fisher L, Byers V, Anasetti C, Appelbaum FR, Beatty PG, Doney K, McDonald GB, Sanders JE, et al. A retrospective analysis of therapy for acute graft-versus-host disease: initial treatment. Blood. 1990;76:1464–1472. [PubMed] [Google Scholar]
  • 42.Weisdorf D, Haake R, Blazar B, Miller W, McGlave P, Ramsay N, Kersey J, Filipovich A. Treatment of moderate/severe acute graft-versus-host disease after allogeneic bone marrow transplantation: an analysis of clinical risk features and outcome. Blood. 1990;75:1024–1030. [PubMed] [Google Scholar]
  • 43.Van Lint MT, Uderzo C, Locasciulli A, Majolino I, Scime R, Locatelli F, Giorgiani G, Arcese W, Iori AP, Falda M, et al. Early treatment of acute graft-versus-host disease with high- or low-dose 6-methylprednisolone: a multicenter randomized trial from the Italian Group for Bone Marrow Transplantation. Blood. 1998;92:2288–2293. [PubMed] [Google Scholar]
  • 44*.Mielcarek M, Storer BE, Boeckh MJ, Carpenter PA, McDonald GB, Deeg HJ, Nash RA, Flowers ME, Doney K, Lee S, et al. Initial therapy of acute graft-versus-host disease with low-dose. prednisone does not compromise patient outcomes. Blood. 2008 doi: 10.1182/blood-2008-07-168401. A retrospective study showing that 1 mg/kg/day prednisone as initial therapy give similar overall survival as compared to 2 mg/kg/day prednisone. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45*.Ibrahim RB, Abidi MH, Cronin SM, Lum LG, Al-Kadhimi Z, Ratanatharathorn V, Uberti JP. Nonabsorbable corticosteroids use in the treatment of gastrointestinal graft-versus-host disease. Biol Blood Marrow Transplant. 2009;15:395–405. doi: 10.1016/j.bbmt.2008.12.487. A review of local corticosteroids used in the treatment of gastrointestinal GVHD. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Bertz H, Afting M, Kreisel W, Duffner U, Greinwald R, Finke J. Feasibility and response to budesonide as topical corticosteroid therapy for acute intestinal GVHD. Bone Marrow Transplant. 1999;24:1185–1189. doi: 10.1038/sj.bmt.1702055. [DOI] [PubMed] [Google Scholar]
  • 47.Hockenbery DM, Cruickshank S, Rodell TC, Gooley T, Schuening F, Rowley S, David D, Brunvand M, Berryman B, Abhyankar S, et al. A randomized, placebo-controlled trial of oral beclomethasone dipropionate as a prednisone-sparing therapy for gastrointestinal graft-versus-host disease. Blood. 2007;109:4557–4563. doi: 10.1182/blood-2006-05-021139. [DOI] [PubMed] [Google Scholar]
  • 48.Shapira MY, Bloom AI, Or R, Sasson T, Nagler A, Resnick IB, Aker M, Zilberman I, Slavin S, Verstanding A. Intra-arterial catheter directed therapy for severe graft-versus-host disease. Br J Haematol. 2002;119:760–764. doi: 10.1046/j.1365-2141.2002.03923.x. [DOI] [PubMed] [Google Scholar]
  • 49.Tfayli A, Selby G, Maqbool F, Bierbaum W, Hamadani M. Role of intra-arterial steroid administration in the management of steroid-refractory acute gastrointestinal graft-versus-host disease. Am J Hematol. 2006;81:959–962. doi: 10.1002/ajh.20711. [DOI] [PubMed] [Google Scholar]
  • 50.Stevens A, Hu C, Isola L, Scigliano E, Grosskreutz C, Osman K, Malone A, Del Toro G, Weintraub Intra-arterial steroids for systemic steroid resistant graft-vs-host-disease. BBMT. 2009;(15):127–128. [Google Scholar]
  • 51.Arai S, Margolis J, Zahurak M, Anders V, Vogelsang GB. Poor outcome in steroid-refractory graft-versus-host disease with antithymocyte globulin treatment. Biol Blood Marrow Transplant. 2002;8:155–160. doi: 10.1053/bbmt.2002.v8.pm11939605. [DOI] [PubMed] [Google Scholar]
  • 52.McCaul KG, Nevill TJ, Barnett MJ, Toze CL, Currie CJ, Sutherland HJ, Conneally EA, Shepherd JD, Nantel SH, Hogge DE, et al. Treatment of steroid-resistant acute graft-versus-host disease with rabbit antithymocyte globulin. J Hematother Stem Cell Res. 2000;9:367–374. doi: 10.1089/15258160050079470. [DOI] [PubMed] [Google Scholar]
  • 53.Van Lint MT, Milone G, Leotta S, Uderzo C, Scime R, Dallorso S, Locasciulli A, Guidi S, Mordini N, Sica S, et al. Treatment of acute graft-versus-host disease with prednisolone: significant survival advantage for day +5 responders and no advantage for nonresponders receiving anti-thymocyte globulin. Blood. 2006;107:4177–4181. doi: 10.1182/blood-2005-12-4851. [DOI] [PubMed] [Google Scholar]
  • 54.Carella AM, Beltrami G, Scalzulli PR, Carella AM, Jr, Corsetti MT. Alemtuzumab can successfully treat steroid-refractory acute graft-versus-host disease (aGVHD) Bone Marrow Transplant. 2004;33:131–132. doi: 10.1038/sj.bmt.1704322. [DOI] [PubMed] [Google Scholar]
  • 55.Wandroo F, Auguston B, Cook M, Craddock C, Mahendra P. Successful use of Campath-1H in the treatment of steroid refractory liver GvHD. Bone Marrow Transplant. 2004;34:285–287. doi: 10.1038/sj.bmt.1704573. [DOI] [PubMed] [Google Scholar]
  • 56.Chae YS, Sohn SK, Kim JG, Cho YY, Moon JH, Yang DH, Lee JJ, Kim YK, Kim HJ, Shin HJ, et al. Impact of alemtuzumab as conditioning regimen component on transplantation outcomes in case of CMV-seropositive recipients and donors. Am J Hematol. 2008;83:649–653. doi: 10.1002/ajh.21215. [DOI] [PubMed] [Google Scholar]
  • 57.Carpenter PA, Appelbaum FR, Corey L, Deeg HJ, Doney K, Gooley T, Krueger J, Martin P, Pavlovic S, Sanders J, et al. A humanized non-FcR-binding anti-CD3 antibody, visilizumab, for treatment of steroid-refractory acute graft-versus-host disease. Blood. 2002;99:2712–2719. doi: 10.1182/blood.v99.8.2712. [DOI] [PubMed] [Google Scholar]
  • 58.Carpenter PA, Lowder J, Johnston L, Frangoul H, Khoury H, Parker P, Jerome KR, McCune JS, Storer B, Martin P, et al. A phase II multicenter study of visilizumab, humanized anti-CD3 antibody, to treat steroid-refractory acute graft-versus-host disease. Biol Blood Marrow Transplant. 2005;11:465–471. doi: 10.1016/j.bbmt.2005.03.002. [DOI] [PubMed] [Google Scholar]
  • 59.Knop S, Hebart H, Gscheidle H, Holler E, Kolb HJ, Niederwieser D, Einsele H. OKT3 muromonab as second-line and subsequent treatment in recipients of stem cell allografts with steroid-resistant acute graft-versus-host disease. Bone Marrow Transplant. 2005;36:831–837. doi: 10.1038/sj.bmt.1705132. [DOI] [PubMed] [Google Scholar]
  • 60.Deeg HJ, Blazar BR, Bolwell BJ, Long GD, Schuening F, Cunningham J, Rifkin RM, Abhyankar S, Briggs AD, Burt R, et al. Treatment of steroid-refractory acute graft-versus-host disease with anti-CD147 monoclonal antibody ABX-CBL. Blood. 2001;98:2052–2058. doi: 10.1182/blood.v98.7.2052. [DOI] [PubMed] [Google Scholar]
  • 61.Macmillan ML, Couriel D, Weisdorf DJ, Schwab G, Havrilla N, Fleming TR, Huang S, Roskos L, Slavin S, Shadduck RK, et al. A phase 2/3 multicenter randomized clinical trial of ABX-CBL versus ATG as secondary therapy for steroid-resistant acute graft-versus-host disease. Blood. 2007;109:2657–2662. doi: 10.1182/blood-2006-08-013995. [DOI] [PubMed] [Google Scholar]
  • 62.Bay JO, Dhedin N, Goerner M, Vannier JP, Marie-Cardine A, Stamatoullas A, Jouet JP, Yakoub-Agha I, Tabrizi R, Faucher C, et al. Inolimomab in steroid-refractory acute graft-versus-host disease following allogeneic hematopoietic stem cell transplantation: retrospective analysis and comparison with other interleukin-2 receptor antibodies. Transplantation. 2005;80:782–788. doi: 10.1097/01.tp.0000173995.18826.de. [DOI] [PubMed] [Google Scholar]
  • 63.Pinana JL, Valcarcel D, Martino R, Moreno ME, Sureda A, Briones J, Brunet S, Sierra J. Encouraging results with inolimomab (anti-IL-2 receptor) as treatment for refractory acute graft-versus-host disease. Biol Blood Marrow Transplant. 2006;12:1135–1141. doi: 10.1016/j.bbmt.2006.06.010. [DOI] [PubMed] [Google Scholar]
  • 64.Funke VA, de Medeiros CR, Setubal DC, Ruiz J, Bitencourt MA, Bonfim CM, Neto JZ, Pasquini R. Therapy for severe refractory acute graft-versus-host disease with basiliximab, a selective interleukin-2 receptor antagonist. Bone Marrow Transplant. 2006;37:961–965. doi: 10.1038/sj.bmt.1705306. [DOI] [PubMed] [Google Scholar]
  • 65.Massenkeil G, Rackwitz S, Genvresse I, Rosen O, Dorken B, Arnold R. Basiliximab is well tolerated and effective in the treatment of steroid-refractory acute graft-versus-host disease after allogeneic stem cell transplantation. Bone Marrow Transplant. 2002;30:899–903. doi: 10.1038/sj.bmt.1703737. [DOI] [PubMed] [Google Scholar]
  • 66.Schmidt-Hieber M, Fietz T, Knauf W, Uharek L, Hopfenmuller W, Thiel E, Blau IW. Efficacy of the interleukin-2 receptor antagonist basiliximab in steroid-refractory acute graft-versus-host disease. Br J Haematol. 2005;130:568–574. doi: 10.1111/j.1365-2141.2005.05631.x. [DOI] [PubMed] [Google Scholar]
  • 67.Przepiorka D, Kernan NA, Ippoliti C, Papadopoulos EB, Giralt S, Khouri I, Lu JG, Gajewski J, Durett A, Cleary K, et al. Daclizumab, a humanized anti-interleukin-2 receptor alpha chain antibody, for treatment of acute graft-versus-host disease. Blood. 2000;95:83–89. [PubMed] [Google Scholar]
  • 68.Ho VT, Zahrieh D, Hochberg E, Micale E, Levin J, Reynolds C, Steckel S, Cutler C, Fisher DC, Lee SJ, et al. Safety and efficacy of denileukin diftitox in patients with steroid-refractory acute graft-versus-host disease after allogeneic hematopoietic stem cell transplantation. Blood. 2004;104:1224–1226. doi: 10.1182/blood-2004-01-0028. [DOI] [PubMed] [Google Scholar]
  • 69.Shaughnessy PJ, Bachier C, Grimley M, Freytes CO, Callander NS, Essell JH, Flomenberg N, Selby G, Lemaistre CF. Denileukin diftitox for the treatment of steroid-resistant acute graft-versus-host disease. Biol Blood Marrow Transplant. 2005;11:188–193. doi: 10.1016/j.bbmt.2004.11.022. [DOI] [PubMed] [Google Scholar]
  • 70.Ellis CN, Krueger GG. Treatment of chronic plaque psoriasis by selective targeting of memory effector T lymphocytes. N Engl J Med. 2001;345:248–255. doi: 10.1056/NEJM200107263450403. [DOI] [PubMed] [Google Scholar]
  • 71.Shapira MY, Resnick IB, Dray L, Aker M, Stepensky P, Elad S, Gesundeheidt B, Samuel S, Or R. A new induction protocol for the control of steroid refractory/dependent acute graft versus host disease with alefacept and tacrolimus. Cytotherapy. 2009;11:61–67. doi: 10.1080/14653240802644669. [DOI] [PubMed] [Google Scholar]
  • 72.Shapira MY, Resnick IB, Bitan M, Ackerstein A, Tsirigotis P, Gesundheit B, Zilberman I, Miron S, Leubovic A, Slavin S, et al. Rapid response to alefacept given to patients with steroid resistant or steroid dependent acute graft-versus-host disease: a preliminary report. Bone Marrow Transplant. 2005;36:1097–1101. doi: 10.1038/sj.bmt.1705185. [DOI] [PubMed] [Google Scholar]
  • 73.Toor AA, Stiff PJ, Nickoloff BJ, Rodriguez T, Klein JL, Gordon KB. Alefacept in corticosteroid refractory graft versus host disease: early results indicate promising activity. J Dermatolog Treat. 2007;18:13–18. doi: 10.1080/09546630601121045. [DOI] [PubMed] [Google Scholar]
  • 74.Patriarca F, Sperotto A, Damiani D, Morreale G, Bonifazi F, Olivieri A, Ciceri F, Milone G, Cesaro S, Bandini G, et al. Infliximab treatment for steroid-refractory acute graft-versus-host disease. Haematologica. 2004;89:1352–1359. [PubMed] [Google Scholar]
  • 75*.Levine JE, Paczesny S, Mineishi S, Braun T, Choi SW, Hutchinson RJ, Jones D, Khaled Y, Kitko CL, Bickley D, et al. Etanercept plus methylprednisolone as initial therapy for acute graft-versus-host disease. Blood. 2008;111:2470–2475. doi: 10.1182/blood-2007-09-112987. A single center phase II trial showing the efficacy of etanercept plus methylprednisolone as initial therapy for acute graft-versus-host disease. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Papadakis KA, Shaye OA, Vasiliauskas EA, Ippoliti A, Dubinsky MC, Birt J, Paavola J, Lee SK, Price J, Targan SR, et al. Safety and efficacy of adalimumab (D2E7) in Crohn's disease patients with an attenuated response to infliximab. Am J Gastroenterol. 2005;100:75–79. doi: 10.1111/j.1572-0241.2005.40647.x. [DOI] [PubMed] [Google Scholar]
  • 77.Sandborn WJ, Feagan BG, Radford-Smith G, Kovacs A, Enns R, Innes A, Patel J. CDP571, a humanised monoclonal antibody to tumour necrosis factor alpha, for moderate to severe Crohn's disease: a randomised, double blind, placebo controlled trial. Gut. 2004;53:1485–1493. doi: 10.1136/gut.2003.035253. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Taddio A, Marchetti F. Adalimumab in juvenile rheumatoid arthritis. N Engl J Med. 2008;359:2495–2496. author reply 2496-2497. [PubMed] [Google Scholar]
  • 79.Sandborn WJ, Feagan BG, Stoinov S, Honiball PJ, Rutgeerts P, Mason D, Bloomfield R, Schreiber S. Certolizumab pegol for the treatment of Crohn's disease. N Engl J Med. 2007;357:228–238. doi: 10.1056/NEJMoa067594. [DOI] [PubMed] [Google Scholar]
  • 80.Keystone E, Heijde D, Mason D, Jr, Landewe R, Vollenhoven RV, Combe B, Emery P, Strand V, Mease P, Desai C, et al. Certolizumab pegol plus methotrexate is significantly more effective than placebo plus methotrexate in active rheumatoid arthritis: findings of a fifty-two-week, phase III, multicenter, randomized, double-blind, placebo-controlled, parallel-group study. Arthritis Rheum. 2008;58:3319–3329. doi: 10.1002/art.23964. [DOI] [PubMed] [Google Scholar]
  • 81.Mitoma H, Horiuchi T, Tsukamoto H, Tamimoto Y, Kimoto Y, Uchino A, To K, Harashima S, Hatta N, Harada M. Mechanisms for cytotoxic effects of anti-tumor necrosis factor agents on transmembrane tumor necrosis factor alpha-expressing cells: comparison among infliximab, etanercept, and adalimumab. Arthritis Rheum. 2008;58:1248–1257. doi: 10.1002/art.23447. [DOI] [PubMed] [Google Scholar]
  • 82.Wong M, Ziring D, Korin Y, Desai S, Kim S, Lin J, Gjertson D, Braun J, Reed E, Singh RR. TNFalpha blockade in human diseases: mechanisms and future directions. Clin Immunol. 2008;126:121–136. doi: 10.1016/j.clim.2007.08.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Bolanos-Meade J, Jacobsohn DA, Margolis J, Ogden A, Wientjes MG, Byrd JC, Lucas DM, Anders V, Phelps M, Grever MR, et al. Pentostatin in steroid-refractory acute graft-versus-host disease. J Clin Oncol. 2005;23:2661–2668. doi: 10.1200/JCO.2005.06.130. [DOI] [PubMed] [Google Scholar]
  • 84.Benito AI, Furlong T, Martin PJ, Anasetti C, Appelbaum FR, Doney K, Nash RA, Papayannopoulou T, Storb R, Sullivan KM, et al. Sirolimus (rapamycin) for the treatment of steroid-refractory acute graft-versus-host disease. Transplantation. 2001;72:1924–1929. doi: 10.1097/00007890-200112270-00010. [DOI] [PubMed] [Google Scholar]
  • 85.Couriel DR, Saliba R, Escalon MP, Hsu Y, Ghosh S, Ippoliti C, Hicks K, Donato M, Giralt S, Khouri IF, et al. Sirolimus in combination with tacrolimus and corticosteroids for the treatment of resistant chronic graft-versus-host disease. Br J Haematol. 2005;130:409–417. doi: 10.1111/j.1365-2141.2005.05616.x. [DOI] [PubMed] [Google Scholar]
  • 86.Basara N, Blau WI, Romer E, Rudolphi M, Bischoff M, Kirsten D, Sanchez H, Gunzelmann S, Fauser AA. Mycophenolate mofetil for the treatment of acute and chronic GVHD in bone marrow transplant patients. Bone Marrow Transplant. 1998;22:61–65. doi: 10.1038/sj.bmt.1701281. [DOI] [PubMed] [Google Scholar]
  • 87.Basara N, Kiehl MG, Blau W, Romer E, Bischoff M, Schmetzer B, Kirsten D, Gunzelmann S, Fauser AA. Mycophenolate Mofetil in the treatment of acute and chronic GVHD in hematopoietic stem cell transplant patients: four years of experience. Transplant Proc. 2001;33:2121–2123. doi: 10.1016/s0041-1345(01)01968-6. [DOI] [PubMed] [Google Scholar]
  • 88.Krejci M, Doubek M, Buchler T, Brychtova Y, Vorlicek J, Mayer J. Mycophenolate mofetil for the treatment of acute and chronic steroid-refractory graft-versus-host disease. Ann Hematol. 2005;84:681–685. doi: 10.1007/s00277-005-1070-0. [DOI] [PubMed] [Google Scholar]
  • 89.Takami A, Mochizuki K, Okumura H, Ito S, Suga Y, Yamazaki H, Yamazaki M, Kondo Y, Asakura H, Nakao S. Mycophenolate mofetil is effective and well tolerated in the treatment of refractory acute and chronic graft-versus-host disease. Int J Hematol. 2006;83:80–85. doi: 10.1532/IJH97.05111. [DOI] [PubMed] [Google Scholar]
  • 90**.Alousi A WD, Logan BR, Bolanos-Meade J, Goldstein SC, Ho VT, Hayes-Lattin B, Wingard JR, Horowitz MM, Levine JE. BMT CTN 0302: A phase II randomized trial evaluating etanercept, mycophenolate mofetil, denileukin diftitox, and pentostatin in combination with corticosteroids in 180 patients with newly diagnosed acute graft versus host disease. Blood. 2008;112:27–28. A phase II randomized BMT CTN trial comparing one of the four agents MMF, etanercept, denileukin diftitox and pentostatin in addition to corticosteroids as initial therapy of acute GVHD. [Google Scholar]
  • 91.Martin PJ, Storer BE, Rowley SD, Flowers ME, Lee SJ, Carpenter PA, Wingard JR, Shaughnessy PJ, Devetten MP, Jagasia M, et al. Evaluation of mycophenolate mofetil for initial treatment of chronic graft-versus-host disease. Blood. 2009 doi: 10.1182/blood-2009-02-202937. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Edelson R, Berger C, Gasparro F, Jegasothy B, Heald P, Wintroub B, Vonderheid E, Knobler R, Wolff K, Plewig G, et al. Treatment of cutaneous T-cell lymphoma by extracorporeal photochemotherapy. Preliminary results. N Engl J Med. 1987;316:297–303. doi: 10.1056/NEJM198702053160603. [DOI] [PubMed] [Google Scholar]
  • 93.Setterblad N, Garban F, Weigl R, Assier E, Drillat P, Charron D, Dickinson A, Greinix H, Mooney N. Extracorporeal photophoresis increases sensitivity of monocytes from patients with graft-versus-host disease to HLA-DR-mediated cell death. Transfusion. 2008;48:169–177. doi: 10.1111/j.1537-2995.2007.01502.x. [DOI] [PubMed] [Google Scholar]
  • 94*.Gatza E, Rogers CE, Clouthier SG, Lowler KP, Tawara I, Liu C, Reddy P, Ferrara JL. Extracorporeal photopheresis reverses experimental graft-versus-host disease through regulatory T cells. Blood. 2008;112:1515–1521. doi: 10.1182/blood-2007-11-125542. An experimental study showing that the transfer of cells treated with ECP reverses established GVHD by increasing donor regulatory T cells and reducing the number of donor effector lymphocytes. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Lamioni A, Parisi F, Isacchi G, Giorda E, Di Cesare S, Landolfo A, Cenci F, Bottazzo GF, Carsetti R. The immunological effects of extracorporeal photopheresis unraveled: induction of tolerogenic dendritic cells in vitro and regulatory T cells in vivo. Transplantation. 2005;79:846–850. doi: 10.1097/01.tp.0000157278.02848.c7. [DOI] [PubMed] [Google Scholar]
  • 96.Greinix HT, Knobler RM, Worel N, Schneider B, Schneeberger A, Hoecker P, Mitterbauer M, Rabitsch W, Schulenburg A, Kalhs P. The effect of intensified extracorporeal photochemotherapy on long-term survival in patients with severe acute graft-versus-host disease. Haematologica. 2006;91:405–408. [PubMed] [Google Scholar]
  • 97.Perfetti P, Carlier P, Strada P, Gualandi F, Occhini D, Van Lint MT, Ibatici A, Lamparelli T, Bruno B, Raiola AM, et al. Extracorporeal photopheresis for the treatment of steroid refractory acute GVHD. Bone Marrow Transplant. 2008;42:609–617. doi: 10.1038/bmt.2008.221. [DOI] [PubMed] [Google Scholar]
  • 98.Calore E, Calo A, Tridello G, Cesaro S, Pillon M, Varotto S, Gazzola MV, Destro R, Marson P, Trentin L, et al. Extracorporeal photochemotherapy may improve outcome in children with acute GVHD. Bone Marrow Transplant. 2008;42:421–425. doi: 10.1038/bmt.2008.174. [DOI] [PubMed] [Google Scholar]
  • 99.Berger M, Pessolano R, Albiani R, Asaftei S, Barat V, Carraro F, Biasin E, Madon E, Fagioli F. Extracorporeal photopheresis for steroid resistant graft versus host disease in pediatric patients: a pilot single institution report. J Pediatr Hematol Oncol. 2007;29:678–687. doi: 10.1097/MPH.0b013e31814d66f5. [DOI] [PubMed] [Google Scholar]
  • 100*.Le Blanc K, Frassoni F, Ball L, Locatelli F, Roelofs H, Lewis I, Lanino E, Sundberg B, Bernardo ME, Remberger M, et al. Mesenchymal stem cells for treatment of steroid-resistant, severe, acute graft-versus-host disease: a phase II study. Lancet. 2008;371:1579–1586. doi: 10.1016/S0140-6736(08)60690-X. The first large clinical trial using cellular component namely MSC for treatment of acute GVHD. [DOI] [PubMed] [Google Scholar]
  • 101.von Bonin M, Stolzel F, Goedecke A, Richter K, Wuschek N, Holig K, Platzbecker U, Illmer T, Schaich M, Schetelig J, et al. Treatment of refractory acute GVHD with third-party MSC expanded in platelet lysate-containing medium. Bone Marrow Transplant. 2009;43:245–251. doi: 10.1038/bmt.2008.316. [DOI] [PubMed] [Google Scholar]
  • 102.Uccelli A, Moretta L, Pistoia V. Mesenchymal stem cells in health and disease. Nat Rev Immunol. 2008;8:726–736. doi: 10.1038/nri2395. [DOI] [PubMed] [Google Scholar]
  • 103.Miura M, Miura Y, Padilla-Nash HM, Molinolo AA, Fu B, Patel V, Seo BM, Sonoyama W, Zheng JJ, Baker CC, et al. Accumulated chromosomal instability in murine bone marrow mesenchymal stem cells leads to malignant transformation. Stem Cells. 2006;24:1095–1103. doi: 10.1634/stemcells.2005-0403. [DOI] [PubMed] [Google Scholar]
  • 104.Tolar J, Nauta AJ, Osborn MJ, Panoskaltsis Mortari A, McElmurry RT, Bell S, Xia L, Zhou N, Riddle M, Schroeder TM, et al. Sarcoma derived from cultured mesenchymal stem cells. Stem Cells. 2007;25:371–379. doi: 10.1634/stemcells.2005-0620. [DOI] [PubMed] [Google Scholar]
  • 105.Paczesny S, Levine JE, Braun TM, Ferrara JL. Plasma biomarkers in graft-versus-host disease: a new era? Biol Blood Marrow Transplant. 2008;15:33–38. doi: 10.1016/j.bbmt.2008.10.027. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106*.Paczesny S, Krijanovski OI, Braun TM, Choi SW, Clouthier SG, Kuick R, Misek DE, Cooke KR, Kitko CL, Weyand A, et al. A biomarker panel for acute graft-versus-host disease. Blood. 2009;113:273–278. doi: 10.1182/blood-2008-07-167098. A study showing a panel of four biomarkers that had high specificity and sensitivity for the diagnosis of GVHD in patients at onset of their clinical symptoms and that provided prognostic information independent of GVHD severity. This is also the first plasma biomarkers study using a discovery and validation sets. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.Zheng Y, Rudensky AY. Foxp3 in control of the regulatory T cell lineage. Nat Immunol. 2007;8:457–462. doi: 10.1038/ni1455. [DOI] [PubMed] [Google Scholar]
  • 108.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;196:401–406. doi: 10.1084/jem.20020090. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109.Edinger M, Hoffmann P, Ermann J, Drago K, Fathman CG, Strober S, Negrin RS. CD4+CD25+ regulatory T cells preserve graft-versus-tumor activity while inhibiting graft-versus-host disease after bone marrow transplantation. Nat Med. 2003;9:1144–1150. doi: 10.1038/nm915. [DOI] [PubMed] [Google Scholar]
  • 110.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;196:389–399. doi: 10.1084/jem.20020399. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111.Jones SC, Murphy GF, Korngold R. Post-hematopoietic cell transplantation control of graft-versus-host disease by donor CD425 T cells to allow an effective graft-versus-leukemia response. Biol Blood Marrow Transplant. 2003;9:243–256. doi: 10.1053/bbmt.2003.50027. [DOI] [PubMed] [Google Scholar]
  • 112.Taylor PA, Lees CJ, Blazar BR. The infusion of ex vivo activated and expanded CD4(+)CD25(+) immune regulatory cells inhibits graft-versus-host disease lethality. Blood. 2002;99:3493–3499. doi: 10.1182/blood.v99.10.3493. [DOI] [PubMed] [Google Scholar]
  • 113.Coghill JM, Carlson MJ, Moran TP, Serody JS. The biology and therapeutic potential of natural regulatory T-cells in the bone marrow transplant setting. Leuk Lymphoma. 2008;49:1860–1869. doi: 10.1080/10428190802272684. [DOI] [PubMed] [Google Scholar]
  • 114.Nguyen VH, Shashidhar S, Chang DS, Ho L, Kambham N, Bachmann M, Brown JM, Negrin RS. The impact of regulatory T cells on T-cell immunity following hematopoietic cell transplantation. Blood. 2008;111:945–953. doi: 10.1182/blood-2007-07-103895. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115.Dieckmann D, Plottner H, Berchtold S, Berger T, Schuler G. Ex vivo isolation and characterization of CD4(+)CD25(+) T cells with regulatory properties from human blood. J Exp Med. 2001;193:1303–1310. doi: 10.1084/jem.193.11.1303. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116.Hoffmann P, Boeld TJ, Eder R, Albrecht J, Doser K, Piseshka B, Dada A, Niemand C, Assenmacher M, Orso E, et al. Isolation of CD4+CD25+ regulatory T cells for clinical trials. Biol Blood Marrow Transplant. 2006;12:267–274. doi: 10.1016/j.bbmt.2006.01.005. [DOI] [PubMed] [Google Scholar]
  • 117.Jonuleit H, Schmitt E, Stassen M, Tuettenberg A, Knop J, Enk AH. Identification and functional characterization of human CD4(+)CD25(+) T cells with regulatory properties isolated from peripheral blood. J Exp Med. 2001;193:1285–1294. doi: 10.1084/jem.193.11.1285. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118.Zorn E, Mohseni M, Kim H, Porcheray F, Lynch A, Bellucci R, Canning C, Alyea EP, Soiffer RJ, Ritz J. Combined CD4+ donor lymphocyte infusion and low-dose recombinant IL-2 expand FOXP3+ regulatory T cells following allogeneic hematopoietic stem cell transplantation. Biol Blood Marrow Transplant. 2009;15:382–388. doi: 10.1016/j.bbmt.2008.12.494. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119.Yi T, Zhao D, Lin CL, Zhang C, Chen Y, Todorov I, LeBon T, Kandeel F, Forman S, Zeng D. Absence of donor Th17 leads to augmented Th1 differentiation and exacerbated acute graft-versus-host disease. Blood. 2008;112:2101–2110. doi: 10.1182/blood-2007-12-126987. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120.Kappel LW, Goldberg GL, King CG, Suh DY, Smith OM, Ligh C, Holland AM, Grubin J, Mark NM, Liu C, et al. IL-17 contributes to CD4-mediated graft-versus-host disease. Blood. 2009;113:945–952. doi: 10.1182/blood-2008-08-172155. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 121.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;113:1365–1374. doi: 10.1182/blood-2008-06-162420. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122.Anderson BE, McNiff J, Yan J, Doyle H, Mamula M, Shlomchik MJ, Shlomchik WD. Memory CD4+ T cells do not induce graft-versus-host disease. J Clin Invest. 2003;112:101–108. doi: 10.1172/JCI17601. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 123.Chen BJ, Cui X, Sempowski GD, Liu C, Chao NJ. Transfer of allogeneic CD62L- memory T cells without graft-versus-host disease. Blood. 2004;103:1534–1541. doi: 10.1182/blood-2003-08-2987. [DOI] [PubMed] [Google Scholar]
  • 124.Zhang Y, Joe G, Zhu J, Carroll R, Levine B, Hexner E, June C, Emerson SG. Dendritic cell-activated CD44hiCD8+ T cells are defective in mediating acute graft-versus-host disease but retain graft-versus-leukemia activity. Blood. 2004;103:3970–3978. doi: 10.1182/blood-2003-09-3135. [DOI] [PubMed] [Google Scholar]
  • 125.Zheng H, Matte-Martone C, Li H, Anderson BE, Venketesan S, Sheng Tan H, Jain D, McNiff J, Shlomchik WD. Effector memory CD4+ T cells mediate graft-versus-leukemia without inducing graft-versus-host disease. Blood. 2008;111:2476–2484. doi: 10.1182/blood-2007-08-109678. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126.Sallusto F, Langenkamp A, Geginat J, Lanzavecchia A. Functional subsets of memory T cells identified by CCR7 expression. Curr Top Microbiol Immunol. 2000;251:167–171. doi: 10.1007/978-3-642-57276-0_21. [DOI] [PubMed] [Google Scholar]
  • 127.Sallusto F, Lenig D, Forster R, Lipp M, Lanzavecchia A. Two subsets of memory T lymphocytes with distinct homing potentials and effector functions. Nature. 1999;401:708–712. doi: 10.1038/44385. [DOI] [PubMed] [Google Scholar]
  • 128.Yakoub-Agha I, Saule P, Depil S, Micol JB, Grutzmacher C, Boulanger-Villard F, Bauters F, Jouet JP, Dessaint JP, Labalette M. A high proportion of donor CD4+ T cells expressing the lymph node-homing chemokine receptor CCR7 increases incidence and severity of acute graft-versus-host disease in patients undergoing allogeneic stem cell transplantation for hematological malignancy. Leukemia. 2006;20:1557–1565. doi: 10.1038/sj.leu.2404308. [DOI] [PubMed] [Google Scholar]
  • 129.Yakoub-Agha I, Saule P, Depil S, Grutzmacher C, Boulanger F, Magro L, Jouet JP, Dessaint JP, Labalette M. Comparative analysis of naive and memory CD4+ and CD8+ T-cell subsets in bone marrow and G-CSF-mobilized peripheral blood stem cell allografts: impact of donor characteristics. Exp Hematol. 2007;35:861–871. doi: 10.1016/j.exphem.2007.03.006. [DOI] [PubMed] [Google Scholar]
  • 130.Morris ES, Macdonald KP, Kuns RD, Morris HM, Banovic T, Don AL, Rowe V, Wilson YA, Raffelt NC, Engwerda CR, et al. Induction of natural killer T cell-dependent alloreactivity by administration of granulocyte colony-stimulating factor after bone marrow transplantation. Nat Med. 2009 doi: 10.1038/nm.1948. [DOI] [PubMed] [Google Scholar]
  • 131.Ringden O, Labopin M, Gorin NC, Le Blanc K, Rocha V, Gluckman E, Reiffers J, Arcese W, Vossen JM, Jouet JP, et al. Treatment with granulocyte colony-stimulating factor after allogeneic bone marrow transplantation for acute leukemia increases the risk of graft-versus-host disease and death: a study from the Acute Leukemia Working Party of the European Group for Blood and Marrow Transplantation. J Clin Oncol. 2004;22:416–423. doi: 10.1200/JCO.2004.06.102. [DOI] [PubMed] [Google Scholar]
  • 132*.Reddy P, Sun Y, Toubai T, Duran-Struuck R, Clouthier SG, Weisiger E, Maeda Y, Tawara I, Krijanovski O, Gatza E, 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;118:2562–2573. doi: 10.1172/JCI34712. An experimental study showing that histone deacetylase inhibitors regulate multiple dendritic cells functions through the induction of IDO and suggest that they may represent a novel class of agents to treat GVHD. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 133.Reddy P, Maeda Y, Hotary K, Liu C, Reznikov LL, Dinarello CA, Ferrara JL. Histone deacetylase inhibitor suberoylanilide hydroxamic acid reduces acute graft-versus-host disease and preserves graft-versus-leukemia effect. Proc Natl Acad Sci U S A. 2004;101:3921–3926. doi: 10.1073/pnas.0400380101. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 134.Sun Y, Chin YE, Weisiger E, Malter C, Tawara I, Toubai T, Gatza E, Mascagni P, Dinarello CA, Reddy P. Cutting edge: Negative regulation of dendritic cells through acetylation of the nonhistone protein STAT-3. J Immunol. 2009;182:5899–5903. doi: 10.4049/jimmunol.0804388. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 135.Wilson J, Cullup H, Lourie R, Sheng Y, Palkova A, Radford KJ, Dickinson AM, Rice AM, Hart DN, Munster DJ. Antibody to the dendritic cell surface activation antigen CD83 prevents acute graft-versus-host disease. J Exp Med. 2009;206:387–398. doi: 10.1084/jem.20070723. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 136.Hadeiba H, Sato T, Habtezion A, Oderup C, Pan J, Butcher EC. CCR9 expression defines tolerogenic plasmacytoid dendritic cells able to suppress acute graft-versus-host disease. Nat Immunol. 2008;9:1253–1260. doi: 10.1038/ni.1658. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 137.Koyama M, Hashimoto D, Aoyama K, Matsuoka K, Karube K, Niiro H, Harada M, Tanimoto M, Akashi K, Teshima T. Plasmacytoid dendritic cells prime alloreactive T cells to mediate graft-versus-host disease as antigen-presenting cells. Blood. 2009;113:2088–2095. doi: 10.1182/blood-2008-07-168609. [DOI] [PubMed] [Google Scholar]
  • 138.Michonneau D, Peffault de Latour R, Porcher R, Robin M, Benbunan M, Rocha V, Ribaud P, Ferry C, Devergie A, Vanneaux V, et al. Influence of bone marrow graft B lymphocyte subsets on outcome after HLA-identical sibling transplants. Br J Haematol. 2009;145:107–114. doi: 10.1111/j.1365-2141.2008.07574.x. [DOI] [PubMed] [Google Scholar]
  • 139.Ratanatharathorn V, Logan B, Wang D, Horowitz M, Uberti JP, Ringden O, Gale RP, Khoury H, Arora M, Spellman S, et al. Prior rituximab correlates with less acute graft-versus-host disease and better survival in B-cell lymphoma patients who received allogeneic peripheral blood stem cell transplantation. Br J Haematol. 2009 doi: 10.1111/j.1365-2141.2009.07674.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 140.Rowe V, Banovic T, MacDonald KP, Kuns R, Don AL, Morris ES, Burman AC, Bofinger HM, Clouston AD, Hill GR. Host B cells produce IL-10 following TBI and attenuate acute GVHD after allogeneic bone marrow transplantation. Blood. 2006;108:2485–2492. doi: 10.1182/blood-2006-04-016063. [DOI] [PubMed] [Google Scholar]

RESOURCES