Allogeneic hematopoietic stem cell transplantation (alloSCT) is an important curative therapy for high-risk hematological malignancies, but the development of severe and/or steroid-refractory acute graft-versus-host disease (aGVHD) remains a significant limitation to optimal outcomes. New approaches to prevent and treat aGVHD remain an unmet need that can be best addressed by understanding the complex disease pathophysiology. It is now clear that chemoradiotherapy used prior to alloSCT induces the release of endogenous alarmins (eg, HMGB-1, ATP, IL-1α, IL-33) from recipient tissue. Exogenous pathogen-derived molecules (eg, lipopolysaccharide, nucleic acids) also translocate from the gastrointestinal tract lumen. Together, these danger signals activate antigen-presenting cells (APCs) to efficiently present alloantigen to donor T cells while releasing cytokines (eg, interleukin-12 [IL-12], IL-23, IL-6, IL-27, IL-10, transforming growth factor-β) that expand and differentiate both pathogenic and regulatory donor T cells. Concurrent costimulatory signals at the APC–T-cell interface (eg, CD80/CD86-CD28, CD40-CD40L, OX40L-OX40, CD155/CD112-DNAM-1) and subsequent coinhibitory signals (eg, CD80/CD86-CTLA4, PDL1/2-PD1, CD155/CD112-TIGIT) are critical to the acquisition of effector T-cell function and ensuing secretion of pathogenic cytokines (eg, IL-17, interferon-γ, tissue necrosis factor, granulocyte-macrophage colony-stimulating factor) and cytolytic degranulation pathway effectors (eg, perforin/granzyme). This review focuses on the combination of cytokine and costimulatory networks at the T-cell surface that culminates in effector function and subsequent aGVHD in target tissue. Together, these pathways now represent robust and clinically tractable targets for preventing the initiation of deleterious immunity after alloSCT.

Allogeneic hematopoietic stem cell transplantation (alloSCT) remains a curative therapy for high-risk hematological malignancies but risks of transplant-related mortality (TRM), largely from graft-versus-host disease (GVHD) and infection, remain. Systemic steroids, the standard treatment of acute GVHD (aGVHD), increase the risk of infection,1  and the prognosis of steroid-refractory aGVHD remains poor with a 2-year overall survival of 25%.2  Thus, preventing severe and steroid-refractory aGVHD is crucial to improving survival.

The pathophysiology of aGVHD generally can be thought of in 3 phases.3  In phase 1, conditioning chemoradiotherapy induces tissue damage and the release of inflammatory cytokines (eg, interleukin-6 [IL-6], tissue necrosis factor [TNF]), alarmins (eg, high-mobility group box-1 [HMGB1], IL-1α, IL-33), and pathogen-associated molecular pattern (PAMP) molecules that enhance alloantigen presentation by host antigen-presenting cells (APCs). In phase 2, transplanted donor CD8 and CD4 T cells recognize alloantigens presented within HLA class I and II, respectively, activate, expand, and differentiate into effector T cells. In phase 3, cytokines (eg, interferon-γ [IFN-γ], TNF, IL-2, IL-17) secreted by differentiated effector T cells and phagocytes (including monocytes and macrophages) contribute to target tissue apoptosis.4 

We have focused this review on costimulation (both cell surface ligands and soluble cytokines) at the APC–T-cell synapse as outlined in detail in “The role of costimulatory and coinhibitory pathways in aGVHD.” We provide context for the cytokine-dependent activation of the APCs prior to this in “The initiation of inflammation by pretransplant conditioning,”, together with a brief context for the role of (T-cell–derived) cytokines in mediating tissue pathology during GVHD in “The impact of T-cell–derived cytokines on GVHD.” The review focuses on donor T cells, and we direct the reader to excellent reviews on the subject of natural killer cell activation and function after alloSCT.5,6 

Multiple signals are required in the second phase of the GVHD process to enable full-donor T-cell activation and the acquisition of effector function. Signal 1 to the T cell involves the ligation of the T-cell receptor (TCR) by major histocompatibility complex (MHC)–peptide complex on the recipient APC surface. Signal 2 involves costimulation delivered by interaction of costimulatory molecules on the recipient APC surface to their cognate ligand expressed by the donor T cell (eg, CD80/CD86-CD28, CD40-CD40L). A third signal is mediated by cytokines secreted by APCs and T cells (eg, IL-2, IL-12, IL-23, IL-6)7  that bind their relevant receptor on donor T cells to induce proliferation and differentiation into effector T cells. These pathways are depicted pictorially in Figure 1.

Figure 1.

The cytokine and costimulatory pathways involved in acute GVHD. Chemoradiotherapy used in conditioning liberates a number of DAMP and PAMP signals that contribute to hematopoietic and nonhematopoietic APC activation (antigen presentation, costimulation molecule expression, and cytokine secretion). Allogeneic peptides presented in MHC are recognized by the TCR on conventional donor T cells in conjunction with a suite of costimulatory molecules (CD40-CD40L, CD80/86-CD28, OX40L-OX40, ICOSL-ICOS) that together with appropriate cytokine signals (eg, IL-12, IL-6, IL-23) drives T-cell differentiation and their secretion of effector cytokines (eg, IFN-γ, IL-17, TNF, GM-CSF) that invoke local inflammation to mediate target tissue apoptosis directly and by recruitment of additional immune effector cells. The activation of Tregs involves MHC/peptide-TCR interactions, costimulatory and coinhibitory molecule engagement (eg, TIGIT and CTLA-4), and additional cytokine signals from IL-33 that enhance cytokine secretion mediating regulatory and repair function (eg, IL-10/TGF-β and amphiregulin, respectively). Over time, many of these coinhibitory molecules are expressed by conventional T effector cells.

Figure 1.

The cytokine and costimulatory pathways involved in acute GVHD. Chemoradiotherapy used in conditioning liberates a number of DAMP and PAMP signals that contribute to hematopoietic and nonhematopoietic APC activation (antigen presentation, costimulation molecule expression, and cytokine secretion). Allogeneic peptides presented in MHC are recognized by the TCR on conventional donor T cells in conjunction with a suite of costimulatory molecules (CD40-CD40L, CD80/86-CD28, OX40L-OX40, ICOSL-ICOS) that together with appropriate cytokine signals (eg, IL-12, IL-6, IL-23) drives T-cell differentiation and their secretion of effector cytokines (eg, IFN-γ, IL-17, TNF, GM-CSF) that invoke local inflammation to mediate target tissue apoptosis directly and by recruitment of additional immune effector cells. The activation of Tregs involves MHC/peptide-TCR interactions, costimulatory and coinhibitory molecule engagement (eg, TIGIT and CTLA-4), and additional cytokine signals from IL-33 that enhance cytokine secretion mediating regulatory and repair function (eg, IL-10/TGF-β and amphiregulin, respectively). Over time, many of these coinhibitory molecules are expressed by conventional T effector cells.

Close modal

The initiation of inflammation by pretransplant conditioning

Current conditioning therapies employ cytotoxic chemoradiotherapy to eradicate residual malignant cells, generate space for donor cell engraftment, and prevent immunological graft rejection. Although hematopoiesis-targeted conditioning may reduce tissue damage,8-12  in clinical practice, cytotoxic therapy (myeloablative or reduced intensity conditioning) is still required for stable engraftment. Cytotoxic conditioning provokes the release of damage associated molecular patterns (DAMPs) and/or alarmin proteins from damaged tissues and promotes the translocation of PAMP molecules across impaired mucosal barriers, particularly within the gastrointestinal (GI) tract. DAMPs include nucleic acids, intracellular proteins (eg, HMGB1, heat shock proteins, histone, actin, adenosine triphosphate [ATP], and reactive oxygen species), and extracellular proteins (eg, hyaluronic acid and biglycan), which bind to sensing receptors, including toll-like receptors (TLRs), NOD-like receptors (NLRs), retinoic acid inducible gene I (RIG-I)-like receptors, C-type lectin receptors, and the receptor for advanced glycation end products (RAGE).13  Alarmins are constitutively expressed endogenous molecules, including some cytokines (IL-1α and IL-33), that are released by degranulation from injured or dead cells.14  PAMPs include bacterial (eg, lipopolysaccharide [LPS], lipoproteins, peptidoglycan, and flagellin), fungal components (eg, β-glucans, α-mannans), and viral nucleic acids.15  In animal models, GVHD has been shown to be exacerbated by the following DAMPs/PAMPs or their signaling pathways: LPS,16  viral/bacterial DNA-mimicking CpG oligodeoxynucleotides,17  α-mannans,18  ATP,19  uric acid,20  NLRP3,20  the ASC inflammasome adaptor protein,20  RAGE21  which is the receptor for HMGB1 and the TLR-signaling molecules, MyD88 and TRIF.21,22  These tissue and microbiota-derived molecules stimulate recipient cells to secrete further cytokines (eg, TNF, IL-1, IL-6, IL-33, IL-12, IL-23, type1 IFN) and chemokines (eg, CCL5), which enhance alloantigen presentation and the expression of costimulatory molecules and cytokines by recipient APCs. Some of the critical and clinically tractable pathways are described in more detail below and in Table 1.

Table 1.

Potential cytokine inhibition/agonists applicable to the prevention of aGVHD

TargetReagentDisease settingClinical trial status/outcomeClinical Trial IDReference
TNF TNFR-Fc GVHD prophylaxis Phase 2, II-IV aGVHD 46%, but steroid-refractory GI GVHD is 23% NCT00639717 26,27  
TNFR-Fc GVHD treatment (as initial therapy) Phase 2, TNFR-Fc was inferior to MMF with lower CR NCT00224874 25  
IL-1β Human rIL-1Ra GVHD prophylaxis Phase 3, no reduction in the incidence of aGVHD  31  
IL-33R Anti-ST2 Ab COPD Phase 2, active, not recruiting NCT03615040  
IL-6R Anti-IL-6 receptor Ab GVHD prophylaxis Phase 1/2, low rates grade II-IV aGVHD NCT02206035* 24,124 * 
IL-12/23 p40 Anti-IL-12/23 p40 Ab GVHD prophylaxis Phase 1/2, improved OS and GRFS NCT01713400 134  
IL-23 p19 Anti-IL-23p19 Ab Psoriasis Phase 3, improved efficacy compared with anti-TNFα Ab NCT02207231 135  
NCT02207244 
Ulcerative colitis Phase 2/3, active, recruiting NCT04033445  
Phase 2, active, recruiting NCT03662542 
Crohn disease Phase 2/3, active, recruiting NCT03466411  
IL-17A Anti-IL-17A Ab Active rheumatoid arthritis Phase 3, secukinumab was inferior to abatacept with lower response rates NCT01350804 154  
Ankylosing spondylitis (AS) Phase 3, improved AS symptoms and low mean progression in spinal radiographic change NCT01358175 153  
Plaque psoriasis Phase 3, effective in 2 randomized trials NCT01365455 152  
NCT01358578 
IL-2 Anti-CD25 Ab GVHD prophylaxis Phase 3, no reduction in aGVHD  144  
IL-22 Human IL-22 IgG2-FC GVHD treatment (II to IV lower GI tract GVHD) Phase 2a, active, not recruiting NCT02406651  
IL-2 Human rIL-2 GVHD prophylaxis Phase 2, eliminated grade II-IV aGVHD (0% vs 13%) NCT00539695 142  
Phase 2, no reduction in the aGVHD NCT01927120 143  
TargetReagentDisease settingClinical trial status/outcomeClinical Trial IDReference
TNF TNFR-Fc GVHD prophylaxis Phase 2, II-IV aGVHD 46%, but steroid-refractory GI GVHD is 23% NCT00639717 26,27  
TNFR-Fc GVHD treatment (as initial therapy) Phase 2, TNFR-Fc was inferior to MMF with lower CR NCT00224874 25  
IL-1β Human rIL-1Ra GVHD prophylaxis Phase 3, no reduction in the incidence of aGVHD  31  
IL-33R Anti-ST2 Ab COPD Phase 2, active, not recruiting NCT03615040  
IL-6R Anti-IL-6 receptor Ab GVHD prophylaxis Phase 1/2, low rates grade II-IV aGVHD NCT02206035* 24,124 * 
IL-12/23 p40 Anti-IL-12/23 p40 Ab GVHD prophylaxis Phase 1/2, improved OS and GRFS NCT01713400 134  
IL-23 p19 Anti-IL-23p19 Ab Psoriasis Phase 3, improved efficacy compared with anti-TNFα Ab NCT02207231 135  
NCT02207244 
Ulcerative colitis Phase 2/3, active, recruiting NCT04033445  
Phase 2, active, recruiting NCT03662542 
Crohn disease Phase 2/3, active, recruiting NCT03466411  
IL-17A Anti-IL-17A Ab Active rheumatoid arthritis Phase 3, secukinumab was inferior to abatacept with lower response rates NCT01350804 154  
Ankylosing spondylitis (AS) Phase 3, improved AS symptoms and low mean progression in spinal radiographic change NCT01358175 153  
Plaque psoriasis Phase 3, effective in 2 randomized trials NCT01365455 152  
NCT01358578 
IL-2 Anti-CD25 Ab GVHD prophylaxis Phase 3, no reduction in aGVHD  144  
IL-22 Human IL-22 IgG2-FC GVHD treatment (II to IV lower GI tract GVHD) Phase 2a, active, not recruiting NCT02406651  
IL-2 Human rIL-2 GVHD prophylaxis Phase 2, eliminated grade II-IV aGVHD (0% vs 13%) NCT00539695 142  
Phase 2, no reduction in the aGVHD NCT01927120 143  

COPD, chronic obstructive pulmonary disease; CR, complete response; GRFS, GVHD/relapse-free survival; OS, overall survival.

*

Drobyski et al124 is related to NCT02206035.

TNF is a pivotal dysregulated cytokine in response to total body irradiation (TBI) in preclinical systems, and inhibition during conditioning can potently inhibit GVHD.23  In contrast, systemic TNF dysregulation is generally not seen in patiebly due to the influence of GVHD prophylaxis,24  and results of clinical studies of TNF inhibition25  have been less dramatic than in murine systems. Phase 2 trials comparing the addition of etanercept to standard GVHD prophylaxis (starting at conditioning),26  or the combination of etanercept (starting on the day of transplant) plus extracorporeal photopheresis,27  have suggested possible reductions in the incidence of steroid-refractory aGVHD without reductions in the incidence of grade II to IV aGVHD.

Activation of the inflammasome pathway (eg, by ATP, crystals [eg, uric acid], NLR family members) is required for IL-1β and IL-18 production.28,29  Preclinical studies have demonstrated that IL-1β promotes GVHD,20  and IL-1β blockade can attenuate GVHD, particularly in conditioning containing cyclophosphamide.30  However, IL-1 blockade commencing during conditioning for up to 2 weeks did not reduce GVHD in a randomized clinical study.31  Most IL-18 produced early posttransplant is recipient-derived32  and in concert with IL-12, IL-18 is critical for T helper type 1 cell (Th1) differentiation and IFN-γ secretion.33  Exogenous IL-18 administration prior to conditioning has been demonstrated to invoke Fas-dependent donor CD4+ T-cell death, that attenuates MHC class II–dependent GVHD while preserving MHC class I–dependent graft-versus-leukemia (GVL) effects.34,35  However, opposing effects that exaggerate CD8+-mediated GVHD36  suggest this is unlikely to be clinically tractable. Thus, it remains to be seen whether modulation of the inflammasome pathway is practical in patients.

IL-33 is an IL-1 family member that is constitutively expressed in the nuclei of endothelial and epithelial cells and released in response to tissue injury.37  IL-33 binds to the IL-33 receptor (also known as growth stimulation expressed gene 2 or serum stimulation-2 [ST2]),38  which has membrane-bound and soluble (sST2) forms. In animal models, conditioning induced intraintestinal IL-33 early posttransplant.39  IL-33 administration preconditioning increased the numbers of recipient regulatory T cells (Tregs) and attenuated aGVHD,40  whereas IL-33 administration postconditioning,39  or preconditioning in the absence of Treg,40  exacerbated GVHD lethality due to enhanced expansion of effector T cells. Thus, the regulatory effects of IL-33 are via the expansion of ST2+ Tregs with enhanced regulatory function,41  together with their expression of amphiregulin42  that enhances tissue repair.43  In contrast, IL-33 can promote inflammation via enhancing type 1 differentiation of T cells.44,45  Importantly, sST2 is a biomarker of severe GVHD46  that acts to antagonize IL-33. Indeed, ST2 inhibition with a monoclonal antibody (Ab) that neutralizes sST2 has been shown to attenuate aGVHD by promoting IL-33 signaling in Th2s and Tregs.47  Well-designed studies will be required to ascertain whether these complex contextual effects of IL-33 can be exploited clinically.

Type I IFN can be generated via endogenous nucleic acid–dependent intracellular signaling of the cyclic guanosine monophosphate–adenosine monophosphate synthase (cGAS)/stimulator of interferon genes (STING) pathway or the RIG-I/mitochondrial antiviral-signaling protein (MAVS) pathway. Type I IFN can also be secreted in response to TLR-ligation at the cell surface by exogenous nucleic acids, such as CpG motifs.48  The absence of MAVS in alloSCT recipients results in enhanced TBI-induced intestinal damage, suggesting that TBI-driven host type I IFN production is protective in tissue.49  Type I IFN signaling of recipient hematopoietic cells (putatively dendritic cells [DC]) also protects recipients from CD4+-mediated GVHD but enhances CD8+ T-cell–mediated GVHD and GVL.50  At this point, further experimental information is needed to understand how type I IFNs modulate GVHD, and the outcome of type I IFNs or RIG-I agonists on clinical GVHD are difficult to predict. Indeed, the administration of type I IFNs after alloSCT are useful to enhance GVL in patients with relapsed leukemia but do so at the expense of GVHD.51 

The role of costimulatory and coinhibitory pathways in aGVHD

The activation and expansion of alloreactive donor T cells requires cognate costimulation at the APC–T-cell synapse. Many of these costimulatory molecules are members of the immunoglobulin superfamily (IgSF) or TNF/TNF receptor superfamilies (TNFSF/TNFRSF).52-54  IgSF members can mediate stimulatory or inhibitory signaling, whereas most TNFSF/TNFRSF signals are stimulatory only, and these pathways are described in detail below and in Figure 1 and Table 2.

Table 2.

Potential costimulatory molecule inhibition in aGVHD

TargetReagentDisease settingStudy or clinical trial status/outcomeClinical Trial IDReference
CD80/86 CTLA4-Ig GVHD prophylaxis Phase 2, low rate of II-IV aGVHD (2 of 10 patients) NCT01012492 65  
Phase 2 (randomized), reduced grade III/IV aGVHD and improved GFS, particularly after 7/8 MUD NCT01743131 66  
CD28 Anti-CD28 Fab Ab NHP: GVHD prophylaxis Preclinical, 100% GVHD-free survival in combination with sirolimus, but infection-related death is high  68  
 Anti-CD28 Fab Ab Healthy subjects Phase 1, safe and tolerated NCT02800811 69  
ICOSL Anti-ICOS ligand Ab SLE Phase 1b, safe, not powered for efficacy NCT01683695 75  
CD40 Anti-CD40 Ab NHP: GVHD prophylaxis Preclinical, combination with CTLA4-Ig and sirolimus, reductions in GVHD  63  
Anti-CD40 Ab Rheumatoid arthritis Phase 2a, safe, not powered for efficacy NCT01751776 105  
CD40L Anti-CD40L Ab GVHD prophylaxis Phase 1/2, recruiting NCT03605927  
OX40L Anti-OX40L Ab NHP: GVHD prophylaxis Preclinical, combination with sirolimus, no clinical GVHD seen  113  
Anti-OX40L Ab Atopic dermatitis Phase 2, recruiting NCT03754309  
DLL4 Anti-DLL4 Ab NHP: GVHD prophylaxis Preclinical, anti-DLL4 Ab monotherapy, prolonged OS  118  
TargetReagentDisease settingStudy or clinical trial status/outcomeClinical Trial IDReference
CD80/86 CTLA4-Ig GVHD prophylaxis Phase 2, low rate of II-IV aGVHD (2 of 10 patients) NCT01012492 65  
Phase 2 (randomized), reduced grade III/IV aGVHD and improved GFS, particularly after 7/8 MUD NCT01743131 66  
CD28 Anti-CD28 Fab Ab NHP: GVHD prophylaxis Preclinical, 100% GVHD-free survival in combination with sirolimus, but infection-related death is high  68  
 Anti-CD28 Fab Ab Healthy subjects Phase 1, safe and tolerated NCT02800811 69  
ICOSL Anti-ICOS ligand Ab SLE Phase 1b, safe, not powered for efficacy NCT01683695 75  
CD40 Anti-CD40 Ab NHP: GVHD prophylaxis Preclinical, combination with CTLA4-Ig and sirolimus, reductions in GVHD  63  
Anti-CD40 Ab Rheumatoid arthritis Phase 2a, safe, not powered for efficacy NCT01751776 105  
CD40L Anti-CD40L Ab GVHD prophylaxis Phase 1/2, recruiting NCT03605927  
OX40L Anti-OX40L Ab NHP: GVHD prophylaxis Preclinical, combination with sirolimus, no clinical GVHD seen  113  
Anti-OX40L Ab Atopic dermatitis Phase 2, recruiting NCT03754309  
DLL4 Anti-DLL4 Ab NHP: GVHD prophylaxis Preclinical, anti-DLL4 Ab monotherapy, prolonged OS  118  

GFS, aGVHD-free survival; MUD, matched unrelated donor; NHP, nonhuman primate; SLE, systemic lupus erythematosus.

IgSF costimulation

CD28 and cytotoxic T-lymphocyte associated protein 4 (CTLA4) are expressed by T cells; CD28 is expressed constitutively on naive CD4+ and CD8+ T cells, and ligation by CD80/86 (B7-1/B7-2) provides the second signal for T-cell growth and survival. In contrast, CTLA4 is induced on activated conventional T cells to provide inhibitory signals52  but is constitutively expressed on Treg.55  Lethal T-cell–mediated aGVHD is almost completely prevented in murine studies when common CD28/CTLA4 ligands, CD80 and CD86, are blocked,56  while aGVHD is only partially reduced when mice received genetically CD28-deficient donor T cells,57,58  likely related to the fact that Treg development and function require constitutional CD28 expression.59-61  Administration of CTLA4-Ig, a fusion protein that binds CD80 and CD86 to block interaction with CD28/CTLA4, does not completely prevent GVHD.62  A nonhuman primate study demonstrated the efficacy of a CTLA4-Ig, anti-CD40 Ab and sirolimus combination for the prevention of aGVHD.63  Indeed, CTLA4-Ig (abatacept) has produced promising clinical results when combined with standard GVHD prophylaxis,64,65  and a randomized phase 2 study found significantly decreased grade II to IV aGVHD after HLA-matched alloSCT.66  Of note, CTLA4-Ig concurrently blocks tolerogenic CTLA4-dependent signaling to Tregs and APCs,67  while CD28-specific inhibition is permissive of this pathway, allowing CD80/86 to continue to bind CTLA4. In nonhuman primates, combining pegylated anti-CD28 Ab (FR104) with sirolimus produced superior GVHD-free survival to a CTLA4-Ig/sirolimus combination, although beneficial effects were limited by infection-related deaths.68  FR104 appears safe in healthy human subjects69  but has not been tested in disease settings. The modulation of this pathway thus shows great promise, although questions in relation to the optimal reagent and timing of administration remain.

Inducible costimulator (ICOS; CD278) binds to ICOS ligand (also known as B7H, B7H2, or B7RP1) and is expressed on activated T cells and constitutively in Tregs and memory CD4+ T cells, where it controls the size of the T-cell pool.70  In murine models, animals receiving ICOS-deficient donor CD4+ T cells or anti-ICOS inhibition develop attenuated aGVHD, with less Th1 cytokine production. In contrast, ICOS-deficient CD8+ T cells (or inhibition during MHC class I–dependent GVHD) promote GVHD by enhancing CD8 T-cell expansion.71-73  This may relate to the fact that CD4+ T cells use PI3K-dependent ICOS signaling, whereas CD8+ T cells use a PI3K-independent pathway.74  Nonetheless, based on these preclinical results, ICOS blockade may attenuate aGVHD when combined with other immune suppressants, such as CTLA4-Ig73  or CD28 inhibition.68  The safety and potential efficacy of anti-ICOS ligand Ab, AMG557, have recently been confirmed in patients with systemic lupus erythematosus.75 

The simultaneous interaction of costimulatory/inhibitory molecules often exerts complex effects. B and T lymphocyte attenuator (BTLA) binds to a TNFRSF member, herpes virus entry mediator (HVEM), which binds BTLA and other molecules, lymphotoxin-like inducible protein that competes with glycoprotein D for herpesvirus entry on T cells (LIGHT), and lymphotoxin-α. These ligands and receptors are expressed on T cells and can bind in cis or trans.52  BTLA can deliver inhibitory signals, but76 administration of either non-blocking77  or blocking76,78  anti-BTLA Abs can prevent GVHD in murine models, reducing T-cell cytotoxicity77  while expanding Foxp3+ Tregs,78  independent of HVEM-binding and its known prosurvival effect.77  On the other hand, a BTLA but not LIGHT-blocking anti-HVEM Ab reduced allogeneic T-cell reactions, including IFN-γ production and cytotoxicity,79  suggesting BTLA-HVEM interactions are inflammatory. Inhibitory agonists thus might be effective against aGVHD, but clinical-grade reagents have not yet been tested.

Costimulatory DNAX accessory molecule 1 (DNAM-1; CD226) is expressed primarily by natural killer and T cells.80  The DNAM-1/CD226 subfamily also includes coinhibitory molecules, T-cell immunoglobulin and immunoreceptor tyrosine inhibitory motif domain (TIGIT; VSTM3), and CD96 molecules, with shared receptors (CD155, CD112, CD111).80  DNAM-1–deficient donor CD8+ T cells81  and Tregs82  ameliorate GVHD in murine alloSCT models. At least 1 TIGIT blocking Ab has been shown to accelerate GVHD mortality.83  TIGIT blocking antibodies are currently being tested as checkpoint inhibitors in patients with cancer, but the inhibition of DNAM-1 or activation of TIGIT may be a useful strategy to prevent aGVHD. Indeed, after alloSCT, high levels of soluble DNAM-1 in sera are associated with grade II to IV aGVHD.84  Furthermore, high TIGIT expression in bone marrow after alloSCT was recently shown to correlate with reduced aGVHD but poor progression-free survival due to high levels of relapse.85 

Programmed cell death protein 1 (PD-1), which binds to programmed death ligand-1 (PD-L1) and PD-L2, is expressed on activated T cells and is a coinhibitory molecule. When expressed in conjunction with functional defects (eg, cytokine secretion), PD-1 is a marker of T-cell exhaustion. Additional molecules expressed during exhaustion include TIM3 (which binds to Galectin 9) and LAG3 (which binds to MHC II). Thus, while PD-1–PD-L1 and CTLA4 inhibitors are currently the most widely used immune checkpoint inhibitors in clinical oncology practice, trials are underway with reagents that inhibit LAG3 and TIM3. In murine models, PD-1–deficient donor T cells or anti–PD-1 Ab treatment pre- or post-alloSCT increase GVL effects, but also augment GVHD severity86,87  and associated morbidity/mortality.88  PD-1 blockade has been widely used for patients with Hodgkin or non-Hodgkin lymphomas, and pretransplant PD-1 blockade is associated with severe aGVHD.89  These patients should receive posttransplant cyclophosphamide-based immune suppression.90,91  Patients with acute myeloid leukemia or myelodysplastic syndromes frequently undergo alloSCT and have shown susceptibility to PD-1 blockade during induction therapy,92  consistent with high PD-1 and TIGIT expression in CD8+ T cells from acute myeloid leukemia patients.93 

V-domain immunoglobulin-containing suppressor of T-cell activation (VISTA) is a novel immune checkpoint ligand/receptor that is expressed on myeloid and T cells. Murine VISTA-Ig fusion protein inhibited cytokine production (IL-2 and IFN-γ) by CD4+ T cells and VISTA-Ig or VISTA-expressing APCs potently suppressed CD4+ and CD8+ T-cell proliferation in vitro.94  Prostate cancers from patients treated with ipilimumab (anti-CTLA4 Ab) increase PD-L1 and VISTA expression on tumor-infiltrated CD4+ and CD8+ T cells and macrophages.95  V-set and Ig domain containing 396,97  and P-selectin glycoprotein ligand-198  have recently been identified as VISTA receptors, and VISTA-Ig may represent a potent therapeutic agent for preventing GVHD. Other suppressive costimulatory pathways, such as B7-H4, have also recently been demonstrated to inhibit GVHD,99  although the cognate receptor on T cells awaits identification.

TNFRSF costimulation

CD40 (TNFRSF5) expressed on APCs binds to its cognate ligand (CD40L; CD154, gp39, or TNFSF5) on activated T cells.54  CD40 signaling potently enhances APC costimulatory expression (CD80/86), and CD40L is critical for B-cell class switching.100  The combination of anti-CD40L Ab and CTLA4-Ig facilitates long-term engraftment in murine cardiac and skin allogeneic transplants101  and is abrogated by calcineurin inhibition. In murine models, CD40L inhibition potently attenuates lethal aGVHD,102,103  especially in combination with CD28 deficiency or LIGHT blockade.103,104  In nonhuman primates, combined (blocking) anti-CD40 Ab, CTLA4-Ig and sirolimus attenuate TRM.63  A phase 2 study has demonstrated the safety of anti-CD40 Ab treatment (BI 655064) and improved end points in patients with rheumatoid arthritis.105  Thus, inhibition of the CD40-CD40L costimulatory pathway seems a highly tractable approach to prevent aGVHD now that a new generation of reagents that do not invoke thrombosis are available (ClinicalTrials: NCT03605927; anti-CD40L Ab; BMS-986004).106-108 

The OX40 (CD134, TNFRSF4) is expressed on activated T cells and produces costimulatory signals upon binding to OX40L, expressed on activated APCs.53  Murine models have shown that OX40 induces effector T-cell expansion, without influencing natural Treg generation, and OX40 signaling in Treg perturbs their suppressive function.109,110  In contrast, OX40 inhibits induced Foxp3+ Treg generation from activated CD4+ T cells in a BATF-dependent and independent manner.111,112  This costimulatory pathway is a highly promising target for inhibition, with confirmation of efficacy also demonstrated in nonhuman primate models of alloSCT.113  In contrast to anti-CD40 Ab plus CTLA4-Ig/Sirolimus combinations that induced severe lymphopenia and poor Treg expansion after transplant,63  an OX40L Ab (KY1005) plus sirolimus combination achieved lymphocyte recovery equivalent to that after autologous transplant, and augmented Treg recovery.113  Together, these findings may explain the ability of OX40 inhibition to preserve Treg and T-cell reconstitution, making inhibition of this axis an attractive target after alloSCT.

Notch-dependent costimulation

Notch is an evolutionally conserved element of cell-to-cell interaction. Mammals have 4 Notch receptors (Notch 1 to 4) with 5 ligands of the Jagged (Jagged 1 and 2) and Delta-like (Delta-like ligand 1 [DLL1], DLL3, and DLL4) families.114  T-cell–specific expression of a pan-Notch inhibitor (DNMAML) reduces aGVHD lethality and tissue pathology in murine models, with attenuated cytokine secretion (IFN-γ, TNF-α, IL-2), despite the maintenance of T-cell proliferation and cytotoxicity that is permissive of GVL.115  The effects primarily relate to Notch 1 inhibition in donor T cells, although Notch 1 is also crucial for intestinal regeneration. DLL4 is largely expressed by nonhematopoietic stromal cells116 ; crypt regeneration was preserved and GVHD was inhibited following administration of an anti-DLL4/DLL1 blocking Ab.117  A subsequent nonhuman primate study confirmed this promise for DLL4 blockade as a strategy to prevent GVHD.118 

Cytokine-dependent control of T-cell differentiation

IL-6 is a proinflammatory cytokine produced by multiple cell types, including endothelial cells, fibroblasts, keratinocytes, DC, macrophages, and T cells.119  Microbial products enhance IL-6 production in tissue,120,121  and systemic IL-6 levels are increased early after allogeneic transplant in animal models122,123  and patients,24,124  which is most profound after TBI-based conditioning.24  Recipient DC appear to be the major cellular source of IL-6 dysregulation125  but donor DC-derived IL-6 also plays a role in T cell expansion and differentiation during aGVHD.21  IL-6 and transforming growth factor-β (TGF-β) induce Th17 differentiation from naive T cells, and IL-6 inhibits TGF-β–driven Treg differentiation.126  Classical IL-6 signaling in donor T cells is critical for the generation of donor Th17/Tc17 and Th22 differentiation after bone marrow transplantation (BMT).125,127,128  Indeed, the beneficial effects of IL-6 inhibition in preclinical studies are related to increased donor Treg/type 1 regulatory T cells (Tr1) numbers125,129,130  and inhibition of Th17/Th22 differentiation.125  Anti–IL-6 receptor Ab (tocilizumab) shows promise in reducing the incidence of acute GVHD in phase 1/2 studies24,124  but requires confirmation in multicenter phase 3 studies.

IL-12 and IL-23 are proinflammatory cytokines that share p40 subunits and are recognized for their key roles in developing Th1 and Th17 subsets of helper CD4+ T cells, respectively. IL-27 is a heterodimeric member of the IL-12 family that contains the IL-27p28 subunit and is secreted by cells of the monocyte-macrophage lineage to drive both proinflammatory effector differentiation and type-1 regulatory T cells.130-132  In the presence of luminal microbiota, intestinal macrophages secrete IL-12/23p40, that induces IFN-γ by lamina propria lymphocytes to control MHC class II expression by intestinal epithelial cells, and this process is enhanced by conditioning irradiation.22  MHC class II expressed by intestinal epithelial cells can initiate CD4-dependent aGVHD in the GI tract and can be attenuated by IL-12/23p40 inhibition.22  IL-23 consists of p40 and p19 subunits. Mice receiving allografts and anti-p19 Ab administration (or IL-23p19 deficient grafts) have attenuated aGVHD in the colon.133  A randomized placebo-controlled clinical study administered anti–IL-12/23p40 Ab (ustekinumab) 1 day pretransplant and again at day 20 posttransplant, resulted in significant reduction of Th17 and Th1 differentiation ex vivo, although the study was not powered to detect reductions in aGVHD.134  Anti–IL-23p19 Ab (Guselkumab) has demonstrated efficacy in psoriasis.135  Of note, IL-23 promotes IL-22 production in the GI tract early after BMT that protects from epithelial damage,136  and so IL-12 itself may represent the most rational target for inhibition in clinical transplantation.

The impact of T-cell–derived cytokines on GVHD

Alloantigen-exposed donor CD4+ and CD8+ T cells differentiate into pathogenic type-1 cells (IL-2, IFN-γ–secreting Th1, and Tc1, respectively) and type-17 cells (IL-17 secreting Th17 and Tc17, respectively), which expedite rapid host cell elimination, promote engraftment and mediate tissue damage in GVHD target organs.137  Although other T-cell differentiation paradigms may play a role in GVHD (Th2, Tfh), they are generally less relevant to aGVHD, and the reader is referred to reviews of cGVHD for a detailed explanation of these lineages after alloSCT.21,138 

IL-2 is primarily secreted by T cells, and particularly CD4+ T cells. The IL-2 receptor, which comprises 3 subunits (the α chain [CD25], β chain [CD122], and the γ subunit [CD132]), is expressed on thymus-derived Treg and antigen-activated T cells.139  IL-2 is critical for thymic Treg development and peripheral homeostasis, including T-cell differentiation. Calcineurin inhibitors, such as tacrolimus and cyclosporin, bind calcineurin and inhibit subsequent nuclear factor of activated T cells (NFAT)–dependent IL-2 transcription, resulting in the blocking of T-cell activation, expansion, and effector function.140  Monoclonal antibodies have been developed to block CD25, and like calcineurin inhibitors, block both conventional T cells but also Treg expansion, creating a potential for both positive and negative effects in transplantation. Conversely, low-dose IL-2 preferentially stimulates Treg and may be tolerogenic, at least when administered late after transplant.141  In contrast, the requirement for IL-2 in effector and regulatory T cell responses has translated to mixed effects on aGVHD when trying to augment or block this cytokine early after transplant.142-144  The effects of therapeutic IL-2 modulation in BMT would appear unpredictable in the absence of mutated IL-2 proteins or Ab with specificity to IL-2 receptor subunits (eg, CD25 agonists to promote Treg expansion).145  In preclinical studies, the transfer of IFN-γ–deficient donor T cells or anti–IFN-γ Ab administration accelerates GVHD lethality due to the promotion of lung injury (idiopathic pneumonia syndrome), but GVHD within the GI tract is markedly attenuated.123,146  Nevertheless, complete blockade of IFN-γ would seem impractical as a clinical strategy.

The IL-17 cytokine family includes 6 members: IL-17A to F; Th17 cell–secreted IL-17A and IL-17F are the best characterized members,147  and the pathogenic role of donor type 17 T cells in aGVHD is well established.127,128,148-150  IL-17 secreted by recipient lymphoid cells, including mucosal-associated invariant T (MAIT) cells, prevent dysbiosis and protect from gut GVHD.150,151  Therefore, although anti-IL17A Ab administration shows the efficacy in multiple autoimmune diseases,152-154  IL-17 blockade itself is unlikely to efficiently prevent GVHD due to the critical role of this cytokine in maintaining mucosal integrity and microbiome diversity.150  The emergence and persistence of donor Th17/Tc17 cells were noted during aGVHD in nonhuman primates following pharmacological GVHD prophylaxis with tacrolimus plus methotrexate.155  Since Tc17 cells exacerbate GVHD but do not mediate GVL, they are good targets to prevent GVHD127  and would require intervention targeting critical transcriptional factors (eg, RORγt) with small molecule inhibitors. During aGVHD, both Th17 and non-Th17 donor lineages are also a major source of granulocyte-macrophage colony-stimulating factor (GM-CSF). GM-CSF expands myeloid populations in peripheral lymph nodes, including donor DC with high alloantigen-presenting capacity that profoundly augments aGVHD in the GI tract.128,156,157  Thus, GM-CSF blockade may be an attractive target to prevent aGVHD, although effects on hematopoiesis would need to be considered.

The immunosuppressive cytokine, IL-10, attenuates tissue damage during inflammation.158  IL-10 is produced predominantly by donor T cells after alloSCT,159  specifically Foxp3+ Treg and Tr1 that are induced in the periphery.130  Although IL-10−/− donor T cells augment lethal GVHD in mice,130,160  systemic IL-10 administration has dose-dependent effects; low doses decrease and high doses enhance aGVHD.160  TGF-β is also an immunosuppressive cytokine that, in the absence of IL-6, promotes Treg differentiation126  and is critical for the induction of tolerance. In murine models, the neutralization of TGF-β early after transplant exacerbates aGVHD, whereas it attenuates fibrosis during chronic skin GVHD.161,162  Thus, inhibition of TGF-β is likely to be difficult to exploit clinically. Given these apparent narrow therapeutic windows, targeting IL-10 delivery to the GI tract, or the adoptive transfer of IL-10/TGF-β–producing regulatory cells (eg, Treg and Tr1) may be the most useful therapeutic approaches after alloSCT.

IL-22 is a member of the IL-20 cytokine subfamily within the larger IL-10 family, which uses receptors with a common subunit (IL-20 receptor β) and links the immune system with epithelial cells to enhance barrier function.158,163  IL-22 secretion by host intestinal innate immune cells (ILCs) protects intestinal stem cells from GVHD damage and promotes epithelial regeneration in a regenerating islet-derived 3γ (REG3γ, mouse homolog of human regenerating islet-derived 3α)-dependent manner, although IL-22–secreting host ILCs are eliminated during GVHD.136,164,165  The systemic administration of F-652, a recombinant human IL-22-dimer–Fc-fusion protein, or mouse recombinant IL-22 reduces gut GVHD in mice when administered after the onset of intestinal GVHD (day 7),164,165  and a clinical trial is underway (Clinical Trials: NCT02406651). In contrast, donor-derived IL-22 can exacerbate GVHD, particularly in the skin, although pathogenic effects in gut have also been suggested.166-168  These effects likely represent IL-22 secretion from pathogenic Th17/Th22 cells125,168  rather than protective ILC3 subsets136  but does represent a theoretical limitation to prolonged IL-22 administration.

The activation, expansion, and differentiation of donor T cells represent the pivotal pathway responsible for acute GVHD. This represents a complex process that starts during conditioning and continues until donor T cells acquire full effector function. The immunological process includes a number of pivotal stimulatory and inhibitory soluble cytokines and surface proteins within the interface of the APCs and T cell that can theoretically be inhibited with clinically available reagents, repurposed for transplantation. Logical targets include pivotal cytokines involved in antigen presentation and T-cell differentiation, such as IL-12, surface costimulatory molecules on APCs, such as CD80/CD86, CD40, OX40L, or their cognate costimulatory T-cell ligands (eg, CD28, CD40L, OX40). Indeed, this field has now generated numerous tractable clinical pathways that are outlined in Table 1 and Table 2. The challenge in the current era is to choose the most appropriate target and concurrent immune suppression, given the recent uptake of augmented immune suppression protocols, including posttransplant cyclophosphamide. It is important to note that meaningful separation of GVHD and GVL has yet to be achieved in the clinic, and early-phase single-arm studies are grossly underpowered and inappropriate to assess relapse. There thus remains a requirement for (ideally blinded) large phase 3 studies to assess promising new approaches to prevent GVHD. Toward the future, it is likely that combinations of therapies that block costimulation and T-cell differentiation may be synergistic; future studies will need to consider a stepwise approach. Finally, we will need to incorporate study end points that measure the potential benefits of using Ab-based approaches that may avoid toxicities inherent in our historical drug-based immune suppression.

The authors thank Laura Green of Fred Hutchinson Cancer Research Center for generation of the graphics.

This work was supported by a research grant from the National Institutes of Health (NIH), National Heart, Lung, and Blood Institute grant R01HL148164.

The content is solely the responsibility of the authors and does not necessarily represent the official views of the NIH. G.R.H. is an Andy Hill CARE Distinguished Researcher.

Contribution: M.K. and G.R.H. wrote the manuscript.

Conflict-of-interest disclosure: M.K. and G.R.H. have submitted a patent application on methods to prevent antigen presentation in the GI tract. G.R.H. has received funding from Roche for a clinical study of Tocilizumab in aGVHD prophylaxis.

Correspondence: Motoko Koyama, Fred Hutchinson Cancer Research Center, 1100 Fairview Ave N, Seattle, WA 98109; e-mail: mkoyama@fredhutch.org; and Geoffrey R. Hill, Fred Hutchinson Cancer Research Center, 1100 Fairview Ave N, Seattle, WA 98109; e-mail: grhill@fredhutch.org.

1.
Matsumura-Kimoto
Y
,
Inamoto
Y
,
Tajima
K
, et al
.
Association of cumulative steroid dose with risk of infection after treatment for severe acute graft-versus-host disease
.
Biol Blood Marrow Transplant
.
2016
;
22
(
6
):
1102
-
1107
.
2.
Rashidi
A
,
DeFor
TE
,
Holtan
SG
,
Blazar
BR
,
Weisdorf
DJ
,
MacMillan
ML
.
Outcomes and predictors of response in steroid-refractory acute graft-versus-host disease
.
Biol Blood Marrow Transplant
.
2019
;
25
(
11
):
2297
-
2302
.
3.
Hill
GR
,
Ferrara
JL
.
The primacy of the gastrointestinal tract as a target organ of acute graft-versus-host disease: rationale for the use of cytokine shields in allogeneic bone marrow transplantation
.
Blood
.
2000
;
95
(
9
):
2754
-
2759
.
4.
Socié
G
,
Blazar
BR
.
Acute graft-versus-host disease: from the bench to the bedside
.
Blood
.
2009
;
114
(
20
):
4327
-
4336
.
5.
Simonetta
F
,
Alvarez
M
,
Negrin
RS
.
Natural killer cells in graft-versus-host-disease after allogeneic hematopoietic cell transplantation
.
Front Immunol
.
2017
;
8
:
465
.
6.
Cooley
S
,
Parham
P
,
Miller
JS
.
Strategies to activate NK cells to prevent relapse and induce remission following hematopoietic stem cell transplantation
.
Blood
.
2018
;
131
(
10
):
1053
-
1062
.
7.
Sckisel
GD
,
Bouchlaka
MN
,
Monjazeb
AM
, et al
.
Out-of-sequence signal 3 paralyzes primary CD4(+) T-cell-dependent immunity
.
Immunity
.
2015
;
43
(
2
):
240
-
250
.
8.
Chen
Y
,
Kornblit
B
,
Hamlin
DK
, et al
.
Durable donor engraftment after radioimmunotherapy using α-emitter astatine-211-labeled anti-CD45 antibody for conditioning in allogeneic hematopoietic cell transplantation
.
Blood
.
2012
;
119
(
5
):
1130
-
1138
.
9.
Chhabra
A
,
Ring
AM
,
Weiskopf
K
, et al
.
Hematopoietic stem cell transplantation in immunocompetent hosts without radiation or chemotherapy
.
Sci Transl Med
.
2016
;
8
(
351
):
351ra105
.
10.
George
BM
,
Kao
KS
,
Kwon
HS
, et al
.
Antibody conditioning enables MHC-mismatched hematopoietic stem cell transplants and organ graft tolerance
.
Cell Stem Cell
.
2019
;
25
(
2
):
185
-
192.e183
.
11.
Czechowicz
A
,
Palchaudhuri
R
,
Scheck
A
, et al
.
Selective hematopoietic stem cell ablation using CD117-antibody-drug-conjugates enables safe and effective transplantation with immunity preservation
.
Nat Commun
.
2019
;
10
(
1
):
617
.
12.
Palchaudhuri
R
,
Saez
B
,
Hoggatt
J
, et al
.
Non-genotoxic conditioning for hematopoietic stem cell transplantation using a hematopoietic-cell-specific internalizing immunotoxin
.
Nat Biotechnol
.
2016
;
34
(
7
):
738
-
745
.
13.
Gong
T
,
Liu
L
,
Jiang
W
,
Zhou
R
.
DAMP-sensing receptors in sterile inflammation and inflammatory diseases
.
Nat Rev Immunol
.
2020
;
20
(
2
):
95
-
112
.
14.
Yang
D
,
Han
Z
,
Oppenheim
JJ
.
Alarmins and immunity
.
Immunol Rev
.
2017
;
280
(
1
):
41
-
56
.
15.
Brubaker
SW
,
Bonham
KS
,
Zanoni
I
,
Kagan
JC
.
Innate immune pattern recognition: a cell biological perspective
.
Annu Rev Immunol
.
2015
;
33
(
1
):
257
-
290
.
16.
Cooke
KR
,
Gerbitz
A
,
Crawford
JM
, et al
.
LPS antagonism reduces graft-versus-host disease and preserves graft-versus-leukemia activity after experimental bone marrow transplantation
.
J Clin Invest
.
2001
;
107
(
12
):
1581
-
1589
.
17.
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
.
18.
Uryu
H
,
Hashimoto
D
,
Kato
K
, et al
.
α-Mannan induces Th17-mediated pulmonary graft-versus-host disease in mice
.
Blood
.
2015
;
125
(
19
):
3014
-
3023
.
19.
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
.
20.
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
.
21.
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
.
22.
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
.
23.
Hill
GR
,
Crawford
JM
,
Cooke
KR
,
Brinson
YS
,
Pan
L
,
Ferrara
JLM
.
Total body irradiation and acute graft-versus-host disease: the role of gastrointestinal damage and inflammatory cytokines
.
Blood
.
1997
;
90
(
8
):
3204
-
3213
.
24.
Kennedy
GA
,
Varelias
A
,
Vuckovic
S
, et al
.
Addition of interleukin-6 inhibition with tocilizumab to standard graft-versus-host disease prophylaxis after allogeneic stem-cell transplantation: a phase 1/2 trial
.
Lancet Oncol
.
2014
;
15
(
13
):
1451
-
1459
.
25.
Alousi
AM
,
Weisdorf
DJ
,
Logan
BR
, et al;
Blood and Marrow Transplant Clinical Trials Network
.
Etanercept, mycophenolate, denileukin, or pentostatin plus corticosteroids for acute graft-versus-host disease: a randomized phase 2 trial from the Blood and Marrow Transplant Clinical Trials Network
.
Blood
.
2009
;
114
(
3
):
511
-
517
.
26.
Choi
SW
,
Stiff
P
,
Cooke
K
, et al
.
TNF-inhibition with etanercept for graft-versus-host disease prevention in high-risk HCT: lower TNFR1 levels correlate with better outcomes
.
Biol Blood Marrow Transplant
.
2012
;
18
(
10
):
1525
-
1532
.
27.
Kitko
CL
,
Braun
T
,
Couriel
DR
, et al
.
Combination therapy for graft-versus-host disease prophylaxis with etanercept and extracorporeal photopheresis: results of a phase II clinical trial
.
Biol Blood Marrow Transplant
.
2016
;
22
(
5
):
862
-
868
.
28.
Guo
H
,
Callaway
JB
,
Ting
JP
.
Inflammasomes: mechanism of action, role in disease, and therapeutics
.
Nat Med
.
2015
;
21
(
7
):
677
-
687
.
29.
Swanson
KV
,
Deng
M
,
Ting
JP
.
The NLRP3 inflammasome: molecular activation and regulation to therapeutics
.
Nat Rev Immunol
.
2019
;
19
(
8
):
477
-
489
.
30.
Hill
GR
,
Teshima
T
,
Gerbitz
A
, et al
.
Differential roles of IL-1 and TNF-alpha on graft-versus-host disease and graft versus leukemia
.
J Clin Invest
.
1999
;
104
(
4
):
459
-
467
.
31.
Antin
JH
,
Weisdorf
D
,
Neuberg
D
, et al
.
Interleukin-1 blockade does not prevent acute graft-versus-host disease: results of a randomized, double-blind, placebo-controlled trial of interleukin-1 receptor antagonist in allogeneic bone marrow transplantation
.
Blood
.
2002
;
100
(
10
):
3479
-
3482
.
32.
Zeiser
R
,
Zambricki
EA
,
Leveson-Gower
D
,
Kambham
N
,
Beilhack
A
,
Negrin
RS
.
Host-derived interleukin-18 differentially impacts regulatory and conventional T cell expansion during acute graft-versus-host disease
.
Biol Blood Marrow Transplant
.
2007
;
13
(
12
):
1427
-
1438
.
33.
Chang
JT
,
Segal
BM
,
Nakanishi
K
,
Okamura
H
,
Shevach
EM
.
The costimulatory effect of IL-18 on the induction of antigen-specific IFN-gamma production by resting T cells is IL-12 dependent and is mediated by up-regulation of the IL-12 receptor beta2 subunit
.
Eur J Immunol
.
2000
;
30
(
4
):
1113
-
1119
.
34.
Reddy
P
,
Teshima
T
,
Kukuruga
M
, et al
.
Interleukin-18 regulates acute graft-versus-host disease by enhancing Fas-mediated donor T cell apoptosis
.
J Exp Med
.
2001
;
194
(
10
):
1433
-
1440
.
35.
Reddy
P
,
Teshima
T
,
Hildebrandt
G
, et al
.
Interleukin 18 preserves a perforin-dependent graft-versus-leukemia effect after allogeneic bone marrow transplantation
.
Blood
.
2002
;
100
(
9
):
3429
-
3431
.
36.
Min
CK
,
Maeda
Y
,
Lowler
K
, et al
.
Paradoxical effects of interleukin-18 on the severity of acute graft-versus-host disease mediated by CD4+ and CD8+ T-cell subsets after experimental allogeneic bone marrow transplantation
.
Blood
.
2004
;
104
(
10
):
3393
-
3399
.
37.
Liew
FY
,
Girard
JP
,
Turnquist
HR
.
Interleukin-33 in health and disease
.
Nat Rev Immunol
.
2016
;
16
(
11
):
676
-
689
.
38.
Griesenauer
B
,
Paczesny
S
.
The ST2/IL-33 axis in immune cells during inflammatory diseases
.
Front Immunol
.
2017
;
8
:
475
.
39.
Reichenbach
DK
,
Schwarze
V
,
Matta
BM
, et al
.
The IL-33/ST2 axis augments effector T-cell responses during acute GVHD [published correction appears in Blood. 2016;128(9):1311]
.
Blood
.
2015
;
125
(
20
):
3183
-
3192
.
40.
Matta
BM
,
Reichenbach
DK
,
Zhang
X
, et al
.
Peri-alloHCT IL-33 administration expands recipient T-regulatory cells that protect mice against acute GVHD
.
Blood
.
2016
;
128
(
3
):
427
-
439
.
41.
Zaiss
DM
,
van Loosdregt
J
,
Gorlani
A
, et al
.
Amphiregulin enhances regulatory T cell-suppressive function via the epidermal growth factor receptor
.
Immunity
.
2013
;
38
(
2
):
275
-
284
.
42.
Yang
J
,
Ramadan
A
,
Reichenbach
DK
, et al
.
Rorc restrains the potency of ST2+ regulatory T cells in ameliorating intestinal graft-versus-host disease
.
JCI Insight
.
2019
;
4
(
5
):
e122014
.
43.
Arpaia
N
,
Green
JA
,
Moltedo
B
, et al
.
A distinct function of regulatory T cells in tissue protection
.
Cell
.
2015
;
162
(
5
):
1078
-
1089
.
44.
Bonilla
WV
,
Fröhlich
A
,
Senn
K
, et al
.
The alarmin interleukin-33 drives protective antiviral CD8+ T cell responses
.
Science
.
2012
;
335
(
6071
):
984
-
989
.
45.
Baumann
C
,
Bonilla
WV
,
Fröhlich
A
, et al
.
T-bet- and STAT4-dependent IL-33 receptor expression directly promotes antiviral Th1 cell responses
.
Proc Natl Acad Sci USA
.
2015
;
112
(
13
):
4056
-
4061
.
46.
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
.
47.
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
.
48.
Crowl
JT
,
Gray
EE
,
Pestal
K
,
Volkman
HE
,
Stetson
DB
.
Intracellular nucleic acid detection in autoimmunity
.
Annu Rev Immunol
.
2017
;
35
(
1
):
313
-
336
.
49.
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
.
50.
Robb
RJ
,
Kreijveld
E
,
Kuns
RD
, et al
.
Type I-IFNs control GVHD and GVL responses after transplantation
.
Blood
.
2011
;
118
(
12
):
3399
-
3409
.
51.
Henden
AS
,
Varelias
A
,
Leach
J
, et al
.
Pegylated interferon-2α invokes graft-versus-leukemia effects in patients relapsing after allogeneic stem cell transplantation
.
Blood Adv
.
2019
;
3
(
20
):
3013
-
3019
.
52.
Chen
L
,
Flies
DB
.
Molecular mechanisms of T cell co-stimulation and co-inhibition [published correction appears in Nat Rev Immunol. 2013;13(7):542]
.
Nat Rev Immunol
.
2013
;
13
(
4
):
227
-
242
.
53.
Ward-Kavanagh
LK
,
Lin
WW
,
Šedý
JR
,
Ware
CF
.
The TNF receptor superfamily in co-stimulating and co-inhibitory responses
.
Immunity
.
2016
;
44
(
5
):
1005
-
1019
.
54.
Zhang
Q
,
Vignali
DA
.
Co-stimulatory and co-inhibitory pathways in autoimmunity
.
Immunity
.
2016
;
44
(
5
):
1034
-
1051
.
55.
Read
S
,
Malmström
V
,
Powrie
F
.
Cytotoxic T lymphocyte-associated antigen 4 plays an essential role in the function of CD25(+)CD4(+) regulatory cells that control intestinal inflammation
.
J Exp Med
.
2000
;
192
(
2
):
295
-
302
.
56.
Blazar
BR
,
Sharpe
AH
,
Taylor
PA
, et al
.
Infusion of anti-B7.1 (CD80) and anti-B7.2 (CD86) monoclonal antibodies inhibits murine graft-versus-host disease lethality in part via direct effects on CD4+ and CD8+ T cells
.
J Immunol
.
1996
;
157
(
8
):
3250
-
3259
.
57.
Yu
XZ
,
Martin
PJ
,
Anasetti
C
.
Role of CD28 in acute graft-versus-host disease
.
Blood
.
1998
;
92
(
8
):
2963
-
2970
.
58.
Speiser
DE
,
Bachmann
MF
,
Shahinian
A
,
Mak
TW
,
Ohashi
PS
.
Acute graft-versus-host disease without costimulation via CD28
.
Transplantation
.
1997
;
63
(
7
):
1042
-
1044
.
59.
Salomon
B
,
Lenschow
DJ
,
Rhee
L
, et al
.
B7/CD28 costimulation is essential for the homeostasis of the CD4+CD25+ immunoregulatory T cells that control autoimmune diabetes
.
Immunity
.
2000
;
12
(
4
):
431
-
440
.
60.
Zhang
R
,
Huynh
A
,
Whitcher
G
,
Chang
J
,
Maltzman
JS
,
Turka
LA
.
An obligate cell-intrinsic function for CD28 in Tregs
.
J Clin Invest
.
2013
;
123
(
2
):
580
-
593
.
61.
Tai
X
,
Cowan
M
,
Feigenbaum
L
,
Singer
A
.
CD28 costimulation of developing thymocytes induces Foxp3 expression and regulatory T cell differentiation independently of interleukin 2
.
Nat Immunol
.
2005
;
6
(
2
):
152
-
162
.
62.
Blazar
BR
,
Taylor
PA
,
Linsley
PS
,
Vallera
DA
.
In vivo blockade of CD28/CTLA4: B7/BB1 interaction with CTLA4-Ig reduces lethal murine graft-versus-host disease across the major histocompatibility complex barrier in mice
.
Blood
.
1994
;
83
(
12
):
3815
-
3825
.
63.
Miller
WP
,
Srinivasan
S
,
Panoskaltsis-Mortari
A
, et al
.
GVHD after haploidentical transplantation: a novel, MHC-defined rhesus macaque model identifies CD28- CD8+ T cells as a reservoir of breakthrough T-cell proliferation during costimulation blockade and sirolimus-based immunosuppression
.
Blood
.
2010
;
116
(
24
):
5403
-
5418
.
64.
Jaiswal
SR
,
Aiyer
HM
,
Rawat
G
,
Gera
A
,
Chakrabarti
S
.
CTLA4Ig-based reduced intensity conditioning and donor lymphocyte infusions for haploidentical transplantation in refractory aggressive B-cell lymphoma relapsing after an autograft: Early results from a pilot study
.
Exp Hematol
.
2019
;
77
:
26
-
35.e1
.
65.
Koura
DT
,
Horan
JT
,
Langston
AA
, et al
.
In vivo T cell costimulation blockade with abatacept for acute graft-versus-host disease prevention: a first-in-disease trial
.
Biol Blood Marrow Transplant
.
2013
;
19
(
11
):
1638
-
1649
.
66.
Watkins
BK
,
Qayed
M
,
Bratrude
B
, et al
.
T cell costimulation blockade with CTLA4-Ig (abatacept) for acute Gvhd prevention in HLA matched and mismatched unrelated donor transplantation: results of the first phase 2 trial [abstract]
.
2019 TCT | Transplantation & Cellular Therapy Meetings of ASBMT and CIBMTR
.
2019
.
Abstract 65
.
67.
Mayer
L
,
Kaser
A
,
Blumberg
RS
.
Dead on arrival: understanding the failure of CTLA4-immunoglobulin therapy in inflammatory bowel disease
.
Gastroenterology
.
2012
;
143
(
1
):
13
-
17
.
68.
Watkins
BK
,
Tkachev
V
,
Furlan
SN
, et al
.
CD28 blockade controls T cell activation to prevent graft-versus-host disease in primates
.
J Clin Invest
.
2018
;
128
(
9
):
3991
-
4007
.
69.
Poirier
N
,
Blancho
G
,
Hiance
M
, et al
.
First-in-human study in healthy subjects with FR104, a pegylated monoclonal antibody fragment antagonist of CD28
.
J Immunol
.
2016
;
197
(
12
):
4593
-
4602
.
70.
Burmeister
Y
,
Lischke
T
,
Dahler
AC
, et al
.
ICOS controls the pool size of effector-memory and regulatory T cells
.
J Immunol
.
2008
;
180
(
2
):
774
-
782
.
71.
Taylor
PA
,
Panoskaltsis-Mortari
A
,
Freeman
GJ
, et al
.
Targeting of inducible costimulator (ICOS) expressed on alloreactive T cells down-regulates graft-versus-host disease (GVHD) and facilitates engraftment of allogeneic bone marrow (BM)
.
Blood
.
2005
;
105
(
8
):
3372
-
3380
.
72.
Yu
XZ
,
Liang
Y
,
Nurieva
RI
,
Guo
F
,
Anasetti
C
,
Dong
C
.
Opposing effects of ICOS on graft-versus-host disease mediated by CD4 and CD8 T cells
.
J Immunol
.
2006
;
176
(
12
):
7394
-
7401
.
73.
Li
J
,
Semple
K
,
Suh
WK
, et al
.
Roles of CD28, CTLA4, and inducible costimulator in acute graft-versus-host disease in mice
.
Biol Blood Marrow Transplant
.
2011
;
17
(
7
):
962
-
969
.
74.
Li
J
,
Heinrichs
J
,
Leconte
J
, et al
.
Phosphatidylinositol 3-kinase-independent signaling pathways contribute to ICOS-mediated T cell costimulation in acute graft-versus-host disease in mice
.
J Immunol
.
2013
;
191
(
1
):
200
-
207
.
75.
Cheng
LE
,
Amoura
Z
,
Cheah
B
, et al
.
Brief report: a randomized, double-blind, parallel-group, placebo-controlled, multiple-dose study to evaluate AMG 557 in patients with systemic lupus erythematosus and active lupus arthritis
.
Arthritis Rheumatol
.
2018
;
70
(
7
):
1071
-
1076
.
76.
Hurchla
MA
,
Sedy
JR
,
Murphy
KM
.
Unexpected role of B and T lymphocyte attenuator in sustaining cell survival during chronic allostimulation
.
J Immunol
.
2007
;
178
(
10
):
6073
-
6082
.
77.
Sakoda
Y
,
Park
JJ
,
Zhao
Y
, et al
.
Dichotomous regulation of GVHD through bidirectional functions of the BTLA-HVEM pathway
.
Blood
.
2011
;
117
(
8
):
2506
-
2514
.
78.
Albring
JC
,
Sandau
MM
,
Rapaport
AS
, et al
.
Targeting of B and T lymphocyte associated (BTLA) prevents graft-versus-host disease without global immunosuppression
.
J Exp Med
.
2010
;
207
(
12
):
2551
-
2559
.
79.
del Rio
ML
,
Jones
ND
,
Buhler
L
, et al
.
Selective blockade of herpesvirus entry mediator-B and T lymphocyte attenuator pathway ameliorates acute graft-versus-host reaction
.
J Immunol
.
2012
;
188
(
10
):
4885
-
4896
.
80.
Dougall
WC
,
Kurtulus
S
,
Smyth
MJ
,
Anderson
AC
.
TIGIT and CD96: new checkpoint receptor targets for cancer immunotherapy
.
Immunol Rev
.
2017
;
276
(
1
):
112
-
120
.
81.
Nabekura
T
,
Shibuya
K
,
Takenaka
E
, et al
.
Critical role of DNAX accessory molecule-1 (DNAM-1) in the development of acute graft-versus-host disease in mice
.
Proc Natl Acad Sci USA
.
2010
;
107
(
43
):
18593
-
18598
.
82.
Koyama
M
,
Kuns
RD
,
Olver
SD
, et al
.
Promoting regulation via the inhibition of DNAM-1 after transplantation
.
Blood
.
2013
;
121
(
17
):
3511
-
3520
.
83.
Levin
SD
,
Taft
DW
,
Brandt
CS
, et al
.
Vstm3 is a member of the CD28 family and an important modulator of T-cell function
.
Eur J Immunol
.
2011
;
41
(
4
):
902
-
915
.
84.
Kanaya
M
,
Shibuya
K
,
Hirochika
R
, et al
.
Soluble DNAM-1, as a predictive biomarker for acute graft-versus-host disease
.
PLoS One
.
2016
;
11
(
6
):
e0154173
.
85.
Hattori
N
,
Kawaguchi
Y
,
Sasaki
Y
, et al
.
Monitoring TIGIT/DNAM-1 and PVR/PVRL2 immune checkpoint expression levels in allogeneic stem cell transplantation for acute myeloid leukemia
.
Biol Blood Marrow Transplant
.
2019
;
25
(
5
):
861
-
867
.
86.
Blazar
BR
,
Carreno
BM
,
Panoskaltsis-Mortari
A
, et al
.
Blockade of programmed death-1 engagement accelerates graft-versus-host disease lethality by an IFN-gamma-dependent mechanism
.
J Immunol
.
2003
;
171
(
3
):
1272
-
1277
.
87.
Asakura
S
,
Hashimoto
D
,
Takashima
S
, et al
.
Alloantigen expression on non-hematopoietic cells reduces graft-versus-leukemia effects in mice
.
J Clin Invest
.
2010
;
120
(
7
):
2370
-
2378
.
88.
Ijaz
A
,
Khan
AY
,
Malik
SU
, et al
.
Significant risk of graft-versus-host disease with exposure to checkpoint inhibitors before and after allogeneic transplantation
.
Biol Blood Marrow Transplant
.
2019
;
25
(
1
):
94
-
99
.
89.
Merryman
RW
,
Kim
HT
,
Zinzani
PL
, et al
.
Safety and efficacy of allogeneic hematopoietic stem cell transplant after PD-1 blockade in relapsed/refractory lymphoma
.
Blood
.
2017
;
129
(
10
):
1380
-
1388
.
90.
Schoch
LK
,
Cooke
KR
,
Wagner-Johnston
ND
, et al
.
Immune checkpoint inhibitors as a bridge to allogeneic transplantation with posttransplant cyclophosphamide
.
Blood Adv
.
2018
;
2
(
17
):
2226
-
2229
.
91.
Herbaux
C
,
Merryman
R
,
Devine
S
, et al
.
Recommendations for managing PD-1 blockade in the context of allogeneic HCT in Hodgkin lymphoma: taming a necessary evil
.
Blood
.
2018
;
132
(
1
):
9
-
16
.
92.
Ravandi
F
,
Assi
R
,
Daver
N
, et al
.
Idarubicin, cytarabine, and nivolumab in patients with newly diagnosed acute myeloid leukaemia or high-risk myelodysplastic syndrome: a single-arm, phase 2 study
.
Lancet Haematol
.
2019
;
6
(
9
):
e480
-
e488
.
93.
Wang
M
,
Bu
J
,
Zhou
M
, et al
.
CD8+T cells expressing both PD-1 and TIGIT but not CD226 are dysfunctional in acute myeloid leukemia (AML) patients
.
Clin Immunol
.
2018
;
190
:
64
-
73
.
94.
Wang
L
,
Rubinstein
R
,
Lines
JL
, et al
.
VISTA, a novel mouse Ig superfamily ligand that negatively regulates T cell responses
.
J Exp Med
.
2011
;
208
(
3
):
577
-
592
.
95.
Gao
J
,
Ward
JF
,
Pettaway
CA
, et al
.
VISTA is an inhibitory immune checkpoint that is increased after ipilimumab therapy in patients with prostate cancer
.
Nat Med
.
2017
;
23
(
5
):
551
-
555
.
96.
Wang
J
,
Wu
G
,
Manick
B
, et al
.
VSIG-3 as a ligand of VISTA inhibits human T-cell function
.
Immunology
.
2019
;
156
(
1
):
74
-
85
.
97.
Mehta
N
,
Maddineni
S
,
Mathews
II
,
Andres Parra Sperberg
R
,
Huang
PS
,
Cochran
JR
.
Structure and functional binding epitope of V-domain Ig suppressor of T cell activation
.
Cell Rep
.
2019
;
28
(
10
):
2509
-
2516.e2505
.
98.
Johnston
RJ
,
Su
LJ
,
Pinckney
J
, et al
.
VISTA is an acidic pH-selective ligand for PSGL-1
.
Nature
.
2019
;
574
(
7779
):
565
-
570
.
99.
Saha
A
,
Taylor
PA
,
Lees
CJ
, et al
.
Donor and host B7-H4 expression negatively regulates acute graft-versus-host disease lethality
.
JCI Insight
.
2019
;
4
(
19
):
e127716
.
100.
Laman
JD
,
Claassen
E
,
Noelle
RJ
.
Functions of CD40 and its ligand, gp39 (CD40L)
.
Crit Rev Immunol
.
2017
;
37
(
2-6
):
371
-
420
.
101.
Larsen
CP
,
Elwood
ET
,
Alexander
DZ
, et al
.
Long-term acceptance of skin and cardiac allografts after blocking CD40 and CD28 pathways
.
Nature
.
1996
;
381
(
6581
):
434
-
438
.
102.
Ohata
J
,
Sakurai
J
,
Saito
K
,
Tani
K
,
Asano
S
,
Azuma
M
.
Differential graft-versus-leukaemia effect by CD28 and CD40 co-stimulatory blockade after graft-versus-host disease prophylaxis
.
Clin Exp Immunol
.
2002
;
129
(
1
):
61
-
68
.
103.
Tamada
K
,
Tamura
H
,
Flies
D
, et al
.
Blockade of LIGHT/LTbeta and CD40 signaling induces allospecific T cell anergy, preventing graft-versus-host disease
.
J Clin Invest
.
2002
;
109
(
4
):
549
-
557
.
104.
Saito
K
,
Sakurai
J
,
Ohata
J
, et al
.
Involvement of CD40 ligand-CD40 and CTLA4-B7 pathways in murine acute graft-versus-host disease induced by allogeneic T cells lacking CD28
.
J Immunol
.
1998
;
160
(
9
):
4225
-
4231
.
105.
Visvanathan
S
,
Daniluk
S
,
Ptaszyński
R
, et al
.
Effects of BI 655064, an antagonistic anti-CD40 antibody, on clinical and biomarker variables in patients with active rheumatoid arthritis: a randomised, double-blind, placebo-controlled, phase IIa study
.
Ann Rheum Dis
.
2019
;
78
(
6
):
754
-
760
.
106.
Kawai
T
,
Andrews
D
,
Colvin
RB
,
Sachs
DH
,
Cosimi
AB
.
Thromboembolic complications after treatment with monoclonal antibody against CD40 ligand
.
Nat Med
.
2000
;
6
(
2
):
114
.
107.
Boumpas
DT
,
Furie
R
,
Manzi
S
, et al;
BG9588 Lupus Nephritis Trial Group
.
A short course of BG9588 (anti-CD40 ligand antibody) improves serologic activity and decreases hematuria in patients with proliferative lupus glomerulonephritis
.
Arthritis Rheum
.
2003
;
48
(
3
):
719
-
727
.
108.
Karnell
JL
,
Rieder
SA
,
Ettinger
R
,
Kolbeck
R
.
Targeting the CD40-CD40L pathway in autoimmune diseases: Humoral immunity and beyond
.
Adv Drug Deliv Rev
.
2019
;
141
:
92
-
103
.
109.
Kinnear
G
,
Wood
KJ
,
Fallah-Arani
F
,
Jones
ND
.
A diametric role for OX40 in the response of effector/memory CD4+ T cells and regulatory T cells to alloantigen
.
J Immunol
.
2013
;
191
(
3
):
1465
-
1475
.
110.
Vu
MD
,
Xiao
X
,
Gao
W
, et al
.
OX40 costimulation turns off Foxp3+ Tregs
.
Blood
.
2007
;
110
(
7
):
2501
-
2510
.
111.
Zhang
X
,
Xiao
X
,
Lan
P
, et al
.
OX40 costimulation inhibits Foxp3 expression and Treg induction via BATF3-dependent and independent mechanisms
.
Cell Rep
.
2018
;
24
(
3
):
607
-
618
.
112.
Jacquemin
C
,
Augusto
JF
,
Scherlinger
M
, et al
.
OX40L/OX40 axis impairs follicular and natural Treg function in human SLE
.
JCI Insight
.
2018
;
3
(
24
):
e122167
.
113.
Tkachev
V
,
Furlan
SN
,
Watkins
B
, et al
.
Combined OX40L and mTOR blockade controls effector T cell activation while preserving Treg reconstitution after transplant
.
Sci Transl Med
.
2017
;
9
(
408
):
eaan3085
.
114.
Radtke
F
,
MacDonald
HR
,
Tacchini-Cottier
F
.
Regulation of innate and adaptive immunity by Notch
.
Nat Rev Immunol
.
2013
;
13
(
6
):
427
-
437
.
115.
Zhang
Y
,
Sandy
AR
,
Wang
J
, et al
.
Notch signaling is a critical regulator of allogeneic CD4+ T-cell responses mediating graft-versus-host disease
.
Blood
.
2011
;
117
(
1
):
299
-
308
.
116.
Chung
J
,
Ebens
CL
,
Perkey
E
, et al
.
Fibroblastic niches prime T cell alloimmunity through Delta-like Notch ligands
.
J Clin Invest
.
2017
;
127
(
4
):
1574
-
1588
.
117.
Tran
IT
,
Sandy
AR
,
Carulli
AJ
, et al
.
Blockade of individual Notch ligands and receptors controls graft-versus-host disease
.
J Clin Invest
.
2013
;
123
(
4
):
1590
-
1604
.
118.
Tkachev
V
,
Kuhnert
F
,
Furlan
SN
, et al
.
Pharmacologic blockade of Notch/Delta-like ligand 4 signaling protects from gastrointestinal acute graft-versus-host disease in non-human primates [abstract]
.
Blood
.
2018
;
132
(suppl 1).
Abstract 2027
.
119.
Kishimoto
T
.
Interleukin-6: from basic science to medicine--40 years in immunology
.
Annu Rev Immunol
.
2005
;
23
(
1
):
1
-
21
.
120.
Hu
B
,
Elinav
E
,
Huber
S
, et al
.
Microbiota-induced activation of epithelial IL-6 signaling links inflammasome-driven inflammation with transmissible cancer [published correction appears in Proc Natl Acad Sci USA. 2013;110(31):12852]
.
Proc Natl Acad Sci USA
.
2013
;
110
(
24
):
9862
-
9867
.
121.
Kuhn
KA
,
Schulz
HM
,
Regner
EH
, et al
.
Bacteroidales recruit IL-6-producing intraepithelial lymphocytes in the colon to promote barrier integrity
.
Mucosal Immunol
.
2018
;
11
(
2
):
357
-
368
.
122.
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
.
123.
Varelias
A
,
Gartlan
KH
,
Kreijveld
E
, et al
.
Lung parenchyma-derived IL-6 promotes IL-17A-dependent acute lung injury after allogeneic stem cell transplantation
.
Blood
.
2015
;
125
(
15
):
2435
-
2444
.
124.
Drobyski
WR
,
Szabo
A
,
Zhu
F
, et al
.
Tocilizumab, tacrolimus and methotrexate for the prevention of acute graft-versus-host disease: low incidence of lower gastrointestinal tract disease
.
Haematologica
.
2018
;
103
(
4
):
717
-
727
.
125.
Wilkinson
AN
,
Chang
K
,
Kuns
RD
, et al
.
IL-6 dysregulation originates in dendritic cells and mediates graft-versus-host disease via classical signaling
.
Blood
.
2019
;
134
(
23
):
2092
-
2106
.
126.
Kang
S
,
Tanaka
T
,
Narazaki
M
,
Kishimoto
T
.
Targeting Interleukin-6 Signaling in Clinic
.
Immunity
.
2019
;
50
(
4
):
1007
-
1023
.
127.
Gartlan
KH
,
Markey
KA
,
Varelias
A
, et al
.
Tc17 cells are a proinflammatory, plastic lineage of pathogenic CD8+ T cells that induce GVHD without antileukemic effects
.
Blood
.
2015
;
126
(
13
):
1609
-
1620
.
128.
Gartlan
KH
,
Varelias
A
,
Koyama
M
, et al
.
Th17 plasticity and transition toward a pathogenic cytokine signature are regulated by cyclosporine after allogeneic SCT
.
Blood Adv
.
2017
;
1
(
6
):
341
-
351
.
129.
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
.
130.
Zhang
P
,
Lee
JS
,
Gartlan
KH
, et al
.
Eomesodermin promotes the development of type 1 regulatory T (TR1) cells
.
Sci Immunol
.
2017
;
2
(
10
):
eaah7152
.
131.
Belle
L
,
Agle
K
,
Zhou
V
, et al
.
Blockade of interleukin-27 signaling reduces GVHD in mice by augmenting Treg reconstitution and stabilizing Foxp3 expression
.
Blood
.
2016
;
128
(
16
):
2068
-
2082
.
132.
Tait Wojno
ED
,
Hunter
CA
,
Stumhofer
JS
.
The immunobiology of the interleukin-12 family: room for discovery
.
Immunity
.
2019
;
50
(
4
):
851
-
870
.
133.
Das
R
,
Komorowski
R
,
Hessner
MJ
, et al
.
Blockade of interleukin-23 signaling results in targeted protection of the colon and allows for separation of graft-versus-host and graft-versus-leukemia responses
.
Blood
.
2010
;
115
(
25
):
5249
-
5258
.
134.
Pidala
J
,
Beato
F
,
Kim
J
, et al
.
In vivo IL-12/IL-23p40 neutralization blocks Th1/Th17 response after allogeneic hematopoietic cell transplantation
.
Haematologica
.
2018
;
103
(
3
):
531
-
539
.
135.
Blauvelt
A
,
Papp
KA
,
Griffiths
CE
, et al
.
Efficacy and safety of guselkumab, an anti-interleukin-23 monoclonal antibody, compared with adalimumab for the continuous treatment of patients with moderate to severe psoriasis: Results from the phase III, double-blinded, placebo- and active comparator-controlled VOYAGE 1 trial
.
J Am Acad Dermatol
.
2017
;
76
(
3
):
405
-
417
.
136.
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
.
137.
Zeiser
R
,
Blazar
BR
.
Acute graft-versus-host disease–biologic process, prevention, and therapy
.
N Engl J Med
.
2017
;
377
(
22
):
2167
-
2179
.
138.
Zeiser
R
,
Blazar
BR
.
Pathophysiology of chronic graft-versus-host disease and therapeutic targets
.
N Engl J Med
.
2017
;
377
(
26
):
2565
-
2579
.
139.
Ross
SH
,
Cantrell
DA
.
Signaling and function of interleukin-2 in T lymphocytes
.
Annu Rev Immunol
.
2018
;
36
(
1
):
411
-
433
.
140.
Clipstone
NA
,
Crabtree
GR
.
Identification of calcineurin as a key signalling enzyme in T-lymphocyte activation
.
Nature
.
1992
;
357
(
6380
):
695
-
697
.
141.
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
.
142.
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
.
143.
Betts
BC
,
Pidala
J
,
Kim
J
, et al
.
IL-2 promotes early Treg reconstitution after allogeneic hematopoietic cell transplantation
.
Haematologica
.
2017
;
102
(
5
):
948
-
957
.
144.
Locke
FL
,
Pidala
J
,
Storer
B
, et al
.
CD25 blockade delays regulatory T cell reconstitution and does not prevent graft-versus-host disease after allogeneic hematopoietic cell transplantation
.
Biol Blood Marrow Transplant
.
2017
;
23
(
3
):
405
-
411
.
145.
Boyman
O
,
Kovar
M
,
Rubinstein
MP
,
Surh
CD
,
Sprent
J
.
Selective stimulation of T cell subsets with antibody-cytokine immune complexes
.
Science
.
2006
;
311
(
5769
):
1924
-
1927
.
146.
Burman
AC
,
Banovic
T
,
Kuns
RD
, et al
.
IFNgamma differentially controls the development of idiopathic pneumonia syndrome and GVHD of the gastrointestinal tract
.
Blood
.
2007
;
110
(
3
):
1064
-
1072
.
147.
Gaffen
SL
.
Structure and signalling in the IL-17 receptor family
.
Nat Rev Immunol
.
2009
;
9
(
8
):
556
-
567
.
148.
Kappel
LW
,
Goldberg
GL
,
King
CG
, et al
.
IL-17 contributes to CD4-mediated graft-versus-host disease
.
Blood
.
2009
;
113
(
4
):
945
-
952
.
149.
Yu
Y
,
Wang
D
,
Liu
C
, et al
.
Prevention of GVHD while sparing GVL effect by targeting Th1 and Th17 transcription factor T-bet and RORγt in mice
.
Blood
.
2011
;
118
(
18
):
5011
-
5020
.
150.
Varelias
A
,
Ormerod
KL
,
Bunting
MD
, et al
.
Acute graft-versus-host disease is regulated by an IL-17-sensitive microbiome
.
Blood
.
2017
;
129
(
15
):
2172
-
2185
.
151.
Varelias
A
,
Bunting
MD
,
Ormerod
KL
, et al
.
Recipient mucosal-associated invariant T cells control GVHD within the colon
.
J Clin Invest
.
2018
;
128
(
5
):
1919
-
1936
.
152.
Langley
RG
,
Elewski
BE
,
Lebwohl
M
, et al;
ERASURE Study Group
;
FIXTURE Study Group
.
Secukinumab in plaque psoriasis--results of two phase 3 trials
.
N Engl J Med
.
2014
;
371
(
4
):
326
-
338
.
153.
Braun
J
,
Baraliakos
X
,
Deodhar
A
, et al;
MEASURE 1 study group
.
Effect of secukinumab on clinical and radiographic outcomes in ankylosing spondylitis: 2-year results from the randomised phase III MEASURE 1 study
.
Ann Rheum Dis
.
2017
;
76
(
6
):
1070
-
1077
.
154.
Blanco
FJ
,
Möricke
R
,
Dokoupilova
E
, et al
.
Secukinumab in active rheumatoid arthritis: a phase III randomized, double-blind, active comparator- and placebo-controlled study
.
Arthritis Rheumatol
.
2017
;
69
(
6
):
1144
-
1153
.
155.
Furlan
SN
,
Watkins
B
,
Tkachev
V
, et al
.
Systems analysis uncovers inflammatory Th/Tc17-driven modules during acute GVHD in monkey and human T cells
.
Blood
.
2016
;
128
(
21
):
2568
-
2579
.
156.
Daro
E
,
Pulendran
B
,
Brasel
K
, et al
.
Polyethylene glycol-modified GM-CSF expands CD11b(high)CD11c(high) but notCD11b(low)CD11c(high) murine dendritic cells in vivo: a comparative analysis with Flt3 ligand
.
J Immunol
.
2000
;
165
(
1
):
49
-
58
.
157.
Gartlan
KH
,
Koyama
M
,
Lineburg
KE
, et al
.
Donor T-cell-derived GM-CSF drives alloantigen presentation by dendritic cells in the gastrointestinal tract
.
Blood Adv
.
2019
;
3
(
19
):
2859
-
2865
.
158.
Ouyang
W
,
O’Garra
A
.
IL-10 family cytokines IL-10 and IL-22: from basic science to clinical translation
.
Immunity
.
2019
;
50
(
4
):
871
-
891
.
159.
Zhang
P
,
Hill
GR
.
Interleukin-10 mediated immune regulation after stem cell transplantation: mechanisms and implications for therapeutic intervention
.
Semin Immunol
.
2019
;
44
:
101322
.
160.
Blazar
BR
,
Taylor
PA
,
Panoskaltsis-Mortari
A
, et al
.
Interleukin-10 dose-dependent regulation of CD4+ and CD8+ T cell-mediated graft-versus-host disease
.
Transplantation
.
1998
;
66
(
9
):
1220
-
1229
.
161.
Banovic
T
,
MacDonald
KP
,
Morris
ES
, et al
.
TGF-beta in allogeneic stem cell transplantation: friend or foe?
Blood
.
2005
;
106
(
6
):
2206
-
2214
.
162.
Alexander
KA
,
Flynn
R
,
Lineburg
KE
, et al
.
CSF-1-dependant donor-derived macrophages mediate chronic graft-versus-host disease
.
J Clin Invest
.
2014
;
124
(
10
):
4266
-
4280
.
163.
Rutz
S
,
Wang
X
,
Ouyang
W
.
The IL-20 subfamily of cytokines--from host defence to tissue homeostasis
.
Nat Rev Immunol
.
2014
;
14
(
12
):
783
-
795
.
164.
Lindemans
CA
,
Calafiore
M
,
Mertelsmann
AM
, et al
.
Interleukin-22 promotes intestinal-stem-cell-mediated epithelial regeneration
.
Nature
.
2015
;
528
(
7583
):
560
-
564
.
165.
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
.
166.
Lamarthée
B
,
Malard
F
,
Gamonet
C
, et al
.
Donor interleukin-22 and host type I interferon signaling pathway participate in intestinal graft-versus-host disease via STAT1 activation and CXCL10
.
Mucosal Immunol
.
2016
;
9
(
2
):
309
-
321
.
167.
Couturier
M
,
Lamarthée
B
,
Arbez
J
, et al
.
IL-22 deficiency in donor T cells attenuates murine acute graft-versus-host disease mortality while sparing the graft-versus-leukemia effect
.
Leukemia
.
2013
;
27
(
7
):
1527
-
1537
.
168.
Gartlan
KH
,
Bommiasamy
H
,
Paz
K
, et al
.
A critical role for donor-derived IL-22 in cutaneous chronic GVHD
.
Am J Transplant
.
2018
;
18
(
4
):
810
-
820
.
Sign in via your Institution