Acute graft-versus-host disease (GVHD) is 1 of the major life-threating complications after allogeneic cell transplantation. Although steroids remain first-line treatment, roughly one-half of patients will develop steroid-refractory GVHD (SR-GVHD), which portends an extremely poor prognosis. Many agents that have shown encouraging response rates in early phase 1/2 trials for prevention and treatment have been unsuccessful in demonstrating a survival advantage when applied in the setting of SR-GVHD. The discovery of novel treatments has been further complicated by the absence of clinically informative animal models that address what may reflect a distinct pathophysiology. Nonetheless, the combined knowledge of established bone marrow transplantation models and recent human trials in SR-GVHD patients are beginning to illuminate novel mechanisms for inhibiting T-cell signaling and promoting tissue tolerance that provide an increased understanding of the underlying biology of SR-GVHD. Here, we discuss recent findings of newly appreciated cellular and molecular mechanisms and provide novel translational opportunities for advancing the effectiveness of treatment in SR-GVHD.

Despite significant progress in allogeneic cell transplantation (HCT; allo-HCT) for the treatment of malignant and nonmalignant disorders, acute graft-versus-host disease (GVHD) remains a major driver of nonrelapse mortality. For decades, high dosages (1-2 mg/kg per day) of prednisone or methylprednisolone have remained a pillar of frontline treatment in the 30% to 50% of allo-HCT recipients who develop GVHD.1  Unfortunately, roughly one-half of patients receiving therapy will not demonstrate an initial response; even fewer (∼30%) will exhibit a durable response that can facilitate withdrawal from glucocorticoids.2,3  Thus, scenarios of nonresponse, progression, or prolonged dependence broadly define steroid-refractory GVHD (SR-GVHD). Overall survival (OS) in SR-GVHD is poor, historically <50% at 6 months, and survival after failure to respond to second-line therapy is dismal (OS < 30%).4,5  Many agents have shown encouraging phase 2 response rates, but none have demonstrated a survival advantage in randomized trials.6,7  The lack of high-quality clinical evidence in the form of controlled trials is compounded by our incomplete understanding of SR-GVHD pathophysiology. To better understand the mechanistic underpinnings of SR-GVHD, we discuss the molecular and intracellular functions of steroids in modulating innate and adaptive immune responses. Second, we discuss potential mechanisms of steroid-mediated regulation of alloreactivity based on our current understanding of GVHD pathophysiology. Here, potential mechanisms of corticosteroid resistance as well as results from basic and translational studies highlight emerging themes in modification of T-cell function, regulation of immune tolerance, and protection of host tissues. Finally, we discuss implications for the prediction, treatment, and prevention of SR-GVHD in the clinic.

Acute GVHD progression can be categorized into 3 phases.8  Host tissue injuries caused by conditioning regimens mediate release of damage-associated molecular patterns (DAMPs), such as adenosine triphosphate,9,10  from injured tissues and pathogen-associated molecular patterns (PAMPs), such as lipopolysaccharide,11  from microbiomes. DAMPs and PAMPs activate recipient and/or donor-derived antigen-presenting cells (APCs), such as dendritic cells (DCs), macrophages (MFs), and host-derived nonhematopoietic cells in epithelial surfaces that in turn produce numerous proinflammatory cytokines (tumor necrosis factor α [TNF-α], interleukin 6 [IL-6]).12-15  Pattern recognition receptors, such as Toll-like receptors (TLRs) and nucleotide-binding oligomerization domain, leucine-rich repeat and pyrin domain–containing 3 (NLRP3), recognize PAMPs and DAMPs.16,17  Specific subsets of DCs (CD103+ DCs, CD8α+ DCs) are capable of alternating roles in either promoting or ameliorating GVHD depending on the inflammatory milieu.18,19  This contrasts certain committed populations such as myeloid-derived suppressor cells and granulocyte colony-stimulating factor (G-CSF)–induced CD34+ regulatory monocytes that exclusively function to constrain GVHD severity.20,21  More recently, activated neutrophils stimulated by bacteria may also function as APCs in an inflammasome-dependent context.22,23  Taken together, activated APCs stimulate newly infused donor-derived naive T cells to respond to host antigens/tissues that characterize GVHD.

The diversity of gut microbiota and viromes has been shown to be dramatically altered after conditioning and may participate in the development of GVHD, especially in the gastrointestinal (GI) tract.24-27  IL-22 produced by innate lymphoid cells protects intestinal stem cells (ISCs) and ameliorates GVHD.28  The ISC compartment located at the crypt base is the primary target of allogeneic donor T cells regulated by mucosal addressin cell adhesion molecule 1 (MAdCAM-1), which is an important adhesion molecule for T-cell migration into the gut.29  In addition, regenerating islet-derived 3α (REG3α) from Paneth cells, induced by IL-22, has been demonstrated to prevent crypt apoptosis and decrease GVHD.30  Moreover, metabolites, such as butyrate or indole, play an important role in protecting ISCs and maintaining tissue homeostasis.31-33  Therefore, host target tissue–intrinsic mechanisms may be crucial for ameliorating GVHD. We and others have demonstrated that, in intestinal epithelial cells, several molecules and pathways, such as inhibitor of apoptosis proteins, NLRP6, retinoic acid–inducible gene I (RIG-I)/mitochondrial antiviral signaling, and stimulator of interferon genes, act as tissue-intrinsic mechanisms that regulate GVHD pathogenesis.27,34-37 

After donor T cells engage APCs they become activated to proliferate. Activated donor CD4+ T cells differentiate into a variety of subsets, such as T-helper 1 (Th1), Th2, Th9, Th17, and Th22 cells whereas CD8+ T cells differentiate into cytotoxic T cells.38  In addition, several transcription factors (TFs) and their regulators, such as MAPK/extracellular signal-regulated kinases, Aurora kinase A/Janus kinase 2 (JAK2), as well as metabolic pathways (glycolysis and mitochondrial oxidative phosphorylation), have been reported to be important in the activation of T cells.38,39  These T cells then migrate into the primary GVHD target organs (intestine, liver, skin) to attack the host. Thus, donor immune cells and host tissue tolerance both emerge as key concepts in regulating GVHD. Because this system uses counterregulatory mechanisms to avoid an excessive immune response, dysfunction in certain cell populations such as regulatory T cells (Tregs), invariant natural killer (NK) T (iNKT) cells, or immune check points (programmed cell death 1 [PD-1]/programmed cell death ligand 1 [PD-L1], CD28/CTLA-4, and CD24/Siglec-G) can disrupt peripheral tolerance and amplify GVHD.40  Decreasing Treg/T effector cell ratios has especially been shown to exacerbate GVHD.39,41,42  Therefore, a major thrust of therapeutics in GVHD has emphasized enhancing Tregs by using ex vivo or in vivo expansion techniques.

The mechanisms leading to steroid-induced suppression of inflammation remain to be elucidated in SR-GVHD.43,44  Immunoregulatory functions of synthetic glucocorticoids (dexamethasone, prednisolone) are initiated through binding glucocorticoid receptors (GRs).43  Cytoplasmic GR activation promotes release from chaperones (heat shock protein 90 [HSP90]) and translocation to the nucleus to interact with DNA and proteins that mediate genomic and nongenomic functions.43-45  GRα is the canonical receptor that binds steroids and modulates immune responses. In contrast, GRβ is a splice variant that binds to DNA to antagonize GRα. Steroid-induced immune regulation can be divided into effects that are genomic and nongenomic.44,45  Genomic effects occur in the nucleus and alter gene expression by 3 mechanisms: (1) direct binding to glucocorticoid response elements to modify gene expression, (2) protein-protein interactions (tethering) with other TFs to alter gene expression of nuclear factor κB (NF-κB) and the signal transducer and activator of transcription (STAT), and (3) binding to composite elements containing a glucocorticoid response element and a response element of another TF. Genomic effects are persistent whereas nongenomic effects arise almost immediately after stimulation, are transient, and do not modify gene expression.46  Thus, steroids regulate inflammation-associated TFs in a manner that can reduce the production of proinflammatory cytokines, chemokines, and adhesion molecules.43,44 

In GVHD, the primary anti-inflammatory mechanism of steroids is mediated by inhibiting NF-κB pathways in APCs and T cells as well as TLR-mediated signaling.47-50  In addition, glucocorticoids can directly regulate activated DCs and MFs by modulating their differentiation and maturation.51-54  Steroids have been shown to decrease expression of major histocompatibility complex (MHC) class II, costimulatory molecules (CD80, CD40) and production of proinflammatory cytokines while enhancing the production of anti-inflammatory cytokines, such as IL-10 in DCs.55  In T cells, steroids suppress activation and proliferation of T cells by dampening key signaling pathways, such as nuclear factor of activated T cells, STAT, lymphocyte-specific protein tyrosine kinase, and mitogen-activated protein kinase/extracellular signal-regulated kinase.56-59  Also, steroids preferentially repress Th1 and Th17 differentiation60,61  but promote Th2 and Tregs.62-64  Finally, steroids reduce the production of chemokines and expression of adhesion molecules in a manner that decreases the migration of donor T cells into target tissues.65  However, although glucocorticoids have numerous effects that mitigate the allogeneic T-cell response, their impact on wound healing and tissue regeneration44,66  and on ameliorating tissue tolerance may in some contexts antagonize recovery from GVHD.40,67 

The pathophysiology of SR-GVHD is complex and enigmatic. The concept of “glucocorticoid resistance” in immunology was first described in the 1970s68  and remains an important area of investigation in autoimmune disease.43,69  Because steroids impair not only effector T cells but also regulatory cells, it is conceivable that their effects may also limit long-term tolerance mediated by immune-suppressor cells.70  Donor alloreactive T cells play a central role in the development of GVHD. However, one possibility is that donor T cells are less crucial in SR-GVHD than in initiation of GVHD due to previous exposure to the suppressive effects of steroids and/or calcineurin inhibitors (Figure 1A). This reasoning is supported by the results of some clinical trials in which broadly depleting donor T cells (antithymocyte globulin [ATG]) and CD25+ activated T cells (inolimomab) did not show benefit in the treatment of SR-GVHD.7  This is also supported by our data that donor T cells are dispensable in murine SR-GVHD.71  Another possibility is that certain subsets, such as Th17 cells,72,73  may not be sufficiently suppressed,64,74  especially glucocorticoid-resistant pathogenic Th17 cells that express the efflux transporter P-glycoprotein75  or glucocorticoid-induced TNF receptor family-related protein. Activation of glucocorticoid-induced TNF receptor family-related protein mediates the opposite effect on the regulation of alloreactive CD4+ and CD8+ T cells in GVHD.76  A recent study suggests that activated steroid-resistant allogeneic donor T cells in the inflamed tissues also undergo markedly altered gene-expression profiles resulting in upregulation of genes involved in T-cell activation (CD28, Tnfrsf9), migration (Cxcr6), and metabolic reprograming (Hif1α) that facilitate increased glycolytic demands.77 

Figure 1.

Mechanisms of SR-GVHD. (A) Cellular mechanisms. Steroids regulate the majority of Th1 responses but can paradoxically increase Th2 and pathogenic Th17-mediated immune responses that may promote GVHD. The role of steroids in CD8+ T cells is uncertain. The combination of steroids with calcineurin inhibitor (CNI) may unintentionally blunt induction of Tregs based on their requirement for IL-2 resulting in loss of peripheral tolerance. (B) Molecular mechanisms. Steroids repress expression of TFs necessary for production of proinflammatory cytokines (IL-6, TNFα). In addition, steroids promote induction of regulatory cell subsets, such as CD103+ DCs and M2 MFs that induce immune tolerance. In refractory disease, long-term use of steroids may paradoxically increase expression of TLRs and NLRP3 that perpetuate inflammation. (C) Target tissue–intrinsic mechanisms. In the GI tract, steroids can impede the reparative processes of the host following T-cell–mediated injury that is associated with loss of Paneth cells, ISCs, and immune-regulatory proteins (α-1-antitrypsin [AAT]). Limited tissue regeneration from long-term suppression of inflammation with ongoing mucosal barrier injury is associated with alterations in the intestinal microbiome and metabolome. Dysbiosis results in loss of protective metabolites (butyrate). Ongoing inflammation can eventually stimulate APCs to increase production of proinflammatory cytokines that further damage host tissue. AP-1, activator protein 1; GCR, glucocorticoid receptor; ROS, reactive oxygen species; Tc, cytotoxic T cell; TCR, T-cell receptor.

Figure 1.

Mechanisms of SR-GVHD. (A) Cellular mechanisms. Steroids regulate the majority of Th1 responses but can paradoxically increase Th2 and pathogenic Th17-mediated immune responses that may promote GVHD. The role of steroids in CD8+ T cells is uncertain. The combination of steroids with calcineurin inhibitor (CNI) may unintentionally blunt induction of Tregs based on their requirement for IL-2 resulting in loss of peripheral tolerance. (B) Molecular mechanisms. Steroids repress expression of TFs necessary for production of proinflammatory cytokines (IL-6, TNFα). In addition, steroids promote induction of regulatory cell subsets, such as CD103+ DCs and M2 MFs that induce immune tolerance. In refractory disease, long-term use of steroids may paradoxically increase expression of TLRs and NLRP3 that perpetuate inflammation. (C) Target tissue–intrinsic mechanisms. In the GI tract, steroids can impede the reparative processes of the host following T-cell–mediated injury that is associated with loss of Paneth cells, ISCs, and immune-regulatory proteins (α-1-antitrypsin [AAT]). Limited tissue regeneration from long-term suppression of inflammation with ongoing mucosal barrier injury is associated with alterations in the intestinal microbiome and metabolome. Dysbiosis results in loss of protective metabolites (butyrate). Ongoing inflammation can eventually stimulate APCs to increase production of proinflammatory cytokines that further damage host tissue. AP-1, activator protein 1; GCR, glucocorticoid receptor; ROS, reactive oxygen species; Tc, cytotoxic T cell; TCR, T-cell receptor.

Close modal

Although the initiating factors of steroid resistance remain obscure, cytokine-induced mechanisms are well investigated in chronic inflammation. Inflammatory cytokines, such as interferon γ, which upregulate the expression of MHC class II and increased antigen presentation in the intestinal epithelium, may be correlated with SR-GVHD.15,78  IL-6, a potential mediator of SR-GVHD, is elevated in the serum during GVHD79  and is also known to directly damage intestinal epithelial tissues80  with IL-6/IL-6 receptor levels enriched in the colonic microenvironment.81  Downstream of IL-6, JAKs possess pleomorphic functions in regulating the immune response and have demonstrated strong rationale in the therapy of SR-GVHD.82-85  The representative cellular mechanisms of SR-GVHD are summarized in Figure 1A.

Compared with cellular mechanisms, relatively less is understood about the underlying molecular features of SR-GVHD. However, recent insights into the immunological impact of steroids, some that paradoxically are immune enhancing, have shed light on these processes (Figure 1B). For example, the TLR4 agonist lipopolysaccharide (LPS) regulates the expression of GRα and β isoforms that prevent the inhibitory effects of glucocorticoids on granulocyte macrophage colony-stimulating factor (GM-CSF) secretion.86  Proinflammatory cytokines also induce glucocorticoid resistance by impairing phosphorylation and function of the GR.87-89  The inflammasome, specifically NLRP3–caspase 1 (CASP1), can deplete cellular levels of GR thus limiting overall cell sensitivity to glucocorticoids.90  Preclinical models suggest that intact recipient GR is necessary to restrain the severity of GVHD,91  and GR single-nucleotide polymorphisms (SNPs) have been associated with clinical outcomes.92  These findings suggest that cytokines, DAMPs, PAMPs, including inflammasome pathways, may drive SR-GVHD.

Target tissue–intrinsic mechanisms are becoming increasingly implicated in SR-GVHD (Figure 1C). Steroids administrated in GVHD are delivered into damaged tissues infiltrated by T cells, DAMPs, and PAMPs. As mentioned, steroids have bilateral effects, not only immunosuppressive but also immune-stimulatory that potentiate the effects of DAMPs and PAMPs, which in turn mediate GR modifications. In addition, steroids promote expression of TLRs, inflammasomes (NLRP3), and purinergic receptor (P2Y2R), which further sensitize cells to potently respond to DAMPs and PAMPs.44  These results suggest that steroids themselves, in certain contexts, may enhance tissue-specific immune responses that perpetuate GVHD. Steroids and other currently available pharmacologic agents have not been shown to terminate ongoing organ damage. Increased serum cytokeratin-18 fragments, associated with epithelial cell apoptosis, can be detected as persistent organ damage occurs in SR-GVHD.93  Angiopoietin-2 (ANG2), which regulates vessel quiescence, is also increased in patients with SR-GVHD,94,95  suggesting that vascular endothelial injury contributes to SR-GVHD. Interestingly, these findings appear associated with the clinical syndrome of transplant-associated thrombotic microangiopathy (TA-TMA), mediated by alternative and terminal complement activation, which is linked with SR-GVHD.96,97 

Disruption of tissue tolerance may explain why intensive immune-suppressive strategies are often ineffective in SR-GVHD, especially involving the GI tract.40,67  Typically, injured tissues have the capacity to regenerate. Once tissue tolerance has been impaired, barrier function, which protects host tissues from invading pathogens and abnormal immune responses, is destroyed. It has recently been recognized that the diversity of intestinal microbiota is altered in GVHD24,27,98  and efforts to correct this dysbiosis with fecal microbiome transplantation (FMT) are being attempted for treatment of SR-GVHD,99-101  suggesting that the microbiota and its associated metabolites such as butyrate may be implicated.31  Long-term exposure to aberrant microbiomes, PAMPs, and DAMPs may mediate prolonged mucosal barrier dysfunction and inflammation. Therefore, disease-promoting tissue-intrinsic mechanisms are becoming an important component to consider in the treatment of SR-GVHD.

In the absence of clinical progression, identifying treatment resistance often requires 7 to 14 days of high-dose steroid therapy.2,102  Responses are insidious, as reductions in diarrhea may be fleeting once attempts are made to reduce steroid dosage.6  Because SR-GVHD often reflects advanced organ injury, early detection with prompt treatment intensification has been postulated as a clinical strategy to reduce alloreactivity and constrain organ injury. Although some studies suggest that early institution of second-line therapy can improve outcome,103  the premise that early prediction can improve outcomes remains an open question being addressed in preemptive trials (NCT03459040). In the following sections, we emphasize biology-based markers for SR-GVHD; other comprehensive reviews are available on the topic of GVHD biomarkers.104,105 

Clinical and histologic risk factors

Patient characteristics known to heighten the probability of steroid resistance include donor-recipient HLA disparity, advanced age, and lower GI tract or hepatic involvement.106  Not surprisingly, organ involvement and severity at GVHD onset strongly predict response. Patients deemed high risk by refined Minnesota score had a 44% probability of response by day 28 compared with 68% in standard risk, although among 1723 patients only 16% were labeled high risk.107  Despite being a clinical diagnosis, histology at GVHD onset may suggest advanced pathobiology heralding a complicated clinical course. For example, severe crypt loss with denudation of the GI epithelium is correlated with higher clinical grade, steroid resistance, and mortality.108  Paneth cells that support adjacent ISCs in the crypt regulate epithelial regeneration and shape microbial ecology through secretion of antimicrobial peptides (α-defensins, REG3α). In line with these observations, loss of Paneth cells in GI biopsies correlates with lack of response to therapy.109  Because Paneth cell loss can influence ISC populations (and regeneration), these findings support tissue injury being a hallmark of clinically resistant phenotypes.

Cellular byproducts of tissue injury

Given the diagnostic challenges in GVHD, there is interest in generating validated serum biomarkers capable of aiding clinical decisions. The Mount Sinai Acute GVHD International Consortium (MAGIC) recently assessed a validated 2-biomarker algorithm involving suppressor of tumorigenicity-2 (ST2) and REG3α to predict treatment resistance.110  High levels of REG3α (stored in Paneth cells and GI mucous) are released into serum during early crypt damage and soluble ST2 is released by alloreactive T cells in the gut.111-114  Patients with high-risk biomarkers 1 week after initiating corticosteroid treatment had significantly higher treatment failure. Interestingly, a high-risk biomarker profile was suggestive of treatment failure independent of early clinical response. If prospectively validated, this information may guide risk-adapted decision-making (tapering steroids vs second-line therapy).

As GI injury is central to fatal GVHD (and repeat endoscopy is challenging), there is interest in informative stool analytes. Fecal calprotectin (CPT), expressed in MFs, monocytes, and the cytoplasm of granulocytes, may provide a real-time surrogate marker of disease activity. Elevated CPT, activated in the presence of DAMPs or PAMPs, serves as a ligand for TLR4 thus suggesting ongoing inflammatory signaling (NF-κB) not constrained by steroids. CPT itself may contribute to mucosal permeability,115  is elevated in lower GI tract GVHD, and is correlated with histopathologic severity116 ; thus, longitudinal assessment may aid in distinguishing steroid refractoriness from other forms of colitis (cytomegalovirus).117  Combining CPT with analytes such as α-1-antitrypsin (AAT), lost during GVHD-induced enteropathy, may further enhance sensitivity and provide an “actionable” protein to supplement.118 

Endothelial cell dysfunction

In addition to epithelial surfaces, inflammation of SR-GVHD involves an endothelium capable of perpetuating injury. TA-TMA is prevalent among patients with SR-GVHD (79% vs 42%; P = .001). Whether TA-TMA is an epiphenomenon of SR-GVHD or an inciting event is unknown, however, damage to the endothelium results in complement activation (BBPlus and C5b-9), release of soluble thrombomodulin, and ANG2 that impedes organ recovery.94,96  In 1 study, elevated levels of ANG2 were detected before onset of SR-GVHD, which may sensitize the endothelium to the injurious effects of proinflammatory cytokines. Variant SNPs in the TM gene have been shown to predict responsiveness to GVHD treatment, suggesting host-intrinsic differences in vulnerability to injury.94 

At present, ruxolitinib represents the only US Food and Drug Administration (FDA)-approved therapy for SR-GVHD, however, no agent has demonstrated superiority in head-to-head trials.119,120  As SR-GVHD reflects complex biology, successful treatment will likely require targeting alternative pathways or combinatorial treatment approaches that overcome putative mechanisms of corticosteroid resistance. Akin to a marathon, and acknowledging inherent limitations in clinical grading systems based on quantifying stool volume, assessing a given intervention will require careful interpretation of both early and late milestones for success. This might be accomplished through varied end points that quantify not only the frequency but depth and durability of response.121  Most SR-GVHD studies report overall response (overall response rate [ORR] or complete response [CR] plus partial response) by day 28 of treatment onset due to its association with improved treatment-related mortality in upfront treatment settings as an early readout of promising activity, particularly when this end point compares favorably against historical rates (∼50% in second-line therapy).4,122  ORR, however, is imperfect as this end point may not be indicative of improved survival if there are subsequent rises in morbidity, late mortality, or GVHD. Thus, treatment trials should increasingly incorporate other qualifiers of success that measure hard end points such as overall and GVHD-free survival, infection, and relapse to identify the most promising agents for larger controlled studies.121,123 

Lessons from existing therapies

Corticosteroids possess potent lymphocytic and anticytokine properties, nonetheless, increasing dosage beyond 2 mg/kg methylprednisolone does not increase response.6,124  T-cell receptor β (TCRβ) sequencing of diagnostic GI-tract biopsies has illuminated the persistence of glucocorticoid-resistant T-cell clonotypes in later stages of GVHD,125  suggesting that targeting alternative or multiple simultaneous signaling pathways may be necessary. T-cell–depleting sera (ATG), CD52 direct antibodies targeting T and B cells (alemtuzumab), or chemotherapies (pentostatin, pulse cyclophosphamide) are all intensive approaches used as second-line therapy.126,127  However, toxicity, namely myelosuppression, opportunistic infection, and interstitial pneumonitis, can reduce their overall efficacy, resulting in <10% long-term OS in some studies.128,129  Therefore, treatment must account for adverse events that accelerate mortality. Due to mucosal barrier injury, ongoing tissue injury from GVHD, and immune suppression, patients are particularly vulnerable to bacteremia, often from pathogenic organisms such as Enterococcus.130,131  Acknowledging these limitations, investigations have also focused on approaches that might selectively block key signaling events in GVHD. Although seemingly innocuous, monoclonal antibodies such as daclizumab, which eliminates activated CD25+ T cells, may heighten infectious mortality or unintentionally deplete Treg populations.132  In a cohort treated with CD25+- or TNFα-directed anticytokine therapy, infection-related mortality was 28% at a median of 88 days.133 

These observations suggest that immunologically intensive therapies are not necessarily ideal in refractory settings, as subtler immunomodulatory methods also illicit response. Extracorporeal photopheresis (ECP) is a commonly used technique that limits host tissues to chemotherapy via ex vivo exposure of apheresed mononuclear cells to the DNA crosslinking agent 8-methoxypsoralen in the presence of UVA light.134  ECP has pleotropic effects that include apoptosis of alloreactive lymphocytes, induction of tolerogenic DC subsets, and expansion of Tregs, which may be suppressed in SR-GVHD. In a meta-analysis of prospective studies conducted as second-line treatment, the response rate for ECP was 69%, with the highest organ-specific response rates observed in the skin (84%), followed by visceral organs (65% in GI; 55% in liver).135  Although it is unclear whether this degree of response can be recapitulated as stand-alone therapy in highly refractory settings, early initiation of ECP is a commonly used adjunct that can facilitate salutary steroid-sparing effects.

New approaches: selective targeting of alloreactive T cells

Select strategies for SR-GVHD are summarized in Table 1. Recently, a phase 1/2 trial in GI and liver GHVD evaluated an anti-CD3/CD7 antibody conjugated to ricin toxin that elicited responses of 60% of SR–acute GVHD with 60% of patients alive after 6 months. This molecule selectively targets activated T- and NK-cell signaling that may enhance potency compared with prior anti-CD3 therapies.136-138  Infectious mortality was relatively low (∼20%), possibly due to only brief periods of lymphopenia after administration. This agent is now being evaluated in an open-label trial for SR-GVHD (CTN 1802). Other proteins such as CD30, expressed on activated T cells (particularly central memory CD8+CD45RO+CD62L+ subsets), are also enriched at GVHD onset.139  In a phase 1 study of brentuximab vedotin, an antibody-drug conjugate targeting CD30, 38% of patients responded at day 28 with an additional 25% experiencing CRs at day 56.140  Finally, integrin expression on lymphocytes may be impaired in corticosteroid resistance in a manner that promotes lymphocyte recruitment to target tissues.44  A phase 2 study of natalizumab directed against α4-integrin chains, administered as a single dose with steroids, produced high response rates (∼75%) in the initial treatment of GI GVHD.141  However, natalizumab inhibits α4β1 and α4β7 integrins, the former via interactions with vascular cell adhesion molecule 1 (VCAM1), which impedes central nervous system trafficking, thereby potentially predisposing patients to John Cunningham virus reactivation and progressive multifocal leukoencephalopathy. In contrast, α4β7 integrins, selectively targeted by the monoclonal antibody vedolizumab, interact with GI mucosal addressin MadCAM-1 expressed in Peyer patches, and lamina propria may have a niche role in treating GI GVHD.29  In a pilot study, 6 of 6 patients responded after 7 to 10 days with 4 experiencing durable remissions.142  A separate retrospective study reported 60% response, however, 34% of patients experienced grade 3-4 infection, frequently bacteremia from Staphylococcus or Enterococcus.143  As several patients had received multiple lines of prior therapy, it is unclear whether infections were attributable to advanced GVHD or possible impairments in immune trafficking.

Table 1.

Novel biology-driven strategies for SR-GVHD

ApproachMechanismClinical responses, %References
Modification of alloreactive T cells    
 Anti-CD3/CD7 antibody conjugated to ricin toxin Apoptosis↑ in T and NK cells ORR, 60; CR, 50 136  
 Brentuximab vedotin CD30 inhibition, central memory ↓ (CD8+CD45RO+CD62L+ T cells) ORR, 38.2 at day 28 139,140  
 Vedolizumab Integrin α4β7 inhibition, donor T-cell homing to GI tract↓ ORR, 100 142  
ORR, 79 176  
ORR, 64 143  
Cytokines    
 Tocilizumab IL-6 signaling↓ (Innate/adaptive response↓) ORR, 67 147  
ORR, 44 146  
CR, 62.5 145  
 F-652 (IL-22 dimer/Fc fusion molecule) Preserved ISCs NCT02406651*  
Gut regeneration↑ 
Combination    
 Ruxolitinib JAK1/2 inhibition ORR, 81.5 85  
ORR, 45 177  
ORR, 57 152  
ORR, 78 178  
ORR, 84 153  
Tissue regeneration    
 Lithium Wnt signaling ↑ by inhibiting GSK3 CR, 50 157  
 AAT Serine protease inhibitor, ↑ Treg, ORR, 66.7 163  
↓APC function   
↓Proinflammatory cytokines (IL-6) ORR, 65 162  
Microbiome    
 FMT Gut microbiome diversity ↑ ORR, 100 99  
ORR, 75 101  
 Resistant starch Modification of metabolome NCT02763033,* NCT02805075*  
ApproachMechanismClinical responses, %References
Modification of alloreactive T cells    
 Anti-CD3/CD7 antibody conjugated to ricin toxin Apoptosis↑ in T and NK cells ORR, 60; CR, 50 136  
 Brentuximab vedotin CD30 inhibition, central memory ↓ (CD8+CD45RO+CD62L+ T cells) ORR, 38.2 at day 28 139,140  
 Vedolizumab Integrin α4β7 inhibition, donor T-cell homing to GI tract↓ ORR, 100 142  
ORR, 79 176  
ORR, 64 143  
Cytokines    
 Tocilizumab IL-6 signaling↓ (Innate/adaptive response↓) ORR, 67 147  
ORR, 44 146  
CR, 62.5 145  
 F-652 (IL-22 dimer/Fc fusion molecule) Preserved ISCs NCT02406651*  
Gut regeneration↑ 
Combination    
 Ruxolitinib JAK1/2 inhibition ORR, 81.5 85  
ORR, 45 177  
ORR, 57 152  
ORR, 78 178  
ORR, 84 153  
Tissue regeneration    
 Lithium Wnt signaling ↑ by inhibiting GSK3 CR, 50 157  
 AAT Serine protease inhibitor, ↑ Treg, ORR, 66.7 163  
↓APC function   
↓Proinflammatory cytokines (IL-6) ORR, 65 162  
Microbiome    
 FMT Gut microbiome diversity ↑ ORR, 100 99  
ORR, 75 101  
 Resistant starch Modification of metabolome NCT02763033,* NCT02805075*  

↓, decrease; ↑, increase; AAT, α-1-antitrypsin; CR, complete response; FMT, fecal microbiome transplantation; GSK3, glycogen synthase kinase 3; ORR, overall response rate; VGPR, very good partial response.

*

Clinical trial number.

Cytokine-based approaches

Considering the redundant roles cytokines play, antagonism of single cytokines may be insufficient to downregulate inflammation in SR-GVHD.144  Antagonizing IL-6 signaling with anti–IL-6 receptor antibody (tocilizumab) or circulating IL-6 (siltuximab) may be 1 exception as this cytokine plays multiple roles in innate and adaptive immunity and may possess tissue-projective effects.80  Production of IL-6, which is transcriptionally downregulated by corticosteroids, is likely to be further elevated during SR-GVHD. Several clinical series now report activity of tocilizumab with responses in 44% to 62% of SR-GVHD.145-147  IL-6 antagonism may be less impactful on immunity because experimentally it does not perturb monocyte DC activation or alloreactive T-cell responses.148  Another cytokine with compelling rationale for treating SR-GVHD is IL-22, owing to its tissue-protective effects. In HCT models, IL-22 restored REG3γ production lost after Paneth cell destruction and facilitated regeneration of gut epithelium.30,149  An IL-22 dimer/Fc fusion molecule (F-652) is currently undergoing testing in newly diagnosed lower GI GVHD (NCT02406651).

Combination therapy with tyrosine kinase inhibition

To date, attempts to combine immunosuppressive agents in second-line therapy have been unsuccessful. No significant elevation in response rate was seen when duel cytokines (IL-2 and TNFα) were interrupted, resulting in high rates of infection and a 6-month OS of 29%.150  However, targeting downstream cytokine-induced signal transduction via small molecule tyrosine kinase inhibitors of JAKs (and STATs) is an emerging approach to overcoming steroid resistance. For example, depletion or inactivation of cytoplasmic GRs may limit tethering to STAT3 proteins that limit transcriptional repression of proinflammatory cytokines.151  After reporting impressively high response rates (81%) in a retrospective survey, a prospective, open label, multicenter trial (REACH1) of the JAK1/2 inhibitor ruxolitinib resulted in FDA approval for treating SR-GVHD.85,152  Overall, 57% of SR-GVHD patients met the primary end point of day 28 overall response including 31% with CR. An impressive finding was that a number of responses were reported to be durable, lasting a median of 345 days. In addition to blood cytopenia, infections were the primary adverse event (∼40%). Whether the high proportion of responses justifies this potential risk remains to be determined.153  As ruxolitinib is applied to increasing patients, studies are needed to guide coadministration with CYP3A4 inhibitors (azole antifungals) that dramatically increase drug levels and optimal withdrawal procedures given theoretical risks for cytokine release. More selective JAK-1 inhibitors (itacitinib) have also similarly displayed encouraging results in pilot studies of SR-GVHD154  and are undergoing phase 3 testing (NCT03139604). Other kinase targets that engage signaling distal to immunoreceptors such as spleen tyrosine kinase (Syk) can elicit immune-suppressive effects on alloreactive T cells and APCs in preclinical models, thus warranting study in SR-GVHD.155,156 

Regeneration of host tissues

The majority of therapies focus on modifying components of the donor immune system. However, a greater emphasis on damaged target tissues may be especially crucial in SR-GVHD, in which profound injury likely participates in clinical pathology. This alters the focus of treatment toward restoration of organ function necessary for functional recovery (alimentation, reduced infection). In 1 study, lithium, capable of promoting Wnt signaling, produced a 50% CR rate in patients with GI mucosal denudation.157  In subset analysis, all patients receiving lithium within 3 days of identifying denuded mucosa had durable CR. Unfortunately, there is a paucity of available agents capable of inducing Wnt signaling, as clinical development has focused on Wnt inhibition for cancer.158 

Dysfunction of the mucosal barrier also promotes loss of immunoregulatory proteins that provide tissue protection. AAT, an acute-phase protein produced by the liver, has increased stool clearance in GI GVHD. In addition to its role as a serine protease inhibitor that prevents organ damage (congenital emphysema) by inhibiting neutrophil elastase, AAT also possesses immunomodulatory functions that suppress proinflammatory cytokines, attenuate DC function, and induce Tregs. Several independent laboratories have demonstrated that exogenous AAT infusion can reduce GVHD-related mortality.159-161  As second-line therapy, 2 trials of AAT demonstrated response rates of 65% (including 50% CR in the GI tract) with low infectious mortality (10% at 6 months).162,163  Placebo-controlled trials of AAT are currently under way in the treatment of high-risk GVHD (NCT04167514). Finally, although we have focused on pharmacological means for treating SR-GVHD, the use of cell-based therapies, Tregs, iNKT, and mesenchymal stem cells (MSCs) hold potential to promote tissue repair.164  Given early success in prevention,165,166  adoptive transfer or in vivo expansion of Tregs remains an attractive strategy, owing to their ability to mitigate GVHD without abrogating CD8+ T-cell–killing function. For example, approaches include targeting TNF superfamily receptor TNFRSF25 using the TL1A-immunoglobulin fusion protein or facilitating iNKT-Treg interactions with a synthetic TCR ligand (α-GalCer),167,168  although generating adequate cell numbers for suppressive activity will remain a challenge. With respect to MSCs, a large meta-analysis composed of nonrandomized studies reported a cumulative survival of 63% at 6 months.169  The immunosuppressive mechanisms remain to be elucidated but may relate to MSC sensitivity to apoptosis, which in turn can promote phagocytosis by host MFs and release of indoleamine 2,3-dioxygenase.170  Further mechanistic and controlled trials are needed to establish the role of MSCs in SR-GVHD.

Treatment of SR-GVHD will remain a challenge despite advances, emphasizing the importance of prevention. Rates of severe GVHD have declined due to use of high-resolution HLA typing, use of ATG, and perhaps the recent application of posttransplant cyclophosphamide.171,172  Although this indirectly impacts SR-GVHD, whether such approaches improve survival is uncertain. Extending concepts of tissue regeneration to tolerance (prevention) may increase thresholds for GVHD, reducing requirements for immune suppression. Approaches that selectively mitigate tissue response to sterile inflammatory mediators (DAMPs) by targeting Siglec-10 on host APCs have been shown to reduce GVHD in murine models.173,174  Use of a CD24 fusion protein (CD24Fc) is now undergoing clinical testing for the reduction of grade III-IV GVHD (NCT02663622). Another approach is to address microbiome-metabolome dysfunction. Here, altering the microbiome to protect vital cell populations (ISCs) may prevent irrevocable injury. For example, preclinical data suggest that short-chain fatty acids, specifically dietary butyrate or butyrate-producing clostridia, are a key energy source for ISCs that prevent GI GVHD.31  Trials testing modification of the host microbiome metabolome through ingestion of resistant starch (NCT02763033) and dietary fructo-oligosaccharides (NCT02805075) are being evaluated for GVHD prevention. Interventions involving transfer of fecal microbiota (FMT) are also associated with responses in SR-GVHD,99,101  but need to address antibiotic use, repeat administrations, and infectious risks in the preventative setting. IL-22 antagonists that induce epithelial regeneration could be logically advanced to the prevention setting because their function depends on intact ISCs. In the future, exploring what combinations of agents (IL-22, Wnt agonists) might best shape the host microbiome to preserve target tissues will be interesting.175 

After several decades, incremental advances in the fundamental mechanistic underpinnings of GVHD, together with new therapeutic approaches that target dysregulated immune biology, bring renewed optimism for progress in SR-GVHD. Strategies that selectively reshape the composition of alloreactive T cells and APCs by targeting key inflammatory mediators (IL-6), that influence cytokine-driven signal transduction (JAK/STAT), or that target organ homing have displayed impressive response rates coupled with manageable toxicity and are currently undergoing testing in larger controlled trials. Despite these advances, SR-GVHD will remain a vexing challenge, owing to its complex biology and often irreversible organ injury; thus, future directions must continue to emphasize methods to prevent or preemptively treat high-risk disease. Future innovation that can leverage emerging knowledge of the host microbiome, tissue-protective strategies, and cellular engineering as well as noninvasive diagnostics to detect subclinical disease hold significant promise for advancing the field. Finally, although common to HCT, SR-GVHD remains an orphan disease with limited numbers of patients to appropriately power larger efficacy trials. Given these limitations, prioritizing the most promising strategies based on compelling biology, rigorous preclinical data, and well-conducted phase 1/2 designs will be important to advance therapies with the greatest potential for success.

The authors thank E. Matsuki for kindly drawing the figure. The authors acknowledge that, given space and citation constraints, some relevant published work was omitted.

This work was supported by Japan Society for the Promotion of Science (JSPS) KAKENHI Grant-in-Aid for Research Activity Start-up grant number JP17H06537 (T.T.), the Mochida Memorial Foundation for Medical and Pharmaceutical Research (T.T.), the Ichiro Kanehara Foundation (T.T.), the SENSHIN Medical Research Foundation (T.T.), the Japanese Society of Hematology research grant (T.T.), the Friends of Leukemia Research Fund (T.T.), National Institutes of Health National Institute of Allergy and Infectious Diseases grant K23 AI123595-04 (J.M.), and a Rogel Cancer Center Scholarship (J.M.).

Contribution: T.T. and J.M. wrote the paper.

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

Correspondence: Tomomi Toubai, Division of Hematology and Cell Therapy, Department of Internal Medicine III, Faculty of Medicine, Yamagata University, 2-2-2 Iida-Nishi, Yamagata, 990-9585, Japan; e-mail: toubai@med.id.yamagata-u.ac.jp; or John Magenau, Division of Hematology and Oncology, Department of Internal Medicine, University of Michigan Medical School, 1500 E. Medical Center Dr, Ann Arbor, MI 48109; e-mail: johnmage@med.umich.edu.

1.
Jagasia
M
,
Arora
M
,
Flowers
ME
, et al
.
Risk factors for acute GVHD and survival after hematopoietic cell transplantation
.
Blood
.
2012
;
119
(
1
):
296
-
307
.
2.
MacMillan
ML
,
Weisdorf
DJ
,
Wagner
JE
, et al
.
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
(
7
):
387
-
394
.
3.
Levine
JE
,
Braun
TM
,
Harris
AC
, et al;
Blood and Marrow Transplant Clinical Trials Network
.
A prognostic score for acute graft-versus-host disease based on biomarkers: a multicentre study
.
Lancet Haematol
.
2015
;
2
(
1
):
e21
-
e29
.
4.
Martin
PJ
,
Rizzo
JD
,
Wingard
JR
, et al
.
First- and second-line systemic treatment of acute graft-versus-host disease: recommendations of the American Society of Blood and Marrow Transplantation
.
Biol Blood Marrow Transplant
.
2012
;
18
(
8
):
1150
-
1163
.
5.
Xhaard
A
,
Rocha
V
,
Bueno
B
, et al
.
Steroid-refractory acute GVHD: lack of long-term improved survival using new generation anticytokine treatment
.
Biol Blood Marrow Transplant
.
2012
;
18
(
3
):
406
-
413
.
6.
Deeg
HJ
.
How I treat refractory acute GVHD
.
Blood
.
2007
;
109
(
10
):
4119
-
4126
.
7.
Socié
G
,
Vigouroux
S
,
Yakoub-Agha
I
, et al
.
A phase 3 randomized trial comparing inolimomab vs usual care in steroid-resistant acute GVHD
.
Blood
.
2017
;
129
(
5
):
643
-
649
.
8.
Ferrara
JL
,
Levine
JE
,
Reddy
P
,
Holler
E
.
Graft-versus-host disease
.
Lancet
.
2009
;
373
(
9674
):
1550
-
1561
.
9.
Wilhelm
K
,
Ganesan
J
,
Müller
T
, et al
.
Graft-versus-host disease is enhanced by extracellular ATP activating P2X7R
.
Nat Med
.
2010
;
16
(
12
):
1434
-
1438
.
10.
Koehn
BH
,
Saha
A
,
McDonald-Hyman
C
, et al
.
Danger-associated extracellular ATP counters MDSC therapeutic efficacy in acute GVHD
.
Blood
.
2019
;
134
(
19
):
1670
-
1682
.
11.
Cooke
KR
,
Hill
GR
,
Crawford
JM
, et al
.
Tumor necrosis factor- alpha production to lipopolysaccharide stimulation by donor cells predicts the severity of experimental acute graft-versus-host disease
.
J Clin Invest
.
1998
;
102
(
10
):
1882
-
1891
.
12.
Teshima
T
,
Ordemann
R
,
Reddy
P
, et al
.
Acute graft-versus-host disease does not require alloantigen expression on host epithelium
.
Nat Med
.
2002
;
8
(
6
):
575
-
581
.
13.
Koyama
M
,
Kuns
RD
,
Olver
SD
, et al
.
Recipient nonhematopoietic antigen-presenting cells are sufficient to induce lethal acute graft-versus-host disease
.
Nat Med
.
2011
;
18
(
1
):
135
-
142
.
14.
Hashimoto
D
,
Chow
A
,
Noizat
C
, et al
.
Tissue-resident macrophages self-maintain locally throughout adult life with minimal contribution from circulating monocytes
.
Immunity
.
2013
;
38
(
4
):
792
-
804
.
15.
Koyama
M
,
Mukhopadhyay
P
,
Schuster
IS
, et al
.
MHC class II antigen presentation by the intestinal epithelium initiates graft-versus-host disease and is influenced by the microbiota
.
Immunity
.
2019
;
51
(
5
):
885
-
898.e7
.
16.
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
;
112
(
8
):
3508
-
3516
.
17.
Jankovic
D
,
Ganesan
J
,
Bscheider
M
, et al
.
The Nlrp3 inflammasome regulates acute graft-versus-host disease
.
J Exp Med
.
2013
;
210
(
10
):
1899
-
1910
.
18.
Koyama
M
,
Cheong
M
,
Markey
KA
, et al
.
Donor colonic CD103+ dendritic cells determine the severity of acute graft-versus-host disease
.
J Exp Med
.
2015
;
212
(
8
):
1303
-
1321
.
19.
Toubai
T
,
Malter
C
,
Tawara
I
, et al
.
Immunization with host-type CD8alpha+ dendritic cells reduces experimental acute GVHD in an IL-10-dependent manner
.
Blood
.
2010
;
115
(
3
):
724
-
735
.
20.
Highfill
SL
,
Rodriguez
PC
,
Zhou
Q
, et al
.
Bone marrow myeloid-derived suppressor cells (MDSCs) inhibit graft-versus-host disease (GVHD) via an arginase-1-dependent mechanism that is up-regulated by interleukin-13
.
Blood
.
2010
;
116
(
25
):
5738
-
5747
.
21.
D’Aveni
M
,
Rossignol
J
,
Coman
T
, et al
.
G-CSF mobilizes CD34+ regulatory monocytes that inhibit graft-versus-host disease
.
Sci Transl Med
.
2015
;
7
(
281
):
281ra42
.
22.
Hülsdünker
J
,
Thomas
OS
,
Haring
E
, et al
.
Immunization against poly-N-acetylglucosamine reduces neutrophil activation and GVHD while sparing microbial diversity
.
Proc Natl Acad Sci USA
.
2019
;
116
(
41
):
20700
-
20706
.
23.
Schwab
L
,
Goroncy
L
,
Palaniyandi
S
, et al
.
Neutrophil granulocytes recruited upon translocation of intestinal bacteria enhance graft-versus-host disease via tissue damage
.
Nat Med
.
2014
;
20
(
6
):
648
-
654
.
24.
Jenq
RR
,
van den Brink
MRM
.
Allogeneic haematopoietic stem cell transplantation: individualized stem cell and immune therapy of cancer
.
Nat Rev Cancer
.
2010
;
10
(
3
):
213
-
221
.
25.
Shono
Y
,
Docampo
MD
,
Peled
JU
, et al
.
Increased GVHD-related mortality with broad-spectrum antibiotic use after allogeneic hematopoietic stem cell transplantation in human patients and mice
.
Sci Transl Med
.
2016
;
8
(
339
):
339ra71
.
26.
Legoff
J
,
Resche-Rigon
M
,
Bouquet
J
, et al
.
The eukaryotic gut virome in hematopoietic stem cell transplantation: new clues in enteric graft-versus-host disease
.
Nat Med
.
2017
;
23
(
9
):
1080
-
1085
.
27.
Shono
Y
,
van den Brink
MRM
.
Gut microbiota injury in allogeneic haematopoietic stem cell transplantation
.
Nat Rev Cancer
.
2018
;
18
(
5
):
283
-
295
.
28.
Hanash
AM
,
Dudakov
JA
,
Hua
G
, et al
.
Interleukin-22 protects intestinal stem cells from immune-mediated tissue damage and regulates sensitivity to graft versus host disease
.
Immunity
.
2012
;
37
(
2
):
339
-
350
.
29.
Fu
YY
,
Egorova
A
,
Sobieski
C
, et al
.
T cell recruitment to the intestinal stem cell compartment drives immune-mediated intestinal damage after allogeneic transplantation
.
Immunity
.
2019
;
51
(
1
):
90
-
103.e3
.
30.
Zhao
D
,
Kim
YH
,
Jeong
S
, et al
.
Survival signal REG3α prevents crypt apoptosis to control acute gastrointestinal graft-versus-host disease
.
J Clin Invest
.
2018
;
128
(
11
):
4970
-
4979
.
31.
Mathewson
ND
,
Jenq
R
,
Mathew
AV
, et al
.
Gut microbiome-derived metabolites modulate intestinal epithelial cell damage and mitigate graft-versus-host disease [published correction appears in Nat Immunol. 2016;17(10):1235]
.
Nat Immunol
.
2016
;
17
(
5
):
505
-
513
.
32.
Fujiwara
H
,
Docampo
MD
,
Riwes
M
, et al
.
Microbial metabolite sensor GPR43 controls severity of experimental GVHD
.
Nat Commun
.
2018
;
9
(
1
):
3674
.
33.
Swimm
A
,
Giver
CR
,
DeFilipp
Z
, et al
.
Indoles derived from intestinal microbiota act via type I interferon signaling to limit graft-versus-host disease
.
Blood
.
2018
;
132
(
23
):
2506
-
2519
.
34.
Toubai
T
,
Rossi
C
,
Oravecz-Wilson
K
, et al
.
IAPs protect host target tissues from graft-versus-host disease in mice
.
Blood Adv
.
2017
;
1
(
19
):
1517
-
1532
.
35.
Toubai
T
,
Fujiwara
H
,
Rossi
C
, et al
.
Host NLRP6 exacerbates graft-versus-host disease independent of gut microbial composition
.
Nat Microbiol
.
2019
;
4
(
5
):
800
-
812
.
36.
Fischer
JC
,
Bscheider
M
,
Eisenkolb
G
, et al
.
RIG-I/MAVS and STING signaling promote gut integrity during irradiation- and immune-mediated tissue injury
.
Sci Transl Med
.
2017
;
9
(
386
):
eaag2513
.
37.
Fischer
JC
,
Bscheider
M
,
Eisenkolb
G
, et al
.
RIG-I/MAVS and STING signaling promote gut integrity during irradiation- and immune-mediated tissue injury
.
Sci Transl Med
.
2017
;
9
(
386
):
eaag2513
.
38.
Zeiser
R
,
Blazar
BR
.
Acute graft-versus-host disease - biologic process, prevention, and therapy
.
N Engl J Med
.
2017
;
377
(
22
):
2167
-
2179
.
39.
Hippen
KL
,
Aguilar
EG
,
Rhee
SY
,
Bolivar-Wagers
S
,
Blazar
BR
.
Distinct regulatory and effector T cell metabolic demands during graft-versus-host disease
.
Trends Immunol
.
2020
;
41
(
1
):
77
-
91
.
40.
Wu
SR
,
Reddy
P
.
Tissue tolerance: a distinct concept to control acute GVHD severity
.
Blood
.
2017
;
129
(
13
):
1747
-
1752
.
41.
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
;
9
(
9
):
1144
-
1150
.
42.
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
(
10
):
3493
-
3499
.
43.
Barnes
PJ
,
Adcock
IM
.
Glucocorticoid resistance in inflammatory diseases
.
Lancet
.
2009
;
373
(
9678
):
1905
-
1917
.
44.
Cain
DW
,
Cidlowski
JA
.
Immune regulation by glucocorticoids
.
Nat Rev Immunol
.
2017
;
17
(
4
):
233
-
247
.
45.
Vandewalle
J
,
Luypaert
A
,
De Bosscher
K
,
Libert
C
.
Therapeutic mechanisms of glucocorticoids
.
Trends Endocrinol Metab
.
2018
;
29
(
1
):
42
-
54
.
46.
Groeneweg
FL
,
Karst
H
,
de Kloet
ER
,
Joëls
M
.
Rapid non-genomic effects of corticosteroids and their role in the central stress response
.
J Endocrinol
.
2011
;
209
(
2
):
153
-
167
.
47.
Vodanovic-Jankovic
S
,
Hari
P
,
Jacobs
P
,
Komorowski
R
,
Drobyski
WR
.
NF-kappaB as a target for the prevention of graft-versus-host disease: comparative efficacy of bortezomib and PS-1145
.
Blood
.
2006
;
107
(
2
):
827
-
834
.
48.
MacDonald
KP
,
Kuns
RD
,
Rowe
V
, et al
.
Effector and regulatory T-cell function is differentially regulated by RelB within antigen-presenting cells during GVHD
.
Blood
.
2007
;
109
(
11
):
5049
-
5057
.
49.
Ogawa
S
,
Lozach
J
,
Benner
C
, et al
.
Molecular determinants of crosstalk between nuclear receptors and toll-like receptors
.
Cell
.
2005
;
122
(
5
):
707
-
721
.
50.
Guiducci
C
,
Gong
M
,
Xu
Z
, et al
.
TLR recognition of self nucleic acids hampers glucocorticoid activity in lupus
.
Nature
.
2010
;
465
(
7300
):
937
-
941
.
51.
Kitajima
T
,
Ariizumi
K
,
Bergstresser
PR
,
Takashima
A
.
A novel mechanism of glucocorticoid-induced immune suppression: the inhibiton of T cell-mediated terminal maturation of a murine dendritic cell line
.
J Clin Invest
.
1996
;
98
(
1
):
142
-
147
.
52.
Werb
Z
,
Foley
R
,
Munck
A
.
Interaction of glucocorticoids with macrophages. Identification of glucocorticoid receptors in monocytes and macrophages
.
J Exp Med
.
1978
;
147
(
6
):
1684
-
1694
.
53.
Luther
C
,
Adamopoulou
E
,
Stoeckle
C
, et al
.
Prednisolone treatment induces tolerogenic dendritic cells and a regulatory milieu in myasthenia gravis patients
.
J Immunol
.
2009
;
183
(
2
):
841
-
848
.
54.
Ehrchen
J
,
Steinmüller
L
,
Barczyk
K
, et al
.
Glucocorticoids induce differentiation of a specifically activated, anti-inflammatory subtype of human monocytes
.
Blood
.
2007
;
109
(
3
):
1265
-
1274
.
55.
Szatmari
I
,
Nagy
L
.
Nuclear receptor signalling in dendritic cells connects lipids, the genome and immune function
.
EMBO J
.
2008
;
27
(
18
):
2353
-
2362
.
56.
Vacca
A
,
Felli
MP
,
Farina
AR
, et al
.
Glucocorticoid receptor-mediated suppression of the interleukin 2 gene expression through impairment of the cooperativity between nuclear factor of activated T cells and AP-1 enhancer elements
.
J Exp Med
.
1992
;
175
(
3
):
637
-
646
.
57.
Bianchi
M
,
Meng
C
,
Ivashkiv
LB
.
Inhibition of IL-2-induced Jak-STAT signaling by glucocorticoids
.
Proc Natl Acad Sci USA
.
2000
;
97
(
17
):
9573
-
9578
.
58.
Löwenberg
M
,
Tuynman
J
,
Bilderbeek
J
, et al
.
Rapid immunosuppressive effects of glucocorticoids mediated through Lck and Fyn
.
Blood
.
2005
;
106
(
5
):
1703
-
1710
.
59.
Kumamaru
E
,
Numakawa
T
,
Adachi
N
,
Kunugi
H
.
Glucocorticoid suppresses BDNF-stimulated MAPK/ERK pathway via inhibiting interaction of Shp2 with TrkB
.
FEBS Lett
.
2011
;
585
(
20
):
3224
-
3228
.
60.
Franchimont
D
,
Galon
J
,
Gadina
M
, et al
.
Inhibition of Th1 immune response by glucocorticoids: dexamethasone selectively inhibits IL-12-induced Stat4 phosphorylation in T lymphocytes
.
J Immunol
.
2000
;
164
(
4
):
1768
-
1774
.
61.
Liberman
AC
,
Refojo
D
,
Druker
J
, et al
.
The activated glucocorticoid receptor inhibits the transcription factor T-bet by direct protein-protein interaction
.
FASEB J
.
2007
;
21
(
4
):
1177
-
1188
.
62.
Ramírez
F
,
Fowell
DJ
,
Puklavec
M
,
Simmonds
S
,
Mason
D
.
Glucocorticoids promote a TH2 cytokine response by CD4+ T cells in vitro
.
J Immunol
.
1996
;
156
(
7
):
2406
-
2412
.
63.
Karagiannidis
C
,
Akdis
M
,
Holopainen
P
, et al
.
Glucocorticoids upregulate FOXP3 expression and regulatory T cells in asthma
.
J Allergy Clin Immunol
.
2004
;
114
(
6
):
1425
-
1433
.
64.
Yosef
N
,
Shalek
AK
,
Gaublomme
JT
, et al
.
Dynamic regulatory network controlling TH17 cell differentiation
.
Nature
.
2013
;
496
(
7446
):
461
-
468
.
65.
Bouazzaoui
A
,
Spacenko
E
,
Mueller
G
, et al
.
Steroid treatment alters adhesion molecule and chemokine expression in experimental acute graft-vs.-host disease of the intestinal tract
.
Exp Hematol
.
2011
;
39
(
2
):
238
-
249.e1
.
66.
Small
GR
,
Hadoke
PW
,
Sharif
I
, et al
.
Preventing local regeneration of glucocorticoids by 11beta-hydroxysteroid dehydrogenase type 1 enhances angiogenesis
.
Proc Natl Acad Sci USA
.
2005
;
102
(
34
):
12165
-
12170
.
67.
Wu
SR
,
Reddy
P
.
Regulating damage from sterile inflammation: a tale of two tolerances
.
Trends Immunol
.
2017
;
38
(
4
):
231
-
235
.
68.
Cidlowski
JA
,
Munck
A
.
Concanavalin A-induced glucocorticoid resistance in rat thymus cells: decreased cytoplasmic and nuclear receptor binding of dexamethasone
.
J Steroid Biochem
.
1976
;
7
(
11-12
):
1141
-
1145
.
69.
Rodriguez
JM
,
Monsalves-Alvarez
M
,
Henriquez
S
,
Llanos
MN
,
Troncoso
R
.
Glucocorticoid resistance in chronic diseases
.
Steroids
.
2016
;
115
:
182
-
192
.
70.
Zeiser
R
,
Nguyen
VH
,
Beilhack
A
, et al
.
Inhibition of CD4+ CD25+ regulatory T-cell function by calcineurin-dependent interleukin-2 production
.
Blood
.
2006
;
108
(
1
):
390
-
399
.
71.
Toubai
T
,
Rossi
C
,
Tawara
I
, et al
.
Murine models of steroid refractory graft-versus-host disease
.
Sci Rep
.
2018
;
8
(
1
):
12475
.
72.
Kappel
LW
,
Goldberg
GL
,
King
CG
, et al
.
IL-17 contributes to CD4-mediated graft-versus-host disease
.
Blood
.
2009
;
113
(
4
):
945
-
952
.
73.
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
(
6
):
1365
-
1374
.
74.
Banuelos
J
,
Shin
S
,
Cao
Y
, et al
.
BCL-2 protects human and mouse Th17 cells from glucocorticoid-induced apoptosis
.
Allergy
.
2016
;
71
(
5
):
640
-
650
.
75.
Ramesh
R
,
Kozhaya
L
,
McKevitt
K
, et al
.
Pro-inflammatory human Th17 cells selectively express P-glycoprotein and are refractory to glucocorticoids
.
J Exp Med
.
2014
;
211
(
1
):
89
-
104
.
76.
Muriglan
SJ
,
Ramirez-Montagut
T
,
Alpdogan
O
, et al
.
GITR activation induces an opposite effect on alloreactive CD4(+) and CD8(+) T cells in graft-versus-host disease
.
J Exp Med
.
2004
;
200
(
2
):
149
-
157
.
77.
Li
H
,
Kaiser
TK
,
Borschiwer
M
, et al
.
Glucocorticoid resistance of allogeneic T cells alters the gene expression profile in the inflamed small intestine of mice suffering from acute graft-versus-host disease
.
J Steroid Biochem Mol Biol
.
2019
;
195
:
105485
.
78.
Toubai
T
,
Tawara
I
,
Sun
Y
, et al
.
Induction of acute GVHD by sex-mismatched H-Y antigens in the absence of functional radiosensitive host hematopoietic-derived antigen-presenting cells
.
Blood
.
2012
;
119
(
16
):
3844
-
3853
.
79.
Imamura
M
,
Hashino
S
,
Kobayashi
H
, et al
.
Serum cytokine levels in bone marrow transplantation: synergistic interaction of interleukin-6, interferon-gamma, and tumor necrosis factor-alpha in graft-versus-host disease
.
Bone Marrow Transplant
.
1994
;
13
(
6
):
745
-
751
.
80.
Tawara
I
,
Koyama
M
,
Liu
C
, et al
.
Interleukin-6 modulates graft-versus-host responses after experimental allogeneic bone marrow transplantation
.
Clin Cancer Res
.
2011
;
17
(
1
):
77
-
88
.
81.
Chen
X
,
Das
R
,
Komorowski
R
, et al
.
Blockade of interleukin-6 signaling augments regulatory T-cell reconstitution and attenuates the severity of graft-versus-host disease
.
Blood
.
2009
;
114
(
4
):
891
-
900
.
82.
Spoerl
S
,
Mathew
NR
,
Bscheider
M
, et al
.
Activity of therapeutic JAK 1/2 blockade in graft-versus-host disease
.
Blood
.
2014
;
123
(
24
):
3832
-
3842
.
83.
Carniti
C
,
Gimondi
S
,
Vendramin
A
, et al
.
Pharmacologic inhibition of JAK1/JAK2 signaling reduces experimental murine acute GVHD while preserving GVT effects
.
Clin Cancer Res
.
2015
;
21
(
16
):
3740
-
3749
.
84.
Betts
BC
,
Bastian
D
,
Iamsawat
S
, et al
.
Targeting JAK2 reduces GVHD and xenograft rejection through regulation of T cell differentiation
.
Proc Natl Acad Sci USA
.
2018
;
115
(
7
):
1582
-
1587
.
85.
Zeiser
R
,
Burchert
A
,
Lengerke
C
, et al
.
Ruxolitinib in corticosteroid-refractory graft-versus-host disease after allogeneic stem cell transplantation: a multicenter survey
.
Leukemia
.
2015
;
29
(
10
):
2062
-
2068
.
86.
Molina
ML
,
Guerrero
J
,
Cidlowski
JA
,
Gatica
H
,
Goecke
A
.
LPS regulates the expression of glucocorticoid receptor α and β isoforms and induces a selective glucocorticoid resistance in vitro
.
J Inflamm (Lond)
.
2017
;
14
:
22
.
87.
Dendoncker
K
,
Timmermans
S
,
Vandewalle
J
, et al
.
TNF-α inhibits glucocorticoid receptor-induced gene expression by reshaping the GR nuclear cofactor profile [published correction appears in Proc Natl Acad Sci USA. 2019;116(31):15745]
.
Proc Natl Acad Sci USA
.
2019
;
116
(
26
):
12942
-
12951
.
88.
Pazdrak
K
,
Straub
C
,
Maroto
R
, et al
.
Cytokine-induced glucocorticoid resistance from eosinophil activation: protein phosphatase 5 modulation of glucocorticoid receptor phosphorylation and signaling
.
J Immunol
.
2016
;
197
(
10
):
3782
-
3791
.
89.
Rider
CF
,
Shah
S
,
Miller-Larsson
A
,
Giembycz
MA
,
Newton
R
.
Cytokine-induced loss of glucocorticoid function: effect of kinase inhibitors, long-acting β(2)-adrenoceptor [corrected] agonist and glucocorticoid receptor ligands [published correction appears in PLoS One. 2015;10(5):e0128728]
.
PLoS One
.
2015
;
10
(
1
):
e0116773
.
90.
Paugh
SW
,
Bonten
EJ
,
Savic
D
, et al
.
NALP3 inflammasome upregulation and CASP1 cleavage of the glucocorticoid receptor cause glucocorticoid resistance in leukemia cells
.
Nat Genet
.
2015
;
47
(
6
):
607
-
614
.
91.
Baake
T
,
Jörß
K
,
Suennemann
J
, et al
.
The glucocorticoid receptor in recipient cells keeps cytokine secretion in acute graft-versus-host disease at bay
.
Oncotarget
.
2018
;
9
(
21
):
15437
-
15450
.
92.
Norden
J
,
Pearce
KF
,
Irving
JAE
, et al
.
The influence of glucocorticoid receptor single nucleotide polymorphisms on outcome after haematopoietic stem cell transplantation
.
Int J Immunogenet
.
2018
;
45
(
5
):
247
-
256
.
93.
Luft
T
,
Conzelmann
M
,
Benner
A
, et al
.
Serum cytokeratin-18 fragments as quantitative markers of epithelial apoptosis in liver and intestinal graft-versus-host disease
.
Blood
.
2007
;
110
(
13
):
4535
-
4542
.
94.
Luft
T
,
Dietrich
S
,
Falk
C
, et al
.
Steroid-refractory GVHD: T-cell attack within a vulnerable endothelial system
.
Blood
.
2011
;
118
(
6
):
1685
-
1692
.
95.
Dietrich
S
,
Falk
CS
,
Benner
A
, et al
.
Endothelial vulnerability and endothelial damage are associated with risk of graft-versus-host disease and response to steroid treatment
.
Biol Blood Marrow Transplant
.
2013
;
19
(
1
):
22
-
27
.
96.
Wall
SA
,
Zhao
Q
,
Yearsley
M
, et al
.
Complement-mediated thrombotic microangiopathy as a link between endothelial damage and steroid-refractory GVHD
.
Blood Adv
.
2018
;
2
(
20
):
2619
-
2628
.
97.
Zeisbrich
M
,
Becker
N
,
Benner
A
, et al
.
Transplant-associated thrombotic microangiopathy is an endothelial complication associated with refractoriness of acute GvHD
.
Bone Marrow Transplant
.
2017
;
52
(
10
):
1399
-
1405
.
98.
Jenq
RR
,
Ubeda
C
,
Taur
Y
, et al
.
Regulation of intestinal inflammation by microbiota following allogeneic bone marrow transplantation
.
J Exp Med
.
2012
;
209
(
5
):
903
-
911
.
99.
Kakihana
K
,
Fujioka
Y
,
Suda
W
, et al
.
Fecal microbiota transplantation for patients with steroid-resistant acute graft-versus-host disease of the gut
.
Blood
.
2016
;
128
(
16
):
2083
-
2088
.
100.
DeFilipp
Z
,
Peled
JU
,
Li
S
, et al
.
Third-party fecal microbiota transplantation following allo-HCT reconstitutes microbiome diversity
.
Blood Adv
.
2018
;
2
(
7
):
745
-
753
.
101.
Qi
X
,
Li
X
,
Zhao
Y
, et al
.
Treating steroid refractory intestinal acute graft-vs.-host disease with fecal microbiota transplantation: a pilot study
.
Front Immunol
.
2018
;
9
:
2195
.
102.
Martin
PJ
,
Schoch
G
,
Fisher
L
, et al
.
A retrospective analysis of therapy for acute graft-versus-host disease: initial treatment
.
Blood
.
1990
;
76
(
8
):
1464
-
1472
.
103.
MacMillan
ML
,
Weisdorf
DJ
,
Davies
SM
, et al
.
Early antithymocyte globulin therapy improves survival in patients with steroid-resistant acute graft-versus-host disease
.
Biol Blood Marrow Transplant
.
2002
;
8
(
1
):
40
-
46
.
104.
Ali
AM
,
DiPersio
JF
,
Schroeder
MA
.
The role of biomarkers in the diagnosis and risk stratification of acute graft-versus-host disease: a systematic review
.
Biol Blood Marrow Transplant
.
2016
;
22
(
9
):
1552
-
1564
.
105.
Paczesny
S
.
Biomarkers for posttransplantation outcomes
.
Blood
.
2018
;
131
(
20
):
2193
-
2204
.
106.
Ho
VT
,
Cutler
C
.
Current and novel therapies in acute GVHD
.
Best Pract Res Clin Haematol
.
2008
;
21
(
2
):
223
-
237
.
107.
MacMillan
ML
,
Robin
M
,
Harris
AC
, et al
.
A refined risk score for acute graft-versus-host disease that predicts response to initial therapy, survival, and transplant-related mortality
.
Biol Blood Marrow Transplant
.
2015
;
21
(
4
):
761
-
767
.
108.
Melson
J
,
Jakate
S
,
Fung
H
,
Arai
S
,
Keshavarzian
A
.
Crypt loss is a marker of clinical severity of acute gastrointestinal graft-versus-host disease
.
Am J Hematol
.
2007
;
82
(
10
):
881
-
886
.
109.
Levine
JE
,
Huber
E
,
Hammer
ST
, et al
.
Low Paneth cell numbers at onset of gastrointestinal graft-versus-host disease identify patients at high risk for nonrelapse mortality
.
Blood
.
2013
;
122
(
8
):
1505
-
1509
.
110.
Major-Monfried
H
,
Renteria
AS
,
Pawarode
A
, et al
.
MAGIC biomarkers predict long-term outcomes for steroid-resistant acute GVHD
.
Blood
.
2018
;
131
(
25
):
2846
-
2855
.
111.
Ferrara
JLM
,
Chaudhry
MS
.
GVHD: biology matters
.
Blood Adv
.
2018
;
2
(
22
):
3411
-
3417
.
112.
Ponce
DM
,
Hilden
P
,
Mumaw
C
, et al
.
High day 28 ST2 levels predict for acute graft-versus-host disease and transplant-related mortality after cord blood transplantation
.
Blood
.
2015
;
125
(
1
):
199
-
205
.
113.
Vander Lugt
MT
,
Braun
TM
,
Hanash
S
, et al
.
ST2 as a marker for risk of therapy-resistant graft-versus-host disease and death
.
N Engl J Med
.
2013
;
369
(
6
):
529
-
539
.
114.
Zhang
J
,
Ramadan
AM
,
Griesenauer
B
, et al
.
ST2 blockade reduces sST2-producing T cells while maintaining protective mST2-expressing T cells during graft-versus-host disease
.
Sci Transl Med
.
2015
;
7
(
308
):
308ra160
.
115.
Azramezani Kopi
T
,
Shahrokh
S
,
Mirzaei
S
,
Asadzadeh Aghdaei
H
,
Amini Kadijani
A
.
The role of serum calprotectin as a novel biomarker in inflammatory bowel diseases: a review study
.
Gastroenterol Hepatol Bed Bench
.
2019
;
12
(
3
):
183
-
189
.
116.
O’Meara
A
,
Kapel
N
,
Xhaard
A
, et al
.
Fecal calprotectin and α1-antitrypsin dynamics in gastrointestinal GvHD
.
Bone Marrow Transplant
.
2015
;
50
(
8
):
1105
-
1109
.
117.
Adam
B
,
Koldehoff
M
,
Ditschkowski
M
, et al
.
Endoscopic and histological findings are predicted by fecal calprotectin in acute intestinal graft-versus-host-disease
.
Dig Dis Sci
.
2016
;
61
(
7
):
2019
-
2026
.
118.
Rodriguez-Otero
P
,
Porcher
R
,
Peffault de Latour
R
, et al
.
Fecal calprotectin and alpha-1 antitrypsin predict severity and response to corticosteroids in gastrointestinal graft-versus-host disease
.
Blood
.
2012
;
119
(
24
):
5909
-
5917
.
119.
Martin
PJ
,
Inamoto
Y
,
Flowers
ME
,
Carpenter
PA
.
Secondary treatment of acute graft-versus-host disease: a critical review
.
Biol Blood Marrow Transplant
.
2012
;
18
(
7
):
982
-
988
.
120.
Macmillan
ML
,
Couriel
D
,
Weisdorf
DJ
, 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
(
6
):
2657
-
2662
.
121.
Martin
PJ
,
Bachier
CR
,
Klingemann
HG
, et al
.
Endpoints for clinical trials testing treatment of acute graft-versus-host disease: a joint statement
.
Biol Blood Marrow Transplant
.
2009
;
15
(
7
):
777
-
784
.
122.
MacMillan
ML
,
DeFor
TE
,
Weisdorf
DJ
.
The best endpoint for acute GVHD treatment trials
.
Blood
.
2010
;
115
(
26
):
5412
-
5417
.
123.
Martin
PJ
.
Study design and endpoints in graft-versus-host disease
.
Best Pract Res Clin Haematol
.
2008
;
21
(
2
):
357
-
372
.
124.
Van Lint
MT
,
Uderzo
C
,
Locasciulli
A
, 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
(
7
):
2288
-
2293
.
125.
Meyer
EH
,
Hsu
AR
,
Liliental
J
, et al
.
A distinct evolution of the T-cell repertoire categorizes treatment refractory gastrointestinal acute graft-versus-host disease
.
Blood
.
2013
;
121
(
24
):
4955
-
4962
.
126.
Bolaños-Meade
J
,
Jacobsohn
DA
,
Margolis
J
, et al
.
Pentostatin in steroid-refractory acute graft-versus-host disease
.
J Clin Oncol
.
2005
;
23
(
12
):
2661
-
2668
.
127.
Mayer
J
,
Krejcí
M
,
Doubek
M
, et al
.
Pulse cyclophosphamide for corticosteroid-refractory graft-versus-host disease
.
Bone Marrow Transplant
.
2005
;
35
(
7
):
699
-
705
.
128.
Weisdorf
D
,
Haake
R
,
Blazar
B
, et al
.
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
(
4
):
1024
-
1030
.
129.
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
(
3
):
155
-
160
.
130.
Holler
E
,
Butzhammer
P
,
Schmid
K
, et al
.
Metagenomic analysis of the stool microbiome in patients receiving allogeneic stem cell transplantation: loss of diversity is associated with use of systemic antibiotics and more pronounced in gastrointestinal graft-versus-host disease
.
Biol Blood Marrow Transplant
.
2014
;
20
(
5
):
640
-
645
.
131.
Vydra
J
,
Shanley
RM
,
George
I
, et al
.
Enterococcal bacteremia is associated with increased risk of mortality in recipients of allogeneic hematopoietic stem cell transplantation
.
Clin Infect Dis
.
2012
;
55
(
6
):
764
-
770
.
132.
Perales
MA
,
Ishill
N
,
Lomazow
WA
, et al
.
Long-term follow-up of patients treated with daclizumab for steroid-refractory acute graft-vs-host disease
.
Bone Marrow Transplant
.
2007
;
40
(
5
):
481
-
486
.
133.
García-Cadenas
I
,
Rivera
I
,
Martino
R
, et al
.
Patterns of infection and infection-related mortality in patients with steroid-refractory acute graft versus host disease
.
Bone Marrow Transplant
.
2017
;
52
(
1
):
107
-
113
.
134.
Schneiderman
J
.
Extracorporeal photopheresis: cellular therapy for the treatment of acute and chronic graft-versus-host disease
.
Hematology Am Soc Hematol Educ Program
.
2017
;
2017
:
639
-
644
.
135.
Abu-Dalle
I
,
Reljic
T
,
Nishihori
T
, et al
.
Extracorporeal photopheresis in steroid-refractory acute or chronic graft-versus-host disease: results of a systematic review of prospective studies
.
Biol Blood Marrow Transplant
.
2014
;
20
(
11
):
1677
-
1686
.
136.
Groth
C
,
van Groningen
LFJ
,
Matos
TR
, et al
.
Phase I/II trial of a combination of anti-CD3/CD7 immunotoxins for steroid-refractory acute graft-versus-host disease
.
Biol Blood Marrow Transplant
.
2019
;
25
(
4
):
712
-
719
.
137.
Carpenter
PA
,
Appelbaum
FR
,
Corey
L
, et al
.
A humanized non-FcR-binding anti-CD3 antibody, visilizumab, for treatment of steroid-refractory acute graft-versus-host disease
.
Blood
.
2002
;
99
(
8
):
2712
-
2719
.
138.
van Oosterhout
YV
,
van Emst
L
,
Schattenberg
AV
, et al
.
A combination of anti-CD3 and anti-CD7 ricin A-immunotoxins for the in vivo treatment of acute graft versus host disease
.
Blood
.
2000
;
95
(
12
):
3693
-
3701
.
139.
Chen
YB
,
McDonough
S
,
Hasserjian
R
, et al
.
Expression of CD30 in patients with acute graft-versus-host disease
.
Blood
.
2012
;
120
(
3
):
691
-
696
.
140.
Chen
YB
,
Perales
MA
,
Li
S
, et al
.
Phase 1 multicenter trial of brentuximab vedotin for steroid-refractory acute graft-versus-host disease
.
Blood
.
2017
;
129
(
24
):
3256
-
3261
.
141.
Kekre
N
,
Kim
HT
,
Ho
VT
, et al
.
Phase II trial of natalizumab (Tysabri (R)) with corticosteroids as initial treatment of gastrointestinal acute graft versus host disease [abstract]
.
Biol Blood Marrow Transplant
.
2018
;
24
(
suppl 3
):
S69
.
142.
Fløisand
Y
,
Lundin
KEA
,
Lazarevic
V
, et al
.
Targeting integrin α4β7 in steroid-refractory intestinal graft-versus-host disease
.
Biol Blood Marrow Transplant
.
2017
;
23
(
1
):
172
-
175
.
143.
Fløisand
Y
,
Lazarevic
VL
,
Maertens
J
, et al
.
Safety and effectiveness of vedolizumab in patients with steroid-refractory gastrointestinal acute graft-versus-host disease: a retrospective record review
.
Biol Blood Marrow Transplant
.
2019
;
25
(
4
):
720
-
727
.
144.
Teshima
T
,
Reddy
P
,
Zeiser
R
.
Reprint of: acute graft-versus-host disease: novel biological insights
.
Biol Blood Marrow Transplant
.
2016
;
22
(
suppl 3
):
S3
-
S8
.
145.
Ganetsky
A
,
Frey
NV
,
Hexner
EO
, et al
.
Tocilizumab for the treatment of severe steroid-refractory acute graft-versus-host disease of the lower gastrointestinal tract
.
Bone Marrow Transplant
.
2019
;
54
(
2
):
212
-
217
.
146.
Roddy
JV
,
Haverkos
BM
,
McBride
A
, et al
.
Tocilizumab for steroid refractory acute graft-versus-host disease
.
Leuk Lymphoma
.
2016
;
57
(
1
):
81
-
85
.
147.
Drobyski
WR
,
Pasquini
M
,
Kovatovic
K
, et al
.
Tocilizumab for the treatment of steroid refractory graft-versus-host disease
.
Biol Blood Marrow Transplant
.
2011
;
17
(
12
):
1862
-
1868
.
148.
Betts
BC
,
St Angelo
ET
,
Kennedy
M
,
Young
JW
.
Anti-IL6-receptor-alpha (tocilizumab) does not inhibit human monocyte-derived dendritic cell maturation or alloreactive T-cell responses
.
Blood
.
2011
;
118
(
19
):
5340
-
5343
.
149.
Lindemans
CA
,
Calafiore
M
,
Mertelsmann
AM
, et al
.
Interleukin-22 promotes intestinal-stem-cell-mediated epithelial regeneration
.
Nature
.
2015
;
528
(
7583
):
560
-
564
.
150.
van Groningen
LF
,
Liefferink
AM
,
de Haan
AF
, et al
.
Combination therapy with inolimomab and etanercept for severe steroid-refractory acute graft-versus-host disease
.
Biol Blood Marrow Transplant
.
2016
;
22
(
1
):
179
-
182
.
151.
Langlais
D
,
Couture
C
,
Balsalobre
A
,
Drouin
J
.
The Stat3/GR interaction code: predictive value of direct/indirect DNA recruitment for transcription outcome
.
Mol Cell
.
2012
;
47
(
1
):
38
-
49
.
152.
Jagasia
M
,
Perales
MA
,
Schroeder
MA
, et al
.
Ruxolitinib for the treatment of steroid-refractory acute GVHD (REACH1): a multicenter, open-label phase 2 trial
.
Blood
.
2020
;
135
(
20
):
1739
-
1749
.
153.
Abedin
S
,
McKenna
E
,
Chhabra
S
, et al
.
Efficacy, toxicity, and infectious complications in ruxolitinib-treated patients with corticosteroid-refractory graft-versus-host disease after hematopoietic cell transplantation
.
Biol Blood Marrow Transplant
.
2019
;
25
(
8
):
1689
-
1694
.
154.
Schroeder
MA
,
Khoury
HJ
,
Jagasia
M
, et al
.
A phase 1 trial of itacitinib, a selective JAK1 inhibitor, in patients with acute graft-versus-host disease
.
Blood Adv
.
2020
;
4
(
8
):
1656
-
1669
.
155.
Mócsai
A
,
Ruland
J
,
Tybulewicz
VL
.
The SYK tyrosine kinase: a crucial player in diverse biological functions
.
Nat Rev Immunol
.
2010
;
10
(
6
):
387
-
402
.
156.
Leonhardt
F
,
Zirlik
K
,
Buchner
M
, et al
.
Spleen tyrosine kinase (Syk) is a potent target for GvHD prevention at different cellular levels
.
Leukemia
.
2012
;
26
(
7
):
1617
-
1629
.
157.
Steinbach
G
,
Hockenbery
DM
,
Huls
G
, et al
.
Pilot study of lithium to restore intestinal barrier function in severe graft-versus-host disease
.
PLoS One
.
2017
;
12
(
8
):
e0183284
.
158.
Krishnamurthy
N
,
Kurzrock
R
.
Targeting the Wnt/beta-catenin pathway in cancer: update on effectors and inhibitors
.
Cancer Treat Rev
.
2018
;
62
:
50
-
60
.
159.
Tawara
I
,
Sun
Y
,
Lewis
EC
, et al
.
Alpha-1-antitrypsin monotherapy reduces graft-versus-host disease after experimental allogeneic bone marrow transplantation
.
Proc Natl Acad Sci USA
.
2012
;
109
(
2
):
564
-
569
.
160.
Marcondes
AM
,
Karoopongse
E
,
Lesnikova
M
, et al
.
α-1-Antitrypsin (AAT)-modified donor cells suppress GVHD but enhance the GVL effect: a role for mitochondrial bioenergetics
.
Blood
.
2014
;
124
(
18
):
2881
-
2891
.
161.
Brennan
TV
,
Lin
L
,
Huang
X
, et al
.
Heparan sulfate, an endogenous TLR4 agonist, promotes acute GVHD after allogeneic stem cell transplantation
.
Blood
.
2012
;
120
(
14
):
2899
-
2908
.
162.
Magenau
JM
,
Goldstein
SC
,
Peltier
D
, et al
.
α1-Antitrypsin infusion for treatment of steroid-resistant acute graft-versus-host disease
.
Blood
.
2018
;
131
(
12
):
1372
-
1379
.
163.
Marcondes
AM
,
Hockenbery
D
,
Lesnikova
M
, et al
.
Response of steroid-refractory acute GVHD to α1-antitrypsin
.
Biol Blood Marrow Transplant
.
2016
;
22
(
9
):
1596
-
1601
.
164.
Blazar
BR
,
MacDonald
KPA
,
Hill
GR
.
Immune regulatory cell infusion for graft-versus-host disease prevention and therapy
.
Blood
.
2018
;
131
(
24
):
2651
-
2660
.
165.
Koreth
J
,
Matsuoka
K
,
Kim
HT
, et al
.
Interleukin-2 and regulatory T cells in graft-versus-host disease
.
N Engl J Med
.
2011
;
365
(
22
):
2055
-
2066
.
166.
Kennedy-Nasser
AA
,
Ku
S
,
Castillo-Caro
P
, et al
.
Ultra low-dose IL-2 for GVHD prophylaxis after allogeneic hematopoietic stem cell transplantation mediates expansion of regulatory T cells without diminishing antiviral and antileukemic activity
.
Clin Cancer Res
.
2014
;
20
(
8
):
2215
-
2225
.
167.
Mavers
M
,
Maas-Bauer
K
,
Negrin
RS
.
Invariant natural killer T cells as suppressors of graft-versus-host disease in allogeneic hematopoietic stem cell transplantation
.
Front Immunol
.
2017
;
8
:
900
.
168.
Wolf
D
,
Barreras
H
,
Bader
CS
, et al
.
Marked in vivo donor regulatory T cell expansion via interleukin-2 and TL1A-Ig stimulation ameliorates graft-versus-host disease but preserves graft-versus-leukemia in recipients after hematopoietic stem cell transplantation
.
Biol Blood Marrow Transplant
.
2017
;
23
(
5
):
757
-
766
.
169.
Hashmi
S
,
Ahmed
M
,
Murad
MH
, et al
.
Survival after mesenchymal stromal cell therapy in steroid-refractory acute graft-versus-host disease: systematic review and meta-analysis
.
Lancet Haematol
.
2016
;
3
(
1
):
e45
-
e52
.
170.
Galleu
A
,
Riffo-Vasquez
Y
,
Trento
C
, et al
.
Apoptosis in mesenchymal stromal cells induces in vivo recipient-mediated immunomodulation
.
Sci Transl Med
.
2017
;
9
(
416
):
eaam7828
.
171.
Gooley
TA
,
Chien
JW
,
Pergam
SA
, et al
.
Reduced mortality after allogeneic hematopoietic-cell transplantation
.
N Engl J Med
.
2010
;
363
(
22
):
2091
-
2101
.
172.
Bolaños-Meade
J
,
Reshef
R
,
Fraser
R
, et al
.
Three prophylaxis regimens (tacrolimus, mycophenolate mofetil, and cyclophosphamide; tacrolimus, methotrexate, and bortezomib; or tacrolimus, methotrexate, and maraviroc) versus tacrolimus and methotrexate for prevention of graft-versus-host disease with haemopoietic cell transplantation with reduced-intensity conditioning: a randomised phase 2 trial with a non-randomised contemporaneous control group (BMT CTN 1203)
.
Lancet Haematol
.
2019
;
6
(
3
):
e132
-
e143
.
173.
Toubai
T
,
Hou
G
,
Mathewson
N
, et al
.
Siglec-G-CD24 axis controls the severity of graft-versus-host disease in mice
.
Blood
.
2014
;
123
(
22
):
3512
-
3523
.
174.
Toubai
T
,
Rossi
C
,
Oravecz-Wilson
K
, et al
.
Siglec-G represses DAMP-mediated effects on T cells
.
JCI Insight
.
2017
;
2
(
14
):
92293
.
175.
Hayase
E
,
Hashimoto
D
,
Nakamura
K
, et al
.
R-Spondin1 expands Paneth cells and prevents dysbiosis induced by graft-versus-host disease
.
J Exp Med
.
2017
;
214
(
12
):
3507
-
3518
.
176.
Danylesko
I
,
Bukauskas
A
,
Paulson
M
, et al
.
Anti-α4β7 integrin monoclonal antibody (vedolizumab) for the treatment of steroid-resistant severe intestinal acute graft-versus-host disease
.
Bone Marrow Transplant
.
2019
;
54
(
7
):
987
-
993
.
177.
Khandelwal
P
,
Teusink-Cross
A
,
Davies
SM
, et al
.
Ruxolitinib as salvage therapy in steroid-refractory acute graft-versus-host disease in pediatric hematopoietic stem cell transplant patients
.
Biol Blood Marrow Transplant
.
2017
;
23
(
7
):
1122
-
1127
.
178.
González Vicent
M
,
Molina
B
,
González de Pablo
J
,
Castillo
A
,
Díaz
MA
.
Ruxolitinib treatment for steroid refractory acute and chronic graft vs host disease in children: clinical and immunological results
.
Am J Hematol
.
2019
;
94
(
3
):
319
-
326
.
Sign in via your Institution